You are on page 1of 36

Periodontology 2000, Vol. 75, 2017, 116–151 © 2017 John Wiley & Sons A/S.

& Sons A/S. Published by John Wiley & Sons Ltd


Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Revisiting the Page & Schroeder


model: the good, the bad and the
unknowns in the periodontal
host response 40 years later
G E O R G E H A J I S H E N G A L L I S & J O N A T H A N M. K O R O S T O F F

The plaque-induced forms of periodontal disease – infiltrating) and structural events progressing from
gingivitis and periodontitis – are extremely prevalent periodontal health to disease. These included: (i)
chronic inflammatory conditions that affect distinct increasing complexity of the composition of the cellu-
components of the periodontium (8). In gingivitis, the lar infiltrate from one dominated by neutrophils (ini-
more benign of the two conditions, the inflammatory tial lesion) to one containing large numbers of
process is limited to the gingival epithelium and con- macrophages and T-cells (early lesion) and finally to
nective tissue. In its most severe form, the clinical one with a preponderance of plasma cells (estab-
manifestations of gingivitis include breakdown of the lished and advanced lesions); (ii) proliferation of
epithelial and connective tissue attachment of the basal cells and eventual apical migration of the junc-
gingiva to the teeth and the formation of gingival tional epithelium; (iii) cytopathologic changes in
pockets. In contrast, the hallmark of periodontitis is fibroblasts and increasing loss of collagen in the gin-
an immunoinflammatory infiltrate of the deeper gival connective tissue; and (iv) bone loss in the
compartments of the periodontium, resulting in advanced lesion.
destruction of the tooth-supporting tissues (cemen- Although there was limited knowledge regarding
tum, periodontal ligament and alveolar bone), tooth the identities and roles of periodontitis-associated
mobility and, ultimately, tooth loss. Furthermore, bacterial species at the time that Page & Schroeder
moderate-to-severe periodontitis is associated with co-authored their landmark paper, they were quick to
increased risk for certain systemic disorders (e.g. realize that the bacteria were an essential, but not a
atherosclerosis and rheumatoid arthritis) (83). sufficient, cause of periodontitis. They observed that
In 1976, Roy Page & Hubert Schroeder published the established lesion (moderate-to-severe gingivitis)
their classic paper ‘Pathogenesis of chronic inflam- did not necessarily progress to periodontitis but could
matory periodontal disease: A summary of the cur- remain stable indefinitely, leading them to conclude
rent work’ (213). Based on evaluation of tissue that the host response to the bacterial challenge plays
obtained from both human and animal models of a key role in determining the extent and severity of
naturally occurring and experimentally induced dis- disease (213).
ease, the authors provided a histopathologic ‘road- Capitalizing on the concept that periodontitis
map’ of the events leading to clinically detectable results from host–microbe interactions, Page, along
gingivitis and periodontitis. In describing the host with Kenneth Kornman and Maurizio Tonetti, pub-
response to the accumulation of dental plaque, they lished a manuscript in 1997 entitled ‘The host
defined four lesions. The initial, early and established response to the microbial challenge in periodontitis:
lesions exemplified distinct stages of gingivitis, and assembling the players’ (137). The authors utilized
the advanced lesion was equated with overt peri- the analogy of putting on a theatrical production and
odontitis. Limited by the available knowledge regard- described how the stage changes as the storyline pro-
ing the induction and regulation of inflammatory and gresses from health through disease progression.
immunologic reactions, the paper provided a descrip- They proposed four ‘scenes’, each of which included
tion of the sequential cellular (stromal and its histologic features, predominant cell types and a

116
Revisiting the Page & Schroeder model

list of key molecular players. As with the original and beyond an initial obstacle to bacterial challenge
paper, these authors stressed that progression from (66, 76, 84, 202). The various components of the
one scene to the next was not an inevitable process. innate response determine if and when an immuno-
Rather, they suggested that bacteria and/or their logic response is necessary and thus play both induc-
products, as well as intrinsic and extrinsic factors tive and regulatory roles in these host responses.
that modify an individual’s response to the bacterial With respect to the adaptive immune system itself,
challenge, collectively determine progression (or not) specialized functional subsets of dendritic cells,
from one scene to the next. The four scenes were: (i) macrophages, T-cells and B-cells have been identified
an acute bacterial challenge phase (representing the that have revolutionized traditional concepts on the
response to the early colonizers of the acquired pelli- roles of antigen-presenting cells and lymphocytes, as
cle); (ii) an acute inflammatory phase (a mild inflam- well as the mechanisms regulating the collective
matory response representing a reactive host response (191, 203). Moreover, it is now appreciated
response to bacterial challenge); (iii) an immune that neutrophils, historically regarded as simply
response phase (activation of numerous types of antimicrobial effector cells, exhibit significant func-
mononuclear cells that mediate and regulate the tional versatility, including regulation of other leuko-
local and systemic immune responses); and (iv) a cytes (93, 241). During this time, it was also
regulation and resolution phase (representing the demonstrated that the junctional epithelium can ini-
point at which a normal protective host response tiate and modulate numerous aspects of plaque-
may deviate toward a destructive chronic immunoin- induced gingival inflammation (22). At the same time,
flammatory process). This interpretation further we came to appreciate that local tissues have a ‘regu-
underscored the complexity of the interactions latory say’ over the host inflammatory response
between the bacterial challenge, host response and through several mechanisms including local
modifying factors that contribute to the ultimate out- production of homeostatic molecules (84, 172, 262).
come, disease protection or progression. Recent In addition, an entirely new field of study – osteoim-
microbiome and mechanistic studies revealed that: munology – has emerged as we have come to
(i) the periodontitis-associated microbiota is much acknowledge the impact of immunoinflammatory
more diverse and complex than traditionally events on the cells that mediate bone formation and
thought; and (ii) its role in disease involves polymi- resorption (194) (Fig. 1).
crobial synergistic and dysbiotic interactions with Therefore, the goal of this review is to update the
the host (4, 49, 52, 90, 143). Although the emerging current state of our knowledge regarding the role of
understanding is that periodontitis represents dys- the host response in periodontal disease pathogenesis
biosis rather than infection, the original concept that by discussing these new concepts in the context of
bacteria are necessary, but not sufficient, for peri- the lesions described by Page & Schroeder 40 years
odontitis (thus requiring a susceptible host) is still ago.
valid.
Although much of what Page & Schroeder proposed
in 1976 (213) has stood the test of time, advances in Neutrophils
the fields of basic and periodontal immunology
necessitate a reassessment of their work as well as its
Recent developments in neutrophil
integration with emerging new concepts. Major
biology
advances have been made regarding the cellular and
molecular mechanisms underlying the induction, reg- Healthy human gingiva displays coordinated gradi-
ulation and effector functions of immune and inflam- ents of chemokines and adhesion molecules that are
matory responses. Chief amongst these are major thought to provide chemotactic and haptotactic cues
developments in our understanding of the mecha- for the directed migration of neutrophils to the gingi-
nisms of innate immunity and how they interface val crevice. The concentration of these molecules
with acquired immune responses. We now recognize increases from the basal cells of the junctional epithe-
that innate immunity is not simply a series of physi- lium toward its outer surface that forms the floor of
cal, chemical and physiologic barriers, but an extre- the crevice and is therefore closer to the bacterial
mely complex and highly regulated network of cells challenge (79, 276, 299). The coordinated and regu-
and molecules (proinflammatory and regulatory lated recruitment of neutrophils is vital for periodon-
cytokines and chemokines, adhesion molecules and tal tissue homeostasis. This notion is supported by
their ligands/counter-receptors) that function over clinical observations in individuals with neutrophil

117
Hajishengallis & Korostoff

Dysbiotic challenge

C
C5a ROS, MMP Connective
N tissue damage
CX G-C
C3a C5a C P
cy SF M TNF, IL-1
tok M
ine PGE2, MMP

TNF, IL-6, MMP

IL
DC s
FB

CCL2, CCL20
PR
C3d

,A
TG

yS

IL- Fβ, IL

BL
6, 7 7
IL- -1
IL-
1 IL-1
23 3
IL-2
IL-6, -6
IL-21 IL-10 IL -23
Protective? IL
IL-17 TGFβ

IL-12
IFNγ
Immune complex- Abs P B Th17 Treg
mediated damage? 17
RA
NK IL- IL
TG -10

RANKL
L

10
-5

-
IL Fβ

IL
,
-4 OBL RANK
IL L RANKL
Th1
OCL
IFNγ
Th2
IL-4
Bone resorption , IL-
13
Th2

Fig. 1. Innate–adaptive immune interplay leading to ligand; B, B-cell; BLyS, B-lymphocyte stimulator; C, com-
inflammatory tissue damage and bone loss in periodonti- plement; CCL, chemokine (C-C motif) ligand; CXC, chemo-
tis. Periodontitis arises from complex interactions between kine (C-X-C motif); DC, dendritic cell; FB, fibroblast; G-
the host and the subgingival dysbiotic microbiota that lead CSF, granulocyte-colony stimulating factor; IFN, inter-
to excessive or dysregulated inflammatory responses feron; IL, interleukin; Mφ, macrophage; MMP, matrix met-
involving elements of both innate (complement and alloproteinase; N, neutrophil; OBL, osteoblast; OCL,
phagocytes) and adaptive (regulatory and effector lympho- osteoclast, P, plasma cell; PGE2, prostaglandin E2; RANKL,
cytes) immunity. Shown is a simplified view of cytokine- receptor activator of nuclear factor-kappaB ligand; ROS,
and chemokine-mediated crosstalk interactions between reactive oxygen species; TGFb, transforming growth fac-
innate and adaptive immune cells leading to destruction tor-beta; Th1, T-helper type 1 cell; Th2, T-helper type 2
of connective tissue and bone in periodontitis. See the text cell; Th17, T-helper type 17 cell; TNF, tumor necrosis fac-
for details. Abs, antibodies; APRIL, a proliferation-inducing tor; Treg, regulatory T-cell.

deficiencies affecting absolute cell numbers (chronic chronic inflammatory disorders, including rheuma-
or cyclic neutropenia) or trafficking capacity (leuko- toid arthritis, psoriasis, atherosclerosis, inflammatory
cyte adhesion deficiencies); these individuals display bowel disease, diabetes and cancer (62, 134, 173, 241,
increased susceptibility to, and severity of, periodon- 279). Indeed, research in the past 10–15 years has
titis (80, 187). However, neutrophils can also be shown that neutrophils can greatly extend their lifes-
involved in periodontal tissue destruction if their pan and exhibit remarkable functional versatility and
recruitment is not properly regulated or if the micro- hitherto unsuspected roles, including regulatory
bial challenge in the periodontium cannot be con- interactions with both innate and adaptive immune
trolled. According to the Page & Schroeder model, the leukocytes (19, 182, 241). Rather than being rapidly
periodontal lesion is initiated as acute inflammation exhausted at peripheral tissues, neutrophils are cur-
characterized by increased numbers of neutrophils rently considered as being capable of migrating to
migrating into the gingival crevice through the junc- lymph nodes where they can interact with dendritic
tional epithelium (213). However, besides hallmark- cells to modulate antigen presentation, thereby par-
ing acute inflammation, neutrophils are now ticipating in the regulation of adaptive immunity
increasingly appreciated as important players in (167).

118
Revisiting the Page & Schroeder model

In addition to stored granule-derived antimicrobial developments that have promoted our understand-
molecules and enzymes, neutrophils are now ing of the fascinating roles of neutrophils in peri-
acknowledged for their de novo biosynthetic capacity odontal inflammation and bone loss.
for chemokines and cytokines with proinflammatory,
anti-inflammatory or immunoregulatory properties
Neutrophil persistence in periodontitis
(241). This previously underappreciated ability of
neutrophils facilitates their ability to network with In response to the tooth-associated biofilm, neu-
tissue-resident cells and other leukocytes. For trophils exit the gingival plexus of the postcapillary
instance, by releasing chemokine (C-C motif) ligand venules and are chemotactically recruited to the gin-
2 and chemokine (C-C motif) ligand 20, neutrophils gival crevice via the junctional epithelium, which,
can induce the recruitment of interleukin-17-produ- under inflammatory conditions, is largely occupied
cing CD4-positive T-helper 17 cells to sites of infec- by transiting neutrophils (48). Neutrophils are actu-
tion or inflammation (219). By secreting the ally the predominant leukocyte type (≥ 95%) in the
cytokines B-lymphocyte stimulator and a prolifera- gingival crevice (48), where they form a ‘defense wall’
tion-inducing ligand (APRIL), neutrophils can pro- against the biofilm, ostensibly to block bacterial inva-
mote the survival, proliferation and development of sion into the underlying gingival connective tissue
B-cells into antibody-secreting plasma cells (105, (236). Although neutrophils provide homeostatic
240) (Fig. 1). Activated neutrophils (e.g. from the immunity in clinically healthy gingiva, their defense
synovial fluid of patients with active rheumatoid mechanisms appear to be insufficient to control a
arthritis) were additionally shown to express mem- dysbiotic microbial community, despite their persis-
brane-bound RANKL, a key osteoclastogenic cyto- tent recruitment and capacity to elicit immune and
kine (16, 194), and thereby able to induce inflammatory responses (81, 236). This implies that
osteoclastic bone resorption (31). Emerging evidence periodontitis-associated bacteria can largely escape
also suggests that the neutrophil population may neutrophil-mediated killing in an inflammatory envi-
not be as homogeneous as traditionally thought but ronment, that is, without causing generalized
rather it is composed of cells that display substantial immunosuppression. In fact, the latter would not be
functional plasticity to form subsets with specialized in ‘their best interest’ because periodontal dysbiotic
cytotoxic or regulatory functions (38, 61, 64, 118, communities rely on inflammation to secure nutri-
167, 223). ents, such as degraded collagen and heme-containing
These recent concepts suggest that neutrophils compounds derived from inflammatory tissue
could contribute to periodontitis not only by initiat- breakdown (82).
ing the lesion but also by participating in its pro- A possible mechanism whereby periodontal bacte-
gression (e.g. by recruiting T-helper 17 cells or ria can uncouple neutrophil-mediated clearance from
promoting the accumulation of B-cells and plasma neutrophil-facilitated inflammation was dissected
cells in the established and advanced lesions). using Porphyromonas gingivalis as a model organism.
Moreover, as neutrophils dwell and become acti- This unique bacterium is a keystone pathogen that
vated within the gingival connective tissue under can manipulate innate immunity in ways that pro-
severe inflammatory conditions (212), these cells mote the conversion of a symbiotic community into a
have the opportunity to contribute to bone dem- dysbiotic one (83, 90). Porphyromonas gingivalis can
ineralization and matrix degradation by releasing co-activate complement C5a receptor-1 and toll-like
matrix metalloproteinases, such as collagenase, and receptor-2 in human neutrophils, resulting in signal-
expressing RANKL (31, 103, 147). Intriguingly, in ing crosstalk that leads to the ubiquitylation and pro-
aggressive forms of periodontitis associated with teasomal degradation of the toll-like receptor-2
neutrophil-related primary immunodeficiencies, the adaptor myeloid differentiation primary response
disease may not actually be the result of impaired protein-88, thereby suppressing a host-protective
neutrophil immune surveillance but may – alterna- antimicrobial response. Moreover, this C5a receptor-
tively or additionally – derive from disruption of 1–toll-like receptor-2 crosstalk utilizes another toll-
homeostatic mechanisms that are normally per- like receptor-2 adaptor (the myeloid differentiation
formed by neutrophils (188). In other words, neu- primary response protein-88-like adaptor protein)
trophils can potentially contribute to periodontitis and activates phosphoinositide 3-kinase, which
through both their presence and absence, suggest- blocks phagocytosis through the inhibition of RhoA
ing that neutrophil homeostasis is key to periodon- GTPase and actin polymerization, while at the same
tal health. Below we discuss in greater detail recent time stimulating the production of inflammatory

119
Hajishengallis & Korostoff

cytokines (162). In mice, oral colonization with P. gin- signaling, which inhibits activation of beta2-integrin
givalis leads to the emergence of a dysbiotic and in response to chemoattractants (286). Endothelial
inflammation-provoking microbiota in the periodon- cell-derived developmental endothelial locus-1 is a
tium, but pharmacological blockade of C5a receptor-1 homeostatic molecule that binds the beta2-integrin
or toll-like receptor-2 leads to near elimination of lymphocyte function-associated antigen-1 (CD11a/
P. gingivalis and reversal of inflammation (162). CD18), and antagonizes its interaction with endothe-
The data from this model suggest a periodontal lial intercellular adhesion molecule-1, thereby
host–microbe interplay that perpetuates chronic inhibiting firm neutrophil adhesion to the endothe-
inflammation and recruitment of neutrophils that are lium (35). It is therefore feasible that the severity of
unable to control the dysbiotic challenge. Sufficient periodontal inflammation and tissue breakdown is
clinical evidence indicates that neutrophils mediate a influenced by the relative abundance of these
substantial portion of periodontal tissue destruction endogenous negative regulators of neutrophil
(101, 147) and that their numbers correlate positively recruitment.
with the severity of the disease (144). Furthermore, In this regard, developmental endothelial locus-1-
owing to persistent inflammation, patients with deficient mice develop spontaneous periodontitis
chronic periodontitis have longer-lived neutrophils in that is characterized by excessive neutrophil infiltra-
oral tissues compared with healthy individuals (140). tion and interleukin-17-mediated inflammatory bone
loss (59). This observation suggests that normal
expression of developmental of endothelial locus-1 is
Supernumerary and dysregulated
crucial for appropriate regulation of neutrophil
neutrophils in periodontitis
recruitment and the local host inflammatory
The extravasation of circulating neutrophils is tightly response. Interestingly, the expression of develop-
regulated and proceeds via the leukocyte adhesion mental endothelial locus-1 and interleukin-17 are
cascade, a sequence of low- and high-affinity adhe- inversely correlated in both human and murine peri-
sive interactions between the neutrophils and the odontal tissue, with developmental endothelial locus-
vascular endothelium (84, 222, 283). The first step 1 dominating in healthy gingiva and interleukin-17
involves transient rolling interactions between the dominating in periodontitis-involved gingiva (59).
neutrophils and the endothelium that is mediated by Moreover, expression of developmental endothelial
the binding of selectin ligands on neutrophils to P- or locus-1 mRNA and protein is decreased in the gingiva
E-selectin on endothelial cells. This rolling-dependent of aged mice (≥ 18 months of age), correlating with
slowing down of neutrophils facilitates transition to excessive neutrophil recruitment and interleukin-17-
firm adhesion on the endothelium, which is followed dependent inflammatory bone loss (59, 253). Accord-
by intraluminal crawling to identify appropriate sites ingly, intragingival administration of recombinant
for extravasation. Firm adhesion and crawling are lar- developmental endothelial locus-1 in old mice inhi-
gely mediated by activated beta2-integrins (CD11/ bits neutrophil infiltration and bone loss (59). Similar
CD18 heterodimers) and endothelial counter-recep- results were obtained by treating nonhuman primates
tors, such as intercellular adhesion molecules-1 and - with recombinant human developmental endothelial
2 (179, 222, 283). The neutrophil recruitment cascade locus-1 (254).
has long been established; however, only recently Although it is currently not known whether
have endogenous mechanisms that down-regulate humans experience decreased expression of develop-
the process been identified. These mechanisms can mental endothelial locus-1 in old age, as seen in
block distinct steps of the leukocyte adhesion cas- mice, recruited neutrophils in old individuals might
cade, such as (i) rolling, (ii) activation and (iii) firm be more likely to cause collateral tissue damage
adhesion. The regulatory molecules that mediate owing to age-related cell-intrinsic defects. In this
these effects are locally expressed by the endothelium regard, neutrophils isolated from elderly subjects
or by the recruited neutrophils upon their interaction (≥ 60 years of age) display unfocused or inaccurate
with endothelial cells (84). For instance, neutrophil- chemotaxis during which they release excessive
derived pentraxin-3 blocks rolling by binding P-selec- amounts of proteases (238). Mechanistically, this
tin on the endothelium and thus interfering with its defect was linked to elevated constitutive phospho-
interaction with P-selectin glycoprotein ligand-1 on inositide 3-kinase activity, as compared with neu-
neutrophils (46). The paired immunoglobulin-like trophils from young controls (238). In a related
type 2 receptor-alpha on neutrophils mediates context, neutrophils from patients with chronic peri-
immunoreceptor tyrosine-based inhibitory motif odontitis have decreased chemotactic accuracy

120
Revisiting the Page & Schroeder model

compared with healthy controls (231). Therefore, the the human interleukin-10 genomic locus could
recruitment of increased numbers of neutrophils with undergo major reorganization rendering it permissive
imprecise chemotaxis could exacerbate neutrophil- to activation (269). In this regard, lipopolysaccharide-
mediated bystander tissue damage. stimulated regulatory T-cells appear to induce chro-
In addition to elevated numbers, neutrophil hyper- matin modifications (H3K4me3, H3AcLys4) specific
activity or hyper-reactivity can also enhance bystan- for transcriptional activation of the interleukin-10
der tissue damage through the release of gene in human neutrophils (149). The biological sig-
inflammatory and cytotoxic mediators and enzymes nificance of the interleukin-10-producing neutrophils
(32, 40, 236, 257). For instance, peripheral blood neu- detected in periodontal pockets is currently uncer-
trophils from patients with chronic periodontitis tain. One possibility is that gram-negative bacteria
release higher levels of proinflammatory cytokines evade host immunity by programming regulatory T-
(e.g. tumor necrosis factor, interleukin-1beta and cells to suppress immune and antimicrobial
interleukin-8) than do healthy controls in response to responses of neutrophils. Alternatively, interleukin-
several stimuli. Interestingly, this hyper-inflammatory 10-producing neutrophils may represent a subset
phenotype persisted, even following successful involved in the mitigation of destructive inflamma-
periodontal therapy (155). Therefore, although of tion and/or in promoting the resolution of inflamma-
uncertain genetic or other mechanistic basis, this tion.
hyper-reactivity appears to be spontaneous rather Additional mechanisms have been described
than secondary to local periodontal inflammation. whereby neutrophils can down-regulate the host
However, long-lasting epigenetic effects on the phe- immune response. Indeed, neutrophils can directly
notype cannot be ruled out. Peripheral blood neu- suppress T-cell responses by releasing arginase-1,
trophils from patients with chronic periodontitis also which depletes arginine (required for T-cell activa-
show higher release of reactive oxygen species than tion) (189, 235), or indirectly by releasing myeloperox-
do those from healthy individuals, even in the idase or elastase, which inhibit the antigen-
absence of exogenous stimulation. Similarly to the presenting function of dendritic cells (163, 204)
hyper-inflammatory phenotype, this reactive oxygen (Fig. 2). Ectosomes (extracellular vesicles with
species-related hyperactivity was not corrected by immunosuppressive properties) released by human
successful periodontal treatment (171). neutrophils inhibit activation and promote an anti-
inflammatory phenotype in several types of innate
immune cells, including macrophages and natural
Neutrophil-dependent regulatory
killer cells (69, 237, 241) (Fig. 2), which are thought to
functions
contribute to destructive periodontal inflammation
A recent study has identified a subset of suppressor (290). Moreover, it was recently shown that a subset
neutrophils at sites of inflammation induced by of human mature neutrophils (CD16bright/CD62Ldim)
gram-negative bacteria (e.g. the periodontal pockets inhibits T-cell activation by delivering hydrogen per-
of patients with chronic periodontitis) but not at sites oxide into the immunological synapse in a Mac-1-
of aseptic inflammation (e.g. the cerebrospinal fluid dependent manner (223) (Fig. 2). It is conceivable,
of patients with neuromyelitis optica) (149). Specifi- therefore, that the presence of neutrophils may be
cally, these investigators showed that human neu- required for restraining excessive, and potentially
trophils produce interleukin-10 following direct harmful, T-cell activation in periodontitis.
contact with lipopolysaccharide-stimulated regula- Other recent developments do indicate a regulatory
tory T-cells. This interaction is mediated by the function for neutrophils in human periodontal dis-
beta2-integrin, Mac-1 (CD11b/CD18), on neutrophils ease, and specifically in the context of an aggressive
and by intercellular adhesion molecule-1 on regula- form of the disease associated with a primary immun-
tory T-cells (149). The ability of human neutrophils to odeficiency, leukocyte adhesion deficiency Type I-
express interleukin-10 was unexpected given that an associated periodontitis. The extent and severity of
earlier study showed that the IL10 genomic locus in attachment loss observed in subjects affected by this
human neutrophils stimulated with lipopolysaccha- condition constitutes unequivocal evidence that neu-
ride and other proinflammatory stimuli remains in an trophils are required for the maintenance of peri-
inactive state, in stark contrast to autologous mono- odontal health (45, 96, 187, 285). Leukocyte adhesion
cytes that can readily be induced to express inter- deficiency Type I is an autosomal-recessive immun-
leukin-10 (269). These authors, nevertheless, did not odeficiency caused by mutations in the ITGB2 gene
preclude as-yet-undefined conditions under which that encode the common subunit, CD18, of beta2-

121
Hajishengallis & Korostoff

Macrophage

Proinflammatory
cytokines
TGFβ1 secretion

Ectosomes

Allostimulation Elastase
TGFβ1 secretion Proinflammatory
Ectosomes cytokines
MPO Anti-inflammatory
Maturation
cytokines NK cell
DC Cytokine production
Neutrophil
Arginase-1
H2O2/Mac-1

Activation
Proliferation

T-cell
Fig. 2. Regulatory neutrophil cross-talk with other leuko- (MPO)- or elastase-dependent mechanisms that inhibit
cyte types. Neutrophils can potentially suppress T-cell acti- dendritic cell (DC) function. Furthermore, neutrophils can
vation by releasing arginase-1 (depletes arginine required release ectosomes that can down-regulate the inflamma-
for T-cell activation) or by delivering hydrogen peroxide tory activity of innate immune cells, such as macrophages
(H2O2) into the immunological synapse in a Mac-1 inte- and natural killer (NK) cells. TGFb1, transforming growth
grin-dependent manner. Neutrophils can also indirectly factor-beta1.
suppress T-cell activation through myeloperoxidase

integrins (96, 249). This deficiency in beta2-integrins (96, 249). Consequently, neutrophils are absent or
impedes normal neutrophil adhesion and extravasa- rarely found in extravascular sites in patients with
tion to peripheral tissues, including the periodontium leukocyte adhesion deficiency Type I who exhibit

Fig. 3. Biological functions of interleukin-17 and their bone by stimulating the production of matrix metallopro-
potential impact on periodontitis. Interleukin-17 acts pre- teinases and RANKL from the indicated stromal cell types.
dominantly on innate immune and stromal cells (e.g. Interleukin-17 is thus an immunological double-edged
fibroblasts, endothelial cells and epithelial cells) to pro- sword with both protective and destructive functions. As
mote innate immunity, especially neutrophil-mediated indicated, the current burden of evidence from human
antimicrobial and inflammatory responses. In this regard, and animal model studies suggests that the net effect of
interleukin-17 promotes granulopoiesis and neutrophil interleukin-17 signaling promotes periodontal disease
recruitment by up-regulating granulocyte-colony stimulat- development. Del-1, developmental endothelial locus-1;
ing factor and C-X-C motif (CXC) chemokines and by G-CSF, granulocyte-colony stimulating factor; ICAM-1,
down-regulating developmental endothelial locus-1, an intercellular adhesion molecule 1; IL, interleukin; LFA-1,
endogenous inhibitor of neutrophil adhesion and extrava- lymphocyte function-associated antigen 1; MMPs, matrix
sation. Moreover, interleukin-17 can induce the produc- metalloproteinases; PGE2, prostaglandin E2; RANKL,
tion of epithelial cell-derived antimicrobial molecules. On receptor activator of nuclear factor-kappaB ligand; TNF,
the other hand, interleukin-17 can contribute to the tumor necrosis factor. From Zenobia and Hajishengallis
destruction of both connective tissue and the underlying (298); used with permission.

122
Revisiting the Page & Schroeder model

Del-1
LFA-1
ICAM-1

Inflammation

CXC Neutrophil
chemokines recruitment
Downregulation
of Del-1 Antimicrobial
expression by functions
endothelial
cells
Granulopoiesis
and release to
circulation

G-CSF
MMPs
IL-17 (MMP-3,-9,-13)
Fibroblasts (and Connective
other cellular sources) tissue
degradation

RANKL

Osteoblasts Bone resorption


by osteoclasts

Proinflammatory
mediators (TNF,
IL-1β, IL-6, PGE2)
Macrophages (and
other cellular sources)

Antimicrobial mediators
(β-defensin-2, S100
proteins, cathelicidin)
Epithelial cells

Antimicrobial
immunity
Inflammation

Periodontitis

123
Hajishengallis & Korostoff

neutrophilia (elevated neutrophil counts in blood), standing puzzle of why individuals with chronic gran-
experience frequent infections at mucosal or skin sur- ulomatous disease, who are susceptible to infections
faces and develop severe periodontitis in childhood elsewhere in the body, do not exhibit increased sus-
(45, 96, 187, 285). Given the established antimicrobial ceptibility to periodontitis relative to the general pop-
activities of neutrophils and the observation that the ulation (201, 257). Although the neutrophils in
periodontal tissue of patients with leukocyte adhe- patients with chronic granulomatous disease have
sion deficiency Type I is specifically devoid of neu- defective oxygen-dependent killing (because of muta-
trophils, this form of periodontitis has been tions in the genes encoding components of the nicoti-
historically attributed to defective neutrophil surveil- namide adenine dinucleotide oxidase), their
lance of the periodontal bacterial infection (44, 45, recruitment to peripheral tissues, such as the gingiva,
129, 145, 201, 258, 285). A recent mechanistic study is intact. Therefore, normal recruitment of neutro-
that examined periodontitis in patients with leuko- phils may be more critical for the homeostasis of the
cyte adhesion deficiency Type I and in relevant periodontium than is the ability of neutrophils to kill
mouse models has linked impaired neutrophil trans- bacteria, a function that could be compensated for by
migration to dysregulated overexpression of predomi- other innate immune cells, such as macrophages. The
nantly T-cell-derived interleukin-17 (187), a notion that leukocyte adhesion deficiency Type I
proinflammatory and bone-resorptive cytokine (298) periodontitis does not represent an uncontrolled
(Fig. 3). Local antibody-mediated neutralization of infection is also consistent with the fact that this form
interleukin-17 in lymphocyte function-associated of periodontitis is not responsive to antibiotics and/
antigen-1-deficient mice, which mimic the human or mechanical removal of the bacterial biofilm (44,
leukocyte adhesion deficiency Type I phenotype, was 45, 187). This does not imply that the bacteria do not
shown to diminish periodontal inflammation and participate in the pathogenesis of leukocyte adhesion
bone loss and, moreover, to reduce the microbial bur- deficiency Type I periodontitis. In fact, the tooth-
den (187). These findings are consistent with disrup- associated microbial community serves as the local
tion of the ‘neutrostat’, a homeostatic mechanism stimulus to unleash the already dysregulated inter-
that senses neutrophil recruitment and clearance in leukin-23–interleukin-17 axis through translocation of
tissues and regulates neutrophil production through released bacterial lipopolysaccharide into the under-
a negative-feedback loop involving a granulopoietic lying gingival connective tissue (92, 185).
cascade of cytokines, specifically the interleukin-23/ As discussed above, mice deficient in developmen-
interleukin-17/granulocyte-colony stimulating factor tal endothelial locus-1 display excessive neutrophil
axis (260). When neutrophils cannot transmigrate to recruitment leading to inflammatory bone loss (59).
peripheral tissues, as in leukocyte adhesion defi- Therefore, with respect to neutrophil infiltration in
ciency Type I, this regulatory circuit breaks down, the periodontium, mice deficient in lymphocyte func-
leading to unrestrained expression of interleukin-23 tion-associated antigen-1 and mice deficient in devel-
and of the downstream cytokines interleukin-17 and opmental endothelial locus-1 display contrasting
granulocyte-colony stimulating factor. Whereas the phenotypes (59, 91, 187). However, mice deficient in
overproduction of granulocyte-colony stimulating lymphocyte function-associated antigen-1 and mice
factor explains the increased granulopoiesis and deficient in developmental endothelial locus-1 both
blood neutrophilia in patients with leukocyte adhe- develop spontaneous periodontitis, whereas wild-
sion deficiency Type I, the local overproduction of type littermate controls, or even their corresponding
interleukin-17 causes inflammation that leads to doubly deficient mice, maintain a healthy periodon-
bone loss and bacterial outgrowth (187). tium (59, 187). These findings suggest that periodon-
In this respect, it should be noted that inflamma- tal tissue homeostasis requires the presence of
tion can promote the growth of ‘inflammophilic’ bac- ‘normal’ numbers of neutrophils; neither ‘too few’
teria that thrive on inflammatory tissue breakdown nor ‘too many’.
products (82). The finding that microbial outgrowth
in leukocyte adhesion deficiency Type I periodontitis
Neutrophil subsets in the periodontium
can be controlled by inhibiting inflammation, despite
the absence of the presumed protective effects of Mounting evidence suggests the existence of distinct
neutrophils, questions earlier assumptions that the neutrophil subsets with diverse roles in infection,
absence of neutrophils promotes disease as a result of inflammation and cancer pathogenesis. In addition
defective surveillance of the periodontal microbiota. to subsets of neutrophils that perform regulatory
This concept also offers a new insight into the long- functions, as discussed above [e.g. the CD16bright/

124
Revisiting the Page & Schroeder model

CD62Ldim subset that inhibits T-cell activation Macrophages


(223)], there is precedent for the polarization of
tumor-associated neutrophils to phenotypes with
anti-tumor or protumorigenic activities (64). It is Macrophages in periodontitis: clinical
thought that tumor-associated neutrophils can be and mechanistic evidence
polarized toward a transforming growth factor-beta- There are relatively few macrophages in healthy gin-
dependent protumoral ‘N1’ or anti-tumoral ‘N2’ giva but their numbers are increased in gingival tissue
state that mirror M1 and M2 macrophages, respec- biopsies from patients with gingivitis or chronic
tively (64, 167). The presence of tumor-associated periodontitis (137). By responding to a variety of
neutrophils with proangiogenic and protumoral microbial conserved structures [e.g. bacterial
functions (N2) was also shown by an independent lipopolysaccharide, lipoproteins and nucleic acids
study (110). Regarding infection control, an investi- that are abundantly present in the periodontium
gation conducted in mice has demonstrated three (126)], macrophages constitute an important source
distinct neutrophil subsets that differ in their of proinflammatory and potentially tissue-destruc-
expression pattern of toll-like receptor, integrin and tive molecules (interleukin-1, tumor necrosis factor,
cytokine, and exhibit dissimilar susceptibilities to matrix metalloproteinases and prostaglandin E2)
methicillin-resistant Staphylococcus aureus (280). that are elevated in the gingival tissue and gingival
More recently, it was shown that human oral neu- crevicular fluid of patients with chronic periodonti-
trophils associated with mucosal health have a dif- tis (11, 76, 137) (Fig. 1). Therefore, macrophages
ferent phenotype from those derived from inflamed have been assumed by Page & Schroeder (213, 214)
mucosae (61). Specifically, in addition to resting/ to participate in the pathogenesis of periodontitis, a
naive circulatory neutrophils, the authors demon- notion that is supported – albeit indirectly – by clin-
strated the presence of distinct neutrophil subsets ical evidence. For instance, in a study that exam-
in the oral cavity: intermediately activated para- ined healthy controls, patients with gingivitis and
inflammatory neutrophils in healthy oral tissues and patients with moderate or severe periodontitis,
proinflammatory neutrophils associated with patho- CD68-positive macrophages were positively corre-
logic inflammation, as in chronic periodontitis. lated with collagen breakdown and the severity of
Para-inflammation is a stress response that displays periodontal disease (250, 295). Moreover, an inde-
some, but not all, of the characteristics of inflam- pendent study showed that the numbers of macro-
mation; in other words, it is an intermediate state phages were significantly increased in sites
between the basal (homeostatic) and inflammatory associated with disease progression (≥ 2 mm change
states (37). Proinflammatory neutrophils can be dis- in probing attachment levels) compared with con-
tinguished from para-inflammatory cells by their tralateral sites that remained stable (297). More
elevated degranulation, phagocytosis, reactive oxy- recently, CD68-positive macrophages were detected
gen species production, formation of neutrophil in abundance in deep periodontal lesions where
extracellular traps and display of a characteristic they express interleukin-23, a cytokine that supports
pattern of cell-surface activation markers (61). the proliferation of T-helper 17 cells, which are also
At present, it is uncertain whether the above-dis- abundantly present in the lesions along with
cussed neutrophil subsets are derived from separate CD20-positive B-cells (5).
lineages of neutrophils, develop from a single neu- In line with the clinical studies, P. gingivalis-
trophil precursor with considerable plasticity or rep- induced periodontitis in mice is associated with
resent different stages of neutrophil development/ recruitment of monocytes/macrophages to the gingi-
maturation. Regarding plasticity, it is possible that val tissue, whereas clodronate liposome-mediated
neutrophils adaptively change their phenotypes in depletion of monocytes/macrophages results in
the course of their response to stressors, such as tis- decreased inflammatory bone loss (142, 261). Inter-
sue injury, inflammation and infection. For instance, estingly, macrophage depletion also leads to a signifi-
even in a clinically healthy periodontium, the con- cant reduction in the numbers of P. gingivalis that
stant presence of a tooth-associated biofilm would colonize the periodontal tissue (142). The association
necessitate the presence of para-inflammatory neu- of macrophages with elevated periodontal inflamma-
trophils. Such cells could ostensibly handle the rela- tion and P. gingivalis colonization is consistent with
tively mild microbial challenge without having to findings that P. gingivalis manipulates toll-like recep-
become fully activated and risk the integrity of the tor-2 signaling in macrophages to escape clearance
host tissue.

125
Hajishengallis & Korostoff

whilst promoting a nutritionally favorable inflamma- relative to M1 cells. Thus, they have immunoregula-
tory response (82, 151, 287). In this regard, moreover, tory properties and promote cell proliferation and tis-
macrophage-specific toll-like receptor-2 signaling is sue regeneration. In fact, the M1 and M2 subsets
linked to inflammatory periodontal bone loss (215). represent extremes of a continuum of different acti-
Specifically, adoptive transfer of toll-like receptor-2- vation states (184, 190, 255). Consequently, the M1/
expressing macrophages to toll-like receptor-2-defi- M2 categorization, although conceptually useful,
cient mice enabled P. gingivalis to cause bone loss in oversimplifies and detracts from the important roles
the recipient toll-like receptor-2-deficient mice (215), that macrophages play in an ever-changing environ-
which are normally resistant to P. gingivalis-induced ment, especially in mucosal tissues, such as the
periodontitis (23). Experiments in the subcutaneous periodontium, that are constantly under microbial
chamber model (89) have suggested additional mech- challenge (107, 124, 184). It has also been argued
anisms whereby P. gingivalis may benefit from the that the M1/M2 classification has largely resulted
presence of macrophages, and explicitly from their from in vitro experiments, whereas in vivo macro-
monocytic precursors. Specifically, it was found that phage phenotypes at sites of inflammation may not
the presence of Ly6Clow/CCR2low/CX3CR1high be as clearly defined (24). For instance, a subset of
monocytes (but not of Ly6Chigh/CCR2high/CX3CR1low macrophages associated with resolution of inflam-
monocytes) was associated with reduced neutrophil- mation (‘resolving macrophages’) in models of acute
mediated phagocytosis of P. gingivalis in the inflammation, such as chemical peritonitis, share
chamber environment (261). Consistent with this properties of both M1 and M2. This macrophage
observation, the absence of the CX3CR1high subset in subset expresses the M1-associated molecules indu-
the gingiva of CX3CR1-deficient mice was associated cible nitric oxide synthase and cyclooxygenase-2,
with significantly decreased P. gingivalis-induced while at the same time expressing mannose recep-
dysbiosis and bone loss compared with the same tor (CD206), arginase-1, transforming growth factor-
parameters in wild-type mice that displayed normal beta and high levels of interleukin-10, all of which
recruitment of CX3CR1high monocytes (261). are associated with M2 macrophages (24, 259).
Moreover, the phenotype of macrophages involved
in inflammation resolution may be tailored to both
Macrophage subsets
the characteristics of the tissue microenvironment
Mature macrophages display heterogeneity and func- and the particular inflammatory and microbial
tional versatility (‘plasticity’), although the role of stimuli encountered (24, 248).
specific macrophage subsets in periodontitis is largely In the above-discussed P. gingivalis-induced
unexplored. Macrophages can alter their functional periodontitis model, M1 macrophages (defined as
activities in response to local microenvironmental CD86-positive) were shown to predominate over M2
factors, and this plasticity allows them to function macrophages (defined as CD206-positive), suggesting
appropriately in distinct conditions (255, 263). In this that M1 macrophages probably represent a periodon-
context, macrophages can undergo M1 (classical) or titis-associated subset (142). Currently, there are no
M2 (alternative) activation (138, 177, 255). M1 macro- clinical studies on the association of different macro-
phages can be induced by microbial agents (e.g. phage subsets with periodontal disease. However,
lipopolysaccharide) or type 1 cytokines (e.g. inter- evaluation of gingival gene expression profiles as indi-
feron-gamma). Relative to M2 macrophages, cells in cators of macrophage variation in nonhuman pri-
the M1 subset display an increased ability to phago- mates showed that M1 macrophage gene-
cytose microbes and enhanced expression of proin- transcription patterns increased significantly with
flammatory cytokines, costimulatory molecules (e.g. aging and with development of periodontitis, whereas
CD86) and antimicrobial molecules (e.g. reactive oxy- changes in M2 gene profiles were relatively modest
gen species and nitric oxide) but a decreased capacity (75). Moreover, a recent study utilizing another mur-
to phagocytose apoptotic cells (efferocytosis). M1 ine model of periodontitis (induced by placing liga-
macrophages are therefore important for protection tures impregnated with P. gingivalis) showed that
against microbial pathogens and are early players in periodontal inflammation is associated with
the course of infection or inflammation. On the other increased presence of both M1- and M2-like macro-
hand, M2 macrophages can be induced by diverse phage phenotypes (relative to control mice) (296).
agonists, including type 2 cytokines (interleukin-4 Nonetheless, because the ratio of inducible nitric
and interleukin-13), and secrete high levels of inter- oxide synthase-expressing (M1-like) to CD206-posi-
leukin-10 and transforming growth factor-beta1 tive (M2-like) macrophages was significantly greater

126
Revisiting the Page & Schroeder model

in the experimental periodontitis group compared osteoblasts, are responsible for bone metabolism.
with the control group, the authors suggested that a Whereas osteoblasts are involved in bone formation,
phenotypic switch to M1 might contribute to disease osteoclasts can resorb bone by demineralizing it
in this model. and degrading its organic matrix. Under homeo-
Although macrophages have been investigated in static conditions, bone resorption is followed by
the context of homeostatic immunity (induction of an osteoblast-mediated bone formation in a process
immune response with timely resolution of inflam- that involves osteoclast–osteoblast communication
mation and restoration of tissue integrity) and as and is known as ‘coupling’ (78). A persisting inflam-
effectors of resolution in various inflammatory dis- matory environment, however, can ultimately dis-
eases and relevant models (191, 259), they have as yet rupt bone homeostasis, resulting in a net loss of
received little attention in the periodontal research bone (i.e. in bone resorption exceeding bone forma-
field. The ability of the host to resolve the inflamma- tion). The fundamental mechanism governing bone
tory response (to infection or injury) in a timely man- resorption involves the regulation of osteoclastogen-
ner is critical because nonresolving inflammation esis by a triad of proteins of the tumor necrosis fac-
underlies the pathogenesis and is the common tor/tumor necrosis factor-receptor family. This
denominator of many chronic diseases, including group of proteins includes RANKL, its functional
periodontitis (251). The successful resolution of receptor RANK and its soluble decoy receptor osteo-
inflammation involves a distinct series of processes, protegerin (194). While osteoclast differentiation
including down-regulation of proinflammatory medi- and activation are promoted by the binding of
ators, up-regulation of anti-inflammatory and RANKL (expressed by osteoblasts and activated T-
pro-resolving mediators, termination of neutrophil cells and B-cells) to RANK on osteoclast precursors,
recruitment, clearance of apoptotic neutrophils by osteoprotegerin acts to block the RANKL–RANK
tissue phagocytes (efferocytosis) and tissue repair interaction, thereby restraining osteoclastogenesis.
(267). Macrophages play a central role in the resolu- A unique feature of the periodontium compared
tion of inflammation. In this regard, efficient efferocy- with other mucosal sites, such as the gastrointesti-
tosis of apoptotic cells by macrophages not only nal and respiratory tracts, is that it consists of both
prevents secondary necrosis and inflammation but mucosal and bone tissue. Thus, it is clear that the
also reprograms the transcriptional profile of the effe- cellular and molecular crosstalk between the peri-
rocytic macrophage (termed resolving macrophage). odontal immune and bone systems plays a critical
Specifically, upon efferocytosis, macrophages are role in the regulation of bone homeostasis. Proin-
reprogrammed to reduce the expression of proin- flammatory cytokines that can induce RANKL
flammatory cytokines and increase the expression of expression in osteoblasts, such as interleukin-1beta,
regulatory cytokines, including transforming growth tumor necrosis factor and interleukin-17, are abun-
factor-beta and interleukin-10 (210, 227, 266). dant in the inflamed periodontium. Therefore,
Macrophage plasticity is therefore crucial for success- osteoblasts can contribute to an increase of the
ful resolution of inflammation and merits intensive RANKL/osteoprotegerin ratio. Interestingly, RANKL
investigation in both human periodontitis and animal and osteoprotegerin were shown to be reciprocally
models of the disease. Further research may reveal regulated by several stimuli. For instance, inter-
novel protective roles by resolving or other leukin-17 induces RANKL and inhibits osteoprote-
as-yet-uncharacterized subsets of macrophages in gerin expression in human periodontal ligament
periodontitis. cells (153), which are also thought to be important
in the regulation of osteoclastogenesis (119). How-
ever, adaptive immune cells appear to be the major
Osteoimmunology: crosstalk source of RANKL in human periodontitis. In this
between the immune and bone regard, approximately 50% of T-cells and 90% of B-
systems cells in diseased gingival tissues of patients with
periodontitis express RANKL (122).
Osteoimmunology is a relatively new field of study RANKL is produced in two different versions: a
that focuses on the molecular understanding of the membrane-bound form; and a soluble form (195).
crosstalk between the immune and the bone sys- Osteoblasts and bone marrow stromal cells express
tems (194). This field has provided important predominantly membrane-bound RANKL, which
insights into how inflammatory events regulate induces osteoclastogenesis through cell contact
osteoclasts (Fig. 1), the cells that, along with with osteoclast precursors. On the other hand,

127
Hajishengallis & Korostoff

Tooth Periodontal
Antibody
pocket Potential antimicrobial humoral immunity
(in pockets and connective tissue) Membrane
RANKL
• Inhibition of colonization Soluble
Biofilm • Activation of complement RANKL
• Opsonization of bacteria
• Neutralization of toxins and enzymes RANK

Junctional Local
epithelium Plasma cell activation
Translocated bacteria
or products (e.g. LPS) Naïve mature
IL-4, IL-6, B cell
APRIL, BLyS T helper cell
Extracellular matrix degradation
Antigen
Activated B cell Neutrophil presentation
IL-1, IL-6, TNF Lysosomal
enzymes

Plasma Effector Effector


MMPs cell B cells T cells

Plasma cell Immune Complement CXCL13 and


complexes other chemokines
Osteoclastogenesis
Osteoblast Osteoclast
IL-1
precursors
IL-6
Activated TNF Blood vessel
osteoclast
Bone
Plasma cell

Lymphocytes activated
Activated B cell in local lymph nodes
Fig. 4. Protective and destructive functions of B-lineage activation of complement and innate immune cells can
cells within the periodontium. The junctional epithelium enhance gingival inflammation and contribute to tissue
is situated in close proximity to the bacterial biofilm that breakdown. Moreover, recent evidence has demonstrated
develops on teeth. Quantitative and compositional the potential for B-lineage cells to express proinflamma-
changes of the microbiota (dysbiosis) induce innate tory cytokines, matrix metalloproteinases and RANKL. B-
immune signaling pathways that lead to the development lineage cells therefore directly and indirectly participate in
of an adaptive immune response within the gingival con- the degradation of the soft- and hard-tissue components
nective tissue. Regarding the humoral component of the of the periodontium, as indicated. The details are dis-
response, pathogen-specific antibody that diffuses into the cussed in the text. APRIL, a proliferation-inducing ligand;
gingival sulcus (or pocket) or remains in the connective BLyS, B-lymphocyte stimulator; CXCL13, chemokine (C-X-
tissue can, in principle, inhibit the bacterial challenge via a C motif) ligand 13; IL, interleukin; LPS, lipopolysaccharide;
number of potential mechanisms (indicated). However, MMPs, matrix metalloproteinases; RANK, receptor activa-
the antibody response has not been associated with pro- tor of nuclear factor-kappaB; RANKL, receptor activator of
tection in clinical studies. In fact, antibody-mediated nuclear factor-kappaB ligand; TNF, tumor necrosis factor.

activated T-cells and B-cells produce both the T-lymphocytes


membrane-bound and soluble forms of RANKL
(Fig. 4). Soluble RANKL can induce osteoclastogen-
esis independently of direct contact between infil- T-cells and homeostatic immunity
trating lymphocytes and osteoclast precursors on To maintain periodontal tissue homeostasis, the
the bone surface. Nevertheless, RANKL-expressing immune system proactively patrols the periodon-
T-helper 17 cells, but not T-helper 1 cells, were tium, which is in constant contact with the tooth-
shown to activate osteoclasts also by direct cell–cell associated microbiota (54). Healthy human gingiva
contact (125). contains a predominantly T-cell-rich inflammatory

128
Revisiting the Page & Schroeder model

infiltrate and a network of antigen-presenting cells major histocompatibility complex class I-restricted
(dendritic cells and macrophages) that orchestrate CD8-positive T-cells or normal controls, thus impli-
local immunity. In contrast, minimal numbers of B- cating CD4-positive T-cells in periodontal tissue
cells and plasma cells are present. Neutrophils are destruction (13).
also found in healthy gingiva (although at lower levels In part, because of our enhanced appreciation of
than in gingivitis and periodontitis) and can poten- the innate-adaptive immune-cell crosstalk (Fig. 1),
tially interact with adaptive immune cells. A small we currently know more about CD4-positive T-cells
population of innate lymphoid cells can also be seen and their functionally distinct subsets and how they
at the gingival mucosal barrier (54). These cells are instructed by antigen-presenting mononuclear
belong to the lymphoid lineage and play important cells to elicit their immune responses (203, 256).
roles in protective immunity and maintenance of tis- Before the discovery of toll-like receptors, innate
sue homeostasis but do not respond in an antigen- immunity was viewed as a nonspecific, transient and
specific manner (106). The majority of T-cells in expedient reaction to ‘buy time’ until the activation
healthy gingiva are CD4-positive T-helper cells, but of adaptive immunity, the system of B- and T-lym-
also present are cytotoxic CD8-positive T-cells and a phocytes that express antigen receptors of exquisite
small population of gamma delta T-cells, unconven- specificity. We now recognize that toll-like and other
tional CD3-positive T-cells involved in local immune pattern-recognition receptors endow innate immu-
surveillance and tissue homeostasis (54). Their poten- nity with adequate specificity to distinguish between
tially protective role notwithstanding, T-cells are also different types of pathogens. Most importantly, it has
implicated in the pathogenesis of periodontal become apparent that innate immune mechanisms
disease. are sophisticated enough to make ‘judgments’ that
instruct the initiation and progression of the adaptive
immune response (174). In contrast to pattern-
T-cells and antigen-presenting cells in
recognition receptors, the exquisite specificity of the
periodontitis
adaptive immune receptors is not the result of
In the Page & Schroeder model, a dense infiltrate of co-evolution with microbes but rather the outcome
lymphocytes and other mononuclear cells is associ- of randomly generated gene recombination. Thus,
ated with gingivitis (‘early lesion’) but not necessarily although the overall B- and T-cell-receptor reper-
with periodontitis. Indeed, this stage, as well as the toires collectively can bind virtually any antigenic
plasma-cell-rich ‘established lesion’, could remain structure (epitope), the cells are essentially ignorant
stable without inevitably progressing to bone loss, the of the biologic context of their antigen specificity. In
hallmark of the ‘advanced lesion’ (213). This key other words, they know what antigen they can
observation is consistent with the concept that peri- respond to but do not have a clue about whether and
odontitis requires a susceptible host, although we do how to respond. This information is predominantly
not fully understand the mechanisms that tip the fine provided by dendritic cells, which mediate between
host–microbe balance from a controlled immunoin- detection of infection and induction of the adaptive
flammatory state to dysbiotic inflammation sufficient response. For instance, in the oral mucosa, dendritic
to cause irreversible damage (bone loss) to the peri- cells encounter and capture oral microbes, then
odontium (143). The association of T-cells expressing migrate to the regional lymph nodes where they pro-
the alpha beta T-cell antigen receptor with periodon- vide costimulatory signals and present processed
titis probably represents a cause-and-effect relation- antigen to CD4-positive T-cells, thereby regulating
ship, as suggested by animal model experiments. their activation and differentiation into different sub-
Studies in the 1990s, using the murine oral gavage sets (43, 291) (Fig. 5). Given the importance of T-cells
model of P. gingivalis-induced periodontitis, indi- in periodontitis, it becomes apparent that dendritic
cated that the periodontal adaptive immune response cells can influence the outcome of this oral disease in
is potentially destructive in periodontitis. Specifically, either protective or destructive directions, and both
severe combined immunodeficient mice, which lack outcomes have been described in animal models (6,
both T- and B-cells, develop less P. gingivalis- 58, 292). Langerhans cells are a specialized subset of
induced bone loss than do immunocompetent dendritic cells that are phenotypically hallmarked by
controls (14). In the same model, major histocompat- their expression of the C-type lectin receptor langerin
ibility complex class II-restricted CD4-positive (CD207). They populate skin epidermis as well as oral
T-cell-deficient mice exhibit reduced P. gingivalis- and mucosal epithelia, survey their environment for
induced bone loss as compared with mice deficient in foreign antigens and can regulate T-cell responses

129
Hajishengallis & Korostoff

Inductive Key transcription Signature Functions Targets Consequences Possible role


cytokines factors cytokines (overactivation) in periodontitis

T-bet IFNγ Cell-mediated Intracellular Delayed-type Uncertain; both


IL-12 Th1 adaptive pathogens hypersensitivity protective and
immunity (viruses, Autoimmunity destructive
bacteria) effects
B
described

Gata3 IL-4 Humoral Extracellular Allergy Uncertain; both


IL-4 IL-5 adaptive pathogens protective and
Naïve Th2
IL-13 immunity (parasites, destructive
DC bacteria) effects
CD4
described
Ag
presentation
Rorγt IL-17A Neutrophilic Extracellular Inflammation May contribute
IL-21
Th17 IL-17F inflammation pathogens Autoimmunity to inflammatory
IL-6, TGFβ IL-21 and innate (bacteria, bone loss
IL-1, IL-23 IL-22 immunity fungi)

Foxp3 IL-10 Immune Effector Immune May contribute


TGFβ
Treg TGFβ regulation T cells suppression to protection
from
inflammatory
bone loss

Fig. 5. Developmental pathways of CD4-positive T-cells, cell; DC, dendritic cell; Foxp3, forkhead box P3; Gata3,
their functions and their associations with particular dis- GATA sequence-binding protein-3; IFNc, interferon-
eases. Na€ıve CD4-positive T-helper cells differentiate into gamma; IL, interleukin; IL-17A, Interleukin-17 isoform A;
distinct lineages depending on the indicated cytokine IL-17F, Interleukin 17 isoform F; Rorct, retinoic acid-
milieu during activation by antigen-presenting cells. As related orphan receptor gammat; T-bet, T-box expressed
shown, each of these effector or regulatory T-cell subsets in T-cells; TGFb, transforming growth factor-beta; Th1,
has a characteristic cytokine-expression pattern and hence T-helper type 1 cell; Th2, T-helper type 2 cell; Th17, T-
distinct functions and roles in autoimmune or inflamma- helper type 17 cell; TNF, tumor necrosis factor; Treg,
tory diseases, including periodontitis. Ag, antigen; B, B- regulatory T-cell.

(120, 291). Consistent with an immunoregulatory role, gamma and are responsible for cell-mediated immu-
in vivo ablation of langerin-expressing cells in a nity to intracellular pathogens but are also implicated
mouse model of P. gingivalis-induced periodontitis in inflammatory and delayed-type hypersensitivity
resulted in increased infiltration of B- and T-cells and reactions. T-helper 2 cells secrete interleukin-4, inter-
enhanced bone loss (7). However, langerin is also leukin-5 and interleukin-13, mediate humoral immu-
expressed by a population of dendritic cells known as nity (including IgE) and mast cell activation, and are
langerin+ dendritic cells, which are present in the also involved in allergic reactions. T-helper 17 cells
mouse gingiva, according to an independent study secrete interleukin-17A, interleukin-17F, interleukin-
(20). In fact, that study showed that specific ablation 21 and interleukin-22 and mediate responses that
of Langerhans cells (thus retaining langerin+ den- recruit neutrophils and activate immunity to extracel-
dritic cells) had no effect on P. gingivalis-induced lular bacterial and fungal infections. Moreover, these
bone loss (20). Therefore, it is plausible that the cells have been implicated in autoimmune, inflam-
observed immunoregulatory effects (7) are attributa- matory and bone-resorptive disorders. CD4-positive,
ble to langerin+ dendritic cells or require the pres- forkhead box P3-expressing regulatory T-cells secrete
ence of both Langerhans cells and langerin+ dendritic interleukin-10 and transforming growth factor-beta
cells. and play important regulatory roles in the mainte-
nance of self-tolerance and suppression of unwar-
ranted inflammation by down-regulating T-helper 1,
T-cell subsets
T-helper 2 and T-helper 17 cell effector functions
Upon activation by antigen-presenting cells, such as (136, 288). The differentiation of each of these effector
dendritic cells, naive CD4-positive T-cells can be T-cell subsets is driven by distinct cytokines and is
polarized into distinct effector T-helper cell subsets – controlled by different sets of transcription factors
T-helper 1, T-helper 2, T-helper 17 and regulatory T (Fig. 5). The differentiation of T-helper 1 and T-helper
cells – depending on the local cytokine milieu. Each 2 subsets is driven by interleukin-12 and interleukin-
of these T-cell subsets has a characteristic cytokine 4, respectively. Whereas interleukin-12 can be derived
expression pattern and hence distinct functions (136, from innate immune sources, such as an antigen-pre-
288) (Fig. 5). T-helper 1 cells secrete interferon- senting dendritic cells, interleukin-4 can be provided

130
Revisiting the Page & Schroeder model

by B-cells or by the naive CD4-positive T-cells them- responses (66, 68, 70, 256, 273) (Fig. 4). Under the
selves. The key transcription factors controlling the T-helper 1/T-helper 2 paradigm, an interesting model
differentiation of T-helper 1 and T-helper 2 cells are was proposed according to which T-helper 1 cells
T-box expressed in T-cells and GATA sequence-bind- predominate in stable early lesions (i.e. when there is
ing protein-3, respectively. Transforming growth fac- a homeostatic balance between the host and the peri-
tor-beta, interleukin-6, interleukin-1, and interleukin- odontal microbiota), whereas T-helper 2 cells are
21 are involved in the differentiation of T-helper 17 associated with disease progression beyond the
cells, whereas interleukin-23 is important for T-helper established lesion, featuring an inflammatory infil-
17 cell expansion and survival. All of these cytokines trate that is particularly rich in B-cells and antibody-
can be derived from innate immune-cell sources, secreting plasma cells (70). However, as alluded to
except for interleukin-21, which feeds back on the above, this and other disease models under the
developing T-helper 17 cells and amplifies the differ- T-helper 1/T-helper 2 paradigm are consistent with
entiation process. The retinoic acid-related orphan only a subset of the clinical and experimental data.
receptor-gammat is the key transcription factor driv-
ing the differentiation of T-helper 17 cells. Regulatory
T-helper 17 cells
T-cells share a reciprocal developmental pathway
with T-helper 17 cells because transforming growth The subsequent discovery of the T-helper 17 and reg-
factor-beta is also required for the differentiation of ulatory T-cell subsets has re-invigorated interest in
naive T-cells into regulatory T-cells, whereas forkhead the role of T-cell subsets in inflammatory diseases,
box P3 controls their development and function (136, including periodontitis. The discovery of T-helper 17
288). cells is particularly relevant in the context of osteoim-
munology, a field of study developed well after the
Page & Schroeder model was proposed. We now have
The T-helper 1/T-helper 2 paradigm
a relatively good understanding of how inflammatory
The T-helper 1/T-helper 2 paradigm, which was events regulate osteoclasts through the osteoprote-
established in the late 1980s, provided a useful con- gerin–RANKL–RANK axis (194) (see above for details).
ceptual framework to help understand T-cell involve- The bone-protective effect of osteoprotegerin is, how-
ment in immunity and inflammation, although ever, diminished in periodontal disease, as the osteo-
diseases of immunologic etiology were occasionally protegerin/RANKL ratio decreases with increasing
pigeonholed into T-helper 1 or T-helper 2 categories periodontal inflammation often dominated by inter-
without adequate supportive evidence (66). Similarly, leukin-17-expressing T-helper 17 cells (5, 16, 27, 34,
periodontitis cannot be readily described in simple T- 207).
helper 1 vs. T-helper 2 dichotomous terms despite T-helper 17 cells constitute a significant source of
over two decades of intensive investigation. Studies in interleukin-17 production in the periodontal tissue
humans and animal models have described both pro- (54, 187). Interleukin-17 potentiates innate immunity,
tective and destructive effects associated with either especially neutrophil-mediated host defenses (123),
subset (66, 68, 71, 256, 273). In some reports, by promoting granulopoiesis and orchestrating neu-
T-helper-1-type cytokines (interferon-gamma and trophil recruitment via up-regulation of granulocyte-
interleukin-12) were negatively correlated with peri- colony stimulating factor and chemokines (284, 294)
odontal disease severity, ostensibly attributed to the and down-regulation of developmental endothelial
ability of these cytokines to promote cell-mediated locus-1, an endogenous inhibitor of the leukocyte
immunity and inhibit osteoclastogenesis. Other adhesion cascade (59, 161) (Fig. 3). However, inter-
investigations, however, attributed destructive effects leukin-17 also has potent osteoclastogenic properties,
to interferon-gamma and T-helper 1 cells in peri- in part because of its capacity to stimulate the expres-
odontitis, consistent with the ability of activated sion of RANKL in osteoblasts and other stromal cells
T-helper 1 cells to express the osteoclastogenic factor (139). Moreover, interleukin-17 can mediate destruc-
RANKL. Certain studies showed that the presence of tion of connective tissue by inducing the expression
T-helper 2 cells was associated with protective effects of matrix metalloproteinases in fibroblasts (178)
attributed to the ability of T-helper 2 cells to secrete (Fig. 3). In addition to secreting interleukin-17,
interleukin-4 and interleukin-13, which can restrain T-helper 17 cells express RANKL and function as a
osteoclastogenesis. Contrasting results linked dedicated osteoclastogenic subset that links T-cell
T-helper 2 cells to the pathogenesis of periodontitis, activation to osteoclast activation and inflammatory
owing to their capacity to support destructive B-cell bone destruction (Fig. 1). Although this concept is

131
Hajishengallis & Korostoff

derived mostly from studies in rheumatoid arthritis conditions (60, 102, 180). In this regard, T-helper 17
(178), multiple investigations have shown increased cells were shown to act as B-cell helpers by inducing
levels of locally produced interleukin-17 in human a robust proliferative response of B-cells in vitro and
periodontal tissue affected with periodontitis as com- by triggering antibody production with IgG class
pared with healthy periodontal tissue (5, 15, 27, 53, switch in vivo (180). The same study showed that
55, 108, 114, 148, 186, 207, 208, 252, 268, 282, 301). inhibition of interleukin-17 signaling leads to a signif-
Although these correlative studies in humans do not icant decrease in both the number and size of B-cell
prove a cause-and-effect relationship, antibody- germinal centers (180). In a similar context, T-helper
mediated neutralization of interleukin-17 in different 17-derived interleukin-21 synergizes with B-lympho-
murine periodontitis models (in which the disease is cyte stimulator in stimulating plasma cell differentia-
induced spontaneously or upon placement of liga- tion (60) (Fig. 1). From a pathologic viewpoint, T-
tures) was shown to block periodontal inflammation helper 17 cells can provide B-cell help in autoanti-
and bone loss (59, 187). It should be noted that inter- body-induced arthritis, whereas neutralization of
leukin-17 is also produced by a variety of adaptive interleukin-17 delays the onset of disease (102). The
and immune cell types, including CD8-positive T- possibility of similar mechanisms occurring in the
cells, gamma delta T-cells, natural killer T-cells and setting of periodontitis has not been investigated,
innate lymphoid cells (42, 167, 265, 270). Therefore, although whether periodontitis includes an autoim-
the protective effects of anti-interleukin-17 interven- mune component remains uncertain (discussed later
tions do not necessarily target T-helper 17 cells in greater detail in the section ‘Abundance of B-lym-
exclusively. phocytes and plasma cells in periodontitis’).
Interestingly, P. gingivalis-induced periodontitis in
Langerhans cell-deficient mice suppresses the num-
Regulatory T-cells
bers of P. gingivalis-specific interleukin-17A-produ-
cing CD4+ T-cells but elevates the numbers of Regulatory T-cells have been identified in chronic
P. gingivalis-specific interferon-gamma-producing inflammatory periodontal lesions (28, 193). These reg-
CD4+ T-cells (compared with wild-type mice), ulatory cells express forkhead box P3, a transcription
although the induced bone loss is comparable in factor that induces expression of genes required for
Langerhans cell-deficient and wild-type mice (20). their immune-suppressive functions. Regulatory T-
This suggests that both T-helper 17 (interleukin-17) cells are classified into natural regulatory T-cells and
and T-helper 1 (interferon-gamma) responses can inducible regulatory T-cells based on the site of matu-
mediate destructive periodontal inflammation, ration. The natural regulatory T-cells are generated
although another possibility is that interleukin-17 was in, and released from, the thymus to the periphery as
produced by other cellular sources in Langerhans a functionally distinct lineage that already expresses
cell-deficient mice, such as gamma delta T-cells, forkhead box P3. The inducible regulatory T-cells
which are abundant in the mouse periodontium and originate from na€ıve T-cells and are induced in the
other mucosal sites (59, 230). periphery by antigen presentation from tolerogenic
At least in principle, the ability of T-helper 17 cells dendritic cells in the presence of transforming growth
to secrete interleukin-17 and interleukin-22 could factor-beta and the absence of interleukin-6 and/or
exert a protective effect in periodontitis. As men- interleukin-21. Interleukin-2 and transforming
tioned earlier, interleukin-17 can stimulate granulo- growth factor-beta are required for induction of fork-
cyte-colony stimulating factor-dependent head box P3 in inducible regulatory T-cells (200). As
granulopoiesis and chemotactic recruitment and acti- the differentiation of T-helper 17 cells is induced by
vation of neutrophils (109, 284) (Fig. 3) and, more- transforming growth factor-beta in combination with
over, both interleukin-17 and interleukin-22 can interleukin-6 and/or other cytokines, such as inter-
induce epithelial cell production of antimicrobial leukin-21, the development of regulatory T-cells and
peptides (136). However, this protective scenario T-helper 17 is reciprocally regulated. In fact, inter-
seems more plausible in the case of acute infections. leukin-6 and interleukin-21 inhibit the expression of
The persistence of T-helper 17 cells at sites of inflam- forkhead box P3 (by activating signal transducer and
mation provides prolonged interleukin-17 signaling, activator of transcription-3) and promote (transform-
which can turn an acute inflammatory response into ing growth factor-beta-induced) retinoic acid-related
chronic immunopathology (156). Moreover, T-helper orphan receptor-gammat expression to induce T-
17 cells have been implicated in promoting B-cell helper 17 cells. In other words, the cytokines control-
responses in autoimmune and inflammatory ling the forkhead box P3/retinoic acid-related orphan

132
Revisiting the Page & Schroeder model

receptor-gammat balance give rise to regulatory T- exacerbates inflammation and bone loss (68). Con-
cells or T-helper 17 cells, respectively. In a related versely, recruitment of regulatory T-cells upon local
context, interleukin-23-activated gamma delta T-cells treatment of mice or dogs with a regulatory T-cell-
were shown to restrain regulatory T-cells and tip the recruiting chemokine [chemokine (C-C motif) ligand
balance in favor of effector T-helper cells (221). More- 22] formulation blocks experimental periodontitis in
over, interleukin-23 can induce expansion and stabi- these models (73). Therefore, therapeutic approaches
lization of the T-helper 17 subset and stimulate their that promote the presence of regulatory T-cells have
production of interleukin-17 (288). It is therefore the potential to mitigate inflammatory tissue damage
plausible that factors which regulate the balance in periodontitis.
between T-helper 17 and regulatory T-cells can, in Our knowledge on the T-cell response in periodon-
principle, influence the pathogenesis of periodontitis. titis has come a long way since the Page & Schroeder
In both human periodontitis and animal models of model was proposed but the roles of T-cells are still
the disease, the number of forkhead box P3-positive enigmatic. Although the discovery of the T-helper 17
regulatory T-cells increases in the inflamed periodon- and regulatory T-cell subsets has offered enhanced
tium in comparison with healthy tissue (28, 53, 133, insights into the pathogenesis of periodontitis, the
193). These findings imply a homeostatic mechanism dissection of the protective and destructive aspects of
aiming to mitigate collateral tissue damage caused by the periodontal T-cell response remains a complex
the overactivation of adaptive immunity. Mechanisti- and challenging endeavor. This may be related to the
cally, immunosuppressive cytokines, such as inter- very nature of periodontal disease, a polymicrobial
leukin-10, produced by regulatory T-cells, can inhibit community-induced dysbiotic malady (88), in which
the induction of RANKL expression by activated T- a potentially protective antimicrobial response could
cells (Fig. 1). Additionally, interleukin-10 may sup- be offset by bystander tissue damage. In summary,
press RANKL-mediated differentiation and activation while T-helper 17 cells and interleukin-17 have the
of osteoclasts (183, 217) (Fig. 1). It should be noted, capacity to mediate protective responses, the current
however, that regulatory T-cells have a substantial burden of evidence from human and animal model
degree of plasticity and may lose their suppressive studies suggests that their net effect promotes disease
function in an inflammatory environment, implying development, whereas the opposite effect (protec-
that an increase in the number of regulatory T-cells tion) appears to be facilitated by regulatory T-cells (5,
does not necessarily mean that these cells will even- 28, 53, 59, 66, 68, 73, 109, 152, 178, 193, 284).
tually control development of inflammatory disease
(200). In this regard, forkhead box P3 was shown to
be overexpressed in active periodontal lesions (along B-Lineage cells
with RANKL, interleukin-17 and other inflammatory
cytokines) compared with inactive lesions (53). Fur-
Abundance of B-lymphocytes and plasma
thermore, it has been suggested that regulatory T-
cells in periodontitis
cells may convert into interleukin-17-producing T-
helper 17 cells in human periodontitis lesions (208). In their classic paper, Page & Schroeder described the
In this study, interleukin-17/forkhead box P3 double- initial appearance of B-cells and plasma cells as a
positive cells were detected in periodontitis lesions, minor component of the lymphoid infiltrate detected
intimating an intermediate stage in the process of within the early lesion (213). Upon progression of the
regulatory T-cell-to-T-helper 17 conversion (208). In disease, however, a dramatic increase in the numbers
contrast, another investigation failed to detect inter- of these cell types was observed such that the ‘distin-
leukin-17/forkhead box P3 double-positive cells in guishing feature of the established lesion is the pre-
the inflammatory infiltrate of human periodontal dominance of B-lymphocytes and of plasma cells
lesions (216). within the affected connective tissue at a stage prior
Studies in animal models of periodontitis have pro- to extensive bone loss’ (213). They reported a further
vided important insights into the possible function of increase in the proportion of B-lineage cells within
regulatory T-cells in this oral disease. In murine the infiltrate, yielding the advanced lesion that was
models of pathogen-induced periodontitis, regulatory associated with overt bone loss. From a histopatho-
T-cells appear in high numbers after the peak emer- logic perspective, plasma cells represented the pre-
gence of RANKL-expressing CD4-positive T-cells dominant cell type within periodontitis lesions. As the
(133), whereas their depletion (by anti-glucocorti- lesions were described on the basis of morphometric
coid-inducible tumor necrosis factor receptor) and ultrastructural observations, it is critical to

133
Hajishengallis & Korostoff

appreciate the limitations of these approaches rela- in their majority. Moreover, conclusive mechanistic
tive to distinguishing B-cells and T-cells from one studies implicating an autoimmune component in
another. periodontitis are lacking and the role of autoantibod-
Soon thereafter, immunohistochemical and ies in the pathogenesis of periodontitis remains
immunofluorescence approaches were utilized by a ambiguous but cannot be ruled out at present.
number of other groups to define more clearly the Another investigation utilized an immunohisto-
composition of the inflammatory infiltrate in gingivi- chemical approach to evaluate the lymphocyte com-
tis and periodontitis lesions. With advancing knowl- position of inflamed human connective tissue within
edge regarding the expression of unique structures on long-standing gingivitis and periodontitis lesions
the surfaces of T, B and plasma cells, specific reagents (274). B-lineage cells (CD20-expressing B-cells and
were developed that allowed investigators not only to CD138-expressing plasma cells) were found in higher
distinguish these cell types more accurately from one numbers in periodontitis lesions relative to gingivitis
another but also to identify the distribution of lym- lesions and in both situations exceeded the number
phocyte subsets within diseased gingival tissue. A of CD3-positive T-cells. Moreover, an independent
majority of these studies has confirmed the observa- study showed that the numbers of plasma cells were
tions of Page & Schroeder, demonstrating the pre- significantly elevated in sites associated with peri-
dominance of B-lineage cells in clinical scenarios odontal disease progression (≥ 2 mm change in prob-
analogous to the established and advanced lesions ing attachment levels) in comparison with
(17, 18, 113, 127, 181, 274, 297). B-lineage cells are contralateral sites in which the number of plasma
estimated to comprise around 60% of the total leuko- cells remained stable (297). Recently, another group
cytes that are present in periodontal lesions associ- sought to describe the make-up of the immune-cell
ated with bone loss (274). We will therefore limit our populations present in clinically healthy human gin-
comments to studies communicating unique giva (54). As the focus of the study was homeostatic
findings. immunity, they evaluated tissue obtained from 50
Utilizing CD19 as a marker for B-lineage cells, it was healthy subjects and only six patients with chronic
shown that B-cells and plasma cells collectively repre- periodontitis, for comparative reasons. Utilizing flow
sented the most common lymphoid cells within the cytometric analysis of collagenase-digested tissue,
inflammatory infiltrate of tissue from patients with they found significant numbers of T-cells but very few
severe chronic periodontitis (50). Interestingly, more- B-cells in healthy samples, an observation consistent
over B-cells outnumbered both plasma cells and T- with Page & Schroeder’s findings regarding the lym-
cells (50). Furthermore, a majority of the B-cells were phocyte population within the initial lesion. Although
also found to express CD5. This observation was taken the numbers of CD19-positive B-cells were increased
to suggest that these cells were analogous to the previ- in periodontitis relative to health, their analysis did
ously described CD5-positive murine B-1a subset of not include plasma cells. Therefore, the overall abun-
B-cells. The B-1a subset has been shown to exhibit sig- dance of B-lineage cells was not estimated and conse-
nificant autoimmune activity in a number of murine quently no comparison could be made with the
models. Thus, the presence of large numbers of CD5- abundance of T-cells.
positive cells in the periodontitis lesions was inter- As discussed in an earlier section of this review, sig-
preted to indicate that autoantibody production has a nificant effort has gone into determining the contri-
role in the pathogenesis of periodontitis (18). Unlike bution of distinct T-cell subsets to the pathogenesis
‘conventional’ B-cells (B-2) that have a hematopoietic of the plaque-induced periodontal diseases. In con-
origin, B-1a cells develop from peritoneal precursors trast, very little is known regarding the distribution of
during the fetal and neonatal phases of life. They typi- B-lineage cells belonging to well-defined subsets
cally respond to common pathogen-associated carbo- within healthy and diseased periodontal tissues. Even
hydrates and tend to produce broadly reactive, but less understood is the role of the cells belonging to
low-affinity, IgM thought to be involved in initial distinct B-cell subsets in the maintenance of gingival
responses to certain bacteria and viruses. It has tissue homeostasis, disease initiation or disease pro-
recently been shown that only a small percentage of gression. In order to shed light on these issues, a very
CD5-expressing human B-cells exhibit the functional recent study evaluated the phenotypes of B-lineage
phenotype of B-1a cells (CD5 is expressed on multiple cells detected in samples of healthy, diseased (gingivi-
human B-cell populations) (234). Therefore, it is tis and periodontitis) and treated (resolved-periodon-
uncertain whether the CD5-expressing cells detected titis) human gingiva (164). Utilizing flow cytometry,
in periodontitis lesions are truly B-1a cells, at least not na€ıve B-cells, memory B-cells, plasmablasts and

134
Revisiting the Page & Schroeder model

plasma cells were identified based upon the differen- notably bacterial polysaccharides, can cross-link mul-
tial expression of a panel of relevant cell-surface tiple antigen receptors and stimulate B-cells in the
molecules (CD19, CD27, CD38, CD138, HLA-DR, IgG absence of T-cells. Additionally, innate signals (such
and IgA). As one would predict, the percentage of B- as lipopolysaccharide and other conserved microbial
lineage cells was highest in cell suspensions prepared structures) may enhance antigen-mediated activation
from tissue affected by periodontitis. Very few na€ıve by binding to toll-like receptors (toll-like receptor-4,
B-cells were detected in any of the samples, while toll-like receptor-7 and toll-like receptor-9) expressed
plasma cells, as expected, were the predominant B- by human B-cells (104). It is well established that the
lineage cell in periodontitis samples. The majority of inflammatory infiltrate within gingivitis and peri-
B-cells observed in healthy, gingivitis and treated odontitis lesions represents the host response to the
samples exhibited the memory cell phenotype. When polymicrobial challenge provided by the plaque bio-
intact healthy tissue was examined using immunohis- film. These organisms express well-defined T-depen-
tochemistry, the memory B-cells were localized adja- dent and T-independent antigens that are likely to
cent to the junctional epithelium, leading the induce local activation and differentiation of B-cells
investigators to speculate that the cells are involved into plasma cells within gingival connective tissue.
in maintaining gingival homeostasis. The mechanism Additionally, bacterial antigens can be transported
(s) through which this occurs was not delineated. via antigen-presenting cells to local lymph nodes,
leading to the generation of activated B-cells and
plasma cells that selectively home to the site of bacte-
Potential mechanisms of B-cell and
rial challenge.
plasma-cell involvement in periodontitis
Chemokine (C-X-C motif) ligand 13 (also known as
As discussed above, an overwhelming amount of liter- B-lymphocyte chemoattractant) is a chemokine
ature supports the original observations of Page & expressed by several cell types (follicular dendritic
Schroeder regarding the predominance of B-lineage cells, endothelial cells, fibroblasts and macrophages)
cells within the inflammatory infiltrate of periodonti- that binds chemokine (C-X-C) receptor 5 on the sur-
tis lesions (17). Over the ensuing four decades, face of mature B-cells and serves to recruit, in a selec-
researchers have attempted to address a number of tive manner, these cells to sites such as the follicular
questions incited by the observations of Page & component of lymph nodes (57) To examine whether
Schroeder. Amongst others, these included: chemokine (C-X-C motif) ligand 13 can serve a similar
 What are the mechanisms underlying the predom- purpose in the periodontium (i.e. to help recruit B-
inance of B-lineage cells in periodontitis lesions? cells), the expression of this chemokine in gingival
 What is the function of the B-lineage cells in peri- connective tissue from subjects with gingivitis or peri-
odontal pathogenesis? odontitis was evaluated (192). Significantly increased
 Are B-lineage cells requisite for alveolar bone loss numbers of chemokine (C-X-C motif) ligand 13-
in periodontitis? expressing cells were detected in periodontitis com-
As these questions are actually intertwined, we will pared with gingivitis. These cells were localized to the
attempt to review the current knowledge in a system- connective tissues underlying the pocket epithelium
atic manner. and, importantly, the numbers of chemokine (C-X-C
Our understanding of the cellular and molecular motif) ligand 13-expressing cells were positively cor-
mechanisms underlying the recruitment, activation related with the number of CD19-positive cells. It was
and differentiation of B-lineage cells in peripheral concluded that enhanced expression of chemokine
lymphoid and nonlymphoid tissues has increased (C-X-C motif) ligand 13 was in part responsible for
dramatically since the description of the four lesions the recruitment to, and distribution of, B-cells within
by Page & Schroeder. Typically, mature B-cells that the diseased tissue (Fig. 4).
exit the bone marrow are activated and induced to A large body of literature currently exists on the role
undergo differentiation into antibody-secreting of cytokines in the pathogenesis of periodontal dis-
plasma cells or memory cells in an antigen-depen- ease. Specifically related to B-lineage cells, increased
dent manner within secondary lymphoid tissues. For expression/production of multiple molecules relevant
most microbial antigens (T-dependent) this requires to their activation, proliferation, differentiation, func-
engagement of the B-cell antigen receptor (cell-sur- tion and survival has been detected in diseased gingi-
face-bound immunoglobulin) and secondary signals val tissue. Classically, most notable are interleukin-4,
derived from T-cells. A small number of antigens (T- interleukin-5 and interleukin-6, which are involved in
independent) with highly repetitive structures, most B-cell differentiation into antibody-secreting plasma

135
Hajishengallis & Korostoff

cells (170) and, furthermore, are up-regulated in peri- innate immune cells. In this regard, advanced peri-
odontitis (65). More recently, two cytokines belonging odontitis is associated with increased deposition of
to the tumor necrosis factor superfamily, APRIL (also IgG immune complexes along with complement acti-
known as tumor necrosis factor ligand superfamily vation fragments (117, 199, 278). Overall, whether
member 13) and B-lymphocyte stimulator (also antibodies specific for periodontitis-associated bacte-
known as B-cell-activating factor or tumor necrosis ria play a protective, a destructive or an innocuous
factor ligand superfamily member 13B) have been role in the pathogenesis of periodontitis remains to
linked to the survival and maturation of human B- be clarified, as do the precise mechanisms involved.
cells (25, 157). Interleukin-6 and APRIL play impor- Since the early 1970s, it has become increasingly
tant roles in maintaining the survival of long-lived obvious that B-lineage cells have immune and
plasma cells within the bone marrow (135). Epithelial inflammatory functions that extend well beyond sim-
cells stimulated by toll-like receptor ligands consti- ply antibody production, such as antigen presenta-
tute a rich source of APRIL (100), while activated mye- tion to T-cells, expression of proinflammatory/
loid cells (e.g. neutrophils, macrophages and osteoclastogenic cytokines and expression of matrix
dendritic cells) express B-lymphocyte stimulator metalloproteinases, thereby having the potential to
(242). It was recently reported that expression and induce degradation of both soft and hard tissue.
production of both APRIL and B-lymphocyte stimula- Thus, it is feasible that these cells participate in the
tor are enhanced in gingival tissue from patients with immunopathogenesis of periodontitis via functions
periodontitis relative to healthy controls (1), and that are independent of their role in humoral
these findings were confirmed by a subsequent inde- immune responses (18, 74). Two of the hallmarks of
pendent study (164). The abundance of the above- the established and advanced lesion described by
discussed cytokines in periodontitis (relative to Page & Schroeder are collagen degradation and
health) can, at least in part, account for the mainte- alveolar bone resorption. Owing to the then-limited
nance and predominance of B-cells and plasma cells information, they were unaware of the potential of
in periodontitis lesions. B-lineage cells to influence in a proactive manner
Despite their abundance in advanced periodontal both of these processes. The association of elevated
lesions, the precise role of B-lineage cells in the numbers of plasma cells with an increase in collagen
pathogenesis of periodontitis remains quite enig- breakdown (113) is consistent with their ability to
matic, although several mechanisms are plausible release (in addition to interleukin-1, tumor necrosis
(Fig. 4). By mediating humoral immunity, B-cells/ factor and interleukin-6) matrix-degrading enzymes,
plasma cells could, in principle, have a protective including collagenase (18, 71, 111). B-lineage cells
function in periodontitis. For instance, the antibody also constitute a major source of both membrane-
response could contribute to the control of the bound and secreted RANKL in the bone-resorptive
polymicrobial dysbiotic communities in periodontal lesions of human periodontitis (122). Indeed, more
pockets and prevent invasion of bacteria into the gin- than 90% of B-cells express RANKL in periodontitis
gival connective tissue, thereby limiting inflammation lesions, whereas the percentage of RANKL-positive B-
and disease. A large number of studies have cells in healthy gingiva is negligible (122). It has
addressed this issue and the topic has been exten- recently been demonstrated that B-cells and plasma
sively reviewed (71, 243, 289). In general, antibody cells in diseased human periodontal tissue constitute
levels have typically been found to be elevated in dis- a significant source of the so-called secreted osteo-
eased individuals relative to healthy controls. Surpris- clastogenic factor of activated T-cells (112), a cytokine
ingly, the high titer of antibody found in patients that induces osteoclast differentiation via a RANKL-
does not appear to ameliorate the disease. It has been independent pathway (229). It is therefore plausible
suggested that antibody responses to periodontal that B-lineage cells may participate in the induction
bacteria in patients with periodontitis are of low of pathological bone loss in periodontitis via both
affinity and/or poor opsonophagocytic capacity (71, RANKL–dependent and -independent effects on
243, 289). In fact, individuals with the highest titers of osteoclast precursors.
antibody tend to exhibit severe alveolar bone loss rel-
ative to low-titer controls (130). Conceptually, anti-
Mechanistic studies in animal models
body, particularly IgG, has the capacity to exacerbate
and clinical intervention
inflammatory reactions and tissue degradation via a
number of mechanisms, including activation of com- The notion that B-lineage cells mediate bone loss in
plement and Fc receptor-mediated activation of periodontitis is supported by findings from animal

136
Revisiting the Page & Schroeder model

model studies. Adoptive transfer of bacterial antigen- lymphocyte stimulator (1, 3, 158), which are all factors
specific B-cells into rats followed by local oral expo- that promote B-cell activation, proliferation and dif-
sure to the same bacterial species (Aggregatibacter ferentiation (25, 29, 33, 157). Accordingly, in a liga-
actinomycetemcomitans) leads to RANKL-dependent ture-induced periodontitis study, gingival B-cells and
periodontal bone loss (98), even when the recipient plasma cells were detected in significant numbers
rats are congenitally athymic and hence T-cell defi- 5 days after initiation of periodontitis in wild-type
cient (94). This suggests that B-cells can contribute to mice, and their numbers increased through day 10,
periodontal bone loss independently of any possible correlating with increased expression of IgG during
cooperation with T-cells. IgD-deficient mice [which the same time interval (1). Consistent with the
have delayed B-cell affinity maturation (232)] exhibit absence of substantial numbers of B-lineage cells in
diminished B-cell activation and are protected wild-type mice during the first 5 days of the study,
against bone loss induced by oral gavage with P. gin- wild-type mice developed bone loss comparable with
givalis compared with wild-type controls (12). The that experienced by B-cell knockout mice at the same
role of B-cells in periodontitis was addressed more time interval (1). In stark contrast, however, wild-type
recently in studies utilizing wild-type and B-cell mice developed significantly more bone loss than did
knockout mice; these mice, also known as lMT, do B-cell knockout mice at day 10 when B-cells/plasma
not express membrane-bound IgM and consequently cells were abundant, suggesting that B-lineage cells
B-cell development is blocked at the pro-B stage (132). contribute to a late phase of ligature-induced alveolar
Utilizing an oral gavage model, one study implicated bone loss (1). In the same study, neutralizing mono-
B-cells in periodontal bone loss in normal mice as the clonal antibodies to APRIL and B-lymphocyte stimu-
results showed that C57BL/6J B-cell knockout mice lator diminished the number of B-cells in the gingival
are protected from P. gingivalis-induced bone loss rel- tissue of wild-type mice and protected them from
ative to wild-type mice, which developed modest bone bone loss. Both antibodies failed to inhibit bone loss
loss compared with sham-infected controls (209). in B-cell knockout mice, indicating that the observed
Another study reported that lean, normoglycemic effects of anti-APRIL and anti-B-lymphocyte stimula-
C57BL/6J B-cell knockout mice raised on a low-fat diet tor were mediated through their action on B-cells. In
were not protected from P. gingivalis-induced bone summary, B-cells and B-cell-stimulatory cytokines
loss, while obese, insulin-resistant B-cell knockout are temporally and causally linked to periodontal
animals were completely protected (302). The investi- bone loss.
gators interpreted the findings to suggest that B-cells Although the animal experiments described above
are ‘able to promote periodontitis only if the hosts are provide compelling evidence that B-cells are involved
primed by obesity’. The discrepancy between the two in the induction of alveolar bone loss, there is mini-
studies is not clear but different experimental condi- mal corroborating data from investigations on
tions, use of different P. gingivalis strains and fre- humans. A very recent study evaluated the periodon-
quency of their oral inoculation, might have tal status of two groups of patients with rheumatoid
contributed to the different outcome. Apparently, the arthritis treated with rituximab (anti-CD20 mono-
experimental protocol used in the latter study did not clonal antibody) (39). Based on the evaluation of a
readily support the development of a B-cell infiltrate number of clinical periodontal parameters, they
in lean mice that is characteristic of human periodon- showed that the majority of subjects exhibited an
titis, consistent with the authors’ comment that ‘later improvement in their periodontal status compared
phases of periodontal disease may be better studied in with that before initiation of rituximab therapy. It was
a model characterized by B-cell-dominated lesions concluded that B-cells play ‘a major role’ in the
that mimic long-term human disease’ (302). immunopathogenesis of periodontitis. Taken together
An alternative to the oral gavage model is the liga- with relevant clinical observations discussed above
ture-induced periodontitis model. The placement of a and animal model-based mechanistic studies, B-cells
ligature facilitates rapid accumulation of a heavy bio- and plasma cells do appear to contribute to the break-
film around the teeth and represents a model of down of soft- and/or hard-tissue components of the
accelerated inflammation and bone loss, which is periodontium, probably via several mechanisms.
more robust than the bone loss seen in the oral gav- In conclusion, the observations reported by Page &
age model and hence more sensitive to detect subtle Schroeder 40 years ago provided the impetus for
differences (2, 77). This model involves early and investigators to evaluate the functional significance of
strong activation of complement and expression of the accumulation of B-lineage cells in periodontitis
high levels of interleukin-6, as well as APRIL and B- lesions. It is now clear that these cells are anything

137
Hajishengallis & Korostoff

but innocent antibody-producing bystanders within promising candidate genes associated with periodon-
inflamed gingival connective tissue. In fact, they have tal disease (300).
the capacity to play an active role in the Also in recent years, complement started to be
immunopathogenesis of the disease (Fig. 4). appreciated as a central system of immune surveil-
lance and homeostasis and a key link between innate
and adaptive immunity (228). Specifically, it was real-
Complement ized that complement is not restricted to a linear cas-
cade of events but rather involves a network of
Page & Schroeder only briefly mentioned comple- interactions with other immune and physiological
ment in their landmark paper (213). Complement systems, which collectively coordinate host responses
was suspected to play a role in the initial lesion, as it to infection or tissue injury. For instance, comple-
was ‘probably present in the extravascular gingival ment activation can amplify immune and inflamma-
tissues’ but there was ‘insufficient evidence to deter- tory responses by synergizing with toll-like receptors
mine the role, if any, that these substances may play (86), form a barrier against the spread of invading
at this stage in the pathogenesis’ (213). Regarding the bacteria by augmenting local clotting (168) and regu-
established lesion, they cited ‘evidence for the pres- late antigen-presenting cells and the activation and
ence of complement and antigen–antibody com- differentiation of T-cell subsets (51). These develop-
plexes, especially around the blood vessels’ but ments greatly facilitated recent complement studies
similarly did not expand on its possible biological sig- in periodontitis.
nificance. However, soon after their publication, sev- As also acknowledged by Page & Schroeder, clinical
eral groups showed that gingival crevicular fluid from periodontal observations are correlative and do not
patients with periodontitis contains complement necessarily establish a cause-and-effect relationship
cleavage products at significantly higher levels than between complement activation and periodontitis.
in gingival crevicular fluid samples from healthy con- Evidence implicating a destructive role of comple-
trols (10, 245–247). Moreover, an abundance of com- ment in periodontitis was generated by recent inter-
plement components and cleavage products was vention studies in relevant animal models, including
demonstrated in chronically inflamed gingiva, nonhuman primates. Briefly, it has been shown that
whereas complement was undetectable or present at complement is exploited by periodontal bacteria in
lower levels in gingival biopsy samples from healthy ways that promote the dysbiotic transformation of
controls (41, 141, 277). Therefore, 5 years later, Page the microbial community and, moreover, comple-
& Schroeder acknowledged that complement may ment signaling pathways synergize with toll-like
play an important role in periodontitis, although ‘the receptors in eliciting an inflammatory response that
extent to which complement participation is protec- leads to alveolar bone destruction (3, 87, 90, 158, 162,
tive in contrast to destructive has not been resolved’ 287). It should be noted that, despite enhanced com-
(214). They discussed complement as a source of plement-dependent inflammation in periodontitis,
endogenous chemotactic substances (C5a) for the important disease-associated bacteria (such as P. gin-
recruitment of neutrophils, noting that its effects may givalis, Tannerella forsythia, Treponema denticola and
not necessarily be destructive because complement Prevotella intermedia) are equipped with several
could also play a protective role (promotion of bacte- mechanisms to protect themselves against comple-
rial phagocytosis through C3b opsonization) (214). ment-mediated opsonophagocytosis or lysis, includ-
Subsequently, it was shown that experimental gin- ing inhibition of opsonization and the ability to
givitis in human volunteers induces progressive com- capture and co-opt physiological inhibitors of com-
plement activation (as determined by C3 conversion) plement (e.g. factor H or C4b-binding protein) (116,
that correlates with elevated clinical inflammatory 166, 176, 224, 225, 244). On the basis of mouse studies
parameters (218). Conversely, but consistently, the that C3-dependent inflammation is critical for long-
resolution of periodontal inflammation in patients term sustenance of periodontal dysbiotic communi-
during successful therapy is associated with reduced ties and for maximal induction of alveolar bone loss
complement activation (198). More recently, a study (158), translational studies were undertaken in non-
using an integrative gene-prioritization method and human primates. These studies showed that local C3
databases from genome-wide association and inhibition (using the drug compstatin Cp40) blocks
microarray approaches has identified the central both inducible and naturally occurring periodontitis
complement component C3 amongst the top 21 most in cynomolgus monkeys, thus unequivocally linking

138
Revisiting the Page & Schroeder model

complement activation to periodontal tissue response. Intriguingly, immune-privileged tissues/or-


destruction (158, 159). gans display abundant expression of developmental
endothelial locus-1 at even higher levels than
peripheral tissues (e.g. lung or the gingiva) (35, 59),
Local tissue control of and developmental endothelial locus-1 deficiency in
inflammation in periodontitis mice promotes experimental autoimmune
encephalomyelitis, a model of multiple sclerosis (36).
This review has focused on the pathogenic role of Therefore, tissues and organs appear to have evolved
leukocytes infiltrating the periodontium, as the main local homeostatic mechanisms for protection against
objective was to revisit the Page & Schroeder model the destructive potential of the immune system. Iden-
of the involvement of myeloid and lymphoid cells in tification of the tissue-specific promoters and inhibi-
inflammatory periodontal disease. That said, it is tors of leukocyte recruitment, as well as the
becoming increasingly apparent that local tissues mechanisms that regulate them, is key to further our
may have greater control (than traditionally acknowl- understanding of the pathogenesis of plaque-induced
edged) over the initiation and properties of the devel- periodontal diseases and develop novel approaches
oping immune response, by virtue of their ability to to treat them.
communicate with and instruct the immune system
through tissue-derived (stromal/parenchymal) sig-
nals, such as cytokines, chemokines, antimicrobial The ‘double-edged sword’ nature of
peptides, growth factors and recently identified the periodontal host response
locally acting homeostatic molecules (84, 172, 262). In
other words, tissues are not passive recipients of Much of the uncertainty on the roles of the various
immune surveillance. In this context, tissue-derived cell types in periodontitis is probably a result of the
‘alarm’ signals (e.g. locally produced cytokines or ‘double-edged sword’ nature of the immune system
chemokines) can educate and appropriately activate and the complex etiology of periodontitis. The notion
antigen-presenting cells to promote a particular type that the host response can be intimately involved in
of host response (172). Conversely, when no longer the destructive process has been conclusively demon-
needed, the recruitment of inflammatory leukocytes strated in animal models. For instance, the inhibition
can be suppressed or arrested by local homeostatic or of the host response through various approaches (e.g.
‘health’ signals (e.g. endothelial-cell-derived develop- blocking complement activation, prostaglandin syn-
mental endothelial locus-1 or locally produced resol- thesis and proinflammatory/osteoclastogenic cytoki-
vins) to promote resolution of inflammation and nes, such as tumor necrosis factor, interleukin-1,
tissue regeneration (59, 172, 179, 251, 262). Stromal interleukin-17 and RANKL) was shown to reduce
cells in the periodontium, such as junctional epithe- inflammatory bone loss in several rodent and nonhu-
lial cells, gingival fibroblasts, periodontal ligament man primate models of inducible or naturally occur-
cells and vascular endothelial cells normally con- ring periodontitis (47, 59, 150, 154, 158, 159, 160, 205,
tribute to periodontal tissue homeostasis. However, 272). On the other hand, an optimally regulated host
under an inflammatory challenge, they appear to response can provide homeostatic immunity and
have the requisite plasticity to alter their phenotypes thus be protective. In this context, Page & Schroeder
and contribute to the host response, which, however, proposed that stable gingivitis may represent a pro-
has significant potential to cause tissue damage (63, tective host response that prevents transition to peri-
115, 145). The notion that local tissues have a ‘regula- odontitis (213). Ample evidence exists which shows
tory say’ over the host inflammatory response is con- that elements of innate or adaptive immunity can
sistent with evidence that immune-privileged sites prevent or ameliorate disease severity. For instance,
(e.g. the brain and the eye) have built-in mechanisms immunization of rats or nonhuman primates against
to promote the appropriate class of immune response P. gingivalis protects against bone loss induced by
that can potentially protect the tissue against infec- inoculation of this bacterium (72, 121, 211, 220, 226).
tion without destructive inflammation (172, 197, 264). Similar protective effects were observed after adop-
In this context, the spontaneous periodontal inflam- tive transfer of T-helper lymphocytes in a rat model
matory phenotype of developmental endothelial of periodontitis (293). Moreover, both humans and
locus-1-deficient mice suggests that tissues express- mice with defective ability to recruit neutrophils to
ing developmental endothelial locus-1 have the the periodontal tissue (e.g. because of leukocyte-
potential to control the local host inflammatory adhesion deficiency) fail to regulate the host response

139
Hajishengallis & Korostoff

properly and develop an aggressive form of periodon- known in the 1970s about crosstalk between leuko-
titis early in life (187). cytes via ‘products of activated lymphocytes’ (cytoki-
There is a fine balance between homeostatic nes) at the time that Page & Schroeder proposed their
immunity and immune pathology and it is possible model of periodontal disease pathogenesis (213), or
that the protective vs. pathogenic actions of leuko- even 5 years later in a subsequent review by the same
cytes in periodontitis reflect appropriate vs. aberrant authors (214). We now have an improved under-
operation of regulatory mechanisms. This, in turn, is standing of how different leukocyte types communi-
probably influenced, to a significant extent, by cate between themselves and with humoral systems,
genetic, epigenetic and environmental factors (128, such as complement, to stage immune and inflam-
146, 206), as well as by the immune-subversive capac- matory responses in the periodontium, and this
ity of the periodontal microbial communities (88). knowledge has offered promising therapeutic targets
Moreover, the ‘double-edged sword’ behavior of a (18, 66, 68, 81, 256, 271). Furthermore, we now appre-
given leukocyte type may be attributed to the activa- ciate that many of the infiltrating cells exhibit func-
tion of different subsets with functionally distinct tions that extend well beyond their classically
roles that could be protective or destructive, depend- accepted roles in the disease process. For example,
ing on the context. In this regard, the recent concept the contribution of neutrophils is not restricted to the
of immune plasticity has led to the realization that initial lesion; rather, neutrophils appear to be signifi-
the function of recruited leukocytes can be tailored to cantly involved in the chronic stages of periodontitis
the requirements of the tissue through intimate and may potentially contribute to the regulation of
crosstalk with tissue-derived factors; this adaptation both T- and B-lymphocytes (85) (Figs 1 and 2), as
can contribute critically to immune homeostasis and shown in other disease settings (239). The lympho-
resolution of inflammation (21, 26, 67, 255, 263). cytes, in turn, come in different ‘flavors’, that is, dif-
However, when the tissue environment is deficient in ferent functional subsets, each of which may play
critical homeostatic molecules, this becomes a pre- distinct roles in periodontal disease pathogenesis
disposing factor for leukocyte-driven inflammatory (Fig. 5). Regulatory T-cells, for instance, may have a
tissue destruction (59). In general, important protective role by preventing or mitigating excessive
advances have been made in the periodontal research inflammation, whereas T-helper cells, especially T-
field that can potentially provide insights as to when helper 17 cells, can become destructive as they are
leukocyte recruitment serves homeostatic immunity important sources of potent proinflammatory cytoki-
as opposed to pathology or when inflammation reso- nes and osteoclastogenic factors, including tumor
lution fails and proceeds to a chronic stage (56, 59, 93, necrosis factor, interleukin-17 and RANKL; the last is
175, 187, 208, 251, 281). Page & Schroeder proposed also produced in large amounts by activated B-cells
their periodontal pathogenesis model in the absence and plasma cells (122, 298) (Fig. 4).
of this recent knowledge (immune plasticity, special- Despite the progress made, there are significant
ized functional leukocyte subsets, local homeostatic gaps in our knowledge on the interrelationships of
mechanisms, etc.). Yet, their model was one that the types of leukocytes involved in periodontitis and
could be improved upon with emerging knowledge how the four lesions evolve. Thus, there are many
but not one that could be ignored or appreciated only exciting challenges ahead in the periodontal field. For
for its historical interest. example, T-helper 17 cells are now implicated as
effective B-cell helpers for antibody responses linked
to autoimmune and inflammatory conditions, such
Conclusions and future directions as arthritis (60, 102, 180), although their involvement
in the establishment of the B-cell/plasma-cell-rich
As alluded to above, the Page & Schroeder model lesions of periodontitis has not yet been addressed.
remains fundamentally relevant with regard to the Similarly to T-lymphocytes, mature macrophages also
main cellular players involved and their order of display heterogeneity and functional versatility (‘plas-
appearance during the development of periodontitis. ticity’). However, the role of specific macrophage sub-
What has changed tremendously in our understand- sets in periodontitis is largely unexplored. In
ing of this disease is the elucidation (albeit still functional analogy to regulatory T-cells, certain popu-
incomplete) of the cross-interactions and roles of the lations of B-cells, collectively termed regulatory B-
various players and that the host response develops cells, can inhibit immunopathology by suppressing
in a setting of dysbiosis rather than infection. In com- the expansion of pathogenic T-cell subsets such as T-
parison with current knowledge, relatively little was helper 1 and T-helper 17 cells (30, 233). Moreover,

140
Revisiting the Page & Schroeder model

another newly discovered subset of B-cells in mice past four decades, but this is an inevitable conse-
and humans, termed age-associated B-cells, is quence of the progress made: the more we under-
thought to represent a memory subset induced by stand, the more we know to ask. Nevertheless, the
nucleic acid-containing antigens in the context of an increased understanding of the cytokine and comple-
inflammatory cytokine environment (196). Age-asso- ment networks for innate-adaptive immune crosstalk,
ciated B-cells accumulate progressively with age, can the dissection (to some extent) of the molecular
have both protective and pathogenic effects (e.g. in mechanisms governing the recruitment, activation
autoimmunity) and appear to favor T-helper-cell and regulation of leukocytes and insights into the
polarization to a T-helper 17 profile (97, 196). The immune regulation of osteoclastogenesis have pro-
existence of age-associated B-cells or regulatory B- duced a number of promising therapeutic targets for
cells (let alone their function) in the periodontium has human periodontitis on the basis of successful pre-
yet to be addressed. Whereas it has long been known clinical studies (73, 78, 95, 99, 131, 169, 179, 254, 275).
that the numbers of mast cells are elevated in active
periodontal lesions (297), they were recently impli-
cated in the pathogenesis of experimental mouse peri- Acknowledgments
odontitis (165). As with age-associated B-cells, their
exact role (e.g. whether they are involved in early or We thank Debbie Maizels (Zoobotanica Scientific
late stages of the disease) remains unknown. Addi- Illustration) for redrawing the figures in this paper.
tionally, little is known about newly identified cell The authors’ research is supported by NIH grants;
types, such as the innate lymphoid cells, a family of AI068730, DE015254, DE021685, DE024153, DE024716
innate lymphocytes enriched at mucosal epithelial and DE026152.
barrier surfaces, where they have been implicated in
both homeostatic immunity and immune pathology.
The family comprises three different subsets, known References
as Group 1, Group 2 and Group 3, which mirror the
functional characteristics of T-helper 1, T-helper 2 1. Abe T, AlSarhan M, Benakanakere MR, Maekawa T, Kinane
and T-helper 17 cells, respectively (9). Interleukin-17- DF, Cancro MP, Korostoff JM, Hajishengallis G. The B cell-
stimulatory cytokines BLyS and APRIL are elevated in
expressing innate lymphoid cells have been identified
human periodontitis and are required for B cell-depen-
in the human and mouse periodontium (54, 187), dent bone loss in experimental murine periodontitis. J
although their exact role awaits elucidation. Immunol 2015: 195: 1427–1435.
Another open issue is the dichotomy between 2. Abe T, Hajishengallis G. Optimization of the ligature-
homeostatic protective and destructive host induced periodontitis model in mice. J Immunol Methods
2013: 394: 49–54.
responses. The components of these responses and
3. Abe T, Hosur KB, Hajishengallis E, Reis ES, Ricklin D, Lam-
mechanisms that regulate them have not yet been bris JD, Hajishengallis G. Local complement-targeted
adequately illuminated. If the microbial challenge is intervention in periodontitis: proof-of-concept using a
controlled by innate and – if necessary – adaptive C5a receptor (CD88) antagonist. J Immunol 2012: 189:
immune mechanisms then the inflammation is 5442–5448.
4. Abusleme L, Dupuy AK, Dutzan N, Silva N, Burleson JA,
resolved and tissue healing ensues. In such cases,
Strausbaugh LD, Gamonal J, Diaz PI. The subgingival
some initial tissue damage would be a small price to
microbiome in health and periodontitis and its relation-
pay. If the inflammatory response, however, does not ship with community biomass and inflammation. ISME J
control the dysbiotic insult, then the stage is set for 2013: 7: 1016–1025.
persistent chronic inflammation and overt periodon- 5. Allam JP, Duan Y, Heinemann F, Winter J, Gotz W, Desch-
titis, owing to collateral release of inflammatory ner J, Wenghoefer M, Bieber T, Jepsen S, Novak N. IL-23-
producing CD68(+) macrophage-like cells predominate
mediators, tissue-destructive enzymes and cytotoxic
within an IL-17-polarized infiltrate in chronic periodonti-
substances from the activated innate and adaptive tis lesions. J Clin Periodontol 2011: 38: 879–886.
leukocytes, as well as the generation of cleavage prod- 6. Alnaeeli M, Park J, Mahamed D, Penninger JM, Teng YT.
ucts of complement activation that have chemoat- Dendritic cells at the osteo-immune interface: implica-
tractant and cell-activating properties. tions for inflammation-induced bone loss. J Bone Miner
Res 2007: 22: 775–780.
In the last 40 years, we have come to know more
7. Arizon M, Nudel I, Segev H, Mizraji G, Elnekave M, Fur-
about the ‘bad’ and ‘good’ players in periodontitis manov K, Eli-Berchoer L, Clausen BE, Shapira L, Wilensky
but are still experiencing a journey of unknowns. A, Hovav AH. Langerhans cells down-regulate inflamma-
Indeed, the periodontal research field has generated tion-driven alveolar bone loss. Proc Natl Acad Sci USA
more questions than it has provided answers in the 2012: 109: 7043–7048.

141
Hajishengallis & Korostoff

8. Armitage GC. Periodontal diagnoses and classification of 25. Cancro MP. Signalling crosstalk in B cells: managing worth
periodontal diseases. Periodontol 2000 2004: 34: 9–21. and need. Nat Rev Immunol 2009: 9: 657–661.
9. Artis D, Spits H. The biology of innate lymphoid cells. Nat- 26. Carding SR, Egan PJ. Gammadelta T cells: functional plas-
ure 2015: 517: 293–301. ticity and heterogeneity. Nat Rev Immunol 2002: 2: 336–
10. Attstrom R, Laurel AB, Lahsson U, Sjoholm A. Comple- 345.
ment factors in gingival crevice material from healthy and 27. Cardoso CR, Garlet GP, Crippa GE, Rosa AL, Junior WM,
inflamed gingiva in humans. J Periodontal Res 1975: 10: Rossi MA, Silva JS. Evidence of the presence of T helper
19–27. type 17 cells in chronic lesions of human periodontal dis-
11. Baeza M, Garrido M, Hernandez-Rios P, Dezerega A, Gar- ease. Oral Microbiol Immunol 2009: 24: 1–6.
cia-Sesnich J, Strauss F, Aitken JP, Lesaffre E, Vanbelle S, 28. Cardoso CR, Garlet GP, Moreira AP, Junior WM, Rossi MA,
Gamonal J, Brignardello-Petersen R, Tervahartiala T, Sorsa Silva JS. Characterization of CD4+ CD25+ natural regula-
T, Hernandez M. Diagnostic accuracy for apical and tory T cells in the inflammatory infiltrate of human
chronic periodontitis biomarkers in gingival crevicular chronic periodontitis. J Leukoc Biol 2008: 84: 311–318.
fluid: an exploratory study. J Clin Periodontol 2016: 43: 34– 29. Carroll MC, Isenman DE. Regulation of humoral immunity
45. by complement. Immunity 2012: 37: 199–207.
12. Baker PJ, Boutaugh NR, Tiffany M, Roopenian DC. B Cell 30. Carter NA, Rosser EC, Mauri C. Interleukin-10 produced
IgD deletion prevents alveolar bone loss following murine by B cells is crucial for the suppression of Th17/Th1
oral infection. Interdiscip Perspect Infect Dis 2009: 2009: responses, induction of T regulatory type 1 cells and
864359. reduction of collagen-induced arthritis. Arthritis Res Ther
13. Baker PJ, Dixon M, Evans RT, Dufour L, Johnson E, Roope- 2012: 14: R32.
nian DC. CD4(+) T cells and the proinflammatory cytoki- 31. Chakravarti A, Raquil MA, Tessier P, Poubelle PE. Surface
nes gamma interferon and interleukin-6 contribute to RANKL of Toll-like receptor 4-stimulated human neu-
alveolar bone loss in mice. Infect Immun 1999: 67: 2804– trophils activates osteoclastic bone resorption. Blood 2009:
2809. 114: 1633–1644.
14. Baker PJ, Evans RT, Roopenian DC. Oral infection with 32. Chapple IL, Matthews JB. The role of reactive oxygen and
Porphyromonas gingivalis and induced alveolar bone loss antioxidant species in periodontal tissue destruction. Peri-
in immunocompetent and severe combined immunodefi- odontol 2000 2007: 43: 160–232.
cient mice. Arch Oral Biol 1994: 39: 1035–1040. 33. Chen-Kiang S. Regulation of terminal differentiation of
15. Beklen A, Ainola M, Hukkanen M, Gurgan C, Sorsa T, human B-cells by IL-6. Curr Top Microbiol Immunol 1995:
Konttinen YT. MMPs, IL-1, and TNF are regulated by IL-17 194: 189–198.
in periodontitis. J Dent Res 2007: 86: 347–351. 34. Cheng WC, Hughes FJ, Taams LS. The presence, function
16. Belibasakis GN, Bostanci N. The RANKL-OPG system in and regulation of IL-17 and Th17 cells in periodontitis. J
clinical periodontology. J Clin Periodontol 2012: 39: 239– Clin Periodontol 2014: 41: 541–549.
248. 35. Choi EY, Chavakis E, Czabanka MA, Langer HF, Fraemohs
17. Berglundh T, Donati M. Aspects of adaptive host response L, Economopoulou M, Kundu RK, Orlandi A, Zheng YY,
in periodontitis. J Clin Periodontol 2005: 32(Suppl. 6): 87– Prieto DA, Ballantyne CM, Constant SL, Aird WC, Papayan-
107. nopoulou T, Gahmberg CG, Udey MC, Vajkoczy P, Quert-
18. Berglundh T, Donati M, Zitzmann N. B cells in periodonti- ermous T, Dimmeler S, Weber C, Chavakis T. Del-1, an
tis: friends or enemies? Periodontol 2000 2007: 45: 51–66. endogenous leukocyte-endothelial adhesion inhibitor,
19. Beyrau M, Bodkin JV, Nourshargh S. Neutrophil hetero- limits inflammatory cell recruitment. Science 2008: 322:
geneity in health and disease: a revitalized avenue in 1101–1104.
inflammation and immunity. Open Biol 2012: 2: 120134. 36. Choi EY, Lim JH, Neuwirth A, Economopoulou M, Chatzi-
20. Bittner-Eddy PD, Fischer LA, Kaplan DH, Thieu K, Costa- georgiou A, Chung KJ, Bittner S, Lee SH, Langer H, Samus
longa M. Mucosal langerhans cells promote differentiation M, Kim H, Cho GS, Ziemssen T, Bdeir K, Chavakis E, Koh
of Th17 cells in a murine model of periodontitis but are JY, Boon L, Hosur K, Bornstein SR, Meuth SG, Hajishengal-
not required for Porphyromonas gingivalis-driven alveolar lis G, Chavakis T. Developmental endothelial locus-1 is a
bone destruction. J Immunol 2016: 197: 1435–1446. homeostatic factor in the central nervous system limiting
21. Bluestone JA, Mackay CR, O’Shea JJ, Stockinger B. The neuroinflammation and demyelination. Mol Psychiatry
functional plasticity of T cell subsets. Nat Rev Immunol 2015: 20: 880–888.
2009: 9: 811–816. 37. Chovatiya R, Medzhitov R. Stress, inflammation, and
22. Bosshardt DD, Lang NP. The junctional epithelium: from defense of homeostasis. Mol Cell 2014: 54: 281–288.
health to disease. J Dent Res 2005: 84: 9–20. 38. Christoffersson G, Vagesjo E, Vandooren J, Liden M,
23. Burns E, Bachrach G, Shapira L, Nussbaum G. Cutting Massena S, Reinert RB, Brissova M, Powers AC, Opde-
edge: TLR2 is required for the innate response to Porphy- nakker G, Phillipson M. VEGF-A recruits a proangio-
romonas gingivalis: activation leads to bacterial persis- genic MMP-9-delivering neutrophil subset that induces
tence and TLR2 deficiency attenuates induced alveolar angiogenesis in transplanted hypoxic tissue. Blood 2012:
bone resorption. J Immunol 2006: 177: 8296–8300. 120: 4653–4662.
24. Bystrom J, Evans I, Newson J, Stables M, Toor I, van Rooi- 39. Coat J, Demoersman J, Beuzit S, Cornec D, Devauchelle-
jen N, Crawford M, Colville-Nash P, Farrow S, Gilroy DW. Pensec V, Saraux A, Pers JO. Anti-B lymphocyte
Resolution-phase macrophages possess a unique inflam- immunotherapy is associated with improvement of peri-
matory phenotype that is controlled by cAMP. Blood 2008: odontal status in subjects with rheumatoid arthritis. J Clin
112: 4117–4127. Periodontol 2015: 42: 817–823.

142
Revisiting the Page & Schroeder model

40. Cooper PR, Palmer LJ, Chapple IL. Neutrophil extracellular 56. Ebersole JL, Kirakodu S, Novak MJ, Stromberg AJ, Shen S,
traps as a new paradigm in innate immunity: friend or foe? Orraca L, Gonzalez-Martinez J, Burgos A, Gonzalez OA.
Periodontol 2000 2013: 63: 165–197. Cytokine gene expression profiles during initiation, pro-
41. Courts FJ, Boackle RJ, Fudenberg HH, Silverman MS. gression and resolution of periodontitis. J Clin Periodontol
Detection of functional complement components in gingi- 2014: 41: 853–861.
val crevicular fluid from humans with periodontal dis- 57. Ebisuno Y, Tanaka T, Kanemitsu N, Kanda H, Yamaguchi
eases. J Dent Res 1977: 56: 327–331. K, Kaisho T, Akira S, Miyasaka M. Cutting edge: the B cell
42. Cua DJ, Tato CM. Innate IL-17-producing cells: the sen- chemokine CXC chemokine ligand 13/B lymphocyte
tinels of the immune system. Nat Rev Immunol 2010: 10: chemoattractant is expressed in the high endothelial
479–489. venules of lymph nodes and Peyer’s patches and affects B
43. Cutler CW, Teng YT. Oral mucosal dendritic cells and peri- cell trafficking across high endothelial venules. J Immunol
odontitis: many sides of the same coin with new twists. 2003: 171: 1642–1646.
Periodontol 2000 2007: 45: 35–50. 58. El-Awady AR, Arce RM, Cutler CW. Dendritic cells: micro-
44. Dababneh R, Al-Wahadneh AM, Hamadneh S, Khouri A, bial clearance via autophagy and potential immunobiolog-
Bissada NF. Periodontal manifestation of leukocyte adhe- ical consequences for periodontal disease. Periodontol
sion deficiency type I. J Periodontol 2008: 79: 764–768. 2000 2015: 69: 160–180.
45. Deas DE, Mackey SA, McDonnell HT. Systemic disease 59. Eskan MA, Jotwani R, Abe T, Chmelar J, Lim JH, Liang S,
and periodontitis: manifestations of neutrophil dysfunc- Ciero PA, Krauss JL, Li F, Rauner M, Hofbauer LC, Choi EY,
tion. Periodontol 2000 2003: 32: 82–104. Chung KJ, Hashim A, Curtis MA, Chavakis T, Hajishengallis
46. Deban L, Russo RC, Sironi M, Moalli F, Scanziani M, Zam- G. The leukocyte integrin antagonist Del-1 inhibits IL-17-
belli V, Cuccovillo I, Bastone A, Gobbi M, Valentino S, Doni mediated inflammatory bone loss. Nat Immunol 2012: 13:
A, Garlanda C, Danese S, Salvatori G, Sassano M, Evange- 465–473.
lista V, Rossi B, Zenaro E, Constantin G, Laudanna C, Bot- 60. Ettinger R, Sims GP, Robbins R, Withers D, Fischer RT,
tazzi B, Mantovani A. Regulation of leukocyte recruitment Grammer AC, Kuchen S, Lipsky PE. IL-21 and BAFF/BLyS
by the long pentraxin PTX3. Nat Immunol 2010: 11: 328– synergize in stimulating plasma cell differentiation from a
334. unique population of human splenic memory B cells. J
47. Delima AJ, Oates T, Assuma R, Schwartz Z, Cochran D, Immunol 2007: 178: 2872–2882.
Amar S, Graves DT. Soluble antagonists to interleukin-1 61. Fine N, Hassanpour S, Borenstein A, Sima C, Oveisi M,
(IL-1) and tumor necrosis factor (TNF) inhibits loss of tis- Scholey J, Cherney D, Glogauer M. Distinct oral neutrophil
sue attachment in experimental periodontitis. J Clin Peri- subsets define health and periodontal disease states. J
odontol 2001: 28: 233–240. Dental Res 2016: 95: 931–938.
48. Delima AJ, Van Dyke TE. Origin and function of the cellu- 62. Fournier BM, Parkos CA. The role of neutrophils during
lar components in gingival crevice fluid. Periodontol 2000 intestinal inflammation. Mucosal Immunol 2012: 5: 354–366.
2003: 31: 55–76. 63. Freire MO, Van Dyke TE. Natural resolution of inflamma-
49. Dewhirst FE, Chen T, Izard J, Paster BJ, Tanner AC, Yu tion. Periodontol 2000 2013: 63: 149–164.
WH, Lakshmanan A, Wade WG. The human oral micro- 64. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L,
biome. J Bacteriol 2010: 192: 5002–5017. Worthen GS, Albelda SM. Polarization of tumor-associated
50. Donati M, Liljenberg B, Zitzmann NU, Berglundh T. B-1a neutrophil phenotype by TGF-beta: “N1” versus “N2”
cells and plasma cells in periodontitis lesions. J Periodon- TAN. Cancer Cell 2009: 16: 183–194.
tal Res 2009: 44: 683–688. 65. Fujihashi K, Kono Y, Beagley KW, Yamamoto M, McGhee
51. Dunkelberger JR, Song WC. Complement and its role in JR, Mestecky J, Kiyono H. Cytokines and periodontal dis-
innate and adaptive immune responses. Cell Res 2010: 20: ease: immunopathological role of interleukins for B cell
34–50. responses in chronic inflamed gingival tissues. J Periodon-
52. Duran-Pinedo AE, Chen T, Teles R, Starr JR, Wang X, Krish- tol 1993: 64: 400–406.
nan K, Frias-Lopez J. Community-wide transcriptome of 66. Gaffen SL, Hajishengallis G. A new inflammatory cytokine
the oral microbiome in subjects with and without peri- on the block: re-thinking periodontal disease and the Th1/
odontitis. ISME J 2014: 8: 1659–1672. Th2 paradigm in the context of Th17 cells and IL-17. J Dent
53. Dutzan N, Gamonal J, Silva A, Sanz M, Vernal R. Over- Res 2008: 87: 817–828.
expression of forkhead box P3 and its association with 67. Galli SJ, Borregaard N, Wynn TA. Phenotypic and func-
receptor activator of nuclear factor-kappa B ligand, inter- tional plasticity of cells of innate immunity: macrophages,
leukin (IL) -17, IL-10 and transforming growth factor-beta mast cells and neutrophils. Nat Immunol 2011: 12: 1035–
during the progression of chronic periodontitis. J Clin Peri- 1044.
odontol 2009: 36: 396–403. 68. Garlet GP. Destructive and protective roles of cytokines in
54. Dutzan N, Konkel JE, Greenwell-Wild T, Moutsopoulos periodontitis: a re-appraisal from host defense and tissue
NM. Characterization of the human immune cell network destruction viewpoints. J Dent Res 2010: 89: 1349–1363.
at the gingival barrier. Mucosal Immunol 2016: 9: 1163– 69. Gasser O, Schifferli JA. Activated polymorphonuclear neu-
1172. trophils disseminate anti-inflammatory microparticles by
55. Dutzan N, Vernal R, Vaque JP, Garcia-Sesnich J, Hernan- ectocytosis. Blood 2004: 104: 2543–2548.
dez M, Abusleme L, Dezerega A, Gutkind JS, Gamonal J. 70. Gemmell E, Yamazaki K, Seymour GJ. Destructive peri-
Interleukin-21 expression and its association with proin- odontitis lesions are determined by the nature of the
flammatory cytokines in untreated chronic periodontitis lymphocytic response. Crit Rev Oral Biol Med 2002: 13:
patients. J Periodontol 2012: 83: 948–954. 17–34.

143
Hajishengallis & Korostoff

71. Gemmell E, Yamazaki K, Seymour GJ. The role of T cells in 89. Hajishengallis G, Lamont RJ, Graves DT. The enduring
periodontal disease: homeostasis and autoimmunity. Peri- importance of animal modelsin understanding periodon-
odontol 2000 2007: 43: 14–40. tal disease. Virulence 2015: 6: 229–235.
72. Gibson FC III, Genco CA. Prevention of Porphyromonas 90. Hajishengallis G, Liang S, Payne MA, Hashim A, Jotwani R,
gingivalis-induced oral bone loss following immuniza- Eskan MA, McIntosh ML, Alsam A, Kirkwood KL, Lambris
tion with gingipain R1. Infect Immun 2001: 69: 7959– JD, Darveau RP, Curtis MA. Low-abundance biofilm spe-
7963. cies orchestrates inflammatory periodontal disease
73. Glowacki AJ, Yoshizawa S, Jhunjhunwala S, Vieira AE, Gar- through the commensal microbiota and complement. Cell
let GP, Sfeir C, Little SR. Prevention of inflammation- Host Microbe 2011: 10: 497–506.
mediated bone loss in murine and canine periodontal dis- 91. Hajishengallis G, Moutsopoulos NM. Etiology of leukocyte
ease via recruitment of regulatory lymphocytes. Proc Natl adhesion deficiency-associated periodontitis revisited: not
Acad Sci USA 2013: 110: 18525–18530. a raging infection but a raging inflammatory response.
74. Gonzales JR. T- and B-cell subsets in periodontitis. Peri- Expert Rev Clin Immunol 2014: 10: 973–975.
odontol 2000 2015: 69: 181–200. 92. Hajishengallis G, Moutsopoulos NM. Role of bacteria in
75. Gonzalez OA, Novak MJ, Kirakodu S, Stromberg A, Nagara- leukocyte adhesion deficiency-associated periodontitis.
jan R, Huang CB, Chen KC, Orraca L, Martinez-Gonzalez J, Microb Pathog 2016: 94: 21–26.
Ebersole JL. Differential gene expression profiles reflecting 93. Hajishengallis G, Moutsopoulos NM, Hajishengallis E,
macrophage polarization in aging and periodontitis gingi- Chavakis T. Immune and regulatory functions of neu-
val tissues. Immunol Invest 2015: 44: 643–664. trophils in inflammatory bone loss. Semin Immunol 2016:
76. Graves D. Cytokines that promote periodontal tissue 28: 146–158.
destruction. J Periodontol 2008: 79: 1585–1591. 94. Han X, Kawai T, Eastcott JW, Taubman MA. Bacterial-
77. Graves DT, Fine D, Teng YT, Van Dyke TE, Hajishengallis responsive B lymphocytes induce periodontal bone
G. The use of rodent models to investigate host-bacteria resorption. J Immunol 2006: 176: 625–631.
interactions related to periodontal diseases. J Clin Peri- 95. Han X, Kawai T, Taubman MA. Interference with immune-
odontol 2008: 35: 89–105. cell-mediated bone resorption in periodontal disease.
78. Graves DT, Li J, Cochran DL. Inflammation and uncou- Periodontol 2000 2007: 45: 76–94.
pling as mechanisms of periodontal bone loss. J Dent Res 96. Hanna S, Etzioni A. Leukocyte adhesion deficiencies. Ann
2011: 90: 143–153. N Y Acad Sci 2012: 1250: 50–55.
79. Greer A, Irie K, Hashim A, Leroux BG, Chang AM, Curtis 97. Hao Y, O’Neill P, Naradikian MS, Scholz JL, Cancro MP. A
MA, Darveau RP. Site-specific neutrophil migration and B-cell subset uniquely responsive to innate stimuli accu-
CXCL2 expression in periodontal tissue. J Dent Res 2016: mulates in aged mice. Blood 2011: 118: 1294–1304.
95: 946–952. 98. Harada Y, Han X, Yamashita K, Kawai T, Eastcott JW,
80. Hajishengallis E, Hajishengallis G. Neutrophil homeostasis Smith DJ, Taubman MA. Effect of adoptive transfer of anti-
and periodontal health in children and adults. J Dent Res gen-specific B cells on periodontal bone resorption. J Peri-
2014: 93: 231–237. odontal Res 2006: 41: 101–107.
81. Hajishengallis G. Immunomicrobial pathogenesis of peri- 99. Hasturk H, Kantarci A, Van Dyke TE. Paradigm shift in the
odontitis: keystones, pathobionts, and host response. pharmacological management of periodontal diseases.
Trends Immunol 2014: 35: 3–11. Front Oral Biol 2012: 15: 160–176.
82. Hajishengallis G. The inflammophilic character of the peri- 100. He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J,
odontitis-associated microbiota. Mol Oral Microbiol 2014: Shan M, Chadburn A, Villanacci V, Plebani A, Knowles
29: 248–257. DM, Rescigno M, Cerutti A. Intestinal bacteria trigger T
83. Hajishengallis G. Periodontitis: from microbial immune cell-independent immunoglobulin A(2) class switching by
subversion to systemic inflammation. Nat Rev Immunol inducing epithelial-cell secretion of the cytokine APRIL.
2015: 15: 30–44. Immunity 2007: 26: 812–826.
84. Hajishengallis G, Chavakis T. Endogenous modulators of 101. Hernandez M, Gamonal J, Tervahartiala T, Mantyla P, Riv-
inflammatory cell recruitment. Trends Immunol 2013: 34: era O, Dezerega A, Dutzan N, Sorsa T. Associations
1–6. between matrix metalloproteinase-8 and -14 and
85. Hajishengallis G, Chavakis T, Hajishengallis E, Lambris JD. myeloperoxidase in gingival crevicular fluid from subjects
Neutrophil homeostasis and inflammation: novel para- with progressive chronic periodontitis: a longitudinal
digms from studying periodontitis. J Leukoc Biol 2015: 98: study. J Periodontol 2010: 81: 1644–1652.
539–548. 102. Hickman-Brecks CL, Racz JL, Meyer DM, LaBranche TP,
86. Hajishengallis G, Lambris JD. Crosstalk pathways between Allen PM. Th17 cells can provide B cell help in autoanti-
Toll-like receptors and the complement system. Trends body induced arthritis. J Autoimmun 2011: 36: 65–75.
Immunol 2010: 31: 154–163. 103. Holliday LS, Welgus HG, Fliszar CJ, Veith GM, Jeffrey JJ,
87. Hajishengallis G, Lambris JD. Microbial manipulation of Gluck SL. Initiation of osteoclast bone resorption by inter-
receptor crosstalk in innate immunity. Nat Rev Immunol stitial collagenase. J Biol Chem 1997: 272: 22053–22058.
2011: 11: 187–200. 104. Hua Z, Hou B. TLR signaling in B-cell development and
88. Hajishengallis G, Lamont RJ. Dancing with the stars: how activation. Cell Mol Immunol 2013: 10: 103–106.
choreographed bacterial interactions dictate nososym- 105. Huard B, McKee T, Bosshard C, Durual S, Matthes T, Myit
biocity and give rise to keystone pathogens, accessory S, Donze O, Frossard C, Chizzolini C, Favre C, Zubler R,
pathogens, and pathobionts. Trends Microbiol 2016: 24: Guyot JP, Schneider P, Roosnek E. APRIL secreted by neu-
477–489. trophils binds to heparan sulfate proteoglycans to create

144
Revisiting the Page & Schroeder model

plasma cell niches in human mucosa. J Clin Invest 2008: Miranda LA, Ernst CW, Izumi Y, Taubman MA. B and T
118: 2887–2895. lymphocytes are the primary sources of RANKL in the
106. Huntington ND, Carpentier S, Vivier E, Belz GT. Innate bone resorptive lesion of periodontal disease. Am J Pathol
lymphoid cells: parallel checkpoints and coordinate inter- 2006: 169: 987–998.
actions with T cells. Curr Opin Immunol 2016: 38: 86–93. 123. Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads
107. Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tis- of innate and adaptive immunity against infectious dis-
sue-specific context. Nat Rev Immunol 2014: 14: 81–93. eases at the mucosa. Mucosal Immunol 2009: 2: 403–411.
108. Ito H, Honda T, Domon H, Oda T, Okui T, Amanuma R, 124. Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly
Nakajima T, Yamazaki K. Gene expression analysis of the DJ, Popovich PG. Identification of two distinct macro-
CD4+ T-cell clones derived from gingival tissues of peri- phage subsets with divergent effects causing either neuro-
odontitis patients. Oral Microbiol Immunol 2005: 20: 382– toxicity or regeneration in the injured mouse spinal cord. J
386. Neurosci 2009: 29: 13435–13444.
109. Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional spe- 125. Kikuta J, Wada Y, Kowada T, Wang Z, Sun-Wada GH,
cialization of interleukin-17 family members. Immunity Nishiyama I, Mizukami S, Maiya N, Yasuda H, Kumanogoh
2011: 34: 149–162. A, Kikuchi K, Germain RN, Ishii M. Dynamic visualization
110. Jablonska J, Leschner S, Westphal K, Lienenklaus S, Weiss of RANKL and Th17-mediated osteoclast function. J Clin
S. Neutrophils responsive to endogenous IFN-b regulate Invest 2013: 123: 866–873.
tumor angiogenesis and growth in a mouse tumor model. 126. Kim PD, Xia-Juan X, Crump KE, Abe T, Hajishengallis G,
J Clin Invest 2010: 120: 1151–1164. Sahingur SE. Toll-like receptor 9-mediated inflammation
111. Jagannathan M, Hasturk H, Liang Y, Shin H, Hetzel JT, triggers alveolar bone loss in experimental murine peri-
Kantarci A, Rubin D, McDonnell ME, Van Dyke TE, Gan- odontitis. Infect Immun 2015: 83: 2992–3002.
ley-Leal LM, Nikolajczyk BS. TLR cross-talk specifically 127. Kim YC, Ko Y, Hong SD, Kim KY, Lee YH, Chae C, Choi Y.
regulates cytokine production by B cells from chronic Presence of Porphyromonas gingivalis and plasma cell
inflammatory disease patients. J Immunol 2009: 183: dominance in gingival tissues with periodontitis. Oral Dis
7461–7470. 2010: 16: 375–381.
112. Jarry CR, Martinez EF, Peruzzo DC, Carregaro V, Sacra- 128. Kinane DF, Demuth DR, Gorr SU, Hajishengallis GN, Mar-
mento LA, Araujo VC, Weitzmann MN, Napimoga MH. tin MH. Human variability in innate immunity. Periodon-
Expression of SOFAT by T- and B-lineage cells may con- tol 2000 2007: 45: 14–34.
tribute to bone loss. Mol Med Rep 2016: 13: 4252–4258. 129. Kinane DF, Hart TC. Genes and gene polymorphisms asso-
113. Joachim F, Barber P, Newman HN, Osborn J. The plasma ciated with periodontal disease. Crit Rev Oral Biol Med
cell at the advancing front of the lesion in chronic peri- 2003: 14: 430–449.
odontitis. J Periodontal Res 1990: 25: 49–59. 130. Kinane DF, Mooney J, Ebersole JL. Humoral immune
114. Johnson RB, Wood N, Serio FG. Interleukin-11 and IL-17 response to Actinobacillus actinomycetemcomitans and
and the pathogenesis of periodontal disease. J Periodontol Porphyromonas gingivalis in periodontal disease. Peri-
2004: 75: 37–43. odontol 2000 1999: 20: 289–340.
115. Jonsson D, Nebel D, Bratthall G, Nilsson BO. The human 131. Kirkwood KL, Cirelli JA, Rogers JE, Giannobile WV. Novel
periodontal ligament cell: a fibroblast-like cell acting as an host response therapeutic approaches to treat periodontal
immune cell. J Periodontal Res 2011: 46: 153–157. diseases. Periodontol 2000 2007: 43: 294–315.
116. Jusko M, Potempa J, Karim AY, Ksiazek M, Riesbeck K, 132. Kitamura D, Roes J, Kuhn R, Rajewsky K. A B cell-deficient
Garred P, Eick S, Blom AM. A metalloproteinase karilysin mouse by targeted disruption of the membrane exon of
present in the majority of Tannerella forsythia isolates the immunoglobulin mu chain gene. Nature 1991: 350:
inhibits all pathways of the complement system. J Immu- 423–426.
nol 2012: 188: 2338–2349. 133. Kobayashi R, Kono T, Bolerjack BA, Fukuyama Y, Gilbert
117. Kajiya M, Giro G, Taubman MA, Han X, Mayer MPA, Kawai RS, Fujihashi K, Ruby J, Kataoka K, Wada M, Yamamoto
T. Role of periodontal pathogenic bacteria in RANKL- M, Fujihashi K. Induction of IL-10-producing CD4+ T-cells
mediated bone destruction in periodontal disease. J Oral in chronic periodontitis. J Dent Res 2011: 90: 653–658.
Microbiol 2010: 2: doi: 10.3402/jom.v2i0.5532. 134. Kolaczkowska E, Kubes P. Neutrophil recruitment and
118. Kalyan S, Kabelitz D. When neutrophils meet T cells: function in health and inflammation. Nat Rev Immunol
beginnings of a tumultuous relationship with underappre- 2013: 13: 159–175.
ciated potential. Eur J Immunol 2014: 44: 627–633. 135. Kometani K, Kurosaki T. Differentiation and maintenance
119. Kanzaki H, Chiba M, Shimizu Y, Mitani H. Dual regulation of long-lived plasma cells. Curr Opin Immunol 2015: 33:
of osteoclast differentiation by periodontal ligament cells 64–69.
through RANKL stimulation and OPG inhibition. J Dent 136. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17
Res 2001: 80: 887–891. Cells. Annu Rev Immunol 2009: 27: 485–517.
120. Kaplan DH. In vivo function of Langerhans cells and der- 137. Kornman KS, Page RC, Tonetti MS. The host response to
mal dendritic cells. Trends Immunol 2010: 31: 446–451. the microbial challenge in periodontitis: assembling the
121. Katz J, Black KP, Michalek SM. Host responses to recombi- players. Periodontol 2000 1997: 14: 33–53.
nant hemagglutinin B of Porphyromonas gingivalis in an 138. Korns D, Frasch SC, Fernandez-Boyanapalli R, Henson
experimental rat model. Infect Immun 1999: 67: 4352– PM, Bratton DL. Modulation of macrophage efferocytosis
4359. in inflammation. Front Immunol 2011: 2: 57.
122. Kawai T, Matsuyama T, Hosokawa Y, Makihira S, Seki M, 139. Kotake S, Yago T, Kawamoto M, Nanke Y. Role of osteo-
Karimbux NY, Goncalves RB, Valverde P, Dibart S, Li YP, clasts and interleukin-17 in the pathogenesis of

145
Hajishengallis & Korostoff

rheumatoid arthritis: crucial ‘human osteoclastology’. J 155. Ling MR, Chapple IL, Matthews JB. Peripheral blood neu-
Bone Miner Metab 2012: 30: 125–135. trophil cytokine hyper-reactivity in chronic periodontitis.
140. Lakschevitz FS, Aboodi GM, Glogauer M. Oral neutrophil Innate Immun 2015: 21: 714–725.
transcriptome changes result in a pro-survival phenotype 156. Lubberts E. IL-17/Th17 targeting: on the road to prevent
in periodontal diseases. PLoS ONE 2013: 8: e68983. chronic destructive arthritis? Cytokine 2008: 41: 84–91.
141. Lally ET, McArthur WP, Baehni PC. Biosynthesis of com- 157. Mackay F, Schneider P, Rennert P, Browning J. BAFF AND
plement components in chronically inflamed gingiva. J APRIL: a tutorial on B cell survival. Annu Rev Immunol
Periodontal Res 1982: 17: 257–262. 2003: 21: 231–264.
142. Lam RS, O’Brien-Simpson NM, Lenzo JC, Holden JA, 158. Maekawa T, Abe T, Hajishengallis E, Hosur KB, DeAngelis
Brammar GC, Walsh KA, McNaughtan JE, Rowler DK, Van RA, Ricklin D, Lambris JD, Hajishengallis G. Genetic and
Rooijen N, Reynolds EC. Macrophage depletion abates intervention studies implicating complement C3 as a
Porphyromonas gingivalis-induced alveolar bone resorp- major target for the treatment of periodontitis. J Immunol
tion in mice. J Immunol 2014: 193: 2349–2362. 2014: 192: 6020–6027.
143. Lamont RJ, Hajishengallis G. Polymicrobial synergy and 159. Maekawa T, Briones RA, Resuello RR, Tuplano JV,
dysbiosis in inflammatory disease. Trends Mol Med 2015: Hajishengallis E, Kajikawa T, Koutsogiannaki S, Garcia CA,
21: 172–183. Ricklin D, Lambris JD, Hajishengallis G. Inhibition of pre-
144. Landzberg M, Doering H, Aboodi GM, Tenenbaum HC, existing natural periodontitis in non-human primates by a
Glogauer M. Quantifying oral inflammatory load: oral neu- locally administered peptide inhibitor of complement C3.
trophil counts in periodontal health and disease. J Peri- J Clin Periodontol 2016: 43: 238–249.
odontal Res 2015: 50: 330–336. 160. Maekawa T, Hajishengallis G. Topical treatment with pro-
145. Larjava H, Koivisto L, Heino J, Hakkinen L. Integrins in biotic Lactobacillus brevis CD2 inhibits experimental peri-
periodontal disease. Exp Cell Res 2014: 325: 104–110. odontal inflammation and bone loss. J Periodontal Res
146. Larsson L, Castilho RM, Giannobile WV. Epigenetics and 2014: 49: 785–791.
its role in periodontal diseases: a state-of-the-art review. J 161. Maekawa T, Hosur K, Abe T, Kantarci A, Ziogas A, Wang B,
Periodontol 2015: 86: 556–568. Van Dyke TE, Chavakis T, Hajishengallis G. Antagonistic
147. Lee W, Aitken S, Sodek J, McCulloch CA. Evidence of a effects of IL-17 and D-resolvins on endothelial Del-1
direct relationship between neutrophil collagenase activity expression through a GSK-3beta-C/EBPbeta pathway. Nat
and periodontal tissue destruction in vivo: role of active Commun 2015: 6: 8272.
enzyme in human periodontitis. J Periodontal Res 1995: 162. Maekawa T, Krauss JL, Abe T, Jotwani R, Triantafilou M,
30: 23–33. Triantafilou K, Hashim A, Hoch S, Curtis MA, Nussbaum
148. Lester SR, Bain JL, Johnson RB, Serio FG. Gingival concen- G, Lambris JD, Hajishengallis G. Porphyromonas gingivalis
trations of interleukin-23 and -17 at healthy sites and at manipulates complement and TLR signaling to uncouple
sites of clinical attachment loss. J Periodontol 2007: 78: bacterial clearance from inflammation and promote dys-
1545–1550. biosis. Cell Host Microbe 2014: 15: 768–778.
149. Lewkowicz N, Mycko MP, Przygodzka P, Cwiklinska H, 163. Maffia PC, Zittermann SE, Scimone ML, Tateosian N, Ami-
Cichalewska M, Matysiak M, Selmaj K, Lewkowicz P. ano N, Guerrieri D, Lutzky V, Rosso D, Romeo HE, Garcia
Induction of human IL-10-producing neutrophils by LPS- VE, Issekutz AC, Chuluyan HE. Neutrophil elastase con-
stimulated Treg cells and IL-10. Mucosal Immunol 2016: 9: verts human immature dendritic cells into transforming
364–378. growth factor-beta1-secreting cells and reduces allostimu-
150. Li KL, Vogel R, Jeffcoat MK, Alfano MC, Smith MA, Collins latory ability. Am J Pathol 2007: 171: 928–937.
JG, Offenbacher S. The effect of ketoprofen creams on 164. Mahanonda R, Champaiboon C, Subbalekha K, Sa-Ard-
periodontal disease in rhesus monkeys. J Periodontal Res Iam N, Rattanathammatada W, Thawanaphong S, Rerkyen
1996: 31: 525–532. P, Yoshimura F, Nagano K, Lang NP, Pichyangkul S.
151. Liang S, Krauss JL, Domon H, McIntosh ML, Hosur KB, Qu Human memory B cells in healthy gingiva, gingivitis, and
H, Li F, Tzekou A, Lambris JD, Hajishengallis G. The C5a periodontitis. J Immunol 2016: 197: 715–725.
receptor impairs IL-12-dependent clearance of Porphy- 165. Malcolm J, Millington O, Millhouse E, Campbell L, Adra-
romonas gingivalis and is required for induction of peri- dos Planell A, Butcher JP, Lawrence C, Ross K, Ramage G,
odontal bone loss. J Immunol 2011: 186: 869–877. McInnes IB, Culshaw S. Mast Cells contribute to Porphy-
152. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joan- romonas gingivalis-induced bone loss. J Dent Res 2016: 95:
nopoulos K, Collins M, Fouser LA. Interleukin (IL)-22 and 704–710.
IL-17 are coexpressed by Th17 cells and cooperatively 166. Malm S, Jusko M, Eick S, Potempa J, Riesbeck K, Blom
enhance expression of antimicrobial peptides. J Exp Med AM. Acquisition of complement inhibitor serine pro-
2006: 203: 2271–2279. tease factor I and its cofactors C4b-binding protein and
153. Lin D, Li L, Sun Y, Wang W, Wang X, Ye Y, Chen X, Xu factor H by Prevotella intermedia. PLoS ONE 2012: 7:
Y. IL-17 regulates the expressions of RANKL and OPG e34852.
in human periodontal ligament cells via TRAF6/TBK1- 167. Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neu-
JNK/NF-kappaB pathways. Immunology 2014: 144: trophils in the activation and regulation of innate and
472–485. adaptive immunity. Nat Rev Immunol 2011: 11: 519–531.
154. Lin X, Han X, Kawai T, Taubman MA. Antibody to receptor 168. Markiewski MM, Nilsson B, Ekdahl KN, Mollnes TE,
activator of NF-jB ligand ameliorates T cell-mediated Lambris JD. Complement and coagulation: strangers
periodontal bone resorption. Infect Immun 2011: 79: 911– or partners in crime? Trends Immunol 2007: 28: 184–
917. 192.

146
Revisiting the Page & Schroeder model

169. Mastellos DC, Ricklin D, Hajishengallis E, Hajishengallis G, 186. Moutsopoulos NM, Kling HM, Angelov N, Jin W, Palmer
Lambris JD. Complement therapeutics in inflammatory RJ, Nares S, Osorio M, Wahl SM. Porphyromonas gingivalis
diseases: promising drug candidates for C3-targeted inter- promotes Th17 inducing pathways in chronic periodonti-
vention. Mol Oral Microbiol 2016: 31: 3–17. tis. J Autoimmun 2012: 39: 294–303.
170. Matsuda T, Yamasaki K, Taga T, Hirano T, Kishimoto T. 187. Moutsopoulos NM, Konkel J, Sarmadi M, Eskan MA, Wild
Current concepts of B cell modulation. Int Rev Immunol T, Dutzan N, Abusleme L, Zenobia C, Hosur KB, Abe T,
1989: 5: 97–109. Uzel G, Chen W, Chavakis T, Holland SM, Hajishengallis
171. Matthews JB, Wright HJ, Roberts A, Ling-Mountford N, G. Defective neutrophil recruitment in leukocyte adhesion
Cooper PR, Chapple IL. Neutrophil hyper-responsiveness deficiency type I disease causes local IL-17–driven inflam-
in periodontitis. J Dent Res 2007: 86: 718–722. matory bone loss. Sci Transl Med 2014: 6: 229ra240.
172. Matzinger P, Kamala T. Tissue-based class control: the 188. Moutsopoulos NM, Lionakis MS, Hajishengallis G. Inborn
other side of tolerance. Nat Rev Immunol 2011: 11: 221– errors in immunity: unique natural models to dissect oral
230. immunity. J Dent Res 2015: 94: 753–758.
173. Mayadas TN, Cullere X, Lowell CA. The multifaceted func- 189. Munder M, Schneider H, Luckner C, Giese T, Langhans
tions of neutrophils. Annu Rev Pathol 2014: 9: 181–218. CD, Fuentes JM, Kropf P, Mueller I, Kolb A, Modolell M,
174. Medzhitov R. Recognition of microorganisms and acti- Ho AD. Suppression of T-cell functions by human granu-
vation of the immune response. Nature 2007: 449: 819– locyte arginase. Blood 2006: 108: 1627–1634.
826. 190. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW,
175. Meyle J, Chapple I. Molecular aspects of the pathogenesis Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T,
of periodontitis. Periodontol 2000 2015: 69: 7–17. Locati M, Mantovani A, Martinez FO, Mege JL, Mosser
176. Miller DP, Bell JK, McDowell JV, Conrad DH, Burgner JW, DM, Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica A, Sut-
Heroux A, Marconi RT. Structure of factor H-binding pro- tles J, Udalova I, van Ginderachter JA, Vogel SN, Wynn TA.
tein B (FhbB) of the periopathogen, Treponema denticola: Macrophage activation and polarization: nomenclature
insights into progression of periodontal disease. J Biol and experimental guidelines. Immunity 2014: 41: 14–20.
Chem 2012: 287: 12715–12722. 191. Murray PJ, Wynn TA. Protective and pathogenic functions
177. Mills CD. M1 and M2 macrophages: oracles of health and of macrophage subsets. Nat Rev Immunol 2011: 11: 723–
disease. Crit Rev Immunol 2012: 32: 463–488. 737.
178. Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in 192. Nakajima T, Amanuma R, Ueki-Maruyama K, Oda T,
chronic inflammation. Nat Rev Drug Discov 2012: 11: 763– Honda T, Ito H, Yamazaki K. CXCL13 expression and follic-
776. ular dendritic cells in relation to B-cell infiltration in peri-
179. Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis odontal disease tissues. J Periodontal Res 2008: 43: 635–
G, Chavakis T. Leukocyte integrins: role in leukocyte 641.
recruitment and as therapeutic targets in inflammatory 193. Nakajima T, Ueki-Maruyama K, Oda T, Ohsawa Y, Ito H,
disease. Pharmacol Ther 2015: 147: 123–135. Seymour GJ, Yamazaki K. Regulatory T-cells infiltrate peri-
180. Mitsdoerffer M, Lee Y, Jager A, Kim HJ, Korn T, Kolls JK, odontal disease tissues. J Dent Res 2005: 84: 639–643.
Cantor H, Bettelli E, Kuchroo VK. Proinflammatory T 194. Nakashima T, Hayashi M, Takayanagi H. New insights into
helper type 17 cells are effective B-cell helpers. Proc Natl osteoclastogenic signaling mechanisms. Trends Endocrinol
Acad Sci USA 2010: 107: 14292–14297. Metab 2012: 23: 582–590.
181. Mizutani Y, Tsuge S, Takeda H, Hasegawa Y, Shiogama K, 195. Nakashima T, Kobayashi Y, Yamasaki S, Kawakami A, Egu-
Onouchi T, Inada K, Sawasaki T, Tsutsumi Y. In situ visual- chi K, Sasaki H, Sakai H. Protein expression and functional
ization of plasma cells producing antibodies reactive to difference of membrane-bound and soluble receptor acti-
Porphyromonas gingivalis in periodontitis: the application vator of NF-kappaB ligand: modulation of the expression
of the enzyme-labeled antigen method. Mol Oral Micro- by osteotropic factors and cytokines. Biochem Biophys Res
biol 2014: 29: 156–173. Commun 2000: 275: 768–775.
182. Mocsai A. Diverse novel functions of neutrophils in immu- 196. Naradikian MS, Hao Y, Cancro MP. Age-associated B cells:
nity, inflammation, and beyond. J Exp Med 2013: 210: key mediators of both protective and autoreactive
1283–1299. humoral responses. Immunol Rev 2016: 269: 118–129.
183. Mohamed SG, Sugiyama E, Shinoda K, Taki H, Hounoki H, 197. Niederkorn JY, Stein-Streilein J. History and physiology of
Abdel-Aziz HO, Maruyama M, Kobayashi M, Ogawa H, immune privilege. Ocul Immunol Inflamm 2010: 18: 19–
Miyahara T. Interleukin-10 inhibits RANKL-mediated 23.
expression of NFATc1 in part via suppression of c-Fos and 198. Niekrash CE, Patters MR. Simultaneous assessment of
c-Jun in RAW264.7 cells and mouse bone marrow cells. complement components C3, C4, and B and their cleavage
Bone 2007: 41: 592–602. products in human gingival fluid. II. Longitudinal changes
184. Mosser DM, Edwards JP. Exploring the full spectrum of during periodontal therapy. J Periodontal Res 1985: 20:
macrophage activation. Nat Rev Immunol 2008: 8: 958– 268–275.
969. 199. Nikolopoulou-Papaconstantinou AA, Johannessen AC,
185. Moutsopoulos NM, Chalmers NI, Barb JJ, Abusleme L, Kristoffersen T. Deposits of immunoglobulins, comple-
Greenwell-Wild T, Dutzan N, Paster BJ, Munson PJ, Fine ment, and immune complexes in inflamed human gingiva.
DH, Uzel G, Holland SM. Subgingival microbial communi- Acta Odontol Scand 1987: 45: 187–193.
ties in leukocyte adhesion deficiency and their relation- 200. Noack M, Miossec P. Th17 and regulatory T cell balance in
ship with local immunopathology. PLoS Pathog 2015: 11: autoimmune and inflammatory diseases. Autoimmun Rev
e1004698. 2014: 13: 668–677.

147
Hajishengallis & Korostoff

201. Nussbaum G, Shapira L. How has neutrophil research 216. Parachuru VP, Coates DE, Milne TJ, Hussaini HM, Rich
improved our understanding of periodontal pathogenesis? AM, Seymour GJ. Forkhead box P3-positive regulatory T-
J Clin Periodontol 2011: 38: 49–59. cells and interleukin 17-positive T-helper 17 cells in
202. O’Neill LA, Golenbock D, Bowie AG. The history of Toll- chronic inflammatory periodontal disease. J Periodontal
like receptors – redefining innate immunity. Nat Rev Res 2014: 49: 817–826.
Immunol 2013: 13: 453–460. 217. Park-Min KH, Ji JD, Antoniv T, Reid AC, Silver RB, Hum-
203. O’Shea JJ, Paul WE. Mechanisms underlying lineage com- phrey MB, Nakamura M, Ivashkiv LB. IL-10 suppresses
mitment and plasticity of helper CD4+ T cells. Science calcium-mediated costimulation of receptor activator NF-
2010: 327: 1098–1102. kappa B signaling during human osteoclast differentiation
204. Odobasic D, Kitching AR, Yang Y, O’Sullivan KM, Muljadi by inhibiting TREM-2 expression. J Immunol 2009: 183:
RC, Edgtton KL, Tan DS, Summers SA, Morand EF, Holds- 2444–2455.
worth SR. Neutrophil myeloperoxidase regulates T-cell- 218. Patters MR, Niekrash CE, Lang NP. Assessment of comple-
driven tissue inflammation in mice by inhibiting dendritic ment cleavage in gingival fluid during experimental gin-
cell function. Blood 2013: 121: 4195–4204. givitis in man. J Clin Periodontol 1989: 16: 33–37.
205. Offenbacher S, Braswell LD, Loos AS, Johnson HG, Hall 219. Pelletier M, Maggi L, Micheletti A, Lazzeri E, Tamassia N,
CM, McClure H, Orkin JL, Strobert EA, Green MD, Odle Costantini C, Cosmi L, Lunardi C, Annunziato F, Romag-
BM. Effects of flurbiprofen on the progression of peri- nani S, Cassatella MA. Evidence for a cross-talk between
odontitis in Macaca mulatta. J Periodontal Res 1987: 22: human neutrophils and Th17 cells. Blood 2009: 115: 335–
473–481. 343.
206. Offenbacher S, Divaris K, Barros SP, Moss KL, March- 220. Persson GR, Engel D, Whitney C, Darveau R, Weinberg A,
esan JT, Morelli T, Zhang S, Kim S, Sun L, Beck JD, Brunsvold M, Page RC. Immunization against Porphy-
Laudes M, Munz M, Schaefer AS, North KE. Genome- romonas gingivalis inhibits progression of experimental
wide association study of biologically-informed peri- periodontitis in nonhuman primates. Infect Immun 1994:
odontal complex traits offers novel insights into the 62: 1026–1031.
genetic basis of periodontal disease. Hum Mol Genet 221. Petermann F, Rothhammer V, Claussen MC, Haas JD,
2016: pii: ddw069. Blanco LR, Heink S, Prinz I, Hemmer B, Kuchroo VK,
207. Ohyama H, Kato-Kogoe N, Kuhara A, Nishimura F, Naka- Oukka M, Korn T. cd T cells enhance autoimmunity by
sho K, Yamanegi K, Yamada N, Hata M, Yamane J, Terada restraining regulatory T cell responses via an interleukin-
N. The involvement of IL-23 and the Th17 pathway in peri- 23-dependent mechanism. Immunity 2010: 33: 351–363.
odontitis. J Dent Res 2009: 88: 633–638. 222. Phillipson M, Kubes P. The neutrophil in vascular inflam-
208. Okui T, Aoki Y, Ito H, Honda T, Yamazaki K. The presence mation. Nat Med 2011: 17: 1381–1390.
of IL-17+/FOXP3+ double-positive cells in periodontitis. J 223. Pillay J, Kamp VM, van Hoffen E, Visser T, Tak T, Lammers
Dent Res 2012: 91: 574–579. JW, Ulfman LH, Leenen LP, Pickkers P, Koenderman L. A
209. Oliver-Bell J, Butcher JP, Malcolm J, MacLeod MK, Adra- subset of neutrophils in human systemic inflammation
dos Planell A, Campbell L, Nibbs RJ, Garside P, McInnes inhibits T cell responses through Mac-1. J Clin Invest 2012:
IB, Culshaw S. Periodontitis in the absence of B cells and 122: 327–336.
specific anti-bacterial antibody. Mol Oral Microbiol 2015: 224. Popadiak K, Potempa J, Riesbeck K, Blom AM. Biphasic
30: 160–169. effect of gingipains from Porphyromonas gingivalis on the
210. Ortega-Gomez A, Perretti M, Soehnlein O. Resolution of human complement system. J Immunol 2007: 178: 7242–
inflammation: an integrated view. EMBO Mol Med 2013: 5: 7250.
661–674. 225. Potempa M, Potempa J, Kantyka T, Nguyen KA, Wawr-
211. Page RC, Lantz MS, Darveau R, Jeffcoat M, Mancl L, Hous- zonek K, Manandhar SP, Popadiak K, Riesbeck K, Eick S,
ton L, Braham P, Persson GR. Immunization of Macaca Blom AM. Interpain A, a cysteine proteinase from Prevo-
fascicularis against experimental periodontitis using a vac- tella intermedia, inhibits complement by degrading com-
cine containing cysteine proteases purified from Porphy- plement factor C3. PLoS Pathog 2009: 5: e1000316.
romonas gingivalis. Oral Microbiol Immunol 2007: 22: 226. Rajapakse PS, O’Brien-Simpson NM, Slakeski N, Hoffmann
162–168. B, Reynolds EC. Immunization with the RgpA-Kgp pro-
212. Page RC, Offenbacher S, Schroeder HE, Seymour GJ, Korn- teinase-adhesin complexes of Porphyromonas gingivalis
man KS. Advances in the pathogenesis of periodontitis: protects against periodontal bone loss in the rat periodon-
summary of developments, clinical implications and titis model. Infect Immun 2002: 70: 2480–2486.
future directions. Periodontol 2000 1997: 14: 216–248. 227. Ravichandran KS, Lorenz U. Engulfment of apoptotic cells:
213. Page RC, Schroeder HE. Pathogenesis of inflammatory signals for a good meal. Nat Rev Immunol 2007: 7: 964–974.
periodontal disease. A summary of current work. Lab 228. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Comple-
Invest 1976: 34: 235–249. ment: a key system for immune surveillance and home-
214. Page RC, Schroeder HE. Current status of the host ostasis. Nat Immunol 2010: 11: 785–797.
response in chronic marginal periodontitis. J Periodontol 229. Rifas L, Weitzmann MN. A novel T cell cytokine, secreted
1981: 52: 477–491. osteoclastogenic factor of activated T cells, induces osteo-
215. Papadopoulos G, Weinberg EO, Massari P, Gibson FC III, clast formation in a RANKL-independent manner. Arthritis
Wetzler LM, Morgan EF, Genco CA. Macrophage-specific Rheum 2009: 60: 3324–3335.
TLR2 signaling mediates pathogen-induced TNF-depen- 230. Roark CL, Simonian PL, Fontenot AP, Born WK, O’Brien
dent inflammatory oral bone loss. J Immunol 2013: 190: RL. Gammadelta T cells: an important source of IL-17.
1148–1157. Curr Opin Immunol 2008: 20: 353–357.

148
Revisiting the Page & Schroeder model

231. Roberts HM, Ling MR, Insall R, Kalna G, Spengler J, Grant 246. Schenkein HA, Genco RJ. Gingival fluid and serum in peri-
MM, Chapple IL. Impaired neutrophil directional chemo- odontal diseases. II. Evidence for cleavage of complement
tactic accuracy in chronic periodontitis patients. J Clin components C3, C3 proactivator (factor B) and C4 in gingi-
Periodontol 2015: 42: 1–11. val fluid. J Periodontol 1977: 48: 778–784.
232. Roes J, Rajewsky K. Immunoglobulin D (IgD)-deficient 247. Schenkein HA, Genco RJ. Complement cleavage products
mice reveal an auxiliary receptor function for IgD in anti- in inflammatory exudates from patients with periodontal
gen-mediated recruitment of B cells. J Exp Med 1993: 177: diseases. J Immunol 1978: 120: 1796.
45–55. 248. Schif-Zuck S, Gross N, Assi S, Rostoker R, Serhan CN, Ariel
233. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, A. Saturated-efferocytosis generates pro-resolving CD11b
and function. Immunity 2015: 42: 607–612. low macrophages: modulation by resolvins and glucocorti-
234. Rothstein TL, Griffin DO, Holodick NE, Quach TD, Kaku coids. Eur J Immunol 2011: 41: 366–379.
H. Human B-1 cells take the stage. Ann N Y Acad Sci 2013: 249. Schmidt S, Moser M, Sperandio M. The molecular basis of
1285: 97–114. leukocyte recruitment and its deficiencies. Mol Immunol
235. Rotondo R, Bertolotto M, Barisione G, Astigiano S, Man- 2012: 55: 49–58.
druzzato S, Ottonello L, Dallegri F, Bronte V, Ferrini S, Bar- 250. Seguier S, Godeau G, Brousse N. Collagen fibers and
bieri O. Exocytosis of azurophil and arginase 1-containing inflammatory cells in healthy and diseased human gingi-
granules by activated polymorphonuclear neutrophils is val tissues: a comparative and quantitative study by
required to inhibit T lymphocyte proliferation. J Leukoc immunohistochemistry and automated image analysis. J
Biol 2011: 89: 721–727. Periodontol 2000: 71: 1079–1085.
236. Ryder MI. Comparison of neutrophil functions in aggres- 251. Serhan CN, Chiang N, Van Dyke TE. Resolving inflamma-
sive and chronic periodontitis. Periodontol 2000 2010: 53: tion: dual anti-inflammatory and pro-resolution lipid
124–137. mediators. Nat Rev Immunol 2008: 8: 349–361.
237. Sadallah S, Eken C, Schifferli JA. Ectosomes as 252. Shaker OG, Ghallab NA. IL-17 and IL-11 GCF levels in
immunomodulators. Semin Immunopathol 2011: 33: 487– aggressive and chronic periodontitis patients: relation to
495. PCR bacterial detection. Mediators Inflamm 2012: 2012:
238. Sapey E, Greenwood H, Walton G, Mann E, Love A, Aaron- 174764.
son N, Insall RH, Stockley RA, Lord JM. Phosphoinositide 253. Shin J, Hosur KB, Pyaram K, Jotwani R, Liang S, Chavakis
3-kinase inhibition restores neutrophil accuracy in the T, Hajishengallis G. Expression and function of the home-
elderly: toward targeted treatments for immunosenes- ostatic molecule Del-1 in endothelial cells and the peri-
cence. Blood 2014: 123: 239–248. odontal tissue. Clin Dev Immunol 2013: 2013: 617809.
239. Scapini P, Bazzoni F, Cassatella MA. Regulation of B-cell- 254. Shin J, Maekawa T, Abe T, Hajishengallis E, Hosur K,
activating factor (BAFF)/B lymphocyte stimulator (BLyS) Pyaram K, Mitroulis I, Chavakis T, Hajishengallis G. DEL-1
expression in human neutrophils. Immunol Lett 2008: 116: restrains osteoclastogenesis and inhibits inflammatory
1–6. bone loss in nonhuman primates. Sci Transl Med 2015: 7:
240. Scapini P, Carletto A, Nardelli B, Calzetti F, Roschke V, 307ra155.
Merigo F, Tamassia N, Pieropan S, Biasi D, Sbarbati A, Soz- 255. Sica A, Mantovani A. Macrophage plasticity and polariza-
zani S, Bambara L, Cassatella MA. Proinflammatory medi- tion: in vivo veritas. J Clin Invest 2012: 122: 787–795.
ators elicit secretion of the intracellular B-lymphocyte 256. Silva N, Abusleme L, Bravo D, Dutzan N, Garcia-Sesnich J,
stimulator pool (BLyS) that is stored in activated neu- Vernal R, Hernandez M, Gamonal J. Host response mecha-
trophils: implications for inflammatory diseases. Blood nisms in periodontal diseases. J Appl Oral Sci 2015: 23:
2005: 105: 830–837. 329–355.
241. Scapini P, Cassatella MA. Social networking of human 257. Sima C, Glogauer M. Neutrophil dysfunction and host sus-
neutrophils within the immune system. Blood 2014: 124: ceptibility to periodontal inflammation: current state of
710–719. knowledge. Curr Oral Health Rep 2014: 1: 95–103.
242. Scapini P, Hu Y, Chu CL, Migone TS, Defranco AL, Cas- 258. Sollecito TP, Sullivan KE, Pinto A, Stewart J, Korostoff J.
satella MA, Lowell CA. Myeloid cells, BAFF, and IFN- Systemic conditions associated with periodontitis in child-
gamma establish an inflammatory loop that exacerbates hood and adolescence. A review of diagnostic possibilities.
autoimmunity in Lyn-deficient mice. J Exp Med 2010: 207: Med Oral Patol Oral Cir Bucal 2005: 10: 142–150.
1757–1773. 259. Stables MJ, Shah S, Camon EB, Lovering RC, Newson J,
243. Schenkein HA. Host responses in maintaining periodontal Bystrom J, Farrow S, Gilroy DW. Transcriptomic analyses
health and determining periodontal disease. Periodontol of murine resolution-phase macrophages. Blood 2011:
2000 2006: 40: 77–93. 118: e192–e208.
244. Schenkein HA, Fletcher HM, Bodnar M, Macrina FL. 260. Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley
Increased opsonization of a prtH-defective mutant of Por- K. Phagocytosis of apoptotic neutrophils regulates granu-
phyromonas gingivalis W83 is caused by reduced degrada- lopoiesis via IL-23 and IL-17. Immunity 2005: 22: 285–
tion of complement-derived opsonins. J Immunol 1995: 294.
154: 5331–5337. 261. Steinmetz O, Hoch S, Avniel-Polak S, Gavish K, Eli-
245. Schenkein HA, Genco RJ. Gingival fluid and serum in peri- Berchoer L, Wilensky A, Nussbaum G. CX3CR1hi mono-
odontal diseases. I. Quantitative study of immunoglobu- cytes support bacterial survival and experimental infec-
lins, complement components, and other plasma tion-driven bone resorption. J Infect Dis 2016: 213: 1505–
proteins. J Periodontol 1977: 48: 772–777. 1515.

149
Hajishengallis & Korostoff

262. Stout RD, Suttles J. Immunosenescence and macrophage 280. Tsuda Y, Takahashi H, Kobayashi M, Hanafusa T, Herndon
functional plasticity: dysregulation of macrophage func- DN, Suzuki F. Three different neutrophil subsets exhibited
tion by age-associated microenvironmental changes. in mice with different susceptibilities to infection by
Immunol Rev 2005: 205: 60–71. methicillin-resistant Staphylococcus aureus. Immunity
263. Stout RD, Watkins SK, Suttles J. Functional plasticity of 2004: 21: 215–226.
macrophages: in situ reprogramming of tumor-associated 281. Van Dyke TE. Proresolving lipid mediators: potential for
macrophages. J Leukoc Biol 2009: 86: 1105–1109. prevention and treatment of periodontitis. J Clin Peri-
264. Streilein JW. Ocular immune privilege: therapeutic oppor- odontol 2011: 38(Suppl. 11): 119–125.
tunities from an experiment of nature. Nat Rev Immunol 282. Vernal R, Dutzan N, Chaparro A, Puente J, Antonieta
2003: 3: 879–889. Valenzuela M, Gamonal J. Levels of interleukin-17 in gingi-
265. Sutton CE, Mielke LA, Mills KH. IL-17-producing gam- val crevicular fluid and in supernatants of cellular cultures
madelta T cells and innate lymphoid cells. Eur J Immunol of gingival tissue from patients with chronic periodontitis.
2012: 42: 2221–2231. J Clin Periodontol 2005: 32: 383–389.
266. Tabas I. Macrophage death and defective inflammation 283. Vestweber D. How leukocytes cross the vascular endothe-
resolution in atherosclerosis. Nat Rev Immunol 2010: 10: lium. Nat Rev Immunol 2015: 15: 692–704.
36–46. 284. von Vietinghoff S, Ley K. Homeostatic regulation of blood
267. Tabas I, Glass CK. Anti-inflammatory therapy in chronic neutrophil counts. J Immunol 2008: 181: 5183–5188.
disease: challenges and opportunities. Science 2013: 339: 285. Waldrop TC, Anderson DC, Hallmon WW, Schmalstieg FC,
166–172. Jacobs RL. Periodontal manifestations of the heritable
268. Takahashi K, Azuma T, Motohira H, Kinane DF, Kitetsu S. Mac-1, LFA-1, deficiency syndrome. Clinical, histopatho-
The potential role of interleukin-17 in the immunopathol- logic and molecular characteristics. J Periodontol 1987: 58:
ogy of periodontal disease. J Clin Periodontol 2005: 32: 400–416.
369–374. 286. Wang J, Shiratori I, Uehori J, Ikawa M, Arase H. Neutrophil
269. Tamassia N, Zimmermann M, Castellucci M, Ostuni R, infiltration during inflammation is regulated by PILRalpha
Bruderek K, Schilling B, Brandau S, Bazzoni F, Natoli G, via modulation of integrin activation. Nat Immunol 2013:
Cassatella MA. Cutting edge: an inactive chromatin config- 14: 34–40.
uration at the IL-10 locus in human neutrophils. J Immu- 287. Wang M, Krauss JL, Domon H, Hosur KB, Liang S, Magotti
nol 2013: 190: 1921–1925. P, Triantafilou M, Triantafilou K, Lambris JD, Hajishengal-
270. Tan Z, Jiang R, Wang X, Wang Y, Lu L, Liu Q, Zheng SG, lis G. Microbial hijacking of complement-toll-like receptor
Sun B, Ryffel B. RORct+IL-17+ neutrophils play a critical crosstalk. Sci Signal 2010: 3: ra11.
role in hepatic ischemia-reperfusion injury. J Mol Cell Biol 288. Weaver CT, Elson CO, Fouser LA, Kolls JK. The Th17 path-
2013: 5: 143–146. way and inflammatory diseases of the intestines, lungs,
271. Taubman MA, Valverde P, Han X, Kawai T. Immune and skin. Annu Rev Pathol 2013: 8: 477–512.
response: the key to bone resorption in periodontal dis- 289. Whitney C, Ant J, Moncla B, Johnson B, Page RC, Engel D.
ease. J Periodontol 2005: 76: 2033–2041. Serum immunoglobulin G antibody to Porphyromonas
272. Teng YT, Nguyen H, Gao X, Kong YY, Gorczynski RM, gingivalis in rapidly progressive periodontitis: titer, avidity,
Singh B, Ellen RP, Penninger JM. Functional human T-cell and subclass distribution. Infect Immun 1992: 60: 2194–
immunity and osteoprotegerin ligand control alveolar 2200.
bone destruction in periodontal infection. J Clin Invest 290. Wilensky A, Chaushu S, Shapira L. The role of natural
2000: 106: R59–R67. killer cells in periodontitis. Periodontol 2000 2015: 69:
273. Teng YTA. Protective and destructive immunity in the 128–141.
periodontium: part 2–T-cell-mediated immunity in the 291. Wilensky A, Segev H, Mizraji G, Shaul Y, Capucha T, Shac-
periodontium. J Dent Res 2006: 85: 209–219. ham M, Hovav AH. Dendritic cells and their role in peri-
274. Thorbert-Mros S, Larsson L, Berglundh T. Cellular compo- odontal disease. Oral Dis 2014: 20: 119–126.
sition of long-standing gingivitis and periodontitis lesions. 292. Xiao W, Dong G, Pacios S, Alnammary M, Barger LA, Wang
J Periodontal Res 2015: 50: 535–543. Y, Wu Y, Graves DT. FOXO1 deletion reduces dendritic cell
275. Tonetti MS, Chapple IL. Biological approaches to the function and enhances susceptibility to periodontitis. Am
development of novel periodontal therapies–consensus of J Pathol 2015: 185: 1085–1093.
the Seventh European Workshop on Periodontology. J Clin 293. Yamashita K, Eastcott JW, Taubman MA, Smith DJ, Cox
Periodontol 2011: 38(Suppl. 11): 114–118. DS. Effect of adoptive transfer of cloned Actinobacillus
276. Tonetti MS, Imboden MA, Lang NP. Neutrophil migration actinomycetemcomitans-specific T helper cells on peri-
into the gingival sulcus is associated with transepithelial odontal disease. Infect Immun 1991: 59: 1529–1534.
gradients of interleukin-8 and ICAM-1. J Periodontol 1998: 294. Ye P, Rodriguez FH, Kanaly S, Stocking KL, Schurr J, Sch-
69: 1139–1147. warzenberger P, Oliver P, Huang W, Zhang P, Zhang J,
277. Toto PD, Lin L, Gargiulo A. Identification of C3a, IgG, IgM Shellito JE, Bagby GJ, Nelson S, Charrier K, Peschon JJ,
in inflamed human gingiva. J Dent Res 1978: 57: 696. Kolls JK. Requirement of interleukin 17 receptor signaling
278. Toto PD, Lin LM, Gargiulo AW. Immunoglobulins and for lung CXC chemokine and granulocyte colony-stimulat-
complement in human periodontitis. J Periodontol 1978: ing factor expression, neutrophil recruitment, and host
49: 631–634. defense. J Exp Med 2001: 194: 519–527.
279. Tseng CW, Liu GY. Expanding roles of neutrophils in aging 295. Younes R, Ghorra C, Khalife S, Igondjo-Tchen-Changotade
hosts. Curr Opin Immunol 2014: 29: 43–48. S, Yousfi M, Willig C, Senni K, Godeau G, Naaman N.

150
Revisiting the Page & Schroeder model

Pertinent cell population to characterize periodontal dis- contributes to periodontal tissue homeostasis. Cell Micro-
ease. Tissue Cell 2009: 41: 141–150. biol 2013: 15: 1419–1426.
296. Yu T, Zhao L, Huang X, Ma C, Wang Y, Zhang J, Xuan D. 300. Zhan Y, Zhang R, Lv H, Song X, Xu X, Chai L, Lv W, Shang
Enhanced activity of the macrophage M1/M2 phenotypes Z, Jiang Y, Zhang R. Prioritization of candidate genes for
and phenotypic switch to M1 in periodontal infection. J periodontitis using multiple computational tools. J Peri-
Periodontol 2016: 87: 1092–1102. odontol 2014: 85: 1059–1069.
297. Zappa U, Reinking-Zappa M, Graf H, Espeland M. Cell 301. Zhao L, Zhou Y, Xu Y, Sun Y, Li L, Chen W. Effect of non-
populations and episodic periodontal attachment loss in surgical periodontal therapy on the levels of Th17/Th1/
humans. J Clin Periodontol 1991: 18: 508–515. Th2 cytokines and their transcription factors in Chinese
298. Zenobia C, Hajishengallis G. Basic biology and role of chronic periodontitis patients. J Clin Periodontol 2011: 38:
interleukin-17 in immunity and inflammation. Periodontol 509–516.
2000 2015: 69: 142–159. 302. Zhu M, Belkina AC, DeFuria J, Carr JD, Van Dyke TE,
299. Zenobia C, Luo XL, Hashim A, Abe T, Jin L, Chang Y, Jin Gyurko R, Nikolajczyk BS. B cells promote obesity-asso-
ZC, Sun JX, Hajishengallis G, Curtis MA, Darveau RP. Com- ciated periodontitis and oral pathogen-associated inflam-
mensal bacteria-dependent select expression of CXCL2 mation. J Leukoc Biol 2014: 96: 349–357.

151

You might also like