You are on page 1of 12

Periodontology 2000, Vol. 0, 2017, 1–12 © 2017 John Wiley & Sons A/S.

ley & Sons A/S. Published by John Wiley & Sons Ltd
Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Bacterial modulators of bone


remodeling in the periodontal
pocket
BRIAN HENDERSON & FRANK KAISER

Bone is one of the most fascinating of natural mechanical stress signals) to ensure that the constant
composite materials, consisting of a largely collage- fatigue fracturing of bone, which occurs as a result of
nous organic matrix in which hydroxyapatite crystals the stresses and strains of living on a 1G planet (or
are embedded, giving bone its rigidity and strength, through other mechanical forces, such as mastica-
allowing it to be sculpted into its complex shapes and tion), is constrained and that bone maintains its opti-
enabling it to be superbly sensitive to stress (63). The mal mechanical strength (37). Unfortunately, this
mechanical properties of bone are, in turn, controlled natural process often goes wrong and creates local
by its major cellular elements. The major cell popula- and systemic pathology in the form of diseases, such
tion in bone is one that is only now coming to the as osteoporosis, rheumatoid arthritis and, of course,
attention of the biomedical community. The osteo- periodontitis. A paradigm shift in thinking about the
cyte comprises some 95% of the cells of bone and is, skeleton, and the cellular basis of its turnover in
in appearance, a dendritic-like cell whose processes health and disease, has given rise to a new discipline
fill the canaliculi of bone. Osteocytes are responsible over the past decade. This is the discipline of
for bone being able to recognize, and respond to, osteoimmunology, which is based on growing evi-
mechanical stress and strain (73). It is also being rec- dence for a role of both innate and adaptive immune
ognized that osteocytes are key bone-regulatory cells mechanisms in controlling normal and pathological
as a result of: (i) their interactions with other bone skeletal turnover and the cells responsible for bone
cells, principally osteoclasts; and (ii) their synthesis remodeling. Osteoimmunological mechanisms have
and secretion of key modulatory proteins (cytokine/ now been suggested to underpin the bone pathology
endocrine-like molecules) that influence bone in osteoporosis (17), rheumatoid arthritis (70) and
remodeling and also phosphate kinetics in the periodontitis (29). This new paradigm, in the context
circulation (68). of periodontitis, also has to encompass the role of
The osteocyte originates from the osteoblast lin- bacteria in the bone loss of this disease. This is the
eages of bone, with the osteoblast being the synthetic point where there starts to be a ‘blurring’ of the
cell of bone responsible for generating the bone pathogenic mechanisms in skeletal diseases, with the
matrix (65). The osteoblast is a mesenchymal cell, as recent finding that the major periodontopathogenic
must be the osteocyte. The third major bone-modu- bacterium, Porphyromonas gingivalis, may play a
lating cell population – the osteoclast – arises from major role in the pathogenesis of rheumatoid arthritis
the myeloid cell lineage and is one of only two types (14). This novel finding will be returned to at a later
of multinucleate cell in mammals. The major function stage in this review.
of the osteoclast is to remove bone matrix and its
functions intermesh with those of the osteoblast and
the osteocyte (51). Bacteria and periodontitis
Bone remodeling is the normal result of the inter-
meshing of these three cell populations with a range With our new understanding of how enormously col-
of cell-surface and secreted mediators (plus onized are the epithelial surfaces of the skin and the

1
Henderson & Kaiser

mucosal membranes of the oral cavity, gut and disease is, it is believed, caused by a response to
urogenital tract, comes a newly developing paradigm members of the oral microbiota, it might be expected
of the role played by the bacterial microbiota in both that bacteria would have a key role to play in driving
health and disease. This developing paradigm has lar- bone destruction. However, it is not known if these
gely focused on the gut, with the evidence suggesting bacteria cause bone breakdown because they directly
that the colonic microbiota plays a major role in con- or indirectly induce inflammation, or because they
trolling the energetics of the mammal (79) and from release factors that can directly interact with bone or,
this context the colonic microbiota is considered a indeed, if they actually are able to interact with bone
bonus for mammals. Indeed, in spite of the huge cells. The idea that bacteria can induce bone destruc-
numbers of diverse bacteria in the colon, the amount tion because they are able to induce gingival inflam-
of tissue pathology thought to be caused by these mation has to be looked at in more detail. Clearly,
bacteria is actually rather small. This contrasts with gingivitis does not induce alveolar bone resorption, so
the much larger association between tissue pathology we may need to look more deeply at the relationship
and bacteria in bacterial vaginosis (54) and in peri- between local inflammation and bone loss. How
odontitis. Indeed, a recent National Health and Nutri- much more deeply can be seen from experimental
tion Examination Study (NHANES) estimated that arthritis studies in which it is well known that it is pos-
47% of the population of the USA suffers from peri- sible to inhibit inflammation of the synovial lining of
odontitis (12). This is a truly staggering amount of the joint (a tissue that can be considered equivalent to
pathology, particularly as periodontitis is a chronic the gingiva) without inhibiting bone resorption and,
disease state. more recently, it has been shown that it is possible to
This raises the obvious question – why do such inhibit bone resorption in the joint without inhibiting
large proportions of the world’s population respond synovitis. This has been shown with osteoprotegerin,
to their oral bacteria and develop a chronic inflamma- the natural inhibitor of the cytokine, RANKL, which is
tory disease of the gums that also causes destruction the major inducer of osteoclast formation (35), and by
of the periodontal ligament and alveolar bone? The use of the Mycobacterium tuberculosis cell stress pro-
obvious answer is that the periodontium is the major tein, chaperonin 60.1 (86). So, the precise relationship
weak spot in the protective mucosa of the body, as a between local inflammation and bone destruction is
result of the interruption of this mucosal layer by the still not mapped. The next three sections of this
teeth. In other tissues, the argument would continue, review cover possible mechanisms linking periodontal
the surface mucosa/epithelium, on which the micro- bacteria with alveolar bone loss.
biota is being supported, prevents the ingress of bac-
teria into tissues and the consequent induction of
Indirect actions of bacteria inducing
inflammation. A counter to this argument is the
bone destruction
recent report that in normal human skin, members of
the skin microbiota can breach the basement mem- What possible mechanisms can link oral bacteria with
brane of the skin and are found within the dermis alveolar bone resorption? The role of immune cells in
and even within the adipose tissues of the subdermal controlling bone remodeling has already been
layer (52). If bacteria can enter the submucosal space alluded to, and it is now clear that most cellular com-
in all mucosal sites, and interact with the submucosal ponents of the innate and, particularly, the adaptive,
cells of the host, it counters the specific argument for immune response (e.g. T-lymphocytes) (55), have
the ‘weakness’ of the gingival mucosal barrier (caused marked influence on bone cells and can produce
by protrusion of the teeth) and also changes the pos- inappropriate bone remodeling. As such, the genera-
sible hypotheses that can be forwarded to explain tion of chronic inflammation in the gingiva, in
how bone loss occurs in periodontitis. response to members of the subgingival microbiota,
may attract and activate appropriate T-cell popula-
tions (particularly T-helper 1 and T-helper 17 cells)
Mechanisms which explain and other leukocytes that are able to drive local alveo-
bacterially-induced bone lar bone destruction. The role of the T-helper 17 lym-
breakdown in periodontitis phocyte (36) in periodontitis is probably to link up
bacterial signaling with bone resorption, and it is
It is surprising that, even at the time of writing, it is interesting that this lymphocyte subpopulation is also
still not completely clear what mechanism, or mecha- implicated in the destruction of bone observed in
nisms, drive the bone loss in periodontitis. As this rheumatoid arthritis (40). It would be assumed that

2
Bacterial modulators of bone remodeling in periodontal pocket

Fig. 1. Cytokines activating osteoclastogenesis in rheuma- induction of T-helper 17 cells. T-helper 17 cells secrete
toid arthritis. Tumor necrosis factor, interleukin-1, inter- interleukin-17, but a main source of synovial interleukin-17
leukin-6 and interleukin-17 up-regulate expression of is probably mast cells. Interleukin-17 induces the expres-
RANKL in osteoblasts and synovial fibroblasts. RANKL sion of RANKL in osteoblasts and fibroblasts and enhances
mediates differentiation, survival and activation of osteo- secretion of proinflammatory cytokines by macrophages.
clasts. Tumor necrosis factor, produced by fibroblasts and Macrophage colony-stimulating factor and interleukin-34
macrophages, promotes differentiation and survival of promote differentiation and activation of osteoclasts;
osteoclasts. Interleukin-1 supports differentiation, survival interleukin-33 supports osteoclast differentiation. IL,
and activation of osteoclasts. Interleukin-6 and interleukin- interleukin; M-CSF, macrophage colony-stimulating factor;
17 promote osteoclastogenesis indirectly. Interleukin-6 is TGF-b, transforming growth factor-beta; Th, T-helper; TNF,
largely produced by fibroblasts and macrophages; it tumor necrosis factor. (From: Braun & Zwerina (5). With
enhances the expression of RANKL and contributes to the permission).

inflammation and associated tissue destruction are bone resorption comes from the study of rheumatoid
caused by the release, by the subgingival bacteria arthritis (5) (Fig. 1). As the signals coming from the
within the periodontal pocket, of one or more of the bacteria in the periodontal pocket, and causing alveo-
pathogen-associated molecular pattern molecules lar bone resorption, will be different from the induc-
that are the evolutionarily stable components of bac- ing signals in the autoimmune disease, rheumatoid
teria which are recognized by the growing number of arthritis, it is to be expected that these two diseases
host pattern-recognition receptors found on immune would exhibit different mechanisms of bone break-
cells (34) and on the epithelial cells supporting the down. However, as stated above, the T-helper 17
microbiotas (57). Binding of pathogen-associated lymphocyte subpopulation is implicated in both
molecular patterns to their pattern-recognition diseases (9), suggesting some commonality of bone
receptors, such as the toll-like receptors, results in the pathogenesis.
production of a range of proinflammatory cytokines,
such as the early response cytokines interleukin-1
Bacterially induced local autoimmunity
beta and tumor necrosis factor alpha, which are
and bone destruction
potent osteolytic agents and could drive bone resorp-
tion. Indeed, it was Dewhirst & Stashenko who The contention above would be different if there
revealed the osteolytic activity of interleukin-1 (11). was evidence that periodontitis is an autoimmune
Since this time, a growing number of nonspecific and disease. Currently, there is only minimal evidence
specific cytokines have been found which can stimu- for aggressive periodontitis showing some autoim-
late the formation/activation of osteoclasts and pro- mune signature (21). However, before discussing
mote bone resorption (Fig. 1). It should be noted that bacterial modulators of bone cells that could be
most of our information on the cytokine biology of directly responsible for aberrant bone remodeling

3
Henderson & Kaiser

in periodontitis, it is important to mention briefly Direct actions of periodontopathic


the emerging evidence for a role for P. gingivalis in bacteria on alveolar bone
autoimmunity and the possibility of its role in peri-
In this section, the role of molecules secreted by bac-
odontal autoimmunity. The amino acid arginine
teria, which can modulate bone-cell function and
can be converted within proteins to citrulline by
which may contribute to pathological bone remodel-
the enzymes known as peptidylarginine deiminases
(80). This protein decoration, known as protein ing in periodontitis, will be discussed. Until recently,
the general finding was that pathogen-associated
citrullination, changes the physicochemical proper-
molecular patterns and other molecules emanating
ties of proteins. Protein citrullination was first
from bacteria promoted bone breakdown generally
shown to enhance immune responsiveness to mye-
by a mechanism that induced the formation of osteo-
lin basic protein, suggesting a role for this protein
decoration in autoimmune encephalomyelitis (94). clasts. Less is known about bacterial molecules that
can inhibit osteoblast bone synthesis. However, in
However, it was quickly established that citrulli-
the past decade, a slow trickle of papers has appeared
nated residues in proteins are essential parts of the
in which a variety of bacterial molecules, including
antigenic determinants recognized by autoantibod-
homologues of well-known osteolytic bacterial patho-
ies present in the blood of patients with rheuma-
gen-associated molecular patterns, have been
toid arthritis (78). It was Travis & Potempa (45),
described to inhibit bone breakdown and osteoclast
who performed pioneering work on the virulence
formation. Thus, it would appear that bacteria can
factors of P. gingivalis known as gingipains, who
promote or inhibit bone resorption depending on the
discovered that this bacterium also produced a
molecules they express; however, it is not known if
peptidylarginine deiminase and suggested that it
this is part of an evolved control system employed by
was probably a virulence factor. However, it was
the rheumatologist, Gerald Weissmann who put bacteria, or whether it has any role in the induction
or pathogenesis of periodontitis.
both strands of this story together to suggest that it
The rest of this section will describe the bone-mod-
was the humoral immune response to the products
ulating properties of the best-known bacterial ‘oste-
of the peptidylarginine deiminase of P. gingivalis
olytic’ components, with an emphasis on those
that could have initiated rheumatoid arthritis (66).
emanating from periodontopathic bacteria.
Biochemical studies have shown that, among the
oral bacteria tested, only P. gingivalis can citrulli-
nate proteins/peptides, and that a combination of Lipopolysaccharides
arginine gingipains and peptidylarginine deiminase
As suggested above, gram-positive and gram-negative
results in the formation of citrullinated peptides
bacteria have a wide range of pathogen-associated
from human fibrinogen and a-enolase. These pep-
molecular patterns and other molecules that can
tides are then proposed to drive the autoimmune
influence bone remodeling. Unfortunately, there is
pathology of rheumatoid arthritis (81). Potentially
increasing evidence for the same apparent class of
the strongest evidence for this hypothesis comes
pathogen-associated molecular pattern having oppos-
from an experimental study showing that wild-type
ing actions on bone. This makes for significant confu-
P. gingivalis exacerbates the pathology of collagen-
sion in the literature and could either be caused by
induced arthritis in mice, while an isogenic mutant
experimental error or because the workers publishing
lacking the gene encoding the P. gingivalis peptidy-
the opposing data are actually not using the same
larginine deiminase did not induce any
molecules. This problem has been best explored using
exacerbation of this experimental autoimmune
the gram-negative outer amphiphilic molecule,
condition (43).
lipopolysaccharide. This forms the outer layer of the
These findings clearly raise the possibility that
outer membrane of gram-negative bacteria and the
autoimmunity to locally induced citrullinated pro-
molecule is a complex consisting of an inner and
teins could be a driving factor in periodontitis and
outer core region and an O-specific side chain. The
therefore the mechanisms of bone destruction in
biologically active component of the lipopolysaccha-
rheumatoid arthritis and periodontitis are likely to be
ride polymer is a small phosphoglycolipid, called lipid
similar. Two recent clinical studies have failed to sup-
A, which has a unique and highly polymorphic struc-
port this contention directly (38, 59) but further stud-
ture and it is in this ability of bacteria to generate vari-
ies are needed in case the autoantigenic proteins
ations on the theme of lipid A that leads to different
driving periodontitis differ from those known to
biological actions of this key pathogen-associated
induce rheumatoid arthritis.

4
Bacterial modulators of bone remodeling in periodontal pocket

molecular pattern (20). Briefly, lipid A is a disaccha- lipopolysaccharide-unresponsive strains of mice (e.g.
ride, which can vary in the composition of its saccha- C3H/HeJ) exist (60). Lipopolysaccharide-induced
ride components, and these sugars are bound to four bone resorption has been shown to be induced both
(R)-3-hydroxyl fatty acids at positions 2,3 and 20 ,30 with in in vitro and in vivo systems. In in vivo experiments,
phosphate groups at positions 1 and 40 . Esterification in which lipopolysaccharide is injected into the gin-
of these fatty acids can occur at the 20 and 30 positions, giva, it has been shown that toll-like receptor 4-defi-
and in this way the number of fatty acyl chains can cient mice exhibit much less bone resorption than do
increase to six (and sometimes even to seven). All lipopolysaccharide-responsive mice, and this is asso-
these modifications of lipid A alter its biological activ- ciated with lower expression of the key osteoclast
ity and its effects on bone cells. A rough rule of thumb inducer/activator, RANKL (50).
is that the more acyl chains a lipopolysaccharide Curiously, in spite of the wealth of data showing
molecule has, the more biologically active it is. Thus, that lipopolysaccharide promotes bone resorption by
seven- and six-chain lipid A molecules are extremely inducing/activating osteoclasts, the story is actually
potent molecules; potency decreases with five-chain more complex, with a growing number of reports
molecules; and with four acyl chains the lipopolysac- that, in given circumstances, lipopolysaccharide can
charides can be almost inactive or even act as actually inhibit osteoclast formation and can have
lipopolysaccharide antagonists (20). bifunctional properties. This was first reported by
The first evidence for lipopolysaccharide being able Zou & Bar-Shavit (95), who used bone marrow-
to promote bone resorption appeared in 1977, when derived osteoclast precursors purified of other cell
it was shown that lipopolysaccharide induced the types (e.g. osteoblasts and stromal cells). Lipopolysac-
formation of multinucleate osteoclasts, suggesting charide not only failed to promote osteoclast differen-
its bone-resorbing mechanism was different to tiation, but it also blocked the osteoclastogenesis
osteoclastic resorption induced by parathyroid hor- promoted by RANKL. However, with RANKL pre-
mone (67). This stimulated research into the oste- treated cells, lipopolysaccharide did indeed promote
olytic effects of lipopolysaccharide molecules from osteoclast formation. The induction of osteoclasts
periodontopathogens (Table 1). It was established, in appeared not to be caused by RANKL, but by the
the 1990s, that lipopolysaccharide signals to cells activity of tumor necrosis factor alpha (75). A key
mainly through binding to toll-like receptor 4 and transcription factor in osteoclast differentiation is
such knowledge was aided by the fact that nuclear factor of activated T-cells 1 (92). This is now

Table 1. Lipopolysaccharides from periodontopathogens and their effect on bone and osteoclast formation/activation

Organism Bone resorption stimulated in Influence on osteoclasts References


culture

Aggregatibacter Yes (Not detected) Kiley & Holt (30)


actinomycetemcomitans

Aggregatibacter (Not detected) Stimulates osteoclast Ueda et al. (77)


actinomycetemcomitans formation

Aggregatibacter Stimulates bone loss in vivo (Not detected) Rogers et al. (64)
actinomycetemcomitans

Aggregatibacter Lipopolysaccharide-induced bone (Not detected) Madeira et al. (42)


actinomycetemcomitans resorption is Myd88-dependent

Capnocytophaga ochracea Yes (Not detected) Iino & Hopps (25)

Eikenalla corrodens Yes (Not detected) Progulske et al. (58)

Fusobacterium nucleatum Yes (Not detected) Sveen & Skaug (74)

Porphyromonas gingivalis (Not detected) Stimulates osteoclastic pit Sismey-Durrrant & Hopps (72)
formation in dentine slices
Treponema denticola (Not detected) Stimulates osteoclast Choi et al. (7)
formation
Veillonella spp. Yes (Not detected) Sveen & Skaug (74)

5
Henderson & Kaiser

seen to be a pivotal factor in this bifunctional mechanisms for this extremely heterogeneous
response of osteoclasts to lipopolysaccharide (41). In amphiphilic molecule to promote bone remodeling.
osteoclast precursors that have not been exposed to Given that the periodontal pocket may contain
RANKL, lipopolysaccharide inhibits nuclear factor of lipopolysaccharide species from several hundred
activated T-cells 1. In contrast, if the same cells have bacteria means that it is difficult to predict what the
been stimulated by RANKL, then lipopolysaccharide overall influence of the lipopolysaccharide molecules
promotes the expression of nuclear factor of activated in their entirety will be. For all we know, there may
T-cells 1. This reveals the complexity of the cell net- be synergy or antagonism between all of these differ-
works involved in osteoclast differentiation. ent lipopolysaccharides, which could lead to very dif-
Not much is known about the periodontopathic ferent biological outcomes for the patients involved.
lipopolysaccharides that promote bone resorption Clearly, we need to know much more about the
through toll-like receptor 4 and osteoclastogenesis. lipopolysaccharide molecule as it relates to bone
However, there is growing knowledge about the remodeling.
unusual properties of the lipopolysaccharide of the
major periodontopathogen, P. gingivalis, whose Wall teichoic acids and lipoteichoic acids
lipopolysaccharide interacts with toll-like receptor 2
(2) as well as toll-like receptor 4. This bacterium Unlike gram-negative bacteria, gram-positive organ-
can make penta-acylated lipopolysaccharide which, isms lack an outer membrane, with its key
surprisingly, is 10- to 100-times less active than an lipopolysaccharide component, and a periplasm. In
almost identical penta-acylated lipopolysaccharide contrast, the layers of peptidoglycan enveloping the
from Bacteroides spp. (4). There is also evidence cytoplasm are very thick and contain anionic gly-
that the penta- and tetra-acylated lipopolysaccha- copolymers that can exit onto the cell surface. One of
rides made by P. gingivalis can differentially target the best studied of these are the teichoic acids – phos-
toll-like receptor 4 and downstream signaling path- phate-rich polymers that come in two varieties: the
ways, suggesting that if this bacterium can modu- lipoteichoic acids, which anchor to the plasma mem-
late its outer lipopolysaccharide, it could markedly brane and extend into the peptidoglycan layers; and
alter its virulence potential, including its effects on the wall teichoic acids, which are bound to the pepti-
bone (22). This may explain, in part, the differential doglycan and extend onto the bacterial surface (6).
effects of P. gingivalis lipopolysaccharide on osteo- These molecules are relatively well-studied pathogen-
clast formation (69). associated molecular patterns with a perceived role in
Thus far, the discussion has been based on the abil- host immune and inflammatory responses to gram-
ity of lipopolysaccharide to induce osteoclast forma- positive bacteria (82). While being important mole-
tion and activate mature osteoclasts. Does cules and, like lipopolysaccharide, with significant
lipopolysaccharide also inhibit the bone-forming structural diversity, the wall teichoic acids and lipote-
actions of osteoblasts or the bone-controlling activity ichoic acids have received relatively scant attention,
of the major osteocyte population? It was reported compared with lipopolysaccharide, in terms of their
that lipopolysaccharide from P. gingivalis was cap- influence on bone remodeling.
able of inhibiting the differentiation of osteoblasts in Given the similarities in the general response of
culture, as assessed by measuring markers of cellular cells to lipopolysaccharide and teichoic acids, the
differentiation, such as alkaline phosphatase, osteo- expectation is that the wall teichoic acids and lipotei-
calcin and osteopontin (26). The mechanism of this choic acids would exert osteolytic activity, presum-
effect is activation of Notch signaling (89). In in vivo ably by promoting osteoclastogenesis. There are only
studies, the inhibition of osteogenesis by lipopolysac- two reports that teichoic acids could promote bone
charide has been shown to be linked to induction of breakdown, the first dating back to the mid-1970s
tumor necrosis factor alpha synthesis (76). Finally, (19). The teichoic acids of the gram-positive organism
there is evidence that lipopolysaccharide is able to Streptococcus mutans (1) have been reported to pro-
inhibit the differentiation of osteoblasts into osteo- mote bone breakdown in vivo. The most important
cytes (3). It will be interesting to find out more details gram-positive bacterium, in relation to bone pathol-
of the role of bacterial lipopolysaccharide on ‘osteo- ogy, is clearly Staphylococcus aureus (88). It would be
cytogenesis’. assumed that the teichoic acids of this organism had
This review of the literature reveals that the small osteolytic activity. However, the lipoteichoic acid
number of lipopolysaccharide species examined is from S. aureus turned out to be inhibitory to osteo-
capable of providing different cellular signaling clast formation, working via a toll-like receptor 2-

6
Bacterial modulators of bone remodeling in periodontal pocket

sensitive mechanism (90). This may be specific for was an unexpected finding of a potent osteolytic
this form of lipoteichoic acid, and clearly a more protein released by the major periodontopathogen,
detailed analysis of wall teichoic acids and lipotei- A. actinomycetemcomitans (33). Of interest, this pro-
choic acids from gram-positive periodontopathogens tein stimulated bone resorption in the C3H/HeJ
is required to give a clearer understanding of the role mouse strain, revealing that the mode of action of
of this gram-positive component in periodontal bone this chaperonin 60 protein was not through activat-
destruction. ing toll-like receptor 4 (33). These chaperonins are
examples of moonlighting proteins, as might be a
number of the bacterial osteolytic proteins. Of inter-
Lipoproteins and lipopeptides
est, and as will be discussed in the next section,
Bacterial lipoproteins and lipopeptides are another some chaperonin 60 proteins can inhibit bone
class of pathogen-associated molecular pattern gen- breakdown. Possibly the most potent osteolytic bac-
erated by all forms of bacteria (61) and for which terial component is the eponymous toxin from Pas-
some evidence exists for a role in bone breakdown teurella multocida (39). This toxin is responsible for
(48). The Omp19 lipoprotein of Brucella abortus was atrophic rhinitis in pigs, a disease in which the turbi-
shown to induce osteoclast formation (10). The direct nate bones are destroyed (15). The P. multocida
role of lipoprotein in S. aureus-induced bone loss toxin was shown to be a potent stimulator of calvar-
in vitro and in vivo has recently been shown using a ial bone resorption in vitro (13) and to promote
lipoprotein-deficient mutant, which failed, unlike the osteoclast formation when administered to rats (44),
wild-type organism, to stimulate bone breakdown suggesting a direct influence on osteoclastogenesis.
(32). An interesting bacterial lipopeptide with a grow- However, it turns out that this toxin, potent as it is,
ing list of biological actions is mycoplasmal macro- has an indirect effect on osteoclast differentiation,
phage activating lipopeptide-2, which interacts with which has been claimed to depend on the action of
toll-like receptor 2/6 (71, 84). When present at pico- osteoblasts (49) and, surprisingly, B-lymphocytes
molar concentrations, this 2-kDa peptide demon- (23).
strated the ability to induce murine calvarial bone One of the intriguing facets of bone-cell biology is
resorption, being one of the most active bone-resorb- the sensitivity of this complex, interrelated cellular
ing agonists recorded (56). system to molecules that would not be expected to
have any influence on bone remodeling. One such
Miscellaneous bacterial osteolytic example is protein A from S. aureus. For most biolo-
molecules gists, all they would know about this molecule is that
it is used for purifying IgA. However, in 2007, staphy-
In addition to the classes of osteolytic molecules lococcal protein A has been shown to have a wide
described above, there is a diverse literature on the range of additional functions, including acting as
ability of other bacterial molecules to either stimu- a B-cell superantigen able to subvert immune
late bone resorption or inhibit osteoblast-induced responses (96) and as a von Willebrand factor-binding
bone synthesis. For example, the capsular polysac- protein (18). Curiously, this protein has recently been
charide of Aggregatibacter actinomycetemcomitans suggested to be critical in mediating the bone loss
promotes osteoclast formation via a mechanism that occurs in S. aureus-induced osteomyelitis by
involving interleukin-1 production (53). The fimbriae inhibiting osteoblasts and stimulating osteoclastoge-
of P. gingivalis have been reported to stimulate bone nesis (8, 83). This is an unexpected effect, and may be
resorption in vitro (28) and have been shown to the sign of things to come as we explore, in more
stimulate osteoclastic bone resorption in a toll-like detail, the bacterial mediators of bone remodeling.
receptor 2-dependent manner (24). Bacterial porins
have also been shown to promote calvarial bone
resorption in vitro (46). The Treponema denticola Bacterial molecules that inhibit
cell-surface protein, Td92, induces osteoclast forma- bone remodeling and breakdown
tion and inhibits osteoprotegerin synthesis (31).
Another class of protein reported to be a potent Thus far, the discussion has been within the para-
stimulator of bone resorption are the molecular digm that bacteria and their skeleton-modulating
chaperones chaperonin 60 (33, 62) and chaperonin molecules act to stimulate bone destruction. How-
10 (47). That chaperonin 60, also known as heat ever, evidence is emerging for bacterial factors that
shock protein 60, could stimulate bone resorption can actually inhibit the cellular processes of bone

7
Henderson & Kaiser

Fig. 2. Bacteria–host and bacteria–bacteria signaling at and between these epithelial cells and the microbiota. In
mucosal surfaces. Complex reciprocal (and higher level) addition, the stability of the composition of the microbiota
dynamic signaling must occur on mucosal surfaces must be controlled by complex dynamic signaling between
between bacteria and associated mucosal epithelial cells these different bacteria.

remodeling. This has already been touched upon Likewise, it has been shown that several bacterial
with lipopolysaccharide and lipoteichoic acid, and proteins, from bacteria implicated in bone-remodel-
these molecules will not be dealt with again. ing pathology, can inhibit osteoclastogenesis. The
There are also a number of bacterial proteins first example of this was the hemoglobin receptor
emerging that inhibit the stimulation of bone remo- protein of P. gingivalis, which is a porphyrin-binding
deling. protein found on the cell surface and a major pro-
A number of low-molecular-mass compounds tein in the supernatant of this organism (27). This
emanating from bacteria, such as reveromycin A, protein was found to inhibit RANKL-induced osteo-
destruxins, mevastatin, FK506, cyclosporin A, prodi- clastogenesis when added within 24 h of stimulation
giosins, concanamycins and symbioimine, some of with RANKL, but not at later time points, suggesting
which are well-known immunosuppressants, have that it interfered with the early events in osteoclast
been shown to inhibit osteoclast differentiation by formation. This effect was associated with inhibition
acting at different stages in the process (87). This of the production of nuclear factor of activated T-
same research group has also reported that a macro- cells 1 (NFATc1) (16). The M. tuberculosis chaper-
lide compound, biselyngbyaside, from a marine onin 60.1 protein also inhibited osteoclastogenesis
cyanobacterium, inhibits osteoclastogenesis and can and this was associated with longer-term inhibition
induce mature osteoclasts to apoptose (91). Anti- of nuclear factor of activated T-cells 1. Indeed, this
coumarins from Streptomyces spp. can also inhibit protein could be added up to 4 days after the addi-
osteoclast differentiation by inhibiting the synthesis tion of RANKL and still be able to inhibit osteoclast
of nuclear factor of activated T-cells 1 (93). formation, suggesting a more comprehensive mode

8
Bacterial modulators of bone remodeling in periodontal pocket

of action than that of the hemoglobin receptor pro- populations in the periodontal pockets in patients
tein (86). Again, like the hemoglobin receptor pro- with various forms of periodontitis, linked with a
tein, inhibition of osteoclast formation, induced by complete genetic profile of the patients with peri-
M. tuberculosis chaperonin 60.1, was associated with odontitis (plus their clinical status). It is only with
inhibition of the transcription of nuclear factor of the foundation of these two interrelated complex
activated T-cells 1 (86). It is interesting that the M. data sets for a sufficient population of patients that
tuberculosis chaperonin 60.1 paralogue, chaperonin it will be possible to tease out the general relation-
60.2, is unable to inhibit or stimulate bone resorp- ships between colonization with periodontopathic
tion, despite the fact that both proteins share 63% bacteria, ‘genetic susceptibilities in periodontitis’
sequence identity (86). and the generation of bone-destructive behavior.
One of the most likely scenarios for explaining the
bacteria/host/bone breakdown equation that is
Summary periodontitis, is that patients with periodontitis have
some complex multigenetic aberration in their abil-
Homo sapiens share a dynamic existence with 1014 ity to perceive and respond to bacteria. This could
bacteria, with these prokaryotes existing on the either be a deficit or an enhanced ability to recog-
exterior epithelial and mucosal surfaces of the body. nize/respond to bacteria. In either case, the out-
Not only are the exterior surfaces of our bodies come is that local immune responses to oral
awash with bacteria, they are awash with an amaz- bacteria are greater than normal and eventually lead
ing diversity of bacteria, with each individual proba- to aberrant signaling that causes pathological bone
bly having a different set of stably colonizing remodeling. It is indeed fortunate that the technol-
prokaryotes as their life companions (85). These ogy, largely in the form of high-throughput nucleic
bacteria are not simply inert organisms disassoci- acid sequencing (next-generation sequencing), now
ated from their host, but must be part of a rich sig- exists to probe human and bacterial genomes, or
naling nexus in which reciprocal signaling between even metagenomes, to identify mutations in the
bacteria and associated epithelial cells occurs and human genome and relate these to the composition
exists within a further network of reciprocal signal- of the microbiota. This can be aided by quantitative
ing between members of the microbiota. It can be transcriptomic analysis of the inflamed gingival and
argued that these mucosal surfaces represent the oral microbiota using RNA sequencing. The entirety
most complex signaling systems outside the nervous of the information derived from such use of next-
system (Fig. 2). With, say, each individual having generation sequencing, if it can be appropriately
200 bacterial species on the gingival mucosa, this analyzed, is likely to provide a clear indication of
equates to almost 1 million bacterial proteins the probable signaling pathways that result in the
(200 9 3000–6000 genes per bacterium) that are aberrant bone breakdown that is periodontitis. All
potentially able to act as signals between bacteria this requires is the will, and the funding organiza-
and between bacteria and the host epithelium. This tions capable of taking up this challenge.
does not include the multitude of low-molecular-
mass signals generated by bacteria. In turn, the sig-
nals received by the mucosal epithelium can be Acknowledgements
directed to local immune cells to control local
immune responsiveness and to maintain normal B.H. and F.K. acknowledge the support of the Medical
alveolar bone status. We know almost nothing Research Council on their ongoing research.
about this immensely complex signaling system,
other than it goes awry in a large proportion of
individuals, resulting in bone pathology. Such References
pathology could be driven either by colonization
with organisms which significantly influence bone 1. Bab IA, Sela MN, Ginsburg I, Dishon T. Inflammatory
remodeling or by some form of complex genetic lesions and bone resorption induced in the rat periodon-
susceptibility to the normal gingival microbiota, or tium by lipoteichoic acid of Streptococcus mutans. Inflam-
mation 1979: 3: 345–358.
by a combination of both factors. It is likely that an
2. Bainbridge BW, Darveau RP. Porphyromonas gingivalis
understanding of the mechanism of bone destruc- lipopolysaccharide: an unusual pattern recognition recep-
tion in periodontitis can only be achieved by having tor ligand for the innate host defense system. Acta Odontol
full knowledge of the identity of the bacterial Scand 2001: 59: 131–138.

9
Henderson & Kaiser

3. Bandow K, Maeda A, Kakimoto K, Kusuyama J, Shamoto M, Protein A is the von Willebrand factor binding protein on
Ohnishi T, Matsuguchi T. Molecular mechanisms of the Staphylococcus aureus. Blood 2000: 96: 2149–2156.
inhibitory effect of lipopolysaccharide (LPS) on osteoblast 19. Hausmann E, Lu € deritz O, Knox K, Weinfeld N. Structural
differentiation. Biochem Biophys Res Commun 2010: 402: requirements for bone resorption by endotoxin and lipotei-
755–761. choic acid. J Dent Res 1975: 54(Spec): 94–99.
4. Berezow AB, Ernst RK, Coats SR, Braham PH, Karimi-Naser 20. Henderson B, Poole S, Wilson M. Bacteria-Cytokine Interac-
LM, Darveau RP. The structurally similar, penta-acylated tions in Health and Disease. London: Portland Press, 1998.
lipopolysaccharides of Porphyromonas gingivalis and Bac- 21. Hendler A, Mulli TK, Hughes FJ, Perrett D, Bombardieri M,
teroides elicit strikingly different innate immune responses. Houri-Haddad Y, Weiss EI, Nissim A. Involvement of
Microb Pathog 2009: 47: 68–77. autoimmunity in the pathogenesis of aggressive periodonti-
5. Braun T, Zwerina J. Positive regulators of osteoclastogene- tis. J Dent Res 2010: 89: 1389–1394.
sis and bone resorption in rheumatoid arthritis. Arthritis 22. Herath TD, Darveau RP, Seneviratne CJ, Wang CY, Wang Y,
Res Ther 2011: 13: 235. Jin L. Tetra- and penta-acylated lipid A structures of Por-
6. Brown S, Santa Maria JP Jr, Walker S. Wall teichoic acids of phyromonas gingivalis LPS differentially activate TLR4-
gram-positive bacteria. Annu Rev Microbiol 2013: 67: 313– mediated NF-jB signal transduction cascade and immuno-
336. inflammatory response in human gingival fibroblasts. PLoS
7. Choi BK, Lee HJ, Kang JH, Jeong GJ, Min CK, Yoo YJ. Induc- ONE 2013: 8: e58496.
tion of osteoclastogenesis and matrix metalloproteinase 23. Hildebrand D, Heeg K, Kubatzky KF. Pasteurella multocida
expression by the lipooligosaccharide of Treponema denti- toxin-stimulated osteoclast differentiation is B cell depen-
cola. Infect Immun 2003: 71: 226–233. dent. Infect Immun 2011: 79: 220–228.
8. Claro T, Widaa A, O’Seaghdha M, Miajlovic H, Foster TJ, 24. Hiramine H, Watanabe K, Hamada N, Umemoto T. Porphy-
O’Brien FJ, Kerrigan SW. Staphylococcus aureus protein A romonas gingivalis 67-kDa fimbriae induced cytokine pro-
binds to osteoblasts and triggers signals that weaken bone duction and osteoclast differentiation utilizing TLR2. FEMS
in osteomyelitis. PLoS ONE 2011: 6: e18748. Microbiol Lett 2003: 229: 49–55.
9. Di Benedetto A, Gigante I, Colucci S, Grano M. Periodontal 25. Iino Y, Hopps RM. The bone-resorbing activities in tissue
disease: linking the primary inflammation to bone loss. Clin culture of lipopolysaccharides from the bacteria Acti-
Dev Immunol 2013: 2013: 503754. nobacillus actinomycetemcomitans, Bacteroides gingivalis
10. Delpino MV, Barrionuevo P, Macedo GC, Oliveira SC, Gen- and Capnocytophaga ochracea isolated from human
aro SD, Scian R, Miraglia MC, Fossati CA, Baldi PC, mouths. Arch Oral Biol 1984: 29: 59–63.
Giambartolomei GH. Macrophage-elicited osteoclastogene- 26. Kadono H, Kido J, Kataoka M, Yamauchi N, Nagata T. Inhi-
sis in response to Brucella abortus infection requires TLR2/ bition of osteoblastic cell differentiation by lipopolysaccha-
MyD88-dependent TNF-a production. J Leukoc Biol 2012: ride extract from Porphyromonas gingivalis. Infect Immun
91: 285–298. 1999: 67: 2841–2846.
11. Dewhirst FE, Stashenko PP, Mole JE, Tsurumachi T. Purifi- 27. Kamaguchi A, Nakano M, Shoji M, Nakamura R, Sagane Y,
cation and partial sequence of human osteoclast-activating Okamoto M, Watanabe T, Ohyama T, Ohta M, Nakayama
factor: identity with interleukin 1 beta. J Immunol 1985: K. Autolysis of Porphyromonas gingivalis is accompanied by
135: 2562–2568. an increase in several periodontal pathogenic factors in the
12. Eke PI, Dye BA, Wei L, Thornton-Evans GO, Genco RJ, Beck supernatant. Microbiol Immunol 2004: 48: 541–545.
J, Douglass G, Page R. Prevalence of periodontitis in adults 28. Kawata Y, Hanazawa S, Amano S, Murakami Y, Matsumoto
in the United States: 2009 and 2010. J Dent Res 2012: 91: T, Nishida K, Kitano S. Porphyromonas gingivalis fimbriae
914–920. stimulate bone resorption in vitro. Infect Immun 1994: 62:
13. Felix R, Fleisch H, Frandsen PL. Effect of Pasteurella multo- 3012–3016.
cida toxin on bone resorption in vitro. Infect Immun 1992: 29. Kayal RA. The role of osteoimmunology in periodontal dis-
60: 4984–4988. ease. Biomed Res Int 2013: 2013: 639368.
14. Farquharson D, Butcher JP, Culshaw S. Periodontitis, Por- 30. Kiley P, Holt SC. Characterization of the lipopolysaccharide
phyromonas, and the pathogenesis of rheumatoid arthritis. from Actinobacillus actinomycetemcomitans Y4 and N27.
Mucosal Immunol 2012: 5: 112–120. Infect Immun 1980: 30: 862–873.
15. Foged NT, Nielsen JP, Jorsal SE. Protection against progres- 31. Kim M, Jun HK, Choi BK, Cha JH, Yoo YJ. Td92, an outer
sive atrophic rhinitis by vaccination with Pasteurella multo- membrane protein of Treponema denticola, induces osteo-
cida toxin purified by monoclonal antibodies. Vet Rec 1989: clastogenesis via prostaglandin E(2)-mediated RANKL/os-
125: 7–11. teoprotegerin regulation. J Periodontal Res 2010: 45: 772–
16. Fujimura Y, Hotokezaka H, Ohara N, Naito M, Sakai E, 779.
Yoshimura M, Narita Y, Kitaura H, Yoshida N, 32. Kim J, Yang J, Park OJ, Kang SS, Kim WS, Kurokawa K, Yun
Nakayama K. The hemoglobin receptor protein of CH, Kim HH, Lee BL, Han SH. Lipoproteins are an impor-
Porphyromonas gingivalis inhibits receptor activator tant bacterial component responsible for bone destruction
NF-kappaB ligand-induced osteoclastogenesis from bone through the induction of osteoclast differentiation and acti-
marrow macrophages. Infect Immun 2006: 74: 2544– vation. J Bone Miner Res 2013: 28: 2381–2391.
2551. 33. Kirby AC, Meghji S, Nair SP, White P, Reddi K, Nishihara T,
17. Geusens P, Lems WF. Osteoimmunology and osteoporosis. Nakashima K, Willis AC, Sim R, Wilson M. The potent
Arthritis Res Ther 2011: 13: 242. bone-resorbing mediator of Actinobacillus actinomycetem-
18. Hartleib J, Ko€ hler N, Dickinson RB, Chhatwal GS, Sixma JJ, comitans is homologous to the molecular chaperone
Hartford OM, Foster TJ, Peters G, Kehrel BE, Herrmann M. GroEL. J Clin Invest 1995: 96: 1185–1194.

10
Bacterial modulators of bone remodeling in periodontal pocket

34. Kishore U. Target Pattern Recognition in Innate Immunity. 49. Mullan PB, Lax AJ. Pasteurella multocida toxin stimulates
Advances in Experimental Medicine and Biology vol 653. bone resorption by osteoclasts via interaction with osteo-
New York, NY: Springer-Verlag New York, 2009. blasts. Calcif Tissue Int 1998: 63: 340–345.
35. Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, Cap- 50. Nakamura H, Fukusaki Y, Yoshimura A, Shiraishi C, Kishi-
parelli C, Li J, Elliott R, McCabe S, Wong T, Campagnuolo G, moto M, Kaneko T, Hara Y. Lack of Toll-like receptor 4
Moran E, Bogoch ER, Van G, Nguyen LT, Ohashi PS, Lacey decreases lipopolysaccharide-induced bone resorption in
DL, Fish E, Boyle WJ, Penninger JM. Activated T cells regulate C3H/HeJ mice in vivo. Oral Microbiol Immunol 2008: 23:
bone loss and joint destruction in adjuvant arthritis through 190–195.
osteoprotegerin ligand. Nature 1999: 402: 304–309. 51. Nakamura I, Takahashi N, Jimi E, Udagawa N, Suda T. Reg-
36. Kurebayashi Y, Nagai S, Ikejiri A, Koyasu S. Recent advances ulation of osteoclast function. Mod Rheumatol 2012: 22:
in understanding the molecular mechanisms of the devel- 167–177.
opment and function of Th17 cells. Genes Cells 2013: 18: 52. Nakatsuji T, Chiang HI, Jiang SB, Nagarajan H, Zengler K,
247–265. Gallo RL. The microbiome extends to subepidermal com-
37. Lanyon L. Strain-related control of bone (re)modeling: partments of normal skin. Nat Commun 2013: 4: 1431.
objectives, mechanisms and failures. J Musculoskelet Neu- 53. Nishihara T, Ueda N, Amano K, Ishihara Y, Hayakawa H,
ronal Interact 2008: 8: 298–300. Kuroyanagi T, Ohsaki Y, Nagata K, Noguchi T. Actinobacil-
38. Lappin DF, Apatzidou D, Quirke AM, Oliver-Bell J, Butcher lus actinomycetemcomitans Y4 capsular-polysaccharide like
JP, Kinane DF, Riggio MP, Venables P, McInnes IB, Culshaw polysaccharidepromotes osteoclast-like cell formation by
S. Influence of periodontal disease, Porphyromonas gingi- interleukin-1 alpha production in mouse marrow cultures.
valis and cigarette smoking on systemic anti-citrullinated Infect Immun 1995: 63: 1893–1898.
peptide antibody titres. J Clin Periodontol 2013: 40: 907–915. 54. Nyirjesy P. Vulvovaginal candidiasis and bacterial vaginosis.
39. Lax A. The Pasteurella multocida toxin: a new paradigm for Infect Dis Clin North Am 2008: 22: 637–652.
the link between bacterial infection and cancer. Curr Top 55. Pacifici R. T cells: critical bone regulators in health and dis-
Microbiol Immunol 2012: 361: 131. ease. Bone 2010: 47: 461–471.
40. Li X, Yuan FL, Lu WG, Zhao YQ, Li CW, Li JP, Xu RS. The role 56. Piec G, Mirkovitch J, Palacio S, Mu € hlradt PF, Felix R.
of interleukin-17 in mediating joint destruction in rheuma- Effect of MALP-2, a lipopeptide from Mycoplasma fer-
toid arthritis. Biochem Biophys Res Commun 2010: 397: mentans, on bone resorption in vitro. Infect Immun 1999:
131–135. 67: 6281–6285.
41. Liu D, Xu JK, Figliomeni L, Huang L, Pavlos NJ, Rogers M, 57. Prescott D, Lee J, Philpott DJ. An epithelial armamentarium
Tan A, Price P, Zheng MH. Expression of RANKL and OPG to sense the microbiota. Semin Immunol 2013: 25: 323–333.
mRNA in periodontal disease: possible involvement in bone 58. Progulske A, Mishell R, Trummel C, Holt SC. Biological
destruction. Int J Mol Med 2003: 11: 17–21. activities of Eikenella corrodens outer membrane and
42. Madeira MF, Queiroz-Junior CM, Cisalpino D, Werneck SM, lipopolysaccharide. Infect Immun 1984: 43: 178–182.
Kikuchi H, Fujise O, Ryffel B, Silva TA, Teixeira MM, Souza 59. Quirke AM, Lugli EB, Wegner N, Hamilton BC, Charles P,
DG. MyD88 is essential for alveolar bone loss induced by Chowdhury M, Ytterberg AJ, Zubarev RA, Potempa J, Cul-
Aggregatibacter actinomycetemcomitans lipopolysaccharide shaw S, Guo Y, Fisher BA, Thiele G, Mikuls TR, Venables PJ.
in mice. Mol Oral Microbiol 2013: 28: 415–424. Heightened immune response to autocitrullinated Porphy-
43. Maresz KJ, Hellvard A, Sroka A, Adamowicz K, Bielecka E, romonas gingivalis peptidylarginine deiminase: a potential
Koziel J, Gawron K, Mizgalska D, Marcinska KA, Benedyk mechanism for breaching immunologic tolerance in
M, Pyrc K, Quirke AM, Jonsson R, Alzabin S, Venables PJ, rheumatoid arthritis. Ann Rheum Dis 2014: 73: 263–269.
Nguyen KA, Mydel P, Potempa J. Porphyromonas gingivalis 60. Qureshi ST, Larivie re L, Leveque G, Clermont S, Moore KJ,
facilitates the development and progression of destructive Gros P, Malo D. Endotoxin-tolerant mice have mutations in
arthritis through its unique bacterial peptidylarginine deim- Toll-like receptor 4 (Tlr4). J Exp Med 1999: 189: 615–625.
inase (PAD). PLoS Pathog 2013: 9: e1003627. 61. Ray A, Cot M, Puzo G, Gilleron M, Nigou J. Bacterial cell
44. Martineau-Doize  B, Caya I, Gagne  S, Jutras I, Dumas G. wall macroamphiphiles: pathogen-/microbe-associated
Effects of Pasteurella multocida toxin on the osteoclast pop- molecular patterns detected by mammalian innate
ulation of the rat. J Comp Pathol 1993: 108: 81–91. immune system. Biochimie 2013: 95: 33–42.
45. McGraw WT, Potempa J, Farley D, Travis J. Purification, 62. Reddi K, Meghji S, Nair SP, Arnett TR, Miller AD, Preuss M,
characterization, and sequence analysis of a potential viru- Wilson M, Henderson B, Hill P. The Escherichia coli chaper-
lence factor from Porphyromonas gingivalis, peptidy- onin 60 (groEL) is a potent stimulator of osteoclast forma-
larginine deiminase. Infect Immun 1999: 67: 3248–3256. tion. J Bone Miner Res 1998: 13: 1260–1266.
46. Meghji S, Henderson B, Nair SP, Tufano MA. Bacterial por- 63. Robling AG, Turner CH. Mechanical signaling for bone
ins stimulate bone resorption. Infect Immun 1997: 65: modeling and remodeling. Crit Rev Eukaryot Gene Expr
1313–1316. 2009: 19: 319–338.
47. Meghji S, White PA, Nair SP, Reddi K, Heron K, Henderson 64. Rogers JE, Li F, Coatney DD, Rossa C, Bronson P, Krieder
B, Zaliani A, Fossati G, Mascagni P, Hunt JF, Roberts MM, JM, Giannobile WV, Kirkwood KL. Actinobacillus actino-
Coates AR. Mycobacterium tuberculosis chaperonin 10 stim- mycetemcomitans lipopolysaccharide-mediated experimen-
ulates bone resorption: a potential contributory factor in tal bone loss model for aggressive periodontitis. J
Pott’s disease. J Exp Med 1997: 186: 1241–1246. Periodontol 2007: 78: 550–558.
48. Millar SJ, Goldstein EG, Levine MJ, Hausmann E. Lipopro- 65. Rosenberg N, Rosenberg O, Soudry M. Osteoblasts in bone
tein: a gram-negative cell wall component that stimulates physiology-mini review. Rambam Maimonides Med J 2012:
bone resorption. J Periodontal Res 1986: 21: 256–259. 3: e0013.

11
Henderson & Kaiser

66. Rosenstein ED, Greenwald RA, Kushner LJ, Weissmann G. Peptidylarginine deiminase from Porphyromonas gingivalis
Hypothesis: the humoral immune response to oral bacteria citrullinates human fibrinogen and a-enolase: implications
provides a stimulus for the development of rheumatoid for autoimmunity in rheumatoid arthritis. Arthritis Rheum
arthritis. Inflammation 2004: 28: 311–318. 2010: 62: 2662–2672.
67. Rowe DJ, Hausmann E. Quantitative analyses of osteoclast 82. Weidenmaier C, Peschel A. Teichoic acids and related cell-
changes in resorbing bone organ cultures. Calcif Tissue Res wall glycopolymers in Gram-positive physiology and host
1977: 23: 283–289. interactions. Nat Rev Microbiol 2008: 6: 276–287.
68. Schaffler MB, Cheung WY, Majeska R, Kennedy O. Osteo- 83. Widaa A, Claro T, Foster TJ, O’Brien FJ, Kerrigan SW. Sta-
cytes: master orchestrators of bone. Calcif Tissue Int 2014: phylococcus aureus protein A plays a critical role in mediat-
94: 5–24. ing bone destruction and bone loss in osteomyelitis. PLoS
69. Scheres N, de Vries TJ, Brunner J, Crielaard W, Laine ML, ONE 2012: 7: e40586.
Everts V. Diverse effects of Porphyromonas gingivalis on 84. Wilde I, Lotz S, Engelmann D, Starke A, van Zandbergen G,
human osteoclast formation. Microb Pathog 2011: 51: 149– Solbach W, Laskay T. Direct stimulatory effects of the
155. TLR2/6 ligand bacterial lipopeptide MALP-2 on neutrophil
70. Schett G. Osteoimmunology in rheumatic diseases. Arthritis granulocytes. Med Microbiol Immunol 2007: 196: 61–71.
Res Ther 2009: 11: 210. 85. Wilson M. Microbial Inhabitants of Humans. Cambridge:
71. Seya T, Matsumoto M. A lipoprotein family from Myco- Cambridge University Press, 2008.
plasma fermentans confers host immune activation 86. Winrow VR, Mesher J, Meghji S, Morris CJ, Maguire M, Fox
through Toll-like receptor 2. Int J Biochem Cell Biol 2002: S, Coates AR, Tormay P, Blake DR, Henderson B. The two
34: 901–906. homologous chaperonin 60 proteins of Mycobacterium
72. Sismey-Durrant HJ, Hopps RM. The effect of lipopolysac- tuberculosis have distinct effects on monocyte differentia-
charide from the oral bacterium Bacteroides gingivalis on tion into osteoclasts. Cell Microbiol 2008: 10: 2091–2104.
osteoclastic resorption of sperm-whale dentine slices 87. Woo JT, Yonezawa T, Cha BY, Teruya T, Nagai K. Pharma-
in vitro. Arch Oral Biol 1987: 32: 911–913. cological topics of bone metabolism: antiresorptive micro-
73. Stern AR, Nicolella DP. Measurement and estimation of bial compounds that inhibit osteoclast differentiation,
osteocyte mechanical strain. Bone 2013: 54: 191–195. function, and survival. J Pharmacol Sci 2008: 106: 547–554.
74. Sveen K, Skaug N. Bone resorption stimulated by 88. Wright JA, Nair SP. Interaction of staphylococci with bone.
lipopolysaccharides from Bacteroides, Fusobacterium and Int J Med Microbiol 2010: 300: 193–204.
Veillonella, and by the lipid A and the polysaccharide part 89. Xing Q, Ye Q, Fan M, Zhou Y, Xu Q, Sandham A. Porphy-
of Fusobacterium lipopolysaccharide. Scand J Dent Res romonas gingivalis lipopolysaccharide inhibits the
1980: 88: 535–542. osteoblastic differentiation of preosteoblasts by activating
75. Takami M, Kim N, Rho J, Choi Y. Stimulation by toll-like Notch1 signaling. J Cell Physiol 2010: 225: 106–114.
receptors inhibits osteoclast differentiation. J Immunol 90. Yang J, Ryu YH, Yun CH, Han SH. Impaired osteoclastogen-
2002: 169: 1516–1523. esis by staphylococcal lipoteichoic acid through Toll-like
76. Tomomatsu N, Aoki K, Alles N, Soysa NS, Hussain A, Naka- receptor 2 with partial involvement of MyD88. J Leukoc Biol
chi H, Kita S, Shimokawa H, Ohya K, Amagasa T. LPS- 2009: 86: 823–831.
induced inhibition of osteogenesis is TNF-alpha dependent 91. Yonezawa T, Mase N, Sasaki H, Teruya T, Hasegawa S, Cha
in a murine tooth extraction model. J Bone Miner Res 2009: BY, Yagasaki K, Suenaga K, Nagai K, Woo JT. Biselyng-
24: 1770–1781. byaside, isolated from marine cyanobacteria, inhibits osteo-
77. Ueda N, Koide M, Ohguchi M, Ishihara Y, Noguchi T, Oka- clastogenesis and induces apoptosis in mature osteoclasts.
hashi N, Nishihara T. Involvement of prostaglandin E2 and J Cell Biochem 2012: 113: 440–448.
interleukin-1 alpha in the differentiation and survival of 92. Zhao Q, Wang X, Liu Y, He A, Jia R. NFATc1: functions in
osteoclasts induced by lipopolysaccharide from Acti- osteoclasts. Int J Biochem Cell Biol 2010: 42: 576–579.
nobacillus actinomycetemcomitans Y4. J Periodontal Res 93. Zheng T, Noh AL, Park H, Yim M. Aminocoumarins inhibit
1998: 33: 509–516. osteoclast differentiation and bone resorption via downreg-
78. van Venrooij WJ, Pruijn GJ. Citrullination: a small change ulation of nuclear factor of activated T cells c1. Biochem
for a protein with great consequences for rheumatoid Pharmacol 2013: 85: 417–425.
arthritis. Arthritis Res 2000: 2: 249–251. 94. Zhou SR, Moscarello MA, Whitaker JN. The effects of citrul-
79. Venema K. Role of gut microbiota in the control of energy lination or variable amino-terminus acylation on the
and carbohydrate metabolism. Curr Opin Clin Nutr Metab encephalitogenicity of human myelin basic protein in the
Care 2010: 13: 432–438. PL/J mouse. J Neuroimmunol 1995: 62: 147–152.
80. Vossenaar ER, Zendman AJ, van Venrooij WJ, Pruijn GJ. 95. Zou W, Bar-Shavit Z. Dual modulation of osteoclast differ-
PAD, a growing family of citrullinating enzymes: genes, fea- entiation by Lipopolysaccharide. J Bone Miner Res 2002: 17:
tures and involvement in disease. BioEssays 2003: 25: 1106– 1211–1218.
1118. 96. Zouali M. B cell superantigens subvert innate functions of B
81. Wegner N, Wait R, Sroka A, Eick S, Nguyen KA, Lundberg K, cells. Chem Immunol Allergy 2007: 93: 92–105.
Kinloch A, Culshaw S, Potempa J, Venables PJ.

12

You might also like