Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Section 1: Biology and Metabolism of Alveolar Bone

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
DOI: 10.1159/000351895

Cellular and Molecular Aspects of Bone


Remodeling
Wenmei Xiao a, d · Yu Wang c, d · Sandra Pacios d, e · Shuai Li b, d · Dana T. Graves d
Departments of a Periodontology and b Implantology, School and Hospital of Stomatology, Peking University, Beijing, and
c Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Jilin, China; d Department of

Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pa., USA; e Department of Periodontology,
School of Dental Medicine, Universitat Internacional de 29 Catalunya, Sant Cugat del Vallès, Spain

Abstract Cell Types Involved in Bone Remodeling


Bone remodeling is a highly coordinated process re-
sponsible for bone resorption and formation. It is initi- Bone remodeling is a lifelong process where old
ated and modulated by a number of factors including bone is removed (resorption) and new bone is
inflammation, changes in hormonal levels and lack of created (formation) [1, 2]. This process is regu-
mechanical stimulation. Bone remodeling involves the lated by different cell types that form bone (osteo-
removal of mineralized bone by osteoclasts followed by blasts), regulate bone homeostasis (osteocytes),
the formation of bone matrix through osteoblasts that resorb bone (osteoclasts) and affect bone resorp-
subsequently becomes mineralized. In addition to the tion and formation (innate and adaptive immune
traditional bone cells (osteoclasts, osteoblasts and os- cells). There are several pathologic processes that
teocytes) that are necessary for bone remodeling, sev- can affect bone remodeling whereby both bone
eral immune cells such as polymorphonuclear neutro- resorption and formation are affected and have
phils, B cells and T cells have also been implicated in been demonstrated in post-menopausal osteopo-
bone remodelling. Through the receptor activator of nu- rosis, arthritis, periodontal disease and micro-
clear factor-κB/receptor activator of the NF-κB ligand/ gravity or disuse [2].
osteoprotegerin system the process of bone resorption Osteoclasts are terminally differentiated my-
is initiated and subsequent formation is tightly coupled. eloid cells that are uniquely adapted to remove
Mediators such as prostaglandins, interleukins, chemo- mineralized bone matrix [1]. They are found in
kines, leukotrienes, growth factors, wnt signalling and pits within the bone surface which are called
bone morphogenetic proteins are involved in the regu- resorption bays, also known as Howship’s Lacu-
lation of bone remodeling. We discuss here cells and me- nae. Osteoclasts resorb bone by producing
diators involved in the cellular and molecular macha- acid that dissolves the mineral content and en-
nisms of bone resorption and bone formation. zymes that remove the organic matrix. Mature
© 2016 S. Karger AG, Basel osteoclasts anchor to the bone through RGD-
149.126.78.17 - 3/9/2016 9:19:57 AM
Downloaded by:
UCL
binding sites to create a sealed microenviron- vascularization phenotype called M1 or M2. M1
ment. macrophages produce cytokines such as IL-1β,
Osteoblasts are bone-forming cells that arise TNF-α and RANKL, whereas M2 monocytes/
from osteoprogenitor cells. RUNX2 (runt-related macrophages produce anti-inflammatory media-
transcription factor 2) and other transcription tors such as IL-10 and IL-4 [5]. Dendritic im-
factors control the differentiation of osteoblasts mune cells have their origin in bone marrow and
[2–4]. During bone formation, a subpopulation are found in a number of different tissues. One of
of osteoblasts undergoes terminal differentiation the first contacts between oral bacteria and the
and becomes engulfed by osteoid, at which time immune system occurs with Langerhans cells, a
they are referred to as osteoid osteocytes. Osteo- subset of dendritic cells found in mucosal surfac-
cytes which reside in lacunae are the most numer- es and skin. They function in activating the im-
ous cell type found in mature bone and are long- mune response as antigen presenting cells and
lived. They have dendritic processes that interact regulate homeostasis by modulating the response
with other osteocytes and bone-lining cells on the to oral bacteria and self-antigens. Once they de-
bone surface. Osteocytes respond to mechanical tect antigen dendritic cells travel to lymph nodes
load. Under resting conditions osteocytes express and present antigen to activate lymphocytes.
sclerostin, which directly prevents Wnt signaling The predominant cells of the adaptive immune
(described in more detail below). Sclerostin ex- response are B and T lymphocytes. Activated T
pression can be inhibited by parathyroid hor- and B cells can express RANKL and various other
mone signaling to remove this inhibitor of Wnt cytokines that typically promote osteoclastogen-
signaling and allow Wnt directed bone formation esis. B cells express TNF-α, IL-6, and RANKL to
to occur. Under basal conditions, osteocytes se- promote osteoclastogenesis. T cells can develop
crete transforming growth factor β (TGF-β), into T-helper cells, Th1, Th2 and Th17 that mod-
which inhibits osteoclastogenesis. However, ulate bone resorption. Th1-type produce IL-1 and
upon stimulation osteoblasts and osteocytes pro- TNF-α that can promote bone resorption. Th17
duce osteoclastogenic factors such as macrophage cells have recently been identified as an effector T
colony-stimulating factor-1 (CSF-1) and receptor helper cell subset characterized by the production
activator of the NF-κB ligand (RANKL) to induce of proinflammatory cytokines. IL-1 and IL-17 me-
bone remodeling [1–4]. diate osteoclast formation through induction of
Innate immune cells (primarily polymorpho- RANKL [6]. Lymphocyte subsets (Th2) produce
nuclear neutrophils (PMNS), monocytes/macro- cytokines that are anti-inflammatory, IL-4 and IL-
phages and dendritic cells) and adaptive immune 10. These cytokines reduce osteoclastogenesis and
cells (primarily lymphocytes) modulate bone re- the severity of bone loss [7].
sorption particularly under inflammatory condi-
tions. PMNs are granular leukocytes that pre-
dominate in the initial acute inflammatory re- Mediators Involved in Bone Resorption
sponse. Like other leukocytes, they are recruited
from the peripheral vasculature by chemotactic Mediators play an important role in the patho-
factors, particularly chemokines. They express a genesis of bone damage. Cytokines stimulate the
number of inflammatory cytokines (e.g. IL-1β, recruitment, formation and activity of the bone-
TNF-α, IL-6) as well as membrane-bound RANKL resorbing cell, the osteoclast. They trigger the
[4]. Macrophages are produced from the differ- chemotaxis of osteoclast precursors, the induc-
entiation of monocytes in tissue. Monocytes/ tion of genes that lead to fusion of these precur-
macrophages can have an inflammatory or a pro- sors, the maturation of osteoclasts and the
149.126.78.17 - 3/9/2016 9:19:57 AM

10 Xiao · Wang · Pacios · Li · Graves


Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895
synthesis of matrix enzymes leading to bone re- OPG, also called tumor necrosis factor recep-
sorption. Understanding these steps has led to the tor superfamily member 11B (TNFRSF11B) is a
development of therapeutic agents that can block soluble cytokine receptor that belongs to the TNF
these osteoclastogenesis and activity reducing family [10]. It is produced by osteoblasts, fibro-
bone loss [8]. blasts and many other cell types. OPG binds to
RANKL and CSF-1 work cooperatively and rep- RANKL preventing RANKL from binding to its
resent one of the dominant pathways that leads to cognate receptor, RANK. Thus, OPG is a natural
osteoclast formation and activity [9]. CSF-1 is cru- decoy receptor (inhibitor) of RANKL [11]. The
cial for the proliferation and survival of osteoclast RANKL/OPG expression ratio to a large degree
precursor cells, while RANKL is essential for osteo- determines the degree of osteoclast formation
clast differentiation [9]. RANKL, also called tumor and activity [4].
necrosis factor ligand superfamily member 11 is a A number of pro-inflammatory cytokines
cytokine that belongs to the TNF family. RANKL is stimulate bone resorption [4] (fig. 1). Most ac-
expressed on the surface of marrow stromal cells, complish this by stimulating production of
monocytes, activated T and B cells, osteocytes and RANKL. Some, such as TNF-α can induce
precursors of bone forming osteoblasts. RANKL RANKL but also stimulate osteoclast formation
may be cleaved and released in a soluble form by directly, independently of RANKL (fig. 1). TNF-α
metalloproteinases. It should be noted that the cell is produced primarily by activated macrophages,
type that expresses RANKL may depend upon the but also by other cell types such as activated T
etiology of the bone resorption. For example, the cells, polymorphonuclear leukocytes, epithelial
cells that produce RANKL differ between bone re- cells, endothelial cells, fibroblasts and bone-lin-
sorption caused by post-menopausal osteoporosis ing cells including osteoblasts. TNF-α upregu-
versus resorption associated with periodontal dis- lates c-fms (CSF-1 receptor) expression and acti-
ease. During physiologic bone resorption, these vates osteoclasts by enhancing RANKL signaling
factors act on osteoblasts/osteocytes to regulate mechanisms. TNF-α induces osteoclast precur-
RANKL and osteoprotegerin (OPG) expression. sors and marrow stromal cells to produce osteo-
CSF-1 also known as macrophage colony- clastogenic cytokines, such as IL-1, RANKL, and
stimulating factor (M-CSF) is a glycoprotein M-CSF. TNF also inhibits the bone-forming
growth factor that specifically regulates the sur- function of osteoblasts. Studies show that TNF-α
vival, proliferation and differentiation of mono- inhibits the differentiation of new osteoblasts
cyte-macrophage lineage cells through a cell sur- from precursor cells. The regulation of RUNX2
face receptor selectively expressed on these cell by TNF could diminish recruitment of osteoblast
types, c-fms. M-CSF is released by osteoblasts and precursors into the pool of mature bone-forming
bone marrow progenitor cells that upon binding cells [12].
to its receptor, c-fms on pre-osteoclasts, activates IL-1, which is encoded by two separate genes,
an intracellular cascade that leads to proliferation IL-1α and IL-1β, is a potent bone resorbing cyto-
of precursors and survival of osteoclasts. RANKL kine produced by various cell types such as mono-
stimulates osteoclastogenesis when it binds to re- cytes, macrophages, polymorphonuclear leuko-
ceptor activator of nuclear factor-κB (RANK), cytes, fibroblasts (gingival and periodontal liga-
which is located on osteoclast precursors to acti- ment), epithelial cells, endothelial cells and
vate NF-κB and other signaling pathways. This osteoblasts. Both isoforms of IL-1 stimulate pro-
binding promotes osteoclast formation, activa- duction of other cytokines and prostaglandins
tion, and survival, thus inducing osteoclast activ- and can induce RANKL expression as well as pro-
ity and bone resorption (fig. 1). duction of degradative enzymes. However, it does
149.126.78.17 - 3/9/2016 9:19:57 AM

Bone Remodeling 11
Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895
Leukocyte
Lipoxin A4 Osteoclast Mesenchymal stem cell
Resolvin E1 IL-1 CXCL8 precursor
Resolvin D1 IL-6 CXCL12 LTB4
Protectin D1 IL-7 CCL3 LTC4 CXCL10

Chemotaxis
IL-11 CCL5 LTD4 CXCL12
IL-17 CCL9 LTDE4 CXCL13
71)į CCL7 CCL5
IL-4 LIF CCL23 PGE2
IL-10

Chemotaxis
OSM CXCL9 PGI2
IL-13

Proliferation
CCL2 3*)į
,)1Dž

Osteoclastogenesis
PGE2 M-CSF
FGF

Differentiation
Wnt/DŽ-catenin PDGF
RANKL BMP-2
OPG TNF
7*)DŽ
IGF

Activity
DKK1 Sclerostin

Bone lining cells New bone

Osteoclasts

Fig. 1. Stimulation of osteoclastogenesis, bone resorption, and coupled bone formation. RANKL, M-CSF, and TNF di-
rectly stimulate the formation of osteoclasts, while other cytokines or lipid-based mediators such as prostaglandins
or leukotrienes indirectly stimulate osteoclastogenesis by effects on RANKL, M-CSF, or TNF-α. Chemokines affect re-
sorption by stimulation recruitment of osteoclast precursors or osteoclast activity. Chemokines such as CXCL10,
CXCL12, CXCL13, and CCL5, may affect bone formation by effects on osteoblast precursors or osteoblasts. Wnt signal-
ing, growth factors such as FGF, PDGF, BMP-2, TGF-β, and IGF play an important role in osteogenesis by stimulating
proliferation of mesenchymal stem cells/osteoblast precursors and inducing osteoblast differentiation or synthesis of
bone matrix. Modified with permission from Graves et al. [30].

not directly induce osteoclastogenesis [13]. IL-6 ulates cells to express receptor activator of
is produced by many of the same cell types that RANKL. IL-17 also reduces bone formation via
produce IL-1. IL-6 has been reported to stimulate inhibition of type I collagen synthesis [15]. Other
bone resorption by enhancing osteoclast forma- osteoclastogenic cytokines that have been shown
tion through a RANKL-dependent mechanism to stimulate activation of RANKL are IL-11, leu-
[14]. IL-7 is an osteoclastogenic cytokine mainly kemia inhibitory factor, and oncostatin M (fig. 1).
produced by stromal cells and osteoblasts that Chemokines are chemotactic cytokines that
promotes RANKL expression. It also inhibits new stimulate recruitment of leukocytes and are pro-
bone formation by down regulation of the osteo- duced by a wide variety of cell types including ep-
blast-specific transcription factor Runx2. IL-17 is ithelial cells, endothelial cells, and many different
produced by activated CD4+ Th17 cells and stim- leukocyte subsets of the innate and adaptive
149.126.78.17 - 3/9/2016 9:19:57 AM

12 Xiao · Wang · Pacios · Li · Graves


Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895
immune response, fibroblasts or bone lining cells PMNs and their clearance by macrophages,
[4]. Some chemokines recruit osteoclast precur- blocking leukotrienes and prostaglandins as well
sors (fig. 1). CCL2 (also known as monocyte che- as reducing cytokine release [21]. For example,
moattractant protein-1, MCP-1) induces recruit- resolvin E1 is produced during the resolution of
ment of osteoclast precursors. Other chemokines the inflammation and blocks stimulation by leu-
that have been shown to stimulate recruitment of kotriene B4. This inhibition attenuates neutro-
osteoclast precursors are CCL5, CXCL8, CCL9, phil migration leading thereby reducing inflam-
CCL7, CCL23, CXCL12, and CXCL9 [4, 16, 17]. mation. Lipoxin acts by binding to lipoxin A4 re-
CCL3 (also called MIP-1α), a pro-inflammatory ceptor inhibiting chemotaxis, transmigration and
chemokine produced at inflammatory sites, ap- blocking activation of nuclear factor kappa-B.
pears to play a crucial role in pathologic osteo- Matrix metalloproteinases (MMPs) are prote-
clastogenesis associated with multiple myelomas. ases produced by different cell types such as mac-
It modulates osteoclast differentiation by binding rophages and fibroblasts [22]. Inflammatory me-
to G-protein coupled receptors, CCR1 and CCR5, diators induce production of MMP from a num-
and activating ERK and AKT signaling pathways. ber of cell types such as fibroblasts and PMNs.
The MIP-1β (CCL4) also can enhance bone re- These molecules degrade extracellular matrix
sorption [16, 17]. proteins such as collagen, elastin, and laminins.
Prostaglandins and leukotrienes are lipid- Osteoclasts secrete MMP contributing to degra-
based mediators derived from fatty acids that are dation of bone matrix following decalcification
produced by different cell types such as macro- by acid production. Furthermore, MMPs can ac-
phages, fibroblasts and gingival epithelial cells tivate chemokines and cytokines to amplify in-
(fig. 1) [18]. Prostaglandin E (PGEs), such as flammation [23]. For example, MMPs activate
prostaglandin E2, prostacyclin and prostaglandin chemokines by cleavage of the N-terminal do-
F2α stimulate osteoclast formation through main.
RANKL and direct effect on osteoclast formation,
also stimulate bone formation. Prostaglandin E2
has been linked to stimulating insulin growth fac- Mediators That Stimulate Bone Formation
tor-1 gene expression, which promotes collagen
synthesis by osteoblasts, thus enhancing bone Wnt signaling, growth factors and bone morpho-
formation [19]. Leukotrienes are another class of genetic proteins play an important role in osteo-
inflammatory lipid mediators and stimulate che- genesis [24, 27, 28] (fig. 1). The Wnt signaling
motaxis of leukocytes and the generation of su- transduction pathway plays an important role in
peroxides in neutrophils. Leukotrienes such as stimulating bone formation and in many other
LTB4, LTC4, LTD4 and LTDE4 enhance osteoclast processes including embryonic development and
formation and activation of mature osteoclasts by tumorigenesis [24]. Wnts are secreted, cysteine-
a RANKL independent mechanism. LTB4 may rich glycoproteins involved in controlling cell
suppress bone formation by inhibiting the prolif- proliferation, cell-fate specification, gene expres-
eration of primary osteoblast precursors and re- sion, and cell survival. Wnt-3a and Wnt-7a are ex-
duce the capacity of osteoblast precursors to dif- pressed in the limb bud and have roles in skeletal
ferentiate into osteoblasts and form new bone pattern determination, while Wnt-14 is involved
[20]. Lipoxins and the D and E series of resolvins in joint formation. And Wnt-3a, Wnt-4, Wnt-5a,
are endogenous anti-inflammatory lipid media- and Wnt-7a all influence cartilage development.
tors that act by controlling the resolution of the Wnt receptors are including low-density lipopro-
inflammation through enhancing apoptosis of tein receptor-related proteins (LRP) and frizzleds
149.126.78.17 - 3/9/2016 9:19:57 AM

Bone Remodeling 13
Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895
(Fzd). LRP are evolutionarily conserved plasma way receptors membrane complex to inhibit Wnt
membrane receptors with a variety of functions activity. Sclerostin, the protein product of the
including lipid metabolism, cargo transport, and Sost gene is also a Wnt inhibitor. It is produced by
cellular signaling. LRP-5 is expressed by osteo- osteocytes and is abundant in osteocytic canalicu-
blasts of the endosteal and trabecular bone sur- lae. Sclerostin binds to Lrp5/6 to block the Wnt/
face. It regulates osteoblastic proliferation, surviv- beta-catenin pathway and inhibit proliferation
al and activity. Fzds are highly versatile seven- and differentiation of osteoblasts and to increase
membrane proteins that contribute to activation their apoptosis [25].
of both β-catenin and non-β-catenin signaling Fibroblast growth factor-2 (FGF-2 also known
pathways by virtue of their interactions with Di- as basic FGF) is involved in numerous cellular
shevelled (Dsh, or Dvl), a cytoplasmic phospho- processes including angiogenesis, tumorigenesis,
protein that acts directly downstream of frizzled cell proliferation, differentiation, wound healing,
receptors. The Wnt pathway has been found to limb formation, and bone biology. In bone, FGF-2
play a central role in controlling embryonic bone is expressed in osteoblasts and mesenchymal
development and bone mass. There are four Wnt cells. Postnatally, FGF-2 is produced by mature
pathways: the canonical Wnt pathway (the Wnt/ osteoblasts and stored in extracellular matrix.
b-catenin pathway) and three non-canonical Wnt FGF-2 along with FGF-18 is a bone anabolic
(β-catenin independent) pathways: the Wnt/ agent that stimulates uncommitted bone marrow
Ca2þ pathway, the Wnt/planar cell polarity (Wnt/ stromal cells to differentiate into osteoblasts and
PCP) pathway, and the Wnt/protein kinase A form osteoid. Thus it plays an important role in
(Wnt/PKA) pathway. Beta-catenin, a key compo- skeletal development by regulating the prolifera-
nent of the canonical Wnt signaling pathway in- tion and differentiation of osteoblasts [26]. FGF-2
teracts with a number of different transcription signaling regulates the Wnt/beta-catenin path-
factors and modulates their activity. This leads to way and activates transcription factors Runx2
the transcription of a number of different target and activating transcription factor 4 (ATF4),
genes that regulate a diverse array of biological thereby promoting osteoblast differentiation
processes. In the osteocyte Wnt/beta-catenin sig- [27].
naling is required for normal bone homeostasis Bone morphogenetic proteins (BMPs) are a
[24]. The Wnt/beta-catenin pathway is anabolic group of protein factors that stimulate bone for-
for bone formation and promotes increased bone mation and are important in a number of other
density and strength. Beta-catenin activation fa- biologic factors such as skin formation and hair
cilitates osteoblast differentiation and enhances follicle development [28] (fig. 1). BMPs released
osteoblast and osteocyte survival in vitro. The by osteoclasts and from resorbing bone matrix in-
Wnt pathway can also affect osteoclastogenesis. teract with specific receptors on the cell surface,
The Wnt/beta-catenin pathway suppresses physi- referred to as bone morphogenetic protein recep-
ologic bone resorption by upregulation of OPG tors (BMPRs). There are type I and type II BMPRs.
expression and downregulation of RANKL in os- Two subclasses of type I receptors have been
teoblasts/osteocytes [25]. identified, type IA and IB (BMPR1A and
The Wnt pathway is inhibited by Dickkopf BMPR1B). Upon ligand binding, the type II re-
factors 1–4 (DKK1, DKK2, DKK3, DKK4) and ceptor forms a heterodimer with the type I recep-
sclerostin. Dkk1 is expressed by synovial cells, en- tor, and the constitutive kinase of the type II acti-
dothelial cells and chondrocytes. Dickkopf fac- vates the type I receptor. The latter initiates a sig-
tors, especially DKK1, bind and sequester the nal transduction cascade by phosphorylating
LRP5/6 and Kremen1/2 (Krm1/2), the Wnt path- downstream cytosolic factors, which then
149.126.78.17 - 3/9/2016 9:19:57 AM

14 Xiao · Wang · Pacios · Li · Graves


Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895
translocate to the nucleus to activate or inhibit expression of RANKL in osteoblasts/osteocytes
transcription. Signaling by BMPs plays an impor- [28]. BMP signaling can be inhibited or modified
tant role in variety of cell types in bone such as in many ways, including inhibition through chor-
osteoblasts and chondrocytes. BMPs have widely din and noggin. These antagonists are regulated
recognized roles in bone formation such as BMP- by BMPs, indicating the existence of local feed-
2, BMP-4, BMP-5, BMP-6 and BMP-7 which back mechanisms to modulate BMP cellular ac-
have an osteogenic capacity. BMPs stimulate in- tivities. For example, noggin binds with various
tracellular signaling by activating the mothers degrees of affinity to BMP-2, BMP-4, BMP-5,
against decapentaplegic (Smad) and mitogen-ac- BMP-6 and BMP-7. Crystallography of noggin
tivated protein kinase (MAPK) pathways. Fol- and BMP-7 reveals that noggin inhibits BMP-7
lowing BMP induction, both the Smad and MAPK signaling by blocking the molecular interfaces of
pathways converge at the Runx2 gene to control the binding epitopes for both type I and type II
its activation. The subsequent signaling induced BMP receptors. Basal noggin expression in osteo-
by BMP (ligand, receptors, intracellular media- blasts is limited but its transcript levels are up-
tors, activation of transcription factors or comod- regulated by BMP-2, BMP-4 and BMP-6 as a po-
ulators) is responsible for the final target gene ex- tential protective mechanism limiting excessive
pression. BMP signaling may also be required for BMP stimulation [29].

References
1 Schett G, Teitelbaum SL: Osteoclasts 9 Takayanagi H: Osteoimmunology: 15 Daoussis D, Andonopoulos AP, Liossis
and arthritis. J Bone Miner Res 2009;24: shared mechanisms and crosstalk be- SN: Wnt pathway and IL-17:novel regu-
1142–1146. tween the immune and bone systems. lators of joint remodeling in rheumatic
2 Jiao H, Xiao E, Graves DT: Diabetes and Nat Rev Immunol 2007;7:292–304. diseases. Looking beyond the RANK-
its effect on bone and fracture healing. 10 Belibasakis GN, Bostanci N: The RANKL-OPG axis. Semin Arthritis
Curr Osteoporos Rep 2015;13:327–335. RANKL-OPG system in clinical peri- Rheum 2010;39:369–383.
3 Karsenty G: Transcriptional control of odontology. J Clin Periodontol 2012;39: 16 Szekanecz Z, Koch AE, Tak PP: Chemo-
skeletogenesis. Annu Rev Genomics 239–248. kine and chemokine receptor blockade
Hum Genet 2008;9:183–196. 11 Simonet WS, Lacey DL, Dunstan CR, et in arthritis, a prototype of immune-me-
4 Graves DT, Oates T, Garlet GP: Review al: Osteoprotegerin: a novel secreted diated inflammatory diseases. Neth J
of osteoimmunology and the host re- protein involved in the regulation of Med 2011;69:356–366.
sponse in endodontic and periodontal bone density. Cell 1997;89:309–319. 17 Szekanecz Z, Vegvari A, Szabo Z, Koch
lesions. J Oral Microbiol 2011;3:10.3402. 12 Karmakar S, Kay J, Gravallese EM: Bone AE: Chemokines and chemokine recep-
5 Benoit M, Desnues B, Mege JL: Macro- damage in rheumatoid arthritis: mecha- tors in arthritis. Front Biosci (Schol Ed)
phage polarization in bacterial infec- nistic insights and approaches to pre- 2010;2:153–167.
tions. J Immunol 2008;181:3733–3739. vention. Rheum Dis Clin North Am 18 Bage T, Kats A, Lopez BS, et al: Expres-
6 Sato K, Suematsu A, Okamoto K, et al: 2010;36:385–404. sion of prostaglandin E synthases in
Th17 functions as an osteoclastogenic 13 Nagai H, Tsukuda R, Mayahara H: Ef- periodontitis immunolocalization and
helper T cell subset that links T cell acti- fects of basic fibroblast growth factor cellular regulation. Am J Pathol 2011;
vation and bone destruction. J Exp Med (bFGF) on bone formation in growing 178:1676–1688.
2006;203:2673–2682. rats. Bone 1995;16:367–373. 19 Minamizaki T, Yoshiko Y, Kozai K, Au-
7 Dinarello CA: Historical insights into 14 Palmqvist P, Persson E, Conaway HH, bin JE, Maeda N: EP2 and EP4 receptors
cytokines. Eur J Immunol 2007;37(suppl Lerner UH: IL-6, leukemia inhibitory differentially mediate MAPK pathways
1):S34–S45. factor, and oncostatin M stimulate bone underlying anabolic actions of prosta-
8 Schett G: Effects of inflammatory and resorption and regulate the expression glandin E2 on bone formation in rat
anti-inflammatory cytokines on the of receptor activator of NF-kappa B li- calvaria cell cultures. Bone 2009;44:
bone. Eur J Clin Invest 2011;41:1361– gand, osteoprotegerin, and receptor ac- 1177–1185.
1366. tivator of NF-kappa B in mouse calvari-
ae. J Immunol 2002;169:3353–3362.
149.126.78.17 - 3/9/2016 9:19:57 AM

Bone Remodeling 15
Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895
20 Jiang J, Lv HS, Lin JH, Jiang DF, Chen 23 Giannobile WV: Host-response thera- 27 Ai-Aql ZS, Alagl AS, Graves DT, Ger-
ZK: LTB4 can directly stimulate human peutics for periodontal diseases. J Peri- stenfeld LC, Einhorn TA: Molecular
osteoclast formation from PBMC inde- odontol 2008;79:1592–1600. mechanisms controlling bone formation
pendent of RANKL. Artif Cells Blood 24 Chen Y, Alman BA: Wnt pathway, an during fracture healing and distraction
Substit Immobil Biotechnol 2005;33: essential role in bone regeneration. J osteogenesis. J Dent Res 2008;87:107–
391–403. Cell Biochem 2009;106:353–362. 118.
21 Serhan CN, Chiang N, Van Dyke TE: 25 Takahashi N, Maeda K, Ishihara A, Ue- 28 Chen G, Deng C, Li YP: TGF-beta and
Resolving inflammation: dual anti-in- hara S, Kobayashi Y: Regulatory mecha- BMP signaling in osteoblast differentia-
flammatory and pro-resolution lipid nism of osteoclastogenesis by RANKL tion and bone formation. Int J Biol Sci
mediators. Nat Rev Immunol 2008;8: and Wnt signals. Front Biosci 2011;16: 2012;8:272–288.
349–361. 21–30. 29 Krause C, Guzman A, Knaus P: Noggin.
22 Manicone AM, McGuire JK: Matrix me- 26 Downey ME, Holliday LS, Aguirre JI, Int J Biochem Cell Biol 2011;43:478–
talloproteinases as modulators of in- Wronski TJ: In vitro and in vivo evi- 481.
flammation. Semin Cell Dev Biol 2008; dence for stimulation of bone resorption 30 Graves DT, Li J, Cochran DL: Inflamma-
19:34–41. by an EP4 receptor agonist and basic tion and uncoupling as mechanisms of
fibroblast growth factor: Implications periodontal bone loss. J Dent Res 2011;
for their efficacy as bone anabolic 90:143–153.
agents. Bone 2009;44:266–274.

Dana Graves
Department of Periodontics, University of Pennsylvania
240 S. 40th St., Levy 122
Philadelphia, PA 19104 (USA)
E-Mail dtgraves@dental.upenn.edu
149.126.78.17 - 3/9/2016 9:19:57 AM

16 Xiao · Wang · Pacios · Li · Graves


Downloaded by:

Kantarci A, Will L, Yen S (eds): Tooth Movement. Front Oral Biol. Basel, Karger, 2016, vol 18, pp 9–16
UCL

DOI: 10.1159/000351895

You might also like