You are on page 1of 12

CRITICAL REVIEWS IN ORAL BIOLOGY & MEDICINE

V. Krishnan1* and Z. Davidovitch2


On a Path to Unfolding the
Biological Mechanisms of
1
Department of Orthodontics, Rajas Dental College,
Tirunelveli District, Tamilnadu, India; and 2Department

Orthodontic Tooth Movement


of Orthodontics, Case Western Reserve University,
Cleveland, OH, USA; *corresponding author, Gourivilasam,
Kudappanakunnu PO, Trivandrum, Kerala State—695043,
India, vikrishnan@yahoo.com

J Dent Res 88(7):597-608, 2009

Abstract
Orthodontic forces deform the extracellular matrix
and activate cells of the paradental tissues, facili­
tating tooth movement. Discoveries in mechano­ INTRODUCTION

T
biology have illuminated sequential cellular and he generation and propagation of signaling cascades and associated tissue
molecular events, such as signal generation and remodeling in response to applied mechanical loads form the central theme of
transduction, cytoskeletal re-organization, gene orthodontic tooth movement. All cell types are capable of reacting when subjected
expression, differentiation, proliferation, synthesis to mechanical stimuli of either intracellular or extracellular origin (Hamill and
and secretion of specific products, and apoptosis. Martinac, 2001; Schwartz and DeSimone, 2008). The main challenges encoun­
Orthodontists work in a unique biological environ­ tered by researchers have been to elucidate the intricate details of the conversion of
ment, wherein applied forces engender remodeling mechanical forces into biochemical signals, to identify the cellular parts involved
of both mineralized and non-mineralized paraden­ in this process, to highlight the signaling pathways, and, ultimately, to determine how
tal tissues, including the associated blood vessels the response mechanisms are activated, leading to tissue remodeling that facilitates
and neural elements. This review aims at identify­ the movement of teeth to different, more desirable locations in the dental arch.
ing events that affect the sequence, timing, and Orthodontic forces affect the extracellular matrix (ECM) and the cells of the
significance of factors that determine the nature of dental pulp, periodontal ligament (PDL), alveolar bone, and gingiva. These
the biological response of each paradental tissue to effects are both physical and biochemical in nature, and are frequently inter­
orthodontic force. The results of this literature twined and interdependent. Recognition of these intimate associations and the
review emphasize the fact that mechanoresponses realization of their importance in defining the details of the biological response
and inflammation are both essential for achieving to applied mechanical loads in vitro and in vivo have been reflected lately by a
tooth movement clinically. If both are working in great increase in the number of articles focusing on this subject (Krishnan and
concert, orthodontists might be able to accelerate Davidovitch, 2006a; Masella and Meister, 2006; Henneman et al., 2008; Wise
or decelerate tooth movement by adding adjuvant and King, 2008). The authors of all these reviews seem to believe that an
methods, whether physical, chemical, or surgical. expanded understanding of these associations may be meritorious for the ortho­
dontist, as well as for his/her patients. For example, a recent review (Henneman
Key words: tooth movement, cellular mechano­ et al., 2008) has listed tissue and cellular responses to mechanical force applica­
transduction, extracellular matrix, cytoskeleton, tion in the following order: (1) matrix strain and fluid flow, (2) cell strain,
nucleic acids, osteocytes, fibroblasts, angiogenesis, (3) cell activation and differentiation, and (4) tissue remodeling. However, that
mechanoreceptors. article concentrated on alveolar bone and PDL cells, largely ignoring other para­
dental tissues known to respond to orthodontic forces.
In contrast, this article is aimed at expanding the overview of the orthodon­
tic tooth movement process, by delineating reactions occurring in mineralized
(alveolar bone) and non-mineralized (PDL and gingiva) paradental tissues,
and their associated neurovascular networks. It presents known information
about the mechanism of cell signaling in response to mechanical loading,
including mechanosensing, transduction, and cellular responses. Furthermore,
the various components of this ECM/cellular interrelated chain of responses
are presented in an organized sequence, highlighting the links between clinical
events and knowledge derived from basic research.

EXTRACELLULAR MATRIX REMODELING


DOI: 10.1177/0022034509338914
Application of external mechanical loads to teeth may alter the effects of
Received August 22, 2008; Last revision November 20, gravitational and internal forces acting on resident cells, leading to changes in
2008; Accepted December 11, 2008 gene expression, and production of proteins that ultimately alter the structure

597
598  Krishnan & Davidovitch J Dent Res 88(7) 2009

and function of the ECM, as well as the jaw bones. The most factor β and insulin-like growth factor mRNA expression are
important ECM macromolecules in determining its mechanical increased (Wildemann et al., 2004). Research reports demon­
properties are collagen (the most abundant molecule), proteo­ strating increases in osteocyte-specific markers have also been
glycans, laminin, fibronectin, elastin, and hyaluronic acid. These published, including E11/gp3.8 (Zhang et al., 2006), dentin
molecules bind to cell adhesion foci, consisting mainly of inte­ matrix protein 1 (found in a tooth movement model) (Gluhak-
grins, to transfer the signals intracellularly (Li et al., 2008; Heinrich et al., 2003), MEPE (Gluhak-Heinrich et al., 2007),
Schwartz and DeSimone, 2008; Wells, 2008; Kong and Vazquez, and sclerostin (Robling et al., 2008). Anabolic signals, such as
2009). Thus, ECMs can be considered to be multi-component nitric oxide, prostaglandins, and ATP, are released within sec­
tissues that enable internal and external mechanical strains onds of osteocyte loading (Bakker et al., 2001). All these signals
to effect changes in organ structure and function, through activate bone-surface cells and osteoblasts, by the network of
mechanotransduction. cell-cell communications. It was proposed recently that the
Wnt/β-catenin pathway initiates, while the SOST/sclerostin
pathway inhibits, new bone formation. Osteocytes are the only
THE CONCEPT OF TENSEGRITY
cells secreting sclerostin—the product of the SOST gene.
The structure and shape of cells are determined by 3 cytoskeletal Sclerostin potently antagonizes and negatively regulates several
molecular structures—microfilaments, microtubules, and inter­ members of the bone morphogenetic protein (BMP) family of
mediate filaments—which link the nucleus to the cell-surface proteins, and also binds to LRP5/LRP6, preventing canonical
adhesion receptors. Since microfilaments are chemically Wnt signaling. Both BMPs and Wnts are critical for osteoblas­
attached to proteins in the cellular and nuclear membranes, they togenesis, since they provide the initial and essential stimulus
are ideal candidates for transfer of tractional forces from inside for the commitment of multipotential mesenchymal progenitors
the cell. These mechanical signals are transferred to the ECM to the osteoblast lineage (Canalis et al., 2003; Li et al., 2005).
through integrins, and to adjacent cells through cadherins, and This makes sclerostin a likely candidate as an osteocyte-derived
are resisted and balanced by the same attachment entities. This regulator of osteoblast function (Tatsumi et al., 2007). It was
physiological pre-existing tensile stress in the cell is known as reported recently (Robling et al., 2008) that SOST transcripts
‘tensegrity’ (tension-dependent cellular integrity), and is consid­ and sclerostin protein levels were dramatically reduced by
ered to be essential for the normal function of cells, tissues, and mechanical loading.
organs, as well as for proper growth and survival. In the absence The fluid flow hypothesis, describing a mechanism by which
of this internal tension, cells often undergo apoptosis (Ingber, osteocytes respond to mechanical forces, states that locally
2006, 2008). The physical properties of the ECM also plays a evoked strain derived from the displacement of fluid in the
significant role in cell behavior, by determining the transmission canaliculi is very important (Goulet et al., 2008). When loading
of stress from the outside to the inside of the cell. According to occurs, interstitial fluid is squeezed through the thin layer of
the tensegrity model of Ingber, a stiffer cytoskeleton is advanta­ non-mineralized matrix surrounding the cell bodies and cell
geous in sensing external mechanical loading, in contrast to a processes, resulting in local strain at the cell membrane and
completely relaxed cytoskeleton. The cells are more sensitive to activation of the affected osteocytes (Weinbaum et al., 1994; Y
dynamic changes in mechanical stress when the effective exter­ Wang et al., 2007; Goulet et al., 2008). Osteocytes may also
nal and internal stresses are similar in magnitude. In contrast, send signals for activation of the bone resorption cascade
the cells in a completely relaxed state are unable to sense an through expression of activator for NF-κB ligand (RANKL),
external stress, because it is not matched by an internal counter- secretion of macrophage colony-stimulating factors (M-CSF),
stress (Anastasi et al., 2008). and through their own apoptosis at the sites of micro-damage or
-cracks (Verborgt et al., 2002; Bonewald, 2007). Gene expres­
sion analysis suggests that osteocytes control osteoclast differ­
MINERALIZED TISSUE RESPONSES
entiation indirectly through modulation of RANKL expression
TO APPLIED MECHANICAL LOADS in osteoblasts. In addition, humoral factors produced and
Alveolar Bone Osteocytes as Mechanosensors released through canaliculi into the bone marrow may regulate
the differentiation and activity of osteoclasts (Tatsumi et al.,
Bone cells (osteoblasts, osteoclasts, and osteocytes) are sensi­ 2007). It has been reported that monocytes are differentiated to
tive to their mechanical environment, and their adaptive response osteoclasts in the presence of RANKL and M-CSF (Narducci
can alter both the mass and the morphology of bones (Rubin and Nicolin, 2009). It is therefore reasonable to conclude that
et al., 2006). The important role played by these bone cells, osteocytes act as chief mechanosensors in bone, which has been
especially osteocytes, as part of mechanosensing or transducing recently confirmed by targeted ablation of osteocytes in a mouse
mechanisms in response to applied mechanical stress, has been model (Tatsumi et al., 2007).
detailed in the literature (Bonewald, 2007; Y Wang et al., 2007;
Robling et al., 2008). Osteocytes are connected to each other
Alveolar Bone Remodeling in Response to Strain
and to cells of the bone surface by cytoplasmic processes or
dendrites. Within a few minutes of the onset of mechanical loading, Applied mechanical stress causes flow of bone interstitial fluid,
glucose 6-phosphate dehydrogenase (a marker of cell metabolism) evoking shear stress in the mineralized ECM and deformation of
is elevated in osteocytes (Dodds et al., 1993), and an increase in the alveolar bone osteocytes in the lacunae and of the dendrites
c-FOS mRNA is observed within 2 hrs (Kawata and Mikuni- in the canaliculae (Fig. 1). This deformation perturbs the integ­
Takagaki, 1998). Soon thereafter, by 4 hrs, transforming growth rin molecules, which act as a tethering protein, opening
J Dent Res 88(7) 2009 Orthodontic Tooth Movement   599

hemichannels in strained osteocytes, allowing for the release of


prostaglandins and facilitating mechanotransduction (Y Wang
et al., 2007; Philips et al., 2008; Schwartz and DeSimone,
2008). Mechanical forces alter the metabolic state of osteocytes,
which in turn secrete osteotropic factors, such as MLO-Y4,
capable of stimulating surface-lining osteoblasts (Heino et al.,
2004). The importance of physical contacts between osteocytes
and surface-lining osteoblasts through gap junction intercellular
communications, formed by members of a family of proteins
known as Connexins (Cx), for a proper bone-forming response
has been stressed (Taylor et al., 2007). Researchers have
detected an increase in the release of PGE2, modulated by hemi­
channels formed by Cx43, enhancing osteoblastic activity as
part of alveolar bone response to mechanical loading (Cherian
et al., 2005; Gluhak-Heinrich et al., 2006). The outcomes of all
these structural modifications are profound changes in cytoskel­
etal organization, as evidenced by increases in alpha actin
(29%), filamin (185%), and vimentin (15%) (Jackson et al.,
2008), altering the metabolic state of the cell, activating signal­
ing molecules inside the cellular cytoplasm, as well as extracel­
lular nucleotides (i.e., ATP and UTP). This promotes a significant
stimulation of Runx2 DNA-binding activity via a mechanism
involving protein kinase C and distinct mitogen-activated pro­
tein kinase cascades (Costessi et al., 2005). Up-regulation
(BMP2, BMP6, ALP, SOX9, MSX1, and VEGFA) and down-
regulation (BMP4 and EGF) of some osteogenic specific genes
in response to cyclic tensile forces have been reported (Wescott
et al., 2007). Time-dependent up-regulation of osterix, an
osteoblast-specific transcription factor, which enhanced alkaline
phosphatase activity, and the mRNA expression of all osteo­
genic marker genes (osteopontin, bone sialoprotein, osteocalcin,
Figure 1. The sequence of bone remodeling stages during orthodontic
and collagen) have been observed (Zhao et al., 2008). The treatment. The roles played by osteocytes, osteoblasts, and osteoclasts
secreted bone matrix proteins consist of type I collagen (about are illustrated.
90% of the organic matrix), which provides strength, structure,
and elasticity to mature bone tissue. Type I collagen also regu­
lates expression/secretion of other non-collagenous proteins, inducing osteoclast recruitment and function, is initiated by
such as osteocalcin, osteopontin, osteonectin, and bone sialopro­ TNF-α (Andrade et al., 2007). The important role of M-CSF in
tein. It is apparent that bone formation is coordinated by the osteoclastogenesis, mediated by TNF-α in response to orthodon­
expression of many molecules, such as growth factors, tran­ tic force application, has been demonstrated (Kitaura et al.,
scription factors, and anti-inflammatory cytokines. Committed 2008). These findings strongly suggest that TNF-α plays a piv­
osteoblasts induce/stimulate osteoclastogenesis through the otal role in the bone resorption process that facilitates orthodon­
RANK/RANKL/OPG pathway, or become highly differentiated tic tooth movement.
cells, persisting for a very long period of time inside the bone Bone degradation and terminal osteoclastogenesis require
matrix as osteocytes (Canalis, 2005). physical contact between osteoclasts and bone matrix. Once dif­
In physiologic conditions, RANKL is released only by osteo­ ferentiated, the capacity of a matrix osteoclast to resorb bone
blasts, but in cases of inflammation (e.g., rheumatoid arthritis, depends on its ability to synthesize and mobilize a series of
orthodontic tooth movement, periodontitis), these molecules are degradative enzymes. The way osteoclasts perform the bone
produced in abundance by T-lymphocytes (Kawai et al., 2006). degradation process has been well-reviewed, and readers are
In addition to RANKL, TNF-α has also been shown to promote referred to these excellent articles for details (Teitelbaum and
osteoclastogenesis in conditions of inflammatory osteolysis Ross, 2003; Teitelbaum, 2007; Kreja et al., 2008).
(Horowitz et al., 2001). This effect is achieved either by stimu­
lation of RANKL release from osteoblasts, or through direct
activation of osteoclast precursors. TNF-α apparently induces NON-MINERALIZED TISSUE RESPONSES
basal osteoclastogenesis via TNF receptor type 1 (p55), while TO MECHANICAL LOADING
suppressing it via TNF receptor type 2 (p75), as has been
Fibroblasts as Mechanosensors and Transducers
demonstrated in a tooth movement model (Abu-Amer et al.,
2000). Synthesis of other inflammatory mediators and chemo­ The changes elaborated in connective tissues in response
kines such as RANTES/CCL5 and MCP-1/CCL2, capable of to mechanical loading are largely mediated through ­fibroblasts.
600  Krishnan & Davidovitch J Dent Res 88(7) 2009

mastication) and long duration (during orthodontic treatment).


In the PDL, integrin binds to fibronectin in the ECM and to
talin intracellularly, as part of establishing a signal transduction
pathway (Sandy, 1998). Applied mechanical forces stretch the
fibronectin molecules in ECM at sites of PDL tension, straight­
ening the initial nodular assembly with quaternary structure, and
unfolding fibronectin type III molecules. The signals transferred
to the inside of the fibroblast result in re-organization of the
cell’s cytoskeleton, leading to changes in cell shape and mobil­
ity, induction of gene expression in the nucleus, and secretion
of newly synthesized proteins, such as fibronectin and collagen
(Smith et al., 2007). A quantity of these newly synthesized colla­
gen fibers is incorporated into the newly formed osteoid, whereas
the rest are embedded in the PDL to form new Sharpey’s fibers
(Meikle, 2006). It has been demonstrated that, after orthodontic
force application, the density of cells expressing positive signals
for type I collagen mRNA is greater in the PDL in tension sites,
followed by expression of type XII collagen (Nakagawa et al.,
1994). Synthesis of all other collagen types has also been dem­
onstrated (types III, V, and VI) in both tension and compression
sites (Bumann et al., 1997). With recognition of several binding
sites, the response of fibronectin to mechanical force is impor­
tant in the mechanosensing and transduction pathway, leading to
ECM remodeling (Smith et al., 2007; Katanosaka et al., 2008).
Degradation of collagen and other macromolecules in the ECM
Figure 2. The responses of periodontal ligament fibroblasts to orthodon- as part of the force-induced PDL remodeling is performed by
tic forces. several enzymes, such as serine proteases, aspartate proteases,
cysteine proteases, and the matrix metalloproteinases (MMPs;
Fibroblasts are considered to be mechanoresponsive, in that collagenase, gelatinase, and stromelysin). Specific up-regulation
the mechanical signals transmitted from the ECM via integrin of MMP-8 and -13 mRNA in PDL compression and tension
receptors influence their morphology, cytoskeletal organiza­ sites during orthodontic tooth movement has been demonstrated
tion, proliferation, differentiation, and gene expression (JH (Takahashi et al., 2003), stressing the role of ECM degradation
Wang et al., 2007). Proteins located at focal adhesion kinases in the remodeling process (Fig. 2).
(FAK) in the inner cell surface act as strain gauges that
‘sense’ stress from the ECM, as well as strain generated by Gingival Fibroblasts and Mechanical Forces
cytoskeletal re-organization. These stretch-induced confor­ Gingival fibroblasts play an important role in determining tissue
mational changes might expose previously hidden phospho­ architecture, by virtue of their attachment to the acellular
rylation sites (Wang et al., 2001; Smith et al., 2007; Li et al., cementum in the cervical part of the dental root, and to the gin­
2008). These newly modified sites are recognized by other gival fibrous matrix at the other end of the fibers (Danciu et al.,
signaling molecules (growth factors and cytokines), which 2004; Grünheid and Zentner, 2005). It has been proposed that if
in turn activate small GTPases of the Rho and Ras families, there is any disruption in the chemico-physical connectivity of
which have multiple effects on cytoskeletal dynamics, gene the marginal gingiva to the dental root, the intracellular tensile
transcription, and cell division and differentiation (Jaffe and forces of gingival fibroblasts are abruptly lowered, changing the
Hall, 2005). Thus, focal adhesions, together with the actin balanced physiological strains existing in the cells (tensegrity)
cytoskeleton, have been suggested to serve as mechanosen­ (Binderman et al., 2002, 2007). In response, molecular changes
sors in ­fibroblasts. may occur, activating propagation of intracellular messengers
that start remodeling responses in the gingiva, as well as in the
alveolar bone. In this situation, the marginal gingiva acts as a
Fibroblasts in the Mechanically Stressed
mechanosensor apparatus, recognizing and responding to abrupt
Periodontal Ligament
modifications in physical stimuli. Once intracellular and cell-
Various investigators studied the mechanoresponse by gene ECM strains are lowered, a molecular signaling pathway, with a
expression in PDL fibroblasts (He et al., 2004; Kim et al., local release of ATP, and up-regulation of a specific p2x4
2007; Ritter et al., 2007). PDL fibroblasts exist in an active purinoreceptor in the gingival fibroblasts are initiated. These
mechanical environment, filling the gap between the dental root events lead to an increase in intracellular calcium influx, trans­
­
cementum and the alveolar bone, and are periodically subjected ducing extracellular signals to the interior, and subsequently to
to compressive, tensile, and shearing forces of both short (during the genome (Fig. 3).
J Dent Res 88(7) 2009 Orthodontic Tooth Movement   601

Blood Vessel Re-organization and Neovascularization


in Orthodontic Tooth Movement
Blood vessels in the PDL, which provide gases and nutrients
required for all metabolic activities of their surrounding tissues,
in health and disease, are active participants in the tissue remod­
eling associated with orthodontic treatment. Furthermore, these
vessels usher immune cells into the strained PDL, extending a
helping hand for their role in the tissue-remodeling process.
They also permit migration into the strained paradental tissues
of hormones and a variety of molecules derived from food and
drugs consumed by the individual. All these molecules—those
that originate from native paradental cells or migratory leuko­
cytes, or from remote endocrine glands, or from consumed
nutrients and drugs—may have profound and long-lasting
effects on the nature of the force-induced tissue remodeling and
the degree of success of orthodontic treatment (Krishnan and
Davidovitch, 2006b).
The ECM surrounding the vessels provides critical support
for the vascular endothelium. Adhesion of the endothelial cell to
the ECM is required for its proliferation, migration, morphogen­
esis, survival, and blood vessel stabilization, to maintain tissue
viability and neovascularization (Davis and Senger, 2005).
During angiogenesis, with the help of the ECM (which serves
as a three-dimensional malleable scaffold), proliferating and
migrating endothelial cells organize to form new three-
dimensional capillary networks. By generating mechanical con­
tractile forces within the ECM, endothelial cells, which acquire
Figure 3. The gingival fibroblastic activity in response to orthodontic
shape changes with the help of the COL1A1 gene, establish
forces.
tension-based guidance pathways that allow them to form inter­
connected cords (Shyy and Chien, 2002; Chien, 2008). A colla­
transmission electron-microscopic observations, Anastasi et al.
gen type I gene is involved in acquiring shape changes by the
(2008) showed a reduction in the number of blood vessels after 72
endothelial cells. Over the course of several days, intercon­
hrs of treatment and an increase in their number after 7 days of
nected cords mature to form hollow tubes through a process
treatment. Modifications of immunofluorescence staining pat­
involving the development and coalescence of intracellular
terns of tested proteins revealed angiogenesis and reparative pro­
vacuoles (Zhou et al., 2006). The key angiogenic cytokine, vas­
cesses, along with thickening of fibrillar matrix, as a defensive
cular endothelial growth factor (VEGF), which induces sprout­
reply to mechanical stress. Through a quantitative representation
ing angiogenesis, also induces microvascular endothelial cells to
of PDL vascular reactions in rats, Ren et al. (2008) discovered
express integrins (Nör et al., 1999). The sequence of neovascu­
that prolonged force applications lead to increased vascularity,
larization steps is outlined below (Davies and Senger, 2005):
which is age-related. In young rats, increased numbers of both
small and large blood vessels were found, while adult rats exhib­
• Collagen type I ligation to integrins in endothelial cells ited an increase in the number of only small blood vessels. These
suppresses cAMP, thereby suppressing activity of cAMP- results confirm earlier findings on the responses of blood vessels
mediated protein kinase A (PKA). to applied mechanical forces, typified by increased permeability,
• PKA activity suppression leads to a marked induction of which enhances fluid extravasation from capillaries into the inter­
actin polymerization, leading to the formation of promi­ stitial space (Cooper and Sims, 1989; Lew, 1989).
nent stress fibers and endothelial cell contractility.
• Endothelial cell organization into multicellular pre-capil-
Neural Responses to Mechanical Forces
lary cords with activation of Src kinases and GTPase Rho
through interaction with integrins, which also results in Innervations of dental and paradental tissues are mainly constituted
cord formation. by neurons that originate in the trigeminal ganglia (located in the
cervical and middle root areas) and mesencephalic trigeminal
Applied mechanical stresses develop strain and activate signal­ nucleus (apical periodontal area). The nerve endings consist mainly
ing pathways, mainly through integrin-dependent mechanisms, fol­ of mechanoreceptors (Ruffini-like endings) and nociceptors. Under
lowed by actin polymerization, activation of type I collagen genes, normal physiological conditions, these receptors remain quiescent,
neovascularization, and remodeling of blood vessels (Fig. 4). The but contain various neuropeptides, such as calcitonin gene-related
entire process is self-limiting, once sufficient changes are obtained peptide (CGRP) and substance P (SP) (Norevall et al., 1995;
to nourish the associated structures. With the help of light and Vandevska-Radunovic, 1999; Hall et al., 2001; Yamaguchi et al.,
602  Krishnan & Davidovitch J Dent Res 88(7) 2009

not uniform throughout the applied region,


but rather develop a ‘quilt’ of areas where
tension or compression prevails, creating
favorable conditions for tissue remodeling.
Great clinical interest exists regarding the
behavior of cells in these strained locations,
since it determines the orthodontic treat­
ment outcome. An insight into this puzzle
may be obtained from investigations on the
responses of paradental cells to conditions
of tension, compression, or torque. It has
been reported that PDL cells respond differ­
ently to tensile and compressive strains, in
Figure 4. Blood vessel response to orthodontic forces. terms of synthesis and degradation of ECM
components (He et al., 2004). A 10% com­
2004). Orthodontic tooth movement alters the neutral state of both pressive strain (0.5 Hz) decreased collagen (COL1A1) mRNA,
mechanoreceptors and nociceptive PDL nerve fibers (Fig. 5), type I collagen, and fibronectin levels in these cells. In contrast,
effecting the release of biologically active proteins, leading to local a 10% tensile strain increased COL1A1 mRNA, MMP2 mRNA,
neurogenic inflammation and the release of neuropeptides such as and TIMP2 mRNA. At the same time, tensile forces result in the
CGRP and SP (Nicolay et al., 1991; Norevall et al., 1995; synthesis of similar levels of both MMP and TIMP, promoting
Vandevska-Radunovic, 1999). Since most PDL neurons are anabolic activity, whereas synthesis of MMP is increased under
wrapped around blood vessels, including capillaries, endothelial compressive loads without a change in TIMP, thus promoting
cells are probably the first to interact with neuropeptides, which in degradation. Recently, an increase in type I collagen immunofluo­
turn bind circulating leukocytes, facilitating their migration from rescence staining intensity was demonstrated in PDL pressure
the capillaries. Chemokines activate integrins on the leukocyte cell sites, while tension sites displayed a diminution of staining after
surface to promote firm adhesion. The migration of leukocytes by 72 hrs of treatment. Type IV collagen staining was reduced in
the chemoattractant gradient and their arrival in the PDL signify the both sites, increasing gradually after 7 days of treatment (Anastasi
onset of an acute inflammation, which is essential for tooth move­ et al., 2008).
ment into newer locations (Middleton et al., 2002). Signaling mol­ Orthodontic forces induce an aseptic inflammatory response,
ecules released by these migrating leukocytes (cytokines, growth rather than a reaction to an invasion by micro-organisms through
factors, and colony-stimulating factors) interact with various dental a wound. Nonetheless, the classic signs of inflammation (red­
and paradental cells and stimulate them to initiate and sustain the ness, swelling, pain, and reduced function) are similar in both
tissue remodeling (Yamaguchi et al., 2004). Monocytes, lympho­ cases. Inflammatory cytokines produced by leukocytes, plate­
cytes, and mast cells express receptors for neuropeptides, which lets, and native cells, which include lymphocyte- and monocyte-
transduce signals intracellularly and evoke cellular responses, such derived factors, colony-stimulating factors, growth factors, and
as cytokine release, production of other neuropeptides, changes in chemotactic chemokines, are often involved in tissue reactions
the expression of mediators, or direct release of inflammatory associated with orthodontic forces (Ren and Vissink, 2008). In
mediators (Yamaguchi et al., 2004; Lee et al., 2007). Moreover, periodontal tissues, cytokines are produced by immune cells,
CGRP and SP also serve as vasodilators, increasing vascular per­ fibroblasts, and osteoblasts, and participate in regular tissue
meability, flow, and plasma extravasations. Often, this plasma turnover and in induced bone-remodeling events (Mundy, 1992).
contains molecules derived from the diet, medications, remedies, Both these procedures are mediated by pro- as well as anti-
drugs, hormones, and inflammatory mediators that originated in inflammatory cytokines under the influence of distinct cytokine
remote diseased organs, which might influence the course of subsets (Andrade et al., 2007; Maeda et al., 2007). Elevated
mechanotherapy, by virtue of their interaction with cells (Krishnan levels of IL-1β, SP, and PGE2 in gingival crevicular fluid from
and Davidovitch, 2006b). both compression and tension sites of moving teeth have been
In addition to this peripheral action, the affected neurons identified, with higher concentrations found in tension sites
rel­ease neurotransmitters centrally, in the trigeminal nucleus, (Dudic et al., 2006). Recently, the differential expression patterns
causing pain shortly after appliance activations in persons of 3 cytokines (TGF-β, IL-10, and TNF-α) in PDL compression
undergoing ortho­­don­tic treatment (Krishnan, 2007). The short and tension sites during tooth movement have been reported
delay in the onset of pain is due to the initial resistance of the (Ren and Vissink, 2008). An augmented expression pattern for
tissue fluids. The gradual move­ment of these fluids from areas TGF-β mRNA has been reported at sites of both PDL compres­
of com­­­­­pression to zones of tension facilitates the development sion and tension, which induces proliferation and chemotaxis of
of ECM strain and the entire process of ­mechanotransduction. PDL cells, up-regulates COL-I, recruits osteoblast precursors,
inducing their differentiation, down-regulates MMPs, and up-
Cellular and Molecular Behavior in Sites of PDL Tension regulates TIMPs, enhancing production of bone matrix proteins
and Compression (Ignotz et al., 1987). In areas of compression, TGF-β inhibits
recruitment of osteoclast precursor cells, suppressing osteoclas­
Orthodontic forces (continuous, interrupted, or intermittent) cre­ tic activity (Kanaan and Kanaan, 2006). Compression-site PDL
ate strains in the teeth and paradental tissues. These strains are cells have been reported to express increased amounts of TNF-α,
J Dent Res 88(7) 2009 Orthodontic Tooth Movement   603

which stimulate the production of MMPs, elevating the levels of


RANKL to be directly involved in bone resorption (Andrade
et al., 2007). In contrast, simultaneous up-regulation of
OPG and down-regulation of RANKL expression by the anti-
inflammatory cytokine IL-10 inhibit bone resorption (Ren and
Vissink, 2008). The dual role of cytokines in the remodeling of
mineralized and non-mineralized connective tissues can be
described as that of a pro-inflammatory cytokine, such as IL-1
and TNF-α, which promotes resorption and inhibits apposition,
while anti-inflammatory cytokines promote apposition and
inhibit resorption.
Chemokines, the superfamily of chemotactic cytokines, are
small structurally related heparin-binding proteins classified
into 4 subfamilies based on the configuration of cysteine resi­
dues near the N-terminal, depending on whether the first 2
cysteines are separated (CXC, CX3C) or not (CC, C). They are
synthesized by a variety of cell types, such as endothelial, epi­
thelial, and stromal cells, fibroblasts, mast cells, bone cells, and
leukocytes. Functionally, chemokines are divided into homeo­
static and inflammatory molecules, which target all types of
leukocytes and lymphocytes through their binding to selective
G-protein-coupled receptors. The activation of these receptors
triggers several intracellular signaling pathways, resulting in
re-organization of the cytoskeleton and cell adhesion, causing
cells to extend pseudopodia and crawl up the chemoattractant
gradient (Terricabras et al., 2004). Chemokine-driven cell Figure 5. Neural response to orthodontic forces.
migration is thought to be an important step in orthodontic-
force-induced paradental tissue remodeling, especially in bone
remodeling and angiogenic events (Krishnan and Davidovitch, PF4, CXCL9/Mig, CXCL10/IL-10, CXCL11/I-TAC, and
2006a; Masella and Meister, 2006; Meikle, 2006). Osteoclast CXCL14/BRAK) activities associated with any sort of chronic
precursors express chemokine receptors such as CCR2 and inflammation (Rosenkilde and Schwartz, 2004). Hence, chemo­
CCR5 and signals provided by chemokines, such as CCL2/ kines appear to emerge as important factors, in more than one
MCP-1, CCL3/MIP-1α, and CXCL12/SDF-1, which are essen­ way, in the remodeling of tissues subsequent to the application
tial for the differentiation of osteoclast progenitors into mature of orthodontic forces.
osteoclasts. High expression patterns have been demonstrated It can be concluded that orthodontic forces are capable
for MCP-1, CCL5/RANTES, and MIP-2 in bone resorption of creating aseptic and transitory inflammation at both PDL
areas associated with PDL compression (Alhashimi et al., 1999; ­compression and tension sites. Although a clear-cut microscopic
Garlet et al., 2008). Osteoblasts also express receptors, such as and/or computerized tomographic demarcation between areas
CXCR4, CXCR5, and CCR5, which function by binding to under compression and tension is not always possible, because
specific chemokines, such as SDF-1 and BCA-1/CXCL13, these zones often overlap (Melsen et al., 2007), the bulk of
resulting in proliferation and type I collagen mRNA expression research outcomes to date points to the fact that the reaction at
(Lisignoli et al., 2006). Moreover, osteoblasts can secrete each of these areas differs clearly on the molecular and cellular
chemokines such as MCP-1, SDF-1 (involved in bone remodel­ levels. Future research will continue to increase our knowledge
ing), KC/CXCL1, LIX/CXCL5, CINC-1/CXCL1, and BCA-1/ about this dichotomy.
CXCL13 (involved in the recruitment of different leukocytes) in
response to mechanical force application. These findings sug­
HOW DO TEETH MOVE ORTHODONTICALLY?
gest an interesting role for osteoblasts in the development of
aseptic inflammatory reaction in the PDL following orthodontic Orthodontic forces strain ECM and cells of the alveolar bone,
force application (Garlet et al., 2008). Chemokines, the known PDL, gingiva, and associated blood vessels and neural elements,
regulators of cell-trafficking and tissue remodeling, play a cru­ the initial effect of which is physical in nature, followed closely
cial role in the migration of cells into specific organs and tissues by a biological response. This interaction generates profound
(Rossi and Zlotnik, 2000). In this capacity, chemokines initiate changes in the structure and function of the ECM, cell mem­
signal transduction events, leading to other biological processes brane, cytoskeletal elements, nucleus, and several other cyto­
such as angiogenesis, cell proliferation, apoptosis, and host plasmic organelles that synthesize and mobilize a variety of
defense (Silva et al., 2007). Chemokines of the CXC family are molecules inside and outside the cells (Kerrigan et al., 2000;
unique in promoting angiogenic (CXCL1/GROα, CXCL2/ Masella and Meister, 2006; Meikle, 2006). Cell adhesion mole­
GROβ, CXCL3/GROγ, CXCL5/ENA 78, CXCL6/GCP-2, cules, like the integrins, transmit tensile, compressive, and shear
CXCL7/NAP-2, and CXCL8/IL-8) and angiostatic (CXCL4/ stresses directly from the ECM into the cell and vice versa, and
604  Krishnan & Davidovitch J Dent Res 88(7) 2009

by which mechanical energy is converted to bio­


chemical and/or electrical signals. In addition, signal­
ing proteins generated in the cell cytoplasm, such as
hedgehog and transforming growth factors, as well as
calcium ions, reach the nuclear matrix and, ultimately,
the genome. The outcome of the entire chain of events
is enhanced or suppressed gene expression, transfer­
ring the signal back to the cytoplasm through mRNAs,
reaching the ribosomes, generating protein synthesis
and secretion, mitosis, cell motility, and programmed
cell death (Hamill and Martinac, 2001; Silver and
Siperko, 2003; Masella and Meister, 2006) (Fig. 6).
The unique feature of this scheme in orthodontics
is the interaction between various tissues, both min­
eralized and non-mineralized, and their associated
neuro­vascular elements, for effecting tooth move­
ment. For such an event to happen, remodeling of var­
ious tissues—performed by cells of the alveolar bone
(osteoblasts, osteoclasts, and osteocytes), PDL, and
gingival fibroblasts, blood vessels (endothelial cells),
and neural tissues (dendritic and neural cells)—must
occur. Additional cell types essential to the body’s
response to applied mechanical loads are derivatives
of the immune system (inflammatory cells and osteo­
clast progenitors). Each of these tissues follows its
own response pattern, as far as the mechanosensing,
transduction, and response mechanisms are concerned
(Fig. 7) (Krishnan and Davidovitch, 2006a; Masella
and Meister, 2006; Meikle, 2006; Henneman et al.,
2008; Wise and King, 2008).
Orthodontic for­ces bend the alveolar bone, and
compress and stretch the PDL. Consequently, there will
be an alteration in the electrical neu­­­trality of the alveo­
lar bone, as the compressed PDL becomes the site of
intense bone resorp­tion, while stretched PDL areas
interface with sites of active osteogenesis. Osteo­­cytes
are key participants in this process, being acu­tely sensi­
tive and responsive to applied mechanical loads. Their
elaborate network of cellular projections facilitates
Figure 6. The sequence followed by cells and tissues in mechanosensing, transduc-
tion, and response.
com­munications with neighboring osteo­cytes, as well
as with alveolar bone surface-lining cells and bone
marrow cavity cells. Osteoblasts, which maintain direct
are considered essential for cellular survival, growth, and mobil­ contact with osteocytes, respond to these signals and initiate
ity. This transmission helps to maintain the cells in active form appositional changes. The activated osteoblasts proceed to convey
(tensegrity), capable of responding rapidly to various mechani­ signals to approaching osteoclasts, enticing these cells to start
cal, physical, and other challenges. Mechanosensing is a process resorbing the alveolar bone, and informing them on the proper
by which cells sense structural changes in the ECM, caused by time to cease their resorptive activities. The origin of osteoclasts
external mechanical loading. This sensing triggers biochemical for bone resorption is attributed to either conversion of mono­
reactions that generate energy, in addition to the energy pro­ cytes/macrophages or migration of progenitor cells from alveolar
vided by the mechanical load, required for the response to the bone marrow cavities to the strained PDL (Xie et al., 2008).
elevated environmental demands (Apodaca, 2002; Poirier and Important components of this osteocyte→osteoblast→osteoclast
Iglesias, 2007). chain are PGs, TNF-α, and the RANK/RANKL/OPG system.
Each cellular system in paradental tissues is equipped with Gingival and periodontal fibroblasts play different roles in
mechanosensors; thus, each mechanical stimulus may activate paradental tissue remodeling. While PDL fibroblasts are pre­
multiple mechanosensors, followed by downstream cellular dominantly involved in the synthesis and degradation of their
events. Subsequent changes in cytoskeletal protein structure and own ECM, gingival fibroblasts participate in bone remodeling
function propagate the signaling process into the nucleus by a events. Up-regulation of 85 genes and down-regulation of 23
process known as mechanotransduction, defined as the process genes associated with protein synthesis in the PDL have been
J Dent Res 88(7) 2009 Orthodontic Tooth Movement   605

Figure 7. The sequence of orthodontic tooth movement, illustrating the roles played by mineralized and non-mineralized tissues along with the
associated blood vessels and neural elements.

identified (de Araujo et al., 2007). Diminution of collagen pro­ (Koyama et al., 2008). Substance P, CGRP, VIP, and other
duction (type I and IV) in the compressed PDL has been released neurotransmitters interact with endothelial cells of the
reported, as well as increased type IV collagen production at paradental capillary network, enticing them to bind circulating
PDL tension sites after 72 hrs of force application (Anastasi leukocytes, promoting their migration into the paradental ECM.
et al., 2008). Upon entering these tissues, the migratory leukocytes produce
The role played by blood vessels in orthodontic tooth move­ ample amounts of chemokines (Andrade et al., 2007; Maeda
ment has received much recent attention. Angiogenesis and et al., 2007) and cytokines (Yamaguchi et al., 2006, 2008).
remodeling of existing blood vessels help them adapt to the new Along with the cytokines produced by the native paradental
environment created by mechanical forces. An initial reduction cells (fibroblasts and osteoblasts), they evoke and maintain the
and a later increase in the number of PDL blood vessels follow­ remodeling events of the PDL and alveolar bone (Garlet et al.,
ing orthodontic force application have been reported (Anastasi 2007). The released neuropeptides also act centrally to produce
et al., 2008; Ren et al., 2008). These blood vessels play a major pain, which in turn restricts the use of the jaws and teeth by the
role in the mechanical force-induced aseptic inflammation by individual, whether for mastication or speech. These reduced
acting as sources of cytokines and chemokines. Increased movements of contraction and relaxation of the PDL might pro­
expression of IL-1β, IL-1 receptor, IL-6, IL-6 receptor, IL-8 voke a loss of transmission of mechanical stresses to the liga­
receptor, IL-11, and TNF-α has been demonstrated in the com­ ment, delaying the cellular responses due to the state of
pressed PDL, confirming the presence of aseptic inflammation inactivity and loss of tensegrity. Increasing the magnitude and
606  Krishnan & Davidovitch J Dent Res 88(7) 2009

frequency of functional force application to orthodontically Canalis E (2005) The fate of circulating osteoblasts. N Engl J Med
treated teeth may restore the pace of tooth movement during 352:2014-2016.
Canalis E, Economides AN, Gazzerro E (2003). Bone morphogenetic pro­
these painful periods. A similar effect can be achieved by teins, their antagonists, and the skeleton. Endocr Rev 24:218-235.
physical, chemical, or surgical means. This phenomenon further Cherian PP, Siller-Jackson AJ, Gu S, Wang X, Bonewald LF, Sprague E,
emphasizes the fact that physical forces of gravity, hemody­ et al. (2005). Mechanical strain opens connexin 43 hemichannels in
namic stresses, and movement play a critical role in tissues, osteocytes: a novel mechanism for the release of prostaglandin. Mol
since the cells use tensegrity architecture for their structural Biol Cell 16:3100-3106.
Chien S (2008). Role of shear stress direction in endothelial mechanotrans­
organization (Anastasi et al., 2008). duction. Mol Cell Biomech 5:1-8.
It can be concluded that orthodontic tooth movement is pro­ Cooper SM, Sims MR (1989). Evidence of acute inflammation in the perio­
duced by mechanical means that evoke biological responses. dontal ligament subsequent to orthodontic tooth movement in rats. Aust
These two entities, mechanics and biology, act in concert to Orthod J 11:107-109.
Costessi A, Pines A, D’Andrea P, Romanello M, Damante G, Cesaratto L,
produce desirable and predictable alterations in the form and
et al. (2005). Extracellular nucleotides activate Runx2 in the osteoblast-
function of the dento-alveolar complex. The actual performers like HOBIT cell line: a possible molecular link between mechanical
of this force-induced remodeling are the native cells of the stress and osteoblasts’ response. Bone 36:418-432.
treated teeth and their surrounding tissues. Other important cel­ Danciu TE, Gagari E, Adam RM, Damoulis PD, Freeman MR (2004).
lular participants in this remodeling process are derivatives of Mechanical strain delivers anti-apoptotic and proliferative signals to
gingival fibroblasts. J Dent Res 83:596-601.
the neuro-vascular and immune systems. Cells and tissues use Davis GE, Senger DR (2005). Endothelial extracellular matrix: biosynthe­
mechanosensing, transduction, and response phenomena to sis, remodeling, and functions during vascular morphogenesis and
respond to applied mechanical forces. This reaction is typified neovessel stabilization. Circ Res 97:1093-1107.
by the aseptic inflammatory reaction, which is initially acute, de Araujo RM, Oba Y, Moriyama K (2007). Identification of genes related
becoming chronic a few days after the activation of the ortho­ to mechanical stress in human periodontal ligament cells using microar­
ray analysis. J Periodontal Res 42:15-22.
dontic appliance. Acute inflammation is re-introduced to the Dodds RA, Ali N, Pead MJ, Lanyon LE (1993). Early loading-related
paradental tissues each time the appliance is reactivated. Hence, changes in the activity of glucose 6-phosphate dehydrogenase and
it may be suggested that mechanoresponse and inflammation are alkaline phosphatase in osteocytes and periosteal osteoblasts in rat fibu­
both essential for achieving the clinical effect of tooth move­ lae in vivo. J Bone Miner Res 8:261-267.
ment. If both mechanisms indeed unfold in concert, orthodon­ Dudic A, Kiliaridis S, Mombelli A, Giannopoulou C (2006). Composition
changes in gingival crevicular fluid during orthodontic tooth move­
tists might be able to accelerate the velocity of tooth movement ment: comparisons between tension and compression sides. Eur J Oral
by utilizing additional external stimuli, whether physical, chem­ Sci 114:416-422.
ical, or surgical. Knowledge pertaining to the subject of mechan­ Garlet TP, Coelho U, Repeke CE, Silva JS, Cunha Fde Q, Garlet GP (2008).
ics and biology in orthodontics is expanding constantly, creating Differential expression of osteoblast and osteoclast chemoattractants in
compression and tension sides during orthodontic movement. Cytokine
an ever-stronger basis for success in the clinical realm.
42:330-335.
Gluhak-Heinrich J, Ye L, Bonewald LF, Feng JQ, MacDougall M, Harris
SE, et al. (2003). Mechanical loading stimulates dentin matrix protein
REFERENCES 1 (DMP1) expression in osteocytes in vivo. J Bone Miner Res 18:
807-817.
Abu Amer Y, Erdmann J, Alexopoulou I, Kollias G, Ross FO, Teitelbaum SL Gluhak-Heinrich J, Gu S, Pavlin D, Jiang JX (2006). Mechanical loading
(2000). Tumor necrosis factor receptors type 1 and 2 differentially stimulates expression of connexin 43 in alveolar bone cells in the tooth
regulate osteoclastogenesis. J Biol Chem 275:27307-27310. movement model. Cell Commun Adhes 13:115-125.
Alhashimi N, Frithiof I, Bridvik P, Bakhiet M (1999). Chemokines are Gluhak-Heinrich J, Pavlin D, Yang W, MacDougall M, Harris SE (2007).
upregulated during orthodontic tooth movement. J Interferon Cytokine MEPE expression in osteocytes during orthodontic tooth movement.
Res 19:1047-1052. Arch Oral Biol 52:684-690.
Anastasi G, Cordasco G, Matarese G, Rizzo G, Nucera R, Mazza M, et al. Goulet GC, Cooper DM, Coombe D, Zernicke RF (2008). Influence of
(2008). An immunohistochemical, histological, and electron-micro­ cortical canal architecture on lacunocanalicular pore pressure and fluid
scopic study of the human periodontal ligament during orthodontic flow. Comput Methods Biomech Biomed Engin 11:379-387.
treatment. Int J Mol Med 21:545-554. Grünheid T, Zentner A (2005). Extracellular matrix synthesis, proliferation
Andrade I, Silva TA, Silva GAB, Teixeira AL, Teixeira MM (2007). The role and death in mechanically stimulated human gingival fibroblasts in
of tumor necrosis factor type 1 in orthodontic tooth movement J Dent vitro. Clin Oral Investig 9:124-130.
Res 86:1089-1094. Hall M, Masella R, Meister M (2001). PDL neuron-associated neurotrans­
Apodaca G (2002). Modulation of membrane traffic by mechanical stimuli. mitters in orthodontic tooth movement: identification and proposed
Am J Physiol Renal Physiol 282:179-190. mechanism of action. Today’s FDA 13:24-25.
Bakker AD, Soejima K, Klein-Nulend J, Burger EH (2001). The production Hamill OP, Martinac B (2001). Molecular basis of mechanotransduction in
of nitric oxide and prostaglandin E (2) by primary bone cells is shear living cells. Physiol Rev 81:685-674.
stress dependent. J Biomech 34:671-677. He Y, Macarak EJ, Korostoff JM, Howard PS (2004). Compression and ten­
Binderman I, Bahar H, Yaffe A (2002). Strain relaxation of fibroblasts in the sion: differential effects on matrix accumulation by periodontal liga­
marginal periodontium is the common trigger for alveolar bone resorp­ ment fibroblasts in vitro. Connect Tissue Res 45:28-39.
tion: a novel hypothesis. J Periodontol 73:1210-1215. Heino TJ, Hentunen TA, Vaananen HK (2004). Conditioned medium from
Binderman I, Bahar H, Jacob-Hirsch J, Zeligson S, Amariglio N, Rechavi G, osteocytes stimulates the proliferation of bone marrow mesenchymal
et al. (2007). P2X4 is up-regulated in gingival fibroblasts after perio­ stem cells and their differentiation into osteoblasts. Exp Cell Res
dontal surgery. J Dent Res 86:181-185. 294:458-468.
Bonewald LF (2007). Osteocytes as dynamic multifunctional cells. Ann NY Henneman S, Von den Hoff JW, Maltha JC (2008). Mechanobiology of tooth
Acad Sci 1116:281-290. movement. Eur J Orthod 30:299-306.
Bumann A, Carvalho RS, Schwarzer CL, Yen EH (1997). Collagen synthe­ Horowitz MC, Xi Y, Wilson K, Kacena MA (2001). Control of osteoclasto­
sis from human PDL cells following orthodontic tooth movement. Eur genesis and bone resorption by members of the TNF family of receptors
J Orthod 19:29-37. and ligands. Cytokine Growth Factor Rev 12:9-18.
J Dent Res 88(7) 2009 Orthodontic Tooth Movement   607

Ignotz RA, Endo T, Massagué J (1987). Regulation of fibronectin and type Masella RS, Meister M (2006). Current concepts in the biology of orthodon­
I collagen mRNA levels by transforming growth factor-beta. J Biol tic tooth movement. Am J Orthod Dentofacial Orthop 129:458-468.
Chem 262:6443-6446. Meikle MC (2006). The tissue, cellular, and molecular regulation of orth­
Ingber DE (2006). Cellular mechanotransduction: putting all the pieces odontic tooth movement: 100 years after Carl Sandstedt. Eur J Orthod
together again. FASEB J 20:811-827. 28:221-240.
Ingber DE (2008). Tensegrity-based mechanosensing from macro to micro. Melsen B, Cattaneo PM, Dalstra M, Kraft DC (2007). The importance of
Prog Biophys Mol Biol 97:163-179. force levels in relation to tooth movement. Semin Orthod 13:220-233.
Jackson WM, Jaasma MJ, Tang RY, Keaveny TM (2008). Mechanical load­ Middleton J, Patterson AM, Gardner L, Schmutz M, Ashton BA (2002).
ing by fluid shear is sufficient to alter the cytoskeletal composition of Leukocyte extravasation: chemokine transport and presentation by the
osteoblastic cells. Am J Physiol Cell Physiol 295:1007-1015. endothelium. Blood 100:3853-3860.
Jaffe AB, Hall A (2005). Rho GTPases: biochemistry and biology. Annu Rev Mundy GR (1992). Cytokines and local factors which affect osteoclast func­
Cell Dev Biol 21:247-269. tion. Int J Cell Cloning 10:215-222.
Kanaan RA, Kanaan LA (2006). Transforming growth factor-beta 1, bone Nakagawa M, Kukita T, Nakasima A, Kurisu K (1994). Expression of the
connection. Med Sci Monit 12:164-169. type I collagen gene in rat periodontal ligament during tooth movement
Katanosaka Y, Bao JH, Komatsu T, Suemori T, Yamada A, Mohri S, et al. as revealed by in situ hybridization. Arch Oral Biol 39:289-294.
(2008). Analysis of cyclic-stretching responses using cell-adhesion- Narducci P, Nicolin V (2009). Differentiation of activated monocytes into
patterned cells. J Biotechnol 133:82-89. osteoclast-like cells on a hydroxyapatite substrate: an in vitro study.
Kawai T, MatsuyamaT, Hosokawa Y, Makihira S, Seki M, Karimbux NY, Ann Anat (in press).
et al. (2006). B and T lymphocytes are the primary sources of RANKL Nicolay O, Shanfeld J, Davidovitch Z, Alley K (1991). SP immunoreactivity
in the bone resorptive lesion of periodontal disease. Am J Pathol 169: in the dental pulp and periodontium during tooth movement. Ann NY
987-998. Acad Sci 632:452-454.
Kawata A, Mikuni-Takagaki Y (1998). Mechanotransduction in stretched Nör JE, Christensen J, Mooney DJ, Polverini PJ (1999). Vascular endothe­
osteocytes—temporal expression of immediate early and other genes. lial growth factor (VEGF)-mediated angiogenesis is associated with
Biochem Biophys Res Commun 246:404-408. enhanced endothelial cell survival and induction of Bcl-2 expression.
Kerrigan JJ, Mansell JP, Sandy JR (2000). Matrix turnover. J Orthod Am J Pathol 154:375-384.
27:227-233. Norevall LI, Forsgren S, Matsson L (1995). Expression of neuropeptides
Kim HJ, Choi YS, Jeong MJ, Kim BO, Lim SH, Kim do K, et al. (2007). (CGRP, substance P) during and after orthodontic tooth movement in
Expression of UNCL during development of periodontal tissue and the rat. Eur J Orthod 17:311-325.
response of periodontal ligament fibroblasts to mechanical stress in Phillips JA, Almeida EA, Hill EL, Aguirre JI, Rivera MF, Nachbandi I, et al.
vivo and in vitro. Cell Tissue Res 327:25-31. (2008). Role for beta1 integrins in cortical osteocytes during acute
Kitaura H, Yoshimatsu M, Fujimura Y, Eguchi T, Kohara H, Yamaguchi A, musculoskeletal disuse. Matrix Biol 27:609-618.
et al. (2008). An anti-c-Fms antibody inhibits orthodontic tooth move­ Poirier CC, Iglesias PA (2007). An integrative approach to understanding
ment. J Dent Res 87:396-400. mechanosensation. Brief Bioinform 8:258-265.
Kong Q, Vazquez M (2009). Flow-induced shear stresses increase the num­ Ren Y, Vissink A (2008). Cytokines in crevicular fluid and orthodontic tooth
ber of cell-cell contacts within extracellular matrix. J Biomed Mater movement. Eur J Oral Sci 116:89-97.
Res A 89:968-979. Ren Y, Maltha JC, Stokroos I, Liem RSB, Kuijpers-Jagtman AM (2008).
Koyama Y, Mitsui N, Suzuki N, Yanagisawa M, Sanuki R, Isokawa K, et al. Effect of duration of force application on blood vessels in young and
(2008). Effect of compressive force on the expression of inflammatory adult rats. Am J Orthod Dentofacial Orthop 133:752-757.
cytokines and their receptors in osteoblastic Saos-2 cells. Arch Oral Ritter N, Mussig E, Steinberg T, Kohl A, Komposch G, Tomakidi P (2007).
Biol 53:488-496. Elevated expression of genes assigned to NF-kappaB and apoptotic
Kreja L, Liedert A, Hasni S, Claes L, Ignatius A (2008). Mechanical regula­ pathways in human periodontal ligament fibroblasts following mechan­
tion of osteoclastic genes in human osteoblasts. Biochem Biophys Res ical stretch. Cell Tissue Res 328:537-548.
Commun 368:582-587. Robling AG, Niziolek PJ, Baldridge LA, Condon KW, Allen MR, Alam I,
Krishnan V (2007). Orthodontic pain: from causes to management—a et al. (2008). Mechanical stimulation of bone in vivo reduces osteocyte
review. Eur J Orthod 29:170-179. expression of Sost/sclerostin. J Biol Chem 283:5866-5875.
Krishnan V, Davidovitch Z (2006a). Cellular, molecular, and tissue-level Rosenkilde MM, Schwartz TW (2004). The chemokine system—a major
reactions to orthodontic force. Am J Orthod Dentofacial Orthop regulator of angiogenesis in health and disease. APMIS 112:481-495.
129:469.e1-469.e32. Rossi D, Zlotnik A (2000). The biology of chemokines and their receptors.
Krishnan V, Davidovitch Z (2006b). The effect of drugs on orthodontic tooth Ann Rev Immunol 18:218-242.
movement. Orthod Craniofac Res 9:163-171. Rubin J, Rubin C, Jacobs CR (2006). Molecular pathways mediating
Lee SK, Pi SH, Kim SH, Min KS, Lee HJ, Chang HS, et al. (2007). mechanical signaling in bone. Gene 367:1-16.
Substance P regulates macrophage inflammatory protein 3alpha/ Sandy JR (1998). Signal transduction. Br J Orthod 25:269-274.
chemokine C-C ligand 20 (CCL20) with heme oxygenase-1 in human Schwartz MA, DeSimone DW (2008). Cell adhesion receptors in mechano­
periodontal ligament cells. Clin Exp Immunol 150:567-575. transduction. Curr Opin Cell Biol 20:551-556.
Lew KK (1989). Orthodontically induced microvascular injuries in the Shyy JY, Chien S (2002). Role of integrins in endothelial mechanosensing
tension zone of the periodontal ligament. J Nihon Univ Sch Dent of shear stress. Circ Res 91:769-775.
31:493-501. Silva TA, Garlet GP, Fukada SY, Silva JS, Cunha FQ (2007). Chemokines
Li J, Zhao Z, Wang J, Chen G, Yang J, Luo S (2008). The role of extracel­ in oral inflammatory diseases: apical periodontitis and periodontal dis­
lular matrix, integrins, and cytoskeleton in mechanotransduction of ease. J Dent Res 86:306-319.
centrifugal loading. Mol Cell Biochem 309:41-48. Silver FH, Siperko LM (2003). Mechanosensing and mechanochemical
Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, et al. (2005). Sclerostin binds transduction: how is mechanical energy sensed and converted into
to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem chemical energy in an extracellular matrix? Crit Rev Biomed Eng
280:19883-19887. 31:255-331.
Lisignoli G, Toneguzzi S, Piacentini A, Cristino S, Grassi F, Cavallo C, Smith ML, Gourdon D, Little WC, Kubow KE, Eguiluz RA, Luna-Morris S,
et al. (2006). CXCl12 (SDF-1) and CXCL 13 (BCA-1) chemokines et al. (2007). Force-induced unfolding of fibronectin in the extracellular
significantly induce proliferation and collagen type 1 expression in matrix of living cells. PLoS Biol 5:e268.
osteoblasts from osteoarthritic patients J Cell Physiol 206:78-85. Takahashi I, Nishimura M, Onodera K, Bae JW, Mitani H, Okazaki M,
Maeda A, Soejima K, Bandow K, Kuroe K, Kakimoto K, Miyawaki S, et al. et al. (2003). Expression of MMP-8 and MMP-13 genes in the
(2007). Force-induced IL-8 from periodontal ligament cells requires periodontal ligament during tooth movement in rats. J Dent Res
IL-1beta. J Dent Res 86:629-634. 82:646-651.
608  Krishnan & Davidovitch J Dent Res 88(7) 2009

Tatsumi S, Ishii K, Amizuka N, Li M, Kobayashi T, Kohno K, et al. (2007). Wescott DC, Pinkerton MN, Gaffey BJ, Beggs KT, Milne TJ, Meikle MC
Targeted ablation of osteocytes induces osteoporosis with defective (2007). Osteogenic gene expression by human periodontal ligament
mechanotransduction. Cell Metab 5:464-475. cells under cyclic tension. J Dent Res 86:1212-1216.
Taylor AF, Saunders MM, Shingle DL, Cimbala JM, Zhou Z, Donahue HJ Wildemann B, Schmidmaier G, Brenner N, Hüning M, Stange R, Haas NP,
(2007). Mechanically stimulated osteocytes regulate osteoblastic et al. (2004). Quantification, localization, and expression of IGF-I and
activity via gap junctions. Am J Physiol Cell Physiol 292:C545- TGF-beta1 during growth factor-stimulated fracture healing. Calcif
C552. Tissue Int 74:388-397.
Teitelbaum SL (2007). Osteoclasts: what do they do and how do they do it? Wise GE, King GJ (2008). Mechanisms of tooth eruption and orthodontic
Am J Pathol 170:427-435. tooth movement. J Dent Res 87:414-434.
Teitelbaum SL, Ross FP (2003). Genetic regulation of osteoclast develop­ Xie R, Kuijpers-Jagtman AM, Maltha JC (2008). Osteoclast differentiation dur­
ment and function. Nat Rev Genet 4:638-649. ing experimental tooth movement by a short-term force application: an
Terricabras E, Benjamin C, Godessart N (2004). Drug discovery and immunohistochemical study in rats. Acta Odontol Scand 66:314-320.
chemokine receptor antagonists: eppur si muove! Autoimmune Rev Yamaguchi M, Kojima T, Kanekawa M, Aihara N, Nogimura A, Kasai K
3:550-556. (2004). Neuropeptides stimulate production of interleukin-1 beta, inter­
Vandevska-Radunovic V (1999). Neural modulation of inflammatory reac­ leukin-6, and tumor necrosis factor-alpha in human dental pulp cells.
tions in dental tissues incident to orthodontic tooth movement. A review Inflamm Res 53:199-204.
of the literature. Eur J Orthod 21:231-247. Yamaguchi M, Yoshii M, Kasai K (2006). Relationship between substance
Verborgt O, Tatton NA, Majeska RJ, Schaffler MB (2002). Spatial distribu­ P and interleukin-1beta in gingival crevicular fluid during orthodontic
tion of Bax and Bcl-2 in osteocytes after bone fatigue: complementary tooth movement in adults. Eur J Orthod 28:241-246.
roles in bone remodeling regulation? J Bone Miner Res 17: 907-914. Yamaguchi M, Ozawa Y, Mishima H, Aihara N, Kojima T, Kasai K (2008).
Wang HB, Dembo M, Hanks SK, Wang Y (2001). Focal adhesion kinase is Substance P increases production of proinflammatory cytokines and forma­
involved in mechanosensing during fibroblast migration. Proc Natl tion of osteoclasts in dental pulp fibroblasts in patients with severe ortho­
Acad Sci USA 98:11295-11300. dontic root resorption. Am J Orthod Dentofacial Orthop 133:690-698.
Wang JH, Thampatty BP, Lin JS, Im HJ (2007). Mechanoregulation of gene Zhang K, Barragan-Adjemian C, Ye L, Kotha S, Dallas M, Lu Y, et al.
expression in fibroblasts. Gene 391:1-15. (2006). E11/gp38 selective expression in osteocytes: regulation by
Wang Y, McNamara LM, Schaffler MB, Weinbaum S (2007). A model for mechanical strain and role in dendrite elongation. Mol Cell Biol
the role of integrins in flow induced mechanotransduction in osteo­ 26:4539-4552.
cytes. Proc Natl Acad Sci USA 104:15941-15946. Zhao Y, Wang C, Li S, Song H, Wei F, Pan K, et al. (2008). Expression of
Weinbaum S, Cowin SC, Zeng Y (1994). A model for the excitation of Osterix in mechanical stress-induced osteogenic differentiation of
osteocytes by mechanical loading-induced bone fluid shear stresses. J perio­dontal ligament cells in vitro. Eur J Oral Sci 116:199-206.
Biomech 27:339-360. Zhou L, Isenberg JS, Cao Z, Roberts DD (2006). Type I collagen is a
Wells RG (2008). The role of matrix stiffness in regulating cell behavior. molecular target for inhibition of angiogenesis by endogenous throm­
Hepatology 47:1394-1400. bospondin-1. Oncogene 25:536-545.

You might also like