You are on page 1of 8

The FASEB Journal • Review

Mechanotransduction in bone repair and regeneration


Chenyu Huang*,†,1 and Rei Ogawa*
*Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan;
and †Department of Plastic Surgery, Meitan General Hospital, Beijing, China

ABSTRACT Mechanotransduction is the process by Such complex mechanisms are responsible for main-
which physical forces are converted into biochemical taining the dynamic balance between bone forma-
signals that are then integrated into cellular responses. tion and bone absorption. A greater understanding
It plays a crucial role in bone repair and regeneration of these processes may lead to the development of
and thus has attracted a great deal of interest from advanced clinical applications that can lead to pre-
researchers in various fields. This report reviews the dictable and reproducible improvements in bone
current clinical evidence that shows the role mechano- structure. Therefore, in this report the clinical evi-
transduction plays in bone processes such as physical dence of bone mechanotransduction and the basic
adaptation, pathological fracture healing, and thera- research into the mechanotransduction mechanisms
peutic distraction osteogenesis. We also outline the involved in bone repair and regeneration are re-
progress that has been made in understanding bone viewed. Moreover, future perspectives of bone mech-
mechanotransduction from both the macro- and mi- anotransduction research are discussed.
croperspectives. Specifically, we describe the theories
that postulate how mechanical force exhibits effects on
bone repair and regeneration (i.e., the tensegrity and CLINICAL EVIDENCE OF
mechanosome theories). We also summarize the recent MECHANOTRANSDUCTION IN BONE REPAIR
advances in our understanding of the molecular signal- AND REGENERATION
ing pathways of mechanotransduction, which include
calcium ion channels, integrins, Wnt/␤-catenin, prosta- How mechanical stimuli influence bone regeneration
glandin, and nitric oxide. A better understanding of has been explored by studying processes such as phys-
skeletal mechanotransduction will facilitate research iological bone adaptation and clinical conditions such
into this promising field and could lead to the develop- as pathological bone fracture and distraction osteogen-
ment of applications that improve bone structures and esis.
functions.—Huang, C., Ogawa, R. Mechanotransduc-
tion in bone repair and regeneration. FASEB J. 24, Physiological bone adaptation
3625–3632 (2010). www.fasebj.org
To meet the functional demands of its mechanical
Key Words: mechanical stimulation 䡠 mechanosensation 䡠 mech- environment, the mass and geometry of bone is physi-
anotransductive pathways 䡠 skeletal adaptation 䡠 bone repara- cally remodeled in a dynamic fashion (Wolff’s law).
tion and reconstruction
Mechanostat theory is a refinement of Wolff’s law and
proposes that bone adapts so that it can function
Bones serve in our body as skeletal structures that mechanically as needed by detecting and responding to
support the body’s weight, connect tendons and mechanical loads (3). Thus, in this dynamic and life-
muscles, provide mechanical protection, assist move- long biological control system, bone formation in terms
ment, store minerals, and produce blood cells. Bone of shape, size, and density can be directed by high
repair and regeneration are undoubtedly influenced mechanical loads. The other side of the coin is that
by intimate interactions between the physiological, immobilization can lead to the loss of bone. For exam-
biochemical, and mechanobiological environments. ple, 120 d of bed rest can induce bone loss by acceler-
In particular, the effect of mechanical force on bone ating bone resorption and retarding bone formation
regeneration has been studied widely, and this has (4). This is also true for spaceflight, where bone loss
led to an increasing awareness of the importance of can be induced because of increased bone resorption
studying mechanotransduction. and decreased calcium absorption (5).
Mechanotransduction is the process by which phys- It is also currently clear that the skeleton needs “time
ical forces are converted into biochemical signals
that are then integrated into cellular responses (1). 1
Correspondence: Department of Plastic, Reconstructive
Consequently, mechanotransduction in the bone in- and Aesthetic Surgery, Nippon Medical School, 1-1-5 Sendagi
cludes 4 phases: mechanocoupling, biochemical cou- Bunkyo-ku, Tokyo 113-8603, Japan. E-mail: huangchenyu2000@
pling, transmission of the signal from the sensor cell gmail.com
to the effector cell, and the effector cell response (2). doi: 10.1096/fj.10-157370

0892-6638/10/0024-3625 © FASEB 3625


off” from mechanical loading. Mechanical loading pre- a special form of bone healing in which gradual trac-
sents a potent osteogenic stimulus, but bone cells tion on the bone leads to osteogenesis and the tandem
desensitize rapidly to mechanical stimulation. Resensi- lengthening of the skin, muscles, and nerves (19, 20).
tization must occur before the cells can transduce It is well known that bone cells consist mainly of
future mechanical signals effectively (6). Consequently, osteoblasts (active osteoblasts and inactive bone-lining
it has been suggested that cyclical or intermittent cells), osteoclasts, and osteocytes. Clinical osteogenesis
loading, which provides the skeleton with regular “time in fracture healing and distraction osteogenesis aims to
off” periods, may be more effective than continuous facilitate bone formation by osteoblast and bone re-
loading in inducing the bone formation needed to modeling together with osteoclast. From the perspec-
promote the growth and repair of the skeleton (7). tive of mechanotransduction, bone tissue is described
Supporting this is that the osteogenic response to as an extensively connected cellular network where the
simulated high-impact exercise over a long period can osteocytes serve as sensory cells and the osteoblasts and
be enhanced by dividing the exercise into brief sessions osteoclasts are the effector cells (21). Loads applied to
of loading that are separated by recovery periods (8). a whole bone are related to the flow past the osteocytic
Factors such as age and gender also affect the mech- processes in their canaliculi (22). The osteocytes can
anotransduction process in bone. Aging can inhibit sense the flow of fluid and then produce signaling
mechanotransduction, as shown by the fact that 19-mo- molecules that regulate osteoclast-mediated bone re-
old rats exhibit over 16-fold less mechanically induced sorption and osteoblast-mediated bone formation. This
bone formation than 9-mo-old rats (9). There also results in adequate bone remodeling (23). That this
appears to be a “window of opportunity” during the process occurs has been supported by the finding that
development of peak bone mass in which the bone is the targeted ablation of osteocytes in mice results in
especially responsive to weight-bearing physical activity defective mechanotransduction and fragile bone with
(10). Furthermore, during advancing age increased osteoblastic dysfunction (24).
remodeling rate and worsening of basic multicellular The mechanical modulation of bone fracture healing
unit (BMU) imbalance increase bone loss and struc- varies depending on the type of fracture, the therapeu-
tural damage, resulting in a predisposition to bone tic fixation, and the loading that follows. A series of
fracture following mechanical stimulation such as min- hypotheses have been proposed to explain mechanical
imal trauma (11). In fact, according to Seeman, 4 of the influence on tissue differentiation in fracture: in these
age-related changes in bone modeling and remodeling hypotheses, hydrostatic pressure and tensile strain (25,
that compromise bone’s material properties and struc- 26), or shear strain and fluid flow, have been identified
tural design are a reduction in bone formation at tissue as the key stimulating mechanical forces (27, 28).
level, reduction in bone formation at cellular level However, to date, far less is understood about the signal
within each BMU, continued resorption in the BMU, mechanotransductive pathways that lead to fracture
and increase in the rate of bone remodeling accompa- repair (29). One study has shown that receptor activa-
nied by worsening of negative bone balance in each tor of nuclear factor ␬B (RANK) signaling is not
BMU (12, 13). In addition, gender affects bone mech- required for the early phase of fracture healing, as
anotransduction because males rather than females evidenced by the repair (callus formation) observed in
exhibit significantly reduced mechanoresponsiveness RANK⫺/⫺ mice with bone fractures; this is supported
compared with controls (14). Conversely, males suffer by the fact that administration of RANK signaling
larger bone mass reductions in both the cortical and inhibitors sufficient to reduce bone resorption is not
cancellous compartments than females when they are detrimental to fracture healing (30). Another study has
subjected to hind limb unloading (15). In fact, the shown that proinflammatory cytokines such as TNF-␣
position of the cortex in relationship to the long axis of and IL-1, which are known to regulate immune func-
the long bone differs in males and females (16). tion and inflammation, participate in fracture healing
Estrogen reduces the osteogenic response induced by because they are expressed at both the very early and
exercise on the periosteal surface of the bone, whereas late phases of the repair process. It then suggests that
it enhances bone formation on the endocortical surface these cytokines are important in the initiation of the
of 8-wk-old male Sprague-Dawley rats (17). Such age- repair process and play important functional roles in
and gender-specific variations may be due to differ- intramembraneous bone formation and remodeling
ences in the efficiency of mechanical sensors and sex (31).
hormone production and/or signaling, respectively. Distraction osteogenesis is specifically character-
However, additional research is needed to clarify this. ized by osseous regeneration that occurs primarily
via intramembranous ossification, which is stimu-
Fracture healing and distraction osteogenesis lated by several mechanotransductive pathways. In
the integrin-mediated, extracellular signal-related ki-
The clinical bone regeneration that occurs in fracture nase (ERK 1/2) -dependent mechanotransduction
healing and distraction osteogenesis is also evidence of pathway, ERK 1/2 is a potential central mediator that
bone mechanotransduction. The healing of a bone acts as a signaling convergence point and regulates
fracture is a unique process whose result is not scarring the osteogenic differentiation of mesenchymal stem
but regeneration (18), while distraction osteogenesis is cells during distraction (32, 33). In addition, in-

3626 Vol. 24 October 2010 The FASEB Journal 䡠 www.fasebj.org HUANG AND OGAWA
creased expression of osteogenic proteins, including end to the extracellular matrix (ECM) (38). Focal
BMP 2/4, can be induced via the c-Src-dependent adhesions are multimolecular complexes that connect
mechanotransduction pathway (34). Mechanical the ECM to the actin cytoskeleton and link integrins to
forces can also stimulate the expression of the early- the ends of contractile microfilament bundles. They
response genes of the activator protein-1 (AP-1) are thought of as mechanosensory organelles (41, 42),
family of transcription factors (35), up-regulate the where mechanochemical signal conversion is carried
expression of Runx2, and initiate the differentiation out in the cell.
of periosteal cells into osteogenic cells (36), thus The forces that are channeled as described above
promoting osseous regeneration. over the ECM and to the integrins are converted into
These observations show the central role mechano- biochemical changes by producing changes in defor-
transduction plays in bone regeneration, physical me- mation of other load-bearing mechantransducer mole-
chanical loading, pathological bone fracture, and the cules, such as stress-sensitive ion channels, protein
surgical intervention of distraction osteogenesis. Stud- kinases, G proteins, and other signaling molecules,
ies that elucidate the fundamental signaling processes inside the cell (42).
that are involved are needed.
Mechanosomes theory

BASIC RESEARCH INTO THE Mechanosomes theory is an appealing hypothesis that


MECHANOTRANSDUCTION MECHANISMS was proposed by Pavalko (43). The key aspect of this
INVOLVED IN BONE REPAIR AND model is the load-induced formation of mechano-
REGENERATION somes, which are multiprotein complexes comprised of
focal adhesion-associated or adherens junction-associ-
Mechanotransduction, particularly in bone, has been a ated proteins. In terms of cellular structure, the load-
focus of interest in fields ranging from molecular induced deformation of bone is converted into the
biophysics to clinical medicine. Research into mech- deformation of the sensor cell membrane, which drives
anotransduction has been approached from 2 direc- conformational changes in membrane proteins. Some
tions. Some researchers have sought to understand it of these membrane proteins are linked to a solid-state
from the macro/system perspective, where it is seen as signaling scaffold that releases protein complexes
a control system that requires feedback and stability. called mechanosomes that are capable of carrying
Others have viewed it from a micro/molecular perspec- mechanical information into the nucleus. In this way,
tive and have sought to understand how it works by “bending bone ultimately bends genes” (43). In terms
characterizing the specialized molecular signaling path- of signal conversion, the solid-state and diffusion-con-
ways that are involved. trolled signaling pathways integrate with the tissue
matrix-mediated transfer of mechanical energy to pro-
Tensegrity theory teins of the adhesion complexes and selected proteins
associated with the cyto- and nucleoskeleton. This
Drawing on control theory and architecture, tenseg- conversion of mechanical energy into chemical energy
rity theory proposes a mechanism that explains how drives changes in protein conformation, phosphoryla-
mechanical stresses applied at the macroscopic level tion, and alterations in DNA geometry and mediates
can influence the molecular structure and function the formation and/or mobilization of nascent signaling
of living cells (37). In this theory, the whole cell, not complexes to the bone cell cytoplasm (43).
just the single specialized mechanotransduction mol-
ecules in isolation, is believed to serve as a mechano- Role of osteocyte in mechanosignal transduction
transducer as it integrates the local signals with other
environmental inputs before eliciting a specific be- Osteocytes orchestrate different cell populations in
havioral response (38). bone in response to mechanical loading (44). They can
The key determinants in tensegrity are architecture be activated by strain amplification through canaliculi.
(the 3-dimensional arrangement of the elements) and The electrically coupled 3-dimensional network of os-
the level of prestress (isometric tension) in the cytoskel- teocytes and lining cells is a communications system for
eton (37). Thus, on the one hand, an even distribution the control of bone homeostasis and for structural
of load protects individual cells from damage, while on strain adaptation, and the effector cells are osteoblasts
the other hand, a small mechanical stimulus is allowed and osteoclasts (45).
to have an effect on a large number of cells (39, 40). Osteocytes are the most abundant cells in bone cells
Integrins and focal adhesions are recognized as and are spaced throughout the mineralized matrix.
being the ubiquitous mediators of mechanosensation. They appear to be the most mechanosensitive cells in
Integrins are a family of ubiquitous, heterodimeric bone and are involved in the transduction of mechan-
glycoproteins that mediate cell attachment to the ex- ical stress into a biological response. Osteocytes, but not
tracellular matrix. They act as mechanoreceptors of the osteoblasts, react to a 1-h pulsating fluid flow, resulting
cell by spanning the cell membrane and being con- in the sustained release of prostaglandin E2. By con-
nected at one end to the cytoskeleton and at the other trast, intermittent hydrostatic compression stimulates

MECHANOTRANSDUCTION IN BONE REPAIR AND REGENERATION 3627


prostaglandin production after 6 and 24 h in osteocytes response to fluid flow and that this can be suppressed
and after 6 h in osteoblasts (46). Osteocytes are more by gadolinium, a stretch-activated channel blocker
responsive to pulsating fluid flow than osteoblasts with (58). In vivo studies have shown that mechanically
respect to the production of soluble factors of nitric induced bone formation in rats is substantially sup-
oxide, which affect both osteoblast proliferation and pressed by verapamil and nifedipine, 2 blockers of
differentiation (44), as well as osteoclast formation and L-type calcium channels (59). Furthermore, inositol
bone resorption (47). Pulsating fluid flow for 15 min 1,4,5-triphosphate (IP3) can mediate intracellular Ca2⫹
stimulates by 3-fold prostaglandin G/H synthase-2 release, which is required for modulating flow-induced
(PGHS-2, COX-2) mRNA expression in osteocytes but responses (60).
does not have the same effect in osteoblasts (48).
Besides, targeted ablation of osteocytes in mice induces Integrin signaling
osteoporosis with defective mechanotransduction (24).
In fact, the main source of signaling induced by oscil- Integrins, as mentioned above, are transmembrane
latory fluid flow in osteoblasts occurs through the focal molecules connected at one end to the ECM and at the
adhesion complex and the focal adhesion kinase (49). other end to the intracellular cytoskeleton. They also
Formation of focal adhesions on fibronectin promotes play an important role in osseous mechanobiology.
fluid shear stress induction of cyclooxygenase (COX)-2 When focal adhesion kinase, an intracellular compo-
and PGE2 release in osteoblasts (50). The linkage of nent of the integrin signaling pathway, was condition-
actin stress fibers to integrins in focal adhesions via ally inactivated, the mechanically induced osteogenic
␣-actinin is a critical link in osteoblasts, because block- response was abolished (61). Moreover, an integrin-
ing this linkage prevents fluid shear-induced COX-2 mediated, extracellular signal-related kinase ERK 1/2-
and c-Fos expression (51). Excitation of osteocytes may dependent mechanotransduction pathway was found to
also be related to interaction of pericellular matrix and play a significant role in distraction osteogenesis (33).
cell process cytoskeleton (52).
Compared to the bone lining cells that cover the Prostaglandin (PG) and nitric oxide (NO) signaling
bone surface, osteocytes located within the bone matrix
would be more efficient sensors. With osteocytes as PG is an important biochemical mediator of mechani-
sensors, remodeling would be more sensitive to exter- cal loading in bone. Fluid flow shear stress can promote
nal loads; such remodeling would lead to adaptation of the release of intracellular PGE2 from osteocytes (62).
the architecture rather than that of strut thickness, and It induces the translocation of Cx43 to the membrane
the remodeling process drifts more easily and effec- surface, and the unapposed hemichannels formed by
tively toward a significantly different morphology. As Cx43 serve as a novel portal for the release of PGE2 in
for changes in load magnitudes only, surface bone response to mechanical strain (63). Moreover, the
lining cells should be equally capable of regulating bone formation that follows brief bouts of mechanical
adaptation as osteocytes. However, adding surface cells loading involves the release of PG from cells at the time
to the osteocyte model makes no difference to the mechanical loading is applied, rather than new PG
output of the model (53). synthesis associated with mechanically induced COX-2
expression (64). Since PGs are synthesized through the
Molecular skeletal mechanotransducers conversion of arachidonic acid by the rate-limiting
COX enzymes, the role of COX in PG signaling re-
Bone, as a mechanosensitive organ, reacts to mechani- mains unclear. One study has shown that a functional
cal stimuli through a series of molecules and crosstalk COX-2 gene is not required for skeletal mechanotrans-
between them. Research into mechanotransduction duction because mice with a null mutation in the
has shown that gaps between physical forces and bio- COX-2 gene respond to loading by inducing skeletal
chemical signals are bridged by intracellular ion chan- COX-1 expression (65). In contrast, another study has
nels such as K⫹ (54) and Ca2⫹ (55) channels, intracel- shown by administering the COX-2 inhibitors NSAIDs
lular signaling, such as that of Wingless-type/␤-catenin or NS-398 that COX-2 does play an important role in
and inositol 1,4,5-triphosphate, mechanically induced the bone formation elicited by mechanical strain (66).
signaling molecules such as that of prostaglandin and NO signaling is also involved, as shown by the fact that
nitric oxide (56), transmembrane molecules such as application of mechanical strain onto bone stromal cells
integrin (57), growth factors such as insulin-like growth regulates NO synthase in synergy with RANK ligand to
factor and bone morphogenetic protein, and systemic promote positive bone remodeling; this process involves
hormones such as parathyroid and estrogen. mitogen-activated protein kinase (67). Moreover,
mechanically induced bone formation can be sup-
Calcium ion signaling pressed by L-NAME, a NO synthesis inhibitor (68).

Several studies have revealed that calcium ion signaling Wingless-type (Wnt)/␤-catenin signaling
plays an important role in osseous mechanobiology. In
vitro studies have shown that the intracellular calcium Wnt signaling through LDL receptor-related protein-5
in osteoblastic cells can be increased within minutes in (LRP5) receptor plays an important and complex role

3628 Vol. 24 October 2010 The FASEB Journal 䡠 www.fasebj.org HUANG AND OGAWA
in skeletal mechanotransduction. One study has shown chanical strain might lead to cellular toxicity. At
that Wnt/␤-catenin signaling is stimulated in calvarial present, the desensitization process is believed to
cells in vitro after they have been subjected to mechan- involve multiple mechanisms in bone cells, most
ical loading such as stretching (69). Wnt signaling was notably G-protein coupled receptor desensitization
also observed in vivo in mice after mechanical loading (79). Moreover, increasing strain to increase cellular
of the tibia; indeed, Wnt signaling enhanced the sensi- toxicity is found in mouse osteoblast, which may
tivity of osteoblasts to mechanical loading (70). In result in decreases in cell number (80). Thus, re-
Lrp5⫺/⫺ mice, the osteogenic responses to mechanical search that identifies desensitization pathways and
loading of the ulna are dramatically impaired (88 and consequently uses specific inhibitors to remove an
99% reductions were observed in males and females, “off switch” would be of great interest. It aims to
respectively) (71). Notably, however, the inhibition of improve osteogenesis by keeping the switch on,
Wnt signaling can also stimulate osteoblast differentia- meanwhile avoiding potential cellular toxicity by
tion and mineral formation. One study has shown that subsequently increased mechanical stimulation.
to enable the formation of a mineralized bone matrix, Mechanical stimuli simultaneously affect other tis-
Wnt signaling in maturing osteoblasts needs to be sues through mechanotransduction, and this could
down-regulated (72). Another study revealed Wnt sig- be manipulated to improve bone regeneration. In-
naling inhibits the osteogenic differentiation of human deed, it is feasible to use mechanical stimuli to
mesenchymal stem cells (73). Moreover, in human promote bone regeneration, although this will occur
osteoblastic cells subjected to 15 min of stretching, only if there is a timely nutrient supply and waste
although there is an initial early increase in ␤-catenin cleaning. This suggests that angiogenesis is needed to
levels, Wnt signaling is ultimately down-regulated. This facilitate the bone regeneration induced by mechan-
biphasic Wnt signaling level is likely to play an impor- ical stimuli. Notably, a study of the chorioallantoic
tant role in end-stage osteoblasts in that the reduced membrane revealed mechanical strain could elicit
Wnt signaling may induce terminal differentiation and angiogenic features (81). Moreover, osteoblasts re-
matrix mineralization (74). sponded in vitro to mechanical strain by increasing
Other signaling pathways, such as those that em- matrix production and regulating angiogenesis (82).
ploy ATP (75), insulin-like growth factor, bone mor- Current research hypothesized that induction of
phogenetic protein (76), and receptors of hormones matrix metalloproteinases after mechanical strain
such as parathyroid hormone (77) or estrogen (78), enhances attraction and penetration of blood vessels
are also of increasing interest in skeletal mechano- through which osteoblasts could reach the chondro-
transduction research. osseous junction and promote ossification (83). Fu-
The observations above show that, regardless of ture research examining whether mechanical stimu-
whether mechanotransduction in bone regeneration lus-induced bone regeneration can be improved by
is approached from the macro- or microperspective, simultaneously activating mechanotransduction
this biological process is highly diverse and complex pathways that promote angiogenesis would be of
and involves multiple overlapping and coordinated considerable clinical interest.
molecular signaling pathways. Further research elu- Much remains to be elucidated in mechanotransduc-
cidating this process is greatly warranted. tive signaling. Some questions of interest are the follow-
ing: 1) Since various signaling cascades can be simulta-
neously up- and down-regulated by mechanical stimuli,
what are correlations and relationship between different
FUTURE PERSPECTIVES OF BONE
pathways? 2) How do different cell types participate in
MECHANOTRANSDUCTION RESEARCH
skeletal mechanotransduction? 3) How do different types
of mechanical stimuli such as hydrostatic pressure and
Mechanical loading activates mechanotransducers
microgravity affect bone regeneration? 4) Genetics prob-
that in turn stimulate bone formation, thereby main-
ably plays a major role in determining the degree of
taining the dynamic skeletal balance between osteo-
skeletal adaptability to the same mechanical stimulus
genesis and bone absorption. This process is regu-
(84), but which set of genes is responsible for this differ-
lated by local cytokines and growth factors and
ence in mechanosensitivity? A deeper understanding of
systemic hormones. Further, mechanical forces are
the skeletal mechanotransductive mechanisms will greatly
even believed to be potent regulators of cell and
facilitate the development of innovative interventions,
tissue development as soluble hormones and insolu-
either pharmaceutical or clinical, that mimic or hamper
ble ECM proteins (42). Should this be the case, there
responses to loading and thereby improve bone structures
are many different avenues future studies in skeletal
and functions.
mechanotransduction can take.
Most current studies have focused on how mechan-
ical stimuli from the environment can lead to molec-
ular effects in bone cells. In other words, they have CONCLUSIONS
studied the “on switch.” In contrast, there is compar-
atively little research into bone cell desensitization Mechanotransduction in bone regeneration is at-
(“off switch”) and the possibility that increased me- tracting increasing interest from a variety of fields.

MECHANOTRANSDUCTION IN BONE REPAIR AND REGENERATION 3629


Clinical evidence has shown that it plays a fundamen- 13. Seeman, E., and Delmas, P. D. (2006) Bone quality-the material
tal role in dynamically modulating and maintaining and structural basis of bone strength and fragility. N. Engl.
J. Med. 354, 2250 –2261
the balance between osteogenesis and bone resorp- 14. Robling, A. G., Warden, S. J., Shultz, K. L., Beamer, W. G., and
tion, thereby leading to physical adaptation, patho- Turner, C. H. (2007) Genetic effects on bone mechanotrans-
logical fracture healing, and therapeutic distraction duction in congenic mice harboring bone size and strength
quantitative trait loci. J. Bone Miner. Res. 22, 984 –991
osteogenesis. Meanwhile, current research has provided 15. David, V., Lafage-Proust, M. H., Laroche, N., Christian, A.,
both macro- and microperspectives in understanding the Ruegsegger, P., and Vico, L. (2006) Two-week longitudinal
mechanotransduction signaling pathways that are in- survey of bone architecture alteration in the hindlimb-unloaded
volved in bone repair and regeneration. According to rat model of bone loss: sex differences. Am. J. Physiol. Endocrinol.
Metab. 290, E440 –E447
tensegrity theory, the whole cells, rather than single 16. Seeman, E. (2008) Bone quality: the material and structural
specialized mechanotransducer molecules, are seen as basis of bone strength. J. Bone Miner. Metab. 26, 1– 8
the mechanotransducers. The mechanosome theory 17. Saxon, L. K., and Turner, C. H. (2006) Low-dose estrogen
treatment suppresses periosteal bone formation in response to
and other signaling pathways such as calcium ion mechanical loading. Bone 39, 1261–1267
channels, integrins, and autocrine and paracrine 18. Isaksson, H., Wilson, W., van Donkelaar, C. C., Huiskes, R., and
messengers such as prostaglandin and nitric oxide Ito, K. (2006) Comparison of biophysical stiuli for mechano-
provide a microview of the mechanotransductive regulation of tissue differentiation during fracture healing.
J. Biomech. 39, 1507–1516
signaling in which molecules play overlapping and 19. Ilizarov, G. A. (1989) The tension-stress effect on the genesis and
coordinated roles. The study of bone mechanotrans- growth of tissues: part I. The influence of stability of fixation and
duction is a highly promising field, and it is likely that soft-tissue preservation. Clin. Orthop. Relat. Res. 238, 249 –281
a greater understanding of the fundamental molec- 20. Ilizarov, G. A. (1989) The tension-stress effect on the genesis
and growth of tissues: part II. The influence of the rate and
ular signal pathways that are involved and collabora- frequency of distraction. Clin. Orthop. Relat. Res. 239, 263–285
tion with other fields of research such as investiga- 21. Mi, L. Y., Basu, M., Fritton, S. P., and Cowin, S. C. (2005)
tion of angiogenesis may facilitate the development Analysis of avian bone response to mechanical loading. Part two:
development of a computational connected cellular network to
of pharmaceutical and clinical applications that can study bone intercellular communication. Biomech. Model. Mech-
improve bone structures and functions. anobiol. 4, 132–146
22. Weinbaum, S., Cowin, S. C., and Zeng, Y. (1994) A model for
the excitation of osteocytes by mechanical loading-induced
bone fluid shear stresses. J. Biomech. 27, 339 –360
23. Santos, A., Bakker, A. D., and Klein-Nulend, J. (2009) The role
of osteocytes in bone mechanotransduction. Osteoporos. Int. 20,
REFERENCES 1027–1031
24. Tatsumi, S., Ishii, K., Amizuka, N., Li, M., Kobayashi, T., Kohno,
1. Huang, H., Kamm, R. D., and Lee, R. T. (2004) Cell mechanics K., Ito, M., Takeshita, S., and Ikeda, K. (2007) Targeted ablation
and mechanotransduction: pathways, probes, and physiology. of osteocytes induces osteoporosis with defective mechanotrans-
Am. J. Physiol. Cell Physiol. 287, C1–C11 duction. Cell. Metab. 5, 464 – 475
2. Turner, C. H., and Pavalko, F. M. (1998) Mechanotransduction 25. Carter, D. R., Beaupre, G. S., Giori, N. J., and Helms, J. A. (1998)
and functional response of the skeleton to physical stress: the Mechanobiology of skeletal regeneration. Clin. Orthop. Relat.
mechanisms and mechanics of bone adaptation. J. Orthop. Sci. 3, Res. 355, S41–S55
346 –355 26. Claes, L. E., and Heigele, C. A. (1999) Magnitudes of local stress
3. Frost, H. M. (2003) Bone’s mechanostat: a 2003 update. Anat. and strain along osseous surfaces predict the course and type of
Rec. A Discov. Mol. Cell. Evol. Biol. 275, 1081–1101 fracture-healing. J. Biomech. 32, 255–266
4. Inoue, M., Tanaka, H., Moriwake, T., Oka, M., Sekiguchi, C., 27. Prendergast, P. J., Huiskes, R., Soballe, K. (1997) Biophysical
Seino, Y. (2000) Altered biochemical markers of bone turnover stimuli on cells during tissue differentiation at implant inter-
in humans during 120 days of bed rest. Bone 26, 281–286 faces. J. Biomech. 30, 539 –548
5. Smith, S. M., Wastney, M. E., O’Brien, K. O., Morukov, B. V., 28. Lacroix, D., Prendergast, P. J. (2002) A mechano-regulation
Larina, I. M., Abrams, S. A., Davis-Street, J. E., Oganov, V., and model for tissue differentiation during fracture healing: analysis
Shackelford, L. C. (2005) Bone markers, calcium metabolism, of gap size and loading. J. Biomech. 35, 1163–1171
and calcium kinetics during extended-duration space flight on 29. Morgan, E. F., Gleason, R. E., Hayward, L. N., Leong, P. L., and
the Mir space station. J. Bone Miner. Res. 20, 208 –218 Palomares, K. T. (2008) Mechanotransduction and fracture
6. Robling, A. G., Hinant, F. M., Burr, D. B., and Turner, C. H. repair. J. Bone Joint Surg. Am. 90, 25–30
(2002) Improved bone structure and strength after long-term 30. Flick, L. M., Weaver, J. M., Ulrich-Vinther, M., Abuzzahab, F.,
mechanical loading is greatest if loading is separated into short Zhang, X., Dougall, W. C., Anderson, D., O’Keefe, R. J., and
bouts. J. Bone Miner. Res. 17, 1545–1554 Schwarz, E. M. (2003) Effects of receptor activator of NFkappaB
7. Saxon, L. K., Robling, A. G., Alam, I., and Turner, C. H. (2005) (RANK) signaling blockade on fracture healing. J. Orthop. Res.
Mechanosensitivity of the rat skeleton decreases after a long period of 21, 676 – 684
loading, but is improved with time off. Bone 36, 454–464 31. Kon, T., Cho, T. J., Aizawa, T., Yamazaki, M., Nooh, N., Graves, D.,
8. Robling, A. G., Hinant, F. M., Burr, D. B., and Turner, C. H. Gerstenfeld, L. C., and Einhorn, T. A. (2001) Expression of
(2002) Shorter, more frequent mechanical loading sessions osteoprotegerin, receptor activator of NF-kappaB ligand (osteo-
enhance bone mass. Med. Sci. Sports Exerc. 34, 196 –202 protegerin ligand) and related proinflammatory cytokines during
9. Turner, C. H., Takano, Y., and Owan, I. (1995) Aging changes fracture healing. J. Bone Miner. Res. 16, 1004 –1014
mechanical loading thresholds for bone formation in rats. 32. Tong, L., Buchman, S. R., Ignelzi, M. A., Jr., Rhee, S., and
J. Bone Miner. Res. 10, 1544 –1549 Goldstein, S. A. (2003) Focal adhesion kinase expression during
10. Nichols, D. L., Sanborn, C. F., and Essery, E. V. (2007) Bone mandibular sistraction osteogenesis: evidence for mechano-
density and young athletic women: an update. Sports Med. 37, transduction. Plast. Reconstr. Surg. 111, 211–222
1001–1014 33. Rhee, S. T., El-Bassiony, L., and Buchman, S. R. (2006) Extra-
11. Seeman, E. (2004) Estrogen, androgen, and the pathogenesis of cellular signal-related kinase and bone morphogenetic protein
bone fragility in women and men. Curr. Osteoporos. Rep. 2, 90 –96 expression during distraction osteogenesis of the mandible: in
12. Seeman, E. (2008) Structural basis of growth-related gain and vivo evidence of a mechanotransduction mechanism for differ-
age-related loss of bone strength. Rheumatology (Oxford) entiation and osteogenesis by mesenchymal precursor cells.
47(Suppl. 4), iv2–iv8 Plast. Reconstr. Surg. 117, 2243–2249

3630 Vol. 24 October 2010 The FASEB Journal 䡠 www.fasebj.org HUANG AND OGAWA
34. Rhee, S. T., and Buchman, S. R. (2005) Colocalization of c-Src 55. El Haj, A. J., Walker, L. M., Preston, M. R., and Publicover, S. J.
(pp60src) and bone morphogenetic protein 2/4 expression (1999) Mechanotransduction pathways in bone: calcium fluxes
during mandibular distraction osteogenesis: in vivo evidence of and the role of voltage-operated calcium channels. Med. Biol.
their role within an integrin-mediated mechanotransduction Eng. Comput. 37, 403– 409
pathway. Ann. Plast. Surg. 55, 207–215 56. Smalt, R., Mitchell, F. T., Howard, R. L., and Chambers, T. J.
35. Lewinson, D., Rachmie, l, A., Rihani-Bisharat, S., Kraiem, Z., (1997) Mechanotransduction in bone cells: induction of nitric
Schenzer, P., Korem, S., and Rabinovich, Y. (2003) Stimulation oxide and prostaglandin synthesis by fluid shear stress, but not
of Fos- and Jun-related genes during distraction osteogenesis. by mechanical strain. Adv. Exp. Med. Biol. 433, 311–314
J. Histochem. Cytochem. 51, 1161–1168 57. Salter, D. M., Robb, J. E., and Wright, M. O. (1997) Electrophys-
36. Kanno, T., Takahashi, T., Ariyoshi, W., Tsujisawa, T., Haga, M., iological responses of human bone cells to mechanical stimula-
and Nishihara, T. (2005) Tensile mechanical strain up-regulates tion: evidence for specific integrin function in mechanotrans-
Runx2 and osteogenic factor expression in human periosteal duction. J. Bone Miner. Res. 12, 1133–1141
cells; implications for distraction osteogenesis. J. Oral Maxillofac. 58. Hung, C. T., Allen, F. D., Pollack, S. R., and Brighton, C. T.
Surg. 63, 499 –504 (1996) Intracellular Ca2⫹ stores and extracellular Ca2⫹ are
37. Ingber, D.E. (2003) Tensegrity, II. How structural networks required in the real-time Ca2⫹ response of bone cells experi-
influence cellular information processing networks. J. Cell Sci. encing fluid flow. J. Biomech. 29, 1411–1417
116, 1397–1408 59. Li, J., Duncan, R. L., Burr, D. B., and Turner, C. H. (2002)
38. Ingber, D. E. (2006) Cellular mechanotransduction: putting all L-type calcium channels mediate mechanically induced bone
the pieces together again. FASEB J. 20, 811– 827 formation in vivo. J. Bone Miner. Res. 17, 1795–1800
39. Myers, K. A., Battner, J. B., Shrive, N. G., and Hart, D. A. (2007) 60. Chen, N. X., Ryder, K. D., Pavalko, F. M., Turner, C. H., Burr,
Hydrostatic pressure sensation in cells: integration into the D. B., Qiu, J., and Duncan, R. L. (2000) Ca(2⫹) regulates fluid
tensegrity model. Biochem. Cell Biol. 85, 543–551 shear-induced cytoskeletal reorganization and gene expression
40. Ingber, D. E. (1997) Tensegrity: the architectural basis of in osteoblasts. Am. J. Physiol. Cell Physiol. 278, C989 –C997
cellular mechanotransduction. Annu. Rev. Physiol. 59, 575– 61. Leucht, P., Kim, J. B., Currey, J. A., Brunski, J., and Helms, J. A.
599 (2007) FAK-Mediated mechanotransduction in skeletal regen-
41. Geiger, B., and Bershadsky, A. (2002) Exploring the neighbor- eration. PLoS ONE 2, e390
hood: adhesion-coupled cell mechanosensors. Cell 110, 139 –142 62. Cheng, B., Kato, Y., Zhao, S., Luo, J., Sprague, E., Bonewald,
42. Ingber, D. E. (2008) Tensegrity-based mechanosensing from L. F., and Jiang, J. X. (2001) Prostaglandin E2 is essential for gap
macro to micro. Prog. Biophys. Mol. Biol. 97, 163–179 junction-mediated intercellular communication between osteo-
43. Pavalko, F. M., Norvell, S. M., and Burr, D. B. (2003) A model cyte-like MLO-Y4 cells in response to mechanical strain. Endo-
for mechanotransduction in bone cells: the load-bearing mech- crinology 142, 3464 –3473
anosomes. J. Cell. Biochem. 88, 104 –112 63. Cherian, P. P., Siller-Jackson, A. J., Gu, S., Wang, X., Bonewald,
44. Vezeridis, P. S., Semeins, C. M., Chen, Q., and Klein-Nulend, J. L. F., Sprague, E., and Jiang, J. X. (2005) Mechanical strain
(2006) Osteocytes subjected to pulsating fluid flow regulate opens connexin 43 hemichannels in osteocytes: a novel mech-
osteoblast proliferation and differentiation. Biochem. Biophys. anism for the release of prostaglandin. Mol. Biol. Cell 16,
Res. Commun. 348, 1082–1088 3100 –3106
45. Cowin, S. C. (2007) The significance of bone microstructure in 64. Li, J., Burr, D. B., and Turner, C. H. (2002) Suppression of
mechanotransduction. J. Biomech. 40, S105–S109 prostaglandin synthesis with NS-398 has different effects on
46. Klein-Nulend, J., van der Plas, A., Semeins, C. M., Ajubi, N. E., endocortical and periosteal bone formation induced by me-
Frangos, J. A., Nijweide, P. J., and Burger, E. H. (1995) chanical loading. Calcif. Tissue Int. 70, 320 –329
Sensitivity of osteocytes to biomechanical stress in vitro. FASEB J. 65. Alam, I., Warden, S. J., Robling, A. G., and Turner, C. H. (2005)
9, 441– 445 Mechanotransduction in bone does not require a functional
47. Tan, S. D., de Vries, T. J., Kuijpers-Jagtman, A. M., Semeins, cyclooxygenease-2 (COX-2) gene. J. Bone Miner. Res. 20, 438 –
C. M., Everts, V., and Klein-Nulend, J. (2007) Osteocytes sub- 446
jected to fluid flow inhibit osteoclast formation and bone 66. Forwood, M. R. (1996) Inducible cyclo-oxygenase (COX-2)
resorption. Bone 41, 745–751 mediates the induction of bone formation by mechanical load-
48. Westbroek, I., Ajubi, N. E., Alblas, M. J., Semeins, C. M., ing in vivo. J. Bone Miner. Res. 11, 1688 –1693
Klein-Nulend, J., Burger, E. H., and Nijweide, P. J. (2000) 67. Rubin, J., Murphy, T. C., Zhu, L., Roy, E., Nanes, M. S., and Fan,
Differential stimulation of prostaglandin G/H synthase-2 in X. (2003) Mechanical strain differentially regulates endothelial
osteocytes and other osteogenic cells by pulsating fluid flow. nitric-oxide synthase and receptor activator of nuclear kappa B
Biochem. Biophys. Res. Commun. 268, 414 – 419 ligand expression via ERK1/2 MAPK. J. Biol. Chem. 278, 34018 –
49. Young, S. R., Gerard-O’Riley, R., Kim, J. B., and Pavalko, F. M. 34025
(2009) Focal adhesion kinase is important for fluid shear 68. Turner, C. H., Takano, Y., Owan, I., and Murrell, G. A. (1996)
stress-induced mechanotransduction in osteoblasts. J. Bone Nitric oxide inhibitor L-NAME suppresses mechanically in-
Miner. Res. 24, 411– 424 duced bone formation in rats. Am. J. Physiol. 270, E634 –E639
50. Ponik, S. M., and Pavalko, F. M. (2004) Formation of focal 69. Hens, J. R., Wilson, K. M., Dann, P., Chen, X., Horowitz, M. C.,
adhesions on fibronectin promotes fluid shear stress induction and Wysolmerski, J. J. (2005) TOPGAL mice show that the
of COX-2 and PGE2 release in MC3T3–E1 osteoblasts. J. Appl. canonical Wnt signaling pathway is active during bone develop-
Physiol. 97, 135–142 ment and growth and is activated by mechanical loading in
51. Pavalko, F. M., Chen, N. X., Turner, C. H., Burr, D. B., Atkinson, vitro. J. Bone Miner. Res. 20, 1103–1113
S., Hsieh, Y. F., Qiu, J., and Duncan, R. L. (1998) Fluid 70. Robinson, J. A., Chatterjee-Kishore, M., Yaworsky, P. J., Cullen,
shear-induced mechanical signaling in MC3T3–E1 osteoblasts D. M., Zhao, W., Li, C., Kharode, Y., Sauter, L., Babij, P., Brown,
requires cytoskeleton-integrin interactions. Am. J. Physiol. 275, E. L., Hill, A. A., Akhter, M. P., Johnson, M. L., Recker, R. R.,
C1591–C1601 Komm, B. S., and Bex, F. J. (2006) Wnt/beta-catenin signaling
52. Han, Y., Cowin, S. C., Schaffler, M. B., and Weinbaum, S. (2004) is a normal physiological response to mechanical loading in
Mechanotransduction and strain amplification in osteocyte cell bone. J. Biol. Chem. 281, 31720 –31728
processes. Proc. Natl. Acad. Sci. U. S. A. 101, 16689 –16694 71. Sawakami, K., Robling, A. G., Ai, M., Pitner, N. D., Liu, D.,
53. Mullender, M. G., Huiskes, R. (1997) Osteocytes and bone Warden, S. J., Li, J., Maye, P., Rowe, D. W., Duncan, R. L.,
lining cells: which are the best candidates for mechano-sensors Warman, M. L., and Turner, C. H. (2006) The Wnt co-receptor
in cancellous bone? Bone 20, 527–532 LRP5 is essential for skeletal mechanotransduction but not for
54. Rezzonico, R., Cayatte, C., Bourget-Ponzio, I., Romey, G., Bel- the anabolic bone response to parathyroid hormone treatment.
hacene, N., Loubat, A., Rocchi, S., Van Obberghen, E., Girault, J. Biol. Chem. 281, 23698 –23711
J. A., Rossi, B., and Schmid-Antomarchi, H. (2003) Focal 72. van der Horst, G., van der Werf, S. M., Farih-Sips, H., van
adhesion kinase pp125FAK interacts with the large conductance Bezooijen, R. L., Lowik, C. W., and Karperien, M. (2005)
calcium-activated hSlo potassium channel in human osteoblasts: Downregulation of Wnt signaling by increased expression of
potential role in mechanotransduction. J. Bone Miner. Res. 18, Dickkopf-1 and -2 is a prerequisite for late-stage osteoblast
1863–1871 differentiation of KS483 cells. J. Bone Miner. Res. 20, 1867–1877

MECHANOTRANSDUCTION IN BONE REPAIR AND REGENERATION 3631


73. De Boer, J., Siddappa, R., Gaspar, C., van Apeldoorn, A., Fodde, 79. Spurney, R. F., Flannery, P. J., Garner, S. C., Athirakul, K., Liu,
R., and van Blitterswijk, C. (2004) Wnt signaling inhibits osteo- S., Guilak, F., and Quarles, L. D. (2002) Anabolic effects of a G
genic differentiation of human mesenchymal stem cells. Bone protein-coupled receptor kinase inhibitor expressed in osteo-
34, 818 – 826 blasts. J. Clin. Invest. 109, 1361–1371
74. Jansen, J. H., Eijken, M., Jahr, H., Chiba, H., Verhaar, J. A., van 80. Singh, S. P., Chang, E. I., Gossain, A. K., Mehara, B. J., Galiano,
Leeuwen, J. P., and Weinans, H. (2010) Stretch-induced inhibi- R. D., Jensen, J., Longaker, M. T., Gurtner, G. C., and Saadeh,
tion of Wnt/beta-catenin signaling in mineralizing osteoblasts. P. B. (2007) Cyclic mechanical strain increases production of
J. Orthop. Res. 28, 390 –396 regulators of bone healing in cultured murine osteoblasts. J. Am.
75. Riddle, R. C., Taylor, A. F., Rogers, J. R., and Donahue, H. J. Coll. Surg. 204, 426 – 434
(2007) ATP release mediates fluid flow-induced proliferation of 81. Baker, A. B., and Sanders, J. E. (2000) Angiogenesis stimulated
human bone marrow stromal cells. J. Bone Miner. Res. 22, by mechanical loading. Microvasc. Res. 60, 177–181
589 – 600 82. Bhatt, K. A., Chang, E. I., Warren, S. M., Lin, S. E., Bastidas,
76. Lau, K. H., Kapur, S., Kesavan, C., and Baylink, D. J. (2006) N., Ghali, S., Thibboneir, A., Capla, J. M., McCarthy, J. G.,
Up-regulation of the Wnt, estrogen receptor, insulin-like and Gurtner, G. C. (2007) Uniaxial mechanical strain: an in
growth factor-I, and bone morphogenetic protein pathways in vitro correlate to distraction osteogenesis. J. Surg. Res. 143,
C57BL/6J osteoblasts as opposed to C3H/HeJ osteoblasts in 329 –336
part contributes to the differential anabolic response to fluid 83. Reich, A., Jaffe, N., Tong, A., Lavelin, I., Genina, O., Pines, M.,
shear. J. Biol. Chem. 281, 9576 –9588 Sklan, D., Nussinovitch, A., and Monsonego-Ornan, E. (2005)
77. Zhang, Y. L., Frangos, J. A., and Chachisvilis, M. (2009) Mechan- Weight loading young chicks inhibits bone elongation and
ical stimulus alters conformation of type 1 parathyroid hormone promotes growth plate ossification and vascularization. J. Appl.
receptor in bone cells. Am. J. Physiol. Cell Physiol. 296, C1391– Physiol. 98, 2381–2389
C1399 84. Robling, A. G., and Turner, C. H. Mechanotransduction in
78. Aguirre, J. I., Plotkin, L. I., Gortazar, A. R., Millan, M. M., bone: genetic effects on mechanosensitivity in mice. (2002) Bone
O’Brien, C. A,. Manolagas, S. C., and Bellido, T. (2007) A novel 31, 562–569
ligand-independent function of the estrogen receptor is essen-
tial for osteocyte and osteoblast mechanotransduction. J. Biol. Received for publication February 21, 2010.
Chem. 282, 25501–25508 Accepted for publication May 20, 2010.

3632 Vol. 24 October 2010 The FASEB Journal 䡠 www.fasebj.org HUANG AND OGAWA

You might also like