Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Acta Biomaterialia 8 (2012) 3876–3887

Contents lists available at SciVerse ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Review

Current views on calcium phosphate osteogenicity and the translation


into effective bone regeneration strategies
Y.C. Chai a,c, A. Carlier b,c, J. Bolander a,c, S.J. Roberts a,c, L. Geris c,d, J. Schrooten c,e, H. Van Oosterwyck b,c,
F.P. Luyten a,c,⇑
a
Laboratory for Skeletal Development and Joint Disorders, KU Leuven, O&N 1, Herestraat 49, PB 813, 3000 Leuven, Belgium
b
Biomechanics Section, KU Leuven, Celestijnenlaan 300 C, PB 2419, 3001 Leuven, Belgium
c
Prometheus, Division of Skeletal Tissue Engineering, KU Leuven O&N 1, Herestraat 49, PB 813, 3000 Leuven, Belgium
d
Biomechanics Research Unit, University of Liege, Chemin des Chevreuils 1 – BAT 52/3, 4000 Liege 1, Belgium
e
Department of Metallurgy and Materials Engineering, KU Leuven, Kasteelpark Arenberg 44, PB 2450, 3001 Leuven, Belgium

a r t i c l e i n f o a b s t r a c t

Article history: Calcium phosphate (CaP) has traditionally been used for the repair of bone defects because of its strong
Received 7 April 2012 resemblance to the inorganic phase of bone matrix. Nowadays, a variety of natural or synthetic CaP-based
Received in revised form 28 June 2012 biomaterials are produced and have been extensively used for dental and orthopaedic applications. This
Accepted 3 July 2012
is justified by their biocompatibility, osteoconductivity and osteoinductivity (i.e. the intrinsic material
Available online 13 July 2012
property that initiates de novo bone formation), which are attributed to the chemical composition, sur-
face topography, macro/microporosity and the dissolution kinetics. However, the exact molecular mech-
Keywords:
anism of action is unknown. This review paper first summarizes the most important aspects of bone
Calcium phosphate
Bone tissue engineering
biology in relation to CaP and the mechanisms of bone matrix mineralization. This is followed by the
Osteoinductivity research findings on the effects of calcium (Ca2+) and phosphate (PO34 ) ions on the migration, prolifera-
Calcium ion tion and differentiation of osteoblasts during in vivo bone formation and in vitro culture conditions. Fur-
Computational modelling ther, the rationale of using CaP for bone regeneration is explained, focusing thereby specifically on the
material’s osteoinductive properties. Examples of different material forms and production techniques
are given, with the emphasis on the state-of-the art in fine-tuning the physicochemical properties of
CaP-based biomaterials for improved bone induction and the use of CaP as a delivery system for bone
morphogenetic proteins. The use of computational models to simulate the CaP-driven osteogenesis is
introduced as part of a bone tissue engineering strategy in order to facilitate the understanding of
cell–material interactions and to gain further insight into the design and optimization of CaP-based bone
reparative units. Finally, limitations and possible solutions related to current experimental and compu-
tational techniques are discussed.
Ó 2012 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction that are essential for vital physiological events, such as ion homeo-
stasis and other intracellular signalling pathways.
Bone is a dynamic, highly vascularized and mineralized tissue In fact, bone is a biocomposite tissue consisting of an organic
that has self-remodelling and healing capacities under normal phase (mainly collagen type-1 fibres, 20%) and an inorganic phase
physiological conditions, with bone loss due to disuse and upon in- (mainly carbonated hydroxyapatite (Ca10(PO4)6(OH)2), (60%) [1]
jury. It provides structural support to the body for locomotion, (Fig. 1A) that are organized in a lamellar cylindrical osteon system
serves as a protective cage for internal organs, and is a site for (i.e. the compact bone) or present as irregular thin trabecular plates
haematopoiesis and endocrine regulation. It also maintains the and struts (i.e. the spongy bone) [2]. This special organization of
acid–base balance of blood, and serves as storage for minerals collagen fibres and mineralized matrix (deposited by the bone-
(mainly calcium (Ca2+) and phosphate (PO34 )) and growth factors forming cells, i.e. the osteoblasts) renders the bone tissue with
relatively high elastic modulus and compressive strength, but low
tensile and shear strength [3,4] (Fig. 1B). These mechanical proper-
⇑ Corresponding author at: Laboratory for Skeletal Development and Joint ties are dependent on the anatomic location [5,6], and are
Disorders, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, influenced among others by the porosity and percentage of the
O&N 1, Herestraat 49, P.O. Box 7003, 3000 Leuven, Belgium. Tel.: +32 16 342541; mineral content within a bone tissue to suit a particular functional-
fax: +32 16 342543.
ity (e.g. 80% mineral content within ossicles for sound transduction
E-mail address: frank.luyten@uz.kuleuven.ac.be (F.P. Luyten).

1742-7061/$ - see front matter Ó 2012 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.actbio.2012.07.002
Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887 3877

Fig. 1. (A) Composition and (B) mechanical properties of human bone [4,6].

by vibration) [7]. In general, a higher mineral content increases the the interior of the fibril, where they solidify into an amorphous
stiffness, but decreases the toughness of the bone [8]. Recently, phase (Fig. 2A). This amorphous phase is then transformed into ori-
these stiffness and energy dissipation properties of bone were ented apatite crystals directed by the collagen fibril arrangement.
found to be attributed to calcium-mediated sacrificial bonds of a The second theory is based on the production of intracellular MV
non-fibrillar organic matrix, which act as a ‘‘glue’’ to hold the containing calcium phosphate crystals by osteoblasts. Three possi-
mineralized fibrils together (through a hidden length mechanism) ble mechanisms for the initiation of MV mediated matrix mineral-
during bone deformation [9,10]. ization have been suggested [15] (Fig. 2B): (i) MV only regulates
Indeed, calcium phosphate [11] plays a critical role in the min- ion concentrations, leading to the formation of soluble molecular
eralization of collagen fibres and thus contributes to physiologi- species which initiate mineral formation in collagen fibrils; (ii)
cally important bone tissue characteristics. Two theories of MV regulates ion compositions, leading to the formation of intra-
collagen fibre biomineralization are reported: (a) direct nucleation vesicular apatite crystals, which leave the vesicle and initiate the
of calcium phosphate crystals onto collagen fibrils [12], and (b) mineralization process; and (iii) MV associate directly with colla-
matrix vesicle (MV) mediated matrix mineralization [13,14]. Direct gen and cooperates to initiate matrix mineralization.
nucleation involves the formation of stable mineral droplets con- These essential matrix mineralization events are tightly regu-
sisting of calcium phosphate cluster–biopolymer complexes that lated by several bone-related proteins and growth factors. For in-
bind to a distinct region on the collagen fibres and diffuse through stance, alkaline phosphatase (ALP) is a periplasmic enzyme

Fig. 2. Theories of biomineralization of collagen fibril. (A) Direct nucleation of CaP crystal: (i) CaP clusters form complexes with the functional biopolymer (e.g. polyaspartic
acid) and (ii) form stable mineral droplets. (iii) The mineral droplets bind to a distinct region on the collagen fibres and enter the fibril and (iv) diffuse through the interior of
the fibril before solidifying into a disordered (amorphous) phase. (v) This amorphous phase is transformed into oriented apatite crystals directed by the collagen orientation
(modified from Colfen [12]). (B) MV-mediated matrix mineralization. Schematic showing the three possible mechanisms for the initiation of MV-mediated matrix
mineralization (adapted from Golub [15]). PPi, pyrophosphate.
3878 Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887

(membrane of cell and matrix vesicle) that hydrolyses pyrophos- and thus the polarization of cell membrane at the leading edge,
phate (a mineralization inhibitor), thereby providing phosphate which was reported to be critical in determining the persistent
ion (PO34 ) to promote mineralization [16–18]. PHOSPHO1 is an- directional cell migration [43,44].
other phosphatase highly expressed in bone [19], which is present Besides the effect on cell chemotaxis, the release of extracellular
within MV and plays a role in the initiation of mineral formation Ca2+ also plays an important role in controlling the proliferation
[20]. Osteocalcin, osteonectin, osteopontin and bone sialoprotein (via c-fos transcription factor expression) and differentiation (via
are the four major non-collagenous bone proteins. Osteocalcin dephosphorylation of NFAT transcription factor) of osteoblasts
and osteonectin were reported to regulate the size and speed of near the bone resorption site (Howship’s lacunae), through the cal-
crystal formation [21], bone sialoprotein was found to act as a crys- cium/calmodulin signalling [41]. These effects are revealed to be
tal nucleator [22], whereas osteopontin influences the type of crys- mediated by the calcium-sensing receptor (CaSR) [45], voltage-
tal formed [23,24]. In contrast, mineralization inhibitors such as gated Ca2+ channels [41,46] or inositol 1,4,5-triphosphate receptors
decorin, a member of the small leucine-rich proteoglycans family, [47] present in the osteoblast, which serve to increase the intracel-
negatively interfere with mineralization by modulating collagen lular Ca2+ level. In recent years, in vitro studies, based on the
assembly [25]. Another inhibitor, Matrix Gla Protein, was associ- addition of elevated Ca2+ levels into osteoblastic cell cultures
ated with parathyroid hormone-mediated inhibition of osteoblast (2–8 mM), demonstrated a profound impact on bone cell fate,
mineralization [26]. Recently, bone-morphogenetic protein-2 which was independent of systemic calciotropic factors in a con-
(BMP-2), a potent osteoinductive growth factor, was found to be centration-dependent way [42,48]. These include osteoblast che-
involved in the control of ALP expression and osteoblast minerali- motaxis [36], DNA synthesis [49], proliferation, differentiation
zation via a Wnt autocrine loop [27], as well as in the enhancement [50] and mineralization of extracellular matrix [51,52]. Interest-
of PO34 transportation into cells for matrix mineralization [28]. ingly, Ca2+ also induced the expression of osteogenic growth fac-
Fibroblast growth factor-2 (FGF-2), is another growth factor that tors, such as parathyroid hormone-related peptide [53], BMP-2
was found to reduce the expression of genes associated with ma- and BMP-4 [52]. Similar effects were observed when osteoblasts
trix mineralization [29]. were cultured on Ca2+-functionalized biomaterials, such as nano-
crystalline CaP glass [54], Ca2+ implanted titanium substrate [55],
Ca2+-exposed collagen gel [51] and CaP-coated implants [56,57].
2. Effect of calcium (Ca2+) and phosphate (PO34 ) on osteoblastic In vivo osseointegration and bone formation were also improved
cell behaviour and its applications when implants were enriched with calcium ions [58]. Encourag-
ingly, Ca2+ has been implicated recently as an important messenger
This section will describe primarily (pre)osteoblasts, since these involved in the non-canonical b-catenin-independent Wnt/calcium
cells play a key role during osteogenesis. Notice that the extracel- signalling for bone formation [59]. This pathway relies on an intra-
lular calcium ion (Ca2+) can be bound to proteins (e.g. albumin). In cellular release of Ca2+ to activate calcium sensitive enzymes such
this complex form, the ion cannot influence cellular behaviour as Ca2+–CaMKII, protein kinase C or calcineurin.
such as differentiation and proliferation [30,31]. The terms ‘‘Ca2+’’ Essentially, the release of PO34 at the resorption site also plays a
and ‘‘PO34 ’’ refer, in the remaining part of this paper, to the active, role in osteoblast proliferation and differentiation. In fact, PO34 has
free ions. been identified as an important signalling molecule that regulates
During in vivo bone resorption, osteoclasts release Ca2+ and cell cycle and proliferation rate, alteration of signal transduction
PO34 derived from bone matrix. This causes a local increase in pathways (e.g. Fos-related antigen-1 (Fra-1) [60] and extracellular
the ion concentration to supra-physiological levels, which has a signal-regulated kinase (ERK1/2) [61]), gene expression (e.g. osteo-
significant impact on the proliferation and differentiation of osteo- pontin) [62], as well as the secretion of bone-related proteins (e.g.
blasts, as well as on the subsequent bone formation process. In fact, matrix Gla protein (MGP) [63]). In contradiction, several in vitro
extracellular Ca2+ gradients are present in a number of distinct studies showed that addition of high concentrations of exogenous
microenvironments and represent potent chemical signals for cell inorganic phosphate (Pi, range from 5 to 7 mM) induced in vitro
migration (chemotaxis) and directed growth [32]. Additionally, osteoblast apoptosis and non-physiological mineral deposition
Ca2+ is an important homing signal, which brings together different [64]. Nevertheless, Pi is believed to play a critical role in physiolog-
cell types required for the initiation of a multicellular process such ical bone matrix mineralization [65]. This mineralization event is
as bone remodelling or wound repair [33]. For instance, high Ca2+ mediated by the enzyme ALP and has been associated with the
concentrations are shown to stimulate pre-osteoblast chemotaxis bone matrix calcification induced by BMP-2, where BMP-2 was
to the site of bone resorption, and their maturation into cells that found to stimulate Pi transport by osteogenic cells primarily via
produce new bone [34]. This chemotactic response to Ca2+ was also the sodium-dependent phosphate transporters [28]. Unfortu-
demonstrated experimentally on different cell types, including nately, the use of Pi-functionalized biomaterials for tissue engi-
monocytes [35], osteoblasts [36], haematopoietic stem cells [37] neering applications is rarely described, possibly because of the
and bone marrow progenitor cells [38]. The latter reported a technical limitations of existing technologies on handling Pi in an
dose-dependent relationship, with a maximal effect achieved at ionic form or the lack of awareness on the potential use of Pi. How-
concentrations from 3–10 mM of Ca2+. These findings indicate that ever, the use of polymers mixed with phosphate salts as a sus-
extracellular Ca2+ is a coupling factor between osteoclasts and tained delivery of Pi to induce in vivo mineralization of the
osteoblasts [39] (apart from other potential coupling factors, such carrier has been reported recently [66].
as mechanical coupling [40]). Mechanistically, extracellular Ca2+ Despite the vast in vitro research findings, the influences of Ca2+
regulates the migration of osteoblasts via the activation of calcium and PO34 differ from cell type to cell type [67]. This implies that
sensing receptors (CaSR) and/or by increasing the influx of Ca2+ there will be not one optimal Ca2+ and Pi concentration that could
[41]. In fact, the CaSR is reported to act as a (gradient) sensor, thus universally drive all cell types toward successful osteogenesis.
triggering chemotaxis of motile cells to critical microenvironments Moreover, the optimal concentration may vary according to the
and transducing the Ca2+ signal to intracellular signalling pathways cellular stage including proliferation and differentiation. Therefore,
that regulate cell function [33]. Interestingly, studies suggest that specific windows of ion concentration need to be determined for a
the CaSR in osteoblasts is functionally similar to, but molecularly specific cell type and its cellular stage, in order to initiate the de-
distinct from, the CaSR present in the parathyroid and the kidney sired in vitro cell behaviour effectively. For instance, the present
[41,42]. Whereas, the influx of Ca2+ elevates intracellular Ca2+ level authors’ group has recently reported on the identification of
Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887 3879

specific Ca2+ and Pi concentrations that could induce higher prolif- CaP-based biomaterial with higher osteoinductivity also targets
eration and osteogenic differentiation of a mesenchymal stem cell- CaP with an appropriately high solubility. This may be more effec-
like human osteoprogenitor [68]. These findings were translated tive in stimulating osteogenic differentiation of stem cells and ini-
into the formulation of bio-instructive media (containing specific tiating bone formation. However, the exact mechanism of
concentrations of Ca2+ and Pi) that could optimally initiate higher osteoinduction by CaP is currently unknown [67,71,76].
proliferation, osteogenic differentiation and bone-like matrix In the mid-1960s, osteoinduction was described as ‘‘a process
deposition, in both two-dimensional (2D) cultures and three- which supports the mitogenesis of undifferentiated perivascular
dimensional (3D) constructs. This represents a novel strategy to mesenchymal cells leading to the formation of osteoprogenitor
produce 3D bone reparative units that may have predictive osteo- cells with the capacity to form new bone’’ [77]. Since the discovery
inductivity for an effective repair of large skeletal defects [69]. of bone morphogenetic proteins (BMP) as potent inducers of ecto-
pic bone formation [78], the term ‘‘osteoinduction’’ has generally
been used to describe an observation of heterotopic bone forma-
3. Rationale for using calcium phosphate for bone regeneration
tion in association with the in vivo ectopic bone inductivity of a
substance, such as growth factors, chemical compounds or bioma-
Bone is a remarkable organ, as it has impressive self-healing
terials. In pathophysiological conditions, ectopic bone formation
capacity without scar formation (when the defect size is not criti-
was induced upon tissue calcification in vivo, such as in tendons
cal). However, delayed healing and non-unions still often develop
and arteries [79]. This phenomenon was associated with the oste-
and will occur more frequently as a result of the ageing of the pop-
ogenic differentiation of the calcified tissue [80], including the
ulation. In the US, approximately six million fractures occur yearly,
expression of BMP-2 by the calcifying cells [81]. In the context of
of which 5–10% develop into a delayed union or non-union. An
osteoinduction by synthetic biomaterials, investigations over past
extrapolation of these numbers to the Indian population results
decades suggest that (i) the presence of a CaP component within
in 240 million fractures a year, of which 12 million are non-unions
a biomaterial (e.g. CaP-based ceramics [82], composite [83], coat-
[70]. Therefore, the need for bone tissue regeneration is continu-
ing [84], coral-derived ceramic [85] and bioactive glass [86]), or
ously increasing, and the emergence of combined engineering
(ii) the ability of a non-CaP-containing biomaterial to induce
and life sciences technologies, such as tissue engineering, may lead
in vivo calcification (e.g. poly-hydroxyethylmethacrylate sponge
to more effective bone-healing therapeutic modalities.
[87], and chemical-treated oxidized titanium substrate [88]) are
Bone tissue engineering aims to offer a better solution for the
prerequisites for heterotopic direct bone formation.
healing of large bone defects and non-unions. This interdisciplinary
Recently, the osteoinductive effect of CaP-based biomaterials
research field applies principles of engineering and life sciences to
and the in vivo host–biomaterial interactions were reviewed to
create an in vivo microenvironment that promotes local bone re-
gain insight into the physicochemical properties that may govern
pair or regeneration [71,72]. In this context, CaP bioceramics ap-
osteoinduction [72,89]. These include the effects of the macro-
pear to be interesting candidates for bone tissue engineering
structures (e.g. dimension, geometry and porosity), the micro/nano
applications, owing to their biomimetic properties, supported by
structures [90] (e.g. microporosity [91], grain size, surface topogra-
the following findings. First, in the event of endochondral ossifica-
phy), and the chemical composition and characteristics of the bio-
tion, mineralized cartilaginous matrix is reported to induce osteo-
materials (e.g. active chemical surface for apatite formation, and
progenitor differentiation and thereafter bone matrix deposition
dissolution kinetics of CaP biomaterials). Additionally, the in vivo
[73]. This phenomenon has been further investigated in order to
osteoinductivity by CaP was reported to be dependent on the ani-
elucidate the necessary physicochemical properties of CaP that
mal model used (higher incidence in large animals, possibly due to
may effectively trigger cellular signalling cascades for bone forma-
higher osteoclastic activity [92]), implantation site [93] and the
tion [74]. Secondly, the bone matrix calcification or mineralization
duration of implantation [85] (higher incidence and faster in intra-
process is a critical stage of bone formation, either through direct
muscular than subcutaneous implantation). At present, the hypo-
mineralization of bone matrix or the preformation of cartilaginous
thetical mechanisms of CaP-driven osteoinduction from a
tissue template that is mineralized at a later stage. Thirdly, the dis-
material perspective can be classified into four categories.
solution of CaP-based biomaterials, either physicochemically or
cell mediated upon implantation, may also resemble the physio-
4.1. Direct effect of the biomaterial
logical bone resorption process [75]. Finally, the data discussed
in the previous section show that the effects of Ca2+ and Pi on oste-
The physical properties of a CaP-based biomaterial, such as
ogenic cell behaviour are appreciable. Therefore, the rationale of
geometry, macroporosity, microporosity, surface topography and
using CaP-based biomaterials for bone engineering strategies is
grain size, have, in combination with its chemical properties, a sig-
clear.
nificant impact on: (i) the nutrient, oxygen and waste exchange for
cells within a biomaterial; (ii) the host blood vessel in-growth; (iii)
4. Historical and hypothetical mechanisms of calcium the total volume of open pores available for cell growth and bone
phosphate osteoinductivity tissue formation; (iv) the effect on osteochondrogenic differentia-
tion of stem cells [94]; (v) the total surface area available for pro-
Carbonated hydroxyapatite (HA) is the prevalent form of CaP tein adsorption (e.g. endogenous BMP), cell attachment (which
mineral found in the bone. It provides mechanical strength to the activates a BMP-2 autocrine loop via a2b1 integrin [95]) and
bone and plays a critical role in the mineralization of the bone ma- growth; and (vi) ion dissolution and re-precipitation [93], which
trix. Because of chemical and biological similarities, HA derived contributes to the formation of a biological apatite layer which,
from natural sources (e.g. bone allograft, autograft or coral) or syn- in turn, stimulates the osteogenic differentiation of stem cells.
thetic HA, is widely used as bone filler for treating skeletal defects.
However, HA is a highly stable CaP mineral and therefore has lower 4.2. Indirect effect of the biomaterial
solubility at the physiological pH (7.2–7.6) compared with other
types of CaP that have higher solubility (such as tricalcium phos- Osteoinductive proteins such as BMP and transforming growth
phate (TCP) and octacalcium phosphate (OCP)). Because the disso- factor-beta are known to have a high affinity to CaP [96]. It has
lution behaviour has been associated with the osteogenicity as been hypothesized that CaP-based biomaterials may act as an
well as the osteoinductivity of CaP [75], the search for an improved in vivo affinity column/concentrator of the endogenous
3880 Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887

osteoinductive molecules upon implantation, which then renders area induce higher in vitro ALP activity and are capable of concen-
the biomaterial osteoinductive. This is supported by a study which trating more osteoinductive molecules that differentiate cells into
showed ectopic bone induction by CaP ceramics implanted the osteogenic lineage [113]. A similar phenomenon was observed
intramuscularly (without cells) in proximity to the fractured fibula, in an in vivo study in which CaP carriers with lower CaP grain size
where osteogenic events were actively ongoing [97]. and higher microporosity (and thus higher ionic dissolution kinet-
ics) were found to be osteoinductive without using stem cell tech-
4.3. Inflammatory response nology [75]. The surface area can also be increased by the
production of CaP microparticles, as their osteoinductive proper-
Upon implantation, CaP-based biomaterials elicit an inflamma- ties are highly correlated to their size. It was shown that micropar-
tory response that attracts the infiltration of mono- and multinu- ticles with size ranges from 80 to 300 lm demonstrated ectopic
cleated cells, and subsequently activates osteoclastogenesis, bone formation, whereas particle sizes >500 lm were not osteoin-
which results in CaP degradation and resorption [98]. Subse- ductive [114,115]. Indeed, several important in vivo studies have
quently, the released Ca2+ and PO34 stimulate osteoprogenitor shown that a CaP-based biomaterial with specific surface area
differentiation and bone matrix deposition [99,100]. above a threshold level of 1.0 m2 g 1 was critical for osteoinduc-
tion (Fig. 3). Unfortunately, the availability of a technology to en-
able high controllability and reproducibility of these desired
4.4. Pathological ossification/bone formation
features is currently lacking.
Also biphasic CaP (BCP) composites, being mixtures of HA with
Osteoinduction by CaP-based biomaterials may resemble a
CaP with higher solubility (such as b-TCP) in a specific ratio, appear
pathological condition of heterotopic bone formation due to tissue
to be an effective alternative to enhance the osteoinductivity of CaP
calcification in vivo, resembling atherosclerosis or calcified ten-
[116]. The introduction of a more soluble phase of CaP may stimu-
donitis. The bone tissue formed often lacks functionality and even-
late osteogenic differentiation of osteoprogenitors [117,118] and
tually may resolve over time. This resorption process may be
meanwhile provide a stable phase of CaP (i.e. the HA), which
related to a foreign body reaction or be due to the depletion of
may act as a homing site for the implanted cells and promote bone
biochemical cues upon implantation.
ingrowth [119]. Indeed, implantation of pure HA or TCP was shown
to be not osteoinductive, due to either too high stability or too high
5. Translation of calcium phosphate osteogenicity for bone a dissolution rate [120], whereas implantation of BCP induced bone
tissue engineering formation with a low inflammatory response, with the newly
formed bone being sustainable in vivo during a long-term animal
In bone tissue engineering, two main forms of CaP materials are study [121]. Unfortunately, no optimized HA:TCP ratio has been re-
used: (i) bulk (fully dense or open porous) CaP biomaterials (e.g. ported until now [122]. This is mainly due to the large variations in
bone fillers, carriers, cement) [101], either as pure CaP or as part the bone-forming capacity in studies using BCP with different
of a composite [102], and (ii) CaP coatings to functionalize bioma- HA:TCP ratios, which is also species and material specific [76]. Ow-
terial surfaces [103]. The use of CaP as growth factor delivery sys- ing to the lack of mechanical strength, these biphasic CaP biomate-
tem [104] is also discussed, specifically BMP-incorporated CaP rials are more suitable for the treatment of skeletal defects at non-
biomaterials for bone formation [105]. load-bearing sites, such as bone fillers for cranial defects. Never-
theless, BCP alone was recently reported in the clinic to heal large
5.1. Bulk CaP bone defects successfully, together with the use of an external fix-
ator [123].
The physicochemical properties of bulk CaP are often modified
during the in vitro production process, aiming to find the optimal
construct geometry for CaP scaffolds that could enhance osteoin-
ductivity and hence improve the clinical outcome [106]. This in-
cludes manipulation of the architecture of biomaterials, such as
the geometry, macro- and microporosity and surface topography
[107]. The aim is to adjust the surface area for the entrapment of
endogenous osteoinductive biomolecules and to accommodate
optimum osteogenic cell density, as well as to fine-tune the disso-
lution kinetics that optimally elicit osteogenic differentiation and
bone matrix deposition. For instance, production of cylindrical
HA scaffolds with a hollow centre (2–4 mm) were reported to en-
hance ectopic bone formation [108]. Also, incorporating HA scaf-
folds with cylindrical tunnels of 90–120 lm and 350 lm induced
endochondral and intramembranous ossification [109]. The use
of biphasic CaP (BCP) scaffolds (HA:TCP = 65:35 wt.%) with cubic
pores of 500 lm resulted in the highest bone formation compared
with the scaffold with lower (100 lm) or higher (1000 lm) pore
sizes [110]. Besides that, the surface topography (i.e. surface
roughness) has also been reported to influence the pattern of bone
formation within CaP-based biomaterials [111]. Moreover, the sur-
face topography is beneficial for the formation of bone-like apatite
and provides a surface area that increases the dissolution of Ca2+ Fig. 3. The influence of specific surface area of CaP-based biomaterials on in vivo
and PO34 ions [112]. This surface area was later found to be asso- ectopic bone formation. Each data point represents one type of CaP implant with
specific surface area and its in vivo bone forming capacity. Based on these studies, a
ciated with the microporosity within the macropores of CaP, which threshold level of specific surface area required to induce bone formation was
affected the material–fluid interface dynamics and triggered oste- chosen at 1.0 m2 g 1 (the cut-off point). Data adapted from Habibovic et al. [91], Li
ogenic differentiation [91]. Indeed, CaP discs with larger surface et al. [113] and Yuan et al. [75].
Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887 3881

5.2. CaP coatings on biomaterials undesirable locations. Moreover, the carrier composition may af-
fect the regenerative response by BMP, as it may modulate protein
Owing to the brittleness of CaP, it is often combined with a stability and variate release kinetics differently. Nonetheless,
mechanically superior biomaterial such as titanium. This is neces- Langer et al. showed as early as 1976 that protein release could
sary, especially in designing an osteoinductive composite for the be sustained when BMP were encapsulated in biocompatible bio-
repair of skeletal defects that receive high mechanical loading polymers [141]. Therefore, an attractive approach is to investigate
in situ [124]. To date, various methods have been proposed for carriers that may favour spatiotemporal physiological protein re-
depositing CaP onto the surface of Ti-based biomaterials [125], lease, but yet promote recruitment of endogenous progenitor cells.
including a recent method based on the immobilization of chemi- These carriers also need to provide a suitable microenvironment
cal moieties to induce mineralization on the surface of biomateri- that promotes efficient cell proliferation and differentiation, as this
als both in vitro [69] and in vivo [126]. These methods aim to optimally may lead to bone formation and turnover [109,136]. In
produce composites containing mineralized components that addition to the collagen sponge used in clinical settings today,
strongly mimic the hierarchies and bioactivity of the mineralized studies have shown promising results from the use of osteocon-
compositions of the bone, in order to provide an inductive environ- ductive and osteoinductive CaP carriers [11,68,142,143]. These car-
ment that is capable of triggering bone formation. This again aises riers possess suitable characteristics (e.g. the interconnected
the principle question regarding what type of CaP has to be depos- microporosity and the variation in CaP ratio) that are critical for
ited onto the biomaterial of choice to ensure sufficient and predic- the release kinetics of BMP, as the electrostatic energy possibly
tive osteoinductivity. Therefore, the controllability and plays a dominant role in carrier-to-BMP interactions (high binding
reproducibility of a technique that deposits CaP with desirable affinity of BMP to CaP), as well as in up-scaled constructs where
physicochemical properties, in the context of achieving optimum multiple carriers may be combined [144,145].
osteoinductivity, is highly demanded. A second consideration is Even though CaP carriers in combination with BMP sounds like
the applicability of a deposition technique to 3D porous structures. a promising approach, an impediment may be variation in acti-
This represents a major drawback for many of the existing meth- vated bone-forming pathways. The osteoinductive effect of CaP
ods, where deposition of CaP onto complex 3D structures is techni- carriers has been shown to form bone mainly via an intramembra-
cally challenging [127]. For instance, in an effort to overcome this nous pathway, whereas BMP induce bone formation mainly via the
major drawback, the present authors’ research group has devel- endochondral pathway. In 2010, however, Eyckmans et al. showed
oped perfusion electrodeposition technology to deposit CaP coat- a close connection between CaP-induced bone formation and BMP
ing homogeneously onto a complex 3D Ti-scaffold porous signalling [71]. In this study, removal of CaP-granules resulted in
structure, which offers high controllability and reproducibility over loss of bone formation in combination with rescinded BMP signal-
the physicochemical properties of the deposited CaP coatings [127] ling as well as abrogated osteoinduction in the construct after inhi-
and displayed a promising osteoinductive effect [128]. bition of endogenous BMP [146,147]. This study displayed that CaP
carriers affect BMP signalling in a model similar to physiological
5.3. BMP-incorporated CaP carriers intramembranous bone formation in a BMP- and CaP-dependent
manner.
As an alternative to existing bone grafting techniques, CaP car- Another hurdle could possibly be that CaP may inhibit the
riers have been investigated for enhancing bone formation through osteoinductive effect of BMP, already displayed by Urist et al.
mediated delivery of bone-inducing factors (including biomole- where partially demineralized bone induced inflammation and
cules and metallic ions [129]), with some promising results re- inhibited osteogenesis [77]. Additional complications regarding
ported to date [130]. An example of such factors are the BMP, these issues were shown by Vehof et al., where the osteoinductive
discovered by Urist et al. as potent proteins with the ability to in- ability in recombinant human BMP-2 (rhBMP-2) coated porous
duce heterotopic bone formation [77]. These proteins are members particles of HA (PPHA) were investigated [148]. Prior to the study,
of the TGF-b family which are secreted by chondrocytes, osteo- PPHA had displayed some ability to induce de novo formed bone
blasts and osteoclasts [131–134], and play pivotal roles throughout through the intramembranous pathway [149,150]. Remarkably,
embryonic skeletogenesis and in postnatal bone formation and PPHA in combination with large amounts of rhBMP-2 in this study
endogenous repair mechanisms [130]. Indeed, BMP are attractive displayed an ability to attract a greater number of inflammatory
for treatments of critical bone defects such as spinal fusions and cells, which increased in time, together with multinucleated cells.
non-unions, and have been successfully combined with substrates Absence of collagen type II prior to hypertrophic chondrocyte dif-
in order to induce bone formation by recapitulating molecular cas- ferentiation and mineralization of the formed cartilage tissue, to-
cades during skeletal development [135–137]. For instance, BMP-7 gether with lack of bone marrow formation was seen. This
(also called osteogenic protein-1 or OP-1) and BMP-2, have been suggests that the de novo formed bone in this construct was not
approved for clinical use and are delivered for spinal fusion and following the usual BMP-induced endochondral pathway, there-
open tibial fractures via bone-derived collagen particles or an fore indicating an intermediate pathway that may have been
absorbable collagen sponge [138]. Recently, these proteins have activated [148].
been studied extensively within the field of tissue engineering in In conclusion, BMP mainly affect bone formation via an endo-
order to mimic natural bone formation from a tissue engineering chondral process, whereas CaP carriers mainly induce an intra-
point of view. When implanted in animal models, these growth membranous pathway. Possibly, the combination of appropriate
factors mainly induce bone formation via the endochondral path- concentrations of specific BMP and well-defined characteristics in
way, through the formation of an intermediate cartilage template CaP carriers may function through an intermediate bone forming
[135,139]. Unfortunately, the currently used delivery system for pathway.
BMP causes rapid protein release and diffusion, which besides
inducing bone formation, also causes inflammation and excessive
bone formation. Consequently, the uncontrollable or improper re- 6. Computer modelling of bone regeneration in CaP scaffolds
lease kinetics resulted in undesirable side effects such as male ste-
rility, cancer and brain injury [140]. An additional reason for this Improvements in computer capacity now enable an increased
could be receptor saturation at higher doses or limited availability model realism and complexity (e.g. 3D calculations, complex
of responding cells, which can lead to stimulation by these BMP at geometries, multi-scale and multi-physics) [151]. As a consequence
3882 Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887

of this technological revolution, there has been an enormous resulting in an overall volumetric reduction, while the scaffold
increase in the use of mathematical models in biology and medicine. geometry remains unaltered. Their calculations show that, as scaf-
These mathematical models can propose and test possible biological fold degradation progresses, the regenerating tissue must take over
mechanisms, contributing to the unravelling of the complex nature the mechanical function of the bone–scaffold system, which would
of biological systems, such as bone regeneration processes inside otherwise collapse due to a lack of mechanical strength. Moreover,
CaP structures. Moreover, they can be used to design and test all three variables (i.e. porosity, Young’s modulus and dissolution
possible experimental strategies in silico before they are tested rate) appear to influence the amount of bone formation in a non-
in vitro or in vivo [152,153]. Eventually, all this knowledge can be intuitive way, demonstrating the need to optimize scaffolds for
used to develop clinically relevant CaP-based bone reparative units site-specific loading requirements. This model was improved by
(Fig. 4). including blood vessel growth, thereby establishing a framework
Currently, there are many computational models of bone forma- to investigate the effect of vascularization on bone formation [159].
tion and regeneration in general [154] or even in scaffolds specifi- Other studies have modelled the bone regeneration process in-
cally [11]. Bohner et al. propose a theoretical approach to side biodegradable polymer-based scaffolds. Stops et al. further
determine the effect of geometrical factors on the resorption rate investigated the influence of mechanical strain and perfusive fluid
of CaP scaffolds [155]. Their theoretical model was based on five flow on cell differentiation and proliferation within a collagen–
assumptions: (i) the sphericity of the pores; (ii) face-centred cubic glycosaminoglycan scaffold [160]. Sanz-Herrera et al. presented a
packing of the pores; (iii) surface-controlled resorption; (iv) multi-scale model of bone regeneration inside a porous scaffold
resorption requires the presence of blood vessels (50 lm in diam- [161]. The degradation mechanism of the biodegradable polymer
eter); and (v) the resorption time is proportional to the net amount scaffold was modelled as a hydrolysis process, i.e. the water
of material [156]. The model calculations show that, based on these content in the polymer chemically reacts and breaks down the
assumptions, the resorption time of a macroporous block depends polymer molecules, resulting in biomaterial bulk erosion. The
on the pore radius, which is determined by the size of the bone mechanical properties of the polymer were assumed to relate lin-
substitute and interpore distance [156]. Subsequently, the model early to its molecular weight. The model was used to predict the
was used to optimize the pore size of CaP scaffolds and was evolution of the bone formation process in a scaffold implanted
validated with experimental data. in the femoral condyle of a rabbit. They found good qualitative
The theoretical model mentioned above, however, looks exclu- agreement between the obtained computational and experimental
sively at geometrical scaffold properties and does not include bio- results. Although further validation is necessary, the proposed
logical variables such as cell or matrix densities. Byrne et al. multi-scale model is a useful tool for investigating the complex
developed a 3D mechanoregulatory model of bone regeneration phenomena that occur at different length and time scales, i.e. the
in a regular scaffold to investigate the effect of porosity, Young’s bone formation and scaffold resorption at the microscopic scale
modulus and dissolution rate on bone regeneration in different and the change in mechanical properties at the macroscopic scale.
loading conditions [157]. They modelled the scaffold as a poroelas- Lacroix et al. reviewed the current techniques used for scaffold
tic material, which resorbs in a linear, load-independent fashion, development: from scaffold optimization of scaffolds by mathe-
i.e. the porosity will be increased by 0%, 0.5%, 1% per iteration for matical models (e.g. FEM) to scaffold design using computer-aided
low, intermediate and high dissolution rates, respectively [158]. design and scaffold characterization by computed tomography (CT)
Consequently, the size of all scaffold elements decreases uniformly, [162].

Fig. 4. The concept of integrating manufacturing process, stem cell technology and computational modelling as a novel strategy for designing and optimizing a CaP-based
bone reparative unit for effective bone regeneration that can also assist the translation to personalized bone regeneration.
Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887 3883

Although the above models can be used to optimize some dynamics of body fluids and blood vessel in-growth, are technically
(mechanical) properties of scaffolds, e.g. the porosity, the microar- challenging to be translated into a simplified in vitro setting. Re-
chitecture, Young’s modulus and dissolution rate, they neglect the cently, the reactivity of CaP-based biomaterials in culture medium
influence of growth factors and other biochemical signals on the was revealed to have significant influence on Ca2+ and PO34 disso-
bone formation process. Moreover, the dissolution process is only lution kinetics and the subsequent cellular behaviour, which was
crudely modelled, neglecting the influence of the degradation not correlated to the ion dissolution behaviour evaluated in simu-
products (e.g. Ca2+ and Pi) on the cellular activities and bone forma- lated body fluid (SBF) or phosphate-buffered saline. This must be
tion processes. Carlier et al. developed and implemented an exper- carefully considered when evaluating the osteoinductive potential
imentally informed bioregulatory model of the effect of calcium of the material [167]. The present authors propose the standardiza-
ions released from CaP-based biomaterials on the activity of oste- tion of in vitro dissolution tests to evaluate the ion release kinetics
ogenic cells and mesenchymal stem cell driven ectopic bone for- of a CaP-based biomaterial, where the in vitro dissolution experi-
mation [163]. The dissolution kinetics of the CaP scaffold were ment should resemble and be customized to the in vivo environ-
modelled by a general empirical equation [164], assuming that ment as close as possible [156]. First, the CaP scaffolds should be
the dissolution rate is proportional to the driving force (i.e. the dif- thoroughly characterized (porosity, pore size distribution, grain
ference between the current and the saturated calcium concentra- size, surface topography and surface area, and composition) before
tion) and that the rate constant is time independent. The amount of and after the dissolution test. Here, the highlight is on the use of
bone formation predicted by the model of Carlier et al. corre- non-invasive methods, including Raman spectroscopy [158] and
sponded to the amount measured experimentally under similar micro- or nano-focus X-ray CT to obtain quantitative characteriza-
conditions. Moreover, experimentally impaired bone formation tion of the material properties [168]. Secondly, a dual solution sys-
due to conditions such as insufficient cell seeding and scaffold tem is proposed for in vitro dissolution testing using SBF [169] and
decalcification, was also retrieved in silico. Subsequently, this mod- culture medium (with and without the presence of cells) [170].
el was used to optimize CaP scaffold selection to make their use in This will allow for the characterization of the intrinsic ionic disso-
combination with cells more clinically relevant. lution behaviour of the biomaterial in a solution with similar ionic
Although computational models can contribute to the general composition to the bodily fluid, followed by understanding of the
knowledge on bone formation inside CaP structures and are useful interactions or the influences of the cells and proteins on the ionic
for customizing the CaP carriers to patient-specific needs as well as dissolution kinetics. In fact, more and more studies have shown the
the particular bone application, experimental research is necessary reduction in Ca2+ in the culture medium, indicating the formation
to establish and validate the mathematical models. The most of new CaP crystals onto the existing CaP substrate [128]. Thirdly,
important parameters and their respective parameter values the experimental setting should also account for the local in vivo
should be experimentally quantified. Consequently, the multidisci- hydrodynamic of the bodily fluid: for instance, at the implantation
plinary problem of optimizing scaffold architecture and seeding site. This can be achieved by performing the dissolution test under
protocols for bone tissue engineering strategies requires an integra- dynamic conditions (e.g. using perfusion system) at a physiologi-
tive approach, which was nicely summarized by Bohner et al. [165]: cally relevant flow rate [118,127].
a combination of mathematical modelling to explain a mechanism Moreover, the in vitro and in vivo experiments should be de-
of biomaterial–cell interactions, with experimental research to pro- signed in such a way that they minimize variability and enable
vide data for the determination of model parameters as well as the quantification, thus providing the essential data for the determina-
validation of the mathematical model. This integrative approach re- tion of model parameters as well as for the model validation. It is of
quires careful design and extensive characterization of the scaffold. note that, as mathematical models predict the dynamics at differ-
Moreover, this process is intrinsically iterative: new experimental ent scales (e.g. molecular, cellular and tissue) as a function of time
results can be fed into the model, and new research hypotheses and space, temporal and spatial quantitative data are crucial to the
can result from thorough model analysis. success of mathematical models. Clearly, computational modelling
plays an essential role to further unravel the complex mechanism
of CaP osteoinduction in vivo. Most of the current models look
7. Limitations, future perspectives and conclusion either at mechanoregulatory or bioregulatory stimuli, depending
on the specific research question that is being answered. In the fu-
As discussed above, it is clear that bone induction by CaP bio- ture, however, these models could be combined to further improve
materials is influenced by the physicochemical properties of the the predictive capabilities of the model.
material and thus the subsequent cellular events of osteogenesis. The limitations mentioned above underline the importance of
However, the exact key determinant(s) of CaP osteoinduction, an interdisciplinary strategy for the optimization of CaP scaffolds
meaning the molecular mechanisms involved and the stem for bone tissue engineering applications [165]. An essential charac-
cell–material–host interactions upon implantation is/are still teristic of these integrative research efforts is that their iterativity:
underdetermined. Therefore, further study to decipher the molec- model analysis can lead to new research hypotheses, and new
ular signalling at the cellular level (such as receptor binding of the experimental findings can be used to improve the predictive capac-
released Ca2+ and PO34 and the activation of critical intracellular ities of the model.
osteogenesis pathway), and to understand the critical biological In conclusion, this review has discussed the importance of CaP
parameters that are essential for implanted cells to communicate for physiological bone homeostasis as well as its potential for bone
and integrate effectively with the host system are required [166]. tissue engineering. Although numerous studies have been devoted
Unfortunately, the translation of the complex in vivo osteogenesis to unravelling the mechanisms of osteoinductivity of CaP scaffolds,
environment into an in vitro system is far from trivial, and the pre- many questions still remain unresolved. To overcome this bottle-
dictiveness of in vitro observations often does not correlate well neck, it is therefore essential that experimental and computational
with the in vivo bone formation capability [89]. This is due to some research are combined so that the complex in vivo biological pro-
critical issues such as the use of correct cell types for a specific type cess of bone regeneration inside CaP constructs can be deciphered
of CaP-based biomaterial and the selection of the correct culture in an effective and systematic manner. This includes computa-
conditions. Therefore, there is a need for customization of the tional modelling and correlation analysis between the physico-
CaP-based biomaterial to overcome these limitations. Moreover, chemical properties of CaP constructs and the influences on
other parameters present in vivo, including the compositions and in vitro and in vivo biological outcomes, thus identifying the
3884 Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887

critical parameters and obtaining a thorough understanding of the [22] Monfoulet L, Malaval L, Aubin JE, Rittling SR, Gadeau AP, Fricain JC, et al. Bone
sialoprotein, but not osteopontin, deficiency impairs the mineralization of
underlying biology that governs osteoinductivity of CaP constructs.
regenerating bone during cortical defect healing. Bone 2010;46:447–52.
Only in this way will it be possible to make CaP a clinically relevant [23] Roach HI. Why does bone matrix contain non-collagenous proteins? The
and predictive tissue engineering construct for effective bone possible roles of osteocalcin, osteonectin, osteopontin and bone sialoprotein
defect repair. in bone mineralisation and resorption. Cell Biol Int 1994;18:617–28.
[24] Rosenthal AK, Gohr CM, Uzuki M, Masuda I. Osteopontin promotes pathologic
mineralization in articular cartilage. Matrix Biol 2007;26:96–105.
Acknowledgements [25] Mochida Y, Parisuthiman D, Pornprasertsuk-Damrongsri S, Atsawasuwan P,
Sricholpech M, Boskey AL, et al. Decorin modulates collagen matrix assembly
and mineralization. Matrix Biol 2009;28:44–52.
This work is funded by the KU Leuven IDO project 05/009- [26] Gopalakrishnan R, Suttamanatwong S, Carlson AE, Franceschi RT. Role of
QuEST, Stem Cell Institute of Leuven—KU Leuven, ENDEAVOUR matrix Gla protein in parathyroid hormone inhibition of osteoblast
project G.0982.11N. Aurélie Carlier and Johanna Bolander are mineralization. Cells Tissues Organs 2005;181:166–75.
[27] Rawadi G, Vayssiere B, Dunn F, Baron R, Roman-Roman S. BMP-2 controls
Ph.D. fellows of the Research Foundation Flanders (FWO Vlaander- alkaline phosphatase expression and osteoblast mineralization by a Wnt
en). The work is part of Prometheus, the Leuven Research and autocrine loop. J Bone Miner Res 2003;18:1842–53.
Development Division of Skeletal Tissue Engineering of KU Leuven: [28] Suzuki A, Ghayor C, Guicheux J, Magne D, Quillard S, Kakita A, et al. Enhanced
expression of the inorganic phosphate transporter Pit-1 is involved in BMP-2-
http://www.kuleuven.be/Prometheus. The authors declare that induced matrix mineralization in osteoblast-like cells. J Bone Miner Res
they have no potential conflict of interest, and they had and will 2006;21:674–83.
have no financial relationships with companies whose products [29] Hughes-Fulford M, Li CF. The role of FGF-2 and BMP-2 in regulation of gene
induction, cell proliferation and mineralization. J Orthop Surg Res 2011;6:8.
are relevant to the subject of this study. [30] Clase CM, Norman GL, Beecroft ML, Churchill DN. Albumin-corrected calcium
and ionized calcium in stable haemodialysis patients. Nephrol Dial
Transplant 2000;15:1841–6.
A. Figures with essential colour discrimination [31] Payne RB, Carver ME, Morgan DB. Interpretation of serum total calcium:
effects of adjustment for albumin concentration on frequency of abnormal
Certain figures in this article, particularly Figs. 1 and 2, are dif- values and on detection of change in the individual. J Clin Pathol
1979;32:56–60.
ficult to interpret in black and white. The full color images can be
[32] Orwar O, Lobovkina T, Gozen I, Erkan Y, Olofsson J, Weber SG. Protrusive
found in the on-line version, at http://dx.doi.org/10.1016/ growth and periodic contractile motion in surface-adhered vesicles induced
j.actbio.2012.07.002. by Ca(2+)-gradients. Soft Matter 2010;6:268–72.
[33] Breitwieser GE. Extracellular calcium as an integrator of tissue function. Int J
Biochem Cell Biol 2008;40:1467–80.
References [34] Dvorak MM, Riccardi D. Ca2+ as an extracellular signal in bone. Cell Calcium
2004;35:249–55.
[1] Murugan R, Ramakrishna S. Development of nanocomposites for bone [35] Olszak IT, Poznansky MC, Evans RH, Olson D, Kos C, Pollak MR, et al.
grafting. Compos Sci Technol 2005;65:2385–406. Extracellular calcium elicits a chemokinetic response from monocytes in vitro
[2] Fratzl P. Bone fracture: when the cracks begin to show. Nat Mater and in vivo. J Clin Invest 2000;105:1299–305.
2008;7:610–2. [36] Godwin SL, Soltoff SP. Extracellular calcium and platelet-derived growth
[3] Rho JY, Kuhn-Spearing L, Zioupos P. Mechanical properties and the factor promote receptor-mediated chemotaxis in osteoblasts through
hierarchical structure of bone. Med Eng Phys 1998;20:92–102. different signaling pathways. J Biol Chem 1997;272:11307–12.
[4] Athanasiou KA, Zhu C, Lanctot DR, Agrawal CM, Wang X. Fundamentals of [37] Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, Poznansky
biomechanics in tissue engineering of bone. Tissue eng 2000;6:361–81. MC, et al. Stem cell engraftment at the endosteal niche is specified by the
[5] Goldstein SA. The mechanical properties of trabecular bone: dependence on calcium-sensing receptor. Nature 2006;439:599–603.
anatomic location and function. J Biomech 1987;20:1055–61. [38] Aguirre A, Gonzalez A, Planell JA, Engel E. Extracellular calcium modulates
[6] Keaveny TM, Morgan EF, Niebur GL, Yeh OC. Biomechanics of trabecular bone. in vitro bone marrow-derived Flk-1+ CD34+ progenitor cell chemotaxis and
Annu Rev Biomed Eng 2001;3:307–33. differentiation through a calcium-sensing receptor. Biochem Biophys Res
[7] Minns RJ, Atkinson A, Steven FS. The role of calcium in the mechanical Commun 2010;393:156–61.
behaviour of bone. Phys Med Biol 1983;28:1057–66. [39] Duncan RL, Akanbi KA, Farach-Carson MC. Calcium signals and calcium
[8] Currey JD. Bones: structure and mechanics. Princeton, NJ: Princeton channels in osteoblastic cells. Semin Nephrol 1998;18:178–90.
University Press; 2006. [40] Huiskes R, Ruimerman R, van Lenthe GH, Janssen JD. Effects of mechanical
[9] Fantner GE, Hassenkam T, Kindt JH, Weaver JC, Birkedal H, Pechenik L, et al. forces on maintenance and adaptation of form in trabecular bone. Nature
Sacrificial bonds and hidden length dissipate energy as mineralized fibrils 2000;405:704–6.
separate during bone fracture. Nat Mater 2005;4:612–6. [41] Zayzafoon M. Calcium/calmodulin signaling controls osteoblast growth and
[10] Fantner GE, Adams J, Turner P, Thurner PJ, Fisher LW, Hansma PK. Nanoscale differentiation. J Cell Biochem 2006;97:56–70.
ion mediated networks in bone: osteopontin can repeatedly dissipate large [42] Dvorak MM, Siddiqua A, Ward DT, Carter DH, Dallas SL, Nemeth EF, et al.
amounts of energy. Nano Lett 2007;7:2491–8. Physiological changes in extracellular calcium concentration directly control
[11] Nakahara H, Dennis JE, Bruder SP, Haynesworth SE, Lennon DP, Caplan AI. In osteoblast function in the absence of calciotropic hormones. Proc Natl Acad
vitro differentiation of bone and hypertrophic cartilage from periosteal- Sci USA 2004;101:5140–5.
derived cells. Exp Cell Res 1991;195:492–503. [43] Ozkucur N, Perike S, Sharma P, Funk RH. Persistent directional cell migration
[12] Colfen H. Biomineralization: a crystal-clear view. Nat Mater 2010;9:960–1. requires ion transport proteins as direction sensors and membrane potential
[13] Stewart AJ, Roberts SJ, Seawright E, Davey MG, Fleming RH, Farquharson C. differences in order to maintain directedness. BMC Cell Biol 2011;12:4.
The presence of PHOSPHO1 in matrix vesicles and its developmental [44] Ozkucur N, Monsees TK, Perike S, Do HQ, Funk RH. Local calcium elevation
expression prior to skeletal mineralization. Bone 2006;39:1000–7. and cell elongation initiate guided motility in electrically stimulated
[14] Anderson HC, Garimella R, Tague SE. The role of matrix vesicles in growth osteoblast-like cells. PLoS One 2009;4:e6131.
plate development and biomineralization. Front Biosci 2005;10:822–37. [45] Yamauchi M, Yamaguchi T, Kaji H, Sugimoto T, Chihara K. Involvement of
[15] Golub EE. Role of matrix vesicles in biomineralization. Biochim Biophys Acta calcium-sensing receptor in osteoblastic differentiation of mouse MC3T3-E1
2009;1790:1592–8. cells. Am J Physiol Endocrinol Metab 2005;288:E608–16.
[16] Siffert RS. The role of alkaline phosphatase in osteogenesis. J Exp Med [46] Barradas AM, Fernandes HA, Groen N, Chai YC, Schrooten J, van de Peppel J,
1951;93:415–26. et al. A calcium-induced signaling cascade leading to osteogenic
[17] Golub EE, Harrison G, Taylor AG, Camper S, Shapiro IM. The role of alkaline differentiation of human bone marrow-derived mesenchymal stromal cells.
phosphatase in cartilage mineralization. Bone Miner 1992;17:273–8. Biomaterials 2012;33:3205–15.
[18] Orimo H. The mechanism of mineralization and the role of alkaline [47] Valerio P, Pereira MM, Goes AM, Leite MF. BG60S dissolution interferes with
phosphatase in health and disease. J Nihon Med Sch 2010;77:4–12. osteoblast calcium signals. J Mater Sci Mater Med 2007;18:265–71.
[19] Houston B, Stewart AJ, Farquharson C. PHOSPHO1—A novel phosphatase [48] Dvorak MM, Chen TH, Orwoll B, Garvey C, Chang W, Bikle DD, et al.
specifically expressed at sites of mineralisation in bone and cartilage. Bone Constitutive activity of the osteoblast Ca2+-sensing receptor promotes loss of
2004;34:629–37. cancellous bone. Endocrinology 2007;148:3156–63.
[20] Roberts S, Narisawa S, Harmey D, Millan JL, Farquharson C. Functional [49] Tsai JA, Bucht E, Torring O, Kindmark H. Extracellular calcium increases free
involvement of PHOSPHO1 in matrix vesicle-mediated skeletal cytoplasmic calcium and DNA synthesis in human osteoblasts. Horm Metab
mineralization. J Bone Miner Res 2007;22:617–27. Res 2004;36:22–6.
[21] Meyer U, Meyer T, Vosshans J, Joos U. Decreased expression of osteocalcin and [50] Titorencu I, Jinga VV, Constantinescu E, Gafencu AV, Ciohodaru C, Manolescu
osteonectin in relation to high strains and decreased mineralization in I, et al. Proliferation, differentiation and characterization of osteoblasts from
mandibular distraction osteogenesis. J Craniomaxillofac Surg 1999;27:222–7. human BM mesenchymal cells. Cytotherapy 2007;9:682–96.
Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887 3885

[51] Maeno S, Niki Y, Matsumoto H, Morioka H, Yatabe T, Funayama A, et al. The [82] Yuan H, Yang Z, Li Y, Zhang X, De Bruijn JD, De Groot K. Osteoinduction by
effect of calcium ion concentration on osteoblast viability, proliferation and calcium phosphate biomaterials. J Mater Sci Mater Med 1998;9:723–6.
differentiation in monolayer and 3D culture. Biomaterials 2005;26:4847–55. [83] Alvis M, Lalor P, Brown MK, Thorn MR, Block JE, Hornby S, et al.
[52] Nakade O, Takahashi K, Takuma T, Aoki T, Kaku T. Effect of extracellular Osteoinduction by a collagen mineral composite combined with isologous
calcium on the gene expression of bone morphogenetic protein-2 and -4 of bone marrow in a subcutaneous rat model. Orthopedics 2003;26:77–80.
normal human bone cells. J Bone Miner Metab 2001;19:13–9. [84] Habibovic P, van der Valk CM, van Blitterswijk CA, De Groot K, Meijer G.
[53] Pekkinen M, Ahlstrom M, Riehle U, Lamberg-Allardt C. Effects of extracellular Influence of octacalcium phosphate coating on osteoinductive properties of
calcium and phosphate on expression and realese of parathyroid hormone- biomaterials. J Mater Sci Mater Med 2004;15:373–80.
related protein in osteoblast progenitor cells. Bone 2007;40:S270. [85] Ripamonti U, Crooks J, Khoali L, Roden L. The induction of bone formation by
[54] Lee YK, Song J, Lee SB, Kim KM, Choi SH, Kim CK, et al. Proliferation, coral-derived calcium carbonate/hydroxyapatite constructs. Biomaterials
differentiation, and calcification of preosteoblast-like MC3T3-E1 cells 2009;30:1428–39.
cultured onto noncrystalline calcium phosphate glass. J Biomed Mater Res [86] Yuan HP, de Bruijn JD, Zhang XD, van Blitterswijk CA, de Groot K. Bone
A 2004;69:188–95. induction by porous glass ceramic made from Bioglass (R) (45S5). J Biomed
[55] Nayab SN, Jones FH, Olsen I. Modulation of the human bone cell cycle by Mater Res 2001;58:270–6.
calcium ion-implantation of titanium. Biomaterials 2007;28:38–44. [87] Winter GD, Simpson BJ. Heterotopic bone formed in a synthetic sponge in the
[56] Jalota S, Bhaduri SB, Tas AC. Osteoblast proliferation on neat and apatite-like skin of young pigs. Nature 1969;223:88–90.
calcium phosphate-coated titanium foam scaffolds. Mat Sci Eng C-Bio S [88] Fujibayashi S, Neo M, Kim HM, Kokubo T, Nakamura T. Osteoinduction of
2007;27:432–40. porous bioactive titanium metal. Biomaterials 2004;25:443–50.
[57] Liu Y, Cooper PR, Barralet JE, Shelton RM. Influence of calcium phosphate [89] Barradas AM, Yuan H, van Blitterswijk CA, Habibovic P. Osteoinductive
crystal assemblies on the proliferation and osteogenic gene expression of rat biomaterials: current knowledge of properties, experimental models and
bone marrow stromal cells. Biomaterials 2007;28:1393–403. biological mechanisms. Eur Cell Mater 2011;21:407–29 [discussion 29].
[58] Park JW, Park KB, Suh JY. Effects of calcium ion incorporation on bone healing [90] Fan H, Ikoma T, Tanaka J, Zhang X. Surface structural biomimetics and the
of Ti6Al4V alloy implants in rabbit tibiae. Biomaterials 2007;28:3306–13. osteoinduction of calcium phosphate biomaterials. J Nanosci Nanotechnol
[59] Piters E, Boudin E, Van Hul W. Wnt signaling: a win for bone. Arch Biochem 2007;7:808–13.
Biophys 2008;473:112–6. [91] Habibovic P, Yuan HP, van der Valk CM, Meijer G, van Blitterswijk CA, de
[60] Conrads KA, Yi M, Simpson KA, Lucas DA, Camalier CE, Yu LR, et al. A Groot K. 3D microenvironment as essential element for osteoinduction by
combined proteome and microarray investigation of inorganic phosphate- biomaterials. Biomaterials 2005;26:3565–75.
induced pre-osteoblast cells. Mol Cell Proteomics 2005;4:1284–96. [92] Akiyama N, Takemoto M, Fujibayashi S, Neo M, Hirano M, Nakamura T.
[61] Beck Jr GR. Inorganic phosphate as a signaling molecule in osteoblast Difference between dogs and rats with regard to osteoclast-like cells in
differentiation. J Cell Biochem 2003;90:234–43. calcium-deficient hydroxyapatite-induced osteoinduction. J Biomed Mater
[62] Beck Jr GR, Knecht N. Osteopontin regulation by inorganic phosphate is ERK1/ Res Part A 2011;96A:402–12.
2-, protein kinase C-, and proteasome-dependent. J Biol Chem [93] Habibovic P, Sees TM, van den Doel MA, van Blitterswijk CA, de Groot K.
2003;278:41921–9. Osteoinduction by biomaterials – physicochemical and structural influences.
[63] Julien M, Khoshniat S, Lacreusette A, Gatius M, Bozec A, Wagner EF, et al. J Biomed Mater Res Part A 2006;77A:747–62.
Phosphate-dependent regulation of MGP in osteoblasts: role of ERK1/2 and [94] Li X, Liu H, Niu X, Fan Y, Feng Q, Cui FZ, et al. Osteogenic differentiation of
Fra-1. J Bone Miner Res 2009;24:1856–68. human adipose-derived stem cells induced by osteoinductive calcium
[64] Liu YK, Lu QZ, Pei R, Ji HJ, Zhou GS, Zhao XL, et al. The effect of extracellular phosphate ceramics. J Biomed Mater Res B Appl Biomater 2011;97:10–9.
calcium and inorganic phosphate on the growth and osteogenic [95] Lu Z, Zreiqat H. The osteoconductivity of biomaterials is regulated by bone
differentiation of mesenchymal stem cells in vitro: implication for bone morphogenetic protein 2 autocrine loop involving alpha2beta1 integrin and
tissue engineering. Biomed Mater 2009;4:025004. mitogen-activated protein kinase/extracellular related kinase signaling
[65] Murshed M, Harmey D, Millan JL, McKee MD, Karsenty G. Unique pathways. Tissue Eng Part A 2010;16:3075–84.
coexpression in osteoblasts of broadly expressed genes accounts for the [96] Urist MR, Huo YK, Brownell AG, Hohl WM, Buyske J, Lietze A, et al.
spatial restriction of ECM mineralization to bone. Genes Dev Purification of bovine bone morphogenetic protein by hydroxyapatite
2005;19:1093–104. chromatography. Proc Natl Acad Sci USA 1984;81:371–5.
[66] Habibovic P, Bassett DC, Doillon CJ, Gerard C, McKee MD, Barralet JE. Collagen [97] Cheng L, Ye F, Yang R, Lu X, Shi Y, Li L, et al. Osteoinduction of hydroxyapatite/
biomineralization in vivo by sustained release of inorganic phosphate ions. beta-tricalcium phosphate bioceramics in mice with a fractured fibula. Acta
Adv Mater 2010;22:1858–62. Biomater 2010;6:1569–74.
[67] Barrere F, van Blitterswijk CA, de Groot K. Bone regeneration: molecular and [98] Ripamonti U, Roden LC. Induction of bone formation by transforming growth
cellular interactions with calcium phosphate ceramics. Int J Nanomedicine factor-beta2 in the non-human primate Papio ursinus and its modulation by
2006;1:317–32. skeletal muscle responding stem cells. Cell Prolif 2010;43:207–18.
[68] Chai YC, Roberts SJ, Schrooten J, Luyten FP. Probing the osteoinductive effect [99] Rueger JM, Siebert HR, Dohr-Fritz M, Schmidt H, Pannike A. Time sequence of
of calcium phosphate by using an in vitro biomimetic model. Tissue Eng Part osteoinduction and osteostimulation elicited by biologic bone replacement
A 2011;17:1083–97. materials. Life Support Syst 1985;3(Suppl 1):471–5.
[69] Chai YC, Roberts SJ, Van Bael S, Chen Y, Luyten FP, Schrooten J. Multi-level [100] Mulliken JB, Kaban LB, Glowacki J. Induced osteogenesis—the biological
factorial analysis of Ca2+/Pi supplementation as bio-instructive media for principle and clinical applications. J Surg Res 1984;37:487–96.
in vitro biomimetic engineering of three-dimensional osteogenic hybrids. [101] Low KL, Tan SH, Zein SH, Roether JA, Mourino V, Boccaccini AR. Calcium
Tissue Eng Part C Methods 2012;18:90–103. phosphate-based composites as injectable bone substitute materials. J
[70] Bhandari M, Jain AK. Bone stimulators: beyond the black box. Indian J Orthop Biomed Mater Res B Appl Biomater 2010;94:273–86.
2009;43:109–10. [102] Coffin JD, Florkiewicz RZ, Neumann J, Mort-Hopkins T, Dorn II GW, Lightfoot
[71] Eyckmans J, Roberts SJ, Schrooten J, Luyten FP. A clinically relevant model of P, et al. Abnormal bone growth and selective translational regulation in basic
osteoinduction: a process requiring calcium phosphate and BMP/Wnt fibroblast growth factor (FGF-2) transgenic mice. Mol Biol Cell
signalling. J Cell Mol Med 2010;14:1845–56. 1995;6:1861–73.
[72] Habibovic P, de Groot K. Osteoinductive biomaterials—properties and [103] LeGeros RZ. Calcium phosphate biomaterials: an update. Int J Oral-Med Sci
relevance in bone repair. J Tissue Eng Regen Med 2007;1:25–32. 2006;4:117–23.
[73] Pazzaglia UE, Zatti G, Ragni P, Ceciliani L. The role of mineralization in [104] Bose S, Tarafder S. Calcium phosphate ceramic systems in growth factor and
experimental models of osteogenetic induction with decalcified bone matrix. drug delivery for bone tissue engineering: a review. Acta Biomater
Ital J Orthop Traumatol 1988;14:369–75. 2012;8:1401–21.
[74] Jukes JM, Both SK, Leusink A, Sterk LM, van Blitterswijk CA, de Boer J. [105] Ginebra MP, Traykova T, Planell JA. Calcium phosphate cements as bone drug
Endochondral bone tissue engineering using embryonic stem cells. Proc Natl delivery systems: a review. J Control Release 2006;113:102–10.
Acad Sci USA 2008;105:6840–5. [106] LeGeros RZ. Calcium phosphate-based osteoinductive materials. Chem Rev
[75] Yuan H, Fernandes H, Habibovic P, de Boer J, Barradas AM, de Ruiter A, et al. 2008;108:4742–53.
Osteoinductive ceramics as a synthetic alternative to autologous bone [107] Chow LC. Next generation calcium phosphate-based biomaterials. Dent
grafting. Proc Natl Acad Sci USA 2010;107:13614–9. Mater J 2009;28:1–10.
[76] Yuan H, van Blitterswijk CA, de Groot K, de Bruijn JD. Cross-species [108] Yoshikawa M, Tsuji N, Shimomura Y, Hayashi H, Ohgushi H. Osteogenesis
comparison of ectopic bone formation in biphasic calcium phosphate (BCP) depending on geometry of porous hydroxyapatite scaffolds. Calcif Tissue Int
and hydroxyapatite (HA) scaffolds. Tissue Eng 2006;12:1607–15. 2008;83:139–45.
[77] Urist MR. Bone: formation by autoinduction. Science 1965;150:893–9. [109] Kuboki Y, Jin Q, Takita H. Geometry of carriers controlling phenotypic
[78] Urist MR, Strates BS. Bone morphogenetic protein. J Dent Res expression in BMP-induced osteogenesis and chondrogenesis. J Bone Joint
1971;50:1392–406. Surg Am 2001;83-A(Suppl 1):S105–15.
[79] Jones W, Roberts RE. Pathological calcification and ossification in relation to [110] Mankani MH, Afghani S, Franco J, Launey M, Marshall S, Marshall GW, et al.
Leriche and Policard’s theory. Proc R Soc Med 1933;26:853–9. Lamellar spacing in cuboid hydroxyapatite scaffolds regulates bone
[80] Vattikuti R, Towler DA. Osteogenic regulation of vascular calcification: an formation by human bone marrow stromal cells. Tissue Eng Part A
early perspective. Am J Physiol Endocrinol Metab 2004;286:E686–96. 2011;17:1615–23.
[81] Zebboudj AF, Shin V, Bostrom K. Matrix GLA protein and BMP-2 regulate [111] Wilson CE, de Bruijn JD, van Blitterswijk CA, Verbout AJ, Dhert WJ.
osteoinduction in calcifying vascular cells. J Cell Biochem 2003;90:756–65. Design and fabrication of standardized hydroxyapatite scaffolds with a
3886 Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887

defined macro-architecture by rapid prototyping for bone-tissue- [142] Fujita N, Matsushita T, Ishida K, Sasaki K, Kubo S, Matsumoto T, et al. An
engineering research. J Biomed Mater Res A 2004;68:123–32. analysis of bone regeneration at a segmental bone defect by controlled
[112] Duan Y, Yao Z, Wang C, Chen J, Zhang X. A study of bone-like apatite release of bone morphogenetic protein 2 from a biodegradable sponge
formation on porous calcium phosphate ceramics in dynamic SBF. Sheng Wu composed of gelatin and beta-tricalcium phosphate. J Tissue Eng Regen Med
Yi Xue Gong Cheng Xue Za Zhi 2002;19:365–9. 2012;6:291–8.
[113] Li X, van Blitterswijk CA, Feng Q, Cui F, Watari F. The effect of calcium [143] Kang Y, Kim S, Khademhosseini A, Yang Y. Creation of bony
phosphate microstructure on bone-related cells in vitro. Biomaterials microenvironment with CaP and cell-derived ECM to enhance human
2008;29:3306–16. bone-marrow MSC behavior and delivery of BMP-2. Biomaterials
[114] Fischer EM, Layrolle P, Van Blitterswijk CA, De Bruijn JD. Bone formation by 2011;32:6119–30.
mesenchymal progenitor cells cultured on dense and microporous [144] Gilbert M, Shaw WJ, Long JR, Nelson K, Drobny GP, Giachelli CM, et al.
hydroxyapatite particles. Tissue Eng 2003;9:1179–88. Chimeric peptides of statherin and osteopontin that bind hydroxyapatite and
[115] Balaguer T, Boukhechba F, Clave A, Bouvet-Gerbettaz S, Trojani C, Michiels JF, mediate cell adhesion. J Biol Chem 2000;275:16213–8.
et al. Biphasic calcium phosphate microparticles for bone formation: benefits [145] Raghunathan V, Gibson JM, Goobes G, Popham JM, Louie EA, Stayton PS, et al.
of combination with blood clot. Tissue Eng Part A 2010;16:3495–505. Homonuclear and heteronuclear NMR studies of a statherin fragment bound
[116] Yuan H, van Blitterswijk CA, de Groot K, de Bruijn JD. A comparison of bone to hydroxyapatite crystals. J Phys Chem B 2006;110:9324–32.
formation in biphasic calcium phosphate (BCP) and hydroxyapatite (HA) [146] Zimmerman LB, De Jesus-Escobar JM, Harland RM. The Spemann organizer
implanted in muscle and bone of dogs at different time periods. J Biomed signal noggin binds and inactivates bone morphogenetic protein 4. Cell
Mater Res A 2006;78:139–47. 1996;86:599–606.
[117] Daculsi G, LeGeros RZ. Biphasic calcium phosphate (BCP) bioceramics: chemical, [147] Brunet LJ, McMahon JA, McMahon AP, Harland RM. Noggin, cartilage
physical and biological properties. Enc Biomat, Biomed Eng 2006:1–9. morphogenesis, and joint formation in the mammalian skeleton. Science
[118] Duan YR, Zhang ZR, Wang CY, Chen JY, Zhang XD. Dynamic study of calcium 1998;280:1455–7.
phosphate formation on porous HA/TCP ceramics. J Mater Sci Mater Med [148] Vehof JW, Takita H, Kuboki Y, Spauwen PH, Jansen JA. Histological
2005;16:795–801. characterization of the early stages of bone morphogenetic protein-induced
[119] Daculsi G, Laboux O, Malard O, Weiss P. Current state of the art of biphasic osteogenesis. J Biomed Mater Res 2002;61:440–9.
calcium phosphate bioceramics. J Mater Sci Mater Med 2003;14:195–200. [149] Kuboki Y, Takita H, Kobayashi D, Tsuruga E, Inoue M, Murata M, et al. BMP-
[120] Arinzeh TL, Tran T, McAlary J, Daculsi G. A comparative study of biphasic induced osteogenesis on the surface of hydroxyapatite with geometrically
calcium phosphate ceramics for human mesenchymal stem-cell-induced feasible and nonfeasible structures: topology of osteogenesis. J Biomed Mater
bone formation. Biomaterials 2005;26:3631–8. Res 1998;39:190–9.
[121] Ye F, Lu X, Lu B, Wang J, Shi Y, Zhang L, et al. A long-term evaluation of [150] Kuboki Y, Saito T, Murata M, Takita H, Mizuno M, Inoue M, et al. Two
osteoinductive HA/beta-TCP ceramics in vivo: 4.5 years study in pigs. J Mater distinctive BMP-carriers induce zonal chondrogenesis and membranous
Sci Mater Med 2007;18:2173–8. ossification, respectively; geometrical factors of matrices for cell-
[122] Lobo SE, Arinzeh TL. Biphasic calcium phosphate ceramics for bone differentiation. Connect Tissue Res 1995;32:219–26.
regeneration and tissue engineering applications. Materials 2010;3:815–26. [151] van der Meulen MC, Huiskes R. Why mechanobiology? A survey article. J
[123] Garrido CA, Lobo SE, Turibio FM, Legeros RZ. Biphasic calcium phosphate Biomech 2002;35:401–14.
bioceramics for orthopaedic reconstructions: clinical outcomes. Int J [152] Geris L, Reed AA, Vander Sloten J, Simpson AH, Van Oosterwyck H.
Biomater 2011;2011:129727. Occurrence and treatment of bone atrophic non-unions investigated by an
[124] El-Ghannam A. Bone reconstruction: from bioceramics to tissue engineering. integrative approach. PLoS Comput Biol 2010;6:e1000915.
Expert Rev Med Dev 2005;2:87–101. [153] Faratian D, Goltsov A, Lebedeva G, Sorokin A, Moodie S, Mullen P, et al.
[125] Kim KH, Ramaswamy N. Electrochemical surface modification of titanium in Systems biology reveals new strategies for personalizing cancer medicine
dentistry. Dent Mater J 2009;28:20–36. and confirms the role of PTEN in resistance to trastuzumab. Cancer Res
[126] Bosetti M, Lloyd AW, Santin M, Denyer SP, Cannas M. Effects of 2009;69:6713–20.
phosphatidylserine coatings on titanium on inflammatory cells and cell- [154] Geris L, Vander Sloten J, Van Oosterwyck H. In silico biology of bone
induced mineralisation in vitro. Biomaterials 2005;26:7572–8. modelling and remodelling: regeneration. Philos Transact A Math Phys Eng
[127] Chai YC, Truscello S, Bael SV, Luyten FP, Vleugels J, Schrooten J. Perfusion Sci 2009;367:2031–53.
electrodeposition of calcium phosphate on additive manufactured titanium [155] Bohner M, Baumgart F. Theoretical model to determine the effects of
scaffolds for bone engineering. Acta Biomater 2011;7:2310–9. geometrical factors on the resorption of calcium phosphate bone
[128] Chai YC, Kerckhofs G, Roberts SJ, Van Bael S, Schepers E, Vleugels J, et al. substitutes. Biomaterials 2004;25:3569–82.
Ectopic bone formation by 3D porous calcium phosphate–Ti6Al4V hybrids [156] Impens S, Chen Y, Mullens S, Luyten F, Schrooten J. Controlled cell-seeding
produced by perfusion electrodeposition. Biomaterials 2012;33:4044–58. methodologies: a first step toward clinically relevant bone tissue engineering
[129] Mourino V, Cattalini JP, Boccaccini AR. Metallic ions as therapeutic agents in strategies. Tissue Eng Part C Methods 2010;16:1575–83.
tissue engineering scaffolds: an overview of their biological applications and [157] Byrne DP, Lacroix D, Planell JA, Kelly DJ, Prendergast PJ. Simulation of tissue
strategies for new developments. J R Soc Interface 2012;9:401–19. differentiation in a scaffold as a function of porosity, Young’s modulus and
[130] Guldberg RE. Spatiotemporal delivery strategies for promoting dissolution rate: application of mechanobiological models in tissue
musculoskeletal tissue regeneration. J Bone Miner Res 2009;24:1507–11. engineering. Biomaterials 2007;28:5544–54.
[131] Enomoto-Iwamoto M, Iwamoto M, Mukudai Y, Kawakami Y, Nohno T, [158] Gentleman E, Swain RJ, Evans ND, Boonrungsiman S, Jell G, Ball MD, et al.
Higuchi Y, et al. Bone morphogenetic protein signaling is required for Comparative materials differences revealed in engineered bone as a function
maintenance of differentiated phenotype, control of proliferation, and of cell-specific differentiation. Nat Mater 2009;8:763–70.
hypertrophy in chondrocytes. J Cell Biol 1998;140:409–18. [159] Checa S, Prendergast PJ. Effect of cell seeding and mechanical loading on
[132] Vukicevic S, Grgurevic L. BMP-6 and mesenchymal stem cell differentiation. vascularization and tissue formation inside a scaffold: a mechano-biological
Cytokine Growth Factor Rev 2009;20:441–8. model using a lattice approach to simulate cell activity. J Biomech
[133] Tan TW, Huang YL, Chang JT, Lin JJ, Fong YC, Kuo CC, et al. CCN3 increases 2010;43:961–8.
BMP-4 expression and bone mineralization in osteoblasts. J Cell Physiol [160] Stops AJ, Heraty KB, Browne M, O’Brien FJ, McHugh PE. A prediction of cell
2012;227:2531–41. differentiation and proliferation within a collagen-glycosaminoglycan
[134] Franceschi RT. The developmental control of osteoblast-specific gene scaffold subjected to mechanical strain and perfusive fluid flow. J Biomech
expression: role of specific transcription factors and the extracellular 2010;43:618–26.
matrix environment. Crit Rev Oral Biol Med 1999;10:40–57. [161] Sanz-Herrera JA, Garcia-Aznar JM, Doblare M. A mathematical model for bone
[135] Sampath TK, Reddi AH. Dissociative extraction and reconstitution of tissue regeneration inside a specific type of scaffold. Biomech Model
extracellular matrix components involved in local bone differentiation. Proc Mechanobiol 2008;7:355–66.
Natl Acad Sci USA 1981;78:7599–603. [162] Lacroix D, Planell JA, Prendergast PJ. Computer-aided design and finite-
[136] Seeherman H, Wozney J, Li R. Bone morphogenetic protein delivery systems. element modelling of biomaterial scaffolds for bone tissue engineering.
Spine (Phila Pa 1976) 2002;27:S16–23. Philos Transact A Math Phys Eng Sci 2009;367:1993–2009.
[137] Sampath TK, Reddi AH. Homology of bone-inductive proteins from human, [163] Carlier A, Chai YC, Moesen M, Theys T, Schrooten J, Van Oosterwyck H, et al.
monkey, bovine, and rat extracellular matrix. Proc Natl Acad Sci USA Designing optimal calcium phosphate scaffold–cell combinations using an
1983;80:6591–5. integrative model-based approach. Acta Biomater 2011;7:3573–85.
[138] Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue [164] Zhang Q, Chen J, Feng J, Cao Y, Deng C, Zhang X. Dissolution and
engineering: the road from laboratory to clinic, part II (BMP delivery). J mineralization behaviors of HA coatings. Biomaterials 2003;24:4741–8.
Tissue Eng Regen Med 2008;2:81–96. [165] Bohner M, Loosli Y, Baroud G, Lacroix D. Commentary: deciphering the link
[139] Wozney JM, Rosen V, Celeste AJ, Mitsock LM, Whitters MJ, Kriz RW, et al. between architecture and biological response of a bone graft substitute. Acta
Novel regulators of bone formation: molecular clones and activities. Science Biomater 2011;7:478–84.
1988;242:1528–34. [166] Chai YC, Roberts SJ, Desmet E, Kerckhofs G, van Gastel N, Geris L, et al.
[140] Carragee EJ, Ghanayem AJ, Weiner BK, Rothman DJ, Bono CM. A challenge to Mechanisms of ectopic bone formation by human osteoprogenitor cells on
integrity in spine publications: years of living dangerously with the CaP biomaterial carriers. Biomaterials 2012;33:3127–42.
promotion of bone growth factors. Spine J 2011;11:463–8. [167] Gustavsson J, Ginebra MP, Engel E, Planell J. Ion reactivity of calcium-
[141] Langer R, Folkman J. Polymers for the sustained release of proteins and other deficient hydroxyapatite in standard cell culture media. Acta Biomater
macromolecules. Nature 1976;263:797–800. 2011;7:4242–52.
Y.C. Chai et al. / Acta Biomaterialia 8 (2012) 3876–3887 3887

[168] Kerckhofs G, Schrooten J, Van Cleynenbreugel T, Lomov SV, Wevers M. [170] Lee JT, Leng Y, Chow KL, Ren F, Ge X, Wang K, et al. Cell culture medium as an
Validation of X-ray microfocus computed tomography as an imaging tool for alternative to conventional simulated body fluid. Acta Biomater
porous structures. Rev Sci Instrum 2008;79:013711. 2011;7:2615–22.
[169] Bohner M, Lemaitre J. Can bioactivity be tested in vitro with SBF solution?
Biomaterials 2009;30:2175–9.

You might also like