Вы находитесь на странице: 1из 18

Biomaterials 32 (2011) 2757e2774

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Review

A review of the biological response to ionic dissolution products from bioactive


glasses and glass-ceramics
Alexander Hoppe, Nusret S. Gldal, Aldo R. Boccaccini*
Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany

a r t i c l e i n f o

a b s t r a c t

Article history:
Received 14 December 2010
Accepted 4 January 2011
Available online 2 February 2011

Several inorganic materials such as special compositions of silicate glasses, glass-ceramics and calcium
phosphates have been shown to be bioactive and resorbable and to exhibit appropriate mechanical
properties which make them suitable for bone tissue engineering applications. However, the exact
mechanism of interaction between the ionic dissolution products of such inorganic materials and human
cells are not fully understood, which has prompted considerable research work in the biomaterials
community during the last decade. This review comprehensively covers literature reports which have
investigated specically the effect of dissolution products of silicate bioactive glasses and glass-ceramics
in relation to osteogenesis and angiogenesis. Particularly, recent advances made in fabricating dense
biomaterials and scaffolds doped with trace elements (e.g. Zn, Sr, Mg, and Cu) and investigations on the
effect of these elements on the scaffold biological performance are summarized and discussed in detail.
Clearly, the biological response to articial materials depends on many parameters such as chemical
composition, topography, porosity and grain size. This review, however, focuses only on the ion release
kinetics of the materials and the specic effect of the released ionic dissolution products on human cell
behaviour, providing also a scope for future investigations and identifying specic research needs to
advance the eld. The biological performance of pure and doped silicate glasses, phosphate based glasses
with novel specic compositions as well as several other silicate based compounds are discussed in
detail. Cells investigated in the reviewed articles include human osteoblastic and osteoclastic cells as well
as endothelial cells and stem cells.
2011 Elsevier Ltd. All rights reserved.

Keywords:
Bone tissue engineering
Bioactive glasses
Bioactivity
Metal ion release
Cell proliferation
Osteoblasts

1. Introduction
The clinical demand on engineered bone tissue has been
growing in recent years in direct relation to the increasing of the
human population [1]. Tissue engineering (TE) is one of the
approaches being investigated to tackle this problem [2,3]. In
common TE strategies a three-dimensional structure, termed
scaffold, fabricated from a suitable articial or natural material
and exhibiting high porosity and pore interconnectivity is used
[4e7]. Ideally, these scaffolds should not only provide a passive
structural support for bone cells, but they also should favourably
affect bone formation by stimulating osteoblastic cell proliferation
and differentiation [8]. Several attempts have been successfully
made to construct porous scaffolds with desired porosity and
appropriate mechanical performance from inorganic materials
such as bioactive ceramics and glasses, from biodegradable

* Corresponding author. Tel.: 49 44 207 594 6731.


E-mail address: aldo.boccaccini@ww.uni-erlangen.de (A.R. Boccaccini).
0142-9612/$ e see front matter 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biomaterials.2011.01.004

polymers and their composites [6,7]. Different fabrication techniques have been proposed including foam replication, sol-gel
foaming, electro-spinning or rapid prototyping methods [2,3]. Due
to their chemical similarity to the inorganic phase of bone, inorganic biomaterials such as calcium phosphates (CaP), e.g.
hydroxyapatite (HAp), a- and b-tricalciumphosphate (TCP), have
been more intensively investigated in respect to their possible
application as bone scaffolds [4,5,9,10]. These materials are bioactive, osteoconductive and are able to bond directly to bone [11].
Moreover, numerous in vitro and vivo studies have shown that HAp
[12e17] and related CaPs [18e21] support the adhesion, differentiation and proliferation of osteogenesis related cells (e.g. osteoblasts, mesenchymal stem cells). Additionally, it has been shown
that biphasic calcium phosphates (BCP), e.g. mixtures of HAp and
TCP, are osteinductive and induce gene expression in bone cells
[20,22,23]. Amongst the common CaPs, hydroxyapatite is the most
thermodynamically stable at physiological pH of 7.4 and it is known
to be bioactive but not bioresorbable [4,5]. The biodegradability of
CaPs depends on many parameters such as crystallinity, porosity,
chemical purity and surface roughness [4]. HAp will degrade faster

2758

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

with increasing porosity, with increasing content of ion substitution in the crystal lattice and increasing content of second phase,
for example when used in BCP mixed with other different CaPs [4].
Bioactive glasses such as 45S5 Bioglass [5] and other compositions based on silicate or phosphate systems represent another
important group of inorganic, bioactive biomaterials used for scaffolds for bone tissue engineering [5,6,24,25]. Bioactive glasses (BG)
exhibit unique properties for this application showing osteoinductive behaviour, ability to bond to soft tissue as well as to hard tissue
and to form a carbonated hydroxyapatite layer (HCA) when exposed
to biological uid [5,11]. This layer is responsible for the strong
bonding between bioactive glasses and human bone [5,9,11]. More
recently, it was observed that ionic dissolution products from Bioglass (e.g. Si, Ca, P) and from other silicate based glasses stimulate
expression of several genes of osteoblastic cells [26]. Furthermore,
bioactive glasses were shown to stimulate angiogenesis in vitro and
in vivo [27e29], whilst possible antibacterial [30e38] and inammatory [39] effects of bioactive glasses have also been investigated.
A schematic overview of biological responses to ionic dissolution
products of bioactive glasses is given in Fig. 1.
Increasing evidence in the literature indicates that ionic dissolution products from inorganic materials are key to understand the
behaviour of these materials in vitro and in vivo, in the context of
tissue engineering applications. Since many trace elements such as
Sr, Cu, Zn or Co present in the human body are known for their
anabolic effects in bone metabolism [40e42], new approaches for
enhancing bioactivity of scaffold materials are being investigated by
introducing therapeutic ions into the scaffold material. As indicated
above, the subsequent release of these ions after exposure to
a physiological environment is believed to favourably affect the
behaviour of human cells and to enhance the bioactivity of the
scaffolds related to both osteogenesis and angiogenesis. Thus,

research efforts are devoted to incorporate these ions in different


CaPs, bioactive silicate glasses and phosphate glasses resulting in the
changed dissolution behaviour of these materials and their modied
(usually improved) biological performance. For example, in vitro and
in vivo studies have shown enhanced bioactivity of Si-doped HAp
[43,44], Sr-doped silicate glasses [45] or Mg-doped phosphate glasses
[46], just to mention only a few examples, whereas a comprehensive
treatment of this topic is presented in the next sections. Clearly, cell
response depends not only on chemical composition, but also on
surface roughness [47] porosity [48], topography, grain size and
crystallinity of the scaffolds [2,48]. However, this review will focus
specically on the biological response to ionic products coming from
the dissolution of different glass compositions based on silicate and
phosphate systems which is relevant in angiogenesis and osteogenesis applied to bone tissue engineering.
The aim of the review is therefore to summarise the information
available in the open literature on stimulation effects of currently
used inorganic biomaterials on osteogenesis and angiogenesis, with
emphasis on the specic effects of therapeutic ionic dissolution
products. Biological performance of pure and ion-doped bioactive
silicate glasses, glass-ceramics, phosphate based glasses containing
different metal oxides and other specic calcium and silica based
compounds will be discussed. Hereby different direct and indirect
methods have been applied in order to asses the cell response to
degradable biomaterials as schematically shown in Fig. 2.
2. Role of inorganic ions in bone metabolism
Human bone is a dynamic, highly vascularised tissue with the
ability to remodel throughout the life by regulated activity of boneforming (osteoblasts) and bone-resorbing cells (osteoclasts) [49].
The processes of bone formation and bone resorption (bone

Fig. 1. Overview of biological responses to ionic dissolution products of bioactive glasses.

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

2759

Fig. 2. Different methods for analysing the cellular response to degradable biomaterials.

remodelling) are regulated by a variety of systematic and local


regulatory agents [50] including growth factors, hormones and
stress actions [51,52]. In addition, single inorganic ions such as
Calcium (Ca) [53e56], Phosphorous (P) [57], Silicon (Si) [58e60],
Strontium (Sr) [50,61e65], Zinc (Zn) [51,66] as well as Boron (B)
[67,68], Vanadium (V) [42,49,69], Cobalt (Co) [70e73] and
Magnesium (Mg) [74e80] are known to be involved in the bone
metabolism and to play a physiological role in angiogenesis, growth
and mineralization of bone tissue. Table 1 gives a summary of
biological responses to single inorganic ions relevant in the context
of this review. In particular, metal ions act as enzyme co-factors and
therefore inuence signalling pathways and stimulate metabolic
effects occurring during tissue formation [41,81]. These effects
make metal ions attractive for use as therapeutic agents in the
elds of hard and soft tissue engineering [82].
Since Ca and P are the main components of biological apatite
(Ca10(PO4,CO3)6OH2), the inorganic phase of human bone, these
ions obviously play an essential role in bone formation and
resorption. Nevertheless it is important to know the specic
extracellular matrix concentration of these ions and the mechanism of their interaction with bone cells and their stimulating effect
on bone formation. This knowledge would allow fabricating
advanced scaffolds with tailored ion release kinetics and controlled
biological response in the relevant physiological environment. For
example, Ca is known to affect osteoblastic cells in vitro. Maeno
et al. [54] found that low (2e4 mmol) and medium (6e8 mmol) Ca
concentrations are suitable for osteoblast proliferation, differentiation and extracellular matrix (ECM) mineralization, respectively,
whereas higher Ca concentrations (>10 mmol) are cytotoxic.

Moreover, extracellular Ca plays an important role in bone


remodelling by directly activating intracellular mechanisms by
affecting Ca-sensing receptors in osteoblastic cells. For example, Ca
increases the expression of Insulin like growth factors IGF-I or IGFII, which regulate human osteoblast proliferation. These ndings
have been recently reviewed in the literature [55]. Similar results
were observed by Valerio et al. [56], who found that extracellular Ca
increases the glutamate release of osteoblast cells. Since glutamate
signalling pathways are known to play an important role for bone
mechanosensitivity [53], extracellular Ca concentration must be
considered as an important regulating agent in bone metabolism.
Inorganic Phosphate (P) was recently shown to stimulate expression of matrix Gla protein (MGP), a key regulator for bone formation, in osteoblastic cells when added (10 mmol) to cell culture
medium [57].
Beside Ca and P, bone apatite is substituted by many different
trace elements occurring in smaller concentration [40]. Hence,
several ions have been considered to be promising agents in
enhancing the bone-forming ability of implant materials and
scaffolds, which can be achieved by controlling the release of
specic ions during in vivo dissolution of the scaffold. These ions
include Si, Sr, Zn, Cu, Mg and B.
Si is known to be an essential element for metabolic processes
associated with the formation and calcication of bone tissue
[58,59]. High Si contents have been detected in early stages of
bone matrix calcication [59], whereby aqueous Si was shown to
be able to induce precipitation of hydroxyapatite, the inorganic
phase of human bone [83]. Additionally, dietary Si intake was
shown to increase the bone mineral density (BMD) in men and

2760

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

Table 1
Effect of selected metallic ions on human bone metabolism and angiogenesis:
summary of literature studies.
Ion

Biological response in vivo / in vitro

Reference

Si

 essential for metabolic processes, formation


and calcication of bone tissue
 dietary intake of Si increases bone mineral
density (BMD)
 aqueous Si induces HAp precipitation
 Si(OH)4 stimulates collagen I formation and
osteoblastic differentiation
 favours osteoblast proliferation, differentiation
and extracellular matrix (ECM) mineralization
 activates Ca-sensing receptors in osteoblast cells,
increases expression of growth factors, e.g.
IGF-I or IGF-II
 stimulates expression of matrix la protein (MGP) a key
regulator in bone formation
 shows anti-inammatory effect and stimulates bone
formation in vitro by activation protein synthesis in
osteoblasts
 increases ATPase activity, regulates transcription
of osteoblastic differentiation genes, e.g. collagen I, ALP,
osteopontin and osteocalcin
 stimulates new bone formation
 increases bone cell adhesion and stability
(probably due to interactions with integrins)
 shows benecial effects on bone cells and bone
formation in vivo
 promising agent for treating osteoporosis
 signicant amounts of cellular Cu are found in
human endothelial cells when undergoing angiogenesis
 promotes synergetic stimulating effects on angiogenesis
when associated with angiogenic growth factor FGF-2
 stimulates proliferation of human endothelial cells
 induces differentiation of mesenchymal cells
towards the osteogenic lineage
 stimulates RNA synthesis in broblast cells
 dietary boron stimulates bone formation

[58,59]

Ca

P
Zn

Mg

Sr

Cu

[84]
[83]
[60]
[54]
[55,56]

[57]
[51]

[92]

[80]
[80,101]
[50,65]
[88]
[93]
[94]
[95]
[96]
[102,103]
[68]

premenopausal women [84]. Another study with Ca-decient rats


showed that Si supplementation caused positive effects on bone
mineral density by reducing bone resorption [85]. Moreover,
Nielsen et al. [86] suggested that Si has a biochemical function in
bone growth processes affecting bone collagen turn over and
sialic acid-containing ECM proteins like osteopontin. Moreover,
orthosilicate acid (Si(OH)4) at physiological concentration of
10 mmol has been shown to stimulate collagen I formation in
human osteoblast cells (HOC) and to stimulate osteoblastic
differentiation [60].
Because of the chemical analogy to Ca (same main group, similar
atomic radius, Ca2 e 1.0 A, Sr e 1.16 A), Sr can accumulate in bone
by exchanging with Ca in the hydroxyapatite crystal lattice [87].
The therapeutic potential of Sr in bone metabolism has been
investigated by Marie et al. [50,65], who showed that Sr exhibits
benecial effects on bone cells and bone formation in vivo. Sr has
also been shown to be a promising agent in treating osteoporosis
[88]. Additionally, Sr based drug such as strontium renalate
enhances bone healing indicated by increased callus resistance in
rat bones which has conrmed Sr as a promising agent in healing
bone fractures [89].
Zinc is also known to play an important role in bone metabolism
[51] and to have anti-inammatory effects [90]. Furthermore, Zn
stimulates bone formation in vitro by activating protein synthesis
in osteoblast cells and increasing ATPase activity in bone [51].
Moreover, Zn shows inhibitory effect on bone resorption inhibiting
the formation of osteoclast cells in mouse marrow cultures [51].
The regulatory effects of Zn on bone cells has suggested the
importance of Zn in gene expression [91]. More recently, zinc was

identied as regulation agent in transcription of osteoblastic


differentiation genes, such as collagen I, ALP, osteopontin and
osteocalcin [92]. It was assumed that Zn can be considered a Runx2
stimulating agent being able to directly stimulate bone formation
through increasing Runx2-targeted osteoblast differentiation gene
transcription [92].
Cu has been shown to play a signicant role in angiogenesis
[93e95]. For example, remarkable distributions of cellular Cu have
been found in human endothelial cells when they were induced to
undergo angiogenesis revealing the importance of the ion as
angiogenic agent [93]. Another recent study revealed that copper,
associated with angiogenesis growth factor FGF-2, promotes
synergetic stimulating effects on angiogenesis in vitro [94]. Moreover, Cu was shown to stimulate proliferation of human endothelial
cells [95] and to induce an increase in differentiation of mesenchymal stem cells (MSC) towards the osteogenic lineage [96]. Cu2
at a concentration of 106 mol l1 has been also shown to inhibit
osteoclast activity [97]. However, a possible positive effect of Cu on
bone metabolism is still not clearly demonstrated. Cashman et al.
[98] for instance found that copper supplements over a period of 4
weeks do not show any effect on biochemical markers of bone
formation or bone resorption [98]. Similarly, Lai and Yamaguchi [99]
have shown that supplementation with copper induced a signicant
decrease in bone tissue of rats showing no anabolic effects on bone
formation in vivo and in vitro and, additionally, it was shown that Cu
reduces anabolic effects of Zn. Other researchers have found that
dietary copper depletion causes a reduction of bone mineral density
(BMD), although no biological markers for bone formation and bone
resorption were explicitly affected by Cu [100].
Mg is essential to bone metabolism and it has been shown to
have stimulating effects on new bone formation [80]. Mg is suggested to interact with integrins of osteoblast cells which are
responsible for cell adhesion and stability [80,101]. Rude et al.
[75e77] observed that Mg depletion results in impaired bone
growth, increased bone resorption and loss in trabecular bone
underlining the signicant role that Mg plays in bone metabolism.
Boron is another element which can be considered as potential
stimulating agent for bone tissue engineering [102]. It was shown
that boron affects the RNA synthesis in broblast cells [103].
Moreover, dietary boron can stimulate bone formation, which was
observed by in vivo studies with rabbits [68]. Boron has been
shown also to increase vertebral resistance to crash force in rats as
observed after dietary intake of boron [104]. Gorustovich et al. [105]
have found that boron-decient diet affects alveolar bone modelling and remodelling in rats. Histological ndings have revealed
that B-deciency results in decreased osteogenic activity and
inactive surfaces indicating that B-deciency alters the dynamic
process of alveolar bone modelling and remodelling due to inhibition of bone formation.
In summary, all mentioned metallic ions have high potential to
be used as therapeutic agents released by resorbable scaffolds to
enhance osteogenesis and in some cases angiogenesis of scaffold
materials. Specic inorganic materials (i.e. bioactive glasses and
glass-ceramics) suitable for TE scaffolds and corresponding tissue
engineering approaches will be discussed in detail in the following
sections.
3. Silicate based glasses and glass-ceramics
3.1. Bioactive glasses based on SiO2eCaOeP2O5
Since Hench et al. [106] discovered the rst bioactive silicate
material, now called 45S5 Bioglass (wt%: 45SiO2e25CaOe25
Na2Oe6P2O5), with its unique biological properties, there has been
extensive research work on bioactive silicate glasses for biomedical

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

applications including bone tissue engineering. However, only


more recently, starting with the Xynos et al. investigation in 2001
[26], the research work has been focussing on the molecular
interactions of ionic dissolution products of bioactive glasses (BG)
and their physiological environment in order to gain greater
understanding of these mechanisms and to be able to fabricate
smart glasses with tailored properties for specic tissue engineering applications, the so-called third generation biomaterials
[24,107]. In this context ionic dissolution products of silicon based
bioactive glasses in the SiO2eCaOeNa2OeP2O5 system have been
shown to change the intracellular ionic concentrations and therefore to mediate cell metabolism. Silver et al. [108] observed higher

2761

Ca concentration in osteoblast cells and enhanced ATP generation


after adding 45S5 Bioglass to the culture medium, whereas other
study showed that an another bioactive glass composition (mol%:
50SiO2e16CaOe6P2O5e5K2Oe2Al2O3e1MgO) led to increased
cellular concentration of phosphorous and sulphur, which are
reecting the phosphorylation state and protein content of the
cells, respectively [109]. Moreover, a large number of studies have
shown the osteogenic effects of bioactive silicate glasses and their
dissolution products [26,47,56,108,110e135], as summarized in
Table 2. One key nding from Xynos et al. [26] studies in 2001 was
that Bioglass dissolution products are able to regulate gene
expression in osteoblastic human cells (HOC). Several genes known

Table 2
Biological responses to ionic dissolution products of silicate based bioactive glasses.
Glass composition
by mol%

45S5
46.1SiO2e24.3Na2Oe
27.0CaOe2.6P2O5

Ions
released

Si
Ca
P
Na

Conc.*[ppm]

Result

Remarks, Conclusions

Ref.

16.5
88.3
30.4
2937.0
16.58
88.35
30.45
2938.0
55.48
72.70
23.45
3341.66

 Ionic dissolution products directly stimulated


the up-regulation of most genes in HOC (up to
vefold) known for their relevant role in bone
metabolism

Which released ion


responsible is unclear

[26]

 Osteoblast number increased to 150%.


 Increase in osteoblast number implies increase
in cell proliferation.

Ionic products of 45S5 Bioglass


may increase proliferation rate.

[134]

 Si concentration shortens osteoblast growth cycle


and promotes the proliferation.

Exact molecular mechanism


should be claried.

[239]

 Upregulation of BSP and ALP in FOB cells

Ionic concentration in culture


medium is not known

[123]

Better proliferation on amorphous


BG is due to slower degradation of
sintered partially crystallized BG

[114]

Osteogenic supplements effect on gene


expression proling and mineralization.

[129]

[110]

 50 % increase in osteoblast proliferation


 Increased collagen production

No glass dissolution studies on the BG


were performed; ion conc. in culture
medium is unknown

[130]

 Extracellular Ca2+ concentration, via bioglass dissolution,


increases the mobilization of intracellular Ca2+
in osteoblast cells

[56]

 Good attachment, proliferation and nodule formation


of osteoblast

[117]

 Good osteoblast attachment on BG scaffold


 Nodules formation and mineralization after 10 days of
seeding (without supplements of mineralization agents)
 Higher Si concentrations (> 120 ppm) of ions lead to
apoptosis

Si concentration is likely the key factor


for mediating mineralization and nodule
formation and cell death

[120]

 No statistically signicant effect on proliferation of HOC

Author suggests higher Si content in


medium to achieve stimulating effects

[115]

 Upregulation of several genes in HOC, e.g. gp130,


MAPK3/ERK1, MAPKAPK2, and IGF-I.

First study to conrm gene expression


of sol-gel derived bioactive glass

[116]

 100% increase in osteoblast proliferation

[111]

 Stimulated differentiation of bone marrow cells into


osteoblast cells in BG (77S as well as control 45S5)
conditioned medium
 77S BG caused inhibition of osteoclast cell formation

Dissolution kinetics and ion release are


not investigated

[136]

14.5 e 19.4
47.5 e 49.9
24.6 e 23.6
4983.8 e 4895.8
MBG 85
85SiO2e10CaO
e5P2O5

BG60S
59.9SiO2e38.4CaO
e1.7P2O5
S520
52SiO2e21Na2Oe7K2O
e18CaOe2P2O5

59.2
64.0
16.9
58.06
70.07
20.02
30
90
10
47.7
0
67.63

58S
60SiO2e36CaOe4P2O5

77S
80SiO2-16CaO-4P2O5

50.02
95.8
26.4
50.2
95.8
26.4
203.11
47.06
6.88
-

 Increased cell proliferation (MG 63) on BG pellets


(2D) compared to control (Thermanox) amongst
which non-crystallized BG shows best results
 Signicantly higher cell proliferation on BG-based
scaffolds (3D) compared to control
 BG extracts create an extracellular environment for
support of osteoblast phenotype expression
 Increased osteoblast differentiation and enhanced
ECM deposition and mineralization
 Increased osteocalcin (OCN) and collagen I synthesis
 High viability of osteoblasts (Saos-2), murine broblasts
(L929) and murine lymphocytes (SR.D10 T) treated with
BG extracts
 But decreased osteoblast and broblast proliferation

*) absolute ion concentration in the cell culture medium after treatment with BG

2762

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

to play a role in osteoblast metabolism, proliferation and cellecell


and matrix-cell adhesion were up-regulated up to 5 fold [26] when
HOC were cultured in Bioglass conditioned medium. Kaufmann
et al. [126] conrmed these results observing expression of several
genes (osteocalcin, osteonectin, osteopontin) in osteoblast-like
cells and increased alkaline phosphatase (ALP) activity and collagen
I formation. Related studies by Jell at al. [123] showed six and
threefold upregulation of osteogenic markers, namely bone sioloprotein (BSP) and ALP, in osteoblasts treated with BG conditioned
culture medium resulting in enhanced cell differentiation.
Similar results have been observed for sol-gel derived bioactive
glasses. For example sol-gel derived 77S (mol%: 80SiO2e16CaOe
4P2O5) bioactive glass has been shown to induce osteogenic
differentiation of bone marrow stromal cells into osteoblast-like
cells and to promote cell mineralization [136]. In addition studies
on 58S (mol%: 60SiO2e36CaOe4P2O5) bioactive glasses have
shown that treatment with their ionic dissolution products results
in cell activation through stimulation of the proliferation of osteoblasts cells [111] and upregulation of the expression of a number
of genes including IGF-I, gpl30 or MAPK3/ERK1 [116]. Previous
work (up to 2005) on the osteogenic stimulating effects of bioactive
silicate glasses has been already reviewed [124].
Additionally, molecular effects of bioactive silicate glasses were
observed in vivo by implanting 13-93 (Vivoxid) glass particles
(wt.%: 53SiO2e6Na2Oe12K2Oe5MgOe20CaOe4P2O5) in bone
defects of rat tibia and by investigating gene expression at the
defect area [132]. In agreement with previous results, the study
revealed an osteoinductive effect of bioactive glass 13-93, where
the defects lled with the glass showed high mRNA expression of
genes for both bone formation and bone resorption, thus affecting
not only osteoblast but also osteoclasts function and enhancing
bone turn over [132]. However, the glass particles were injected
along with bone morphogenic protein 2 (BMP-2), so that synergetic
effects of addition of bioactive glass and BMP-2 on bone formation
were evaluated. Other studies have conrmed the stimulating
ability of bioactive glass extracts on osteogenesis, which are
believed to be related to gene expression as previously observed,
showing enhanced proliferation of human osteoblast cells (HOC)
and murine osteoblast cells [111,130] and increased collagen I
formation in osteoblastic cells [113,130]. It has been shown that
Bioglass conditioned culture media with Si concentrations of 15
and 20 mg l1 promoted higher metabolic activity, expression of the
Cbfa1 factor and enhanced formation of mineralized bone nodules
in primary fetal osteoblasts [129]. Additionally, it has been
observed that sol-gel derived phosphate-free glass 70S30C (mol%:
70SiO2-30CaO) is able to enhance osteoblast maturation and
differentiation as well as production of bone-like minerals by
osteoblasts indicating that P may be not necessarily required for in
vitro mineralization of the extracellular matrix [125].
Since it has become clear that successful clinical application of
scaffolds in bone tissue engineering highly depends on a functional
vascularised network, relevant research is being expanded to
include investigations on the angiogenic effects of biomaterials
[137]. Vasculature of the hard tissue plays a key role in bone
remodelling through acting as reservoir and conduit for bone cells,
growth factors and providing key signals involved in bone metabolism [138]. Thus, functional vascularisation of the scaffold
implants would result in enhanced bone metabolism and bone
formation. For this reason several studies have been carried out in
order to investigate the potential stimulating effects of bioactive
glasses on angiogenesis, using both endothelial cells and broblasts. [27e29,118,139,140]. An early study showing the angiogenic
effect of Bioglass was provided by Day et al. [140] revealing
increased neovascularization into Bioglass coated polymer
meshes after being subcutaneously implanted in rats. In further

investigations Day [27] showed that Bioglass stimulates the


secretion of angiogenic growth factors from human broblasts
cells, providing further evidence that bioactive glass enhances
angiogenesis. In the study of Day [27], broblast cells (CCD-18Co)
cultured in Bioglass particles containing medium secreted
increased amounts of vascular endothelial growth factor (VEGF)
and basic broblast growth factor (bFGF). Subsequent culturing of
endothelial cells in conditioned medium obtained from Bioglass
stimulated broblast growth resulting in increased endothelial cell
proliferation and formation of anastomosed networks of cell
tubules thus indicating enhanced angiogenesis. The studies of Day
et al. [27,140] were extended by Leu and Leach [29,139], who found
similar results. These and other studies on angiogenic effects of
bioactive glasses have been comprehensively reviewed recently
[28]. In recent investigations, Deb et al. [118] performed coculturing of osteoblast and endothelial cells showing increased
proliferation of both, human osteoblast cells and human umbilical
vein endothelial cells (HUVEC) seeded on Bioglass based scaffolds
compared to HAp scaffolds.
The large amount of results on the biological performance of
bioactive glasses discussed above conrms the hypotheses that
ionic dissolution products released from bioactive glasses stimulate the genes of cells towards a path of regeneration and selfrepair as recently discussed by Hench [141]. It is now well accepted
that bioactive silicate glasses are able to stimulate ostegenesis as
well as angiogenesis and therefore they exhibit unique properties
which make them highly attractive for bone tissue engineering
applications. However, it remains unclear which ionic products
(and in which concentration) are responsible for cell activation and
which are the exact mechanisms of interaction between the ionic
products and cells. In fact, the importance of single dissolution
products from bioactive glasses is still controversially discussed.
Based on Xynos et al. [26,134,135] studies, Hench [141] stated that
eluted Ca and Si concentrations of 60e88 ppm and 17e21 ppm,
respectively, are critical for upregulation of several osteogenic
genes. Valerio et al. [131] suggested that higher osteoblastic
proliferation and collagen secretion after treatment with BG60S
dissolution products is related to Si contact, since despite higher Ca
concentration no increased osteoblast activity was observed in
presence of BCP (no Si release). Indeed, as mentioned in the
previous section, Si has been shown to be an essential element for
metabolic processes associated with the formation and calcication
of bone tissue [58,59] and to be present in early stages of bone
matrix calcication [59]. Moreover, high Ca concentrations (of
88e109 ppm) have been shown to reduce Saos-2 osteoblast
proliferation when exposed to bioactive glass MBG 85 (mol%:
85SiO2e10CaOe5P2O5) conditioned culture, whereas a Si concentration of 60 ppm did not have any negative effect on the cell
proliferation [110]. Despite the fact that a too high Ca concentration
is toxic for human osteoblastic cells, Ca has been shown to increase
the glutamate release of osteoblast cells [56]. Since glutamate signalling pathways are known to play an important role in bone
mechanosensitivity [53], the extracellular Ca concentration must
be considered as an important stimulating agent in bone formation.
Phosphor has also been suggested to be essential for calcium
phosphate deposition and extracellular matrix mineralization,
whereby higher inorganic Phosphor (P) concentrations in osteoblast culture medium resulted in more rapid mineral deposition
[142]. Elevated phosphate levels have been shown to increase Nrf2
expression in osteoblastic cells (MC3T3-E1) when treated with
10 mmol phosphate [143]. More recently, P has been shown to
upregulate Glvr-1 and Glvr-2 in murine odontoblast-like cells
correlated with ERK1/2 phosphorylation and CaP crystallisation
[144]. However, this effect requires the presence of Ca indicating
that both, P and Ca, are essential in signalling pathways occurring in

Table 3
Biological response to ion-substituted silicate based bioactive glasses.
Glass composition (mol%)U
U
if not otherwise specidied
6.4SiO2-1.1P2O5-26.4Na2O-23.1SrO
45S5.6Sr
45SiO2e24.5Na2Oe18.5CaOe6P2O5-6SrO

Ion(s)
released

Si
Ca
P
Sr

Conc.* [ppm]

Result

Remarks

Ref.

w
w
w
w

 increased osteoblast (Saos2) activity (increased proliferation


and ALP activity) after treatment with BG extracts
 reduced osteoclast activity (RAW264.7)

[45]

 good bone bonding ability in vivo after 30 days implantation


in rat tibia bone marrow
 increased proliferation of rat calvaria osteoblastic cells
 enhanced the cell differentiation and ALP activity

No signicant difference in bioactivity


compared to undoped glass was observed
Sr release into the culture medium is not
investigated and remains unknown

21.2
85.3
19.37
0.31

 increased proliferation and differentiation of osteoblast cells

No dissolution studies in cell culture


medium were performed; thus ionic
concentration remains unknown.

[148]

 Zn doping results in increased surface area


 favors HAP formation and growth

More investigations on different


concentrations of Zn in a cellular
medium are required to evaluate specic
effect of Zn on cells

[151]

15
28
1.4
2.7

 higher Zn content (20%) decreases cell (Endothelial cells)


proliferation rate

Zn content in culture medium is


suggested to be cytotoxic

[146]

Cell response depends on the form BG is


applied, e.g. as supsension, glass extract

[159]

20
30
5
80

(wt.%): 58.8SiO2-22.3CaO-P2O5-7.9SrO

64SiO2-26CaO-P2O5-5ZnO

(wt.%): 73.7SiO2-25.3-CaO-1ZnO
74SiO2-21.2CaO-4.8ZnO

Si
Ca
P
Zn

[165]

67SiO2-5P2O5-17CaO-11ZnO/CuO

Zn
Cu

w13
w58-23

 Reduced TNF production and Il10 following pretreatment with


doped BG compared to undoped control indicating
prophylactic inammatory effect of the Zn and Cu containing
glasses
However, no therapeutic effect after post treatment with Zn
and Cu glasses detected, likely due to toxic Cu and Zn levels
released into the culture medium

(Zn Sr)- BG
40SiO2-0CaO-28SrO-32ZnO

Si
Ca
Zn
Sr

 no inammatory response In vivo with juveline rats


 showed immediate bone formation response due to higher cell
viability

Further in vivo studies are planned

[150]

 Up to 99.8 % reduction of bacteria (Staphylococcus aureus,


Escherichia coli) adhesion and growth
 Ag surface doping has no effect on acellular in vitro bioactivity.

[181]

Ag-BG
57SiO2-34CaO-6Na2O-3Al2O3
surface is doped with Ag
Ag-BG (S70C30)
70SiO2-28CaO-2Ag2O
45S5.2B
wt%: 45SiO2-24.5CaO-24.5-Na2O-P2O52B2O3

Si
Ca
Ag
B

71
155
1
-

13-93B2
wt%: 36B2O3-18SiO2-22CaO-6Na2O8MgO-8K2O-2P2O5-

0.65 mmol

HCaCaF2
46.2-SiO2-24.3 Na2O, (26.9x)CaO2.6P2O5-xCaF2
(x 0, 5, 10, 15)

15.4 (for x5)


30.4(for x10)
40.9(for x15)
after 24 h

(wt.%): 45SiO2-24.5CaO-6P2O5-24.5Na2O5TiO2/Fe2O3/B2O3
43SiO2-14.5-CaO-5.77P2O5-23.64Na2O13.1CaF2

Si
Ca
Na
P

 No cytotoxic effect on human epidermal keratinocytes


Larger area and increased thickness of neoformed bone tissue
around B-BG compared to undoped control indicating
enhanced bone formation
 Boron concentration above 0.65 mmol inhibit the growth and
proliferation of BMSCs and MLO-A5 cells
 Fluoride release causes oxidative damage of MG-73 cells
indicated through:
 Increased extracellular LDH (index of cytotoxicity) levels and
accumulation of intracellular MDA (index of lipoperoxidation) and
 Release of ROS (indicator of stress) and inhibition of PPP
(signal of cell response to oxidative stress) activity
_
Increased
osteoblast proliferation and osteoblast expression
on Ti-BG
B-BG, Fe-BG and F-Bg exhibit lower osteoblast proliferation
and activity

Different treatments to the silver ion


release may cause different cellular
responses.
Increased rate of bone formation can be
due to the enhanced degradation of
bioglass after B substitution to Si.
Threshold Boron concentration of 0.65
mmol is achieved through deluting BG
extracts indicating a too high B2O3
amount in the BG composition

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

37SiO2-18CaO-4P2O5-18Na2O-20ZnO

[164]

[180]
[191]

[188]

[195]

[198]

2763

[133]

bone metabolism. Even though phosphate-free sol-gel derived


bioactive glasses have been shown to induce extracellular matrix
mineralization even without presence of P [125], inorganic phosphates should be considered important regulating agents in bone
metabolism.
Taking all these facts into account one can state that one of the
challenges for developing scaffold materials for bone regeneration
is to achieve controlled dissolution and ion release kinetics making
sure that required critical concentrations of certain ions are
released into the physiological environment. Future in vitro studies
should therefore explicitly investigate in greater detail ion release
kinetics and the respective concentrations in cultured medium to
make results comparable within the biomaterials scientic
community, which will help to identify the specic role of single
dissolution products of bioactive glasses and inorganic materials in
general.

Enhanced expression is most likely due


to the dissoluted ions.
Only Si conc. signicantly higher than
control medium

[173]

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

Mg containing glass samples are not


compared to undoped standard bioactive
glass ceramics
In vitro results are not compared to Mgfree glass, so that effect of Mg doping on
cell behavior remains unclear

[171]

2764

6P53-b (LBL)
(wt.%): 52.7-SiO2-10.3Na2O-18CaO-6P2O52.8K2O-10.2MgO-)

*) absolute ion concentration in the cell culture medium after treatment with BG

 Enhanced stimulation of osteogenic markers expression


including Collagen I, ECM formation, ALP activity and osteocalcin
 No differences between the experimental composition and pure
Bioglass control were detected
33.4  3.8
4458  186
298.3  34.9
57.1  2.8
32.7  5.4
32.6  9.3

64SiO2-26CaO-5P2O5-5MgO

45.98SiO2-43.30CaO-10.72MgO

Mg
Si
Ca
P
Ca
Mg
Si
Na
K
Ca
Mg
P

 Increased viability and proliferation of osteoblast seeded on


glass wafers compared to control (pure culture medium).
 increased ALP activity after 7 days culturing
 Increased proliferation and differentiation of hFOB cells
seeded on glass samples compared to PS control
 No toxic effects with L929 cells

3.2. Ion-doped bioactive silicate based glasses


In order to enhance the bioactivity of bioactive glasses towards
a specic biological response in relevant physiological environments many approaches have been investigated incorporating
various metal ions in the silicate network. The main goal is to
increase the stimulating effects of bioactive glasses on osteogenesis, angiogenesis and the promotion of antibacterial properties. These approaches consider diverse bioactive glass
compositions including Co-BG [145], Zn-BG [146e161], Cu-BG
[159,162], Sr-BG [45,150,156,163e169], (Zn Sr)-BG [150,156,170],
Mg-BG [133,161,162,171e175], Ag-BG [176e184], Ce-BG [185] as
well as B-BG [154,186e194] and F-BG [195,196]. Table 3 gives
a representative overview of silicate based BGs containing specic
metal ions and the respective biological response to their ionic
dissolution products.
Diverse silicate glasses based on CaOeMgOeSiO2 with different
additive agents (B2O3, CaF2, Na2O and P2O5) have been shown to
exhibit appropriate level of acellular bioactivity proved through
standard bioactivity testing by soaking the material in simulated
body uid, according to Kokubo et al. [11,197], in order to prove the
formation of surface HCA layer [186]. In the study of Agathopoulos
et al. [186], for example, it was observed that increasing amount of
phosphates favours the deposition of HCA while increasing
contents of CaO and SiO2 inhibit its deposition. However no biological data through in vitro cell test were provided for the glass
compositions investigated. Moreover, Vrouwenvelder et al. [198]
reported on the osteoblast behaviour on 45S5 bioactive glass
doped with iron, titanium, uorine and boron giving (to our
knowledge) the rst overview of the biological response to iondoped bioactive glasses. It was shown that Ti-BG exhibited higher
proliferation and osteoblast expression probably due to more
controlled release of the ionic glass products, whereas doping with
B, Fe and F resulted in lower osteoblast activity compared to
undoped control 45S5 bioactive glass.
Cobalt containing glasses co-doped with Zn and Mg [145] have
been fabricated in order to create hypoxia-mimicking (low
oxygen pressure environment) biomaterials to be used in bone
tissue engineering. Creating hypoxia conditions is suggested to be
a strategy for activating pro- and anti-angiogenic genes [199]. Co
ions (known for their ability to mimic hypoxia) have been introduced to bioactive glass and it has been shown that Co ions are
released in proportional manner to the CoO content (incorporated
into the BG network) into TRIS solution used as dissolution medium
[145]. Co2 concentrations in the range of 10e14 ppm in the
medium were measured, which are believed to be within the biologically active limits. Moreover, HAp formation was delayed
through the incorporation of CoO, ZnO and MgO as it was revealed

Table 4
Biological response to phosphate based bioactive glasses.
Glass composition by mol%
Ions released
Phosphate glasses based on P2O5eCaOeNa2O (PBG)
ACaO-(55-A)Na2O-45P2O5
A: 30 e 48

Ca
P
Na

50P2O5-(50-X)CaO-XNa2O-

Biological response
 More soluble glasses with less than 20% CaO inhibit growth and bone
antigen expression in osteoblasts (MG63 and HOS)
 Less soluble glasses increase cell proliferation and expression of BSP,
ON and FN osteogenic factors
 PBGs containing less than 40% CaO do not support cell adhesion and
growth of human osteosarcoma cells (MG-63)
 Human oral osteoblasts (HOB) and broblasts show well spread
morphology on higher calcium containing glasses.
 All PBG investigated inhibit cell adhesion and proliferation and
increase cell death of hBMSC and HFOB 1.19 compared to TCP and
HA control

Remarks

Ref.

[210]

No dissolition were performed; ion release kinetic


and ion concentration in culture medium remains
unknwown

[211]

[212]

Cell behavior differs when applied directly to the


materials or to the materials extracts

[213]

[215]

[214]

No comparison to control samples and undoped glass,


role of Zn not specically investigated

[218]

Cytotoxicity of Mg-free PGG is likely due to higher


dissolution rates

[46]

PBG containing different metal oxides

*)

Ti-PBG
44.5P2O5e44.5CaOe6Na2Oe5TiO2

Ca
P
Na
Ti

60.1  0.06
32.3  0.003
3350.7  0.6
not detected

Ti-PBG
27.5CaO-33 P2O5-22.5-Na2O-9.5MgO5.5TiO2

Ca
P
Na
Mg

Ti-PBG
44.5CaO-44.5P2O5-6Na2O-5TiO2

Ca
P
Na
Ti

4.1  0.36
5.4  0.12
90.3  1.84
Not detected

Zn-PBG
48CaOe45P2O5e5Na2Oe2ZnO

Mg-PBG
40P2O5-25CaO-ANa2O-BMgO
A: 5 e 35
B: 0 e 30

Ca
P
Na
Mg

 No toxic effect of the glass extracts, but however slightly toxic cell
response when seeded directly on glass disc (toxic effect lower on Ti
doped glass)
 better human skin broblast cell adhesion on Ti-free glass (more soluble)
 Proliferation of MC3T3-E1.4 osteoblast like cells do not changed for
polished samples for all glasses investigated.
 MC3T3-E1.4 osteoblast proliferation is higher on porous glass samples
with lower solubility, namely BG containing 5.5 mol% TiO2.
 However for all samples (porous and non-porous) the proliferation rate
was always lower than the positive control TCP control
 Non-cytotoxic effects on osteoblast cells (SAOS-2) of glass extracts
and of the glass scaffolds in the direct testing are detected
 Cells directly seeded on the glass scaffold exhibit higher viability
(after 3 days of culturing) compared to control medium
 High degree of adhesion and spreading of mouse-derived MC3T3-E1
cells on scaffold surface
 No HCA formation after soaking in SBF for 28 days indicating
insufcient acellular bioactivity
 Decrease in dissolution rate is observed with increasing MgO content
 Cytocompatibility of Mg-PBG comparable to TCP control; Mg-free
PBG shows higher cytotoxicity

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

XCaO-(50-X)Na2O-50XP2O5
X: 30 e 48

Conc.* [ppm]

absolute ion concentration in the cell culture medium after treatment with BG

2765

2766

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

by SBF studies [145], which is, in fact, desirable for soft tissue
engineering applications, e.g. cartilage regeneration.
In the following paragraphs different studies on particular ions
and their effect on cell behaviour are summarized and the results
discussed regarding their relevance for bone tissue engineering.
3.2.1. ZinceBG
Bioactive silicate glasses doped with Zinc (Zn-BG) have been
shown to enhance acellular formation of calcium phosphate layer
on the BG surface after soaking BG substrates in biological uids
(Dulbeccos Modied Eagles Medium, DMEM) [151] and in simulated body uid (SBF) [149,155], which is a fundamental requirement for bioactive glasses to bond to bone [5]. Similarly, Zn and Fe
containing glass compositions have been shown to increase acellular bioactivity with increasing the combined Zn-Fe content [158].
However, the results on the acellular bioactivity of Zn-doped BG
are not consistent. Aina et al. [146], for example, have shown that
Zn content reduces the overall leaching activity of the glass
inhibiting the formation of the HCA layer on its surface. Haimi
et al. [154] obtained similar results showing a delayed formation of
HCA which is related to the slower degradation prole of the Zndoped bioactive glasses. The analysis of the limited literature
available indicates that further specic studies on the relationship
between microstructure and physicochemical properties of Zndoped BG are needed to determine with accuracy the inuence of
Zn-doping on the dissolution behaviour of bioactive glasses in
order to control the ion release kinetics and the biological
performance.
Independently of the acellular behaviour investigations, in vitro
cell culture studies have shown the anti-inammatory effect of Zndoped bioactive sol-gel derived 58S glass [159]. In this study,
murine macrophage cells (RAW 264.7) stimulated with lippopolysaccharide (LPS) endotoxin were cultivated in DMEM treated with
BG suspension (1 mg ml1) and glass ionic dissolution products,
respectively, resulting in decreased secreted amount of tumor
necrosis factor TNF-a and interleukin-1 which indicated the
inammatory effect on the cells. Glass compositions with 5mol-%
and 11mol-% Zn did not have any therapeutic effects on cells since
the TNF-a and IL-1 contents were not signicantly decreased after
treatment with the BG suspension. On the other hand the pretreatment of cells with bioactive glasses resulted in a signicantly
decreased amount of TNF and IL-1 indicating a prophylactic antiinammatory effect of the Zn-doped glasses compared to the
undoped control glasses. However, cell treatments with ionic
dissolution products of the same glass did not show any positive
anti-inammatory effects of the Zn-doped glasses compared to
undoped control glass, which might be related to the high
concentrations of Zn (up to 16 ppm) released into the cell medium
being toxic for cells. Other studies have also shown that Zn
concentrations in the range of 2e8 ppm can cause damage in
human osteoblasts via oxidative stress [147]. Similar results were
observed for endothelial cells seeded on wells in the presence of
Zn-doped glass slabs revealing that bioactive glasses doped with
20 wt.% Zn increase adhesion of cells but decrease the proliferation
rate probably due to toxic Zn concentration of 2.7 ppm released into
the culture medium, whereas 5Zn-BG promoted well cell adhesion
as well as high cell proliferation of endothelial cells at Zn concentrations of 1.1 ppm [146]. However, results on cell adhesion related
to the undoped glass composition, indicated that all glass samples,
undoped 58S, 5%Zn-58S and 20%Zn-58S, exhibited poorer cell
adhesion and proliferation than the silica control samples.
Additionally, Zn-releasing bioactive glass scaffolds have been
investigated in relation to their possible osteogenic effects
[148,153e155,157]. It has been shown that sol-gel derived glasses
containing 5 mol% ZnO resulted in increased ALP activity and

increased osteoblast proliferation [148] indicating the possible


stimulating effect of Zn-BG on osteoblast cells.
In related research, Oki et al. [157] observed that Zn containing
bioactive glass (5 mol%) increased AP activity of human fetal osteoblastic cells (hFOB 1.19) when seeded on glass discs in relation to
polystyrene control. However, these results were not compared to
undoped glass samples, so that no denitive statement on the
stimulatory effect of Zn can be made based on the available
experimental evidence. More recently, Haimi et al. [154] investigated the effects of Zn-doped BG scaffolds on human adipose stem
cells proliferation and osteogenic differentiation revealing no
signicant effect of Zn-doping on cell activity when cells were
seeded on the Zn-doped BG scaffolds. The authors suggested that
the possible stimulatory effect of Zn is inhibited through the
decreased degradation prole of the bioactive glass caused by the
Zn addition. Similar results were observed by Lusvardi et al. [155],
who found that Zn-doping provides no signicant effect on adhesion and proliferation of osteoblast-like cells (MC3T3-E1)
compared to undoped control glasses, probably due to the slowed
degradation of the BG caused by Zn addition resulting in a negligible amount of Zn ions released. However, the degradation
kinetics was investigated only in SBF (release of 4.6 ppm after 70
days) and no cell cultures treated with ionic dissolution products of
the Zn-doped glasses were carried out missing a specic investigation of possible anabolic effect of Zn. Despite the fact that in vitro
studies with systematic supplements of Zn ions showed osteogenic
effect of Zn (as previously described), to our knowledge no clear
data has been presented to date to conrm in a denite way the
osteogenic effect of Zn released as ionic dissolution product from
Zn-doped bioactive silicate glasses.
3.2.2. Strontium-BG
Sr-doped glasses have been shown to exhibit enhanced acellular
bioactivity and to release critical concentrations of Sr ions in the
range of 1e5 ppm into the dissolution medium [166,167]. Moreover, Gentleman et al. [45] have shown that ion release from Srdoped silicate glasses enhances bone cell activity. They investigated
the in vitro behaviour of SiO2eP2O5eNa2OeCaO silicate glass in
which Ca was systematically replaced by Sr up to 100%. In this
study, treatment with ionic dissolution products of Sr-doped
glasses (ion release in culture medium in the range of 5e23 ppm)
resulted in enhanced osteoblast (Saos-2 cell line) activity and
inhibited osteoclasts differentiation. Moreover, these Sr-doped
glasses promoted osteoblast proliferation and ALP activity when
directly applied in contact with cells as solid BG-discs. Similarly, it
has been shown that Sr-doped BGs produce increased proliferation
of rat calvaria osteoblastic cells and enhance cell differentiation and
ALP activity [165]. In vivo studies have also conrmed the high
biocompatibility of Sr-containing 45S5 Bioglass expressed
through strong bonding to bone via HCA layer without any
inammatory affects, though no differences of the in vivo behaviour compared to undoped Bioglass control were observed [164].
(Zn Sr)-co-doping has been also applied to create novel
bioactive glass compositions. For example, Murphy et al. [156]
showed that SiO2eCaOeNa2OeSrOeZnO2 glasses release Zn and
Sr concentrations in the 3e18 ppm and 0e3500 ppm ranges,
respectively, having therapeutic potential with capacity for
controlled release of Zn and Sr ions. Moreover, another study by
Murphy et al. [170] ovserved higher viability of mouse broblast
cells (L929) when ionic extracts of these ZneSr doped glasses were
applied to the cell culture medium. However, no specic stimulating effect of the a certain ion released in to the culture medium
could be identied and the authors suggest that the improved
biocompatibility of the Sr and Zn containing glasses compared to
Novabone is likely due to synergetic effects of all ions released

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

from the glass as dissolution products. Boyd et al. [150] observed


that 0.4SiO2-0.32ZnO-0.28SrO (mol%) glass exhibits higher cell
viability compared to 0.4SiO2-0.32ZnO-0.28CaO and 0.4SiO20.32ZnO-0.14CaO-0.14SrO glasses as well as to Novabone
(commercial Bioglass product) control, indicating the positive
effects of the Sr-doping. However, no dissolution studies were
performed and it was only assumed from the authors that the lower
cytotoxicity of the Sr-doped glass was due to higher chemical
stability resulting in a lower amount of Sr ions released into the
culture medium.
3.2.3. Magnesium-BG
Bioactive glasses doped with Mg (Mg-BG) have been shown to
have improved dissolution behaviour due to the Mg effect disrupting the silica network [172]. On the other hand these glasses
exhibit decreased crystallisation of the HAp layer on the BG surface
[172]. Varanasi et al. [133] observed enhanced expression of
collagen I, ALP, Runx2 and osteocalcin osteogenic markers after
treatment of the cell culture medium with Mg-BG-dissolution
products indicating their signicant effect on the osteoblast
differentiation. However, Mg concentration in culture medium did
not change signicantly after treating with BG, so that the osteogenic effect of this particular BG could be related to increased levels
of Ca and Si. Other studies have shown an increased osteoblast
proliferation and differentiation [171] as well as enhanced ALP
activity in human fetal osteoblastic cells (hFOB 1.19) [173] and
enhanced expression of osteogenic markers including OP, OC, ON in
human bone derived cells (HBDC) [200]. These ndings conrm the
good biocompatibility of BGs with MgO addition. However, Mgdoped samples have not been compared to undoped control
glasses; thus, it remains unclear whether or not Mg released from
these bioactive glasses has a specic osteogenic effect.
3.2.4. Flouride-BG (F-BG)
CaF2 has also been considered as promising doping agent to
enhance the biocompatibility of bioactive glasses, since CaF2 is
known to inhibit the formation of alveolar cavities [201] and to
provide higher acidic resistance of enamel by substituting OH sites
in dental apatite [202]. Fluorides are also known for stimulating
effects on osteoblast cells when applied at moderate concentrations (25e500 ng ml1), whereas higher concentrations (>500 ng
ml1) suppress osteoblast activity [203]. However, CaF2 doped
bioactive glasses have been shown to be cytotoxic and to promote
oxidative cell damage as indicated by increased lactate dehydrogenase (LDH, indicator of cytoxicity) formation and accumulated
extracellular malonyldialdehyde (index of lipoperoxidation) when
investigated in contact with osteoblasts (MG 63) [195]. Only acellular bioactivity studies on F-doped bioactive glasses showed that
incorporation of F in silicate glasses results in a smaller pH raise
(which is favourable for the cells) and favoured formation of
ourapatite (FAp), which is chemically more stable than HAp and
thus a very promising material for dental applications [196].
3.2.5. Boron-BG
Boron doped 45S5 Bioglass (B-BG) materials with different
boron contents have been shown to exhibit increased acellular
bioactive behaviour as the rate of HAp formation on the surface
during soaking in P-rich solution increases [187]. However, with
increasing B-content the proliferation of osteoblastic like cells
decreased, with B-concentration of 1.5 mmol in the culture
medium having inhibiting effect on cell proliferation. Similar
results were found by Fu et al. [188] who observed accelerated HAp
formation of boron doped BG but inhibited cell proliferation with
increasing B-content in culture medium. In the study of Fu et al.
[188] a boron concentration of 0.65 mmol in the cell culture

2767

medium was shown to be the threshold concentration, whereas B


values below 0.65 mmol supported the proliferation of preosteocytic MLO-A5 cells indicating good biocompatibility of the
boron doped glass. Moreover, a Boron concentration above
0.65 mmol was shown to decrease the growth and proliferation
rate of bone marrow cells (BMSc). However, the dissolution products of the processed BG with a Boron oxide content of 36 mol%
were diluted up to a ratio of 1:8 indicating that the B2O3 content in
the BGs investigated was too high [188]. Clear evidences of
enhanced biological performance of boron doped BG have been
observed in vivo by Gorustovich et al. [189,191], who found
a signicant increase of quantity of bone tissue formed around
Boron-BG particles after implantation in wist rats indicating
simulating effects of boron doped bioactive glasses on osteogenesis.
However, more in vitro studies on boron modied bioactive glasses
are needed to clearly identify the effect of these glasses on human
cells and to conrm the potential simulating effect of boron, as
discussed previously (Section 2).
3.2.6. Silver-BG
Since Ag ions are known for their antibacterial effects
[204e206] Ag-doped silicate glasses (Ag-BG) have been developed
to create bioactive glasses exhibiting inhibitory effects on bacterial
growth using different techniques including sol-gel [176e180] and
ion exchange processes [181e184]. It has been conrmed that Ag
ions with concentrations in the range of 0.05e0.20 mg ml1
released as dissolution products from Ag-doped BGs inhibit the
growth of different bacterial strains (Escherichia coli, Pseudomonas
aeruginosa, Staphylococcus aureus) [176]. Moreover, an ion exchange
process has been applied to glass-ceramic scaffolds in order to
fabricate Ag-containing porous structures with antibacterial properties for bone tissue engineering [183]. Thus, Ag-doped BGs are
considered highly promising materials for tissue engineering
applications with anti-inammatory properties that are attractive
for wound healing applications as well.
All ion-doped bioactive glasses discussed above have been
shown to exhibit sufcient levels of acellular bioactivity indicated
through the formation of carbonated hydroxapapatite layer after
soaking in SBF which is an essential requirement of biomaterials to
form a strong bonding to bone [5]. Moreover, doping with different
metal ions seems to be a promising approach to enhance the
biocompatibility of the glasses and to stimulate cell proliferation, as
it was shown for a few specic novel glass compositions discussed
above. However, more quantitative experimental data on the biological response of relevant cells to ion-doped bioactive glasses are
needed to conrm the specic role of the ions released as dissolution products from those silicate systems in the context of bone
tissue engineering.
4. Phosphate based glasses
4.1. Basic compositions: (CaO)-(Na2O)-(P2O5)
Phosphate based glasses (PBG) have gained high interest as
bone lling material and for fabricating scaffolds for bone tissue
engineering because of their high solubility, which can be tailored
by varying the composition [207], and their chemical similarity to
the inorganic phase of human bone [208]. Moreover, early studies
on phosphate glasses have shown that they are non toxic with
regard to macrophage cells causing low level of activation and
promoting adequate support for osteoblastic cell responses [209].
Ionic dissolution products of highly soluble phosphate glasses have
been shown to affect osteoblastic cell behaviour in a dissolution
rate depending manner. Salih et al. [210] observed that glass
compositions with lower CaO content, and therefore lower

2768

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

dissolution rate, up-regulated the proliferation of osteoblast cells


and expression of bone sioloprotein (BSP), osteonectin (ON),
bronectin (FN) genes, whereas glasses with higher CaO content
(and higher dissolution rate) caused inhibition of osteoblastic cell
growth and bone antigen expression. Similar results were obtained
by Bitar et al. [211] who found that phosphate based glasses containing at least 46 mol% CaO do not produce any toxic effect on cells,
but support attachment and proliferation of MG63 cells and human
oral osteoblasts (HOB). However, studies carried out by Skelton
et al. [212] revealed that the presence of PBGs with varying
amounts of Na2O in the culture medium reduced the adhesion and
proliferation and increased the death of human bone marrow
stromal cells (hBMSC) and human felt osteoblast (HFOB 1.19)
compared to TCP and HAp control.
In order to tailor the solubility of phosphate glasses several
oxides such as TiO2 [213e215], Al2O3 [216], B2O3 [217], ZnO [218],
MgO [46,219,220], SrO [221], CuO [222] and Fe2O3 [219,223] have
been investigated as additives incorporated into the phosphate
based glass compositions in order to stabilize the glass network and
to control the degradation rate. The biological performance of the
resulting phosphate based glass compositions is discussed in the
following section. Table 4 gives an overview of the biological
response to phosphate based bioactive glasses containing different
metal oxides.
4.2. Variant compositions
Clearly, the varying solubility of bioactive phosphate glasses
affects their biological performance since, as mentioned above,
ionic dissolution products are known to stimulate angiogenesis and
osteogenesis in vivo and in vitro. For example, addition of TiO2 has
been shown to reduce the solubility of the P2O5eCaOeNa2OeTiO2
system and to affect the cellular response of this glass [213]. The
addition of 5 mol% TiO2 resulted in a slower degradation of the
glass. Additionally, the more soluble glass exhibited a higher toxic
effect on human broblast cells than the undoped glass (when
applied as glass extracts), but, surprisingly, an increased cell
attachment was observed when cells were directly seeded on the
glass samples. Another study by Navarro et al. [214] conrmed the
non-cytotoxicity of the same glass composition by obtaining positive results from both cell treatment with glass extracts and direct
cell seeding on the glass samples. Brauer et al. [215] investigated
the inuence of SiO2 and TiO2 doping on the degradation behaviour
and the biological performance of P2O5eCaOeMgOeNa2O glass.
The authors reported that addition of TiO2 decreased the solubility
of the glass in water and SBF, while SiO2 doping resulted in
increased dissolution rates. However, the cell culture test revealed
that proliferation of MC3T3-E1.4 osteoblast-like cells did not
change for dense samples of all glasses investigated. The cell
proliferation on porous samples, however, was changed by the
varying chemistry of the glasses fabricated. It was shown that cell
proliferation was higher on glass samples with lower solubility,
namely PBG containing 5.7 mol% TiO2. However, for the cells seeded
on all samples, porous and non-porous, the proliferation rate was
always lower than the one on the positive control material (Tissue
Culture Polysterene, TCPS).
Addition of MgO to P2O5eCaOeNa2O glasses has also been
shown to decrease the degradation rate and thus to inuence the
cell response [46]. Compared to TCP positive control, PBG compositions with O mol% MgO caused the lowest anabolic activity of
osteoblast cells, while MgO contents of 10, 20 and 30 mol% resulted
in osteoblast activity at similar levels as the positive control. Leonardi et al. [220] investigated phosphate based glasses with SiO2,
MgO and K2O additions observing good cell adhesion and viability
of hMSCs seeded on the glass samples though it was lower

compared to the positive TCP control. Moreover, gene expression


levels of collagen and osteocalcin, presented in early osteogenic cell
differentiation, were higher compared to TCP indicating stimulating effects of the glass dissolution products on osteogenesis.
However, cell culture tests were carried out using the direct contact
method and the concentration of the ions released into the culture
medium were not measured. Abou Neel et al. [221e223] extensively reported on degradation kinetics of phosphate glasses
modied with several oxides including Fe2O3, CuO or SrO and their
biological performance. It has been shown that adding 1e5 mol%
Fe2O3 to P2O5eCaOeNa2O glass bres reduced the degradation rate
of the material in water [223]. Inclusion of 1e10 mol% CuO [222]
resulted in decreased glass degradation but, on the other hand, it
was shown to increase the amount of Cu2 ions released due to the
increased surface area to volume ratio. The release of Cu2 ions has
been shown to have antibacterial effects reducing the number of
Staphylococcus epidermis cultured on the glass bres, whereby the
addition of 10 mol% CuO exhibited the strongest effect amongst the
samples tested. Doping with 1e5 mol% SrO has been shown to
increase the degradation rate of the glass and to exhibit better
cellular response related to HOS viability compared to Sr-free
samples [221]. However, the cellular response was still lower than
that on the positive control (not specied in the study).
Pickup et al. [224] reported on Ga-doped PBG containing molar
fractions of Ga2O3 in the range of 0.01e0.05. The Gallium doping
resulted in an increased stability of the glass network, which is
relevant for potential use of Ga-PBG materials as delivery system of
antibacterial Ga3 ions for biomedical applications. In fact, recent
studies by Mourino et al. [225] on Ga-alignate coated bioactive
glass scaffolds have conrmed the antibacterial characteristics of
Ga3 ions.
5. Other calcium and silicate based compounds
Apart from bioactive silicate and phosphate based glass
compositions discussed above numerous other inorganic materials
and their dissolution products are being considered as promising
materials for bone tissue engineering applications, including
sulphates [226], silicate ceramics [160,227e230], xerogels [231]
and cements [232]. Relevant studies are summarized and discussed in this section.
Mesoporous silica xerogels (SiO2eCaOeP2O5, SXC) with varying
amounts of Ca (0, 5, 10 and 15), for example, have been studied in
order to investigate the role of calcium on murine osteoblast cell
(MC3T3-E1) behaviour [231]. It has been shown that samples
containing small amounts of Ca (5 wt.%) can stimulate cell differentiation indicated through increased ALP activity, whereas higher
Ca contents tend to decrease ALP stimulation levels. Moreover, the
nitric oxide (NO) levels of cultured cells has been shown to increase
with increasing Ca content of the silica xerogels [231], which can be
of interest for biomedical applications, as NO is known to play
a signicant role in several physiological processes including
neuronal communication, neurotransmission, inammation as well
as angiogenesis, stem cell proliferation and differentiation [233].
Several approaches have been put forward to use synthetic materials, including metallic and silica nano-particles, polymeric particles and carbon nanotubes (CNT), as NO release agents for
biomedical applications as recently reviewed in the literature [233].
Shin et al. [234,235] and Hetric et al. [236] for instance have
synthesized and characterized NO-containing silica nano-particles
and have shown that NO release from these scaffolds induces
strong antibacterial effects against Pseudomonas Aeruginosa, while
being not toxic to mouse broblast cells (L929) [236].
Further it has been shown that Zn-doped calcium sulphate
cements exhibit higher cell viability and increased ALP activity

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

when applied to the culture medium of human osteosarcoma cells


(G-292) [226]. Hereby samples with 0.74 wt.% Zn showed the best
result whereas higher Zn concentrations (>w2 wt.% Zn) resulted in
decreased cell viability indicating toxic behaviour of these samples.
Dicalcium silicate coatings [229] have been also used to
generate cell cultures containing critical Ca, Si and P concentrations
and to stimulate cell activity in vitro. Sun et al. [229] have shown
that culture medium with ionic products of CaSiO4 coatings can
stimulate osteoblast-like cells (MG 63) towards an enhanced
transition from the G1 to the S stage and from G2 to the M stage of
the cell cycle, respectively, indicating stimulated cell proliferation.
Sol-gel derived hardystonite ceramics (Zn stabilised calcium
silicate, Ca2ZnSiO7) have also been considered as promising materials for bone tissue engineering [227,228]. These materials have
been shown to increase the proliferation rate of MSCs when
exposed to Ca2ZnSiO7 immersed culture medium [227] and to
induce osteogenic differentiation of MSCs [227]. Moreover, hardystonite ceramics have been shown to support cell attachment,
increase cell proliferation and differentiation and to stimulate
expression of ALP, osteocalcin and collagen I when in contact with
HOB cells [228].
Zreiqat et al. [160] reported on the biological response of Srdoped hardystonite ceramic based scaffolds. It has been shown that
Sr-doped Hardystonite (Sr-HT, Sr-Ca2ZnSiO7) showed acellular
bioactivity indicated by HAp formation in SBF compared to HT
samples without Sr substitution (Ca2ZnSiO7) which did not show
HAp precipitations. Moreover, Sr-Ca2ZnSiO7 scaffolds have been
shown to increase the proliferation rate of HOB cells seeded on the
scaffolds compared to pure Ca2ZnSiO8 samples. In vitro studies on
the differentiation of HOB cells revealed that although the ALP
activity of cells seeded on Sr-HT and HT were below the TCP control,
Sr-HT samples showed signicant upregulation of osteocalcin (an
osteoblast maturation marker) compared to HT, b-TCP and tissue
culture plastic (TCP) control. In vivo studies conrmed the high
biocompatibility of pure and Sr-Hardystonite scaffolds revealing
increased osteoconductivity compared to b-TCP control, indicated
through greater bone in-growth into the scaffolds after 3 and 6
weeks of implantation in tibia bone defects in rats [160].
Strontium silicate powders (SrSiO3) have also been developed in
order to create novel resorbable materials for bone regeneration
[230]. SrSiO3 compounds, synthesized through chemical precipitation methods, have been evaluated in terms of acellular bioactivity and their effect on mouse broblast cells (L929) and rabbit
bone marrow stem cells (rMSC). SrSiO3 powders were shown to
form carbonated hydroxyapatite layer after 7 days of immersion in
SBF, which is an indication of sufcient acellular bioactivity.
Moreover, SrSiO3 powders showed no cytotoxicity when their ionic
extracts were applied to the L929 cell culture and they even
increased the cell proliferation of rMSCs. In this context Sr
concentrations in the range of 6.25e50 mg have been shown to
exhibit benecial effects on cellular response, whereas higher Sr
contents showed deleterious effects on cells.
6. Discussion
The control of metallic ion release from inorganic scaffolds is
being considered as an attractive approach to enhance the biological capability of scaffolds for bone tissue engineering. The present
review has discussed comprehensively reports in the open literature which have investigated specically ion release effects on
bioactivity from bioactive glasses and glass-ceramics.
Ionic dissolution products from bioactive silicate glasses have
been shown to stimulate osteogenesis and angiogenesis via activation of several genes associated with bone formation and vascularisation, as schematically shown in Fig. 1, making bioactive

2769

glasses (including also phosphate glasses) a group of materials of


choice for tissue repair and bone tissue engineering applications.
Moreover, new approaches have been put forward to advance the
eld of bioactive glass based scaffolds through introducing active
metal ions into the glass network in order to use the therapeutic
effects of these ions and to improve the biological performance of
the materials towards a specic host response. Indeed, the dissolution of ion-doped bioactive glasses has been shown to result in
controlled release of critical concentrations of metal ions showing
in some cases increased osteoblast activity (Sr-BG) or benecial
anti-inammatory effects (Zn-BG). However, more experimental
evidences are necessary to conrm the therapeutic effects of single
biologically active metal ions released as dissolution products from
bioactive glasses. Hereby, the accurate evaluation of the results is
hindered by the fact that the biological performance of the material
depends on whether it is applied as dense substrate, porous scaffolds, particle suspension or ionic extract [153] and whether cell are
seeded directly on the material or are applied to liquid extracts
containing the dissolution products [159,213,228]. Clearly, applying
glass extracts to the cells does not consider factors like surface
roughness, surface energy, surface electric charge, substrate stiffness as well as pore curvature and pore size [237,238] which all are
known to inuence the cell behaviour. Moreover, adding high
amounts of metal oxides to the glass network results in overall
changed dissolution behaviour of the glass, making hard to
compare the results to those on unmodied control glass [146,154].
Several different methods, schematically shown in Fig. 2, are being
applied by investigators worldwide to investigate the biological
response to biomaterials regarding their ionic dissolution products
leading to a large amount of experimental data which are difcult
to compare. The development of standardised methods for testing
the biological response to ionic dissolution products of inorganic
materials is suggested to improve the quantitative evaluation of the
experimental results and to gain a more complete understanding of
the cell response to these biomaterials.
7. Conclusions
A comprehensive analysis of the available literature on the effect
of ionic dissolution products of inorganic bioactive glasses and
glass-ceramics (silicate and phosphate systems) in the context of
bone tissue engineering has been presented. Taking into account
the experimental evidence summarized in this review and the
discussion above, a signicant challenge for developing scaffolds
for bone regeneration based on these inorganic materials is to
achieve controlled ion dissolution and release kinetics ensuring
that required critical concentrations of certain ions are released
into the physiological environment in a predetermined manner.
Future in vitro studies should therefore explicitly investigate the
release kinetics of specic ions (and combination of ions) and their
respective concentrations in cultured medium in order to make
results comparable among different materials. A systematic
approach like this will help to identify the specic role of single
dissolution products of bioactive glasses (and inorganic materials in
general). Fabricating polymer/glass-ceramic scaffolds has been
approached to achieve controlled dissolution behaviour of the
composite scaffold and the desired ion release proles [6].
However, in this case the effect of the degrading polymer matrix on
the biological performance of the composite material must be
considered, as there will be a combined effect of polymer and
inorganic material degradation kinetics on cellular behaviour.
The greater understanding of molecular mechanisms and
chemical pathways dictating the interaction between materials and
cells will allow constructing tissue specic scaffolds with tailored
chemistry, surface topography, porosity and ion release kinetics,

2770

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

which will induce the desired biological effect in the physiological


environment. In this regard, in future material-based approaches,
bioactive glasses and glass-ceramics can serve as valid biomaterial
platforms for the delivery of tailored concentrations of predetermined ions for targeted cellular or tissue responses.
Appendix
Figures with essential colour discrimination. Certain gures in
this article, particularly Figs. 1and 2 are difcult to interpret in black
and white. The full colour images can be found in the online
version, at doi:10.1016/j.biomaterials.2011.01.004.
References
[1] Palangkaraya A, Yong J. Population ageing and its implications on aggregate
health care demand: empirical evidence from 22 OECD countries. Int J Health
Care Finance Econ 2009;9(4):391e402.
[2] Hutmacher DW. Scaffolds in tissue engineering bone and cartilage. Biomaterials 2000;21(24):2529e43.
[3] Moroni L, de Wijn JR, van Blitterswijk CA. Integrating novel technologies to
fabricate smart scaffolds. J Biomater Sci Polym Ed 2008;19:543e72.
[4] Dorozhkin SV. Bioceramics of calcium orthophosphates. Biomaterials 2010;
31(7):1465e85.
[5] Hench LL. Bioceramics. J Am Ceram Soc 1998;81(7):1705e28.
[6] Rezwan K, Chen QZ, Blaker JJ, Boccaccini AR. Biodegradable and bioactive
porous polymer/inorganic composite scaffolds for bone tissue engineering.
Biomaterials 2006;27(18):3413e31.
[7] Gerhardt L-C, Boccaccini AR. Bioactive glass and glass-ceramic scaffolds for
bone tissue engineering. Materials 2010;3(7):3867e910.
[8] Jones JR, Hench LL. Regeneration of trabecular bone using porous ceramics.
Curr Opin Solid State Mater Sci 2003;7(4e5):301e7.
[9] Hench LL. Bioceramics: from concept to clinic. J Am Ceram Soc 1991;74
(7):1487e510.
[10] Suchanek W, Yoshimura M. Processing and properties of hydroxyapatitebased biomaterials for use as hard tissue replacement implants. J Mater Res
1998;13(1):94e117.
[11] Kokubo T. Apatite formation on surfaces of ceramics, metals and polymers in
body environment. Acta Mater 1998;46(7):2519e27.
[12] Annaz B, Hing KA, Kayser M, Buckland T, Di Silvio L. Porosity variation in
hydroxyapatite and osteoblast morphology: a scanning electron microscopy
study. J Microsc (Oxf) 2004;215:100e10.
[13] Hott M, Noel B, BernacheAssolant D, Rey C, Marie PJ. Proliferation and
differentiation of human trabecular osteoblastic cells on hydroxyapatite.
J Biomed Mater Res 1997;37(4):508e16.
[14] Rouahi M, Champion E, Hardouin P, Anselme K. Quantitative kinetic analysis
of gene expression during human osteoblastic adhesion on orthopaedic
materials. Biomaterials 2006;27(14):2829e44.
[15] Shu R, McMullen R, Baumann MJ, McCabe LR. Hydroxyapatite accelerates
differentiation and suppresses growth of MC3T3-E1 osteoblasts. J Biomed
Mater Res A 2003;67A(4):1196e204.
[16] Sun JS, Liu HC, Chang WHS, Li J, Lin FH, Tai HC. Inuence of hydroxyapatite
particle size on bone cell activities: an in vitro study. J Biomed Mater Res
1998;39(3):390e7.
[17] Ogata K, Imazato S, Ehara A, Ebisu S, Kinomoto Y, Nakano T, et al. Comparison
of osteoblast responses to hydroxyapatite and hydroxyapatite/soluble
calcium phosphate composites. J Biomed Mater Res A 2005;72A(2):127e35.
[18] Ehara A, Ogata K, Imazato S, Ebisu S, Nakano T, Umakoshi Y. Effects of alphaTCP and TetCP on MC3T3-E1 proliferation, differentiation and mineralization. Biomaterials 2003;24(5):831e6.
[19] Okuda T, Ioku K, Yonezawa I, Minagi H, Kawachi G, Gonda Y, et al. The effect
of the microstructure of beta-tricalcium phosphate on the metabolism of
subsequently formed bone tissue. Biomaterials 2007;28(16):2612e21.
[20] Takami M, Mochizuki A, Yamada A, Tachi K, Zhao B, Miyamoto Y, et al.
Osteoclast differentiation induced by synthetic octacalcium phosphate
through receptor activator of NF-kB ligand expression in osteoblasts. Tissue
Eng Part A 2009;15(12):3991e4000.
[21] Wang J, Jd Boer, Kd Groot. Proliferation and differentiation of osteoblast-like
MC3T3-E1 cells on biomimetically and electrolytically deposited calcium
phosphate coatings. J Biomed Mater Res A 2009;90A(3):664e70.
[22] Kondo N, Ogose A, Tokunaga K, Umezu H, Arai K, Kudo N, et al. Osteoinduction with highly puried beta-tricalcium phosphate in dog dorsal
muscles and the proliferation of osteoclasts before heterotopic bone
formation. Biomaterials 2006;27(25):4419e27.
[23] Sun L, Wu L, Bao C, Fu C, Wang X, Yao J, et al. Gene expressions of collagen
type I, ALP and BMP-4 in osteo-inductive BCP implants show similar pattern
to that of natural healing bones. Mater Sci Eng C 2009;29(6):1829e34.
[24] Hench LL, Xynos ID, Polak JM. Bioactive glasses for in situ tissue regeneration.
J Biomater Sci Polym Ed 2004;15:543e62.

[25] Boccaccini AR, Blaker JJ. Bioactive composite materials for tissue engineering
scaffolds. Expert Rev Med Devices 2005;2:303e17.
[26] Xynos ID, Edgar AJ, Buttery LDK, Hench LL, Polak JM. Gene-expression
proling of human osteoblasts following treatment with the ionic products
of Bioglass 45S5 dissolution. J Biomed Mater Res 2001;55(2):151e7.
[27] Day RM. Bioactive glass stimulates the secretion of angiogenic growth factors
and angiogenesis in vitro. Tissue Eng 2005;11(5e6):768e77.
[28] Gorustovich AA, Roether JA, Boccaccini AR. Effect of bioactive glasses on angiogenesis: a review of in vitro and in vivo evidences. Tissue Eng Part B Rev; 2009.
[29] Leu A, Leach J. Proangiogenic potential of a collagen/bioactive glass substrate.
Pharm Res 2008;25(5):1222e9.
[30] Allan I, Newman H, Wilson M. Antibacterial activity of particulate Bioglass
against supra- and subgingival bacteria. Biomaterials 2001;22(12):1683e7.
[31] Gorriti MF, Lpez JMP, Boccaccini AR, Audisio C, Gorustovich AA. In vitro
study of the antibacterial activity of bioactive glass-ceramic scaffolds. Adv
Eng Mater 2009;11(7):B67e70.
[32] Hu S, Chang J, Liu M, Ning C. Study on antibacterial effect of 45S5 Bioglass.
J Mater Sci Mater Med 2009;20(1):281e6.
[33] Jones J, Ehrenfried L, Saravanapavan P, Hench L. Controlling ion release from
bioactive glass foam scaffolds with antibacterial properties. J Mater Sci Mater
Med 2006;17(11):989e96.
[34] Leppranta O, Vaahtio M, Peltola T, Zhang D, Hupa L, Hupa M, et al. Antibacterial effect of bioactive glasses on clinically important anaerobic bacteria
in vitro. J Mater Sci Mater Med 2008;19(2):547e51.
[35] Munukka E, Leppranta O, Korkeamki M, Vaahtio M, Peltola T, Zhang D,
et al. Bactericidal effects of bioactive glasses on clinically important aerobic
bacteria. J Mater Sci Mater Med 2008;19(1):27e32.
[36] Stoor P, Sderling E, Salonen JI. Antibacterial effects of a bioactive glass paste
on oral microorganisms. Acta Odontol Scand 1998;56(3):161e5.
[37] Yli-Urpo H, Nrhi T, Sderling E. Antimicrobial effects of glass ionomer
cements containing bioactive glass (S53P4) on oral micro-organisms in vitro.
Acta Odontol Scand 2003;61(4):241e6.
[38] Zhang D, Lepparanta O, Munukka E, Ylanen H, Viljanen MK, Eerola E, et al.
Antibacterial effects and dissolution behavior of six bioactive glasses.
J Biomed Mater Res A 2010;93(2):475e83.
[39] Day RM, Boccaccini AR. Effect of particulate bioactive glasses on human
macrophages and monocytes in vitro. J Biomed Mater Res A 2005;73A
(1):73e9.
[40] Saltman PD, Strause LG. The role of trace minerals in osteoporosis. J Am Coll
Nutr 1993;12(4):384e9.
[41] Beattie JH, Avenell A. Trace element nutrition and bone metabolism. Nutr Res
Rev 1992;5(01):167e88.
[42] Nielsen F. New essential trace elements for the life sciences. Biol Trace Elem
Res 1990;26e27(1):599e611.
[43] Balas F, Prez-Pariente J, Vallet-Reg M. In vitro bioactivity of siliconsubstituted hydroxyapatites. J Biomed Mater Res A 2003;66A(2):364e75.
[44] Vallet-Regi M, Arcos D. Silicon substituted hydroxyapatites. A method to
upgrade calcium phosphate based implants. J Mater Chem 2005;15(15):
1509e16.
[45] Gentleman E, Fredholm YC, Jell G, Lotbakhshaiesh N, ODonnell MD, Hill RG,
et al. The effects of strontium-substituted bioactive glasses on osteoblasts
and osteoclasts in vitro. Biomaterials 2010;31(14):3949e56.
[46] Ahmed I, Parsons A, Jones A, Walker G, Scotchford C, Rudd C. Cytocompatibility and effect of increasing MgO content in a range of quaternary invert
phosphate-based glasses. J Biomater Appl 2010;24(6):555e75.
[47] Gough JE, Notingher I, Hench LL. Osteoblast attachment and mineralized
nodule formation on rough and smooth 45S5 bioactive glass monoliths.
J Biomed Mater Res A 2004;68A(4):640e50.
[48] Karageorgiou V, Kaplan D. Porosity of 3D biomaterial scaffolds and osteogenesis. Biomaterials 2005;26(27):5474e91.
[49] Cortizo AM, Molinuevo MS, Barrio DA, Bruzzone L. Osteogenic activity of
vanadyl(IV)-ascorbate complex: evaluation of its mechanism of action. Int J
Biochem Cell Biol 2006;38(7):1171e80.
[50] Marie PJ, Ammann P, Boivin G, Rey C. Mechanisms of action and therapeutic
potential of strontium in bone. Calcif Tissue Int 2001;69(3):121e9.
[51] Yamaguchi M. Role of zinc in bone formation and bone resorption. J Trace
Elem Exp Med 1998;11(2e3):119e35.
[52] Jerosch J, Bader A, Uhr G. Knochen, curasan Taschenatlas spezial. Stuttgart:
Thieme Verlag; 2002.
[53] Hinoi E, Takarada T, Yoneda Y. Glutamate signaling system in bone.
J Pharmacol Sci 2004;94(3):215e20.
[54] Maeno S, Niki Y, Matsumoto H, Morioka H, Yatabe T, Funayama A, et al. The
effect of calcium ion concentration on osteoblast viability, proliferation and
differentiation in monolayer and 3D culture. Biomaterials 2005;26(23):
4847e55.
[55] Marie PJ. The calcium-sensing receptor in bone cells: a potential therapeutic
target in osteoporosis. Bone 2010;46(3):571e6.
[56] Valerio P, Pereira MM, Goes AM, Leite MF. Effects of extracellular calcium
concentration on the glutamate release by bioactive glass (BG60S) preincubated osteoblasts. Biomed Mater 2009;4:045011.
[57] Julien M, Khoshniat S, Lacreusette A, Gatius M, Bozec A, Wagner EF, et al.
Phosphate-dependent regulation of MGP in osteoblasts: role of ERK1/2 and
Fra-1. J Bone Miner Res 2009;24(11):1856e68.
[58] Carlisle E. Silicon: a requirement in bone formation independent of vitamin
D1. Calcif Tissue Int 1981;33(1):27e34.

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774


[59] Carlisle EM. Silicon: a possible factor in bone calcication. Science 1970;167
(3916):279e80.
[60] Reftt DM, Ogston N, Jugdaohsingh R, Cheung HFJ, Evans BAJ, Thompson RPH,
et al. Orthosilicic acid stimulates collagen type 1 synthesis and osteoblastic
differentiation in human osteoblast-like cells in vitro. Bone 2003;32(2):
127e35.
[61] Delannoy P, Bazot D, Marie PJ. Long-term treatment with strontium ranelate
increases vertebral bone mass without deleterious effect in mice. Metabolism 2002;51(7):906e11.
[62] Grynpas MD, Marie PJ. Effects of low doses of strontium on bone quality and
quantity in rats. Bone 1990;11(5):313e9.
[63] Shahnazari M, Sharkey NA, Fosmire GJ, Leach RM. Effects of strontium on
bone strength, density, volume, and microarchitecture in laying hens. J Bone
Miner Res 2006;21(11):1696e703.
[64] Verberckmoes SC, De Broe ME, DHaese PC. Dose-dependent effects of
strontium on osteoblast function and mineralization. Kidney Int 2003;64
(2):534e43.
[65] Marie PJ. Strontium ranelate: a physiological approach for optimizing bone
formation and resorption. Bone 2006;38(2, Suppl. 1):10e4.
[66] Brando-Neto J, Stefan V, Mendona BB, Bloise W, Castro AVB. The essential
role of zinc in growth. Nutr Res 1995;15(3):335e58.
[67] Choi MK, Kim MH, Kang MH. The effect of boron supplementation on bone
strength in ovariectomized rats fed with diets containing different calcium
levels. Food Sci Biotechnol 2005;14(2):242e8.
[68] Uysal T, Ustdal A, Sonmez MF, Ozturk F. Stimulation of bone formation by
dietary boron in an orthopedically expanded suture in rabbits. Angle Orthod
2009;79(5):984e90.
[69] Barrio DA, Etcheverry SB. Vanadium and bone development: putative
signaling pathways. Can J Physiol Pharmacol 2006;84(7):677e86.
[70] Buttyan R, Chichester P, Stisser B, Matsumoto S, Ghafar MA, Levin RM. Acute
intravesical infusion of a cobalt solution stimulates a hypoxia response,
growth and angiogenesis in the rat bladder. J Urol 2003;169(6):2402e6.
[71] Patntirapong S, Habibovic P, Hauschka PV. Effects of soluble cobalt and cobalt
incorporated into calcium phosphate layers on osteoclast differentiation and
activation. Biomaterials 2009;30(4):548e55.
[72] Peters K, Schmidt H, Unger RE, Otto M, Kamp G, Kirkpatrick CJ. Softwaresupported image quantication of angiogenesis in an in vitro culture system:
application to studies of biocompatibility. Biomaterials 2002;23(16):3413e9.
[73] Tanaka T, Kojima I, Ohse T, Ingelnger JR, Adler S, Fujita T, et al. Cobalt
promotes angiogenesis via hypoxia-inducible factor and protects tubulointerstitium in the remnant kidney model. Lab Invest 2005;85(10):
1292e307.
[74] Hartwig A. Role of magnesium in genomic stability. Mutat Res Fundam Mol
Mech Mutagen 2001;475(1e2):113e21.
[75] Rude RK, Gruber HE, Norton HJ, Wei LY, Frausto A, Kilburn J. Dietary
magnesium reduction to 25% of nutrient requirement disrupts bone and
mineral metabolism in the rat. Bone 2005;37(2):211e9.
[76] Rude RK, Gruber HE, Norton HJ, Wei LY, Frausto A, Mills BG. Bone loss
induced by dietary magnesium reduction to 10% of the nutrient requirement
in rats is associated with increased release of substance P and tumor necrosis
factor-alpha. J Nutr 2004;134(1):79e85.
[77] Rude RK, Gruber HE, Wei LY, Frausto A, Mills BG. Magnesium deciency:
effect on bone and mineral metabolism in the mouse. Calcif Tissue Int
2003;72(1):32e41.
[78] Staiger MP, Pietak AM, Huadmai J, Dias G. Magnesium and its alloys as
orthopedic biomaterials: a review. Biomaterials 2006;27(9):1728e34.
[79] Tsuboi S, Nakagaki H, Ishiguro K, Kondo K, Mukai M, Robinson C, et al.
Magnesium distribution in human bone. Calcif Tissue Int 1994;54(1):34e7.
[80] Zreiqat H, Howlett CR, Zannettino A, Evans P, Schulze-Tanzil G, Knabe C, et al.
Mechanisms of magnesium-stimulated adhesion of osteoblastic cells to
commonly used orthopaedic implants. J Biomed Mater Res 2002;62(2):175e84.
[81] Sun ZL, Wataha JC, Hanks CT. Effects of metal ions on osteoblast-like cell
metabolism and differentiation. J Biomed Mater Res 1997;34(1):29e37.
[82] Mourino V, Boccaccini AR. Bone tissue engineering therapeutics: controlled
drug delivery in three-dimensional scaffolds. J Royal Soc Interface 2010;7
(43):209e27.
[83] Damen JJM, Ten Cate JM. Silica-induced precipitation of calcium phosphate
in the presence of inhibitors of hydroxyapatite formation. J Dent Res 1992;71
(3):453e7.
[84] Jugdaohsingh R, Tucker KL, Qiao N, Cupples LA, Kiel DP, Powell JJ. Dietary
silicon intake is positively associated with bone mineral density in men and
premenopausal women of the framingham offspring cohort. J Bone Miner
Res 2004;19(2):297e307.
[85] Kim M-H, Bae Y-J, Choi M-K, Chung Y-S. Silicon supplementation improves
the bone mineral density of calcium-decient ovariectomized rats by
reducing bone resorption. Biol Trace Elem Res 2009;128(3):239e47.
[86] Nielsen FH, Poellot R. Dietary silicon affects bone turnover differently in
ovariectomized and sham-operated growing rats. J Trace Elem Exp Med
2004;17(3):137e49.
[87] Boivin G, Deloffre P, Perrat B, Panczer G, Boudeulle M, Mauras Y, et al.
Strontium distribution and interactions with bone mineral in monkey iliac
bone after strontium salt (S 12911) administration. J Bone Miner Res 1996;11
(9):1302e11.
[88] Meunier PJ, Slosman DO, Delmas PD, Sebert JL, Brandi ML, Albanese C, et al.
Strontium ranelate: dose-dependent effects in established postmenopausal

[89]

[90]

[91]
[92]

[93]

[94]
[95]
[96]

[97]
[98]

[99]

[100]

[101]

[102]
[103]

[104]

[105]

[106]
[107]
[108]

[109]

[110]

[111]

[112]

[113]

[114]

[115]

[116]

2771

vertebral osteoporosiseA 2-year randomized placebo controlled trial. J Clin


Endocrinol Metab 2002;87(5):2060e6.
Habermann B, Kafchitsas K, Olender G, Augat P, Kurth A. Strontium ranelate
enhances callus strength more than PTH 1-34 in an osteoporotic rat model of
fracture healing. Calcif Tissue Int 2010;86(1):82e9.
Lang C, Murgia C, Leong M, Tan L-W, Perozzi G, Knight D, et al. Antiinammatory effects of zinc and alterations in zinc transporter mRNA in
mouse models of allergic inammation. Am J Physiol Lung Cell Mol Physiol
2007;292(2):L577e84.
Cousins RJ. A role of zinc in the regulation of gene expression. Proc Nutr Soc
1998;57(02):307e11.
Kwun I-S, Cho Y-E, Lomeda R-AR, Shin H-I, Choi J-Y, Kang Y-H, et al. Zinc deciency suppresses matrix mineralization and retards osteogenesis transiently
with catch-up possibly through Runx 2 modulation. Bone 2010;46(3):732e41.
Finney L, Vogt S, Fukai T, Glesne D. Copper and angiogenesis: unravelling
a relationship key to cancer progression. Clin Exp Pharmacol Physiol 2009;36
(1):88e94.
Grard C, Bordeleau L-J, Barralet J, Doillon CJ. The stimulation of angiogenesis
and collagen deposition by copper. Biomaterials 2010;31(5):824e31.
Hu G-f. Copper stimulates proliferation of human endothelial cells under
culture. J Cell Biochem 1998;69(3):326e35.
Rodrguez JP, Ros S, Gonzlez M. Modulation of the proliferation and
differentiation of human mesenchymal stem cells by copper. J Cell Biochem
2002;85(1):92e100.
Zhang JC, Huang JA, Xu SJ, Wang K, Yu SF. Effects of Cu2 and pH on osteoclastic bone resorption in vitro. Prog Nat Sci 2003;13(4):266.
Cashman KD, Baker A, Ginty F, Flynn A, Strain JJ, Bonham MP, et al. No effect
of copper supplementation on biochemical markers of bone metabolism in
healthy young adult females despite apparently improved copper status. Eur
J Clin Nutr 2001;55(7):525e31.
Lai YL, Yamaguchi M. Effects of copper on bone component in the femoral
tissues of rats: anabolic effect of zinc is weakened by copper. Biol Pharm Bull
2005;28(12):2296e301.
Smith BJ, King JB, Lucas EA, Akhter MP, Arjmandi BH, Stoecker BJ. Skeletal
unloading and dietary copper depletion are detrimental to bone quality of
mature rats. J Nutr 2002;132(2):190e6.
Yamasaki Y, Yoshida Y, Okazaki M, Shimazu A, Uchida T, Kubo T, et al.
Synthesis of functionally graded MgCO3 apatite accelerating osteoblast
adhesion. J Biomed Mater Res 2002;62(1):99e105.
Nielsen FH. Is boron nutritionally relevant? Nutr Rev 2008;66(4):183.
Dzondo-Gadet M, Mayap-Nzietchueng R, Hess K, Nabet P, Belleville F,
Dousset B. Action of boron at the molecular level. Biol Trace Elem Res
2002;85(1):23e33.
Chapin RE, Ku WW, Kenney MA, McCoy H, Gladen B, Wine RN, et al. The
effects of dietary boron on bone strength in rats. Fundam Appl Toxicol
1997;35(2):205e15.
Gorustovich AA, Steimetz T, Nielsen FH, Guglielmotti MB. A histomorphometric
study of alveolar bone modelling and remodelling in mice fed a boron-decient
diet. Arch Oral Biol 2008;53(7):677e82.
Hench LL, Splinter RJ, Allen WC, Greenlee TK. Bonding mechanism at the
interface of ceramic prosthetic materials. J Biomed Mater Res 1972;2:117e41.
Hench LL, Polak JM. Third-generation biomedical materials. Science
2002;295(5557):1014e7.
Silver IA, Deas J, Erecinska M. Interactions of bioactive glasses with osteoblasts in vitro: effects of 45S5 Bioglass (R), and 58S and 77S bioactive glasses
on metabolism, intracellular ion concentrations and cell viability. Biomaterials 2001;22(2):175e85.
Laquerriere P, Jallot E, Kilian L, Benhayoune H, Balossier G. Effects of bioactive glass particles and their ionic products on intracellular concentrations.
J Biomed Mater Res A 2003;65A(4):441e6.
Alcaide M, Portols P, Lpez-Noriega A, Arcos D, Vallet-Reg M, Portols MT.
Interaction of an ordered mesoporous bioactive glass with osteoblasts,
broblasts and lymphocytes, demonstrating its biocompatibility as a potential bone graft material. Acta Biomater 2010;6(3):892e9.
Bielby RC, Christodoulou IS, Pryce RS, Radford WJP, Hench LL, Polak JM.
Time- and concentration-dependent effects of dissolution products of 58S
solegel bioactive glass on proliferation and differentiation of murine and
human osteoblasts. Tissue Eng 2004;10(7e8):1018e26.
Bielby RC, Pryce RS, Hench LL, Polak JM. Enhanced derivation of osteogenic
cells from murine embryonic stem cells after treatment with ionic dissolution
products of 58S bioactive solegel glass. Tissue Eng 2005;11(3e4):479e88.
Bosetti M, Zanardi L, Hench L, Cannas M. Type I collagen production by
osteoblast-like cells cultured in contact with different bioactive glasses.
J Biomed Mater Res A 2003;64A(1):189e95.
Chen QZ, Efthymiou A, Salih V, Boccaccini AR. Bioglass-derived glassceramic scaffolds: study of cell proliferation and scaffold degradation in
vitro. J Biomed Mater Res A 2008;84A(4):1049e60.
Christodoulou I, Buttery LDK, Saravanapavan P, Tai G, Hench LL, Polak JM.
Dose- and time-dependent effect of bioactive gel-glass ionic-dissolution
products on human fetal osteoblast-specic gene expression. J Biomed Mater
Res B Appl Biomater 2005;74B(1):529e37.
Christodoulou I, Buttery LDK, Tai G, Hench LL, Polak JM. Characterization of
human fetal osteoblasts by microarray analysis following stimulation with
58S bioactive gel-glass ionic dissolution products. J Biomed Mater Res B Appl
Biomater 2006;77B(2):431e46.

2772

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

[117] Clupper DC, Gough JE, Hall MM, Clare AG, LaCourse WC, Hench LL. In vitro
bioactivity of S520 glass bers and initial assessment of osteoblast attachment. J Biomed Mater Res A 2003;67A(1):285e94.
[118] Deb S, Mandegaran R, Di Silvio L. A porous scaffold for bone tissue engineering/45S5 Bioglass(A (R)) derived porous scaffolds for co-culturing osteoblasts and endothelial cells. J Mater Sci Mater Med 2010;21(3):893e905.
[119] Gorustovich AA. Imaging resin-cast osteocyte lacuno-canalicular system at
bone-bioactive glass interface by scanning electron microscopy. Microsc
Microanal 2010;16(02):132e6.
[120] Gough JE, Jones JR, Hench LL. Nodule formation and mineralisation of human
primary osteoblasts cultured on a porous bioactive glass scaffold. Biomaterials 2004;25(11):2039e46.
[121] Grootveld M, Silwood CJL, Winter WT. High-resolution H NMR investigations
of the capacity of dentifrices containing a ldquosmartrdquo bioactive glass to
inuence the metabolic prole of and deliver calcium ions to human saliva.
J Biomed Mater Res B Appl Biomater 2009;91B(1):88e101.
[122] Haimi S, Moimas L, Pirhonen E, Lindroos B, Huhtala H, Rty S, et al. Calcium
phosphate surface treatment of bioactive glass causes a delay in early
osteogenic differentiation of adipose stem cells. J Biomed Mater Res A 2009;
91A(2):540e7.
[123] Jell G, Notingher I, Tsigkou O, Notingher P, Polak JM, Hench LL, et al. Bioactive
glass-induced osteoblast differentiation: a noninvasive spectroscopic study.
J Biomed Mater Res A 2008;86A(1):31e40.
[124] Jell G, Stevens M. Gene activation by bioactive glasses. J Mater Sci Mater Med
2006;17(11):997e1002.
[125] Jones JR, Tsigkou O, Coates EE, Stevens MM, Polak JM, Hench LL. Extracellular
matrix formation and mineralization on a phosphate-free porous bioactive
glass scaffold using primary human osteoblast (HOB) cells. Biomaterials
2007;28(9):1653e63.
[126] Kaufmann E, Ducheyne P, Shapiro IM. Evaluation of osteoblast response to
porous bioactive glass (45S5) substrates by RT-PCR analysis. Tissue Eng
2000;6(1):19e28.
[127] Meretoja VV, Malin M, Seppl JV, Nrhi TO. Osteoblast response to
continuous phase macroporous scaffolds under static and dynamic culture
conditions. J Biomed Mater Res A 2009;89A(2):317e25.
[128] Reilly GC, Radin S, Chen AT, Ducheyne P. Differential alkaline phosphatase
responses of rat and human bone marrow derived mesenchymal stem cells
to 45S5 bioactive glass. Biomaterials 2007;28(28):4091e7.
[129] Tsigkou O, Jones JR, Polak JM, Stevens MM. Differentiation of fetal osteoblasts and
formation of mineralized bone nodules by 45S5 Bioglass (R) conditioned medium
in the absence of osteogenic supplements. Biomaterials 2009;30(21):3542e50.
[130] Valerio P, Goes AM, Pereira MM, Leite MF. Cytological and biochemical
evaluation of osteoblast in contact with ionic products of bioactive ceramics.
In: BenNissan B, Sher D, Walsh W, editors. Bioceramics, vol. 15. ZurichUetikon: Trans Tech Publications Ltd; 2003. p. 703e6.
[131] Valerio P, Pereira MM, Goes AM, Leite MF. The effect of ionic products from
bioactive glass dissolution on osteoblast proliferation and collagen production. Biomaterials 2004;25(15):2941e8.
[132] Vlimki V-V, Yrjans JJ, Vuorio EI, Aro HT. Molecular biological evaluation of
bioactive glass microspheres and adjunct bone morphogenetic protein 2
gene transfer in the enhancement of new bone formation. Tissue Eng
2005;11(3e4):387e94.
[133] Varanasi VG, Saiz E, Loomer PM, Ancheta B, Uritani N, Ho SP, et al. Enhanced
osteocalcin expression by osteoblast-like cells (MC3T3-E1) exposed to
bioactive coating glass (SiO2eCaOeP2O5eMgOeK2OeNa2O system) ions.
Acta Biomater 2009;5(9):3536e47.
[134] Xynos ID, Edgar AJ, Buttery LDK, Hench LL, Polak JM. Ionic products of
bioactive glass dissolution increase proliferation of human osteoblasts and
induce insulin-like growth factor II mRNA expression and protein synthesis.
Biochem Biophys Res Commun 2000;276(2):461e5.
[135] Xynos ID, Hukkanen MVJ, Batten JJ, Buttery LD, Hench LL, Polak JM. Bioglass
45S5 stimulates osteoblast turnover and enhances bone formation in vitro:
implications and applications for bone tissue engineering. Calcif Tissue Int
2000;67(4):321e9.
[136] Bosetti M, Cannas M. The effect of bioactive glasses on bone marrow stromal
cells differentiation. Biomaterials 2005;26(18):3873e9.
[137] Santos MI, Reis RL. Vascularization in bone tissue engineering: physiology,
current strategies, major hurdles and future challenges. Macromol Biosci
2010;10(1):12e27.
[138] Brandi ML, Collin-Osdoby P. Vascular biology and the skeleton. J Bone Miner
Res 2006;21(2):183e92.
[139] Leu A, Stieger SM, Dayton P, Ferrara KW, Leach JK. Angiogenic response to
bioactive glass promotes bone healing in an irradiated calvarial defect. Tissue
Eng Part A 2009;15(4):877e85.
[140] Day RM, Boccaccini AR, Shurey S, Roether JA, Forbes A, Hench LL, et al.
Assessment of polyglycolic acid mesh and bioactive glass for soft-tissue
engineering scaffolds. Biomaterials 2004;25(27):5857e66.
[141] Hench LL. Genetic design of bioactive glass. J Eur Ceram Soc 2009;29(7):
1257e65.
[142] Chung C-H, Golub EE, Forbes E, Tokuoka T, Shapiro IM. Mechanism of action
of beta-glycerophosphate on bone cell mineralization. Calcif Tissue Int
1992;51(4):305e11.
[143] Beck GR, Moran E, Knecht N. Inorganic phosphate regulates multiple genes
during osteoblast differentiation, including Nrf2. Exp Cell Res 2003;288
(2):288e300.

[144] Wittrant Y, Bourgine A, Khoshniat S, Alliot-Licht B, Masson M, Gatius M, et al.


Inorganic phosphate regulates Glvr-1 and -2 expression: role of calcium and
ERK1/2. Biochem Biophys Res Commun 2009;381(2):259e63.
[145] Azevedo MM, Jell G, ODonnell MD, Law RV, Hill RG, Stevens MM. Synthesis
and characterization of hypoxia-mimicking bioactive glasses for skeletal
regeneration. J Mater Chem 2010;20(40):8854e64.
[146] Aina V, Malavasi G, Pla AF, Munaron L, Morterra C. Zinc-containing bioactive
glasses: surface reactivity and behaviour towards endothelial cells. Acta
Biomater 2009;5(4):1211e22.
[147] Aina V, Perardi A, Bergandi L, Malavasi G, Menabue L, Morterra C, et al.
Cytotoxicity of zinc-containing bioactive glasses in contact with human
osteoblasts. Chem Biol Interact 2007;167(3):207e18.
[148] Balamurugan A, Balossier G, Kannan S, Michel J, Rebelo AHS, Ferreira JMF.
Development and in vitro characterization of sol-gel derived CaOeP2O5e
SiO2eZnO bioglass. Acta Biomater 2007;3(2):255e62.
[149] Bini M, Grandi S, Capsoni D, Mustarelli P, Saino E, Visai L. SiO2P2O5CaO glasses
and glass-ceramics with and without ZnO: relationships among composition,
microstructure, and bioactivity. J Phys Chem C 2009;113(20):8821e8.
[150] Boyd D, Carroll G, Towler MR, Freeman C, Farthing P, Brook IM. Preliminary
investigation of novel bone graft substitutes based on strontium-calciumzinc-silicate glasses. J Mater Sci Mater Med 2009;20(1):413e20.
[151] Courthe
coux L, Lao J, Nedelec JM, Jallot E. Controlled bioactivity in zincdoped solgel-derived binary bioactive glasses. J Phys Chem C 2008;112
(35):13663e7.
[152] Du RL, Chang J. Preparation and characterization of Zn and Mg doped
bioactive glasses. J Inorg Mater 2004;19(6):1353e8.
[153] Du RL, Chang J, Ni SY, Zhai WY, Wang JY. Characterization and in vitro
bioactivity of Zinc-containing bioactive glass and glass-ceramics. J Biomater
Appl 2006;20(4):341e60.
[154] Haimi S, Gorianc G, Moimas L, Lindroos B, Huhtala H, Rty S, et al. Characterization of zinc-releasing three-dimensional bioactive glass scaffolds and
their effect on human adipose stem cell proliferation and osteogenic
differentiation. Acta Biomater 2009;5(8):3122e31.
[155] Lusvardi G, Malavasi G, Menabue L, Menziani MC, Pedone A, Segre U, et al.
Properties of zinc releasing surfaces for clinical applications. J Biomater Appl
2008;22(6):505e26.
[156] Murphy S, Boyd D, Moane S, Bennett M. The effect of composition on ion
release from CaeSreNaeZneSi glass bone grafts. J Mater Sci Mater Med
2009;20(11):2207e14.
[157] Oki A, Parveen B, Hossain S, Adeniji S, Donahue H. Preparation and in vitro
bioactivity of zinc containing sol-gel-derived bioglass materials. J Biomed
Mater Res A 2004;69A(2):216e21.
[158] Singh RK, Srinivasan A. Bioactivity of SiO2eCaOeP2O5eNa2O glasses containing zinc-iron oxide. Appl Surf Sci 2010;256(6):1725e30.
[159] Varmette EA, Nowalk JR, Flick LM, Hall MM. Abrogation of the inammatory
response in LPS-stimulated RAW 264.7 murine macrophages by Zn- and Cudoped bioactive sol-gel glasses. J Biomed Mater Res A 2009;90A(2):317e25.
[160] Zreiqat H, Ramaswamy Y, Wu C, Paschalidis A, Lu Z, James B, et al. The
incorporation of strontium and zinc into a calcium-silicon ceramic for bone
tissue engineering. Biomaterials 2010;31(12):3175e84.
[161] Erol M, Ozyuguran A, Celebican O. Synthesis, characterization, and in vitro
bioactivity of sol-gel-derived Zn, Mg, and Zn-Mg co-doped bioactive glasses.
Chem Eng Technol 2010;33(7):1066e74.
[162] Li X, Wang X, He D, Shi J. Synthesis and characterization of mesoporous
CaOeMOeSiO2eP2O5 (M Mg, Zn, Cu) bioactive glasses/composites. J Mater
Chem 2008;18(34):4103e9.
[163] Fredholm YC, Karpukhina N, Law RV, Hill RG. Strontium containing bioactive
glasses: glass structure and physical properties. J Non Cryst Solids 2010;356
(44e49):2546e51.
[164] Gorustovich AA, Steimetz T, Cabrini RL, Lpez JMP. Osteoconductivity of
strontium-doped bioactive glass particles: a histomorphometric study in
rats. J Biomed Mater Res A 2010;92A(1):232e7.
[165] Hesaraki S, Alizadeh M, Nazarian H, Shari D. Physico-chemical and in vitro
biological evaluation of strontium/calcium silicophosphate glass. J Mater Sci
Mater Med 2010;21(2):695e705.
[166] Lao J, Jallot E, Nedelec J- M. Strontium-delivering glasses with enhanced
bioactivity: a new biomaterial for antiosteoporotic applications? Chem
Mater 2008;20(15):4969e73.
[167] Lao J, Nedelec JM, Jallot E. New strontium-based bioactive glasses: physicochemical reactivity and delivering capability of biologically active dissolution products. J Mater Chem 2009;19(19):2940e9.
[168] ODonnell MD, Candarlioglu PL, Miller CA, Gentleman E, Stevens MM.
Materials characterisation and cytotoxic assessment of strontium-substituted bioactive glasses for bone regeneration. J Mater Chem 2010;20(40):
8934e41.
[169] Pan HB, Zhao XL, Zhang X, Zhang KB, Li LC, Li ZY, et al. Strontium borate
glass: potential biomaterial for bone regeneration. J Royal Soc Interface
2010;7(48):1025e31.
[170] Murphy S, Wren A, Towler M, Boyd D. The effect of ionic dissolution products
of CaeSreNaeZneSi bioactive glass on in vitro cytocompatibility. J Mater Sci
Mater Med 2010;21(10):2827e34.
[171] Chen X, Liao X, Huang Z, You P, Chen C, Kang Y, et al. Synthesis and characterization of novel multiphase bioactive glass-ceramics in the
CaOeMgOeSiO2 system. J Biomed Mater Res B Appl Biomater 2010;93B(1):
194e202.

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774


[172] Dietrich E, Oudadesse H, Lucas-Girot A, Mami M. In vitro bioactivity of meltderived glass 46S6 doped with magnesium. J Biomed Mater Res A 2009;88A
(4):1087e96.
[173] Saboori A, Rabiee M, Moztarzadeh F, Sheikhi M, Tahriri M, Karimi M.
Synthesis, characterization and in vitro bioactivity of sol-gel-derived
SiO2eCaOeP2O5eMgO bioglass. Mater Sci Eng C 2009;29(1):335e40.
[174] Soulie J, Nedelec JM, Jallot E. Inuence of Mg doping on the early steps of
physico-chemical reactivity of sol-gel derived bioactive glasses in biological
medium. Phys Chem Chem Phys 2009;11(44):10473e83.
[175] Watts SJ, Hill RG, ODonnell MD, Law RV. Inuence of magnesia on the structure
and properties of bioactive glasses. J Non Cryst Solids 2010;356(9e10):517e24.
[176] Bellantone M, Williams HD, Hench LL. Broad-spectrum bactericidal activity
of Ag2O-doped bioactive glass. Antimicrob Agents Chemother 2002;46
(6):1940e5.
[177] Blaker JJ, Nazhat SN, Boccaccini AR. Development and characterisation of
silver-doped bioactive glasscoated sutures for tissue engineering and wound
healing applications. Biomaterials 2004;25(7e8):1319e29.
[178] Delben JRJ, Pimentel OM, Coelho MB, Candelorio PD, Furini LN, dos Santos FA,
et al. Synthesis and thermal properties of nanoparticles of bioactive glasses
containing silver. J Therm Anal Calorim 2009;97(2):433e6.
[179] Kawashita M, Tsuneyama S, Miyaji F, Kokubo T, Kozuka H, Yamamoto K.
Antibacterial silver-containing silica glass prepared by sol-gel method.
Biomaterials 2000;21(4):393e8.
[180] Lohbauer U, Jell G, Saravanapavan P, Jones JR, Hench LL. Indirect cytotoxicity
evaluation of silver doped bioglass Ag-S70C30 on human primary keratinocytes. In: Li P, Zhang K, Colwell CW, editors. Bioceramics, 17. ZurichUetikon: Trans Tech Publications Ltd; 2005. p. 431e4.
[181] Vern E, Miola M, Vitale Brovarone C, Cannas M, Gatti S, Fucale G, et al.
Surface silver-doping of biocompatible glass to induce antibacterial properties. Part I: massive glass. J Mater Sci Mater Med 2009;20(3):733e40.
[182] Vern E, Nunzio SD, Bosetti M, Appendino P, Vitale Brovarone C, Maina G,
et al. Surface characterization of silver-doped bioactive glass. Biomaterials
2005;26(25):5111e9.
[183] Vitale-Brovarone C, Miola M, Balagna C, Vern E. 3D-glass-ceramic scaffolds
with antibacterial properties for bone grafting. Chem Eng J 2008;137(1):
129e36.
[184] Di Nunzio S, Vitale Brovarone C, Spriano S, Milanese D, Vern E, Bergo V,
et al. Silver containing bioactive glasses prepared by molten salt ionexchange. J Eur Ceram Soc 2004;24(10e11):2935e42.
[185] Leonelli C, Lusvardi G, Malavasi G, Menabue L, Tonelli M. Synthesis and
characterization of cerium-doped glasses and in vitro evaluation of bioactivity. J Non Cryst Solids 2003;316(2e3):198e216.
[186] Agathopoulos S, Tulyaganov DU, Ventura JMG, Kannan S, Karakassides MA,
Ferreira JMF. Formation of hydroxyapatite onto glasses of the
CaOeMgOeSiO2 system with B2O3, Na2O, CaF2 and P2O5 additives. Biomaterials 2006;27(9):1832e40.
[187] Brown RF, Rahaman MN, Dwilewicz AB, Huang W, Day DE, Li Y, et al. Effect of
borate glass composition on its conversion to hydroxyapatite and on the
proliferation of MC3T3-E1 cells. J Biomed Mater Res A 2009;88A(2):392e400.
[188] Fu H, Fu Q, Zhou N, Huang W, Rahaman MN, Wang D, et al. In vitro evaluation of borate-based bioactive glass scaffolds prepared by a polymer foam
replication method. Mater Sci Eng C 2009;29(7):2275e81.
[189] Gorustovich A, Guglielmotti MB, Lopez JMP, Cabrini RL. Increased osteogenesis elicited by boron-modied bioactive glass particles in the
SiO2eCaOeP2O5eNa2O system: a histomorphometric study in rats. In: Li P,
Zhang K, Colwell CW, editors. Bioceramics, 17. Zurich-Uetikon: Trans Tech
Publications Ltd; 2005. p. 913e6.
[190] Gorustovich A, Steitnetz T, Lopez JP, Guglielmotti M, Cabrini R. Osteogenic
response to bioactive glass particles modied by boron. Bone 2006;38(5):1.
[191] Gorustovich AA, Lopez JMP, Guglielmotti MB, Cabrini RL. Biological performance of boron-modied bioactive glass particles implanted in rat tibia bone
marrow. Biomed Mater 2006;1(3):100e5.
[192] Liang W, Rahaman MN, Day DE, Marion NW, Riley GC, Mao JJ. Bioactive
borate glass scaffold for bone tissue engineering. J Non Cryst Solids 2008;354
(15e16):1690e6.
[193] Marion NW, Liang W, Reilly GC, Day DE, Rahaman MN, Mao JJ. Borate glass
supports the in vitro osteogenic differentiation of human mesenchymal stem
cells. Mech Adv Mater Struct 2005;12(3):239e46.
[194] Ryu HS, Lee JK, Seo JH, Kim H, Hong KS, Kim DJ, et al. Novel bioactive and
biodegradable glass ceramics with high mechanical strength in the
CaOeSiO2eB2O3 system. J Biomed Mater Res A 2004;68A(1):79e89.
[195] Bergandi L, Aina V, Garetto S, Malavasi G, Aldieri E, Laurenti E, et al. Fluoridecontaining bioactive glasses inhibit pentose phosphate oxidative pathway
and glucose 6-phosphate dehydrogenase activity in human osteoblasts.
Chem Biol Interact 2010;183(3):405e15.
[196] Brauer DS, Karpukhina N. ODonnell MD, Law RV, Hill RG. Fluoride-containing bioactive glasses: effect of glass design and structure on degradation,
pH and apatite formation in simulated body uid. Acta Biomater 2010;6
(8):3275e82.
[197] Kokubo T, Takadama H. How useful is SBF in predicting in vivo bone
bioactivity? Biomaterials 2006;27(15):2907e15.
[198] Vrouwenvelder WCA, Groot CG, de Groot K. Better histology and biochemistry for osteoblasts cultured on titanium-doped bioactive glass: bioglass
45S5 compared with iron-, titanium-, uorine- and boron-containing
bioactive glasses. Biomaterials 1994;15(2):97e106.

2773

[199] Jain RK, Au P, Tam J, Duda DG, Fukumura D. Engineering vascularized tissue.
Nat Biotechnol 2005;23(7):821e3.
[200] Knabe C, Stiller M, Berger G, Reif D, Gildenhaar R, Howlett CR, et al. The effect
of bioactive glass ceramics on the expression of bone-related genes and
proteins in vitro. Clin Oral Implants Res 2005;16(1):119e27.
[201] Pessan JP, Al-Ibrahim NS, Buzalaf MAR, Toumba KJ. Slow-release uoride
devices: a literature review. J Appl Oral Sci 2008;16:238e44.
[202] Hicks J, Garcia-Godoy F, Flaitz C. Biological factors in dental caries: role of
saliva and dental plaque in the dynamic process of demineralization and
remineralization (part 1). J Clin Pediatr Dent 2004;28(1):47e52.
[203] Pak CYC, Zerwekh JE, Antich P. Anabolic effects of uoride on bone. Trends
Endocrinol Metab 1995;6(7):229e34.
[204] Siddhartha S, Bera T, Roy A, Singh G, Ramachandrarao P, Dash D. Characterization of enhanced antibacterial effects of novel silver nanoparticles.
Nanotechnology 2007;18(22):225103.
[205] Lok C-N, Ho C-M, Chen R, He Q-Y, Yu W-Y, Sun H, et al. Silver nanoparticles:
partial oxidation and antibacterial activities. J Biol Inorg Chem 2007;12
(4):527e34.
[206] Yang XH, Yang WJ, Wang Q, Li HM, Wang KM, Yang L, et al. Atomic force
microscopy investigation of the characteristic effects of silver ions on
Escherichia coli and Staphylococcus epidermidis. Talanta 2010;81(4e5):
1508e12.
[207] Uo M, Mizuno M, Kuboki Y, Makishima A, Watari F. Properties and cytotoxicity of water soluble Na2OeCaOeP2O5 glasses. Biomaterials 1998;19
(24):2277e84.
[208] Peters F, Schwarz K, Epple M. The structure of bone studied with synchrotron
X-ray diffraction, X-ray absorption spectroscopy and thermal analysis.
Thermochim Acta 2000;361(1e2):131e8.
[209] Gough JE, Christian P, Scotchford CA, Rudd CD, Jones IA. Synthesis, degradation, and in vitro cell responses of sodium phosphate glasses for craniofacial bone repair. J Biomed Mater Res 2002;59(3):481e9.
[210] Salih V, Franks K, James M, Hastings GW, Knowles JC, Olsen I. Development
of soluble glasses for biomedical use part II: the biological response of
human osteoblast cell lines to phosphate-based soluble glasses. J Mater Sci
Mater Med 2000;11(10):615e20.
[211] Bitar M, Salih V, Mudera V, Knowles JC, Lewis MP. Soluble phosphate glasses:
in vitro studies using human cells of hard and soft tissue origin. Biomaterials
2004;25(12):2283e92.
[212] Skelton KL, Glenn JV, Clarke SA, Georgiou G, Valappil SP, Knowles JC, et al.
Effect of ternary phosphate-based glass compositions on osteoblast and
osteoblast-like proliferation, differentiation and death in vitro. Acta Biomater
2007;3(4):563e72.
[213] Navarro M, Ginebra M-P, Planell JA. Cellular response to calcium phosphate
glasses with controlled solubility. J Biomed Mater Res A 2003;67A(3):1009e15.
[214] Navarro M, Sd Valle, Martnez S, Zeppetelli S, Ambrosio L, Planell JA, et al.
New macroporous calcium phosphate glass ceramic for guided bone
regeneration. Biomaterials 2004;25(18):4233e41.
[215] Brauer DS, Rssel C, Li W, Habelitz S. Effect of degradation rates of resorbable
phosphate invert glasses on in vitro osteoblast proliferation. J Biomed Mater
Res A 2006;77A(2):213e9.
[216] Manupriya Thind KS, Singh K, Sharma G, Rajendran V. Inuence of addition
of Al2O3 on physical, structural, acoustical and in-vitro bioactive properties
of phosphate glasses. Physica A (Amsterdam) 2009;206(7):1447e55.
[217] Saranti A, Koutselas I, Karakassides MA. Bioactive glasses in the system
CaOeB2O3eP2O5: preparation, structural study and in vitro evaluation. J Non
Cryst Solids 2006;352(5):390e8.
[218] Shu C, Wenjuan Z, Xu G, Wei Z, Wei J, Dongmei W. Dissolution behavior and
bioactivity study of glass ceramic scaffolds in the system of
CaOeP2O5eNa2OeZnO prepared by sol-gel technique. Mater Sci Eng C 2010;
30(1):105e11.
[219] Ahmed I, Collins CA, Lewis MP, Olsen I, Knowles JC. Processing, characterisation and biocompatibility of iron-phosphate glass bres for tissue engineering. Biomaterials 2004;25(16):3223e32.
[220] Leonardi E, Ciapetti G, Baldini N, Novajra G, Vern E, Baino F, et al. Response
of human bone marrow stromal cells to a resorbable P2O5eSiO2eCaOe
MgOeNa2OeK2O phosphate glass ceramic for tissue engineering applications. Acta Biomater 2010;6(2):598e606.
[221] Abou Neel EA, Chrzanowski W, Pickup DM, ODell LA, Mordan NJ,
Newport RJ, et al. Structure and properties of strontium-doped phosphatebased glasses. J Royal Soc Interface 2009;6(34):435e46.
[222] Abou Neel EA, Ahmed I, Pratten J, Nazhat SN, Knowles JC. Characterisation of
antibacterial copper releasing degradable phosphate glass bres. Biomaterials 2005;26(15):2247e54.
[223] Abou Neel EA, Ahmed I, Blaker JJ, Bismarck A, Boccaccini AR, Lewis MP, et al.
Effect of iron on the surface, degradation and ion release properties of
phosphate-based glass bres. Acta Biomater 2005;1(5):553e63.
[224] Pickup DM, Moss RM, Qiu D, Newport RJ, Valappil SP, Knowles JC, et al.
Structural characterization by x-ray methods of novel antimicrobial gallium-doped phosphate-based glasses. J Chem Phys 2009;130(6).
[225] Mourino V, Newby P, Boccaccini AR. Preparation and characterization of
gallium releasing 3-D alginate coated 45S5 bioglass (R) based scaffolds for
bone tissue engineering. Adv Eng Mater 2010;12(7):B283e91.
[226] Hesaraki S, Nemati R, Nazarian H. Physico-chemical and in vitro biological
study of zinc-doped calcium sulfate bone substitute. J Biomed Mater Res B
Appl Biomater 2009;91B(1):37e45.

2774

A. Hoppe et al. / Biomaterials 32 (2011) 2757e2774

[227] Lu Hongxu, Kawazoe N, Tateishi T, Chen Guoping, Jin Xiaogang, Chang Jiang.
In vitro proliferation and osteogenic differentiation of human bone marrowderived mesenchymal stem cells cultured with hardystonite (Ca2ZnSi2O7)
and b-TCP ceramics. J Biomater Appl 2010;25(1):39e56.
[228] Ramaswamy Y, Wu C, Zhou H, Zreiqat H. Biological response of human bone cells
to zinc-modied Ca-Si-based ceramics. Acta Biomater 2008;4(5):1487e97.
[229] Sun JY, Li JY, Liu XY, Wei L, Wang GC, Meng FH. Proliferation and gene
expression of osteoblasts cultured in DMEM containing the ionic products of
dicalcium silicate coating. Biomed Pharmacother 2009;63(9):650e7.
[230] Zhang ML, Zhai WY, Lin KL, Pan HB, Lu W, Chang J. Synthesis, in vitro
hydroxyapatite forming ability, and cytocompatibility of strontium silicate
powders. J Biomed Mater Res B Appl Biomater 2010;93B(1):252e7.
[231] Zhou H, Wei J, Wu X, Shi J, Liu C, Jia J, et al. The bio-functional role of calcium
in mesoporous silica xerogels on the responses of osteoblasts in vitro.
J Mater Sci Mater Med 2010;21(7):2175e85.
[232] Clarkin O, Boyd D, Towler MR. Strontium-based glass polyalkenoate
cements for luting applications in the skeleton. J Biomater Appl 2010;24
(6):483e502.

[233] Seabra AB, Duran N. Nitric oxide-releasing vehicles for biomedical applications. J Mater Chem 2010;20(9):1624e37.
[234] Shin JH, Metzger SK, Schoensch MH. Synthesis of nitric oxide-releasing
silica nanoparticles. J Am Chem Soc 2007;129(15):4612e9.
[235] Shin JH, Schoensch MH. Inorganic/organic hybrid silica nanoparticles as
a nitric oxide delivery scaffold. Chem Mater 2007;20(1):239e49.
[236] Hetrick EM, Shin JH, Stasko NA, Johnson CB, Wespe DA, Holmuhamedov E,
et al. Bactericidal efcacy of nitric oxide-releasing silica nanoparticles. ACS
Nano 2008;2(2):235e46.
[237] Schneider A, Francius G, Obeid R, Schwint P, Hemmerl J, Frisch B, et al.
Polyelectrolyte multilayers with a tunable youngs modulus: inuence of
lm stiffness on cell adhesion. Langmuir 2005;22(3):1193e200.
[238] Mitrossilis D, Fouchard J, Guiroy A, Desprat N, Rodriguez N, Fabry B, et al.
Single-cell response to stiffness exhibits muscle-like behavior. Proc Natl
Acad Sci U S A 2009;106(43):18243e8.
[239] Sun J-Y, Yang Y-S, Zhong J, Greenspan DC. The effect of the ionic products of
Bioglass dissolution on human osteoblasts growth cycle in vitro. J Tissue Eng
Regen Med 2007;1(4):281e6.

Вам также может понравиться