Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Acta Biomaterialia 44 (2016) 97–109

Contents lists available at ScienceDirect

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

Full length article

Novel bioceramic-reinforced hydrogel for alveolar bone regeneration


Giorgio Iviglia a,b,⇑, Clara Cassinelli a, Elisa Torre a, Francesco Baino b, Marco Morra a,
Chiara Vitale-Brovarone b
a
Nobil Bio Ricerche Srl, 14037 Portacomaro, Italy
b
Department of Applied Science and Technology, Institute of Materials Physics and Engineering, Politecnico di Torino, 10121 Torino, Italy

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

Article history: The osseointegration of dental implants and their consequent long-term success is guaranteed by the
Received 22 February 2016 presence, in the extraction site, of healthy and sufficient alveolar bone. Bone deficiencies may be the
Received in revised form 25 July 2016 result of extraction traumas, periodontal disease and infection. In these cases, placement of titanium
Accepted 10 August 2016
implants is contraindicated until a vertical bone augmentation is obtained. This goal is achieved using
Available online 10 August 2016
bone graft materials, which should simulate extracellular matrix (ECM), in order to promote osteoblast
proliferation and fill the void, maintaining the space without collapsing until the new bone is formed.
Keywords:
In this work, we design, develop and characterize a novel, moldable chitosan-pectin hydrogel reinforced
Composite
Calcium phosphate
by biphasic calcium phosphate particles with size in the range of 100–300 lm. The polysaccharide nature
Hydrogel of the hydrogel mimics the ECM of natural bone, and the ceramic particles promote high osteoblast pro-
Scaffold liferation, assessed by Scanning Electron Microscopy analysis. Swelling properties allow significant
Alveolar bone regeneration adsorption of water solution (up to 200% of solution content) so that the bone defect space can be filled
by the material in an in vivo scenario. The incorporation of ceramic particles makes the material stable at
different pH and increases the compressive elastic modulus, toughness and ultimate tensile strength.
Furthermore, cell studies with SAOS-2 human osteoblastic cell line show high cell proliferation and adhe-
sion already after 72 h, and the presence of ceramic particles increases the expression of alkaline phos-
phatase activity after 1 week. These results suggest a great potential of the developed moldable
biomaterials for the regeneration of the alveolar bone.

Statement of Significance

The positive fate of a surgical procedure involving the insertion of a titanium screw still depends on the
quality and quantity of alveolar bone which is present in the extraction site. Available materials are basi-
cally hard scaffold materials with un-predictable behavior in different condition and difficult shaping
properties. In this work we developed a novel pectin-chitosan hydrogel reinforced with ceramic particles.
Polysaccharides simulate the extracellular matrix of natural bone and the extensive in vitro cells culture
study allows to assess that the incorporation of the ceramic particles promote a pro-osteogenic response.
Shape control, easy adaption of the extraction site, predictable behavior in different environment condi-
tion, swelling properties and an anti-inflammatory response are the significant characteristics of the
developed biomaterial.
Ó 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction reported that 69% of adults aged 35–44 have lost at least one of
their permanent teeth and, by age 74, almost 26% of adults have
The restoration of teeth by using titanium dental implants is lost all of their permanent teeth. More than 300,000 dental
nowadays a quite common procedure in oral surgery [1]. The implants are placed per year worldwide and, until 2020, this num-
American Association of Oral and Maxillofacial Surgeons recently ber is expected to increase [1,2]. Tooth loss is a possible conse-
quence of traumas or periodontal diseases, such as gingivitis,
⇑ Corresponding author at: Nobil Bio Ricerche srl, Via Valcastellana, 26, 14037 periodontitis or tissue decay. The rate of success of dental implants
Portacomaro (AT), Italy. is around 98%, but it should be taken into account that the positive
E-mail address: giorgio.iviglia@polito.it (G. Iviglia). fate of a surgical procedure involving the insertion of a titanium

http://dx.doi.org/10.1016/j.actbio.2016.08.012
1742-7061/Ó 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
98 G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109

screw still depends on the quality and quantity of alveolar bone ECM [21,27]. In order to stabilize the structure, we coupled chi-
which is present in the extraction site. In order to get a successful tosan with a natural polyanionic polysaccharide, pectin, which is
implant insertion, the alveolar ridge should have a minimum a major component of cell walls of citrus or apple peel by-
dimension, which is of 5 mm of width in the maxilla and of products. Pectin consists in a poly(D-galacturonic acid) chain, with
10 mm of bone height [3]. If these dimensions are not available, a partially methoxylated carboxyl group [28]. We exploited the
an augmentation strategy will be necessary using grafting proce- ionic interaction that occurs between chitosan and pectin [29–
dures. A huge alveolar bone loss of around 2 mm in vertical and 31] to develop a novel hydrogel mimicking an ECM-like environ-
almost 50% in horizontal occurs in the first 6 months after surgery; ment for osteoblast cells, while biphasic calcium phosphate parti-
a continuous bone resorption occurs if no treatment is provided cles were added to mimic the inorganic phase of bone. Here, the
during the next year after the extraction procedure, and an average physico-chemical, mechanical and in vitro biological properties of
of 60% of bone loss could be reached in 3 years [4–9]. this new HA/bTCP-reinforced hydrogel were systematically inves-
These data show the importance of implementing a strategy to tigated and a comparison with the base hydrogel was reported.
augment the alveolar bone volume in order to provide a stable sup-
port for the implant and the future dental crown. The common use 2. Materials and methods
of autografts or allografts is affected by the morbidity and the risk
of disease transmission associated with the donor site, thus the use 2.1. Materials
of man-made bone graft biomaterials is more and more attractive
[10–12]. A bone graft material should be biocompatible, degrad- b-Tricalcium phosphate (bTCP), chitosan with medium molecu-
able, osteoconductive, osteoinductive and should mimic the ECM lar weight from crab shell (Mw = 400 kDa), pectin from citrus peel
of bone, in order to allow cell infiltration, proliferation and new and all other chemicals were purchased from Sigma-Aldrich. HA
bone formation. Hydroxyapatite (HA) and b-tricalcium phosphate particles (average size 5 lm) were purchased from Fluidinova.
(bTCP) are well known ceramic materials, widely used in bone tis-
sue engineering as bone grafts in the form of particles or three-
2.2. Preparation of calcium phosphate particles
dimensional scaffolds [13–15]. HA (Ca10(PO4)6(OH)2) is the most
abundant component in native bone (around 65% of the inorganic
HA and b-TCP were mixed in a percentage of 25 wt.% and 75 wt.
phase) but, despite many studies showing osteoconduction stimu-
% respectively, on the basis of a previous study by Morra et al. [15].
lation and good mechanical properties, the slow degradation rate
Briefly, the preparation of the ceramic slurry involved the mixing
of HA limits its use as a bone filler only [16–18].
of the HA and b-TCP powders (31 wt.%) in ultrapure water (69%).
Combination of HA with bTCP allows the degradation properties
Guar gum was dissolved previously in ultrapure water (3 wt.% of
to be managed, since bTCP materials have a degradation rate 3–12
the water content), and used a pore forming agent. The slurry
times faster than HA, and the mechanical properties to be main-
was then desiccated in oven at 80 °C for 8 h, and then shaped in cir-
tained good [19]. The ratio between HA and bTCP is a key point
cular disks using a hydraulic press (Mignon EA/SSN) with a pres-
to obtain satisfactory mechanical strength and resorption kinetics
sure of 17.5 MPa for 10 s. The ceramic disks were then sintered
and, ultimately, to stimulate osteointegration. Animal studies
in a furnace at 1100 °C for 1 h in air (heating rate 1 °C/min) [15].
demonstrated that HA/bTCP biphasic materials in the rate of
Sintered disks were ground in particles using a grinder machine
25/75 wt.% allow new bone formation [15]. In the post-extractive
(GM200, Retsch) and the ceramic powder was sieved in order to
site, it is important to use materials which can not only fill the void,
obtain a range of particles between 100 and 300 lm. Eventually,
but also be shaped in an easy manner and be not prone to migra-
the biphasic calcium phosphate particles obtained were washed
tion, hence particles and three-dimensional rigid scaffolds are not
in ultrapure water and desiccated overnight at room temperature
the best choice.
under laminar flow.
To overcome this issue, we developed a pectin/chitosan-based
polyelectrolyte hydrogel reinforced with biphasic calcium phos-
phate particles. Combining ceramics with polymeric materials pro- 2.3. Preparation of composite hydrogel
vides – at least potentially – many advantages, in particular for
dental practice, such as shape control, optimization of adhesion We dissolved 7 wt.% of pectin powder and 4 wt.% of chitosan
between implant and surrounding bone tissue, easy adaptation of powder in acetate buffer solution to obtain a pectin/chitosan ratio
the biomaterial to the extraction site, promotion of clot formation of 20/80. The solution was stirred for 6 h, then it was centrifuged
and prevention of ceramic particles migration [16,20–23]. Few and the polyelectrolyte (PEI) precipitated was collected. PEI com-
composite products for bone regeneration are currently available plex (10 wt.%) was mixed using a Thinky Mixer (Retsch) with
on the market; perhaps the most popular is Bi-Oss CollagenÒ that HA/bTCP particles (90 wt.%). The resulting slurry was poured in a
exhibits an excellent biocompatibility but tends to resorb very circular shape polyester mold and freeze-dried with Lyo5P for
quickly, with consequent loss of the mechanical properties and 12 h, at 0.06 mbar and 56 °C in order to obtain pectin/chi-
migration of the inorganic particles into the surrounding environ- tosan_ceramic particles (PCC) composite hydrogels. For the
ment [24]. pectin_chitosan based hydrogel (PC), we poured the PEI solution
In the attempt to obtain composite biomaterials with better into a circular-shape polyester mold and freeze-dried the sample
performances, in this work we developed a new hydrogel formula- by Lyo5P for 12 h, at 0.06 mbar and 56 °C.
tion based on two natural polymers, chitosan and pectin, to be
combined with HA/bTCP particles. Chitosan is a natural polyca- 2.4. Chemical characterization
tionic material derived from chitin extracted from crustaceous
exoskeleton, composed of b–(1,4)–glucosamine and N-acetyl-D- Attenuated Total Reflectance Infrared spectroscopy (ATR-IR)
glucosamine; it has recently aroused great interest in bone tissue was recorded using a Nicolet iS 10 produced by Thermo Scientific
engineering applications due to its biocompatibility, intrinsic and equipped with a diamond crystal. Raw and processed materi-
antibacterial nature, and ability not to stimulate a foreign body als (PC and PCC) were placed on the crystal and kept in place by a
reaction and, on the contrary, to promote cell adhesion, prolifera- specific crimping tool. Experimental setup was conducted by
tion and differentiation [25,26]. Furthermore, chitosan has a back- acquiring 32 scans in the range of 500–4000 cm1, with a resolu-
bone similar to glycosaminoglycan, the major component of bone tion of 4 cm1.
G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109 99

2.5. Morphological analysis 0.49 g of sodium phosphate dihydrate, 13.8 g of potassium phos-
phate and 0.49 g of sodium chloride were dissolved in 1000 ml of
The microstructure of the PCC and PC materials was studied in a ultrapure water. The solution at pH 7.4 was prepared by dissolving
nondestructive manner by micro-computed tomography (l-CT), 0.78 g of sodium phosphate dihydrate, 0.097 g of potassium phos-
with a desktop l-CT scanner (SkyScan 1174, Aartselaar, Belgium). phate and 4 g of sodium chloride in 500 ml of ultrapure water. The
The scanner was set at a voltage of 50 kV and a current of samples, three for each type, were cut in a cylindrical shape
800 lA; the sample was rotationally scanned (0–180°) at (10 mm of diameter and 2 mm of height) and immersed in the
7.55 lm pixel resolution. The exposure time per projection was three different solutions, after recording their initial weight (W0).
2300 ms and no filter was used. No contrasting agents were used The degradation study was carried out for 1 week, during which
and the sample had a cylindrical size of 10 mm of diameter and samples were taken out at specific time intervals, freeze-dried
10 mm of height. Sample volume reconstruction in 3D was per- and weighted (Wf). The mass loss percentage was calculated by
formed by making use of N-Recon software. We quantified the using the following equation:
microstructural parameters of the samples in 3D by making use
Mass Loss ð%Þ ¼ ½ðW0  Wf Þ=W0   100 ð1Þ
of CT-Analyzer software, after having binarized the 2D tomo-
graphic images (‘‘slices”). The threshold levels of the grayscale
images were equally adjusted for all the samples to allow a reliable 2.8. Hydration properties
measurement and comparison of the porosity. Scanning Electron
Microscopy (SEM) analysis was performed to analyze the morphol- For hydration kinetics, PC and PCC samples (10 mm diameter,
ogy of PCC and PC hydrogels. The samples were mounted on alu- 2 mm thickness, n = 3) were allowed swelling in three different
minum stubs and sputtered with gold at 15 mA for 2 min by solutions (the same used for the degradation test pH 2.5, 5.5 and
using Agar Sputter Coater. The morphology of pores and the distri- 7.4) after recording the initial weight (W0) at 37 °C. The swollen
bution of particles in the PEI structure were captured using a scan- disks were taken out of the solution at regular time intervals, blot-
ning electron microscope (EVO MA10 system, Zeiss) equipped with ted with filter paper to remove excess surface water, and their
a micro-analysis system AZTec (Oxford University UK). swollen weights (Ws) were noted. The water uptake by the PCC
and PC networks was determined by the following equation:
2.6. Mechanical characterization
Hydration Degree ð%Þ ¼ ½ðWs  W0Þ=Ws  100 ð2Þ
The mechanical properties of PCC and PC materials were evalu-
ated through unconfined compression and cyclic compression test- 2.9. In vitro studies
ing by using Bose Electroforce equipped with a 100-N load cell. For
unconfined compressive tests, PCC and PC samples were cut in a Osteoblast-like SAOS-2 cells were used in cell growth experi-
circular shape with 10 mm of diameter, and 10 mm of gauge length ments. Experimental cell culture medium (BIOCHROM KG, Berlin)
(n = 3). The mechanical properties were assessed in both dry and consisted of Minimum Eagle’s Medium without L-glutamine, 10%
hydrated conditions (soaked in PBS at 37 °C for 24 h). In prelimi- fetal bovine serum, streptomycin (100 lg/l), penicillin (100 U/
nary tests no sample could reach a failure, so we decided to per- ml), and 2 mM L-glutamine in 250-ml plastic culture flasks (Corn-
form the compressive test until 40% of the height of the sample, ingTM). Cells were cultured at 37 °C in a humidified incubator equi-
which is a sufficient value to see elastic and plastic deformation librated with 5% CO2. Cell suspension was obtained by adding 2 mL
of the samples. A constant crosshead speed of 0.2 mm/min was of a sterile 0.5% Trypsin-EDTA solution (GIBCO by LIFE TECHNOL-
applied. Stress (r (MPa)) was obtained by dividing the applied OGY, ref. 15400-054) to a 250 mL cell culture flask, resuspended
force (N) by the cross-sectional area (mm2). Compressive elastic in the experimental cell culture medium, and diluted to 1  105 -
modulus was calculated from the linear region of the stress- cells/ml. For these experiments, 5 ml of the cell suspension were
strain curve between 0.5 and 5% of strain, and the stress at 40% seeded into 6-well tissue culture polystyrene plates (9.6 cm2 of
of strain was cautiously assumed as collapse stress (this is an growth area; FalconTM), containing the samples. Experiments were
approach commonly followed in the literature to assess the com- performed in triplicate. After 72 h and 2 weeks, samples were care-
pressive strength of polymer/bioceramic composite scaffold fully rinsed with PBS and fixed in 5% glutaraldehyde-PBS. Samples
[32,33]). The area under the stress-strain curve was calculated were dehydrated using increasing concentration of ethanol in
and related to the toughness of the sample. For cycling testing, water-ethanol solutions up to 100% ethanol. Dehydrated samples
100 loading and unloading cycles were performed on PC and PCC were gold sputter-coated (Agar Sputter Coater) and SEM analysis
samples in hydrated condition (n = 3). A first load until 16% of ini- was performed using a EVO MA10 (Zeiss), equipped with a
tial height was applied, and then a sinus cyclic loading curve with a micro-analysis system AZTec (Oxford Instruments, UK). The
frequency of 0.5 Hz and a constant crosshead speed of 1.2 mm/min expression of ALP gene as a cell differentiation marker was
was applied between 16 and 41% of initial height. Stress-strain assessed by using the real time reverse transcription polymerase
curves for PCC and PC samples were reported; the amount of chain reaction (qRT-PCR). Osteoblast–like SAOS-2 cells were cul-
energy adsorbed and the percentage of recovery during the defor- tured as previously described and the total RNA was extracted after
mation cycle were calculated every 10 cycles. 1 week by using MagMax Total RNA Isolation Kit (Applied Biosys-
tems, Milan, Italy) following the manufacturer’s instruction. RNA
2.7. Degradation properties quality was assessed by checking the A260/A280 ratio (1.6–2.0).
Then total RNA was used as a template for cDNA synthesis using
The stability of PCC and PC hydrogels was analyzed at three dif- random hexamers as primer and multiscript reverse transcriptase
ferent pH (2.5, 5.5 and 7.4) at 37 °C (n = 3) following an experimen- (high capacity cDNA RT Kit from Applied Biosystems). cDNA ampli-
tal protocol proposed elsewhere [34]. Briefly, for the solution at pH fication and relative gene quantification were performed by using
2.5, 21 g of citric acid monohydrate were dissolved in 500 ml of Taq Man probe and primers from Applied Biosystems (Hs
ultrapure water in a 1000 ml volumetric flask, then 200 ml of 01029144_m1, ALP). Real time PCR was performed in duplicate
sodium hydroxide 1 M were added and the solution diluted to for all sample and targets on a Step-One instrument (Applied
the mark with water; 40.4 ml of this solution were mixed with Biosystems) by using software Step-One version 2.1. PCRs were
59.6 ml of 0.1 M hydrochloric acid. For the solution at pH 5.5, carried out in a total volume of 20 ll and the amplification was
100 G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109

performed as follows: after an initial denaturation at 95 °C for (Life Technologies), and then reverse transcripted by using the
10 min, the PCR was run for 40 cycles at 95 °C for 15 s and at High-Capacity cDNA Reverse Transcription Kit (Applied Biosys-
60 °C for 1 min. We normalized the content of cDNA samples by tems), according to the manufacturer’s instructions. Total RNA con-
making use of the comparative threshold (Ct) cycle method, con- centration was measured by using a spectrophotometer (UV-1700,
sisting in the normalization of the number of target gene copies Shimadzu) and RNA quality was assessed by evaluating A260/A280
versus the endogenous reference gene GAPDH. The Ct is defined ratio ranging from 1.8 to 2.1. Real-time PCR was performed with
as the fractional cycle number at which the fluorescence generated the TaqMan Gene Expression Master Mix (Applied Biosystems)
by cleavage of the probe passes a fixed threshold baseline when with the Applied Biosystems StepOne Plus instrument (Applied
amplification of the PCR product is first detected. For comparative Biosystems). The primer sets for the Real-time PCR of mouse
analysis of gene expression, data were obtained using DCT method. interleukin-1b (IL-1b), interleukin-6 (IL-6), interleukin-10 (IL-10),
and glyceraldehyde-3-phosphate dehydrogenase (GAPDH)
2.10. Inflammatory response (Applied Biosystems Assay’s ID: Mm01336189_m1,
Mm99999064_m1, Mm99999056_m1, Mm03302249_g1 respec-
The murine macrophage cell line J774.1A (European Collection tively) were chosen from the collection of the TaqMan Gene
of Cell Cultures) was maintained in Dulbecco’s modified Eagle’s Expression Assays (Applied Biosystems). The analysis was con-
medium (Gibco Invitrogen, Cergy-Pontoise, France) supplemented ducted using the method of comparative CT (DCT), which was
with 10% fetal bovine serum, penicillin (100 U ml1), streptomycin designed following the manufacturer’s instructions.
(100 lg ml1) and 4 mM L-glutamine. Cells were grown in a 100%
humidified incubator at 37 °C with 10% CO2 and passaged 2–3 days
before use. The J774.1A cells (2  104 cells ml1) were seeded into 2.11. Statistical analysis
6-well tissue culture polystyrene plates (9.6 cm2 of growth area;
FalconTM), containing the samples. RNA extraction was performed Experimental data were presented as mean ± standard devia-
on the cells grown on the polystyrene in presence or absence of tion. Statistical differences between groups were analyzed using
PC or PCC scaffolds. The extraction protocol was already reported two-way ANOVA using Tukey’s multiple comparison test and Stu-
in the previous paragraph. Briefly, after 4 h, RNA was isolated from dent’s t-test. Statistical significance was represented as *p < 0.05,
**
J774.1A cell line by using the MagMax-96 Total RNA Isolation Kit p < 0.01, ***p < 0.001, ****p < 0.0001.

Fig. 1. Chemical characterization: (a) ATR-IR spectra of pectin and chitosan powders, HA/bTCP ceramic particles, PC hydrogels, PCC composite scaffolds; (b) ATR-IR spectra
focus between 2000 cm1 and 500 cm1, where the shift of the amine band to 1557 cm1 and the bands belonging to the inorganic phase (950 cm1 and 1140 cm1) are
clearly visible.
G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109 101

3. Results and discussion and carboxyl acid (COOH), while the band at 1606 cm1 belongs
to the stretching vibration of the carbonyl group of the carboxylate
3.1. Chemical characterization ion (COO) (Fig. 1b). Concerning the chitosan spectrum, the band at
1647 cm1 is due to the C@O stretching vibration of amide I, while
Fig. 1a shows the ATR-IR spectra of pectin powder, chitosan the band at 1580 cm1 is due to the NH bending amide II, maybe
powder, pectin/chitosan polyelectrolyte complex (PC), HA/bTCP overlapped to the N–H vibration of the amine groups (Fig. 1 b).
particles and pectin/chitosan_HA/bTCP composite hydrogel (PCC). Band assignment is consistent with available literature [36,37].
The analysis confirms that all peaks belong to inorganic material; PC spectra show the formation of PEI complex; a shift of amine
in particular, triply degenerated asymmetric stretching mode (m3) band to 1557 cm1 due to the interaction between the positive
of the P–O bond of the phosphate group is associated to the peaks charge of chitosan, NH+3, and the negative charge of pectin, COO,
at 1125 cm1 for tricalcium phosphate and at 1025–1010 cm1 for was detected (Fig. 1b). As expected, the spectrum of PCC sample
hydroxyapatite [15,35]. Typical spectra of polysaccharide were show bands associated to both the PEI complex (the band at
shown in the case of pectin and chitosan powder. The region 1557 cm1) and the inorganic phase (bands between 950 cm1
between 3700 cm1 and 3000 cm1 for pectin and chitosan is and 1140 cm1) (Fig. 1b) [38].
assigned to the O–H stretching vibration (mOH), while the region
between 3000 and 2800 cm1 belongs to C–H stretching vibration 3.2. Morphological analysis
(mCH) (Fig. 1 a). Deeper analysis on pectin spectrum shows two
bands associated with the stretching vibration at 1740 cm1 of car- SEM analyses at different magnifications for PC and PCC sam-
bonyl group, corresponding to the methyl ester group (COOCH3) ples are reported in Fig. 2. The addition of ceramic particles reduces

Fig. 2. SEM investigation of PC and PCC scaffolds. Highly interconnected pores are shown for PC scaffold; the incorporation of ceramic particles reduces the porosity and pore
size of the material.
102 G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109

the pore size and the total porosity, but reinforces the trabeculae broader distribution of pore size, with most pores located between
and the 3D porous structure of the materials. SEM pictures suggest 200 and 400 lm and the maximum pore size around 650 lm
that the dispersed particles are well distributed in the PEI matrix. (Fig. 3b). Direct comparison of mean pore sizes (St.Sp) further con-
In vitro and in vivo studies from the available literature show that firms the decrease of pore dimension due to the introduction of
a porosity up to 50%, and pore size in the range between 100 and ceramic particles (250 vs. 120 lm). The total porosity decreases
400 lm are the optimum for bone healing and show higher alka- by adding ceramic particles, too, from around 74% for PC sample
line phosphatase activity and new bone formation compared with to 55% in the case of PCC scaffold. Three-dimensional porous scaf-
materials with a porosity lower than 100 lm and higher than folds should allow cell infiltration and facilitate vascular invasion.
400 lm [39]. Furthermore, it is necessary that the ceramic particles are homoge-
A lCT study (Fig. 3a–c) confirmed the results of SEM analysis. nous dispersed in the polymeric matrix – which was observed in
Fig. 3a and c reveal that the bioceramic particles are uniformly this study (Fig. 3a and c) – in order to promote uniform osteogen-
and homogeneously distributed throughout the sample volume. esis. Both micro-porosity and macro-porosity are important mor-
Most pores of PCC sample are in the range of 50–200 lm (the lar- phological properties [40,41]. It has been shown that
gest pores do not exceed 350 lm), while the PC scaffold has a microporosity plays an important role in influencing cell adhesion

Fig. 3. Non-destructive tomographical analysis: (a) lCT images reveal uniform distribution of the ceramic particles in the pectin–chitosan PEI matrix, thus confirming the
results from SEM investigation; (b) Pore size distribution of PC and PCC scaffold, the addition of ceramic scaffold reduces the pore size. (c) 2-D development of PC and PCC
scaffolds, obtained by placing the analysis planes [xy] and [yz] in the mid-length, and moving the [xz] plane from the bottom to the middle and until the top of the length of
the scaffold; 3-D reconstruction of the central (approximately mid-length) cross-section along the [xy], [xz] and [yz] orthogonal planes is also shown.
G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109 103

to the biomaterial surface, while macro-porosity promotes blood material, both in as prepared and hydrated conditions. Compres-
vessel infiltration, nutrients transportation and clot formation, sive elastic modulus for PC scaffolds was 1005 ± 250 kPa in as pre-
which is important to stimulate the overall healing process, pared condition, instead it was twofold higher for PCC scaffold,
besides allowing cell infiltration and new bone growth [42–44]. 2559 ± 596 kPa. Hydration kinetics data show that the water
uptake at pH 7.4 was higher in both cases, and the compressive
3.3. Mechanical characterization elastic modulus decreases to 33 ± 8 kPa for PC material and to
65 ± 9 kPa for PCC scaffolds. Alveolar bone regeneration is a low-
The incorporation of ceramic particles into soft hydrogel scaf- bearing application, where providing stability and three-
folds is a valuable strategy to improve the mechanical properties dimensional shape for functional and aesthetic reasons is more
of the base material [45,46]. A compressive test was performed important than mechanical strength [16,47]. PC materials show a
on PC and PCC scaffolds, and the results are shown in Fig. 4a and soft structure, and the incorporation of ceramic particles inside
in Table 1. The addition of HA/bTCP particles increases the elastic the PEI matrix makes the material tougher (Fig. 4a). For example,
modulus, the compressive strength and the toughness of the the toughness was 34.3 ± 4.0 kJ/m3 and 222.8 ± 1.9 kJ/m3 for PC

Fig. 4. Compressive test: (a) Stress–strain curves for PC and PCC scaffolds in as prepared and hydrated conditions. (b) loading and unloading curves for PC and PCC samples
subjected to cyclic strain (100 cycles) in as prepared and fully hydrated conditions; (c) energy absorbed and recovery percentage (monitored every 10 cycles) for PC and PCC
samples (the incorporation of calcium phosphate particles increases the strength of PCC scaffold, but does not reduce its recovery ability which is similar to that of PC
material). The data are represented as mean ± standard deviation (n = 3).

Table 1
Compressive modulus, collapse stress (at 40% strain) and toughness for PC and PCC materials in both dry and hydrated conditions.

Samples As prepared Hydrated


Compressive modulus [kPa] Stress40% [kPa] Toughness [kJ/m3] Compressive modulus [kPa] Stress40% [kPa] Toughness [kJ/m3]
PC 1005 ± 250 127 ± 6 34.3 ± 4.0 33 ± 8 13 ± 3 2.4 ± 0.5
PCC 2559 ± 596 1525 ± 61 222.8 ± 1.9 65 ± 9 121 ± 27 12.0 ± 2.1
104 G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109

and PCC scaffolds, respectively, in as prepared condition. There is a furthermore, PC and PCC materials, after being left without load
relative paucity of literature dealing with the toughness of bone to recover the (original) shape for 5 min, showed a variation of
tissue engineering scaffolds and, thus, comparison with previous the final diameter with respect to the initial one of 1.4 ± 0.59%
works is difficult; however, it is interesting to point out that the and 7.52 ± 1.32%, respectively (more than 90% of recovery for both
toughness of PCC scaffold is ten times higher than that assessed samples). This means that in vivo the PCC scaffold is able to main-
for single-phase glass-ceramic porous scaffolds [48]. After 24 h at tain the shape and to avoid the collapse of the site. Furthermore,
37 °C in a solution with pH 7.4, both hydrated samples show a tooth extraction site has a non-regular shape, hence ensuring the
decrease of toughness (2.4 ± 0.5 kJ/m3 for PC scaffold and possibility to customize the dimension of the implant material in
12.0 ± 2.1 kJ/m3 for PCC scaffold) compared to as prepared materi- order to fit the socket is an important characteristic for dental
als. A similar behavior was also found by Liu et al. on bioglass scaf- practice.
folds [49]. The toughness of PCC material in hydrated condition is
comparable, as order of magnitude, to the value found by other 3.4. Hydration studies at different pH
authors for hydroxyapatite scaffolds [50]. Ceramic particles, as
expected, reduce the porosity but reinforce mechanically the scaf- A high degree of swelling allows cell infiltration into the scaf-
folds, maintaining a range and a degree of porosity that should still folds and maximizes the probability of cell growth in a three-
allow cell infiltration and proliferation [40]. dimensional structure [22]. Furthermore, high swelling behavior
Cyclic stress is a common situation to which the scaffolds used improves the capability of the scaffold to adsorb nutrients from
for alveolar bone regeneration are subjected. The stability and the culture media. Chitosan/pectin complexes are formed by a
integrity of the scaffolds are the most important properties that ionic interaction between positive charges of chitosan and negative
they should have, since material fragmentation could provoke an charges of pectin; hence, the PEI network exhibits a pH-sensitive
inflammatory response and a huge deformation could cause the swelling [34,51]. We tested the swelling behavior in three different
collapse of the defect. Hydrogel PC scaffolds show a stable energy solutions with three different pH (2.5, 5.5 and 7.4), and the results
adsorbed (and recovery network) from the 5th to the 100th cycle; are shown in Fig. 5. PC hydrogels showed a significantly different
the addition of 90% of ceramic particles does not affect too much behavior depending on the pH of the solution. If the pH of the solu-
the trend, although the energy adsorbed was slightly higher for tion changes, the degree of interaction between pectin and chi-
the PCC composite due to the reduced mobility of the PEI network tosan changes, too, and the swelling increases or decreases
(Fig. 4b, c). The results are affected by the deformation of the depending on the degree of dissociation of the complex (Fig. 5a).
swollen surface of the samples that do not return instantaneously At low pH value (2.5), pectin is neutralized and free positive
to the original shape after cessation of compression stress. After charges (NH3+) appear in the network; COOH from pectin chain
100 cycles no fragmentation was detected for both samples; allows the swelling of the material until the value of

Fig. 5. Swelling properties: (a) swelling kinetics of PC and PCC scaffolds tested in different pH conditions (2.5, 5.5 and 7.4) over 24 h; (b) summary of the swelling degrees at
24 h; (c) optical images show the swollen sample at different pH, and (d) proof of the easy mouldability of composite scaffolds, along with the high hydration properties
(water colored with few drops of red ink was used for this qualitative hydration test) and the capacity to recover the initial shape after a compressive stress. The results are
reported as mean ± standard deviation (n = 3), (*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, two-way ANOVA with Tukey’s multiple comparison test).
G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109 105

1460.42 ± 81.33% of the initial mass, after 6 h. After 24 h, PC hydro- almost 10-fold lower than the value reported for PC hydrogel at
gels soaked in a strongly acidic solution (pH 2.5) showed a huge the same pH. The maximum percentage of solution uptake for
increase in the water uptake; this behavior could be associated PCC material was reached after 2 h and was stable until 24 h. As
to the total degradation of pectin and to the loss of PEI network already observed for PC hydrogels, also in the case of PCC samples
that allows the highest water adsorption, 2495.34 ± 33.87%. At the lowest value of water uptake was shown for the sample soaked
alkaline pH (7.4), free negative charges appear inside the network, in the solution at pH 5.5.
since at higher pH chitosan is partially neutralized and the PEI net- Providing a stable material could allow the dental technician to
work loosens, thus leading to a hydration degree of use the scaffold independently of the environment (especially the
1880.2 ± 218.8%, after 6 h. At pH 7.4, the partial neutralization of pH), since it will work always in the same manner avoiding unex-
NH+3 has not caused a total loss of the PEI network, since chitosan pected behaviors (Fig. 5c). Furthermore, the swelling of the PCC
has still some ionized groups bonded with COO of the pectin scaffold is high enough to ensure cell infiltration and nutrient
chain. However, after 6 h, the swelling ratio started to decrease transportation, and a sufficient mechanical strength is still main-
due to an initial degradation of the network. Since the pKa of pectin tained. The incorporation of ceramic particles into the PC matrix
is 4.0 and that of chitosan is 6.0, at the pH of 5.5 over 99% of pectin reduces the swelling capability of the materials, which is maybe
is still in its ionized form and chitosan exists in both ionized (NH+3) due to the interaction between the particles and the network. In
and unionized forms (NH2). The swelling percentage of PC network particular, pectin carboxylic groups could be ionically crosslinked
in the solution with a pH of 5.5 was 1443.8 ± 14.4% after 6 h; then, by calcium ions (Ca2+), thereby forming the so-called ‘‘egg box”
the capability of the network to adsorb the solution is reduced structure, where a divalent cation is bonded with different car-
compared to the other cases. Specifically, due to the presence in boxylic anions [28]. Furthermore, the high percentage of rigid,
the network at pH 5.5 of intramolecular H-bonding between non-deformable ceramic particles (which cannot swell) used in
COOH3 and OH, which makes the PEI matrix more stable, the PC this work (90 wt.%) reduces the influence of the PC matrix in the
hydrogel after 24 h at pH 5.5 show a significantly lower swelling swelling properties. In alveolar bone regeneration, the use of
degree (687.69%) compared to the same material at the pH of 7.4 sponges is desirable, since they could easily fit into the alveolar
(around 1450%) and 2.5 (around 2500%). bone defect, be simply cut by scissors or a lancet, and be easily
Addition of HA/bTCP particles stabilizes, in general, the swelling molded in the periodontal cavity (Fig. 5d) [16,43,52]. The addition
properties of the PC-based hydrogel (maximum swelling degree of of calcium phosphate particles does not influence the moldability
the composite around 300%) (Fig. 5 a). The swelling behavior of the PC hydrogel base material (Fig. 5d).
between PC and PCC materials is significantly different, while no
statistically significant differences were assessed among PCC sam- 3.5. Degradation properties at different pH
ples during the hydration kinetics in the three different solutions
(pH of 2.5, 5.5, 7.4) (Fig. 5 b). As expected, the maximum water After implantation, the composite hydrogel should degrade, and
uptake for PCC scaffold was lower compared to the PC hydrogel new bone should form and replace it. Furthermore, during degra-
in all the conditions considered: for example the swelling degree dation the osteconductive HA/bTCP ceramic particles come into
for PCC scaffold at pH 7.4 was about 275.05 ± 35.45% after 6 h, direct contact with the newly formed bone and should further

Fig. 6. Degradation properties at different pH: (a) degradation behavior of the PC and PCC materials over 1 week in different pH conditions; (b) summary of mass losses after
soaking for 1 week; (c) optical images of PC and PCC scaffolds during the degradation test. The results are reported as mean ± standard deviation (n = 3), (*p < 0.05, **p < 0.01,
***
p < 0.001, ****p < 0.0001, two-way ANOVA with Tukey’s multiple comparison test).
106 G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109

promote osseointegration. The degradation behaviors of PC and environment. Furthermore, the ionic interaction between the two
PCC samples have been tested in three different conditions of pH polymer chains is reversible and non-permanent, so in aqueous
(2.5, 5.5, 7.4) for 1 week and the results are shown in Fig. 6. During solution this association is gradually reduced and then lost with
the degradation process, the mechanisms are similar to those time. PCC scaffolds tested in the solution at pH of 5.5 and 7.4
obtained during the hydration kinetics, since the solutions used showed a similar percentage of initial mass loss, around 10%. The
were the same [53]. PC hydrogel shows the highest degradation addition of ceramic particles makes the materials more resistant
rate at pH 2.5 with almost 100% mass loss after 1 week; this mass and less susceptible to pH variation (Fig. 6c).
loss was twice compared to that of PC treated at pH 7.4 and almost
three times with respect to the hydrogel soaked at pH 5.5, which 3.6. In vitro studies
lost around 40% of the initial mass (Fig. 6a, b). At the pH of 2.5,
the PEI network loosens due to the neutralization of the pectin. Adhesion, spreading and morphology of osteoblast-like cells
The same behavior is reported for the PCC sample, that lost around cultured for 14 days on PC and PCC are shown in Fig. 7. SEM images
40% of the initial mass after 1 week in the solution at the pH of 2.5. show that, after 72 h, SAOS-2 cells seeded on the PC scaffold sur-
These results are due in part to the neutralization of COO ions, face exhibit a round shape and low density, followed by high pro-
and in part to the dissolution of ceramic particles in a highly acidic liferation and formation of a network of cells. In particular, SEM

Fig. 7. Biological tests with SAOS-2 cells: adhesion and proliferation of osteoblast-like cells were investigated by SEM analysis on (a) PC and (b) PCC scaffolds after 72 h and
2 weeks of culture. HA/bTCP particles promote adhesion and proliferation of SAOS 2 cells after 72 h and a confluent layer of cells was detected after 14 days.
G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109 107

images after 2 weeks show the formation of large cords of cells, 3.7. Inflammatory response
which prefer to stay together rather than adhere and spread on
the surface of the materials (Fig. 7a). The addition of ceramic par- Gene expression analysis on IL1b, IL6 and IL10 is reported in
ticles increases the adhesion and proliferation of osteoblast cells at Fig. 9. Inflammatory response was evaluated on the cells grown
72 h, and the SEM images reveal that, after 2 weeks, SAOS-2 cells on the polystyrene in presence of PC, PCC and without the scaffold.
are completely adhered on the surface and infiltrated inside the The test was performed in order to evaluate the possible inflamma-
porosity, forming bridges and creating a strong and consistent tion which could be provoked on the tissue surrounding the
layer of cells (Fig. 7b). The higher attachment and proliferation of implanted material. Degradation products, chemical composition
cells on the PCC materials are due to the presence of the calcium and swelling behavior could influence the response of the sur-
phosphate phase that conducts osteoblast proliferation [54] Fur- rounding tissue; if the material does not stimulate any cytokines
thermore, the addition of micro-particles increases the roughness expression, a foreign body reaction is avoided. As shown in Fig. 9,
and the area of the surface, which promote the osteoblast prolifer- no material elicits a pro-inflammatory response, and the expres-
ation and adhesion [23]. Since PC and PCC materials show different sions of IL1b and IL6 are comparable between the cells grown in
cell morphology at two weeks but both demonstrated cytocompat- presence of PC/PCC and on the control (polystyrene alone). A slight
ibility and high proliferation, we thus performed ALP gene- increase in the expression of IL10 was reported for PCC material
expression after 7 days of SAOS-2 cell culture. The results are compared with the polystyrene control, conversely no significance
reported in the bar graph in Fig. 8 and demonstrated that the pres- was assessed to the increase of IL10 for PC material. Bone resorp-
ence of HA/bTCP particles in the PCC composite scaffolds promotes tion occurs when inflammatory mediators reach a critical concen-
ALP activity, which is 3-fold higher than that of PC-based hydro- tration that depends on the expression of pro-inflammatory
gels. In vitro studies show a good biocompatibility of PCC scaffold, cytokines, such as the interleukin (IL) family, of which IL1b is the
furthermore the addition of ceramic particles could promote alve- most studied member due to its role in acute and chronic
olar bone formation and infiltration inside the PCC composite inflammatory and autoimmune disorders. On the opposite site,
hydrogel. the inflammatory level is controlled by the expression of

Fig. 8. ALP gene expression after 1 week of osteoblast-like SAOS-2 cell culture. The results are reported as mean ± standard deviation (n = 4), (*p < 0.05, **p < 0.01, Student’s t-
test).

Fig. 9. Inflammatory response of murine macrophage cells surrounding the scaffolds (gene expression of IL1b, IL6 and IL10 of the cells grown in contact with PC and PCC
materials). The results are reported as mean ± standard deviation (n = 4) (*p < 0.05, two-way ANOVA with Tukey’s multiple comparison test).
108 G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109

anti-inflammatory cytokines, such as IL10. In normal physiological [7] A. Tallgren, The continuing reduction of the residual alveolar ridges in
complete denture wearers: a mixed-longitudinal study covering 25 years, J.
conditions, there is a balance between bone formation and bone
Prosthet. Dent. 89 (2003) 427–435.
resorption, but this balance can be altered in certain inflammatory [8] S. Bernstein, J. Cooke, P. Fotek, H.-L. Wang, Vertical bone augmentation: where
conditions. This equilibrium is altered by the action of pro- are we now?, Implant Dent 15 (2006) 219–228.
inflammatory cytokines, such as IL1b that induces osteoclastogen- [9] F.G. Draenert, D. Huetzen, A. Neff, W.E.G. Mueller, Vertical bone augmentation
procedures: basics and techniques in dental implantology, J. Biomed. Mater.
esis, while counteraction by anti-inflammatory mediators, such as Res. A 102 (2014) 1605–1613.
IL10, inhibits osteoclastogenesis. An anti-inflammatory response [10] F.-M. Chen, Y. Jin, Periodontal tissue engineering and regeneration: current
seems to occur in both PC and PCC scaffolds, although a slight approaches and expanding opportunities, Tissue Eng. B: Rev. 16 (2010) 219–
255.
increase of IL10 gene expression was detected in the latter com- [11] J.D. Bashutski, H.L. Wang, Periodontal and endodontic regeneration, J. Endod.
pared to the former (Fig. 9). The hypothesis is that this anti- 35 (2009) 321–328.
inflammatory behavior could be due to the properties of pectin, [12] S. Elangovan, S. Srinivasan, S. Ayilavarapu, Novel regenerative strategies to
enhance periodontal therapy outcome, Expert Opin. Biol. Ther. 9 (2009) 399–
which has been shown to exhibit an anti-inflammatory action in 410.
many studies [55–57]. Further and in-depth studies have to be [13] D. Tadic, M. Epple, A thorough physicochemical characterisation of 14 calcium
done in order to fully elucidate this behavior, but this preliminary phosphate-based bone substitution materials in comparison to natural bone,
Biomaterials 25 (2004) 987–994.
work confirms that PCC could be used as a safe bone graft for alve- [14] H. Yuan, M. Van Den Doel, S. Li, C.A. Van Blitterswijk, K. De Groot, J.D. De
olar bone regeneration, without the risk of inducing inflammation Bruijn, A comparison of the osteoinductive potential of two calcium phosphate
in the surrounding tissue. ceramics implanted intramuscularly in goats, J. Mater. Sci. – Mater. Med. 13
(2002) 1271–1275.
[15] M. Morra, G. Giavaresi, M. Sartori, A. Ferrari, A. Parrilli, D. Bollati, R.R.Y. Baena,
4. Conclusions C. Cassinelli, M. Fini, Surface chemistry and effects on bone regeneration of a
novel biomimetic synthetic bone filler, J. Mater. Sci. – Mater. Med. 26 (2015).
[16] T. Matsuno, K. Omata, Y. Hashimoto, Y. Tabata, T. Satoh, Alveolar bone tissue
Alveolar bone regeneration is a procedure that requires a bio- engineering using composite scaffolds for drug delivery, Jpn. Dent. Sci. Rev. 46
material with some peculiar characteristics such as mouldability (2010) 188–192.
[17] L. Zhang, N. Hanagata, M. Maeda, T. Minowa, T. Ikoma, H. Fan, X. Zhang, Porous
in order to fill the irregular void, biodegradability, swelling capac- hydroxyapatite and biphasic calcium phosphate ceramics promote ectopic
ity to promote infiltration of nutrients, absence of inflammatory osteoblast differentiation from mesenchymal stem cells, Sci. Technol. Adv.
response on the surrounding tissue, osteoconduction and stimula- Mater. 10 (2009) 025003.
[18] M. Alcaide, M.C. Serrano, R. Pagani, S. Sánchez-Salcedo, M. Vallet-Regí, M.T.
tion of new bone formation. Composite scaffolds comprising a Portolés, Biocompatibility markers for the study of interactions between
pectin/chitosan base hydrogel filled with HA/bTCP particles (PCC) osteoblasts and composite biomaterials, Biomaterials 30 (2009) 45–51.
was successfully prepared and characterized. The results show that [19] S. Schaefer, R. Detsch, F. Uhl, U. Deisinger, G. Ziegler, How degradation of
calcium phosphate bone substitute materials is influenced by phase
PCC material can be easily molded to fit the defect size/shape and composition and porosity, Adv. Eng. Mater. 13 (2011) 342–350.
has a good stability in different pH condition, with a high swelling [20] J.A. Killion, S. Kehoe, L.M. Geever, D.M. Devine, E. Sheehan, D. Boyd, C.L.
degree (up to 300% of the initial weight). Mechanical characteriza- Higginbotham, Hydrogel/bioactive glass composites for bone regeneration
applications: synthesis and characterisation, Mater. Sci. Eng., C 33 (2013)
tion demonstrated that the addition of ceramic particles increases
4203–4212.
the mechanical strength compared to the base hydrogel (toughness [21] L. Peng, Preparation and evaluation of porous chitosan/collagen scaffolds for
of PCC scaffold was around 220 kJ/m3, and compressive elastic periodontal tissue engineering, J. Bioact. Compat. Polym. 21 (2006) 207–220.
modulus was 2.5 MPa in dry condition). The scaffold morphology [22] M. Peter, N.S. Binulal, S.V. Nair, N. Selvamurugan, H. Tamura, R. Jayakumar,
Novel biodegradable chitosan-gelatin/nano-bioactive glass ceramic composite
and porosity as well as the presence of osteoconductive HA/bTCP scaffolds for alveolar bone tissue engineering, Chem. Eng. J. 158 (2010) 353–
micro-particles promote highly osteoblast adhesion and prolifera- 361.
tion with 2-fold higher ALP gene expression at 1 week compared to [23] S. Sowmya, P.T.S. Kumar, K.P. Chennazhi, S.V. Nair, H. Tamura, R. Jayakumar,
Biocompatible b-chitin hydrogel/nanobioactive glass ceramic nanocomposite
PC scaffold. Gene expression results demonstrated that PCC scaf- scaffolds for periodontal bone regeneration, Trends Biomater. Artif. Organs 25
fold elicits anti-inflammatory and pro-osteogenic responses; this (2011) 1–11.
results confirm that PCC biomaterial could be a promising tool [24] M.G. Araújo, B. Liljenberg, J. Lindhe, Dynamics of bio-Oss collagen
incorporation in fresh extraction wounds: an experimental study in the dog,
for application in alveolar bone regeneration. Clin. Oral Implants Res. 21 (2010) 55–64.
[25] P.K. Dutta, J. Duta, V.S. Tripathi, Chitin and chitosan: chemistry, properties and
applications, J. Sci. Ind. Res. (India) 63 (2004) 20–31.
Acknowledgments [26] J. Finlay, L. Miller, J. Poupard, A review of the antimicrobial activity of chitosan,
J. Antimicrob. Chemother. 52 (2003) 18–23.
GI would like to acknowledge Nobil Bio Ricerche srl and Politec- [27] E. Khor, L.Y. Lim, Implantable applications of chitin and chitosan, Biomaterials
24 (2003) 2339–2349.
nico di Torino, Department of Applied Science and Technology, for [28] P. Sriamornsak, Chemistry of pectin and its pharmaceutical uses: a review,
the PhD fellowship. This research was funded by Nobil Bio Ricerche Silpakorn Univ, J. Soc. Sci. Humanit. Arts 3 (2003) 206–228.
srl. [29] R.K. Mishra, A.K. Banthia, A.B. Majeed, Pectin based formulations for
biomedical applications: a review, Asian J. Pharm. Clin. Res. 5 (2012) 1–7.
[30] F. Munarin, S.G. Guerreiro, M.A. Grellier, M.C. Tanzi, M.A. Barbosa, P. Petrini, P.
References L. Granja, Pectin-based injectable biomaterials for bone tissue engineering,
Biomacromolecules 12 (2011) 568–577.
[31] L. Liu, M.L. Fishman, J. Kost, K.B. Hicks, Pectin-based systems for colon-specific
[1] L. Gaviria, J.P. Salcido, T. Guda, J.L. Ong, Current trends in dental implants, J.
drug delivery via oral route, Biomaterials 24 (2003) 3333–3343.
Korean Assoc. Oral Maxillofac. Surg. 40 (2014) 50.
[32] P. Gentile, M. Mattioli-Belmonte, V. Chiono, C. Ferretti, F. Baino, C. Tonda-Turo,
[2] A. Gupta, M. Dhanraj, G. Sivagami, Status of surface treatment in endosseous
C. Vitale-Brovarone, L. Pashkuleva, R.L. Reis, G. Ciardelli, Bioactive glass/
implant: a literary overview, Indian J. Dent. Res. 21 (2010) 433–438.
polymer composite scaffolds mimicking bone tissue, J. Biomed. Mater. Res.,
[3] G.M. Raghoebar, R.H. Batenburg, A. Vissink, H. Reintsema, Augmentation of
Part A 100A (2012) 2654–2667.
localized defects of the anterior maxillary ridge with autogenous bone before
[33] P. Gentile, V. Chiono, C. Tonda-Turo, C. Mattu, F. Baino, C. Vitale-Brovarone, G.
insertion of implants, J. Oral Maxillofac. Surg. 54 (1996) 1180–1185. discussion
Ciardelli, Bioresorbable glass effect on the physico-chemical properties of
1185–1186.
bilayered scaffolds for osteochondral regeneration, Mater. Lett. 89 (2012) 74–
[4] J. Liu, D.G. Kerns, Mechanisms of guided bone regeneration: a review, Open
76.
Dent. J. 8 (2014) 56–65.
[34] F. Bigucci, B. Luppi, T. Cerchiara, M. Sorrenti, G. Bettinetti, L. Rodriguez, V.
[5] F. Van Der Weijden, F. Dell’Acqua, D.E. Slot, Alveolar bone dimensional changes
Zecchi, Chitosan/pectin polyelectrolyte complexes: selection of suitable
of post-extraction sockets in humans: a systematic review, J. Clin. Periodontol.
preparative conditions for colon-specific delivery of vancomycin, Eur. J.
36 (2009) 1048–1058.
Pharm. Sci. 35 (2008) 435–441.
[6] L. Schropp, A. Wenzel, L. Kostopoulos, T. Karring, Bone healing and soft tissue
[35] S. Koutsopoulos, Synthesis and characterization of hydroxyapatite crystals: a
contour changes following single-tooth extraction: a clinical and radiographic
review study on the analytical methods, J. Biomed. Mater. Res. 62 (2002) 600–
12-month prospective study, Int. J. Periodontics Restorative Dent. 23 (2003)
612.
313–323.
G. Iviglia et al. / Acta Biomaterialia 44 (2016) 97–109 109

[36] S.S. Rashidova, R.Y. Milusheva, L.N. Semenova, M.Y. Mukhamedjanova, N.L. mechanical and cell adhesion characteristics, Mater. Sci. Eng., C 33 (2013)
Voropaeva, S. Vasilyeva, R. Faizieva, I.N. Ruban, Characteristics of interactions 1800–1807.
in the pectin-chitosan System, Chromatographia 59 (2004) 779–782. [47] D.A. Wahl, J.T. Czernuszka, Collagen-hydroxyapatite composites for hard tissue
[37] G. Morris, S. Kök, S. Harding, G. Adams, Polysaccharide drug delivery repair, Eur. Cells Mater. 11 (2006) 43–56.
systems based on pectin and chitosan, Biotechnol. Genet. Eng. Rev. 27 [48] C. Vitale-Brovarone, G. Ciapetti, E. Leonardi, N. Baldini, O. Bretcanu, E. Verné, F.
(2010) 257–284. Baino, Resorbable glass-ceramic phosphate-based scaffolds for bone tissue
[38] P. Coimbra, P. Ferreira, H.C. de Sousa, P. Batista, M.A. Rodrigues, I.J. Correia, M. engineering: synthesis, properties, and in vitro effects on human marrow
H. Gil, Preparation and chemical and biological characterization of a stromal cells, J. Biomater. Appl. 26 (2011) 465–489.
pectin/chitosan polyelectrolyte complex scaffold for possible bone tissue [49] X. Liu, M.N. Rahaman, G.E. Hilmas, B.S. Bal, Mechanical properties of bioactive
engineering applications, Int. J. Biol. Macromol. 48 (2011) 112–118. glass (13–93) scaffolds fabricated by robotic deposition for structural bone
[39] E. Tsuruga, H. Takita, H. Itoh, Y. Wakisaka, Y. Kuboki, Pore size of porous repair, Acta Biomater. 9 (2013) 7025–7034.
hydroxyapatite as the cell-substratum controls BMP-induced osteogenesis, J. [50] H.-W. Kim, J.C. Knowles, H.-E. Kim, Hydroxyapatite porous scaffold engineered
Biochem. 121 (1997) 317–324. with biological polymer hybrid coating for antibiotic vancomycin release, J.
[40] V. Karageorgiou, D. Kaplan, Porosity of 3D biomaterial scaffolds and Mater. Sci. – Mater. Med. 16 (2005) 189–195.
osteogenesis, Biomaterials 26 (2005) 5474–5491. [51] A. Ghaffari, K. Navaee, M. Oskoui, K. Bayati, M. Rafiee-Tehrani, Preparation and
[41] J.R. Woodard, A.J. Hilldore, S.K. Lan, C.J. Park, A.W. Morgan, J.A.C. Eurell, S.G. characterization of free mixed-film of pectin/chitosan/Eudragit RS intended for
Clark, M.B. Wheeler, R.D. Jamison, A.J.W. Johnson, The mechanical properties sigmoidal drug delivery, Eur. J. Pharm. Biopharm. 67 (2007) 175–186.
and osteoconductivity of hydroxyapatite bone scaffolds with multi-scale [52] Z. Sheikh, C. Sima, M. Glogauer, Bone replacement materials and techniques
porosity, Biomaterials 28 (2007) 45–54. used for achieving vertical alveolar bone augmentation, Materials (Basel) 8
[42] A. Scabbia, L. Trombelli, A comparative study on the use of a (2015) 2953–2993.
HA/collagen/chondroitin sulphate biomaterial (Biostite) and a bovine- [53] P.-H. Chen, T.-Y. Kuo, J.-Y. Kuo, Y.-P. Tseng, D.-M. Wang, J.-Y. Lai, H.-J. Hsieh,
derived HA xenograft (Bio-Oss) in the treatment of deep intra-osseous Novel chitosan–pectin composite membranes with enhanced strength,
defects, J. Clin. Periodontol. 31 (2004) 348–355. hydrophilicity and controllable disintegration, Carbohydr. Polym. 82 (2010)
[43] T. Matsuno, T. Nakamura, K. Kuremoto, S. Notazawa, T. Nakahara, Y. 1236–1242.
Hashimoto, T. Satoh, Y. Shimizu, Development of beta-tricalcium [54] S.V. Dorozhkin, Biphasic, triphasic and multiphasic calcium orthophosphates,
phosphate/collagen sponge composite for bone regeneration, Dent. Mater. J. Acta Biomater. 8 (2012) 963–977.
25 (2006) 138–144. [55] N.L. Wang, H. Kiyohara, T. Matsumoto, H. Otsuka, M. Hirano, H. Yamada,
[44] F. Baino, C. Vitale-Brovarone, Three-dimensional glass-derived scaffolds for Polyclonal antibody against a complement-activating pectin from the roots of
bone tissue engineering: current trends and forecasts for the future, J. Biomed. Angelica acutiloba, Planta Med. 60 (1994) 425–429.
Mater. Res. A 97 (2011) 514–535. [56] M.H. Sakurai, T. Matsumoto, H. Kiyohara, H. Yamada, B-cell proliferation
[45] H.H.K. Xu, C.G. Simon, Fast setting calcium phosphate-chitosan scaffold: activity of pectic polysaccharide from a medicinal herb, the roots of Bupleurum
mechanical properties and biocompatibility, Biomaterials 26 (2005) 1337– falcatum L. and its structural requirement, Immunology 97 (1999) 540–547.
1348. [57] H. Salman, M. Bergman, M. Djaldetti, J. Orlin, H. Bessler, Citrus pectin affects
[46] A.K. Gaharwar, C. Rivera, C.J. Wu, B.K. Chan, G. Schmidt, Photocrosslinked cytokine production by human peripheral blood mononuclear cells, Biomed.
nanocomposite hydrogels from PEG and silica nanospheres: structural, Pharmacother. 62 (2008) 579–582.

You might also like