ArticlePDF Available

A BMSCs‐laden Quercetin/Duck's Feet Collagen/Hydroxyapatite Sponge for Enhanced Bone Regeneration

Authors:

Abstract and Figures

Treating critical‐sized bone defects is an important issue in the field of tissue engineering and bone regeneration. From the various biomaterials for bone regeneration, collagen is an important and widely used biomaterial in biomedical applications, hence, it has numerous attractive properties including biocompatibility, hyper elastic behavior, prominent mechanical properties, support cell adhesion, proliferation, and biodegradability. In the present study, collagen was extracted from duck's feet (DC) as a new collagen source and combined with quercetin (Qtn), a type of flavonoids found in apple and onions and has been reported to affect the bone metabolism, for increasing osteogenic differentiation. Further, improving osteoconductive properties of the scaffold Hydroxyapatite (HAp) a biodegradable material was used. We prepared 0, 25, 50 and 100 μM Qtn/DC/HAp sponges using Qtn, DC, and HAp. Their physiochemical characteristics were evaluated using scanning electron microscopy (SEM), compressive strength, porosity, and Fourier transform infrared spectroscopy (FT‐IR). To assess the effect of Qtn on osteogenic differentiation, we cultured bone marrow mesenchymal stem cells (BMSCs) on the sponges and evaluated by ALP, MTT assay, and RT‐PCR. Additionally, they were studied implanting in rat, analyzed through Micro‐CT and histological staining. From our in vitro and in vivo results, we found that Qtn has an effect on bone regeneration. Among the different experimental groups, 25 μM Qtn/DC/HAp sponge was found to be highly increased in cell proliferation and osteogenic differentiation compared with other groups. Therefore, 25 μM Qtn/DC/HAp sponge can be used as an alternative biomaterial for bone regeneration in critical situations. This article is protected by copyright. All rights reserved.
Content may be subject to copyright.
ORIGINAL ARTICLE
A BMSCs-laden quercetin/duck's feet collagen/hydroxyapatite
sponge for enhanced bone regeneration
Jeong Eun Song
1
| Jingwen Tian
2
| Yeon Ji Kook
1
| Muthukumar Thangavelu
1
|
Joo Hee Choi
1
| Gilson Khang
1
1
Department of BIN Convergence Technology,
Department of Polymer Nano Science &
Technology and polymer Materials Fusion
Research Center, Chonbuk National
University, Jeonju-si, Jeollabuk-do, Republic of
Korea
2
Department of Nuclear Medicine, Molecular
Imaging and Therapeutic Medicine Research
Center, Cyclotron Research Center, Institute
for Medical Science, Biomedical Research
Institute, Chonbuk National University Medical
School and Hospital, Jeonju-si, Jeollabuk-do,
Republic of Korea
Correspondence
Gilson Khang, Department of BIN
Convergence Technology, Department of
Polymer Nano Science & Technology and
polymer Materials Fusion Research Center,
Chonbuk National University, 567 Baekje-
daero, Deokjin-gu, Jeonju-si, Jeollabuk-do
54896, Republic of Korea.
Email: gskhang@chonbuk.ac.kr
Funding information
Korea Health Industry Development Institute,
Grant/Award Number: HI15C2996; Ministry
of Science, ICT and Future Planning, Grant/
Award Number: 2017K1A3A7A03089427
Abstract
Treating critical-sized bone defects is an important issue in the field of tissue engi-
neering and bone regeneration. From the various biomaterials for bone regeneration,
collagen is an important and widely used biomaterial in biomedical applications,
hence, it has numerous attractive properties including biocompatibility, hyper elastic
behavior, prominent mechanical properties, support cell adhesion, proliferation, and
biodegradability. In the present study, collagen was extracted from duck's feet
(DC) as a new collagen source and combined with quercetin (Qtn), a type of flavo-
noids found in apple and onions and has been reported to affect the bone metabo-
lism, for increasing osteogenic differentiation. Further, improving osteoconductive
properties of the scaffold hydroxyapatite (HAp) a biodegradable material was used.
We prepared 0, 25, 50, and 100 μM Qtn/DC/HAp sponges using Qtn, DC, and
HAp. Their physiochemical characteristics were evaluated using scanning electron
microscopy, compressive strength, porosity, and Fourier transform infrared spectros-
copy. To assess the effect of Qtn on osteogenic differentiation, we cultured bone
marrow mesenchymal stem cells on the sponges and evaluated by alkaline phospha-
tase, 3-4-2, 5-diphenyl tetrazolium bromide assay, and real-time polymerase chain
reaction. Additionally, they were studied implanting in rat, analyzed through Micro-
CT and histological staining. From our in vitro and in vivo results, we found that Qtn
has an effect on bone regeneration. Among the different experimental groups,
25 μM Qtn/DC/HAp sponge was found to be highly increased in cell proliferation
and osteogenic differentiation compared with other groups. Therefore, 25 μM
Qtn/DC/HAp sponge can be used as an alternative biomaterial for bone regeneration
in critical situations.
KEYWORDS
bone marrow mesenchymal stem cells, bone regeneration, duck's feet collagen,
hydroxyapatite, quercetin
1|INTRODUCTION
One among the primary goals of tissue engineering and regeneration
is to develop a variety of techniques to reconstruct defected bone
(Amini, Laurencin, & Nukavarapu, 2012; Bose, Roy, & Bandyopadhyay,
2012; Henkel et al., 2013; Polo-Corrales, Latorre-Esteves, & Ramirez-
Vick, 2014; Ranganathan, Ferrari, & Decuzzi, 2013; Vaquette et al.,
2012). Skeletal development and fracture repair include the coordina-
tion of migration, differentiation, and activation of multicell types and
tissues (Dirckx, Van Hul, & Maes, 2013; Ivkovic et al., 2003; Kanczler &
Received: 7 June 2019 Revised: 27 November 2019 Accepted: 29 November 2019
DOI: 10.1002/jbm.a.36857
J Biomed Mater Res. 2019;111. wileyonlinelibrary.com/journal/jbma © 2019 Wiley Periodicals, Inc. 1
Oreffo, 2008; Ozasa et al., 2014). Currently, autografts are widely
used to treat defected bone in clinics. However, it has some draw-
backs, like limited availability and the risk of second surgery (Rabkin,
Reid, & Doty, 2015; Temponi et al., 2017; Yanamadala et al., 2017).
Therefore, scaffolds arise as a viable alternative for bone tissue engi-
neering to treat bone-related diseases (Hollister, 2005; Murphy,
O'Brien, Little, & Schindeler, 2013; Saranya, Saravanan, Moorthi,
Ramyakrishna, & Selvamurugan, 2011; Wang et al., 2007).
Currently, numerous tissue engineering techniques have been
used to produce three-dimensional (3D) scaffolds for the defective
bone (Jones et al., 2009). The application of 3D scaffolds offers tem-
porary supports for cell growth and new tissue development. When
we choose bone graft scaffolds to treat fractures in human body, the
desired physical and chemical characteristic of scaffolds must be con-
sidered such as biocompatibility, biodegradability, and nontoxicity
(Chen et al., 2013; Hoornaert, d'Arros, Heymann, & Layrolle, 2016;
Mistry, Mikos, & Jansen, 2007; Thi Hiep et al., 2017). Collagen is the
most renowned material used in biomaterial research (Behring, Junker,
Walboomers, Chessnut, & Jansen, 2008; Dong-Soo et al., 2015;
Elango et al., 2016; Inzana et al., 2014). Collagen comprises the mate-
rial of bone, teeth, and connected tissue and can be extracted from
porcine, duck's feet and human tissues (Kim et al., 2016; Song et al.,
2016). The unique collagen properties include prominent mechanical
properties. Hyper-elastic behavior similar to natural human tissue,
facilitate and encourage cell adhesion, nontoxic, biocompatible, and
biodegradable. Collagen is considered as one of the best useful bioma-
terials. Collagen can be prepared in sponge's foams, nanofibrous pow-
ders, viscous solution, and dispersions (Kook et al., 2017), in this
study, collagen is extracted from duck's feet and sponges were pre-
pared. Usually, duck's feet are discarded as byproducts, and they are
very cheap when used commercially. When comparing the character-
istics of collagen extracted from commercially available animals, there
is no significant difference (El-Jawhari, Sanjurjo-Rodríguez, Jones, &
Giannoudis, 2016; Sultan et al., 2018).
HAp is one of the main inorganic components of bone, with a lot
of application in the field of bone tissue engineering owing to its excel-
lent biocompatibility, osteoconductivity, and biointegration properties.
HAp has attracted considerable attention as a bone substitute (Ambre,
Katti, & Katti, 2015; Fu, Rahaman, Dogan, & Bal, 2008; Michel, Penna,
Kochen, & Cheung, 2015). HAp helps cell adhesion, proliferation due
to its surface roughness, and has been known to enhance osteoblast
adhesion, proliferation, and differentiation (Chen et al., 2013).
Quercetin (Qtn) is flavonoid materials, and it induced mineral depo-
sition and also enhances osteoblast response of calcium phosphate
(Forte et al., 2016; Gupta, Kumar, & Mishra, 2017; Wong & Rabie,
2008a; Zhou et al., 2015). Qtn has been reported to favor adhesion
and proliferation of osteoblast by positively interacting with them and
effectively slowing-down or inhibiting the osteoclast activity (Kanter,
Altan, Donmez, Ocakci, & Kartal, 2007; Liang et al., 2011; Napimoga
et al., 2013; Siddiqui et al., 2011; Wong, 2011; Wong & Rabie, 2008b).
Therefore, if Qtn and Hap are used in bone tissue engineering applica-
tions, it could activate the metabolism of bone cells and it will promote
osteoblast proliferation, differentiation, and mineralization.
Bone marrow mesenchymal stem cells (BMSCs) have shown great
potential in the clinical application upon activation by pharmacological or
biological means, result in improved bone healing by modulating their dif-
ferentiation into osteoblasts. In this study, unique collagen isolated from
DC which has superior mechanical properties compared with fish and
chicken collagen (Theng, Huda, Wariyah, Hashim, & Nik Muhammad,
2018), that makes it highly suitable for biomedical application and the col-
lagen fibers were used to create biocomposite material. In addition, they
were coated with Hap, an osteoinductive and osteoconductive material
that facilitates cell adhesion and proliferation, they are combined differ-
ent ratio of Qtn in our developed DC/HAp sponges. They prepared spon-
ges were physio chemically characterized for its mechanical behavior,
biocompatibility in an in vitro model by seeding BMSCs and further eval-
uated in in vivo rat as an animal model for the bone regeneration.
2|MATERIALS AND METHODS
2.1 |Materials
Duck's feet were purchased from Korean local market. Qtn, acetone,
NaOH, methanol, citric acid, chloroform, pepsin, and acetic acid were
obtained from Sigma-Aldrich. All other reagents used in this study
were of high-performance liquid chromatography grade.
2.2 |Collagen isolation from ducks feet
DC was extracted using our previously reported protocol (Kook et al.,
2017; Song et al., 2016). The percentage of DC to be used in this
study was selected from our earlier study (Kook et al., 2017; Song
et al., 2016), where we have found that 2% DC have good mechanical
and cell viability properties. Therefore, we fabricated 2% of DC. First,
iced duck's feet were washed with distilled water several times and
their feet were cut using a blade and then immersed in 70% alcohol to
disinfect for 3 hr. Followed by, immersing them in 0.5M NaOH solu-
tion for 24 hr to remove fat from the tissue by placing in stirring at
60 rpm. Later, it was washed for a moment with methanol:chloroform
in 3:1 ratio. For collagen extraction, the feet were submerged again in
5% citric acid for 24 hr. Later, they were treated with 3 g of pepsin for
48 hr. Following the acid treatment, the supernatant was centrifuged
at 12,000 rpm for 15 min. The precipitated collagen was treated with
alcohol and centrifuged again at 3500 rpm for 5 min. Subsequently,
precipitated collagens were stored at 80C for 24 hr. Then, the
extracted collagen was freeze-dried for 5 days.
2.3 |Fabrication of Qtn/DC/HAp sponges
We dissolved collagen in 0.5M acetic acid to make 2% collagen solu-
tion followed by stirring for 72 hr at room temperature in 100 rpm.
Qtn was prepared in dimethyl sulfoxide (DMSO) under stirring at
60 rpm for 5 min at room temperature. To make a different ratio
2SONG ET AL.
(0, 25, 50, and 100 μM), Qtn solution was mixed into 2% collagen
solution. The mixture was then poured into 48 well plates (1 ml/well),
freeze at 80C, later freeze-dried. Afterward, the Qtn/DC/HAp
sponges were prepared following the methods: briefly, 2% DC or
Qtn/DC sponges were immersed in 1X stimulated body fluid (SBF) for
24 hr at room temperature, then washed with distilled water and
lyophilized for 48 hr. In this study, DC/HAp sponges and a different
ratio of Qtn/DC/HAp were referred as 0 μM Qtn/DC/HAp, 25 μM
Qtn/DC/HAp, 50 μM Qtn/DC/HAp, and 100 μM Qtn/DC/HAp. All
these characterization were carried out using the dry samples.
2.4 |Sponge characterizations
The surface morphology and pore size of sponges were examined by
scanning electron microscopy (SEM; Bio-LV SEM, Hitachi, S-2250N,
Japan). The chemical properties of the sponges were analyzed by Fou-
rier transform infrared spectroscopy (FTIR; Spectrum GX, Perkin Elmer).
The compressive mechanical strength of each sponge was measured
using TMS-Pro instrument (Food Technology Corporation, Sterling,
VA), by moving the samples down at a target distance to specimen of
1.5 mm with a speed of 10 mm/s at a force of 0.5N for calculating the
strength and height of the sponges. Porosity was evaluated by Archi-
medes principles via fluid displacement measurement techniques.
Sponges were immersed in a known volume of water (V
1
) for 10 min.
Then the total water volume along with the immersed sponge was
referred to as V
2
. Then the water impregnated sponges were removed,
and the remaining volume of water was recorded as V
3
. The porosity of
sponges was estimated by using the following equation.
Porosity %ðÞ=V1V3
V2V3
×100
2.5 |Cell culture
BMSCs were isolated from the femurs and tibias of 6 weeks old
female New Zealand white rabbits. Isolated femurs and tibias moved
into the clean bench and washed three times with phosphate buffered
saline (PBS). After cutting both ends of the bone, the bone marrow
was isolated using a 20-G needle. Then bone marrow was incubated
in culture plate containing Alpha-MEM (Lonza, Walkersville, MD) sup-
plemented with 20% fetal bovine serum, and 1% penicillin/streptomy-
cin (Invitrogen, Carlsbad, CA). After 3 hr, bone marrow was
resuspended using 100 μm cell strainer in the medium. Isolated
BMSCs were plated in the culture medium at 37Cin5%CO
2
. The
culture medium was replaced once in every 3 days.
2.6 |In vitro cell adhesion and proliferation
The surface morphology and the initial attachment of BMSCs on the
sponges were observed by SEM (S-2250N, Hitachi, Tokyo, Japan).
BMSCs were seeded on the sponges at a concentration of
5×10
4
cells/sponge (n= 3). After 21 days of culture, the cultured
sponges were washed with PBS and fixed using 2.5% glutaraldehyde
(Sigma-Aldrich, Saint Louis) at room temperature for 24 hr. Fixed sam-
ples were dehydrated in ethanol graded series (50, 60, 70, 80, 90, and
100%) for 30 min each. Dried samples were coated with gold using a
plasma sputter under argon gas. The cell morphology on sponge was
observed by SEM.
The cell viability and proliferation on the sponges was measured
using MTT (3-4-2, 5-diphenyl tetrazolium bromide, Sigma-Aldrich)
assay. The density of BMSC per sponge was seeded at 5 ×10
4
. After
1, 7, 14, and 21 days of culture, 100 μl of MTT solution was added on
the sponges. Then it was incubated for 4 hr at 37C in 0.5% CO
2
.
After the formation of purple crystals, the solution was removed,
sponges were carefully washed with PBS followed by addition of 1 ml
DMSO and incubated for 24 hr. Then 100 μl of the solution was
transferred into 96-well plate, and the absorbance was measured at
570 nm using Synergy Mx monochromator-based multimode micro-
plate reader (Biotek Instuments, Inc.).
2.7 |Alkaline phosphatase activity
The osteogenic differentiation of BMSCs on the sponges was ana-
lyzed using alkaline phosphatase (ALP) Assay Kit (Takara Bio Inc.,
Tokyo, Japan) according to the manufacturer's instruction. BMSCs
were seeded on the sponges at the concentration of 5 ×10
4
cells/
sponge (n= 3) and cultured for 1, 7, 14, and 21 days. At each time
point, the samples were retrieved and rinsed using PBS. The protein
from each BMSCs seeded on the sponges were extracted using 5%
ALP extraction solution and para-nitrophenyl phosphate (pNPP) solu-
tion was added. The reaction solution incubated at 37Cin5%CO
2
for 1 hr. The reaction was stopped by adding 0.9N NaOH. The absor-
bance was measured using a microplate reader at 405 nm.
2.8 |Gene expression using real-time polymerase
chain reaction
Osteogenic gene expression was evaluated using real-time polymer-
ase chain reaction (RT-PCR). On 1, 7, 14, and 21 days of BMSCs
(5 ×10
4
) cultured on sponges (n= 3), the total RNA was extracted
using a Trizol reagent (Takara Bio, Inc., Tokyo, Japan) and 0.2 ml chlo-
roform. The supernatants were precipitated using 0.5 ml of isopropanol
(Sigma-Aldrich). Each of primers; glyceraldehyde 3-phosphate dehydro-
genase (GAPDH), type-I collagen (COLI), runt-related transcription fac-
tor 2 (Runx2), and osteocalcin (OCN) were purchased from Genotec
(Daejeon, Korea) and extended using TOPscriptOne-step RT PCR
DryMIX (Enzynomics, Korea) by Authorized Thermal Cycler (TP 600,
Takara Bio, Inc., Japan). PCR products were examined by electrophore-
sis on 1.2% (wt/vol) agarose gel containing ethidium bromide and visu-
alized under UV light (FluorChem HD2 Gel Imaging System, Alpha
Innotech) at 300 nm.
SONG ET AL.3
2.9 |In vivo bone regeneration-micro-CT
The sponges were cut into 0.5 cm thick and 4 mm diameter of
round shapes. Two pieces of 0.5 cm ×4 mm calvarial bone defects
were carved off from each spraguedawley rats (SD rat, 6 weeks,
female, Hanil laboratory animal center, Wanju, Korea). Scaffolds
were implanted and fixed with silicone caps (Summit medical;
n= 2). Then, the wounds were sutured (Ethicon) to protect the
implanted sites from external contamination. The in vivo minerali-
zation and compatibility of 0 μM Qtn DC/HAp and 25 μM
Qtn/DC/HAp sponges were evaluated by micro-CT (SkyScan 1173,
SkyScan, Belgium) after 2 and 8 weeks of implantation. The 3D
images of calvarial defects implanted sponges were built from cor-
onal images to evaluate bone formation in the defects. We ana-
lyzed bone volume (BV), and bone mineral density (BMD), through
μ-CT analysis.
2.10 |Histological evaluation
To evaluate the bone regeneration of 0 μM Qtn DC/HAp and 25 μM
Qtn/DC/HAp, sponges were seeded with BMSCs at 2 ×10
5
(cells/
sponge), and sponges were implanted at the calvarial defected site of
SD rat with 4 mm biopsy punch (n= 2). Histological evaluation of SD
rat cranial section was performed after 8 weeks postsurgery. Sponges
along with the surrounding tissues were collected and fixed using
10% formaldehyde (Sigma-Aldrich), and dehydrated with a series of
graded ethanol, and embedded in paraffin blocks, sectioned into
8 mm. Then samples were deparaffinized and stained with Hematoxy-
lin and Eosin (H&E) and Masson's trichrome stain (MTS). All the animal
experimental procedures were performed with the approval from the
Chonbuk National University Animal Care Committee, Jeonju, South
Korea.
2.11 |Statistical analysis
The data were expressed as means ± standard deviation (SD) of exper-
iments performed in triplicate (n= 3). Statistical significance was ana-
lyzed with one-way analysis of variance using GraphPad Prism 5.0
(San Diego, CA). Probability (p) values of <.05(*), <.01(**), and <.001
(***) were considered as statistically significant.
3|RESULTS
3.1 |Morphology of Qtn/DC/HAp sponges
SEM analysis was used to observe the surface morphology of the
scaffold and the pore size that is shown in Figure 1, showing the gross
images of 0, 25, 50, and 100 μM Qtn/DC/HAp sponges (photographic
images of the scaffolds are presented as an insert in Figure 1). All the
sponges presented smooth, soft surface, and sponge-like morphology
with the 2 mm thickness and 6 mm wide length. Also, all sponge's
pore shape showed uniform elliptical shapes in the cross-sectional
images. All the scaffold had a pore structure with well-oriented pores
that are enclosed by a thin wall from the sponge surface to inside,
the majority of the pores observed are of uniform size. This special
orientation of the scaffolds structure helps cells moving inside the
pores of the scaffold and form adhesion and proliferation. For an ideal
biomaterial scaffold must possess a suitable 3D structure and pore
size for the nutrients transport and prevent cell loss.
3.2 |Physiochemical properties
Figure 2a FTIR spectra illustrated the change in the chemical structure
of Qtn, hydroxyapatite, and different concentration of Qtn/DC/HAp
FIGURE 1 Scanning electron microscopy images of 0, 25, 50, and 100 μM Qtn/DC/HAp sponges showing in ×100 and their imagined images
at ×200 (Insert shows the gross images of sponges)
4SONG ET AL.
sponges. DC spectrum displays characteristic peaks at 1645 cm
1
(amide-I; C O stretch), 1561 cm
1
(amide-II; N H bend), 1469 cm
1
(C H bond), and 1249 cm
1
(amide-III; C N stretch), respectively.
The peaks at 1740 and 1680 cm
1
corresponded to the stretch vibra-
tions of C O and C C groups. Qtn spectrum at ~3422 cm
1
cor-
responded to the stretch vibration of the O H bond. Also, at 1663,
1609, 1574, and 1523 cm
1
corresponding to its benzene ring and
CO group. Hydroxyapatite spectrum showed PO
4
3
at 1033 and
571 cm
1
. The peaks at 3400 cm
1
corresponded to O H. In the
spectra of the different ratios of Qtn/DC/HAp sponges, the 100 μM
Qtn/DC/HAp sponge exhibited comparatively higher intensity at the
same absorption bands that were observed in Qtn spectrum. More-
over, the absorption peaks at 3325, 1642 cm
1
were observed in the
Qtn/DC/HAp sponge due to O H and C O stretch by the interac-
tion of Qtn and collagen, and the effect of hydroxyapatite was rela-
tively small. This sponge was coated with SBF solution by immersing
the sponge for 1 day instead of direct addition of hydroxyapatite and
was expected to have low absorption. We confirmed Qtn, HAp, and
DC peaks in the fabricated Qtn/DC/HAp. And the Qtn peak intensity
was increased at higher concentration of Qtn. So it is considered that
all sponges have unique properties of Qtn, DC, and HAp during the
production process. Mechanical strength of the scaffold plays a vital
role in bone tissue engineering. Figure 2b shows the compressive
strength of the fabricated sponges. The compressive strength was
found to 0.178 ± 0.02, 0.173 ± 0.02, 0.150 ± 0.02, and 0.122
± 0.01 MPa for 0, 25, 50, and 100 μM Qtn/DC/HAp sponges, respec-
tively. The compressive strength of the scaffolds is observed to be
decreasing with increasing concentration of Qtn in the scaffold. Scaf-
fold containing 0% Qtn had the highest compressive strength 0.178
± 0.02 MPa compared with other scaffolds but they have the lowest
porosity, pore size. Figure 2c presents the porosity of the 0, 25,
50, and 100 μM Qtn/DC/HAp sponges. We observed that the porosi-
ties of all groups of Qtn/DC/HAp sponges were over 70%. The poros-
ities of 0, 25, 50, and 100 μM Qtn/DC/HAp sponges were 71.36
± 1.93, 73.46 ± 1.28, 76.36 ± 5.14, and 80.90 ± 4.89%, respectively.
Most of the pores were of uniform size.
3.3 |Cell proliferation and adhesion of BMSCs on
Qtn/DC/HAp sponges
To further study the biocompatibility, cell adhesion, proliferation, and
cytotoxicity of the sponges, SEM analysis was carried out to confirm
the cell adherence and morphology of BMSC in Qtn/DC/HAp spon-
ges. As a result, BMSCs were attached to the surface of all sponges in
a round shape, and the extracellular matrix was observed to extend
like a spider web (Figure 3). The images clearly showed cells randomly
distributed on the surface and inside the walls of the sponge pores.
In order to evaluate the effect of Qtn on cell growth and their
toxicity, we performed MTT assay during 21 days in different time
point (1, 7, 14, and 21 days) by culturing BMSCs on 0, 25, 50, and
100 μM Qtn/DC/HAp sponges. OD values of all the three different
groups showed not much statistically significant difference (Figure 4a)
over time compared with the control groups without Qtn, the prolifer-
ation of BMSCs seeded on the Qtn/DC/HAp sponges gradually
increased. All of the experimental groups showed similar proliferation
on day 1.
3.4 |Osteogenic differentiation on Qtn/DC/HAp
sponges
The previous experimental results confirmed that the prepared scaf-
fold possessed good physiochemical properties with good biocompati-
bility. Further, study to evaluate its ability to promote osteogenic
differentiation of BMSCs in vitro model was carried out. The osteo-
genic differentiation of BMSCs on Qtn/DC/HAp sponges was evalu-
ated by culturing BMSCs on the sponges for 21 days and they were
analyzed on different time point (1, 7, 14, and 21 days). BMSCs grown
in osteoblast-like environments undergo osteoblast differentiation
into osteoblasts cells. ALP activity is an important analysis carried out
for evaluating osteoblast differentiation. In general, unlike BMSCs, dif-
ferentiated osteoblasts have ALP that hydrolyzes other substances to
produce phosphoric acid. The BMSCs present in the Qtn/DC/HAp
FIGURE 2 (a) Fourier transform infrared spectroscopy spectra, (b) Compressive strength, and (c) Porosity of 0, 25, 50, and 100 μM Qtn/DC/
HAp sponges (n=3,**p< .005)
SONG ET AL.5
sponge was evaluated by measuring ALP activity using the fact that
the number of BMSCs differentiated into osteoblasts was increased
thereby increasing ALP activity with the same trend. Figure 4b shows
that BMSCs in the Qtn/DC/HAp sponge gradually increased osteo-
blast differentiation. A total of 25 μM Qtn/DC/HAp sponge had the
highest ALP value compared with other sponges studied at all-time
point.
3.5 |mRNA expression of BMSCs on Qtn/DC/
HAp sponges
To analyze the ability of osteogenic differentiation of BMSCs on
0, 25, 50, and 100 μM Qtn/DC/HAp sponges, RT-PCR was performed
(Figure 5). To study, whether the sponges affected the osteogenic dif-
ferentiation of rBMSCs, the expressions of bone-specific genes such
as OCN, COL1, and RUNX-2 were examined on 21 days of cell culture
(GAPDH was used as a housekeeping gene). Runx2 and COL1 are
considered as early markers of osteogenic differentiation, whereas
OCN is middle and late markers (Komori, 2010). Comparing the gene
expression on the different concentration of the sponges analyzed,
25 μM Qtn/DC/HAp sponges real-time quantitative values for COL1
(1.05-fold), OCN (1.22-fold), and Runx2 (1.11-fold) showed higher
expression. These results suggested that 25 μM Qtn/DC/HAp possess
stronger stimulating effects on osteogenic differentiation than 0 μM
Qtn /DC/HAp as a control.
3.6 |Sponge scaffold improved bone regeneration
in vivo
To investigate osteoconduction ability of the Qtn/DC/HAp sponges,
sponges were implanted in a full thickness rat calvarial bone defect.
All rats survived the surgery and recovered well postoperatively,
FIGURE 3 SEM images of BMSCs cultured on the surface of 0, 25, 50, and 100 μM Qtn/DC/HAp sponges over a period of 21 days, showing
enhanced cell attachment and proliferation (magnifications with ×500 and ×1000). BMSCs, bone marrow mesenchymal stem cells; SEM, scanning
electron microscopy
FIGURE 4 Biocompatibility and osteogenic differentiation of BMSCs cultured on 0, 25, 50, and 100 μM Qtn/DC/HAp sponges for 1, 7,
14, and 21 days studied by (a) MTT and (b) ALP assay (*p< .05, **p< .005, ***p< .001). ALP, alkaline phosphatase; BMSCs, bone marrow
mesenchymal stem cells; MTT, 3-4-2, 5-diphenyl tetrazolium bromide
6SONG ET AL.
without infection. All the animals were sacrificed on 8 weeks of post-
surgery for further examination. Micro-CT images were performed to
analyze the new bone formation in the defected region (Figure 6). In
the control group, the newly formed bones appeared a little around
the hole. At 8 weeks postsurgery, the BMD of the rate on the
25 μM Qtn/DC/HAp group was almost 3.65 fold higher than con-
trol groups and 2.5 fold higher than 0 μM Qtn/DC/HAp group
showing the significantly largest area of new bone regeneration. BV
(mm
3
) on the 0 μM Qtn/DC/HAp group showed a slight increase
(2 weeks: 0.12 ± 2.1 mm
3
, 8 weeks: 0.76 ± 0.32 mm
3
). While BV on
the 25 μM Qtn/DC/HAp sponge showed a significant increase
(2 weeks: 2.92 ± 0.99 mm
3
, 8 weeks: 3.92 ± 1.63 mm
3
). These
results indicate the 25 μM Qtn/DC/HAp sponge could stimulate
new bone formation.
3.7 |Histological analysis
To evaluate bone regeneration in the rat calvarial defect model,
12 SD-rats were sacrificed at 8 weeks postsurgery. H&E staining rev-
eled significantly more new calcified bone formation in implanted
defected models and inside the porous scaffold. In the H&E staining,
the blank group with no sponge implanted in the defective part no vis-
ible newly produced bone were seen, and the group with 0 μM
Qtn/DC/HAp inserted into the defective part and the group with
25 μM Qtn/DC/HAp, it was observed and confirmed that the sponge
pores were filled with a pale red color. In particular, this new bone for-
mation was found to be regenerated significantly higher at 25 μM
Qtn/DC/HAp sponge compared with 0 μM Qtn/DC/HAp sponge.
MTS staining showed new bone (red), collagen sponge (blue), and
FIGURE 5 mRNA expression of BMSCs bone-specific markers analyzed from the BMSCs cultured on 0, 25, 50, and 100 μM Qtn/DC/HAp
sponges for 21 days. (a) RT-PCR results of glyceraldehyde 3-phosphate dehydrogenase (GAPDH), type-I collagen (COLI), osteocalcin (OCN), and
runt-related transcription factor 2 (Runx2), (b) quantitative analysis of COL1, OCN, and Runx2 were normalized using GAPDH (**p< .005,
***p< .001). BMSCs, bone marrow mesenchymal stem cells; RT-PCR, real-time polymerase chain reaction
FIGURE 6 Micro-CT images of SD-Rat cranial new bone regeneration after implantation at 2 and 8 weeks: (a) Micro-CT images and (b) Bone
mineral density (BMD) and (c) Bone volume (BV; ***p< .001)
SONG ET AL.7
tissue (blue) in part of the implanted area in 0 μM Qtn/DC/HAp and
25 μM Qtn/DC/HAp sponges (Figure 7). When comparing blank with
0μM Qtn/DC/ HAp and 25 μM Qtn/DC/HAp in MT staining, it was
observed that only the tissue was filled with the bone defects. The
0μM Qtn/DC/HAp sponge confirmed that the new bone tissue
appeared as a red color between the collagen sponge pores and that
the 25 μM Qtn/DC/HAp sponge had more new bones between the
sponge pores and the red color part was more distributed than
DC/HAp sponges.
4|DISCUSSION
In bone tissue engineering, cells cultured with a specific scaffold/
sponge/biomaterial to construct composite materials to repair bone
defects have achieved initial success. There is an urgent need for
developing suitable scaffolds with good physiochemical, nontoxic, and
biocompatible properties for bone tissue engineering applications. An
ideal bone scaffold/sponge should possess biocompatibility, optimal
mechanical properties, and good morphology for cell adhesion, prolif-
eration, and ensuring the integration of the scaffold with bone tissue
(Ho-Shui-Ling et al., 2018). For tissue bone engineering sponge/scaf-
fold, minimal porosity, appropriate pore size, and well interconnecting
pores are very important for ingrowth, cell adhesion, proliferation, and
metabolic transport of the cells (Gomes, Bossano, Johnston, Reis, &
Mikos, 2006; Przekora, 2019). In our present study, 25 μM Qtn/DC/
HAp sponge had well-interconnected pores and a porosity (73.46
± 1.28%), compared with cancellous bone having a porosity of
5090%. Considering that the porosity of the cancellous bone was
reported to be >75% (Polo-Corrales, Latorre-Esteves, & Ramirez-Vick,
2014; Uth, Mueller, Smucker, & Yousefi, 2017), our finding was so
close to the cancellous bone supporting that it can be used for tissue
engineering application. This uniform pore shape was formed through
the 3 hr refrigeration, freeze-drying method. The pore size of each
sponge was increased by the increasing content of Qtn. The reason
for the different pore size of each sponge is due to DMSO, the sol-
vent used to dissolve Qtn. It affects the ice particle size during
freeze-dry.
The change in functional groups was confirmed by the FTIR spec-
trum. The spectrum clearly shows the respective peaks of Qtn, DC,
and HAp, and their intensity was observed to be increasing with the
increasing concentration of Qtn in the sponge materials. We found
the addition of Qtn slightly decreased the compressive strength of the
sponges. It is known that DMSO enhanced the hydrogen bonding of
water to strengthen the binding force between the molecules of colla-
gen (Ho-Shui-Ling et al., 2018; Parimaladevi & Srinivasan, 2016) nev-
ertheless, as the concentration of Qtn increased, the compressive
strength of the sponge decreased slightly. This indicates that the pore
size has a larger influence on the compressive strength than the
above-mentioned bond between collagen molecules. The porosity
was increased with the increasing concentration of Qtn in the sponge.
These results are also owing to the influence of the pore size.
The MTT assay was performed to evaluate the proliferation and
biocompatibility of the sponges by seeding BMSCs on the surface of
the materials in vitro. The activity of the cells (live cells) presented
through observance reading of the different groups on several time
points. On day 1, all the groups showed similar proliferative activities
with no significant difference. On day 7, there was a significant differ-
ence in proliferative activities between the groups. We observed a
decrease in cell proliferative activities on increasing in Qtn concentra-
tions. But over time, among the four groups, 25 μM Qtn/DC/HAp
sponges exhibited the highest proliferation rate. Compared with
BMSCs cultured without sponges in cell culture plates, the cell prolif-
erative activities of BMSCs cultured on the sponges was not
FIGURE 7 Histological evaluation of 0 and 25 μM Qtn/DC/HAp sponges implanted calvarial defected SD-Rat for 8 weeks (Red arrow
indicates the edge of the defect, magnification ×40)
8SONG ET AL.
decreased and after day 7 compared with control, the cell proliferative
activities was observed to be increasing in 25 μM Qtn/DC/HAp sam-
ples, the same trend was followed till day 21. However, cell prolifera-
tion was inhibited with the increase of Qtn concentration. Since Qtn
inhibits cell proliferation at higher concentrations, different mecha-
nisms have been proposed, including cell cycle arrest, DNA strand
breakage, induction of apoptosis, and possibly influencing the activity
of various kinases including phosphatidylinositol-3-kinase, tyrosine
protein kinase, and protein kinase C (van der Woude et al., 2005).
Thus, it is considered that the higher amount of Qtn is suppressed cell
proliferation in the DC/HAp sponges. SEM images showed BMSCs to
be well adhered and proliferated and integrated within the pores
sponge.
In vitro ALP activities of the 25 μM, Qtn/DC/HAp sponge con-
firmed a stimulatory effect on the differentiation of osteoblasts. Also,
when the content of Qtn was increased in the sponge, the degree of
bone differentiation gradually decreased. These results suggest that
the Qtn/DC/HAp sponge with Qtn tends to inhibit BMSC prolifera-
tion by Qtn, but the present BMSCs further promote bone differentia-
tion into osteoblasts. Stem cells were optimized in 25 μM Qtn/DC/
HAp sponge. This suggests that Qtn reduced osteoclastic differentia-
tion by RANKL, and osteoblastic differentiation is indirectly induced
for that reason, thus promoting bone morphogenesis of bone
marrow-derived stem cells in the sponge (Wattel et al., 2004). The
gene expression results revealed among the sponge analyzed 25 μM
Qtn/DC/HAp sponge showed better results. These results indicated
that Qtn could enhance osteogenic differentiation at various stages,
additionally, the concentration of 25 μM Qtn/DC/HAp was found to
be the optimum concentration for COL1, Runx2, and OCN expression.
Micro-CT results revealed that the implanted 0 μM Qtn /DC/HAp and
25 μM Qtn/DC/HAp sponges with rBMSCs promotes more bone
regeneration than the blank group. These results indicated that the
density and area of tissue regenerated in the 25 μM Qtn/DC/HAp
sponge were the highest, and their osteogenic activity was strong.
The results confirm the integration between the surrounding tissues
and implanted sponges. Histological analysis results confirmed that
compared with control the 25 μM Qtn/DC/HAp sponge treated
enhanced defect closure and mineralization and the analysis comple-
mented the Micro-CT results, this suggests that Qtn has an effect on
the osteogenic differentiation of BMSC, resulted in an increase in
bone regeneration.
5|CONCLUSION
In this study, we fabricated porous composite Qtn/DC/HAp sponges
by a freeze-drying process with the aim of biomaterial for bone regen-
eration. We found that 25 μM Qtn/DC/HAp sponge is the most effi-
cient promoter of BMSCs osteogenic differentiation with
osteoinductive and osteoconductive features as evidenced by the
in vitro and in vivo outcomes without any negative effects on rats. In
spite of fulfilling all the desired properties of an ideal tissue engineer-
ing scaffold, Qtn/DC/HAp sponges has, 3D structure with pore size
for the nutrients transport, preventing cell loss, biocompatible, non-
toxic, good morphology for cell adhesion, proliferation, and ensuring
the integration of the scaffold with bone tissue. Thus, this bio-
engineered Qtn/DC/HAp sponge might be applicable for bone regen-
eration in larger animals and future clinical trials. In conclusion, the
proper amount of the Qtn incorporated with collagen sponges may
price clinical support for the patients with various bone defects and
might be applied in several tissue engineering fields.
ACKNOWLEDGMENTS
This research was supported by a grant of the Korea Health Technol-
ogy R&D Project (HI15C2996) through the Korea Health Industry
Development Institute (KHIDI), the International Research & Develop-
ment Program of the National Research Foundation of Korea (NRF)
funded by the Ministry of Science, ICT & Future Planning (NRF-
2017K1A3A7A03089427).
REFERENCES
Ambre, A. H., Katti, D. R., & Katti, K. S. (2015). Biomineralized hydroxyapa-
tite nanoclay composite scaffolds with polycaprolactone for stem cell-
based bone tissue engineering. Journal of Biomedical Materials Research
Part A,103(6), 20772101.
Amini, A. R., Laurencin, C. T., & Nukavarapu, S. P. (2012). Bone tissue engi-
neering: Recent advances and challenges. Critical Reviews in Biomedical
Engineering,40(5), 363408.
Behring, J., Junker, R., Walboomers, X. F., Chessnut, B., & Jansen, J. A.
(2008). Toward guided tissue and bone regeneration: Morphology,
attachment, proliferation, and migration of cells cultured on collagen
barrier membranes. A systematic review. Odontology,96(1), 111.
Bose, S., Roy, M., & Bandyopadhyay, A. (2012). Recent advances in
bone tissue engineering scaffolds. Trends in Biotechnology,30(10),
546554.
Chen, L., Zhu, W.-M., Fei, Z.-Q., Chen, J.-L., Xiong, J.-Y., Zhang, J.-F.,
Zeng, Y. (2013). The Study on Biocompatibility of Porous nHA/PLGA
Composite Scaffolds for Tissue Engineering with Rabbit Chondrocytes
In Vitro. BioMed Research International,2013,6.
Dirckx, N., Van Hul, M., & Maes, C. (2013). Osteoblast recruitment to sites
of bone formation in skeletal development, homeostasis, and regener-
ation. Birth Defects Research. Part C, Embryo Today,99(3), 170191.
Dong-Soo, P., Jung-Chul, P., Jung-Seok, L., Tae-Wan, K., Ki-Joon, K.,
Byung-Joo, J., ChangSung, K. (2015). Effect of FGF-2 on collagen
tissue regeneration by human vertebral bone marrow stem cells. Stem
Cells and Development,24(2), 228243.
Elango, J., Zhang, J., Bao, B., Palaniyandi, K., Wang, S., Wenhui, W., &
Robinson, J. S. (2016). Rheological, biocompatibility and osteogenesis
assessment of fish collagen scaffold for bone tissue engineering. Inter-
national Journal of Biological Macromolecules,91,5159.
El-Jawhari, J. J., Sanjurjo-Rodríguez, C., Jones, E., & Giannoudis, P. V.
(2016). Collagen-containing scaffolds enhance attachment and prolif-
eration of non-cultured bone marrow multipotential stromal cells.
Journal of Orthopaedic Research,34(4), 597606.
Forte, L., Torricelli, P., Boanini, E., Gazzano, M., Rubini, K., Fini, M., &
Bigi, A. (2016). Antioxidant and bone repair properties of quercetin-
functionalized hydroxyapatite: An in vitro osteoblastosteoclast
endothelial cell co-culture study. Acta Biomaterialia,32, 298308.
Fu, Q., Rahaman, M. N., Dogan, F., & Bal, B. S. (2008). Freeze-cast
hydroxyapatite scaffolds for bone tissue engineering applications. Bio-
medical Materials,3(2), 025005.
Gomes, M. E., Bossano, C. M., Johnston, C. M., Reis, R. L., & Mikos, A. G.
(2006). In vitro localization of bone growth factors in constructs of
biodegradable scaffolds seeded with marrow stromal cells and
SONG ET AL.9
cultured in a flow perfusion bioreactor. Tissue Engineering,12(1),
177188.
Gupta, S. K., Kumar, R., & Mishra, N. C. (2017). Influence of quercetin and
nanohydroxyapatite modifications of decellularized goat-lung scaffold
for bone regeneration. Materials Science and Engineering: C,71,
919928.
Henkel, J., Woodruff, M. A., Epari, D. R., Steck, R., Glatt, V.,
Dickinson, I. C., Hutmacher, D. W. (2013). Bone regeneration based
on tissue engineering conceptionsA 21st century perspective. Bone
Res,1(3), 216248.
Hollister, S. J. (2005). Porous scaffold design for tissue engineering. Nature
Materials,4(7), 518524.
Hoornaert, A., d'Arros, C., Heymann, M.-F., & Layrolle, P. (2016). Biocom-
patibility, resorption and biofunctionality of a new synthetic biode-
gradable membrane for guided bone regeneration. Biomedical
Materials,11(4), 045012.
Ho-Shui-Ling, A., Bolander, J., Rustom, L. E., Johnson, A. W.,
Luyten, F. P., & Picart, C. (2018). Bone regeneration strategies:
Engineered scaffolds, bioactive molecules and stem cells current stage
and future perspectives. Biomaterials,180, 143162.
Inzana, J. A., Olvera, D., Fuller, S. M., Kelly, J. P., Graeve, O. A.,
Schwarz, E. M., Awad, H. A. (2014). 3D printing of composite cal-
cium phosphate and collagen scaffolds for bone regeneration. Bioma-
terials,35(13), 40264034.
Ivkovic, S., Yoon, B. S., Popoff, S. N., Safadi, F. F., Libuda, D. E.,
Stephenson, R. C., Lyons, K. M. (2003). Connective tissue growth
factor coordinates chondrogenesis and angiogenesis during skeletal
development. Development,130(12), 27792791.
Jones, A. C., Arns, C. H., Hutmacher, D. W., Milthorpe, B. K.,
Sheppard, A. P., & Knackstedt, M. A. (2009). The correlation of pore
morphology, interconnectivity and physical properties of 3D ceramic
scaffolds with bone ingrowth. Biomaterials,30(7), 14401451.
Kanczler, J. M., & Oreffo, R. O. (2008). Osteogenesis and angiogenesis: The
potential for engineering bone. European Cells & Materials,15,100114.
Kanter, M., Altan, M. F., Donmez, S., Ocakci, A., & Kartal, M. E. (2007). The
effects of quercetin on bone minerals, biomechanical behavior, and
structure in streptozotocin-induced diabetic rats. Cell Biochemistry and
Function,25(6), 747752.
Kim, S. H., Park, H. S., Lee, O. J., Chao, J. R., Park, H. J., Lee, J. M.,
Park, C. H. (2016). Fabrication of duck's feet collagensilk hybrid bio-
material for tissue engineering. International Journal of Biological Mac-
romolecules,85, 442450.
Komori, T. (2010). Regulation of osteoblast differentiation by Runx2.
Advances in Experimental Medicine and Biology,658,4349.
Kook, Y. J., Lee, D. H., Song, J. E., Tripathy, N., Jeon, Y. S., Jeon, H. Y.,
Khang, G. (2017). Osteogenesis evaluation of duck's feet-derived
collagen/hydroxyapatite sponges immersed in dexamethasone. Bioma-
terials Research,21(1), 2.
Liang, W., Luo, Z., Ge, S., Li, M., Du, J., Yang, M., Luo, Z. (2011). Oral
administration of quercetin inhibits bone loss in rat model of diabetic
osteopenia. European Journal of Pharmacology,670(1), 317324.
Michel, J., Penna, M., Kochen, J., & Cheung, H. (2015). Recent advances in
hydroxyapatite scaffolds containing mesenchymal stem cells. Stem
Cells International,2015, 13.
Mistry, A. S., Mikos, A. G., & Jansen, J. A. (2007). Degradation and biocom-
patibility of a poly(propylene fumarate)-based/alumoxane
nanocomposite for bone tissue engineering. Journal of Biomedical
Materials Research Part A,83A(4), 940953.
Murphy, C. M., O'Brien, F. J., Little, D. G., & Schindeler, A. (2013). Cell-
scaffold interactions in the bone tissue engineering triad. European
Cells & Materials,26, 120132.
Napimoga, M. H., Clemente-Napimoga, J. T., Macedo, C. G., Freitas, F. F.,
Stipp, R. N., Pinho-Ribeiro, F. A., Verri, W. A., Jr. (2013). Quercetin
inhibits inflammatory bone resorption in a mouse periodontitis model.
Journal of Natural Products,76(12), 23162321.
Ozasa, Y., Gingery, A., Thoreson, A. R., An, K. N., Zhao, C., & Amadio, P. C.
(2014). A comparative study of the effects of growth and differentia-
tion factor 5 on muscle-derived stem cells and bone marrow stromal
cells in an in vitro tendon healing model. The Journal of Hand Surgery,
39(9), 17061713.
Parimaladevi, P., & Srinivasan, K. (2016). Understanding the crystallization
behavior of α-lactose monohydrate (α-LM) through molecular interac-
tion in selected solvents and solvent mixtures under different growth
conditions. CrystEngComm,18(13), 23122318.
Polo-Corrales, L., Latorre-Esteves, M., & Ramirez-Vick, J. E. (2014). Scaf-
fold design for bone regeneration. Journal of Nanoscience and Nano-
technology,14(1), 1556.
Przekora, A. (2019). The summary of the most important cell-biomaterial
interactions that need to be considered during in vitro biocompatibility
testing of bone scaffolds for tissue engineering applications. Materials
Science and Engineering: C,97, 10361051.
Rabkin, D. G., Reid, B. B., & Doty, J. R. (2015). Acute on chronic pulmonary
autograft dissection. Interactive Cardiovascular and Thoracic Surgery,20
(4), 563564.
Ranganathan, S. I., Ferrari, M., & Decuzzi, P. (2013). Design maps for scaf-
fold constructs in bone regeneration. Biomedical Microdevices,15(6),
10051013.
Saranya, N., Saravanan, S., Moorthi, A., Ramyakrishna, B., &
Selvamurugan, N. (2011). Enhanced osteoblast adhesion on polymeric
nano-scaffolds for bone tissue engineering. Journal of Biomedical Nano-
technology,7(2), 238244.
Siddiqui, J. A., Swarnkar, G., Sharan, K., Chakravarti, B., Gautam, A. K.,
Rawat, P., Dwivedi, A. K. (2011). And others. A naturally occurring
rare analog of quercetin promotes peak bone mass achievement and
exerts anabolic effect on osteoporotic bone. Osteoporosis International,
22(12), 30133027.
Song, J. E., Lee, S. E., Cha, S. R., Jang, N. K., Tripathy, N., Reis, R. L., &
Khang, G. (2016). Inflammatory response study of gellan gum impreg-
nated duck's feet derived collagen sponges. Journal of Biomaterials Sci-
ence, Polymer Edition,27(15), 14951506.
Sultan, M. T., Jeong, J. Y., Seo, Y. B., Lee, O. J., Ju, H. W., Park, H. J.,
Park, C. H. (2018). Fabrication and characterization of the porous
duck's feet collagen sponge for wound healing applications. Journal of
Biomaterials Science, Polymer Edition,29(79), 960971.
Temponi, E. F., Camelo, N., Tuteja, S., Thaunat, M., Daggett, M.,
Fayard, J. M., Sonnery-Cottet, B. (2017). Reconstruction of chronic
patellar tendon rupture with contralateral bone-tendon-bone auto-
graft. Knee Surgery, Sports Traumatology, Arthroscopy,25(8),
24682473.
Theng, C., Huda, N., Wariyah, C., Hashim, H., & Nik Muhammad, N. A.
(2018). Physicochemical properties of duck feet collagen with different
soaking time and its application in Surimi. International Journal on
Advanced Science, Engineering and Information Technology,8, 832841.
Thi Hiep, N., Chan Khon, H., Dai Hai, N., Byong-Taek, L., Van Toi, V., &
Thanh, H. L. (2017). Biocompatibility of PCL/PLGA-BCP porous scaf-
fold for bone tissue engineering applications. Journal of Biomaterials
Science. Polymer Edition,28(9), 864878.
Uth, N., Mueller, J., Smucker, B., & Yousefi, A. M. (2017). Validation of
scaffold design optimization in bone tissue engineering: finite element
modeling versus designed experiments. Biofabrication,9(1), 015023.
van der Woude, H., ter Veld, M. G. R., Jacobs, N., van der Saag, P. T.,
Murk, A. J., & Rietjens, I. M. C. M. (2005). The stimulation of cell prolif-
eration by quercetin is mediated by the estrogen receptor. Molecular
Nutrition & Food Research,49(8), 763771.
Vaquette, C., Fan, W., Xiao, Y., Hamlet, S., Hutmacher, D. W., &
Ivanovski, S. (2012). A biphasic scaffold design combined with cell
sheet technology for simultaneous regeneration of alveolar
bone/periodontal ligament complex. Biomaterials,33(22), 55605573.
Wang, H., Li, Y., Zuo, Y., Li, J., Ma, S., & Cheng, L. (2007). Biocompatibility
and osteogenesis of biomimetic nano-hydroxyapatite/polyamide
10 SONG ET AL.
composite scaffolds for bone tissue engineering. Biomaterials,28(22),
33383348.
Wattel, A., Kamel, S., Prouillet, C., Petit, J.-P., Lorget, F., Offord, E., &
Brazier, M. (2004). Flavonoid quercetin decreases osteoclastic differ-
entiation induced by RANKL via a mechanism involving NFκB and AP-
1. Journal of Cellular Biochemistry,92(2), 285295.
Wong, R. W. K. (2011). Re: Effect of Quercetin on bone formation: JOR
25:10611066, 2008 and other publications reporting the effects of
various materials on bone formation in rabbit Calvarial defects. Journal
of Orthopaedic Research,29(2), 157157.
Wong, R. W. K., & Rabie, A. B. M. (2008a). Effect of quercetin on bone for-
mation. Journal of Orthopaedic Research,26(8), 10611066.
Wong, R. W. K., & Rabie, A. B. M. (2008b). Effect of quercetin on
preosteoblasts and bone defects. The Open Orthopaedics Journal,2,
2732.
Yanamadala, V., Rozman, P. A., Kumar, J. I., Schwab, J. H., Lee, S. G.,
Hornicek, F. J., & Curry, W. T., Jr. (2017). Vascularized fibular strut
autografts in spinal reconstruction after resection of vertebral
Chordoma or Chondrosarcoma: A retrospective series. Neurosurgery,
81(1), 156164.
Zhou, Y., Wu, Y., Jiang, X., Zhang, X., Xia, L., Lin, K., & Xu, Y. (2015). The
effect of Quercetin on the Osteogenesic differentiation and angio-
genic factor expression of bone marrow-derived Mesenchymal stem
cells. PLoS One,10(6), e0129605.
How to cite this article: Song JE, Tian J, Kook YJ,
Thangavelu M, Choi JH, Khang G. A BMSCs-laden quercetin/
duck's feet collagen/hydroxyapatite sponge for enhanced
bone regeneration. J Biomed Mater Res. 2019;111. https://
doi.org/10.1002/jbm.a.36857
SONG ET AL.11
... In addition, Jeong Eun Song et al. incorporated quercetin into collagen/hydroxyapatite sponge to enhance bone metabolism and osteogenic differentiation of the scaffold. The results showed that the prepared sponge had good compressive strength and high porosity and could significantly increase the expression of COL-1, OCN, and Runx-2, promoting the repair of rat skull defects [100]. R.W.K. Wong et al. used collagen sponge loaded with naringin, quercetin, or puerarin to treat full-thickness parietal bone defects in rabbits. ...
Article
Full-text available
Skeletons play an important role in the human body, and can form gaps of varying sizes once damaged. Bone defect healing involves a series of complex physiological processes and requires ideal bone defect implants to accelerate bone defect healing. Traditional grafts are often accompanied by issues such as insufficient donors and disease transmission, while some bone defect implants are made of natural and synthetic polymers, which have characteristics such as good porosity, mechanical properties, high drug loading efficiency, biocompatibility and biodegradability. However, their antibacterial, antioxidant, anti-inflammatory and bone repair promoting abilities are limited. Flavonoids are natural compounds with various biological activities, such as antitumor, anti-inflammatory and analgesic. Their good anti-inflammatory, antibacterial and antioxidant activities make them beneficial for the treatment of bone defects. Several researchers have designed different types of flavonoid-loaded polymer implants for bone defects. These implants have good biocompatibility, and they can effectively promote the expression of angiogenesis factors such as VEGF and CD31, promote angiogenesis, regulate signaling pathways such as Wnt, p38, AKT, Erk and increase the levels of osteogenesis-related factors such as Runx-2, OCN, OPN significantly to accelerate the process of bone defect healing. This article reviews the effectiveness and mechanism of biomaterials loaded with flavonoids in the treatment of bone defects. Flavonoid-loaded biomaterials can effectively promote bone defect repair, but we still need to improve the overall performance of flavonoid-loaded bone repair biomaterials to improve the bioavailability of flavonoids and provide more possibilities for bone defect repair.
... In view of the aforementioned, optimization of synthetic bone substitutes with nano-formulated QT can represent a good approach to locally aid bone regeneration in bone fractures. Previous in vitro studies proved a successful incorporation of QT into ceramic bone substitutes [14,[22][23][24]. To our knowledge it is the first time to incorporate QT loaded NLCs within CPC. ...
Article
The prevalence of bone injuries is greatly increasing each year and the proper healing of fractures without any complications is very challenging. Self-setting calcium phosphate cements (CPCs) have attracted great attention as bioactive synthetic bone substitutes. Quercetin (QT) is a multipurposed drug with reported bone-conserving properties. The loading of QT and QT-phospholipid complex within nanostructured lipid carriers (NLC) was proposed to overcome the poor physical properties of the drug and to introduce the use of bioactive excipients as phospholipids and olive oil. The aim of this work was to formulate a regenerative scaffold loaded with nano-formulated QT for local treatment of orthopedic fractures. For the first time, scaffolds composed of brushite CPC were prepared and loaded with quercetin lipid nano-systems. In vitro tests proved that the addition of lipid nano-systems did not deteriorate the properties of CPC where QT-NLC/CPC showed an adequate setting time, appropriate compressive strength, and porosity. The scanning electron microscope confirmed maintenance of nanoparticles integrity within the cement. Using a rat femur bone defect animal model, the histological results showed that the QT-NLC/CPC had a superior bone healing potential compared to crude unformulated QT/CPC. In conclusion, QT-NLC /CPC are promising lipid nano-composite materials that could enhance bone regeneration.
Article
The purpose of this research was to observe the clinical effect of iRoot-BP combined with collagen sponge for the treatment of periapical inflammation with lesions larger than 10 mm and undeveloped apical foramen. 26 teeth from 25 patients with periapical inflammation meeting the inclusion criteria were randomly assigned into two groups with 13 teeth per group. In collagen sponge group, the apical barrier was performed using iRoot-BP combined with collagen sponge, and in non-collagen sponge group was only treated with iRoot-BP. Patients in two groups were followed up at 3, 6 and 12 months respectively, and the therapeutic effect was judged according to the clinical and imaging findings. The prognosis of periapical lesions indicated that there were 13 of treated teeth in collagen sponge group and 11 of treated teeth in non-collagen sponge group show no signs of clinical or radiographic failure ( χ ² = 2.167, p = 0.14). The observation results of continued root growth suggested that the average root growth of collagen group was 2 mm, while no continuous root growth was observed in non-collagen sponge group ( p < 0.05). Application of iRoot-BP combined with collagen sponge has obvious advantages in treatment of large-scale periapical inflammation with undeveloped apical foramen, which can promote the formation of immature roots closer to the normal roots.
Article
Nanostructured inorganic biomaterial emerged as the most essential platform to address traumatic and non-traumatic conditions of hard tissues in the current scenario. Synthetic inorganic biomaterials serve as an efficient and pathogen-free choice that overcomes the obstructions associated with autografts and allografts to promote new tissue regeneration, since nano-hydroxyapatite (nHAp) is a biomaterial that mimics the natural mineral composition of bones and teeth of human hard tissues, which is widely employed in orthopedics and dentistry. The nHAp-based materials exhibit bioactive, biocompatible, and osteoconductive features under in vitro and in vivo conditions. The brittle nature of synthetic nHAp leads to weak mechanical properties, which eventually confines the utility of nHAp in load-bearing applications. Hence, this review focuses on the recent trends in the fabrication and investigation of nHAp-based polymer nanocomposite scaffolds for bone regeneration. Employing different polymers and fabrication strategies would efficiently tailor the physicochemical properties, and tailor-made mechanical properties in competence with biodegradation, thereby enhancing their potential in biomedical utility, and exploring their efficacy under in vitro and in vivo conditions to make “HAp-based smart-biomaterials” for bone tissue engineering.
Article
Full-text available
Bone defects are a persistent challenge in clinical practice. Although repair therapies based on tissue-engineered materials, which are known to have a crucial role in defective bone regeneration, have gathered increased attention, the current treatments for massive bone defects have several limitations. In the present study, based on the immunomodulatory inflammatory microenvironment properties of quercetin, we encapsulated quercetin solid lipid nanoparticles (SLNs) in a hydrogel. Temperature-responsive poly(ε-caprolactone-co-lactide)-b-poly(ethylene glycol)-b-poly(ε-caprolactone-co-lactide) (PCLA) modifications were coupled to the main chain of hyaluronic acid (HA) hydrogel, constructing a novel, injectable bone immunomodulatory hydrogel scaffold. Extensive in vitro and in vivo data showed that this bone immunomodulatory scaffold forms an anti-inflammatory microenvironment by decreasing M1 polarization, while elevating the M2 polarization. Synergistic effects on angiogenesis and anti-osteoclastic differentiation were observed. These findings further proved that administering quercetin SLNs encapsulated in a hydrogel can aid bone defect reconstruction in rats, providing new insights for large-scale bone defect repair.
Article
Full-text available
Conventional intake of drugs and active substances is most often based on oral intake of an appropriate dose to achieve the desired effect in the affected area or source of pain. In this case, controlling their distribution in the body is difficult, as the substance also reaches other tissues. This phenomenon results in the occurrence of side effects and the need to increase the concentration of the therapeutic substance to ensure it has the desired effect. The scientific field of tissue engineering proposes a solution to this problem, which creates the possibility of designing intelligent systems for delivering active substances precisely to the site of disease conversion. The following review discusses significant current research strategies as well as examples of polymeric and composite carriers for protein and non-protein biomolecules designed for bone tissue regeneration.
Article
Collagen is commonly used as a regenerative biomaterial due to its excellent biocompatibility and wide distribution in tissues. Different kinds of hybridization or cross-links are favored to offer improvements to satisfy various needs of biomedical applications. Previous reviews have been made to introduce the sources and structures of collagen. In addition, biological and mechanical properties of collagen-based biomaterials, their modification and application forms, and their interactions with host tissues are pinpointed. However, there is still no review about collagen-based biomaterials for tissue engineering. Therefore, we aim to summarize and discuss the progress of collagen-based materials for tissue regeneration applications in this review. We focus on the utilization of collagen-based biomaterials for bones, cartilages, skin, dental, neuron, cornea, and urological applications and hope these experiences and outcomes can provide inspiration and practical techniques for the future development of collagen-based biomaterials in related application fields. Moreover, future improving directions and challenges for collagen-based biomaterials are proposed as well.
Article
Full-text available
Duck feet are considered as waste products because people normally do not consume duck feet. It is highly available in Malaysia and thus has the high potential to be the alternative source to produce collagen other than fish, bovine or porcine. The main objective of this study is to evaluate the physicochemical properties of different treated duck feet collagen and its effect on the quality of sardine surimi. Duck feet was stirred and soaked in 5% lactic acid solution for different time (12, 24, 36 and 48 h) at 4ºC. Another treatment for the duck feet is demineralization. Duck feet collagen was demineralized by 0.5M EDTA solution for 24 h prior soaking in lactic acid solution for 24 h. Soaking time 24, 36 and 48 h has successfully decreased fat content of duck feet collagen significantly compared to 12 h. However, no differences in yield were observed for the samples. Prolong soaking time produced duck feet collagen with higher content of imino acid (hydroxyproline + proline). The intensity of lightness (L*) for the demineralized duck feet collagen is lower compared to duck feet collagen which was not treated by EDTA. Duck feet collagen has the ability to improve the quality of sardine surimi gel by enhancing its lightness, water holding capacity, gel strength and texture with low cooking loss and expressible moisture. The folding test score of sardine surimi gel has been increased from 3.00 to 5.00 by the addition of duck feet collagen. The gel strength for sardine surimi gel with the addition of duck feet collagen is in the range of 2625.90 g.mm to 2681.58 g.mm, which is approximately 9 times higher than the gel strength of sardine surimi without collagen (280.43 g.mm).
Article
Full-text available
There are several artificial dermis commonly use to cover the wound and promote healing. The major goal of wound management is fast and scarless healing. However, there is no ideal skin substitute, that is effective to accelerate wound healing without scar formation. Artificial dermis substitute also has some drawbacks, such as high cost, insufficient available period and donor pathogen infection. To overcome these problems, we developed duck's feet collagen (DFC) sponge as artificial dermal substitutes for the treatment of full-thickness skin defects. We measured these DFC sponge's comparative characteristics and performances with an artificial dermis Colladerm by carried out SEM-EDX analyze, water-binding abilities and porosity test. Biocompatibility test was also performed using CCK-8 cytotoxicity assay. We also evaluated its wound healing effects for a full-thickness skin wound and compared with Colladerm in a rat model. Histological studies were carried via H&E and MT staining. Although the wound healing effect of the DFC sponge was almost similar to that of Colladerm, the DFC sponge did not induce scar formation and wound contracture like Colladerm. We suggest that DFC sponge can be used as an ideal dermal substitute to the treatment of full-thickness skin wound.
Article
Full-text available
Background The aim of this study was to investigate the osteogenesis effects of DC and DC/HAp sponge immersed in without and with dexamethasone. Methods The experimental groups in this study were DC and DC/HAp sponge immersed in without dexamethasone (Dex(−)DC and Dex(−)-DC/HAp group) and with dexamethasone (Dex(+)-DC and Dex(+)-DC/HAp group). We characterized DC and DC/HAp sponge using compressive strength, scanning electron microscopy (SEM). Also, osteogenic differentiation of BMSCs on sponge (Dex(−)DC, Dex(−)-DC/HAp, Dex(+)-DC and Dex(+)-DC/HAp group) was assessed by SEM, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazoliumbromide (MTT) assay, alkaline phosphatase (ALP) activity assay and reverse transcription-PCR (RT-PCR). ResultsIn this study, we assessed osteogenic differentiation of BMSCs on Duck’s feet-derived collagen (DC)/HAp sponge immersed with dexamethasone Dex(+)-DC/HAp. These results showed that Dex(+)-DC/HAp group increased cell proliferation and osteogenic differentiation of BMSCs during 28 days. Conclusion From these results, Dex(+)-DC/HAp can be envisioned as a potential biomaterial for bone regeneration applications.
Article
Full-text available
This study reports the development of biological/synthetic scaffolds for bone tissue engineering via 3D bioplotting. These scaffolds were composed of poly(L-lactic-co-glycolic acid) (PLGA), type I collagen, and nano-hydroxyapatite (nHA) in an attempt to mimic the extracellular matrix of bone. The solvent used for processing the scaffolds was 1,1,1,3,3,3-Hexafluoro-2-propanol (HFP). The produced scaffolds were characterized by scanning electron microscopy, microcomputed tomography, thermogravimetric analysis, and unconfined compression test. This study also sought to validate the use of finite-element optimization in COMSOL Multiphysics for scaffold design. Scaffold topology was simplified to three factors: nHA content, strand diameter, and strand spacing. These factors affect the ability of the scaffold to bear mechanical loads and how porous the structure can be. Twenty four scaffolds were constructed according to an I-optimal, split-plot designed experiment (DE) in order to generate experimental models of the factor-response relationships. Within the design region, the DE and COMSOL models agreed in their recommended optimal nHA (30%) and strand diameter (460 μm). However, the two methods disagreed by more than 20% in strand spacing (923 μm for DE; 601 μm for COMSOL). Seven scaffolds were 3D-bioplotted to validate the predictions of DE and COMSOL models (4.5 - 9.9 MPa measured moduli). The predictions for these scaffolds showed relative agreement for scaffold porosity (mean absolute percentage error of 4% for DE and 13% for COMSOL), but were substantially poorer for scaffold modulus (52% for DE; 21% for COMSOL), partly due to some simplifying assumptions made by the models. Expanding the design region in future experiments (e.g., higher nHA content and strand diameter), developing an efficient solvent evaporation method, and exerting a greater control over layer overlap could allow developing PLGAnHA- collagen scaffolds to meet the mechanical requirements for bone tissue engineering.
Article
Full-text available
Membranes for guided bone regeneration (GBR) were prepared from the synthetic biodegradable polymer poly-D,L-lactic/glycolic acid (PLGA). This GBR membrane has a bi-layered structure with a dense film to prevent gingival fibroblast ingrowth and ensure mechanical function, and a micro-fibrous layer to support colonization by osteogenic cells and promote bone regeneration. Hydrolysis and biodegradation were both studied in vitro through soaking in phosphate buffered saline (PBS) and in vivo by implantation in the subcutis of rats for 4, 8, 16, 26, 48 and 52 weeks. Histology revealed an excellent colonization of the micro-fibrous layer by cells with a minimal inflammatory reaction during resorption. GBR using the synthetic PLGA membrane was evaluated on critical-size calvaria defects in rats for 4 and 8 weeks. Radiographs, micro-computed tomography and histology showed bone regeneration with the PLGA membrane, while the defects covered with a collagen membrane showed a limited amount of mineralized bone, similar to that of the defect left empty. The biofunctionality of the PLGA membranes was also compared to collagen membranes in mandible defects in rabbits, associated or not with beta-tricalcium phosphate granules. This study revealed that the bi-layered synthetic membrane made of PLGA was safer, more biocompatible, and had a greater controlled resorption rate and bone regeneration capacity than collagen membranes. This new PLGA membrane could be used in pre-implantology and peri-odontology surgery.
Article
Tissue engineered products (TEPs), which mean biomaterials containing either cells or growth factors or both cells and growth factors, may be used as an alternative to the autografts taken directly from the bone of the patients. Nevertheless, the use of TEPs needs much more understanding of biointeractions between biomaterials and eukaryotic cells. Despite the possibility of the use of in vitro cellular models for initial evaluation of the host response to the implanted biomaterial, it is observed that most researchers use cell cultures only for the evaluation of cytotoxicity and cell proliferation on the biomaterial surface, and then they proceed to animal models and in vivo testing of bone implants without fully utilizing the scientific potential of in vitro models. In this review, the most important biointeractions between eukaryotic cells and biomaterials were discussed, indicating molecular mechanisms of cell adhesion, proliferation, and biomaterial-induced activation of immune cells. The article also describes types of cellular models which are commonly used for biomaterial testing and highlights the possibilities and drawbacks of in vitro tests for biocompatibility evaluation of novel scaffolds. Finally, the review summarizes recent findings concerning the use of adult mesenchymal stem cells for TEP generation and compares the potential of bone marrow- and adipose tissue-derived stem cells in regenerative medicine applications.
Article
Bone fractures are the most common traumatic injuries in humans. The repair of bone fractures is a regenerative process that recapitulates many of the biological events of embryonic skeletal development. Most of the time it leads to successful healing and the recovery of the damaged bone. Unfortunately, about 5-10% of fractures will lead to delayed healing or non-union, more so in the case of co-morbidities such as diabetes. In this article, we review the different strategies to heal bone defects using synthetic bone graft substitutes, biologically active substances and stem cells. The majority of currently available reviews focus on strategies that are still at the early stages of development and use mostly in vitro experiments with cell lines or stem cells. Here, we focus on what is already implemented in the clinics, what is currently in clinical trials, and what has been tested in animal models. Treatment approaches can be classified in three major categories: i) synthetic bone graft substitutes (BGS) whose architecture and surface can be optimized; ii) BGS combined with bioactive molecules such as growth factors, peptides or small molecules targeting bone precursor cells, bone formation and metabolism; iii) cell-based strategies with progenitor cells combined or not with active molecules that can be injected or seeded on BGS for improved delivery. We review the major types of adult stromal cells (bone marrow, adipose and periosteum derived) that have been used and compare their properties. Finally, we discuss the remaining challenges that need to be addressed to significantly improve the healing of bone defects.
Article
In this study, biomimic porous polycaprolactone/poly (lactide-co-glycolide) loading biphasic tricalcium phosphate (PCL/PLGA-BCP) scaffolds were fabricated successfully by solvent evaporation method. The distribution of biphasic tricalcium phosphate (BCP) in polycaprolactone/poly (lactide-co-glycolide) (PCL/PLGA) scaffold was confirmed by micro-computed tomography (micro-CT) scanning, scanning electron microscope (SEM) observation and Energy-dispersive X-ray Spectroscopy (EDS) analysis. The hydrophilicity of the scaffolds was confirmed by contact angle measurement. In in vitro experiments, proliferation of human bone marrow mesenchymal stem cell (hBMSCs) and its osteoblastic differentiation on scaffold were assessed for 1, 2 and 3 weeks using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, fluorescence observation, hematoxylin & eosin (H&E) staining and real-time polymerase chain reaction (RT-PCR). In in vivo experiments, ossification was observed using micro-CT analysis and histological staining.
Article
Background: Margin-free en bloc resection is the best medical practice for primary vertebral chordoma and chondrosarcoma. Spinal reconstruction following total spondylectomy requires reconstructive interbody graft (allograft, devascularized autograft, vascularized autograft, or cage constructs) and instrumentation. An important consideration when choosing grafts and instrumentation is the durability and the long-term success of the fusion without subsidence. Objective: To evaluate the potential use of vascularized fibular autograft as a reconstructive strategy after en bloc resection. Methods: We present a series of 16 patients who underwent spondylectomy for primary vertebral chordoma or chondrosarcoma with reconstruction using a vascularized fibular autograft and anterior/posterior instrumentation between January 2011 and April 2014. We report postoperative neurological outcome, 6-mo rates of fusion and graft subsidence, and other complications. Results: Two patients passed away prior to 6-mo follow-up, and 1 patient was lost to follow-up. The mean follow-up time for the remaining 13 patients was 32 mo. Of these patients, 9 (69%) had evidence of fusion on the 6-mo follow-up computed tomography (CT) scan. Of the 4 patients who did not fuse, 2 had undergone surgery for new tumor diagnoses, 1 for hardware failure, and 1 for graft nonunion. Two patients (15%) had eventual graft subsidence along with hardware failure. Conclusions: Vascularized fibular strut grafts are a viable method for reconstruction following spondylectomy. We present the largest series of patients to date utilizing this technique. Further comparative studies examining vascularized grafts vs nonvascularized grafts or metallic cage constructs will be important in choosing the best reconstructive strategy.
Article
In the present study, goat-lung scaffold was fabricated by decellularization of lung tissue and verified for complete cell removal by DNA quantification, DAPI and H&E staining. The scaffold was then modified by crosslinking with quercetin and nanohydroxyapatite (nHAp), and characterized to evaluate the suitability of quercetin-crosslinked nHAp-modified scaffold for regeneration of bone tissue. The crosslinking chemistry between quercetin and decellularized scaffold was established theoretically by AutoDock Vina program (in silico docking study), which predicted multiple intermolecular hydrogen bonding interactions between quercetin and decellularized scaffold, and FTIR spectroscopy analysis also proved the same. From MTT assay and SEM studies, it was found that the quercetin-crosslinked nHAp-modified decellularized scaffold encouraged better growth and proliferation of bone-marrow derived mesenchymal stem cells (BMMSCs) in comparison to unmodified decellularized scaffold, quercetin-crosslinked decellularized scaffold and nHAp-modified decellularized scaffold. Alkaline Phosphatase (ALP) assay results showed highest expression of ALP over quercetin-crosslinked nHAp-modified scaffold among all the tested scaffolds (unmodified decellularized scaffold, quercetin-crosslinked decellularized scaffold and nHAp-modified decellularized scaffold) indicating that quercetin and nHAp is very much efficient in stimulating the differentiation of BMMSCs into osteoblast cells. Alizarin red test quantified in vitro mineralization (calcium deposits), and increased expression of alizarin red over quercetin-crosslinked nHAp-modified scaffold indicating better stimulation of osteogenesis in BMMSCs. The above findings suggest that quercetin-crosslinked nHAp-modified decellularized goat-lung scaffold provides biomimetic bone-like microenvironment for BMMSCs to differentiate into osteoblast and could be applied as a potential promising biomaterial for bone regeneration.