You are on page 1of 9

Ceramics International 49 (2023) 22961–22969

Contents lists available at ScienceDirect

Ceramics International
journal homepage: www.elsevier.com/locate/ceramint

Osseointegration and anti-infection of dental implant under osteoporotic


conditions promoted by gallium oxide nano-layer coated titanium dioxide
nanotube arrays
Litao Yao a, b, c, 1, Abdullrahman M. Al-Bishari a, 1, Jiating Shen a, Zhen Wang a, Tingting Liu a,
Lieping Sheng b, Gang Wu c, Lei Lu a, *, Lihua Xu d, **, Jinsong Liu a
a
School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
b
Department of Dentistry, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China
c
Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam
(VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands
d
Department of General Medicine, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China

A R T I C L E I N F O A B S T R A C T

Handling Editor: Dr P. Vincenzini The insufficient osteointegration between titanium (Ti) based dental implants and surrounding bone tissue under
osteoporotic conditions, as well as implant-associated infections usually associate with dental implant failure.
Keywords: Gallium (Ga) has drawn increasing attention due to its antibacterial ability and pro-osteogenic property. Herein,
Gallium oxide we established a facile strategy to deposit gallium oxide (Ga2O3) nano-layer on titanium dioxide nanotube (TNT)
Nanotube
arrays by magnetron sputtering. Both Ga2O3@TNT/50 W and Ga2O3@TNT/100 W specimens were prepared at
Titanium
different powers (50 W and 100 W), respectively, which all achieved a sustained release of Ga3+ ion for over 14
Magnetron sputtering
Antibacterial days. In vitro results showed that Ga2O3@TNT/50 W sample has good biocompatibility and osteogenic activity,
Osteointegration compared with TNT and Ga2O3@TNT/100 W samples. Moreover, both Ga2O3@TNT/50 W and Ga2O3@TNT/
100 W samples possess potent antibacterial properties, due to the released Ga3+ ion. Furthermore, in vivo results
indicated that the Ga2O3@TNT/50 W showed excellent in vivo antibacterial ability and promoted new bone
formation effectively compared with TNT implants in an osteoporotic rat model with Staphylococcus aureus
(S. aureus) infection. Therefore, such Ga2O3 deposition coating strategy provides a valuable guidance for the
potential future development of Ga-doped dental implants and TNT based drug loading surfaces for clinical
applications.

1. Introduction mechanical properties, and corrosion resistance [4]. However, the po­
tential infection and insufficient bone formation capability limits their
Osteoporosis is a common systemic disease affects the elderly, clinical performances [5,6]. The bone defects around dental implants
especially postmenopausal women. It is characterized by destruction of caused by osteoporosis could affect the bone integration of Ti implant
the microstructure of bone tissue, decreased of bone mass and density and lead to implant failure. Another major risk is bacterial colonization
[1,2]. With the condition of osteoporosis, the absorption and atrophy of and infection associated peri-implantitis, a common postoperative
alveolar bone on the upper and lower jaw was enhanced, resulting in a complication which usually results in implant failure [7]. Hence, it is
reduced thickness of bone trabecular and increased separation of bone essential to endow Ti with good antibacterial property as well as
trabecula [3]. anti-osteoclastic property [8,9].
Pure titanium (Ti) and its alloys are still the “gold standards” for For decades, highly ordered nanotubes with desired diameters on the
dental implant materials because of their excellent biocompatibility, Ti and its alloy surface based on cost-effective electrochemical anodic

* Corresponding author.
** Corresponding author.
E-mail addresses: llu2@foxmail.com (L. Lu), lihuaxu@wmu.edu.cn (L. Xu).
1
Co-first authors: These authors contributed equally to this work.

https://doi.org/10.1016/j.ceramint.2023.04.121
Received 8 February 2023; Received in revised form 12 April 2023; Accepted 14 April 2023
Available online 14 April 2023
0272-8842/© 2023 Elsevier Ltd and Techna Group S.r.l. All rights reserved.
L. Yao et al. Ceramics International 49 (2023) 22961–22969

oxidation, namely TiO2 nanotubes (TNT), exhibit good biocompatibility, Magnetron sputtering is also a conventional metallic coating method
and has been reported to be conducive to the adhesion, proliferation, for Ti and its alloys, which can also directly form evenly distributed
differentiation and mineralization of osteoblasts [10]. Besides, TNT can nano-scaled particles inside or on the outer edge of the TNTs [15,32,33].
also be used for loading and release various drugs and bioactive com­ The thin-film coating prepared by magnetron sputtering has the char­
pounds such as bioactive metal ions and functional peptides, to enhance acteristics of uniform deposition, uniform thickness, and high bonding
the therapeutic effect of implants [11–14]. Owing to its inherent strength. Herein, the electrochemical anodic oxidation and magnetron
biocompatibility and drug-loading/local release function, TNTs has sputtering were used to modify the Ti surface. The Ga2O3 coated TNT
received increasing attention as an ideal surface for commercialized drug loading system (Ga2O3@TNT) was constructed by combining
orthopedic and dental implant [15,16]. The key developmental chal­ Ga2O3 with TNT via magnetron sputtering. Furthermore, Ga2O3 reduced
lenges for the long term success of TNT based dental implants in clinical the diameter of TNT, promoting sustained release of Ga3+ ions, which
dentistry are simultaneously ensuring the osseointegration while has both antibacterial and osteogenic properties (Fig. 1).
improving the long-term immunomodulation, local antibacterial prop­
erty and minimized cytotoxicity [16,17]. Various bioactive metallic ions 2. Materials and methods
incorporated biomaterials has enhanced interest in specific biomedical
applications including anti-inflammation, anti-infection, enhancing tis­ 2.1. Sample preparation
sue regeneration and angiogenesis for the last few decades [18,19].
Gallium (Ga), a semi-metallic element that widely used in semi­ Pure Ti samples (99.99%) were laser cut into sizes of 10 mm × 10
conductor industry, has shown therapeutic effects towards various dis­ mm × 0.5 mm, polished with sandpaper of 300, 500, 1000 grits. The
orders, such as diagnostic and therapeutic in cancer like cancer of the TNT samples were prepared through anodization at an operating voltage
lymph system, disorders of calcium and accelerated bone resorption of 20 V for 1 h at 37 ◦ C. The anode of the DC power supply was con­
[20–22]. Ga3+ ions share certain similarities with Fe3+ ions, which nected with a pure Ti sheet fixed by an electrode clamp, and the cathode
competes with Fe3+ in iron-binding proteins and enzymes, and disrupts was connected with a pure platinum sheet. Then, TNT was coated with
their functions in cells. For bacteria, which highly depends on the Fe3+ Ga2O3 through magnetron sputtering. The control power was 50 W and
mediated metabolic and signaling, resulting in inhibition of several 100 W for 25 min, respectively, the argon pressure was constant at 0.5
iron-dependent redox pathways [23]. Specifically, Ga compounds have Pa, the distance between TNT and the target was 10 cm, and the tem­
displayed anti-inflammatory and immunosuppressive, as well as the perature was constant at 40 ◦ C. After sputtering was over, the sample
multi-target antimicrobial properties against multidrug-resistant (MDR) was taken out, vacuum-dried, and stored.
pathogens which attracted attention of researchers to develop Ga based
non-antibiotic-dependent antibacterial strategies to combat drug resis­ 2.2. Surface characterization
tance [19,23–25]. Moreover, some studies have reported that Ga com­
pound has enhanced the effect on the activity of osteoblasts, which play The morphology of substrates was conducted using scanning electron
an essential role in bone remodeling [20]. Currently, intravenous gal­ microscopy (SEM, S-4800, Hitachi, Japan) and transmission electron
lium nitrate (Ga(NO₃)₃) is widely used in treatments of osteoporosis, microscopy (TEM, JEM-2100 F, JEOL, Japan). X-ray photoelectron
bone metabolic diseases, multiple myeloma, etc. [26–28], and was spectroscopy (XPS, Model PHI 5400, PerkinElmer, USA), and energy-
approved by the United States Food and Drug Administration (FDA) for dispersive X-ray spectroscope (EDS, S-4800, Hitachi, Japan), attenu­
clinical use in 2003 [23]. However, treatments with high dose of Ga ated total reflectance Fourier transform infrared spectrometer (ATR-
(NO₃)₃ was usually associated with potential clinical toxicities and side FTIR, Tensor II, Bruker, Germany) were used to identify chemical
effects. Recently, it has been reported that TNT loading with Ga(NO₃)₃ components. The phase composition and water contact angle were
successfully enhanced the anti-bacterial property and soft-tissue inte­ measured using an X-ray diffraction (XRD, D/Max 2500 PC, Rigaku,
gration of Ti surfaces in vitro [29,30]. In contrast to Ga(NO₃)₃, gallium Japan) and contact angle goniometer (SDC-200 S, Sindin, China),
oxide (Ga2O3) is a stable metallic compound with low solubility which respectively.
had been doped into bioactive glasses to improve the cancer inhibition
and osteointegration [25]. For example, a micro-arc oxidation (MAO) 2.3. The release profile of Ga3+
coating containing TiO2, Ga2O3 and tantalum oxide (Ta2O5) on the ti­
tanium surface showed excellent cyto-biocompatibility, osteogenic Five pieces of the Ga2O3 modified samples (Ga2O3@TNT/50 W and
bioactivity, and corrosion resistance [31]. Thus, we hypothesized that Ga2O3@TNT/100 W) were soaked in 15 mL of PBS solution, respec­
the sustained release of Ga3+ ions from the Ga2O3 modified TNT on the tively. The samples were fetched into a new PBS solution at 1-, 4-, 7-, and
surface of Ti implant could enhance its osteogenic and antibacterial 14-day time points. Finally, the collected liquid is accessed by induc­
effects. tively coupled plasma mass spectrometry (ICP-MS, Agilent 7500,

Fig. 1. Schematic illustration of the preparation of Ga2O3@TNT coatings with enhanced antibacterial and osteogenic properties, in vitro and in vivo.

22962
L. Yao et al. Ceramics International 49 (2023) 22961–22969

Agilent). internal reference gene for standardization were detected by SYBR


Premix ExTM TaqII kits (Takara, Kyoto, Japan). The primers are shown
2.4. In vitro osteogenesis assays in Table S1.

2.4.1. Cell culture 2.5. Antibacterial assays


Pre-osteoblastic MC3T3-E1 cells were used to evaluate the biocom­
patibility of materials and promote osteoblast function. The cells were Gram-positive Staphylococcus aureus (S. aureus) and gram-negative
cultured in an α-MEM medium, supplemented by 10% fetal bovine Escherichia coli (E. coli) were cultured onto substrates at an initial den­
serum (FBS) and 1% penicillin/streptomycin (P/S). MC3T3-E1 cells sity of 1 × 106 CFU/mL at 37 ◦ C before evaluation.
were stored at 37 ◦ C in a humidified incubator under 5% CO2.
2.5.1. Antibacterial rates
2.4.2. Cell morphology After culturing for 4 h and 24 h, the adherent bacteria were collected
Both SEM and fluorescence microscope (FM) was used for cell via a vibrate ultrasonication for 5 min. The diluted bacteria were then
morphology observation. After the MC3T3-E1 were cultured at density seeded onto sterile agar plates for 24 h, then observed and counted. The
(1 × 103 cells/well) for 3 days, different samples were fixed in 4% antibacterial rates were calculated as following:
paraformaldehyde. Samples were dehydrated by gradient ethanol,
C = (A − B)/A × 100%
sprayed with gold before SEM observation. For FM morphology obser­
vation, cells were stained by fluorescein isothiocyanate (FITC) labeled (C: antibacterial rate; A: average colony number of TNT group; B:
phalloidin and 4′ ,6-diamidino-2-phenylindole (DAPI), respectively average colony number of Ga2O3@TNT/50 W or Ga2O3@TNT/100 W
before analysis (OLYMPUS IX71, Japan). groups).

2.4.3. Cell proliferation 2.5.2. Live/dead staining of bacteria


After 1, 4, and 7 days of osteogenic induction at density of 2 × 104 After culturing for 4 h and 24 h, S. aureus and E. coli on different
cells/well with the α-MEM medium, the MC3T3-E1 cell viability on each samples were fixed with 4% paraformaldehyde and stained using a
sample was tested using the MTT assay. After gently washing with PBS, LIVE/DEAD Baclight™ Bacterial Viability Kit (thermos fisher, USA). The
300 μL of incomplete medium and 30 μL of MTT solution was added to stained bacteria were then observed using FM. Five visual fields were
each sample and incubated at 37 ◦ C for 4 h. Then, 1 mL of DMSO was randomly selected for fluorescence observation.
used to dissolve the formazan, and the optical density was measured
using a microplate reader (Bio-Rad 680, USA) at a wavelength of 490 2.5.3. Bacterial morphology
nm. After 24 h of incubation, S. aureus and E. coli on different samples
were fixed with 4% paraformaldehyde, and dehydrated by gradient
2.4.4. ALP activities ethanol solutions, sprayed with gold, and observed using SEM.
Samples were sterilized with 75% ethanol and ultraviolet disinfec­
tion. MC3T3-E1 cells were seeded on different substrates at density of 2 2.6. In vivo studies
× 104 cells/well. After 7 days of osteogenic induction, the cells cultured
on the sample were washed 3 times with PBS and fixed with 4% para­ 2.6.1. Preparation of osteoporotic rat bone defect model
formaldehyde for 40 min. Then samples were stained with BCIP/NBT Ovariectomy were performed to model osteoporosis as described
ALP color kit (Beyotime, China) for 40 min. Five random views were previously [1,2]. Briefly, female Sprague-Dawley (SD) rats (~300 g)
selected to photograph by using the stereomicroscope. were anesthetized by intraperitoneal injection of 1% chloral hydrate
For quantitative experiments, after 7 and 14 days of culturing, the prior to bilateral ovariectomy operations (OVX). After 3 months, the
cells on the surface of different samples were lysed by 1% Triton X-100 osteoporotic model was established. The bone density of rats was
on ice for 40 min of osteogenic induction, respectively. ALP activity was characterized by Micro-CT, tissue section analysis, etc. All animal pro­
evaluated using the ALP assay kit (Nаnjing Jiаncheng Bioengineering tocols were reviewed and approved by the Animal Ethics Committee of
Institute, China) and the BCA protein assay kit (Beyotime, China). Wenzhou Medical University followed the China animal management
Finally, the total intracellular protein content determined by the BCA and committee guidelines.
protein assay kit was used to standardize the ALP results.
2.6.2. Implantation surgery and infection model
2.4.5. Mineralization After intraperitoneal injection anesthesia with 10% chloral hydrate
MC3T3-E1 cells were seeded on different substrates with the α-MEM (3.4 mL/kg), OVX SD rats were randomly divided into two groups. TNT
medium at density of 2 × 104 cells/well. After 14 days for osteogenic and Ga2O3@TNT/50 W samples (diameter = 1 mm, length = 10 mm)
induction, alizarin red S was used to stain the fixed samples for 60 min. were inserted into the cylindrical holes through articular surface of the
Five random views were selected using the stereomicroscope to observe femur. S. aureus suspension (10 μL, 1*105 CFU/mL) carefully injected
the mineralized nodules. into the cavity to prepare an osteoporosis model with a bacterial
In the quantitative experiment, the mineralized nodules on the infection, and then muscle tissue and skin were sewed. For in vivo
sample were dissolved by 1 mL of cetylpyridinium chloride solution antibacterial analysis, Giemsa and HE staining of bone tissue was
(10%). And the absorbance of the resulting solution was measured by applied after 3 days of implantation.
using a microplate reader (Bio-Rad 680, USA) at a wavelength of 540
nm. 2.6.3. Micro-CT analysis
Samples were collected and fixed after 1 month of implantation. The
2.4.6. RT-PCR osteogenesis around implants in the OVX SD rats were observed via
MC3T3-E1 cells were seeded on different substrates with the α-MEM Micro-CT (Skyscan 1176, Bruker, Germany) scanning. New bone volume
medium at density of 3 × 104 cells/well. After 14 days of cell culture, the (mm2), bone connectivity density (Conn-Dens), trabecular width
RNAiso extraction kit is used to obtain total mRNA, and the RNA con­ (Tb⋅Th), and trabecular separation (Tb.Sp) were calculated.
centration was determined by Nano-drop 2000. Then, PrimeScriptTM
RT kit was used to reverse transcribe mRNA into cDNA. Bone formation- 2.6.4. Osteogenesis assessment in vivo
related genes ALP, OCN, OSX, OPG, RunX2, COL I, and GAPDH as the Samples were decalcified for 21 days using 12%

22963
L. Yao et al. Ceramics International 49 (2023) 22961–22969

ethylenediaminetetraacetic acid (EDTA, pH 7.4). After implants were 3. Results and discussion
carefully removed, samples were dehydrated in graded ethanol solutions
and embedded in paraffin, followed by hematoxylin and eosin (HE) and Magnetron sputtering technique has been widely used for thin-film
Masson’s trichrome (MT) staining. deposition associated applications, such as manufacture of semi­
conductors and the surface modification of dental implants [34,35]. It
2.7. Statistical analysis has recently reported that TNT loading with Ga(NO₃)₃ could signifi­
cantly enhanced the antibacterial property of Ti implants in vitro [29,
All experiments had at least 3 repeats, and quantitative data were 30]. Compared with Ga(NO₃)₃, Ga2O3 has a much lower solubility and
given as mean ± SD. The data was evaluated and quantified using one- higher stability which may benefits the sustained release of Ga3+ ions
way analysis of variance (ANOVA) and Tukey’s multiple comparison test while reduces its initial burst release [25]. In this study, Ga2O3 was
(GraphPad Prism 7.0, San Diego, CA, USA) with a 95% confidence level. uniformly deposited on the surface of TNT by magnetron sputtering, and
the coating thickness was controlled by the constant deposition time for
25 min, and power range from 50 to 100 W [36].

Fig. 2. Characterization of TNT, Ga2O3@TNT/50 W and Ga2O3@TNT/100 W. (a) SEM and TEM images; (b) EDS mapping spectra; (c, d) XPS spectra; (e) water
contact angle; and (f) in vitro Ga3+ release of samples. The significance was evaluated for * p < 0.05, **p < 0.01, ***p < 0.001, and n.s. P > 0.05.

22964
L. Yao et al. Ceramics International 49 (2023) 22961–22969

3.1. Surface characterization Ga2O3@TNT/100 W, as well as the emerging O1s (Ga–O) at 530.7 eV
which indicates the successful deposition of Ga2O3 [37]. The hydro­
The morphology changes of samples with different power level were philicity of the TNT arrays was significantly reduced after Ga2O3
observed by SEM and TEM. Fig. 2a shows the successful preparation of deposition (Fig. 2e), which may due to the decreased tube diameter
TNT arrays, and the outmost surface of TNT was uniformly coated with [38]. The XRD crystal forms of different samples were detected. The TNT
scattered Ga2O3 nano-particles after sputtering. There was a slightly sample shows the characteristic peak of rutile and anatase, and there
change on the morphology and tube diameter at the outer edge of was no new diffraction peak in the spectra after deposition, which in­
Ga2O3@TNT/50 W by the deposited Ga2O3, while the tube diameter at dicates that the thin layer of Ga2O3 has little effect on the crystal
the outer edge was dramatically reduced in Ga2O3@TNT/100 W group. structure of TNT (Fig. S1) [39].
TEM analysis reveals that the scattered Ga2O3 nano-particles were
formed inside of the nanotubes after magnetron sputtering. The nano­ 3.2. In vitro Ga3+ release
layer coating does not completely seal the tube opening and preserves
the hollow structure of the TNT. As a result, such Ga2O3 nanolayer As show in Fig. 2f, Ga3+ ions were mainly released in the first 7 days.
coated TNT arrays still possess the capability of drug loading. The amount of released Ga3+ ions of Ga2O3@TNT/100 W was 1.5 times
The surface chemical composition of the different samples was higher than Ga2O3@TNT/50 W in the first 7 days, which may due to the
further investigated by EDS and XPS. The EDS elemental mapping show higher amount of Ga2O3 on sample surface. After 7 days, both samples
that the Ga element was evenly distributed on the surfaces after depo­ achieved a plateau in Ga3+ ion release. This might be because of the
sition, and the content of Ga in Ga2O3@TNT/50 W and Ga2O3@TNT/ diffusion barrier that forms or because of the depletion of Ga2O3 on the
100 W groups was 0.45 ± 0.06 at% and 1.15 ± 0.12 at%, respectively sample surface. The release profile of Ga3+ ions from surfaces per time
(Fig. 2b, Table S2). As shown in Fig. 2c, the representative XPS spectra and surface area was also shown in Fig. S2. These results show that the
show major characteristic peaks of TiO2: Ti2p3, at 464.3 eV and 458.7 release of Ga3+ ions from the samples can be managed by changing the
eV, and O1s at 531.0 eV and 530.0 eV, respectively [23]. The Ga peaks at amount of Ga2O3 on the surface of the samples.
1145.2 eV and 1118.3 eV were prominent in Ga2O3@TNT/50 W and

Fig. 3. In vitro evaluation of osteogenesis. (a) FM images of MC3T3-E1 cells on samples; the statistical analysis of (b) cell viability and (c) cell spreading; The activity
and distribution of ALP and mineralization level of each sample: (d) images (white arrows: mineralized nodules); statistical analysis of ALP activity (e) and
mineralization; (f) osteogenic differentiation related RT-PCR results. The significance was evaluated for * p < 0.05, **p < 0.01, ***p < 0.001, and n.s. P > 0.05.

22965
L. Yao et al. Ceramics International 49 (2023) 22961–22969

3.3. In vitro osteoblast assays and OCN. These results indicate that the Ga2O3@TNT/50 W samples
effectively enhanced the osteogenic differentiation, which may due to
3.3.1. Osteoblast morphology and viability the reduced tube diameter and released Ga3+ ions. Moreover, it seems
The FM and SEM results showed that Ga2O3@TNT/50 W signifi­ that COL-I was the main marker enhanced by the released Ga3+ ions,
cantly promoted MC3T3-E1 cell spreading compared to the TNT and the which was corresponding to previous studies [46,47].
Ga2O3@TNT/100 W groups (Fig. 3a), this may due to the additional cell
binding site from nano-scaled Ga2O3 particles on surface [40–42].
3.4. Antibacterial assessment
The cell viability of Ga2O3@TNT/50 W group after 3 and 7 days
increased significantly compared to Ga2O3@TNT/100 W group. How­
Ga3+ and its compounds are novel broad-spectrum metallic antimi­
ever, there was no significant difference in cell viability between TNT
crobials. Their pharmacological properties rely on Fe atom mimicry,
and Ga2O3@TNT/50 W groups (Fig. 3b). Moreover, the average area
thus interference the essential enzymatic processes of bacterial infection
and aspect ratio of adhered cells on Ga2O3@TNT/50 W samples are
that dependent on Fe [42,48–50]. The antibacterial performance of
significantly higher than they are on the Ga2O3@TNT/100 W samples
TNT, Ga2O3@TNT/50 W and Ga2O3@TNT/100 W samples was tested
(Fig. 3c). The inhibited cell proliferation and cell spreading of
against Gram-positive strain S. aureus and Gram-negative strain E. coli,
Ga2O3@TNT/100 W may attributed to the destruction of nanotube
which are commonly associated with implants infection.
structure [43], and the excessive released Ga3+ ions, which could affect
The morphology changes of S. aureus and E. coli after 24 h of incu­
cell viability [35,44].
bation was determined by SEM (Fig. 4a). Compared with the TNT group,
the number of adherent bacteria on Ga2O3@TNT/50 W and
3.3.2. Osteogenic differentiation
Ga2O3@TNT/100 W was reduced, which may be attributed to the
The early osteogenic differentiation of MC3T3-E1 cells was investi­
release of Ga3+ ions. In TNT group, the surfaces of spherical S. aureus
gated by measuring their ALP activity, the early indicator for osteogenic
and rodlike E. coli are smooth and damage-free. However, for
differentiation. The activity and distribution of ALP (7 days, Fig. 3 d and
Ga2O3@TNT/50 W and Ga2O3@TNT/100 W groups, the morphologies
e) and mineralization level (14 days, Fig. 3 d and f) in Ga2O3@TNT/50
of cells have changed significantly the appearance of rumples, dis­
W group were significantly improved than other groups after 7 days of
configuration and shrunken were observed.
incubation. However, after 14 days of osteogenic induction, the ALP
Fig. 4b–d shows the colonies of S. aureus and E. coli E. coli after 4 and
activity of Ga2O3@TNT/50 W was lower than TNT, which may be
24 h of culturing. TNT demonstrated non-antibacterial activity that
related to the transformation of cells to mineralization (Fig. 3e) [9,44].
bacterial colonies are all over the agar plates. However, the agar plates
Correspondingly, the mineralization level of Ga2O3@TNT/50 W group
from the Ga2O3@TNT/50 W and Ga2O3@TNT/100 W samples host
was significantly improved than other groups after 14 days of incubation
minimal bacterial colonies indicated their excellent antibacterial
(Fig. 3f).
activities.
The RT-PCR results of each sample was shown in Fig. 3g. The
Live and dead staining result showed that nearly 100% S. aureus in
Ga2O3@TNT/50 W group significantly enhanced osteogenic genes
TNT group were stained in green fluorescence, indicating they were
expression including RUNX2, OPN, and COL-I. It has been reported that
alive. However, 89.3 ± 2.5% and 94.0 ± 2.6% S. aureus were killed in
Ga promotes the expression of COL-I, which is associated with bone
Ga2O3@TNT/50 W and Ga2O3@TNT/100 W groups, respectively.
collagen formation and mineralization deposition [45]. However, the
Similar results were observed with E. coli., 92.7 ± 4.0% and 95.7 ± 2.1%
deposition of Ga2O3 had no significant effect on the expression of ALP
of bacteria was dead in Ga2O3@TNT/50 W and Ga2O3@TNT/100 W

Fig. 4. Antibacterial efficacy of TNT, Ga2O3@TNT/50 W and Ga2O3@TNT/100 W. (a) The adhesion and morphology of S. aureus and E. coli after 24 h of incubation;
(b) typical images of cultivated S. aureus and E. coli colonies from the surface of samples after 4 and 24 h of incubation; (c, d) antibacterial rates of various specimens
against S. aureus and E. coli respectively; (e) live and dead staining of adherent bacteria, and (f) the relevant statistical results. The significance was evaluated for * p
< 0.05, **p < 0.01, ***p < 0.001, and n.s. P > 0.05.

22966
L. Yao et al. Ceramics International 49 (2023) 22961–22969

groups, while it was almost all alive in TNT group (Fig. 4 e and f). Thus, implantation, we sacrificed the rats at 3 days and 1 month after im­
our research suggests that the released Ga3+ ions from Ga2O3 nano­ plantation, and collected femoral samples for histology analysis and
particles contribute to the TNT array with good antibacterial ability. micro-CT.
In summary, the Ga2O3@TNT/50 W group effectively promotes
osteogenic differentiation and has excellent antibacterial properties in 3.5.1. In vivo antibacterial activity
vitro. The Ga2O3@TNT/100 W group was discarded in subsequent in vivo The in vivo antibacterial property of samples was evaluated by
experiments due to its obvious cytotoxicity. osteoporotic rat model with S. aureus infection. As shown by Giemsa
staining in Fig. 5a, many bacteria were distributed around the TNT
implant (red arrow). However, there was no apparent bacterial aggre­
3.5. In vivo results gation observed around the Ga2O3@TNT/50 W implant, indicating that
its early antibacterial property. We speculate that the rapid release of
In this study, the ovariectomized (OVX) rat model was selected Ga3+ ions in the first 3 days around the implant leads to abnormal
because it is of great significance to the pathophysiological study of bacterial metabolism and affects bacteria proliferation and survival due
implant osseointegration and drug treatment, which was considered as to the competitive inhibition of the uptake of Fe3+ ions by bacteria. On
the most suitable model for simulating postmenopausal osteoporosis by the other hand, the high surface-to-volume ratio of Ga2O3 nanoparticles
the US Food and Drug Administration [24,51,52]. could physically destroys the bacterial membrane as well [54].
During osteoporotic conditions, the bone microenvironment effects It was observed by HE staining results that many monocytes and
on the transformation of monocytes into osteoclasts, resulting in bone other inflammatory cells were aggregated around the TNT implant
loss and reduced bone density, which affects the initial stability of the (green arrow), indicating an early severe inflammatory response
implant. The decrease in the initial stability of the implant will result in induced by infection. However, no monocyte aggregation was observed
difficulty in the formation of new bone around the implant and the in the bone tissue nearby Ga2O3@TNT/50 W implant, suggesting no
decline of the osseointegration capacity of the implant, which will apparent inflammation occurred, which may due to the inhibitory effect
seriously affect the success rate of implant surgery. of released Ga3+ on macrophages and T lymphocytes [52].
Peri-implantitis induced by infections hurts the reconstruction of
tissues surrounding implants, which often leads to implant failure and 3.5.2. Micro-CT results
other serious dental diseases. The formation of plaque biofilm on the The newly formed bone around the implants were analyzed via
surface of implant enhanced the resistance of bacteria to host’s defense micro-CT 3D images. As shown in Fig. 5b, there is an increased new bone
and antibiotics [53]. Therefore, the OVX rat model with bacterial formation around the Ga2O3@TNT/50 W group compared to TNT
infection after implantation surgery was used to evaluate the effect of group. This observation was confirmed using bone volume measure
released Ga3+ on antibacterial and osseointegration in vivo. In order to softwares, CTan, and CTVox. The quantified data of bone volume/total
check the early anti-inflammatory and osseointegration status after

Fig. 5. The therapeutical efficacy of TNT and Ga2O3@TNT/50 W implants in vivo. (a) histological images of TNT and Ga2O3@TNT/50 W after surgical implantation
for 3 days; (b) micro-CT 3D images after surgical implantation for 1 month; (c) the statistical analysis of micro-CT evaluation; (d) histological images of bone tissue
after surgical implantation for 1 month. The significance was evaluated for * p < 0.05, **p < 0.01, ***p < 0.001, and n.s. P > 0.05.

22967
L. Yao et al. Ceramics International 49 (2023) 22961–22969

volume (BV/TV%), trabecular thickness (Tb⋅Th), bone connectivity Appendix A. Supplementary data
density (Conn-Dens), and trabecular space (Tb.Sp) was shown in Fig. 5c,
indicating that Ga2O3@TNT/50 W had the significant better osteoin­ Supplementary data to this article can be found online at https://doi.
ductive ability than TNT. org/10.1016/j.ceramint.2023.04.121.

3.5.3. Analysis of early osseointegration tissue References


Both Masson and HE staining were used to further confirm the new
bone formation around the implants. As illustrated in Fig. 5d, in Masson [1] Y. Jin, L. Xu, X. Hu, S. Liao, J.L. Pathak, J. Liu, Lithium chloride enhances bone
regeneration and implant osseointegration in osteoporotic conditions, J. Bone
staining, the blue part of the bone interface around the implant repre­ Miner. Metabol. 35 (2017) 497–503.
sents the immature trabecular bone, while the red part represents [2] J. Liu, J.L. Pathak, X. Hu, Y. Jin, Z. Wu, M.A. Al-Baadani, S. Wu, H. Zhang,
mature trabecular bone. It was found that more blue part tissue, namely S. Farkasdi, Y. Liu, Sustained release of zoledronic acid from mesoporous TiO2-
layered implant enhances implant osseointegration in osteoporotic condition,
immature trabecular bone tissue was distributed around the J. Biomed. Nanotechnol. 14 (2018) 1965–1978.
Ga2O3@TNT/50 W implant compared to TNT implant. Moreover, the [3] J.S. Khurana, L.A. Fitzpatrick, Osteoporosis and Metabolic Bone Disease, Bone
bone interface of Ga2O3@TNT/50 W implant was significantly thicker Pathology, Springer, 2009, pp. 217–237.
[4] R. Rasouli, A. Barhoum, H. Uludag, A review of nanostructured surfaces and
than TNT group. For HE staining, it showed an increased bone-like tissue materials for dental implants: surface coating, patterning and functionalization for
around Ga2O3@TNT/50 W implant compared to TNT implant, which improved performance, Biomater. Sci. 6 (2018) 1312–1338.
consisted with micro-CT results as well. [5] A. Czan, O. Babík, M. Miklos, L. Záušková, V. Mezencevová, Assessment of surface
area characteristics of dental implants with gradual bioactive surface treatment,
Micro-CT and histological analysis showed that Ga2O3@TNT/50 W
Technol. Eng. 14 (2017) 35–39.
can promote new bone formation and improve implantation in the [6] M. Rottmar, E. Müller, S. Guimond-Lischer, M. Stephan, S. Berner, K. Maniura-
infected osteoporosis rat model, which may due to the enhanced initial Weber, Assessing the osteogenic potential of zirconia and titanium surfaces with an
stability of the implantation and the appropriate tissue regeneration advanced in vitro model, Dent. Mater. 35 (2019) 74–86.
[7] P. Augat, U. Simon, A. Liedert, L. Claes, Mechanics and mechano-biology of
microenvironment. As described previous, the sustained release of Ga3+ fracture healing in normal and osteoporotic bone, Osteoporos. Int. 16 (2005)
ions played a role in anti-infection and regulation of inflammatory re­ S36–S43.
sponses in the early stage, which is beneficial to enhance the subse­ [8] K. Page, R.G. Palgrave, I.P. Parkin, M. Wilson, S.L. Savin, A.V. Chadwick, Titania
and silver–titania composite films on glass—potent antimicrobial coatings,
quence osseointegration process. On the other hand, the promotion of J. Mater. Chem. 17 (2007) 95–104.
COL-I and other osteogenesis-related gene expression from osteoblasts [9] R.O. Darouiche, Treatment of infections associated with surgical implants, N. Engl.
by the release Ga3+ ions from Ga2O3@TNT/50 W directly stimulates J. Med. 350 (2004) 1422–1429.
[10] P. Roy, S. Berger, P. Schmuki, TiO2 nanotubes: synthesis and applications, Angew.
new bone formation. In addition, it has been reported the regulation of Chem. Int. Ed. 50 (2011) 2904–2939.
bone mineral and matrix properties by Ga3+ ions inhibits the activity of [11] G. Balasundaram, C. Yao, T.J. Webster, TiO2 nanotubes functionalized with
osteoclasts and reduces bone resorption [24,51]. regions of bone morphogenetic protein-2 increases osteoblast adhesion, J. Biomed.
Mater. Res. 84 (2008) 447–453.
[12] S.-K. Min, H.K. Kang, D.H. Jang, S.Y. Jung, O.B. Kim, B.-M. Min, I.-S. Yeo, Titanium
4. Conclusions surface coating with a laminin-derived functional peptide promotes bone cell
adhesion, BioMed Res. Int. (2013) 2013.
[13] B. Wang, A. Bian, F. Jia, J. Lan, H. Yang, K. Yan, L. Xie, H. Qiao, X. Chang, H. Lin,
In summary, we successfully sputtered Ga2O3 nanoparticles on TNT Dual-functional” strontium titanate nanotubes designed based on fusion peptides
surface via magnetron sputtering. We found that both Ga2O3@TNT/50 simultaneously enhancing anti-infection and osseointegration, Biomaterials
W and Ga2O3@TNT/100 W had superior antibacterial ability in vitro. Advances 133 (2022), 112650.
[14] B. Wang, Z. Wu, S. Wang, S. Wang, Q. Niu, Y. Wu, F. Jia, A. Bian, L. Xie, H. Qiao,
Meanwhile, it was indicated that the sustained release of Ga3+ ions from Mg/Cu-doped TiO2 nanotube array: a novel dual-function system with self-
Ga2O3@TNT/50 W could significantly improve the osteogenic differ­ antibacterial activity and excellent cell compatibility, Mater. Sci. Eng. C 128
entiation ability of MC3T3-E1, whereas Ga2O3@TNT/100 W group (2021), 112322.
[15] Y. Li, Y. Yang, R. Li, X. Tang, D. Guo, Y. Qin, Enhanced antibacterial properties of
showed cytotoxicity. Moreover, in vivo experiments have confirmed that orthopedic implants by titanium nanotube surface modification: a review of
Ga2O3@TNT/50 W group has excellent antibacterial effect and effec­ current techniques, Int. J. Nanomed. (2019) 7217–7237.
tively promotes new bone formation in an infected rat model of osteo­ [16] K. Gulati, S. Ivanovski, Dental implants modified with drug releasing titania
nanotubes: therapeutic potential and developmental challenges, Expet Opin. Drug
porosis in comparison with TNT group. Our work provides a novel
Deliv. 14 (2017) 1009–1024.
strategy to effectively deposits Ga2O3 on the surface of Ti implants to [17] D. Chopra, K. Gulati, S. Ivanovski, Understanding and optimizing the antibacterial
enhance its antibacterial and bone integration properties in osteoporotic functions of anodized nano-engineered titanium implants, Acta Biomater. 127
(2021) 80–101.
situations. The future studies may focus on the regulation mechanism of
[18] G. Janarthanan, I. Noh, Recent trends in metal ion based hydrogel biomaterials for
such systems on osteoclasts and related immune cells, exploring their tissue engineering and other biomedical applications, J. Mater. Sci. Technol. 63
potential drug loading abilities, and improving their mechanical prop­ (2021) 35–53.
erties for clinical application. [19] V.M. Schatkoski, T.L. do Amaral Montanheiro, B.R.C. de Menezes, R.M. Pereira, K.
F. Rodrigues, R.G. Ribas, D.M. da Silva, G.P. Thim, Current advances concerning
the most cited metal ions doped bioceramics and silicate-based bioactive glasses for
bone tissue engineering, Ceram. Int. 47 (2021) 2999–3012.
Declaration of competing interest [20] M.B. Aldrich, J.C. Rasmussen, C.E. Fife, S.F. Shaitelman, E.M. Sevick-Muraca, The
development and treatment of lymphatic dysfunction in cancer patients and
survivors, Cancers 12 (2020).
The authors declare that they have no known competing financial [21] C. Xie, P. Li, Y. Liu, F. Luo, X. Xiao, Preparation of TiO2 nanotubes/mesoporous
interests or personal relationships that could have appeared to influence calcium silicate composites with controllable drug release, Mater. Sci. Eng. C 67
the work reported in this paper. (2016) 433–439.
[22] Y. Dong, H. Ye, Y. Liu, L. Xu, Z. Wu, X. Hu, J. Ma, J.L. Pathak, J. Liu, G. Wu, pH
dependent silver nanoparticles releasing titanium implant: a novel therapeutic
Acknowledgement approach to control peri-implant infection, Colloids Surf. B Biointerfaces 158
(2017) 127–136.
[23] F. Li, F. Liu, K. Huang, S. Yang, Advancement of gallium and gallium-based
This work is supported by the National Natural Science Foundation compounds as antimicrobial agents, Front. Bioeng. Biotechnol. 10 (2022).
of China (81870810 and 82071170), Zhejiang Provincial Science and [24] Y. Kaneko, M. Thoendel, O. Olakanmi, B.E. Britigan, P.K. Singh, The transition
Technology Project for Public Welfare (LGF21H140004, metal gallium disrupts Pseudomonas aeruginosa iron metabolism and has
antimicrobial and antibiofilm activity, J. Clin. Invest. 117 (2007) 877–888.
LTGY23H140001), Wenzhou Public Welfare Science and Technology [25] C.R. Chitambar, Medical applications and toxicities of gallium compounds, Int. J.
Project (ZY2019009, 2021Y0441 and Y20210114) and Traditional Environ. Res. Publ. Health 7 (2010) 2337–2361.
Chinese Medicine Science and Technology Program of Zhejiang Prov­
ince (2023ZR109).

22968
L. Yao et al. Ceramics International 49 (2023) 22961–22969

[26] A. Rahimnejad Yazdi, L. Torkan, W. Stone, M.R. Towler, The impact of gallium [40] E. Verron, J. Bouler, J. Scimeca, Gallium as a potential candidate for treatment of
content on degradation, bioactivity, and antibacterial potency of zinc borate osteoporosis, Drug Discov. Today 17 (2012) 1127–1132.
bioactive glass, J. Biomed. Mater. Res. B Appl. Biomater. 106 (2018) 367–376. [41] L. Todorov, I. Kostova, M. Traykova, Lanthanum, Gallium and their impact on
[27] G. Lusvardi, G. Malavasi, L. Menabue, S. Shruti, Gallium-containing oxidative stress, Curr. Med. Chem. 26 (2019) 4280–4295.
phosphosilicate glasses: functionalization and in-vitro bioactivity, Mater. Sci. Eng. [42] E. Verron, A. Loubat, G. Carle, C. Vignes-Colombeix, I. Strazic, J. Guicheux,
C 33 (2013) 3190–3196. N. Rochet, J. Bouler, J. Scimeca, Molecular effects of gallium on osteoclastic
[28] K.S. Rana, L.P.D. Souza, M.A. Isaacs, F.N. Raja, A.P. Morrell, R.A. Martin, differentiation of mouse and human monocytes, Biochem. Pharmacol. 83 (2012)
Development and characterization of gallium-doped bioactive glasses for potential 671–679.
bone cancer applications, ACS Biomater. Sci. Eng. 3 (2017) 3425–3432. [43] H.C. Blair, S.L. Teitelbaum, H.L. Tan, P.H. Schlesinger, Reversible inhibition of
[29] A. Jayasree, M.N. Gómez-Cerezo, E. Verron, S. Ivanovski, K. Gulati, Gallium-doped osteoclastic activity by bone-bound gallium (III), J. Cell. Biochem. 48 (1992)
dual micro-nano titanium dental implants towards soft-tissue integration and 401–410.
bactericidal functions, Materials Today Advances 16 (2022), 100297. [44] N. Wang, H. Li, W. Lü, J. Li, J. Wang, Z. Zhang, Y. Liu, Effects of TiO2 nanotubes
[30] S. Maher, D. Linklater, H. Rastin, P. Le Yap, E.P. Ivanova, D. Losic, Tailoring with different diameters on gene expression and osseointegration of implants in
additively manufactured titanium implants for short-time pediatric implantations minipigs, Biomaterials 32 (2011) 6900–6911.
with enhanced bactericidal activity, ChemMedChem 17 (2022), e202100580. [45] H. Yu, X. Huang, X. Yang, H. Liu, M. Zhang, X. Zhang, R. Hang, B. Tang, Synthesis
[31] F. Wang, X. Wang, E. Xie, Q. Gan, S. Ping, J. Wei, F. Li, Z. Wang, Simultaneous and biological properties of Zn-incorporated micro/nano-textured surface on Ti by
incorporation of gallium oxide and tantalum microparticles into micro-arc high current anodization, Mater. Sci. Eng. C 78 (2017) 175–184.
oxidation coating of titanium possessing antibacterial effect and stimulating [46] H. Qiao, C. Zhang, X. Dang, H. Yang, Y. Wang, Y. Chen, L. Ma, S. Han, H. Lin,
cellular response, Biomaterials Advances (2022), 212736. X. Zhang, Gallium loading into a polydopamine-functionalised SrTiO3 nanotube
[32] M. Nycz, K. Arkusz, D.G. Pijanowska, Influence of the silver nanoparticles (AgNPs) with combined osteoinductive and antimicrobial activities, Ceram. Int. 45 (2019)
formation conditions onto titanium dioxide (TiO2) nanotubes based electrodes on 22183–22195.
their impedimetric response, Nanomaterials 9 (2019) 1072. [47] F. He, C. Qiu, T. Lu, X. Shi, J. Ye, Conjunction of gallium doping and calcium
[33] C. Pan, T. Liu, Y. Yang, T. Liu, Z. Gong, Y. Wei, L. Quan, Z. Yang, S. Liu, silicate mediates osteoblastic and osteoclastic performances of tricalcium
Incorporation of Sr2+ and Ag nanoparticles into TiO2 nanotubes to synergistically phosphate bioceramics, Biomed. Mater. 17 (2021), 015012.
enhance osteogenic and antibacterial activities for bone repair, Mater. Des. 196 [48] Y. Zhu, X. Liu, K.W. Yeung, P.K. Chu, S. Wu, Biofunctionalization of carbon
(2020), 109086. nanotubes/chitosan hybrids on Ti implants by atom layer deposited ZnO
[34] A. Abdal-hay, N. Abbasi, M. Gwiazda, S. Hamlet, S. Ivanovski, Novel nanostructures, Appl. Surf. Sci. 400 (2017) 14–23.
polycaprolactone/hydroxyapatite nanocomposite fibrous scaffolds by direct melt- [49] L.G. Jenis, C.E. Waud, G.S. Stein, J.B. Lian, D.T. Baran, Effect of gallium nitrate in
electrospinning writing, Eur. Polym. J. 105 (2018) 257–264. vitro and in normal rats, J. Cell. Biochem. 52 (1993) 330–336.
[35] Y. Zhu, Y. Gu, S. Qiao, L. Zhou, J. Shi, H. Lai, Bacterial and mammalian cells [50] A. Cochis, B. Azzimonti, C. Della Valle, E. De Giglio, N. Bloise, L. Visai, S. Cometa,
adhesion to tantalum-decorated micro-/nano-structured titanium, J. Biomed. L. Rimondini, R. Chiesa, The effect of silver or gallium doped titanium against the
Mater. Res. 105 (2017) 871–878. multidrug resistant Acinetobacter baumannii, Biomaterials 80 (2016) 80–95.
[36] E. Verron, M. Masson, S. Khoshniat, L. Duplomb, Y. Wittrant, M. Baud’Huin, [51] R. García-Contreras, B. Pérez-Eretza, E. Lira-Silva, R. Jasso-Chávez, R. Coria-
Z. Badran, B. Bujoli, P. Janvier, J.C. Scimeca, Gallium modulates osteoclastic bone Jiménez, A. Rangel-Vega, T. Maeda, T.K. Wood, Gallium induces the production of
resorption in vitro without affecting osteoblasts, Br. J. Pharmacol. 159 (2010) virulence factors in Pseudomonas aeruginosa, Pathogens and disease 70 (2014)
1681–1692. 95–98.
[37] I. Lopez, A.D. Utrilla, E. Nogales, B. Mendez, J. Piqueras, A. Peche, J. Ramírez- [52] A. Król, P. Pomastowski, K. Rafińska, V. Railean-Plugaru, B. Buszewski, Zinc oxide
Castellanos, J.M. Gonzalez-Calbet, In-doped gallium oxide micro-and nanoparticles: synthesis, antiseptic activity and toxicity mechanism, Adv. Colloid
nanostructures: morphology, structure, and luminescence properties, J. Phys. Interface Sci. 249 (2017) 37–52.
Chem. C 116 (2012) 3935–3943. [53] K.R. Raghupathi, R.T. Koodali, A.C. Manna, Size-dependent bacterial growth
[38] Y. Li, Y. Qi, Q. Gao, Q. Niu, M. Shen, Q. Fu, K. Hu, L. Kong, Effects of a micro/nano inhibition and mechanism of antibacterial activity of zinc oxide nanoparticles,
rough strontium-loaded surface on osseointegration, Int. J. Nanomed. 10 (2015) Langmuir 27 (2011) 4020–4028.
4549. [54] D. Thompson, H. Simmons, C. Pirie, H. Ke, FDA Guidelines and animal models for
[39] K. Zhang, Y. Zhu, X. Liu, Z. Cui, K.W. Yeung, H. Pan, S. Wu, Sr/ZnO doped titania osteoporosis, Bone 17 (1995) S125–S133.
nanotube array: an effective surface system with excellent osteoinductivity and
self-antibacterial activity, Mater. Des. 130 (2017) 403–412.

22969

You might also like