You are on page 1of 12

Smart Materials in Medicine 2 (2021) 280–291

Contents lists available at ScienceDirect

Smart Materials in Medicine


journal homepage: www.keaipublishing.com/en/journals/smart-materials-in-medicine/

Graphene oxide as a promising material in dentistry and tissue regeneration:


A review
Xuanyu Qi a, Fei Jiang a, b, c, Mingliang Zhou a, Wenjie Zhang a, **, Xinquan Jiang a, *
a
Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong
University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Engineering Research
Center of Advanced Dental Technology and Materials, No. 639 Zhizaoju Road, Shanghai, 200011, China
b
Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, No. 140, Han Zhong Road, Nanjing, 210029, China
c
Department of Polyclinic, Affiliated Hospital of Stomatology, Nanjing Medical University, No. 136, Han Zhong Road, Nanjing, 210029, China

A R T I C L E I N F O 1 . A B S T R A C T

Keywords: Current treatments for common dental diseases such as caries, periodontal diseases, tooth defects, missing teeth
Dental materials and bone defects cannot replenish dental tissue with artificial alternatives while maintaining the biological and
Nanomaterials mechanical properties of natural tissue. In recent years, in addition to existing conventional treatments, tissue
Tissue regeneration
engineering and regenerative medicine have also been applied in this field to recover real tissue. Graphene oxide
Graphene oxide
(GO) is considered to be a promising material in dentistry and related tissue regeneration due to its outstanding
properties. In this review, we first describe the physicochemical properties of GO and then explain the biological
properties that derive from the physiochemical properties, including biocompatibility, controlled release of
medicine, antibacterial properties, osteogenic differentiation properties and odontogenic differentiation proper-
ties. We also highlight the potential applications of graphene oxide in dentistry and related tissue regeneration.
For bacterial resistance in dentistry, GO has a wide range of applications. For reconstructing tooth defects,
conventional dental materials such as resin and adhesives have been optimized with GO. To treat missing teeth,
GO surface modification of dental implants is adopted to enhance not only the antibacterial properties but also
osseointegration. To repair maxillofacial bone defects, GO can enhance the osteogenic properties of engineered
tissue scaffolds through both surface and composite modifications. Some obstacles that inhibit the progression of
this technology from the laboratory to the clinic have also been described.

1. Introduction With a survival rate greater than 89 %, dental implants have been a
conventional treatment for patients suffering from missing teeth [3–5].
Dentistry is a field with multiple problems to be solved by optimizing However, there is still an 11 % failure rate in dental implant treatment
materials. In modern dentistry, both the early prevention of caries and and removal, which brings patients both physical and mental discomfort
the development of new and efficient restorative materials are sought. along with additional economic loss [6,7]. Many factors result in implant
Although tremendous efforts have been made to promote oral hygiene failures, and a major factor is the harmful influences of anaerobic plaque
and fluoridation, the prevention of early-stage tooth decay lesions is still bacteria on peri-implant tissues [8]. To address this problem, mechanical
a challenge for dental research and public health [1]. As a result, treating cleaning to remove biofilms from implants, antibiotic therapy to kill in-
tooth defects resulting from caries has become one of the most common fectious bacteria and regenerative surgery to rebuild bone-implant in-
treatments in dentistry. The mechanical properties of materials applied in terfaces are routinely adopted [9–13]. Reliable antibacterial surface
treating tooth defects, such as resin-based composites, are related to their modification strategies and efficient cleaning methods should be
clinical application and the longevity of the restoration [2]. improved for future applications. In addition, conventional dental im-
More serious than caries and some caused by caries, periodontitis or plants made of titanium cannot fully meet clinical needs due to their
trauma, tooth loss is a problem that negatively affects human life quality. limited osseointegration and osteoinductive properties, especially under

* Corresponding author. Shanghai Jiao Tong University, School of Medicine Department of Prosthodontics, 639 Zhizaoju Road, Shanghai, 200011, China.
** Corresponding author. Shanghai Jiao Tong University, School of Medicine Department of Prosthodontics, 639 Zhizaoju Road, Shanghai, 200011, China.
E-mail addresses: zhangwenjie586@126.com (W. Zhang), xinquanjiang@aliyun.com (X. Jiang).

https://doi.org/10.1016/j.smaim.2021.08.001
Received 10 June 2021; Received in revised form 27 July 2021; Accepted 3 August 2021
Available online 8 August 2021
2590-1834/© 2021 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

poor or inadequate bone conditions. Materials that positively promote 2. The physicochemical properties of GO
osseointegration and have osteoinduction properties are urgently needed
to decorate implants. GO has a high specific surface area (890 m2 g1) [30], and abundant
In addition to existing conventional treatments, tissue engineering functional groups, such as hydroxyl and epoxy groups, as well as slight
and regenerative medicine have also been applied in dentistry. Tissue- amounts of carboxyl, carbonyl, phenol, lactone, and quinone [31], were
engineered bone grafts represent an ideal choice for bone augmenta- clearly observed by FT-IR spectroscopy [32–34]. These functional groups
tion and create both robust and sufficient bone for dental implantation. were created by the oxidation of graphene, which also increased the
Maxillofacial bone defects resulting from traumatic injuries [14] or hydrophilicity of the surface. The GO properties are enormously changed
dental and surgical treatments [15,16] require tissue engineered scaf- due to the presence of these oxygenated groups, which helps the for-
folds with optimized properties, including biocompatibility, osteoin- mation of stable dispersion in aqueous media and other polar solvents
duction, and controlled drug release, to achieve bone regeneration with [35–37]. Biochemical and bioconjugation reactions were therefore
higher efficiency. allowed to occur at both its basal plane and its edges [33], which pro-
Graphene is an allotrope of carbon composed of single-atom-thick moted the functionalization of the GO surface with proteins, antibodies
carbon nanosheets with a honeycomb structure and was first success- and DNA fragments [38,39]. Functional moieties (i.e., hydroxyl and
fully prepared by Geim and Novoselov in 2004 [17]. When modified by carboxyl groups) on GO can establish chemical bonds with polymers in
oxygen-containing groups such as hydroxyl (-OH), epoxy (-O-), carboxyl scaffold matrices, which can provide scaffolds with enhanced interfacial
(-COOH) and carbonyl (C– – O), the graphene sheet transforms into a strength [40]. GO has also been reported to enhance the mechanical
derivative known as graphene oxide (GO). As a result of the modification, properties of various materials, including chitosan [41,42], gelatin [43,
the highly conjugated graphene structure is damaged, but the GO 44], chitin [45], collagen [46], hydroxyapatite [47,48] and PEEK [49].
nanosheets are also rendered with better chemical stability and solubility A high physical adsorption capacity for proteins was observed on GO.
in water [18]. GO has many attractive physicochemical and biological The mechanism depends on the GO morphology, oxidation degree [50],
properties. First, it has been reported that GO has great biocompatibility, hydrophobicity and type of absorbed protein [51–53]. Regarding the
can promote cell adhesion and proliferation, and even induce the type of protein, polypeptides can be adsorbed on the GO surface via four
directional differentiation of stem cells [19,20] Remarkably, GO alone mechanisms; the main mechanism is hydrophobic-hydrophobic interac-
can induce the osteogenic differentiation of stem cells [21]. Second, GO tion. 1) Hydrophobic-hydrophobic interactions benefit from the sp2 hy-
has been indicated to have excellent antibacterial properties through bridization of GO and are affected by the electron density of proteins:
different mechanisms [22–25]. Third, thanks to a large surface area (on Bovine albumin shows reliable adsorption to GO, while low molecular
both sides of the sheet) for the physical adsorption of nucleobases [26, weight heparin, which has a low electron density, exhibits a decreased
27] and aromatic compounds via surface adsorption, hydrogen bonding, hydrophobic interaction [54]. 2) van der Waals interactions are the
and other types of interactions [28], GO also has potential for drug de- second mechanism by which GO bonds with molecules; these in-
livery [29]. In summary, the properties of GO wonderfully meet the re- teractions can be weakened by the oxygenated moieties formed during
quirements of dentistry and related tissue engineering. the oxidation process [55]. 3) Third, electrostatic interactions are also
In this review, we first describe the physicochemical properties and common when GO serves as a vehicle, and when pH < 6.0, sorption can
biological properties of GO and then focus on the application of GO in be enhanced [56]. 4) The fourth mechanism involves π–π stacking in-
different aspects of dentistry and related tissue regeneration. We further teractions due to the abundant π electrons on the basal plane of the GO
summarize the future possible development direction of GO nano- surface, as described for the individual hexagonal cells of the GO basal
materials in dentistry and related tissue regeneration. planes and the glucose oxidase [57].

281
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Fig. 1. Bone regeneration of Ti ring implants with or without GO coating and BMP-2/SP loading in a mouse calvarial defect model. Notes: Histological analysis with
Goldners trichrome staining of mouse calvarial defects with implants 8 weeks after treatment. (A) Lower magnification (  12.5) images of the implants. The black
arrowheads and red arrows indicate the implant and newly formed bone, respectively. Scale bars ¼ 1 mm. (B) Higher magnification (  100) images of the implants.
Scale bars ¼ 100 μm. (C) The area of new bone formation was determined using histomorphometric analysis (n ¼ 5). *P < 05 between designated two groups.
Abbreviations: Ti, titanium; GO, graphene oxide; BMP-2, bone morphogenetic protein-2; SP, substance P[68]. Copyright © 2014, Dove Medical Press Limited.

GO can also mechanically form a covalent conjugation on its surface 3.2. Controlled release property
with substances, which is general in functionalization associated with GO
[58]. The binding strength between GO and other substances is therefore The therapeutic efficacy of drugs usually relies on the drug delivery.
enhanced, and the stability improves even with varying pH, organic Owing to its excellent biocompatibility, low toxicity, and large dosage
solvents, and storage conditions [59]. loading capacity [65,66], GO group has been considered as a method in
therapeutic medicine for the delivery of therapeutic medicine with high
3. The biological properties of GO efficiency. Many kinds of medicines have been successfully loaded on GO
and released in a controlled manner for bone tissue engineering; These
3.1. Biocompatibility medicines include BMP-2 [67–69], aspirin [70], vancomycin [71], and
siRNA [72].
Biocompatibility is a fundamental requirement for both implants The mechanism behind the ability of GO to load and sustain protein
and tissue engineering scaffolds and is also the basis for the design and release is divided into two parts. First, the ionized groups on the GO
improvement of novel materials. An increasing number of studies have enable protein delivery by binding through electrostatic interactions
shown that GO has some cytotoxicity, but its toxic manifestation is [73–75]. Second, the hydrophobic domains on GO can interact with
concentration-dependent and correlates with the shape of the GO [60, those of proteins by hydrophobic π–π stacking [76,77]. La et al. coated
61]. Wang et al. [62] cocultured three different concentration gradients titanium substrates with GO through layer-by-layer assembly of posi-
of GO with human fibroblasts and found that GO had no toxic effect on tively (GO-NH3þ) and negatively charged (GO-COO) GO sheets and
human fibroblasts at doses below 20 μg/mL, while doses above loaded BMP-2 on the outermost coating layer of GO-COO. Compared to
50 μg/mL showed some cytotoxicity. Further in vivo studies in mice the hBMMSCs cultured on Ti substrates and directly loaded with BMP-2,
showed no significant toxicity in the low (0.1 mg) and medium GO enhanced the in vitro osteogenic differentiation when it was used as a
(0.25 mg) dose groups, while the high (0.4 mg) dose group showed coat, and it led to the higher conformational protein stability, higher
chronic toxicity mainly in the lung, liver and kidney, leading to mor- bioactivity, and increased local concentration of BMP-2 on surfaces
tality and lung granuloma formation in mice. In addition, the testing of modified with GO. When implanted into mouse calvaria, the GO group
the GO cytotoxicity in mammalian cells indicated that microsized GO showed a sustained release of BMP-2, which resulted in more robust bone
led to higher cytotoxicity than nanosized GO, the reason of which may formation [67]. To improve the stem cell recruitment of the material,
be the faster sedimentation rate and the formation of compact GO ag- substance P was then introduced into the same group, and it was reported
gregates on top of the adherent cells in the wells, inhibiting the nutrient to be effective in promoting recruitment of MSCs to damaged tissues.
availability for the growth of cells [63]. Remarkably, GO has an auto- Loading of BMP-2 and SP with GO further enhanced bone formation in
degradation pathway in aqueous conditions. After prolonged water vivo [68] (Fig. 1.). Recently, through layer by layer (LBL) self-assembly
exposure, GO lattice sizes have been shown to decrease over time due to techenique, polydopamine(PDA), GO, and type I collagen(Col I) was
C–C bond cleavage; the resulting structure is similar to humid acid, a loaded on the surface of Ti-based implants. Furthermore, the encapsu-
benign degradation product of all organic matter [64] This property lated amount and release rate of bioactive substances including silver
gives GO moderate biocompatibility, and it can be cleared in vivo on a (Ag) ions and bovine serum albumin (BSA) could be precisely controlled
timescale of months. by the layer number of the nanofilms [66].

282
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Fig. 2. Live/dead fluorescent staining images. (A, B) S. mutans, (C, D) F. nucleatum, and (E, F) P. gingivalis cells were treated with GO nanosheets and isotonic saline
(control) for 2 h. Representative fluorescence microscopy images of bacterial cells stained with SYTO 9 (green channel) and PI (red channel) for 15 min in the dark. All
treated cases had the same GO dose of 80 μg/mL. Scale bar ¼ 50 μm. (G) TEM images of S. mutans, F. nucleatum, and P. gingivalis cells after incubation with GO
nanosheet dispersion (right side) for 2 h and after incubation with saline solution for 2 h as a control (left side). All treated cases had the same GO dose of 80 μg/mL.
Scale bar ¼ 500 nm. Reprinted with permission from Ref. [90]. Copyright © 2015, American Chemical Society.

Apart from titanium, in recent years, our group has also chosen a GO differentiation of stem cells in vitro and new bone formation in vivo [72].
delivery system as a modification of silk fibroin electrospun scaffolds in To overcome the problem that GO cannot be used as a gene carrier to load
bone tissue engineering. To avoid the shortcomings of BMP-2, which double-stranded DNA (dsDNA) due to its poor loading ability [80–82],
includes a complex preparation process, low production rate and side Santo et al. employed microfluidic mixing and synthesized hybrid NPs
effects at large doses, we chose a new polypeptide known as P24 with a consisting of GO nanosheets coated with a double layer of canonic lipids
simple synthesis procedure and the ability to increase osteogenic dif- [83]. We are looking forward to the application of the results of the study
ferentiation as the loaded medicine. The negatively charged BMP-2 in bone regeneration.
polypeptide-grafted GO was assembled on positively charged CS-coated
SF electrospun scaffolds through electrostatic interactions. The scaf- 3.3. Antibacterial properties
folds both exhibited satisfying cellular performance as well as excellent
bone regeneration properties in critical-sized bone defects [69]. Graphene-based materials share a similar antibacterial mechanism
Chemical medicine is another example of molecules that can be with CNTs; this mechanism has been considered to have synergy of both
loaded on substrates with GO via π–π stacking for the enhancement of “physical” and “chemical” effects [84]. The physical mechanisms include
bone regeneration properties. Ten et al. conjugated GO to modified ti- a sharp edge-mediated cutting effect and cell entrapment [85–87].
tanium with the aid of 3-APTES functional groups and then loaded Smaller-sized GO with a higher edge density has a stronger cutting effect,
aspirin, which has a benzene ring, onto the Ti-GO surface via π-π stack- while larger-sized GO with wider lateral dimensions has a higher po-
ing. The release of aspirin was sustained for three days, and in vitro cell tential to entrap bacterial cells [88,89]. He et al. observed common
studies revealed that aspirin-loaded Ti-GO materials promoted MC3T3- dental pathogens, including S. mutans, P. gingivalis and F. nucleatum,
E1 cell proliferation and osteogenic differentiation [70]. In recent exposed to GO nanosheet dispersion [90] (Fig. 2). The TEM images
years, inspired by mussels, Han et al. introduced polydopamine (PDA), indicate a common intracellular density decrease in the GO-treated
which has a structure similar to that of mussel adhesion proteins, to groups, which indicates the loss of intracellular substances and is
GO-based titanium modification. The molecular mechanism is thought to considered the result of the destruction of the cell wall and membrane.
involve the Michael addition and/or a Schiff base reaction, hydrogen Oxidative stress is a chemical mechanism that contributes substantially
bonding, or π-π stacking [78]. Because of its universal adhesiveness, PDA to the antibacterial activity of GO. Graphene-based materials are able to
served as an intermediate layer and provided biointeractive and chemi- oxidize GSH; this process induces superoxide anion-independent oxida-
cally reactive surfaces for secondary modification of GO [71]. Finally, tive stress in bacterial cells [91]. In contrast to the physical mechanisms,
chitosan-coated GelMS (CGelMS) that encapsulated BMP-2 and vanco- oxidative stress has no significant relationship with GO size; however, it
mycin (Van) were assembled on the GO/Ti scaffold via electrostatic in- can be substantially inhibited by reductive agents. In addition to the
teractions, which not only facilitated CGelMS immobilization but also physical mechanism, there is a parabolic relationship between the size
favored cell activity. The scaffold achieved independent release of BMP-2 and antibacterial activity of GO [88], and further studies remain to be
and Van, which was demonstrated by the superior in vitro and in vivo conducted to optimize the GO size for the highest degree of antibacterial
osteogenic properties [71]. activity.
Finally, GO has also been studied in gene delivery systems with safety
and high transfection activity. Pristine GO has been reported to be able to 3.4. Osteogenic differentiation properties
absorb single-stranded DNA and RNA through π–π stacking [79]. Poly-
ethylene glycol (PEG) and polyethylenimine (PEI) dual-functionalized The use of GO as a potential material for tissue engineering and
graphene oxide (GO) (nGO-PEG-PEI) are promising siRNA vectors. regenerative medicine has increased rapidly in recent years, particularly
Zhang et al. successfully synthesized GO that was dually-functionalized in directing the fate of stem cells [92]. Many studies have demonstrated
by PEG and PEI as a novel siRNA nanovector for implant surface bio- the advantages of GO in bone tissue engineering. La et al. showed that GO
modification through cathodic electrodeposition. When loaded with can reduce the required dose and optimize the use of bone morphoge-
osteogenic Ckip-1 siRNA, the coated implant enhanced the osteogenic netic protein 2 (BMP-2) to induce osteogenic differentiation in MSCs

283
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Fig. 3. (A) Radiographic evaluation of the implants on various samples after treatment for 4 weeks; (B) micro-CT tomography analysis of various samples after
treatment for 4 weeks; (C) Masson staining of longitudinal sections from various samples after treatment for 4 weeks; (D) VG staining of transverse sections from
various samples after treatment for 4 weeks [112]. Copyright © 2019 American Academy of Periodontology.

[93]. GO has been proven to induce BMSC, PDLSC and DPSC osteogenic graphene oxide to confirm the influences on mechanical properties.
differentiation in vitro [94–96]. However, the molecular mechanisms Alshahrani et al. used SEM, micro-Raman spectroscopy and microtensile
have not reached a consensus. It has been shown that GO can promote bond strength analysis to assess the differences between GO-added ad-
osteogenic differentiation of MSCs by enhancing the immunomodulatory hesive and control adhesive. The results indicate that the GO-modified
properties of MSCs [97]. Some researchers believe that hydroxyl and adhesive shows comparable bond strength and durability of resin
epoxy groups on the planes and carboxylic acid groups at the edges dentine bonds [107]. GO was also added to an adhesive containing 5 wt%
enhance interactions of GO with molecules and proteins in osteogenic HA at 0.5 wt% (HA-GO-0.5 %) and 2 wt% GO (HA-GO-2%). The
induction medium through hydrophobic and electrostatic interactions HA-GO-2% group showed uniform diffusion in adhesive and comparable
[98–100], potentially enhancing stem cell osteogenic differentiation resin tag development to the controls. The addition of GO has been
[101,102]. proven to provide a higher μTBS and adequate durability [106]. Apart
from adhesives, the primers that are used for surface pretreatment and
therefore require long-term bonding of resin composites to dental pros-
3.5. Odontogenic differentiation properties theses remain to be optimized. Khan et al. infused GO sheets into
commercially available silane premiers and influenced the surface
Inspired by the properties of GO to promote osteogenic differentia- morphology, which was observed to exhibit increased surface roughness
tion, our group tried to investigate its potential odontogenic differenti- and slightly increased water contact angle measurements. The highest
ation properties. First, we prepared a modified titanium-based material enclosed mold shear bond strength (EM-SBS) values were found, with
using the microarc oxidation (MAO) technique and self-assembling GO. mean (SD) EM-SBS values of 26.4 (3.7) MPa and 21.5 (5.1) MPa after 2
The upregulation of markers in DPSCs cultured on the group with GO, and 4 months of storage, respectively, which indicates improved me-
including dentin sialophosphoprotein (DSPP), dentine matrix protein 1 chanical properties [105]. Remarkably, Nizami et al. assessed the effi-
(DMP-1), ALP, OPN, and OCN, demonstrated a positive correlation with cacy of functionalized graphene oxide (f-GO) nanocomposites on the
GO content on coatings [103]. A similar study was reported by Ahn et al., decalcification of dentin, such as GO-silver (Ag), GO-Ag-calcium fluoride
who built mesoporous bioactive glass nanoparticle (MBN)/GO compos- (CaF2), GO-CaF2, GO-zinc, and GO-tricalcium phosphate (Ca3(PO4)2)
ites. Both the mRNA and protein levels of odontogenic markers showed [108]. The abilities of GO-Ag, GO-Ag-CaF2, and GO-CaF2 nano-
upregulation in hDPSCs in response to growth on the MBN/GO com- composites were most preventive for decalcification. In addition, GO-Ag
posites. Furthermore, they also revealed that the Wnt/β-catenin signaling and GO-Ag-CaF2 almost completely inhibited S. mutans growth.
pathway is involved in the mechanism behind the odontoblast differen- Furthermore, these f-GO nanocomposites exhibited less or no discolor-
tiation property of the material. Radunovic et al. synthetized collagen ation of dentin, although commonly used silver diamine fluoride causes
membranes coated with GO. Unlike the group that did not have a GO discoloration of dentin to black. The research represents an attractive and
coating, the GO-coated group exhibited not only biocompatibility and interesting direction of GO clinical transformation in dentistry, which
osteoblast differentiation but also odontoblast differentiation [104]. innovatively play to the strengths of GO and make up for the weaknesses
of traditional dental materials.
4. The application of GO in dentistry and related tissue
engineering
4.2. Antibacterial applications of GO
4.1. Cooperation of conventional dental materials and GO
Apart from the GO aqueous dispersion mentioned before, the appli-
In modern dentistry, tremendous efforts have been made to prevent cation of GO coatings or mixed components utilizing other mechanisms
oral diseases and promote oral hygiene. Restorative materials with in dental biomaterial and tissue engineering scaffolds for the enhance-
excellent biological properties, improved mechanical properties and ment of antimicrobial adhesives has also been explored. Introducing
longer service life have always been sought. The mechanical influence of either hydrophilic or zwitterionic surfaces generating a hydration layer
GO in dental materials should be confirmed before introducing it in on the surface are two usually adopted strategies. A physical or energetic
practice and utilizing its multiple biological properties. Conventional barrier preventing microbial adhesion can be created by this tightly
materials in dentistry, such as resin-based composites [47], silane bound water layer [107,109]. Oxygen-containing functional groups such
primers [105], and adhesives [106,107], have been combined with as the hydroxyl (-OH) and carboxyl (-COOH) groups on GO represent the

284
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Fig. 4. The GO-Cu nanocomposite-coated CPC scaffolds. (a) The appearance of the CPC scaffold under a stereoscopic zoom microscope. (b) The appearance of the GO-
coated CPC scaffold under a stereoscopic zoom microscope. (c) SEM observation of the GO-Cu-coated CPC scaffold, and nanoscale Cu particles can be observed under
higher magnification. (d) Release kinetics of the Cu ions from the GO-Cu-coated CPC scaffolds (n ¼ 6; *represents p < 0.05; **represents p < 0.01) [127].© 2016
WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. Implantation of GO scaffold to extraction socket. (e) Photograph of implanted scaffold. (g) Macroscopic view at 2
weeks postsurgery. (f) Radiographic images immediately after operations. (h) Radiographic image at 2 weeks postsurgery. The socket applied with the GO scaffold
showed increased radiopacity. Abbreviation: GO, graphene oxide [129]. Copyright © 2016, Dove Medical Press Limited.

capability of introducing hydrophilicity [110]. Qian et al. processed pure graphene as a photothermal agent for effective bacterial capture and
titanium with NaOH and APS to endow it with a positively charged killing [123]. Ran et al. demonstrated a hyaluronidase-triggered photo-
surface, which caused GO, with its oxygen-containing functional groups thermal platform based on AgNPs and graphene oxide (GO), which
and negatively charged surfaces, to adhere to the titanium surfaces resulted in high mortality of S. aureus as well as low toxicity to
through nucleophilic substitution reactions and electrostatic interactions mammalian cells under locally raised temperatures upon illumination
[111]. Minocycline hydrochloride was then loaded on the GO-modified with NIR light [124].
titanium surface, which showed slow release behavior; the observed
superior antibacterial activity against S. aureus, S. mutans and E. coli is 4.3. Osteogenic GO coating on dental implants and tissue-engineered
considered to be a synergistic effect of contact killing by GO and release scaffolds
killing by minocycline hydrochloride [111]. Furthermore, they estab-
lished a peri-implantitis model in beagles and treated them with implants Successful dental implant treatment is highly dependent on stable
that were equipped with abutments coated with graphene oxide film and osseointegration and reliable infection inhibition. In the process of dental
loaded with minocycline hydrochloride [112]. Compared to other implant treatment, failure may occur under the negative effects of oral
groups, the MH/GO/Ti group demonstrated the least marginal bone loss bacteria such as E. coli, C. albicans, S. aureus, and S. mutans or marginal
in radiographic analysis and nearly no neutrophils in the histological bone loss due to the poor osteoinduction property of conventional tita-
analysis [112] (Fig. 3). nium material. Among the nanocarbons that share antibacterial proper-
The oral cavity, including teeth, root canals, mucosa, periodontal ties, graphenic materials also hold exceptional promise in biomaterials
tissues, saliva and dentures, is a complex ecosystem. The oral bacteria due to their unique physicochemical properties, which include hydro-
themselves and substances produced or gathered by them can form oral philic functional groups, considerable mechanical strength, large surface
bacterial biofilms [113]. Pure GO nanosheets have been found to be toxic area and excellent biocompatibility [119,120]. In recent years, many
to the development of mature biofilms from planktonic cells [114]. publications have illustrated the enhanced osteogenic properties of
Bacterial biofilms have high pathogenicity because they are less sus- GO-containing composites. Biomimetic modifications could also offer
ceptible to antibiotics and even more resistant to physical treatments mechanical and chemical environments encouraging osteogenesis.
[115–117]. Qin et al. substantially removed polymicrobial biofilms that GO-based materials have become a promising regeneration scaffold
remained on previously contaminated titanium surfaces by utilizing competitor for bone tissue engineering.
256 μg/mL GO dispersion with brushing. The bone marrow–derived Due to the attractive properties of GO, some studies have introduced
mesenchymal stem cell osteogenic potential was regained or even it as a coating on dental and orthopedic implants as well as on bone tissue
enhanced on the titanium surfaces treated in vitro, which represents engineering scaffolds. A pioneering study demonstrated that graphene
another application of GO nanosheets in addition to killing planktonic and GO can accelerate MSC osteogenic differentiation to different de-
pathogens and inhibiting the formation of bacterial biofilms, and this grees due to π–π stacking, hydrogen bonding, and electrostatic in-
application might provide a new strategy for treating periimplantitis teractions with proteins [121], which may be the underlying mechanism
[118]. Di Giulio et al. reported that during the biofilm formation, GO supporting the coating application.
reduces the S. aureus and P. aeruginosa growth of 55.05 %  4.73 and As a widely accepted material applied in dental and orthopedic im-
44.18 %  3.91 compared to the control. In mature biofilm, GO affects S. plants, Ti has been modified in many strategies to enhance its biocom-
aureus and P. aeruginosa by reducing their growth of 70.24 %  4.47 and patibility and osteointegration. GO has been proven to be an attractive
63.68 %  17.56, respectively [22]. surface decoration on titanium, which is usually deposited electropho-
Graphene consists of a single layer of tightly packed carbon atoms and retically [122] or physically [123]. Recently, Carlo et al. activated tita-
possesses unique optical properties, which gives it the capability to nium surfaces with piranha solution or using a UV/ozone lamp and
absorb light irradiation and release it as heat [119–123]. Therefore, the coated them with GO via covalent bond formation. In the presence of a
photothermal effect has become another mechanism of GO activity in GO coating, the titanium discs showed outstanding properties in pro-
bacterial therapy. Wu et al. applied magnetic nanoparticle-functionalized moting the osteogenic differentiation of DPSCs, which was indicated by

285
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Fig. 5. SEM images of the newly formed


bone growth into the (a), (b), (c) scaffold
HA/rGO-6/0.3 and (d), (e), (f) scaffold HA-6
(M means material, NB means new bone).
Rendered 3D images and representative
tomographic images of micro-CT of the
implanted scaffold (g) HA/rGO-6/0.3 and
(h) HA-6 after 8 weeks. (i) Quantification of
newly formed bone in the implant. BV/TV
refers to bone volume/total volume, Tb. Th
refers to the trabecular thickness and Tb. Sp
refers to the trabecular separation. (j) Dia-
gram of the HA/rGO composite scaffold
substitution and new bone formation mech-
anism. (k) Mechanical test performance of
the surgical femur integrated with scaffold
HA/rGO-6/0.3 after 6 months. Reprinted
with permission from Ref. [148]. Copyright
© 2019, American Chemical Society.

the increased gene expression of TGFβ and BMP2 at the early stage and 4.4. Tissue-engineered scaffolds consisting of GO composites
the positive regulation of the RUNX2 transcription factor. Furthermore,
following RUNX2 upregulation, the subsequent SP7 increased only when Some studies also reported that GO rich in functional groups such as
DPSCs were cultured on titanium surfaces modified with GO [125]In carbonyls (COOH) and hydroxyls (OH) can provide biomimetic
addition to the three strategies mentioned above, Li et al. also introduced mineralization using simulated body fluid (SBF), which has many active
large particle sandblasting and acid etching (SLA) combined with the sites [131]. Remarkably, graphene particles have the ability to induce
ultrasonic atomization spraying technique, which is commonly used in stem cell osteogenesis similar to that of bone morphogenic protein
clinical practice, in GO coatings. The GO coating endowed the SLA tita- (BMP-2) and other bioactive inorganic materials, such as hydroxyapatite
nium surface with enhanced proliferation, adhesion and osteogenic dif- and nanosilicates [132,133]. In addition, the graphene family can exert
ferentiation of BMSCs in vitro, which has been proven to be involved in distinct molecular effects on immune cells [134–136], which is advan-
the FAK/P38 pathways. In vivo, the SLA/GO implants also showed tageous for new immune-based strategies in osteogenesis and bone
excellent osteointegration performance [126]. regeneration.
GO coating has also been applied in modifying scaffolds for bone GO has been used to form composites with biopolymers such as
tissue engineering; such scaffolds include CaP [127,128], collagen [129] collagen [131,137], chitosan [138], gelatin [139] and alginate [140].
and poly(3-hydroxybutyrate-Co-4-hydroxybutyrate) [130]. Our group The introduction of GO can help enhance the mechanical properties of
fabricated aqueous soluble graphene oxide-copper nanocomposites biopolymer materials so they can better bear the force and provide
(GO-Cu) and used them to coat porous calcium phosphate (CaP) scaf- possibilities for different designed constructures when serving as scaf-
folds. The composite scaffolds therefore obtained the ability to release Cu folds for bone tissue engineering. Recently, our group prepared
ions in the long term and were reported to enhance the adhesion and gelatin-reduced graphene oxide (GOG) and constructed composites that
osteogenic differentiation of rat BMSCs. Remarkably, by activating the mimicked the procallus by combining GOG with a photocrosslinked
Erk1/2 signaling pathway, the GO-Cu nanocomposites upregulated the gelatin hydrogel [139]. The bidirectional differentiation of BMSCs,
expression of Hif-1α in BMSCs, which resulted in the secretion of VEGF including osteogenesis and angiogenesis, was proven to be activated
and BMP-2 proteins. Finally, when transplanted into rats with through the Erk1/2 and AKT pathways. Furthermore, due to the large
critical-sized calvarial defects, the composite scaffolds (CPC/GO-Cu) specific surface area and the strong π-π conjugation with the drug mol-
promoted angiogenesis and osteogenesis [127](Fig. 4 a-d). Nishida et al. ecules [141,142] as well as the tendency of GO to be taken up by cells
fabricated a GO-coated collagen sponge scaffold and then implanted it through endocytosis [136], we also loaded the GOG with methyl vanil-
into a dog tooth extraction socket. Dog bone formation tests showed that late (MV), which contributed to the bioactive signals of the biomimetic
1 μg/mL GO scaffold implantation enhanced bone formation. New bone procallus by priming the osteogenesis of BMSCs [139]. Another study
formation following GO scaffold implantation was enhanced fivefold demonstrated that the synthesis of injectable hydrogels containing
compared to that in control subjects [129] (Fig. 4e-h). poly(N-isopropylacrylamide) (PNIPAAm)-based copolymers, graphene

286
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Table 1
Combination of different native materials and GO.
Native materials Incorporated Cell/bacteria Biological effects GO preparing method Ref.
materials

GO dispersion N/A S. mutans; Antimicrobial activity Modified Hummer's method [90]


P. gingivalis;
F. nucleatum
GO suspensions N/A S. mutans Antimicrobial activity Bath sonication [88]
GO coating N/A fibroblast cells; Biocompatible; High-density atmospheric plasma [162]
S. mutans Antibacterial deposition
PMMA GO E. coli; Sustained antimicrobial-adhesive Purchased [163]
C. albicans;
S. aureus;
S. mutans;
Hyaluronidase Ag NP/GO S. aureus Antibacterial Purchased [124]
composites
GO dispersion N/A S. mutans; Antibacterial; Purchased [118]
P. gingivalis; Reosteogenesis
F. nucleatum;
BMSCs
Titanium plates GO S. aureus; Antibacterial Purchased [111]
S. mutans;
E. coli;
Human gingival
fibroblasts
Type I collagen GO rBMSCs Biomineralization; Modified Hummer's method [46]
Biocompatibility;
Osteogenic ability
Chitosan GO/HAP/Au C3H10T1/2; Biocompatibility; Hummer's method [138]
E.coli; Osteogenic Differentiation;
S.mutans; Antibacterial
S.aureus;
P.aeruginosa
Alginate GO hMSCs Proliferation and survival in oxidative Purchased [140]
environment;
Osteogenic differentiation
Hydrogel Chitosan/GO hDPSCs Proliferation; Differentiation to the Hummer's method [143]
osteoblasts
Fibrin hydrogel GO, IONPs, nHAP NIH-3T3; Biocompatible; Modified Hummer's method [144]
MG-63 cells Osteogenic property;
Calcium deposits
HA rGO MC3T3-E1 cells Increasing ALP; mineralization; osteopontin; Modified Hummer's and Offeman's [147]
osteocalcin methods
β-TCP GO hBMSCs Proliferation; Purchased [150]
Osteogenic property
HA rGO BMSCs Improved adhesion; proliferation; Two-step oxidation of NG [148]
spontaneous osteogenic differentiation flakes
CaP GO hMSCs; monocytes Activation of monocytes; Purchased [151]
Osteogenesis
Poly(L-lactic-co-glycolic Tussah silk fibroin; Mouse MSCs Enhanced adhesion; proliferation; Modified Hummer's method [157]
acid) GO ALP;
Mineral deposition
Titanium GO DPSCs Biocompatibility; Purchased [125]
Osteogenic differentiation
Titanium GO rBMSCs Proliferation; N/A [126]
Adhesion;
Osteogenic differentiation;
Osteointegration
Titanium GO PDLSCs Proliferation; Modified Hummer's method [96]
Osteogenic differentiation;
CaP Cu-GO BMSCs Adhesion; Modified Hummer's method [127]
Osteogenic differentiation
CaP rGO MC3T3-E1 cells Accelerated new bone formation Modified Hummer's and Offeman's [128]
methods
P34HB GO BMSCs Improved cellular performance; Purchased [130]
Osteogenic differentiation
Titanium GO/BMP-2 hBMMSCs Osteogenic differentiation; Modified Hummer's method [67]
Osteointegration
Titanium plates GO/BMP-2/SP hBMMSCs Osteogenic differentiation; Modified Hummer's method [68]
Osteointegration
Silk fibroin GO/P24 BMSCs Biocompatibility; Modified Hummer's method [69]
Adhesion;
Proliferation;
Osteogenic differentiation
Titanium GO/aspirine MC3T3-E1 cells Proliferation; osteogenic differentiation Modified Hummer's method [70]
Titanium GO/BMP-2/ BMSCs Osteogenic property; Modified Hummer's method [71]
vancomycin Antibacterial
Titanium nGO-PEG-PEI/siRNA MC3T3-E1 cells Osteogenic differentiation; Modified Hummer's method [72]
Osteointegration

287
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

oxide (GO) and chitosan through free-radical copolymerization resulted preosteoblasts [155]. The GO incorporated into poly(vinyl alcohol) and the
in materials with porosity, swelling and biocompatibility. Increased increased porosity formed by selective laser sintering improved the me-
expression of osteogenic genes such as Runx 2 and OCN as well as ALP chanical properties of the composite as well as the attachment and growth of
activity and calcium deposition in hDPSCs demonstrated osteogenic po- MG-67 human bone fibroblasts [156]. In recent years, Shao et al. fabricated
tential [143]. A similar conclusion was also drawn by Pathmanapan an ultrafine nanofiber scaffold by electrospinning a blend of
et al., who incorporated GO with a fibrin hydrogel by the interaction poly(L-lactic-co-glycolic acid), tussah silk fibroin, and graphene oxide (GO).
between the –COOH groups of the GO and the -NH2 groups of fibrin The data indicated that the modifications boosted the Young's modulus and
[144]. Chitosan is an attractive biopolymer with properties such as tensile strength by nearly 4-fold and 3-fold, respectively, and enhanced
nontoxicity, biodegradability, biocompatibility, and flexibility, but its adhesion and proliferation in mouse mesenchymal stem cells along with
poor mechanical properties limit its application in tissue engineering. promoted biomineralization-relevant ALP and mineral deposition [157].
Some strategies, including constructing nanocomposite polymeric films In addition to being used in the decoration of single ceramic or
and nanoparticles, have been used to improve the mechanical properties polymeric materials, in recent years, GO has also been incorporated into
[145,146]. Prakash et al. adopted a hydrothermal method to uniformly composites to enhance their properties by utilizing the different advan-
disperse nanorod-like HAP and Au NPs on the surfaces of GO and pre- tages of different materials. The same materials, such as the combination
pared GO/HAP/Au ternary nanocomposites. Furthermore, they incor- of gelatin and alginate [158], chitosan and hydrogel [159], and different
porated the obtained GO/HAP/Au ternary nanocomposites with kinds of materials, including the combination of hydroxyapatite and silk
polymeric film by a gel casting technique, the product of which exhibited fibroin [160], hydroxyapatite and chitosan [161], have been reported to
good chemical stability, improved mechanical properties, good antibac- successfully serve as scaffold materials used in bone tissue engineering.
terial properties against infection-causing pathogens and improved
osteoblast cell viability [138]. 5. Conclusions and future perspectives
Other strategies have included combining GO with ceramic materials
to enhance the osteogenesis of composites. The combination of GO with We have highlighted the recent progress in GO as strategies to
different kinds of ceramic materials has been reported, including HA improve the properties of dental materials and bone tissue engineered
[147–149] and CaP [150,151]. Owing to its similarities to the inorganic scaffolds. First, we described its physicochemical properties, which are
components of bone tissues and its high biocompatibility, degradability the basis of its diverse applications. Then, the biological properties were
and osteoconductivity [152,153], hydroxyapatite (HA) was chosen discussed, including biocompatibility, controlled release of medicine,
explicitly by many researchers as a partner for GO to create and enhance antibacterial properties, osteogenic differentiation properties and odon-
the mechanical and osteogenic properties. For instance, Lee et al. soni- togenic differentiation properties. After a preliminary description of its
cated prepared reduced graphene oxide (rGO) and mixed it with hy- applications, we further described the applications of GO in dentistry and
droxyapatite (HA). The in vitro behaviors of the composites have been related tissue engineering. Antibacterial properties were discussed due to
proven, including promoting the differentiation of MC3T3-E1 preosteo- their importance in dentistry applications, as dentistry covers areas with
blasts and increasing alkaline phosphatase, mineralization, osteopontin large amounts of bacteria. Researchers then investigated the cooperation
and osteocalcin. Enhanced in vivo formation of new bone in of GO and conventional dental materials mainly in terms of the effects on
full-thickness calvarial defects without inflammatory responses after the the mechanical properties. With the increasing number of attempts at GO
implantation of rGO/HA grafts was also observed [147]. In the study application, GO coatings for dental implants and tissue engineering
conducted by Zhou et al., when incorporated with rGO, the hierarchically scaffolds were proven to promote bone formation. Scaffolds consisting of
porous hydroxyapatite hybrid scaffold improved the adhesion, prolifer- GO were then summarized according to the matrices, which included
ation and spontaneous osteogenic differentiation of BMSCs. After im- ceramic, polymeric, biopolymeric materials and composite materials
plantation in vivo, the scaffold was crushed, degraded and wrapped by (Table 1).
the newly formed bone, the breaking strength of which with the healing Although the integration of GO within scaffolds can reduce problems
of scaffold HA/rGO-6/0.3 was satisfactory (20.0 MPa) and much higher related to physical properties, including cytotoxicity, biodistribution,
than that of HA-6 (9.8 MPa)[148]. (Fig. 5). biotransformation and immune response, there are still no distinct
In addition to HA, calcium phosphate (CaP) has also been introduced chemical structures for the prepared GO, and the variation resulting from
in combination with GO. Wu et al. successfully prepared β-TCP-GO disks methods or laboratory conditions requires a deep exploration to ensure
and 3D-printed scaffolds through a simple GO/water suspension soaking safety in further clinical use. In addition, recently, the delivery of genes
method and heat treatment. Significantly enhanced proliferation, alka- loaded on GO showed great potential, and this method may have great
line phosphatase activity and osteogenic gene expression of human potential in dental tissue engineering when used to deliver osteogenic-
marrow stromal cells (hBMSCs) was observed in the β-TCP-GO disk group related or odontogenic-related genes on implants or scaffolds. With
compared to the control group with only β-TCP and without GO modi- respect to the novel odontogenic properties of GO, although several kinds
fication, which was attributed to the activation of the Wnt/β-catenin of materials have been studied as substrates in vitro, there is still no in
signaling pathway. When implanted in the cranial bone defects of rab- vivo experiment to verify the properties and investigate the possibility of
bits, β-TCP-GRA scaffolds led to an increased rate of new bone formation their clinical application. In summary, the development of GO is an
compared to β-TCP controls in vivo [150]. In addition to MSCs, mono- exciting and promising material with the potential to revolutionize
cytes have also been demonstrated to be involved in the osteogenic clinical practice, which remains to be further explored.
properties of GO owing to their immunological properties. Bordoni et al.
found that in the presence of monocytes, the ALP activity and the
Declaration of competing interest
expression of osteogenic markers increased on the GO-CaP material along
with the overproduction of Oncostatin M, a pro-osteogenic factor pro-
We declare that we have no financial and personal relationships with
duced by monocytes [151].
other people or organizations that can inappropriately influence our
Polymeric materials are another matrix that has been studied for
work, there is no professional or other personal interest of any nature or
incorporation into GO. GO-doped poly(lactic-co-glycolic acid) nanofiber
kind in any product, service and/or company that could be construed as
scaffolds formed through electrospinning were reported to increase the
influencing the position presented in, or the review of, the manuscript
adhesion and proliferation of human MSCs [154].
entitled, “Graphene Oxide: A Promising Material in Dentistry and Related
Polydopamine-functionalized rGO increased the osteogenesis of MC3T3-E1
Tissue Regeneration.

288
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

Acknowledgements [28] Yanwu, Zhu, Shanthi, et al., Graphene-based materials: graphene and graphene
oxide: synthesis, properties, and applications, Adv. Mater. 22 (35) (2010), 35/
2010). Advanced Materials.
This work was supported by the National Key Research and Devel- [29] W. Hu, Q. Huang, C. Fan, Nanomaterial-Based Antibacterial Paper[M], Springer
opment Program of China (2016YFC1102900), the National Natural Berlin Heidelberg, 2012.
Science Foundation of China (81921002, 82022015,81900960) and the [30] P. Bradder, S.K. Ling, S. Wang, et al., Dye adsorption on layered graphite oxide,
J. Chem. Eng. Data 56 (1) (2011) 138–141.
Natural Science Foundation of Jiangsu Province (BK20180671). [31] V.B. Mohan, K-t Lau, D. Hui, D. Bhattacharyya, Graphene-based materials and
their composites: a review on production, applications and product limitations,
References Compos. B Eng. 142 (2018) 200–220.
[32] Li Fang, Jinlu, Controllable synthesis of reduced graphene oxide, Curr. Appl.
Phys.: Off. J. Kor. Phys. Soc. 16 (9) (2016) 1152–1158.
[1] T. Lancet, GBD 2016 disease and injury incidence and prevalence collaborators, [33] S. Park, R.S. Ruoff, Chemical methods for the production of graphenes, Nat.
Global Health Metrics 390 (2017) 1211, 10100. Nanotechnol. 4 (4) (2009) 217–224.
[2] J.L. Ferracane, Resin-based composite performance:are there somethings we can't [34] Deepali, Sharma, Suvardhan, et al., Insight into the biosensing of graphene oxide:
predict? Dent. Mater. 29 (1) (2013) 51–58. present and future prospects, Arab. J. Chem. 9 (2) (2016) 238–261.
[3] I. Miletic, I. Anic, S. Pezelj-Ribaric, S. Jukic, Survival of the branemark implant in [35] D. Chen, H. Feng, J. Li, Graphene oxide: preparation, functionalization, and
partially edentulous jaws: a 10-year prospective multicenter study, Int. J. Oral electrochemical applications, Chem. Rev. 112 (11) (2012) 6027–6053.
Maxillofac. Implants 14 (1999) 639–645. [36] B. Konkena, S. Vasudevan, Understanding aqueous dispersibility of graphene
[4] K.D.J. Snauwaert, D. van Steenberghe, M. Quirynen, I. Naert, Time dependent oxide and reduced graphene oxide through pKa measurements, J. Phys. Chem.
failure rate and marginal bone loss of implant supported prostheses: a 15-year Lett. 3 (7) (2012) 867–872.
follow-up study, Clin. Oral Invest. 4 (2000) 13–20. [37] J.A. Sungjin Park, Inhwa Jung, Richard D. Piner, Sung Jin An, Xuesong Li,
[5] E.S.C.S. Rosenberg, N. Elian, Z.N. Jalbout, S. Froum, C.I. Evian, A comparison of Aruna Velamakanni, Rodney S. Ruoff, Colloidal suspensions of highly reduced
characteristics of implant failure and survival in periodontally compromised and graphene oxide in a wide variety of organic solvents, Nano Lett. 9 (4) (2009)
periodontally healthy patients: a clinical report, Int. J. Oral Maxillofac. Implants 1593–1597.
19 (2004) 873–879. [38] X. Zhang, J. Yin, P. Cheng, et al., Distribution and biocompatibility studies of
[6] T.P.L. Berglundh, B. Klinge, A systematic review of the incidence of biological and graphene oxide in mice after intravenous administration[J], Carbon 49 (3) (2011)
technical complications in implant dentistry reported in prospective longitudinal 986–995.
studies of at least 5 years, J. Clin. Periodontol. 29 (2002) 197–212. [39] Z. Liu, J. Robinson, X. Sun, H. Dai, PEGylated nano-graphene oxide for delivery of
[7] A.S.P. O'Mahony, Osseointegrated implant failures, J. Ir. Dent. Assoc. 45 (1999) water insoluble cancer drugs, J. Am. Chem. Soc. 130 (33) (2008) 10876–10877.
44–51. [40] M. Gu, Y. Liu, T. Chen, F. Du, X. Zhao, C. Xiong, et al., Is graphene a promising
[8] A.L.N. Mombelli, The diagnosis and treatment of peri-implantitis, Periodontol. nano-material for promoting surface modification of implants or scaffold materials
2000 17 (1998) 63–76. in bone tissue engineering? Tissue Eng. B Rev. 20 (5) (2014) 477–491.
[9] A M, Microbiology and antimicrobial therapy of peri-implantitis, Periodontol. [41] M. Li, Y. Wang, Q. Liu, Q. Li, Y. Cheng, Y. Zheng, et al., In situ synthesis and
2000 28 (2002) 177–189. biocompatibility of nano hydroxyapatite on pristine and chitosan functionalized
[10] A.M.R.S. Roos-Jansaker, J. Egelberg, Treatment of peri-implant infections: a graphene oxide, J. Mater. Chem. B 1 (4) (2013) 475–484.
literature review, J. Clin. Periodontol. 30 (2003) 467–485. [42] J. Ruan, X. Wang, Z. Yu, et al., Enhanced physiochemical and mechanical
[11] N.P.M.A. Lang, M.S. Tonetti, U. Bragger, C.H. Hammerle, Clinical trials on performance of chitosan-grafted graphene oxide for superior osteoinductivity,
therapies for peri- implant infections, Ann. Periodontol. 2 (1997) 343–356. Adv. Funct. Mater. 26 (7) (2016) 1085–1097.
[12] C.S.B. Isehed, P. Lundberg, A. Holmlund, Surgical treatment of peri-implantitis [43] S.R. Shin, B. Aghaei-Ghareh-Bolagh, T.T. Dang, S.N. Topkaya, X. Gao, S.Y. Yang, et
using enamel matrix derivative, an RCT: 3-and 5-year follow-up, J. Clin. al., Cell-laden microengineered and mechanically tunable hybrid hydrogels of
Periodontol. 45 (2018) 744–753. gelatin and graphene oxide, Adv. Mater. 25 (44) (2013) 6385–6391.
[13] O.C.W.J. Koldsland, A.M. Aass, Surgical treatment of peri-implantitis: prognostic [44] D. Jiao, A. Zheng, Y. Liu, et al., Bidirectional differentiation of BMSCs induced by
indicators of short-term results, J. Clin. Periodontol. 45 (2018) 100–113. a biomimetic procallus based on a gelatin-reduced graphene oxide reinforced
[14] S. Chukwulebe, C. Hogrefe, The diagnosis and management of facial bone hydrogel for rapid bone regeneration, Bioact. Mater. 6 (7) (2020) 2011–2028. .
fractures, Emerg. Med. Clin. 37 (1) (2019) 137–151. Published 2020 Dec 31.
[15] M.G. Araújo, J. Lindhe, Dimensional ridge alterations following tooth extraction. [45] W. Zhang, B. Yin, Y. Xin, L. Li, G. Ye, J. Wang, et al., Preparation, mechanical
An experimental study in the dog, J. Clin. Periodontol. 32 (2005) 212–218. properties, and biocompatibility of graphene oxide-reinforced chitin
[16] D. Rolski Jk-J, P. Zawadzki, K. Zyci nska, E. Mierzwi nska-Nastalska, The monofilament absorbable surgical sutures, Mar. Drugs 17 (4) (2019).
Management of Patients after Surgical Treatment of Maxillofacial Tumors, 2016, [46] S. Liu, C. Zhou, S. Mou, J. Li, M. Zhou, Y. Zeng, et al., 405 Biocompatible graphene
p. 7, 2016. oxide-collagen composite aerogel for enhanced stiffness and in situ bone
[17] A.K. Novoselov Ksg, S.V. Morozov, D. Jiang, Y. Zhang, S.V. Dubonos, regeneration, Mater. Sci. Eng. C Mater. Biol. Appl. 105 (2019) 110137.
I.V. Grigorieva, A. Firsov, Electric field effect in atomically thin carbon films, [47] N. Ilie, C. Sarosi, M.C. Rosu, M. Moldovan, Synthesis and characterization of
Science 306 (2004) 666–669. graphene oxide-zirconia (GO-ZrO2) and hydroxyapatite-zirconia (HA-ZrO2) nano-
[18] S. Zhu Ym, W. Cai, X. Li, J.W. Suk, J.R. Potts, R.S. Ruoff, Graphene and graphene fillers for resin-based composites for load-bearing applications, J. Dent. 105
oxide: synthesis, properties, and applications, Adv. Mater. 22 (2010) 3906–3924. (2021) 103557.
[19] R. Mancinelli, E. Filippo, M. Tumedei, M. Marrone, S. Fulle, Human dental pulp [48] Y.F. AlFawaz, B. Almutairi, H.F. Kattan, et al., Dentin bond integrity of
stem cell osteogenic differentiation seeded on equine bone block with graphene hydroxyapatite containing resin adhesive enhanced with graphene oxide nano-
and melatonin, Appl. Sci. 11 (7) (2021) 3218. particles-an SEM, EDX, micro-Raman, and microtensile bond strength study,
[20] A. Di Crescenzo, S. Zara, C. Di Nisio, V. Ettorre, A. Ventrella, B. Zavan, et al., Polymers 12 (12) (2020) 2978. Published 2020 Dec 14.
Graphene oxide foils as osteoinductive stem cells substrate, Acs Appl. Bio. Mater. 2 [49] W. Qin, Y. Li, J. Ma, et al., Osseointegration and biosafety of graphene oxide
(4) (2019) 1643–1651. wrapped porous CF/PEEK composites as implantable materials: the role of surface
[21] H. Elkhenany, L. Amelse, A. Lafont, S. Bourdo, M. Caldwell, N. Neilsen, et al., structure and chemistry, Dent. Mater. 36 (10) (2020) 1289–1302.
Graphene supports in vitro proliferation and osteogenic differentiation of goat [50] H. Yan, X. Tao, Z. Yang, K. Li, H. Yang, A. Li, et al., Effects of the oxidation degree
adult mesenchymal stem cells: potential for bone tissue engineering, J. Appl. of graphene oxide on the adsorption of methylene blue, J. Hazard Mater. 268
Toxicol. 35 (4) (2015) 367–374. (2014) 191–198.
[22] M. Di Giulio, S. Di Lodovico, A. Fontana, et al., Graphene oxide affects [51] F. Emadi, A. Amini, A. Gholami, Y. Ghasemi, Functionalized graphene oxide with
Staphylococcus aureus and Pseudomonas aeruginosa dual species biofilm in chitosan for protein nanocarriers to protect against enzymatic cleavage and retain
lubbock chronic wound biofilm model, Sci. Rep. 10 (1) (2020) 18525. Published collagenase activity, Sci. Rep. 7 (2017) 42258.
2020 Oct 28. [52] X. Xu, X. Mao, Y. Wang, D. Li, Z. Du, W. Wu, et al., Study on the interaction of
[23] A. Sx, A. Xl, G.B. Lu, C. Jl, A. Ymd, E. Jl, et al., Graphene oxide as antibacterial graphene oxidesilver nanocomposites with bovine serum albumin and the
sensitizer: mechanically disturbed cell membrane for enhanced poration efficiency formation of nanoparticle-protein corona, Int. J. Biol. Macromol. 116 (2018)
of melittin, Carbon 149 (2019) 248–256. 492–501.
[24] T.R. Nayak, H. Andersen, V.S. Makam, et al., Graphene for controlled and [53] M. Lv, L. Yan, C. Liu, C. Su, Q. Zhou, X. Zhang, Non-covalent functio- nalized
accelerated osteogenic differentiation of human mesenchymal stem cells[J], ACS graphene oxide (GO) adsorbent with an organic gelator for co-adsorption of dye,
Nano 5 (6) (2011) 4670–4678. endocrine-disruptor, pharmaceutical and metal ion, Chem. Eng. J. 349 (2018)
[25] W.C. Lee, C. Lim, H. Shi, et al., Origin of enhanced stem cell growth and 791–799.
differentiation on graphene and graphene oxide[J], ACS Nano 5 (9) (2011) [54] D.Y. Lee, Z. Khatun, J.H. Lee, et al., Blood compatible graphene/heparin conjugate
7334–7341. through noncovalent chemistry, Biomacromolecules 12 (2) (2011) 336–341.
[26] S. Wu, N. Duan, X. Ma, Y. Xia, H. Wang, Z. Wang, et al., Multiplexed fluorescence [55] S. Stankovich, D.A. Dikin, R.D. Piner, K.A. Kohlhaas, A. Kleinhammes, Y. Jia, et al.,
resonance energy transfer aptasensor between upconversion nanoparticles and Synthesis of graphene-based nanosheets via chemical reduction of exfoliated
graphene oxide for the simultaneous determination of mycotoxins, Anal. Chem. 84 graphite oxide, Carbon 45 (7) (2007) 1558–1565.
(14) (2012) 6263–6270. [56] S. Yu, X. Wang, Y. Ai, Y. Liang, Y. Ji, J. Li, et al., Spectroscopic and theoretical
[27] H. Wang, T. Chen, S. Wu, et al., A novel biosensing strategy for screening G- studies on the counterion effect of Cu(ii) ion and graphene oxide interaction with
quadruplex ligands based on graphene oxide sheets, Biosens. Bioelectron. 34 (1) titanium dioxide, Environ. Sci.: Nano 3 (6) (2016) 1361–1368.
(2012) 88–93.

289
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

[57] T.M. Simsikova, Interaction of Graphene Oxide with Proteins and Applications of [88] C.H. Yu, G.Y. Chen, M.Y. Xia, et al., Understanding the sheet size-antibacterial
Their Conjugates, 2017. activity relationship of graphene oxide and the nano-bio interaction-based
[58] R. Di Carlo, S. Zara, A. Ventrella, et al., Covalent decoration of cortical membranes physical mechanisms, Colloids Surf. B Biointerfaces 191 (2020) 111009.
with graphene oxide as a substrate for dental pulp stem cells, Nanomaterials 9 (4) [89] M. Di Giulio, R. Zappacosta, S. Di Lodovico, et al., Antimicrobial and antibiofilm
(2019) 604. Published 2019 Apr 12. efficacy of graphene oxide against chronic wound microorganisms, Antimicrob.
[59] S.F. Oliveira, G. Bisker, N.A. Bakh, S.L. Gibbs, M.P. Landry, M.S. Strano, Protein Agents Chemother. 62 (7) (2018) e00547, 18.
functionalized carbon nanomaterials for biomedical applications, Carbon 95 [90] J. He, X. Zhu, Z. Qi, et al., Killing dental pathogens using antibacterial graphene
(2015) 767–779. oxide, ACS Appl. Mater. Interfaces 7 (9) (2015) 5605–5611.
[60] T. Fahmi, D. Branch, Z.A. Nima, D.S. Jang, A.V. Savenka, A.S. Biris, et al., [91] S. Liu, T.H. Zeng, M. Hofmann, et al., Antibacterial activity of graphite, graphite
Mechanism of graphene-induced cytotoxicity: role of endonucleases, J. Appl. oxide, graphene oxide, and reduced graphene oxide: membrane and oxidative
Toxicol. 37 (11) (2017) 1325–1332. stress, ACS Nano 5 (9) (2011) 6971–6980.
[61] R. Zhou, H. Gao, Cytotoxicity of graphene: recent advances and future perspective, [92] A. Halim, Q. Luo, Y. Ju, G. Song, A mini review focused on the recent applications
Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 6 (5) (2014) 452–474. of graphene oxide in stem cell growth and differentiation, Nanomaterials 8 (9)
[62] J.R. Kan Wang, Hua Song, Jiali Zhang, Wo Yan, Shouwu Guo, Daxiang Cui, (2018) 736. Published 2018 Sep. 18.
O. Access, Biocompatibility of graphene oxide, Nanoscale Res. Lett. 6 (2011) 1–8. [93] W.G. La, M.J. Jung, J.K. Yoon, et al., Bone morphogenetic protein-2 for bone
[63] K.H. Liao, Y.S. Lin, C.W. Macosko, C.L. Haynes, Cytotoxicity of graphene oxide regeneration – dose reduction through graphene oxide-based delivery[J], Carbon
and graphene in human erythrocytes and skin fibroblasts, ACS Appl. Mater. 78 (2014) 428–438.
Interfaces 3 (7) (2011) 2607–2615. [94] V. Rosa, H. Xie, N. Dubey, et al., Graphene oxide-based substrate: physical and
[64] A.M. D Imiev, L.B. Alemany, J.M. Tour, Graphene oxide. Origin of acidity, its surface characterization, cytocompatibility and differentiation potential of dental
instability in water, and a new dynamic structural model, ACS Nano 7 (1) (2013) pulp stem cells, Dent. Mater. 32 (8) (2016) 1019–1025.
576–588. [95] C. Wei, Z. Liu, F. Jiang, B. Zeng, M. Huang, D. Yu, Cellular behaviours of bone
[65] W. Zhang, Z. Guo, D. Huang, Z. Liu, X. Guo, H. Zhong, Synergistic effect of chemo- marrow-derived mesenchymal stem cells towards pristine graphene oxide
photothermal therapy using PEGylated graphene oxide, Biomaterials 32 (33) nanosheets, Cell Prolif 50 (5) (2017), e12367.
(2011) 8555–8556. [96] Q. Zhou, P. Yang, X. Li, H. Liu, S. Ge, Bioactivity of periodontal ligament stem cells
[66] J. Hong, N.J. Shah, A.C. Drake, et al., Graphene multilayers as gates for multi- on sodium titanate coated with graphene oxide, Sci. Rep. 6 (2016) 19343.
week sequential release of proteins from surfaces, ACS Nano 6 (1) (2011) 81–88. Published 2016 Jan 14.
[67] La Wan-Geun, S. Park, Hee-Hun Yoon, et al., Delivery of a therapeutic protein for [97] D. Xue, E. Chen, H. Zhong, et al., Immunomodulatory properties of graphene oxide
bone regeneration from a substrate coated with graphene oxide, Small 9 (2013). for osteogenesis and angiogenesis, Int. J. Nanomed. 13 (2018) 5799–5810,
[68] W.G. La, M. Jin, S. Park, H.H. Yoon, G.J. Jeong, S.H. Bhang, et al., Delivery of bone https://doi.org/10.2147/IJN.S170305. Published 2018 Sep. 26.
morphogenetic protein-2 and substance P using graphene oxide for bone [98] D.R. Dreyer, S. Park, C.W. Bielawski, R.S. Ruoff, The chemistry of graphene oxide,
regeneration, Int. J. Nanomed. 9 (Suppl 1) (2014) 107–116. Chem. Soc. Rev. 39 (1) (2010) 228–240.
[69] J. Wu, A. Zheng, Y. Liu, D. Jiao, D. Zeng, X. Wang, et al., Enhanced bone [99] D. Li, R.B. Kaner, Materials science. Graphene-based materials, Science 320
regeneration of the silk fibroin electrospun scaffolds through the modification of (5880) (2008) 1170–1171.
the graphene oxide functionalized by BMP-2 peptide, Int. J. Nanomed. 14 (2019) [100] J. Hong, N.J. Shah, A.C. Drake, et al., Graphene multilayers as gates for multi-
733–751. week sequential release of proteins from surfaces, ACS Nano 6 (1) (2012) 81–88.
[70] L. Ren, S. Pan, H. Li, Y. Li, L. He, S. Zhang, et al., 419 Effects of aspirin-loaded [101] W.C. Lee, C.H. Lim, H. Shi, et al., Origin of enhanced stem cell growth and
graphene oxide coating of a titanium surface on proliferation and osteogenic differentiation on graphene and graphene oxide, ACS Nano 5 (9) (2011)
differentiation of MC3T3-E1 cells, Sci. Rep. 8 (1) (2018) 15143. 7334–7341.
[71] L. Han, H. Sun, P. Tang, P. Li, C. Xie, M. Wang, et al., 420 Mussel-inspired [102] D. Mohammadrezaei, H. Golzar, M. Rezai Rad, et al., In vitro effect of graphene
graphene oxide nanosheet-enwrapped Ti scaffolds with drug-encapsulated gelatin structures as an osteoinductive factor in bone tissue engineering: a systematic
microspheres for bone regeneration, Biomater. Sci. 6 (3) (2018) 538–549. review, J. Biomed. Mater. Res. 106 (8) (2018) 2284–2343.
[72] L. Zhang, Q. Zhou, W. Song, K. Wu, Y. Zhang, Y. Zhao, 385 dual-functionalized [103] N. Sun, S. Yin, Y. Lu, W. Zhang, X. Jiang, Graphene oxide-coated porous titanium
graphene oxide based siRNA delivery system for implant surface biomodification for pulp sealing: an antibacterial and dentino-inductive restorative material,
with enhanced osteogenesis, ACS Appl. Mater. Interfaces 9 (40) (2017) J. Mater. Chem. B 8 (26) (2020) 5606–5619.
34722–34735. [104] M. Radunovic, M. De Colli, P. De Marco, C. Di Nisio, A. Fontana, A. Piattelli,
[73] W.G. La, S. Park, H.H. Yoon, et al., Delivery of a therapeutic protein for bone A. Cataldi, S. Zara, Graphene oxide enrichment of collagen membranes improves
regeneration from a substrate coated with graphene oxide, Small 9 (23) (2013) DPSCs differentiation and controls inflammation occurrence, J. Biomed. Mater.
4051–4060. Res. 105 (8) (2017 Aug) 2312–2320, https://doi.org/10.1002/jbm.a.36085. Epub
[74] J. Hong, N.J. Shah, A.C. Drake, et al., Graphene multilayers as gates for multi- 2017 May 30. PMID: 28380697.
week sequential release of proteins from surfaces, ACS Nano 6 (1) (2012) 81–88. [105] A.A. Khan, A.A. Al-Khureif, S.A. Saadaldin, et al., Graphene oxide-based
[75] D. Weber, A. Kotzsch, J. Nickel, et al., A silent H-bond can be mutationally experimental silane primers enhance shear bond strength between resin composite
activated for high-affinity interaction of BMP-2 and activin type IIB receptor, BMC and zirconia, Eur. J. Oral Sci. 127 (6) (2019) 570–576.
Struct. Biol. 7 (2007) 6. [106] A. Alshahrani, M.S. Bin-Shuwaish, R.S. Al-Hamdan, et al., Graphene oxide nano-
[76] C. Mücksch, H.M. Urbassek, Adsorption of BMP-2 on a hydrophobic graphite filler based experimental dentine adhesive. A SEM/EDX, Micro-Raman and
surface: a molecular dynamics study, Chem. Phys. Lett. 510 (4–6) (2011) microtensile bond strength analysis, J. Appl. Biomater. Funct. Mater. 18 (2020),
252–256. 2280800020966936.
[77] T. Utesch, G. Daminelli, M.A. Mroginski, Molecular dynamics simulations of the [107] A S C, B L L, C C Z, et al., Surface hydration: principles and applications toward
adsorption of bone morphogenetic protein-2 on surfaces with medical relevance, low-fouling/nonfouling biomaterials - ScienceDirect[J], Polymer 51 (23) (2010)
Langmuir 27 (21) (2011) 13144–13153. 5283–5293.
[78] Y. Liu, K. Ai, L. Lu, Polydopamine and its derivative materials: synthesis and [108] M.Z.I. Nizami, Y. Nishina, T. Yamamoto, Y. Shinoda-Ito, S. Takashiba,
promising applications in energy, environmental, and biomedical fields, Chem. Functionalized graphene oxide shields tooth dentin from decalcification, J. Dent.
Rev. 114 (9) (2014) 5057–5115. Res. 99 (2) (2020) 182–188.
[79] H. Yue, X. Zhou, M. Cheng, D. Xing, Graphene oxide-mediated Cas9/sgRNA [109] C. Leng, S. Sun, K. Zhang, S. Jiang, Z. Chen, Molecular level studies on interfacial
delivery for efficient genome editing, Nanoscale 10 (3) (2018) 1063–1071. hydration of zwitterionic and other antifouling polymers in situ, Acta Biomater. 40
[80] Z. Liu, J.T. Robinson, X. Sun, H. Dai, PEGylated nanographene oxide for delivery (2016) 6–15.
of water-insoluble cancer drugs, J. Am. Chem. Soc. 130 (33) (2008) [110] D.R. Dreyer, S. Park, C.W. Bielawski, R.S. Ruoff, The chemistry of graphene oxide,
10876–10877. Chem. Soc. Rev. 39 (1) (2010) 228–240.
[81] X. Sun, Z. Liu, K. Welsher, et al., Nano-graphene oxide for cellular imaging and [111] W. Qian, J. Qiu, J. Su, X. Liu, Minocycline hydrochloride loaded on titanium by
drug delivery, Nano Res 1 (3) (2008) 203–212. graphene oxide: an excellent antibacterial platform with the synergistic effect of
[82] C.H. Lu, H.H. Yang, C.L. Zhu, X. Chen, G.N. Chen, A graphene platform for sensing contact-killing and release-killing, Biomater. Sci. 6 (2) (2018) 304–313.
biomolecules, Angew Chem. Int. Ed. Engl. 48 (26) (2009) 4785–4787. [112] W. Qian, J. Qiu, X. Liu, Minocycline hydrochloride-loaded graphene oxide films on
[83] R. Di Santo, L. Digiacomo, S. Palchetti, et al., Microfluidic manufacturing of implant abutments for peri-implantitis treatment in beagle dogs, J. Periodontol. 91
surface-functionalized graphene oxide nanoflakes for gene delivery, Nanoscale 11 (6) (2020) 792–799.
(6) (2019) 2733–2741. [113] P.D. Marsh, Dental plaque: biological significance of a biofilm and community life-
[84] C.D. Vecitis, K.R. Zodrow, S. Kang, M. Elimelech, Electronic-structure-dependent style, J. Clin. Periodontol. 32 (Suppl 6) (2005) 7–15.
bacterial cytotoxicity of single-walled carbon nanotubes, ACS Nano 4 (9) (2010) [114] I.E. Mejías Carpio, C.M. Santos, X. Wei, D.F. Rodrigues, Toxicity of a polymer-
5471–5479. graphene oxide composite against bacterial planktonic cells, biofilms, and
[85] X. Lu, X. Feng, J.R. Werber, et al., Enhanced antibacterial activity through the mammalian cells, Nanoscale 4 (15) (2012) 4746–4756, https://doi.org/10.1039/
controlled alignment of graphene oxide nanosheets, Proc. Natl. Acad. Sci. U. S. A. c2nr30774j.
114 (46) (2017) E9793–E9801. [115] I. Struzycka, The oral microbiome in dental caries, Pol. J. Microbiol. 63 (2) (2014)
[86] M.U. Farid, S. Jeong, D.H. Seo, et al., Mechanistic insight into the in vitro toxicity 127–135.
of graphene oxide against biofilm forming bacteria using laser-induced breakdown [116] D. Davies, Understanding biofilm resistance to antibacterial agents, Nat. Rev. Drug
spectroscopy, Nanoscale 10 (9) (2018) 4475–4487. Discov. 2 (2) (2003) 114–122.
[87] M. Tahriri, M. Del Monico, A. Moghanian, et al., Graphene and its derivatives: [117] R.P. Allaker, K. Memarzadeh, Nanoparticles and the control of oral infections, Int.
opportunities and challenges in dentistry, Mater. Sci. Eng. C Mater. Biol. Appl. 102 J. Antimicrob. Agents 43 (2) (2014) 95–104, https://doi.org/10.1016/
(2019) 171–185. j.ijantimicag.2013.11.002.

290
X. Qi et al. Smart Materials in Medicine 2 (2021) 280–291

[118] W. Qin, C. Wang, C. Jiang, J. Sun, C. Yu, T. Jiao, Graphene oxide enables the [142] J. Linares, M.C. Matesanz, M. Vila, et al., Endocytic mechanisms of graphene oxide
reosteogenesis of previously contaminated titanium in vitro, J. Dent. Res. 99 (8) nanosheets in osteoblasts, hepatocytes and macrophages, ACS Appl. Mater.
(2020) 922–929. Interfaces 6 (16) (2014) 13697–13706.
[119] K.P. Loh, Q. Bao, G. Eda, M. Chhowalla, Graphene oxide as a chemically tunable [143] N. Amiryaghoubi, N. Noroozi Pesyan, M. Fathi, Y. Omidi, Injectable
platform for optical applications, Nat. Chem. 2 (12) (2010) 1015–1024. thermosensitive hybrid hydrogel containing graphene oxide and chitosan as dental
[120] S.R. Ryoo, Y.K. Kim, M.H. Kim, D.H. Min, Behaviors of NIH-3T3 fibroblasts on pulp stem cells scaffold for bone tissue engineering, Int. J. Biol. Macromol. 162
graphene/carbon nanotubes: proliferation, focal adhesion, and gene transfection (2020) 1338–1357.
studies, ACS Nano 4 (11) (2010) 6587–6598. [144] S. Pathmanapan, P. Periyathambi, S.K. Anandasadagopan, Fibrin hydrogel
[121] W.C. Lee, C.H. Lim, H. Shi, et al., Origin of enhanced stem cell growth and incorporated with graphene oxide functionalized nanocomposite scaffolds for
differentiation on graphene and graphene oxide, ACS Nano 5 (9) (2011) bone repair - in vitro and in vivo study, Nanomedicine 29 (2020) 102251.
7334–7341. [145] M.A. Nazeer, E. Yilgr, I. Yilgr, Intercalated chitosan/hydroxyapatite
[122] M.S. Bahrami, P. Eshghinejad, H.R. Bakhsheshi-Rad, et al., Electrophoretic nanocomposites: promising materials for bone tissue engineering applications[J],
deposition of bioglass/graphene oxide composite on Ti-alloy implants for Carbohydr. Polym. 175 (2017) 38–46.
improved antibacterial and cytocompatible properties[J], Mater. Technol. 35 (2) [146] Pistone, Celesti, Piperopoulos, et al., Engineering of chitosan-hydroxyapatite-
(2019) 1–6. magnetite hierarchical scaffolds for guided Bone Growth[J], Materials 12 (14)
[123] K. Rho, P. Chan, K. Alam, et al., Biological effects of plasma-based graphene oxide (2019) 2321.
deposition on titanium[J], J. Nanomater. 2019 (3) (2019) 1–7. [147] J.H. Lee, Y.C. Shin, S.M. Lee, et al., Enhanced osteogenesis by reduced graphene
[124] X. Ran, Y. Du, Z. Wang, et al., Hyaluronic acid-templated Ag nanoparticles/ oxide/hydroxyapatite nanocomposites, Sci. Rep. 5 (2015) 18833. Published 2015
graphene oxide composites for synergistic therapy of bacteria infection, ACS Appl. Dec 21.
Mater. Interfaces 9 (23) (2017) 19717–19724. [148] K. Zhou, P. Yu, X. Shi, et al., Hierarchically porous hydroxyapatite hybrid scaffold
[125] R. Di Carlo, A. Di Crescenzo, S. Pilato, et al., Osteoblastic differentiation on incorporated with reduced graphene oxide for rapid bone ingrowth and repair,
graphene oxide-functionalized titanium surfaces: an in vitro study, Nanomaterials ACS Nano 13 (8) (2019) 9595–9606.
10 (4) (2020) 654. Published 2020 Apr 1. [149] T. Wu, B. Li, W. Wang, et al., Strontium-substituted hydroxyapatite grown on
[126] Q. Li, Z. Wang, Involvement of FAK/P38 signaling pathways in mediating the graphene oxide nanosheet-reinforced chitosan scaffold to promote bone
enhanced osteogenesis induced by nano-graphene oxide modification on titanium regeneration, Biomater. Sci. 8 (16) (2020) 4603–4615.
implant surface, Int. J. Nanomed. 15 (2020) 4659–4676. [150] Yin Xiao, Han, et al., Graphene-oxide-modified beta-tricalcium phosphate
[127] W. Zhang, Q. Chang, L. Xu, et al., Graphene oxide-copper nanocomposite-coated bioceramics stimulate in vitro and in vivo osteogenesis, Carb. Int. J. Spons. Am.
porous CaP scaffold for vascularized bone regeneration via activation of hif-1α, Carbon Soc. 93 (2015) 116–129.
Adv. Healthc. Mater. 8 (5) (2019), e1900067. [151] V. Bordoni, G. Reina, M. Orecchioni, et al., Stimulation of bone formation by
[128] J.W. Kim, Y.C. Shin, J.J. Lee, et al., The effect of reduced graphene oxide-coated monocyte-activator functionalized graphene oxide in vivo[J], Nanoscale (2019)
biphasic calcium phosphate bone graft material on osteogenesis, Int. J. Mol. Sci. 11.
18 (8) (2017) 1725. Published 2017 Aug 8. [152] N. Shao, J. Guo, Y. Guan, H. Zhang, X. Li, X. Chen, D. Zhou, Y. Huang,
[129] E. Nishida, H. Miyaji, A. Kato, et al., Graphene oxide scaffold accelerates cellular Development of organic/inorganic compatible and sustainably bioactive
proliferative response and alveolar bone healing of tooth extraction socket, Int. J. composites for effective bone regeneration, Biomacromolecules 19 (2018)
Nanomed. 11 (2016) 2265–2277. Published 2016 May 24. 3637–3648.
[130] T. Zhou, G. Li, S. Lin, et al., Electrospun poly(3-hydroxybutyrate-co-4- [153] M. Degli Esposti, F. Chiellini, F. Bondioli, D. Morselli, P. Fabbri, Highly porous
hydroxybutyrate)/graphene oxide scaffold: enhanced properties and promoted in PHB-based bioactive scaffolds for bone tissue engineering by in situ synthesis of
vivo bone repair in rats, ACS Appl. Mater. Interfaces 9 (49) (2017) 42589–42600. hydroxyapatite, Mater. Sci. Eng. C 100 (2019) 286–296.
[131] C. Zhou, S. Liu, J. Li, et al., Collagen functionalized with graphene oxide enhanced [154] Y. Luo, H. Shen, Y. Fang, Y. Cao, J. Huang, M. Zhang, J. Dai, X. Shi, Z. Zhang,
biomimetic mineralization and in situ bone defect repair, ACS Appl. Mater. Enhanced proliferation and osteogenic differentiation of mesenchymal stem cells
Interfaces 10 (50) (2018) 44080–44091. on graphene oxide-incorporated electrospun poly(lactic-co-glycolic acid)
[132] T.R. Nayak, H. Andersen, V.S. Makam, et al., Graphene for controlled and nanofibrous mats, ACS Appl. Mater. Interfaces 7 (2015) 6331–6339.
accelerated osteogenic differentiation of human mesenchymal stem cells, ACS [155] J. Cheng, H. Liu, B. Zhao, R. Shen, D. Liu, J. Hong, H. Wei, P. Xi, F. Chen, D. Bai,
Nano 5 (6) (2011) 4670–4678. MC3T3-E1 preosteo- blast cell-mediated mineralization of hydroxyapatite by poly-
[133] Y.C. Shin, J.H. Lee, O.S. Jin, et al., Synergistic effects of reduced graphene oxide dopamine-functionalized graphene oxide, J. Bioact. Compat Polym. 30 (2015)
and hydroxyapatite on osteogenic differentiation of MC3T3-E1 preosteoblasts[J], 289–301.
Carbon 95 (2015) 1051–1060. [156] C. Shuai, P. Feng, C. Gao, X. Shuai, T. Xiao, S. Peng, Graphene oxide reinforced
[134] M Orecchioni, D Bedognetti, L Newman, et al., Single-cell mass cytometry and poly(vinyl alcohol): nano- composite scaffolds for tissue engineering applica-
transcriptome profiling reveal the impact of graphene on human immune cells, tions, RSC Adv. 5 (2015) 25416–25423.
Nature communications 8 (1) (2017) 1109. [157] W. Shao, J. He, F. Sang, et al., Enhanced bone formation in electrospun poly(L-
[135] M. Orecchioni, C. Menard-Moyon, L.G. Delogu, A. Bianco, Graphene and the lactic-co-glycolic acid)-tussah silk fibroin ultrafine nanofiber scaffolds
immune system: challenges and potentiality, Adv. Drug Deliv. Rev. 105 (Pt B) incorporated with graphene oxide, Mater. Sci. Eng. C Mater. Biol. Appl. 62 (2016)
(2016) 163–175, https://doi.org/10.1016/j.addr.2016.05.014. 823–834.
[136] M. Orecchioni, R. Cabizza, A. Bianco, L.G. Delogu, Graphene as cancer theranostic [158] S.D. Purohit, R. Bhaskar, H. Singh, I. Yadav, M.K. Gupta, N.C. Mishra,
tool: progress and future challenges, Theranostics 5 (7) (2015) 710–723. Development of a nanocomposite scaffold of gelatin-alginate-graphene oxide for
Published 2015 Mar 28. bone tissue engineering, Int. J. Biol. Macromol. 133 (2019) 592–602.
[137] S. Liu, C. Zhou, S. Mou, et al., Biocompatible graphene oxide-collagen composite [159] S. Saravanan, S. Vimalraj, D. Anuradha, Chitosan based thermoresponsive
aerogel for enhanced stiffness and in situ bone regeneration, Mater. Sci. Eng. C hydrogel containing graphene oxide for bone tissue repair, Biomed.
Mater. Biol. Appl. 105 (2019) 110137. Pharmacother. 107 (2018) 908–917.
[138] J. Prakash, D. Prema, K.S. Venkataprasanna, K. Balagangadharan, [160] W. Qian, Y. Chu, J. He, et al., A graded graphene oxide-hydroxyapatite/silk fibroin
N. Selvamurugan, G.D. Venkatasubbu, Nanocomposite chitosan film containing biomimetic scaffold for bone tissue engineering, Mater. Sci. Eng. C 80 (nov)
graphene oxide/hydroxyapatite/gold for bone tissue engineering, Int. J. Biol. (2017) 232.
Macromol. 154 (2020) 62–71. [161] P. Yu, R.Y. Bao, X.J. Shi, et al., Self-assembled high-strength hydroxyapatite/
[139] D. Jiao, A. Zheng, Y. Liu, et al., Bidirectional differentiation of BMSCs induced by a graphene oxide/chitosan composite hydrogel for bone tissue engineering,
biomimetic procallus based on a gelatin-reduced graphene oxide reinforced Carbohydr. Polym. 155 (2017) 507–515.
hydrogel for rapid bone regeneration, Bioact. Mater. 6 (7) (2020) 2011–2028. [162] K. Alam, Y.Y. Jo, C.K. Park, et al., Synthesis of graphene oxide using atmospheric
Published 2020 Dec 31. plasma for prospective biological applications, Int. J. Nanomed. 15 (2020)
[140] G. Choe, S. Oh, J.M. Seok, S.A. Park, J.Y. Lee, Graphene oxide/alginate composites 5813–5824.
as novel bioinks for three-dimensional mesenchymal stem cell printing and bone [163] J.H. Lee, J.K. Jo, D.A. Kim, et al., Nano-graphene oxide incorporated into PMMA
regeneration applications, Nanoscale 11 (48) (2019) 23275–23285. resin to prevent microbial adhesion, Dent. Mater. 34 (4) (2018).
[141] L. Zhang, J. Xia, Q. Zhao, L. Liu, Z. Zhang, Functional graphene oxide as a
nanocarrier for controlled loading and targeted delivery of mixed anticancer
drugs, Small 6 (4) (2010) 537–544.

291

You might also like