Graphene based scaffolds on bone tissue engineering

0 downloads 0 Views 859KB Size Report
Nov 30, 2017 - for biomimetic mineralization of hydroxyapatite. (HA). A detailed structural and morphological charac- terization of the mineralized composite ...
Bioengineered

ISSN: 2165-5979 (Print) 2165-5987 (Online) Journal homepage: http://www.tandfonline.com/loi/kbie20

Graphene based scaffolds on bone tissue engineering Nasrin Shadjou, Mohammad Hasanzadeh & Balal Khalilzadeh To cite this article: Nasrin Shadjou, Mohammad Hasanzadeh & Balal Khalilzadeh (2018) Graphene based scaffolds on bone tissue engineering, Bioengineered, 9:1, 38-47, DOI: 10.1080/21655979.2017.1373539 To link to this article: https://doi.org/10.1080/21655979.2017.1373539

© 2018 The Author(s). Published with license by Taylor & Francis© Nasrin Shadjou, Mohammad Hasanzadeh, and Balal Khalilzadeh Accepted author version posted online: 02 Nov 2017. Published online: 30 Nov 2017. Submit your article to this journal

Article views: 143

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=kbie20

BIOENGINEERED 2018, VOL. 9, NO. 1, 38–47 https://doi.org/10.1080/21655979.2017.1373539

COMMENTARY

Graphene based scaffolds on bone tissue engineering Nasrin Shadjoua,b, Mohammad Hasanzadeh

,c and Balal Khalilzadehd

a Department of Nanochemistry, Nano Technology Research Center, Urmia University, Urmia, Iran; bDepartment of Nanochemistry, Faculty of Science, Urmia University, Urmia, Iran; cDrug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; dStem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran

ABSTRACT

ARTICLE HISTORY

Tissue engineering has been emerging as a valid approach to the current therapies for bone regeneration/substitution. Tissue-engineered bone constructs have the potential to alleviate the demand arising from the shortage of suitable autograft and allograft materials for augmenting bone healing. Scaffolds play a central role in tissue engineering research, they not only provide as structural support for specific cells but also provide as the templates to guide new tissue growth and construction. In this survey we describe application of graphene based nano-biomaterials for bone tissue engineering. In this article, application of different graphene based materials on construction of manufacture scaffolds for bone tissue engineering was discussed. It begins by giving the reader a brief background on tissue engineering, followed by a comprehensive description of all the relevant components of graphene based materials, going from materials to scaffolds and from cells to tissue engineering strategies that will lead to “engineered” bone. In this survey, more recent studies on the effects of graphene on surface modifications of scaffold materials was discused. The ability of graphene to improve the biological properties of scaffold materials, and its ability to promote the adhesion, proliferation, and osteoblasts have been demonstrated in several studies which we discuss in this survey article. We further highlight how the properties of graphene are being exploited for scaffolds in bone tissue engineering, comprehensively surveying recent experimental works featuring graphene and graphene derivatives. Bone tissue engineering, for the purpose of this survey, is the use of a scaffolding material to either induce formation of bone from the surrounding tissue or to act as a carrier or template for implanted bone cells or other agents. Materials used as bone tissue-engineered scaffolds may be injectable or rigid, the latter requiring an operative implantation procedure.

Received 28 July 2017 Revised 13 August 2017 Accepted 21 August 2017

Introduction Scaffolds in tissue engineering

Scaffolds play a central role in tissue engineering research, they not only provide as structural support for specific cells but also provide as the templates to guide new tissue growth and construction [1]. Scaffolds provide support to the regenerating tissue and could also be used to deliver bioactive molecules to accelerate the healing process. In addition, the scaffolds for bone tissue engineering need to be biocompatible, osteoinductive, osteoconductive, and osteointegrative. In order to achieve tissue reconstruction and functional recovery of damaged tissues, stem cell-based tissue engineering requires three-dimensional (3D) functional scaffolds to improve the regenerative CONTACT Mohammad Hasanzadeh, Scientist University of Medical Sciences, Tabriz 51664, Iran.

KEYWORDS

bone scaffold; biochemical science; engineered bone; graphene; nano-biomaterials; tissue engineering

potential of stem cells. Such functional 3D scaffolds can be constructed via the systematic control and optimization of the biochemical, biophysical, and mechanical characteristics of the scaffolds. Biophysical and mechanical cues including elasticity, stiffness, and topography can regulate stem cell phenotype and functions such as self-renewal, proliferation, and differentiation of stem cells [2]. Graphene based materials for use in scaffolds

The success of bone tissue engineering highly depends on the functionality of the scaffold. Identifying new scaffold materials with properties like good biocompatibility, controlled nontoxic biodegradation, ability to support cell differentiation, growth, and proliferation, and suitable mechanical strength, is crucial for

[email protected]; [email protected]

Drug Applied Research Center, Tabriz

© 2018 Nasrin Shadjou, Mohammad Hasanzadeh, and Balal Khalilzadeh. Published with license by Taylor & Francis This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/ 4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.

BIOENGINEERED

39

Scheme 1. Biomedical application of graphene.

the efficiency of tissue regeneration process [3]. One potential functional scaffold material for bone tissue engineering applications is graphene. Among different carbon allotropes, graphene is a novel and potentially useful nanomaterial, the application of which in biomedical sciences and biotechnology is beginning to be realized. Graphene materials have been explored for various biomedical applications, such as biosensors, tissue engineering, and drug delivery (see Scheme 1) [4,5]. Graphene has emerged as a subject of enormous scientific interest because of its exceptional electron transport, physicochemical and mechanical properties, and high surface area (Table 1). Of the cell types tested, it was found that cells adhered to and proliferated better when cultured on graphene films than when the cells were cultured on a SiO2 substrate [6]. In addition, these atomically

thin carbon sheets can significantly improve the physical properties of host polymers at extremely small concentrations when incorporated appropriately. Bone tissue engineering promises to restore bone defects that are caused by severe trauma, congenital malformations, and so forth. Many researchers are studying the ways to confer a pro-osteodifferentiation or osteoinductive capability on scaffold materials, where osteogenesis of seed cells is promoted. Graphene provides a new kind of coating material that may confer the proosteo differentiation capability on Table 1. Some of specific properties of graphene. Specific surface area Band gap Thermal conductivity Transparency Charge carrier mobility

»2630 m2/g 0 »5000 Watts per meter-kelvin (W/(m. K)) »97.4% »200 000 cm2/V¢s

40

N. SHADJOU ET AL.

scaffold materials by surface modification. Here, in this article, we survey more recent studies on the effects of graphene on surface modifications of scaffold materials. The ability of graphene to improve the biological properties of scaffold materials, and its ability to promote the adhesion, proliferation, and osteoblasts have been demonstrated in several studies which we discuss in this survey. We further highlight how the properties of graphene are being exploited for scaffolds in bone tissue engineering, comprehensively surveying recent experimental works featuring graphene and graphene derivatives. In this survey, more recent studies on the effects of graphene on surface modifications of scaffold materials was discussed. The ability of graphene to improve the biological properties of scaffold materials, and its ability to promote the adhesion, proliferation, and osteoblasts have been demonstrated in several studies which we discuss in this survey article. We further highlight how the properties of graphene are being exploited for scaffolds in bone tissue engineering, comprehensively surveying recent experimental works featuring graphene and graphene derivatives. Bone tissue engineering, for the purpose of this survey, is the use of a scaffolding material to either induce formation of bone from the surrounding tissue or to act as a carrier or template for implanted bone cells or other agents. Materials used as bone tissue-engineered scaffolds may be injectable or rigid, the latter requiring an operative implantation procedure. The present survey intends to provide the reader an overview of the current state (during 2013 to 2017) of the graphene-based scafolds in bone tissue engineering, its limitations and hopes as well as the future research trends for this exciting field of science. The aim of this effort is to provide the reader with a clear and concise view of new advances in areas ranging from bone tissues engineering to bone cements. More importantly, different aspects of the graphene-based biomaterials such as functionalization techniques, and bio-activation of glasses and etc. are discussed in detail. Moreover, we have attempted to highlight areas of the latest and significant development of enhanced bone tissues engineering process that inspire broader interests across various disciplines. Such novel and recent advances are important for the development of bone research that open up new paths for future research. As mentioned above, it has been reported that due to its aromatic scaffold nature, graphene and graphene

oxide (GO) are potential for promoting the cell behavior including attachment, growth, proliferation, and differentiation [7,8]. Recently, GO nanoflakes (0.5 and 1 wt%) were incorporated into a gelatin- hydroxyapatite (GHA) matrix through a freeze drying technique and its effect to enhance mechanical strength and osteogenic differentiation was studied [9]. The GHA matrix with GO demonstrated less brittleness in comparison to GHA scaffolds. There was no significant difference mechanical strength between GOGHA0.5 and GOGHA1.0 scaffolds. Results of et al. [9], show that when the scaffolds were immersed in phosphate buffered saline (to mimic physiologic condition) for 60 days, around 50–60% of GO was released in sustained and linear manner and the concentration was within the toxicity limit as reported earlier. Further, GOGHA0.5 scaffolds were continued for cell culture experiments, wherein the scaffold induced osteogenic differentiation of human adipose derived mesenchymal stem cells without providing supplements like dexamethasone, L-ascorbic acid and b-glycerophosphate in the medium. The level of osteogenic differentiation of stem cells was comparable to those cultured on GHA scaffolds with osteogenic supplements. Thus biocompatible, biodegradable and porous GO reinforced gelatin-gelatin-hydroxyapatite 3D scaffolds (graphene oxide (GO) incorporated into a gelatinhydroxyapatite (GHA) matrix) may serve as a suitable candidate in promoting bone regeneration in orthopaedics. It seems that this is the first report which showing the osseopromotive property of GO incorporated 3D gelatin-HA scaffolds without providing any osteogenic factors in the medium. Also, results of researchers indicated that GO coating improved several biomedical properties of collagen scaffold including surface structure, compressive strength and cell ingrowth [9]. A low concentration GO film did not inhibit cell proliferation or differentiation in vitro, and enhanced biocompatibility and biodegradability. Therefore, scaffold modified by a suitable concentration of GO holds promise as a biomaterial for tissue engineering. Also, Xie and coworkers reported a simple method to synthesize free-standing G/HA hydrogels based on colloidal chemistry, with unprecedented homogeneity in their 3D structure [10]. In this report, the HA NPs are encapsulated in the graphene hydrogels thanks to the formation of a thick, graphite-like shell during the

BIOENGINEERED

hydrothermal treatment. The resulting graphene-HA gels are highly porous, strong, electrically conductive and biocompatible, making them promising scaffolds for bone tissue engineering. The biological bone minerals form within a complex microenvironment that consists of various ions and template molecules [10]. Silk fibroin provides an attractive template for biomimetic hydroxyapatite synthesis. On the other hand, it is known that some cationic ions (e.g., Zn2+, Mg2+, and Sr2+) can easily enter the apatite crystal structure, and they are found to play important roles in biological functions. These ions can either be included in the growing stable apatite lattice during the ageing process or remain in the hydrated layer. Due to the very high specific area of the crystals, the mineral could participate in several basic equilibria, involving for example specific proteins or mineral ions adsorption and release. Thus, some cationic ions could remain on the surface of the apatite crystals. Then the global composition of such crystals can differ from the proposed chemical formulae which do not consider surface ions although they can represent an important fraction in nanocrystals. Shepherd et al. [11], reveal the influence of zinc substitution on the mineralization process of silk fibroin. Herein, silk fibroin is used as the primary template because (1) the fibroin molecule is able to regulate the growth of apatite and (2) the mineralized silk fibroin is a biocompatible biomaterial. Using cationic substitution, it is possible for us to tune the properties of the obtained minerals, such as morphology, structure and crystallinity. Furthermore, the incorporation of zinc ions into hydroxyapatite could add more biological functions to the nanoparticles, such as immunological modulation [10], antibacterial property [12], and osteoblast response [11]. The calcium phosphate precipitates could maintain the apatite phase when Zn/(Zn+Ca) reached 15–20 mol % [13]. Recently, a novel approach presented for the synthesis of zinc substituted hydroxyapatite and demonstrated the mechanisms of calcium phosphate precipitation regulated by silk fibroin and sodium alginate [14]. In this work, graphene-like zinc substituted hydroxyapatite crystals were prepared using silk fibroin and sodium alginate as template molecules, and the resulting products were investigated. The alginate ions interacted with silk fibroin, and the additional zinc ions also influenced the crystal formation of hydroxyapatite. The graphene-like sheets with hydroxyapatite phase

41

were approximately 3 nm in thickness, and the size was more than 100 nm. The biomimetic method, which develops materials by mimicking the structure and composition of natural tissue, has been a major goal in the bone tissue engineering. The mechanism of the bone formation process is thought to be due to the mechanical signals provided by the self-assembled collagen [15] and the presence of the charged proteins in the extracellular matrix, which facilitate the nucleation of HA [9]. Therefore, it is imperative to design multifunctional biomaterials which can induce and assemble bonelike apatite that is close to natural bone. In the previous report by Chen et al. [16], these researchers have demonstrated the functionalization of GO by several bioactive molecules, such as dopamine and carrageenan, which have catechol groups and sulfate groups, respectively. These charged groups can mimic the proteins present in the extracellular matrix to induce mineralization [17]. It has been found that the biointerfaces with different charged groups can result in different structures of HA by mineralization. In this case, a facile modification of graphene oxide (GO) by gelatin to mimic charged proteins present in the extracellular matrix during bone formation was reported by Liu and coworkers [18]. In this report, the bioinspired surface of GO-gelatin (GO-Gel) composite was used for biomimetic mineralization of hydroxyapatite (HA). A detailed structural and morphological characterization of the mineralized composite was performed. Additionally, MC3T3-E1 cells were cultured on the GO-Gel surfaces to observe various cellular activities and HA mineralization. Higher cellular activities such as cell adhesion, cell proliferation, and alkaline phosphatase activity (ALP) were observed on the GO-Gel surface compared with the GO or glass surface. The increase of ALP confirms that the proposed GO-Gel promotes the osteogenic differentiation of MC3T3-E1 cells. Moreover, the evidence of mineralization evaluated by scanning electron microscopy (SEM) and alizarin red staining (ARS) corroborate the idea that a native osteoid matrix is ultimately deposited. All these data suggest that the GO-Gel hybrids will have great potential as osteogenesis promoting scaffolds for successful application in bone surgery. Also, nanofibrous biocomposite scaffolds of poly (vinyl alcohol) (PVA) and graphene oxide (GO) were prepared by using electrospinning method [19]. The mechanical properties were investigated by tensile

42

N. SHADJOU ET AL.

testing. In this work, mouse Osteoblastic Cells (MC3T3-E1) attachment and proliferation on the nanofibrous scaffolds were investigated by MTT [3(4,5-dimeth-ylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assay. SEM images show the three-dimensional porous fibrous morphology, and the average diameter of the composite fibers decreases with increasing the content of GO. The mechanical properties of the scaffolds are altered by changing the content of GO as well. The tensile strength and elasticity modulus increase when the content of GO is lower than 1 wt %, but decrease when GO is up to 3 and 5 wt %. MC3T3-E1 cells attach and grow on the surfaces of the scaffolds, and the adding of GO do not affect the cells’ viability. Also, MC3T3-E1 cells are likely to spread on the PVA/GO composite scaffolds. Above all, these unique features of the PVA/GO nanofibrous scaffolds prepared by electrospinning would open up a wide variety of future applications in bone tissue engineering. Despite great recent progress with carbon nanotubes and other nanoscale fillers, the development of strong, durable, and cost-efficient multifunctional nanocomposite materials has yet to be achieved. The challenges are to achieve molecule-level dispersion and maximum interfacial interaction between the nanofiller and the matrix at low loading. Here, the preparation of poly(vinyl alcohol) (PVA) nanocomposites with graphene oxide (GO) using a simple water solution processing method open up a wide variety of future applications in bone tissue engineering. Recently, Cheng et al. [20] reported a biomimetic mineralization of hydroxyapatite induced by polydopamine-functionalized reduced graphene oxide (RGO-PDA). Graphene oxide was first simultaneously reduced and surface functionalized by one-step oxidative polymerization of dopamine. The resultant RGOPDA was further used as a bioinspired surface to mimic the mineralization of hydroxyapatite during bone formation. MC3T3-E1 cells were cultured on the RGO-PDA substrates to observe various cellular activities and hydroxyapatite mineralization. The MC3T3-E1 cells on RGOPDA substrates show higher cellular activities such as proliferation, adhesion, and osteogenic differentiation over the bare glass and graphene oxide substrates. Results of this work suggested the potentials of using RGO-PDA as osteogenesis-promoting scaffolds for successful applications in bone tissue regeneration. The study of structure-function relationships in bone tissue engineering has been promoted the

development of bioactive substitutes and engineered biomaterials. Among the possible routes to homogeneously coat highly porous 45S5 Bioglass®- derived glass ceramic scaffolds with graphene sheets without influencing the shape and dimension of the pores, the hybrid sol-gel technology has several advantages, mainly related to the mild conditions associated with this wet chemical process. In fact the classical sol-gel process involving metal alkoxides, typically tetraethoxysilane (TEOS) hydrolysis and polycondensation reactions was shown to be versatile enough for an efficient incorporation of polymer chains bearing reactive groups that are involved in the hydrolysis-polycondensation reactions and act as flexible domains inside the resulting metal oxide three dimensional network [21,22]. These materials are also known as phase- interconnected organicinorganic nanocomposites because of the high level of interconnection between the two phases with domain size approaching the nanometer scale, which are the basis of their optical transparency and high toughness. These sol-gel organic–inorganic hybrids have been successfully used for the preparation of tough, transparent, flexible coatings for both plastic and glassceramic substrates, and were demonstrate to be highly adherent and resistant to wear and abrasion. Moreover, the solution chemical process for their preparation easily allows the inclusion of functional objects or molecules inside their formulation. In particular, solgel hybrids with poly(ethylene glycol) (PEG) domains covalently bonded to a silica network are very flexible and resistant [23] and are particularly suited for the coating of biomedical devices and implants because of their medical safety and biocompatibility [23,24]. According to these advantages, highly porous 45S5 Bioglass®-based scaffolds fabricated by a foam replication technique were coated with electrically conductive organic-inorganic hybrid layers containing graphene by a solution method. a,v-Triethoxysilane terminated poly(ethylene glycol) and tetraethoxysilane were used as the precursors of the organic- inorganic hybrid coatings, that contained 1.5 wt.% of homogeneously dispersed graphene nanoplatelets [25]. Obtained results by this report show that the presence of graphene did not impair the bioactivity of the scaffolds in simulated body fluid. Initial tests carried out using MG-63 cells demonstrated that both uncoated scaffolds and scaffolds coated with organic/inorganic hybrids containing graphene offered the cultured cells

BIOENGINEERED

an adequate surface for cell attachment, spreading and expression of extracellular matrix. Furthermore, in the case of graphene coated scaffolds the cells seem to be oriented. The results of this report showed that scaffolds coated with graphene are biocompatible and they can support cellular activity. Also, the electrical conductivity introduced by the coating might have the potential to increase tissue growth when cell culture is carried out under an applied electric field. Initial tests carried out using MG-63 cells demonstrated that both uncoated scaffolds and scaffolds coated with organic/ inorganic hybrids containing graphene offered the cultured cells an adequate surface for cell attachment, spreading and expression of extracellular matrix. 58Sbioactive glass (58S) (58% SiO2, 33% CaO and 9% P2O5, based on mol%) has received special attention as scaffold material owing to its good biodegradability, excellent bioactivity and bonebonding ability. [68, 69] It reacts with physiological fluids to form direct bonds to bone tissue in the early time after implantation without toxicity, inflammation and foreign-body response [26]. The fast surface reactions in vivo lead to rapid ionic dissolution and formation of hydroxyl-carbonated apatite (HCA) layer [27]. The release of soluble Si, Ca, and P ions can activate gene expression and stimulate osteoblast proliferation for rapid bone formation [28]. Moreover, 58S in nano scale (nano-58S) exhibits better bioactivity in terms of cell growth, osteogenic differentiation and HCA formation [29]. The major hurdles of 58S are intrinsic brittleness, low fracture toughness and crack resistance to sustain the loads transmitted from surrounding bone tissue [30], which is considered to be one of the main requirements of scaffolds during the period of new bone formation. Thus, there has been a strong impetus to improve the mechanical properties of 58S scaffold in the past years. Current attempts focused on improving the mechanical properties of 58S by incorporating second phase reinforcements including polymers and metallic oxides. O’Shea and coworkers developed a poly(lactic-co-glycolic acid) (PLGA)-coated 58S scaffold. The addition of PLGA coating improved the compressive strength of 58S scaffold to 0.25 MPa, which was twice that of uncoated 58S scaffold (0.12 MPa) but still lies toward the lower limit of cancellous bone [31]. Increasing efforts were also devoted to improve the mechanical properties by surface modification using organic molecular, which promoted the dispersion of 58S particles in the composites [32].

43

Nevertheless, this method was also accompanied by a weakened capability for calcium precipitation [33]. So far, few of these scaffolds fulfill both the mechanical and biological requirements for load bearing applications. Recently, nano-58S was combined with graphene in order to enhance its poor mechanical properties for bone tissue engineering applications [34]. In this work, 3D porous composite scaffolds of graphene/nano-58S were fabricated using SLS technique. The optimum compressive strength and fracture toughness reached 48.65 6 3.19 MPa and 1.94 6 0.10 MPa/m0.5 with graphene content of 0.5 wt%, indicating significant improvements by 105% and 38% respectively. The mechanisms of pull-out, crack bridging, crack deflection and crack tip shielding were found to be responsible for the mechanical enhancement. Simulated body fluid and cell culture tests indicated favorable bioactivity and biocompatibility of the composite scaffold. The results suggest a great potential of graphene/nano-58S composite scaffold for bone tissue engineering applications. More recently, the contribution of BG with BG-graphene nanoplatelets (GNP) was investigated by Porwal and coworkers [35]. In this work, fully dense BG nanocomposites with GNP loading of 1, 3 and 5 vol% were consolidated using Spark plasma sintering(SPS). SPS avoided any structural damage of GNP as confirmed using Raman spectroscopy. GNP increased the viscosity of BG-GNP composites resulting in an increase in the sintering temperature by about 50 C compared to pure BG. Electrical conductivity of BGGNP composites increased with increasing concentration of GNP. The highest conductivity of 13 S/m was observed for BG-GNP (5 vol%) composite which is about 9 orders of magnitude higher compared to pure BG. For both BG and BG-GNP composites, in vitro bioactivity testing was done using simulated body fluid for1 and 3 days. Results of this report indicated that GNP increased the electrical conductivity of BG-GNP composites without affecting the bioactivity thus opening the possibility to fabricate bioactive and electrically conductive scaffolds for bone tissue engineering. Concluding remarks and future directions

The field of tissue engineering and in particular, bone tissues engineering, is rapidly growing. Bone tissues engineering-based products are beginning to be used

44

N. SHADJOU ET AL.

in clinical practice. Based on the current success, even more bone tissues engineering technologies are expected to become available to patients in the next few years. Current efforts are focused on developing effective strategies for bone tissues engineering, but we predict that the future discussion will turn toward the identification of the most cost-effective bone tissues engineering strategies. Although the race to make bone tissues engineering a clinical reality is well warranted, significant challenges and limitations in this field still exist. An unresolved matter for tissue engineering is the translation of 3D cultures from the academia to the pharmaceutical industry. To achieve this goal, 3D cultures should meet a set of requirements, apart from biological relevance: standardization, high throughput applicability and economic feasibility. Hence, major efforts are being made in this direction. Nowadays many questions about cancer biology remain to be answered, but TE modeling enriches the toolbox to understand disease progression and, thus, improve therapeutic approaches. To achieve this goal, it would be interesting to use more extendedly 3D models within the scientific community. The above-mentioned 3D cultures are based on combining cells, scaffolds and biomolecules. However, we can reach a higher degree of complexity by integrating microchip and microfluidic approaches to TE. First of all, microfabrication include techniques such as photolithography, replica molding and microcontact printing, which enable the creation of structures with well-defined shapes on the micrometer scale, so that we can control cell position, morphology and function. Secondly, microfluidics consists of manipulating small amounts (10¡9 to 10¡18 L) of fluids in hollow chambers and, therefore, allows us to generate and precisely tune spatiotemporal gradients of soluble effector molecules (nutrients and oxygen). The combination of both techniques can lead to organ-on-chips microdevices, which notably improve the level of cell differentiation and organization achieved with 3D models and constitute potential substitutes for animals in drug screening processes. Current efforts are focused on developing effective strategies for tissue engineering, but we predict that the future discussion will turn toward the identification of the most cost-effective tissue engineering strategies. Although the race to make tissue engineering a clinical reality is well warranted, significant challenges and limitations in this field still exist.

Current limitations and challenges facing the field of bone tissue engineering. Generally, the field of tissue engineering has undergone tremdous advances in the last several decades, especially with simple tissues (i.e., skin). Engineering bone tissue, however, is not only based on principles of cellular and molecular developmental biology and morphogenesis, it is very much guided by bioengineering and biome-chanics. Bone tissue structure and mechanical strength varies by distinct and dynamic loading conditions, as well as location in the body. Perhaps one of the largest challenges facing bone tissue engineering is developing mechanically strong porous scaffolds that retain proper vascularization and host integration properties. Currently, the vast majority of reported mechanically strong tissue engineering scaffolds experience bone tissue regeneration that is limited to the periphery of the scaffold upon implantation, due to lack of sufficient and timely vascularization of the construct. In addition, the incorporation of immunomodulatory strategies is becoming increasingly popular for modulating the host’s foreign-body response (i.e., fibrous tissue encapsulation), an event that is often observed to be an inhibitory factor for optimal tissue regeneration and integration. Scientists are attempting to tackle both enhanced vascularization and inhibition of fibrous tissue formation by incorporating growth factors via the scaffold or genetically modified cells that release increased levels of angiogenic VEGF, or even by coating the scaffold with anti-inflammatory molecules, such as dexamethasone. Animal models pose another critical challenge to testing various tissue engineering approaches pre-clinically. In pre-clinical studies, load-bearing large animal models should generally be used to assess graft functionality because research on small animals (i.e., mice) does not yield relevant results due to major differences in graft size and healing properties. Although many tissue engineering strategies have been investigated, so far only a few have been approved for clinical use. These are mostly singlecomponent strategies involving cells, factors, or defect-filling materials. For tissue engineering to become a widespread clinical reality, it must incorporate the recent technologies that utilize all the necessary components (i.e., scaffolds, cells, and growth factors) for successful bone repair and regeneration. One concern is that the technologies that include more components may have difficulty obtaining

BIOENGINEERED

regulatory approval. Furthermore, tissue engineering may even pose as a health care burden in its current form, as it comes with high manufacturing costs and is patient specific. To increase efficiency, patient-independent methods need to be considered. In addition, more effective cell isolation, seeding, and culturing methods need to be developed to streamline the engineering process and to decrease the safety risks associated with the handling the constructs during the preimplantation period. Bioreactors that can combine all three steps have been proposed for this purpose and may drive the way for safer and more effective bone tissue engineering. Ultimately, however, the best bioreactor for tissue engineering scaffolds is bone itself with the idea that a scaffold could indeed mature into a normal bone tissue if an adequate environment is provided in vivo. Perhaps the quickest route to clinical success will avoid utilizing the in vitro bioreactor approach. Therefore, further efforts must be made to establish efficient intraoperative cell seeding methods to minimize in vitro culture of the tissue engineering constructs, and allow for maximized bone tissue regeneration in vivo. Generally, the field of tissue engineering has undergone enormous advances in the last several decades, especially with simple tissues. Engineering bone tissue, however, is not only based on principles of cellular and molecular developmental biology and morphogenesis, it is very much guided by bioengineering and biomechanics. Bone tissue structure and mechanical strength varies by distinct and dynamic loading conditions, as well as location in the body. Perhaps one of the largest challenges facing bone tissue engineering is developing mechanically strong porous scaffolds that retain proper vascularization and host integration properties. Currently, the vast majority of reported mechanically strong bone tissues engineering scaffolds experience bone tissue regeneration that is limited to the periphery of the scaffold upon implantation, due to lack of sufficient and timely vascularization of the construct. In addition, the incorporation of immunomodulatory strategies is becoming increasingly popular for modulating the host’s foreign-body response, an event that is often observed to be an inhibitory factor for optimal tissue regeneration and integration. Scientists are attempting to tackle both enhanced vascularization and inhibition of fibrous tissue formation by incorporating growth factors via the scaffold or genetically modified cells that release increased levels

45

of angiogenic vascular endothelial growth factor, or even by coating the scaffold with anti-inflammatory molecules, such as dexamethasone. Animal models pose another critical challenge to testing various bone tissues engineering approaches pre-clinically. In preclinical studies, load-bearing large animal models should generally be used to assess graft functionality because research on small animals (i.e., mice) does not yield relevant results due to major differences in graft size and healing properties. As described above examples graphene-based nanobiomaterials are excellent candidates to be used as starting materials for the manufacture of scaffolds for bone tissue engineering. Within this application, special attention should be given to the possibility of covalently grafting osteoinductive agents (peptides, proteins and growth factors) to the surface of the scaffolds, which would act as attractive signals for bone cells and promote the bone regeneration process. Overall, mechanical and electrical properties of graphene materials can be useful in reinforcing tissue engineering scaffolds. Recent literature indicates that graphene-based composites interfaced with micro/ nanofabrication technologies may lead to development of scaffolds with properties fine-tuned for target organ/tissues. However, along with detailed in vitro characterization of scaffolds, more emphasis should be placed on their evaluation in vivo with respect to inflammatory responses, biocompatibility and regenerative potential. As it was described bone tissue is complex, as well as its various structural arrangements. At this moment, this is probably one of the most challenging aspects to develop a bioartificial tissue engineered bone. Although a great advance in the knowledge of bone biology has been achieved until now, further steps need to be taken in order to better understand what is needed to develop a commercial tissue engineered bone. In this article, we summarized application of different type of graphen-based nonobiomaterials on bone tissues engineering. More importantly, different aspects of the graphene based biomaterials such as functionalization techniques, and bioactivation of glasses and etc. were discussed in detail. Also, several outstanding properties of the bioactive glasses and their research opportunities as well as the development potential and prospects were discussed. In addition, this survey challenged the possibility of covalently grafting different osteoinductive agents to the

46

N. SHADJOU ET AL.

graphene based scaffold surface that act as attracting signals for bone cells to promote the bone regeneration process.

[11]

Acknowledgements We gratefully acknowledge the support of this work by Tabriz University of Medical Sciences.

[13]

ORCID Mohammad Hasanzadeh 1239

[12]

http://orcid.org/0000-0003-4918[14]

References [1] Shadjou N, Hasanzadeh M. Graphene and its nanostructure derivatives for use in bone tissue engineering: Recent advances. J Biomed Mater Res A. 2015;104:1250–1275. doi:10.1002/jbm.a.35645. [2] Sill TJ, von Recum HA. Electrospinning: applications in drug delivery and tissue engineering. Biomaterials. 2008;29:1989–2006. doi:10.1016/j.biomaterials.2008.01.011. PMID:18281090. [3] Kim H, Jiao A, Hwang N, et al.. Nanotopography-guided tissue engineering and regenerative medicine. Adv Drug Delivery Rev. 2013;65:536. doi:10.1016/j.addr.2012.07.014. [4] Hasanzadeh M, Pournaghi-Azar MH, Shadjou N, et al.. A new mechanistic approach to elucidate furosemide electrooxidation on magnetic nanoparticles loaded on graphene oxide modified glassy carbon electrode. RSC Adv. 2014;4:6580–90. doi:10.1039/c3ra46973e. [5] Saghatforoush L, Hasanzadeh M, Shadjou N. ß-Cyclodextrin/graphene oxide grafted sulfonic acid: Application for electro-oxidation and determination of cadaverine in fish samples. J Electroanalytical Chem. 2014;714:79–84. doi:10.1016/j.jelechem.2013.12.021. [6] Kalbacova M, Broz A, Kong J, et al. Graphene substrates promote adherence of human osteoblasts and mesenchymal stromal cells. Carbon. 2010;48:4323–9. doi:10.1016/j. carbon.2010.07.045. [7] Ryoo S-R, Kim Y-K, Kim M-H, et al. Behaviors of NIH-3T3 fibroblasts on graphene/carbon nanotubes: proliferation, focal adhesion, and gene transfection studies. ACS Nano. 2010;4:6587–98. doi:10.1021/nn1018279. PMID:20979372. [8] Wang K, Ruan J, Song H. Biocompatibility of graphene oxide. Nanoscale Res Lett. 2011;6(1):8. PMID:27502632. [9] Nair M, Nancy D, Krishnan AG, et al. Graphene oxide nanoflakes incorporated gelatin-hydroxyapatite scaffolds enhance osteogenic differentiation of human mesenchymal stem cells. Nanotechnology. 2015;26:161001. doi:10.1088/ 0957-4484/26/16/161001. PMID:25824014. [10] Xie X, Hu K, Fang D, et al. Graphene and hydroxyapatite self-assemble into homogeneous, free standing nanocomposite hydrogels for bone tissue

[15]

[16]

[17]

[18]

[19]

[20]

[21]

[22]

[23]

[24]

engineering. Nanoscale. 2015;7:7992–8002. doi:10.1039/C5NR01107H. PMID:25864935. Shepherd D, Best SM. Production of zinc substituted hydroxyapatite using various precipitation routes. Biomed Materials. 2013;8:025003. doi:10.1088/17486041/8/2/025003. Velard F, Maquin DL, Guillaume C, et al. Zinc doped hydroxyapatite as immunomodulatory biomaterial. Int J Artificial Organs. 2009;32:457. Webster TJ, Massa-Schlueter EA, Smith JL, et al. Osteoblast response to hydroxyapatite doped with divalent and trivalent cations. Biomaterials. 2004;25:2111–21. doi:10.1016/j.biomaterials.2003.09.001. PMID:14741626. Ren F, Xin R, Ge X, et al. Characterization and structural analysis of zinc-substituted hydroxyapatites. Acta Biomaterialia. 2009;5:3141–9. doi:10.1016/j.actbio.2009.04.014. PMID:19446055. Ma J, Qin J. Graphene-like zinc substituted hydroxyapatite. Crystal Growth Design. 2015;15:1273–9. doi:10.1021/cg501659x. Bose S, Roy M, Bandyopadhyay A. Recent advances in bone tissue engineering scaffolds. Trends Biotechnol. 2012;30:546–54. doi:10.1016/j.tibtech.2012.07.005. PMID:22939815. Liu H, Xi P, Xie G, et al. Simultaneous reduction and surface functionalization of graphene oxide for hydroxyapatite mineralization. J Phys Chem C. 2012;116:3334–41. doi:10.1021/jp2102226. Liu H, Cheng J, Chen F, et al. Gelatin functionalized graphene oxide for mineralization of hydroxyapatite: biomimetic and in vitro evaluation. Nanoscale. 2014;6:5315– 22. doi:10.1039/c4nr00355a. PMID:24699835. Qi Y, Tai Z, Sun D, et al. Fabrication and characterization of poly (vinyl alcohol)/graphene oxide nanofibrous biocomposite scaffolds. J Applied Polymer Sci. 2013;127:1885–94. doi:10.1002/app.37924. Cheng J, Liu H, Zhao B, et al. MC3T3-E1 preosteoblast cell-mediated mineralization of hydroxyapatite by polydopamine-functionalized graphene oxide. J Bioactive Compatible Polymers. 2015;30:289–301. doi:10.1177/ 0883911515569918. Hasanzadeh M, Shadjou N, de la Guardia M. Electrochemical biosensing using hydrogel nanoparticles. Trends Anal. Chem. 2015;62;11–19. doi:10.1016/j.trac.2014.06.011. Shadjou N, Hasanzadeh M, Silica based mesoporous nanobiomaterials as promoter of bone regeneration process. J Biomed Mater Res A. 2015;103:3703–3716. doi:10.1002/jbm.a.35504. PMID:26011776. Iotti M, Fabbri P, Messori M, et al. Organic-inorganic hybrid coatings for the modification of barrier properties of poly (lactic acid) films for food packaging applications. J Polymers Environment. 2009;17:10–9. doi:10.1007/ s10924-009-0120-4. Kortesuo P, Ahola M, Karlsson S, et al. Silica xerogel as an implantable carrier for controlled drug delivery-evaluation of drug distribution and tissue effects after

BIOENGINEERED

[25]

[26]

[27]

[28]

[29]

implantation. Biomaterials. 2000;21:193–8. doi:10.1016/ S0142-9612(99)00148-9. PMID:10632401. Fabbri P, Valentini L, Hum J, et al. 45S5 Bioglass®derived scaffolds coated with organic-inorganic hybrids containing graphene. Materials Sci Engineering: C. 2013;33:3592–600. doi:10.1016/j.msec.2013.04.028. Faure J, Drevet R, Potiron S, et al. Electrophoretic deposition of bioactive glass coatings on Ti12Mo5Ta alloy. Key Engineering Materials: Trans Tech Publ. 2012;507:135–40. doi:10.4028/www.scientific. net/KEM.507.135. Gerhardt L-C, Widdows KL, Erol MM, et al The proangiogenic properties of multi-functional bioactive glass composite scaffolds. Biomaterials. 2011;32:4096–108. doi:10.1016/j.biomaterials.2011.02.032. PMID:21411138. Hoppe A, G€ uldal NS, Boccaccini AR. A review of the biological response to ionic dissolution products from bioactive glasses and glass-ceramics. Biomaterials. 2011;32:2757– 74. doi:10.1016/j.biomaterials.2011.01.004. PMID:21292319. Misra SK, Mohn D, Brunner TJ, et al. Comparison of nanoscale and microscale bioactive glass on the properties of P (3HB)/Bioglass® composites. Biomaterials. 2008;29:1750–61. doi:10.1016/j.biomaterials.2007.12.040. PMID:18255139.

47

[30] Ma J, Chen C, Wang D, et al. Influence of the sintering temperature on the structural feature and bioactivity of sol–gel derived SiO 2–CaO–P 2 O 5 bioglass. Ceramics Int. 2010;36:1911–6. doi:10.1016/j.ceramint.2010.03.017. [31] O’Shea TM, Miao X. Preparation and characterisation of plga-coated porous bioactive glass-ceramic scaffolds for subchondral bone tissue engineering. Ceramic Materials and Components for Energy and Environmental Applications: John Wiley & Sons Inc. 2010;5:517–23. [32] Boccaccini AR, Erol M, Stark WJ, et al. Polymer/bioactive glass nanocomposites for biomedical applications: a review. Composites Sci Technol. 2010;70:1764–76. doi:10.1016/j.compscitech.2010.06.002. [33] Zhou Y, Gao Y, Chang J. Effects of hydrolysis on dodecyl alcohol-modified bioactive glasses and PDLLA/modified bioactive glass composite films. J Materials Sci. 2010;45:6411–6. doi:10.1007/s10853-010-4724-9. [34] Gao C, Liu T, Shuai C, et al. Enhancement mechanisms of graphene in nano-58S bioactive glass scaffold: mechanical and biological performance. Sci Rep. 2014;4:4712–22. [35] Porwal H, Grasso S, Cordero-Arias L, et al. Processing and bioactivity of 45S5 Bioglass®-graphene nanoplatelets composites. J Mater Sci Med. 2014;25:1403–13. doi:10.1007/s10856-014-5172-x.