Elsevier

Biomaterials

Volume 277, October 2021, 121082
Biomaterials

Graphene oxide-functionalized nanocomposites promote osteogenesis of human mesenchymal stem cells via enhancement of BMP-SMAD1/5 signaling pathway

https://doi.org/10.1016/j.biomaterials.2021.121082Get rights and content

Abstract

Biomaterials that can harness the intrinsic osteogenic potential of stem cells offer a promising strategy to accelerate bone regeneration and repair. Previously, we had used methacrylated gelatin (GelMA)-based scaffolds to achieve bone formation from human mesenchymal stem cells (hMSCs). In this study, we aimed to further enhance hMSC osteogenesis by incorporating graphene oxide (GO)-based nanosheets into GelMA. In vitro results showed high viability and metabolic activities in hMSCs encapsulated in the newly developed nanocomposites. Incorporation of GO markedly increased mineralization within hMSC-laden constructs, which was further increased by replacing GO with silica-coated graphene oxide (SiGO). Mechanistic analysis revealed that the nanosheet enhanced the production, retention, and biological activity of endogenous bone morphogenetic proteins (BMPs), resulting in robust osteogenesis in the absence of exogenous osteoinductive growth factors. Specifically, the osteoinductive effect of the nanosheets was abolished by inhibiting the BMP signaling pathway with LDN-193189 treatment. The bone formation potential of the technology was further tested in vivo using a mouse subcutaneous implantation model, where hMSCs-laden GO/GelMA and SiGO/GelMA samples resulted in bone volumes 108 and 385 times larger, respectively, than the GelMA control group. Taken together, these results demonstrate the biological activity and mechanism of action of GO-based nanosheets in augmenting the osteogenic capability of hMSCs, and highlights the potential of leveraging nanomaterials such as GO and SiGO for bone tissue engineering applications.

Introduction

As the median age of the world's population increases, the need for bone repair and regeneration is trending steeply upward [1]. Conventional bone defect management includes bone transplants, with autografts being the “gold standard” [2]. Although these natural grafts usually lead to increased osteoinduction and osteoconduction, they have a number of drawbacks, such as limited availability and donor site morbidity, as well as risks of infection and disease transmission [3]. As an alternative, stem cell-based tissue engineering offers a promising approach to treating bone damage. In particular, human mesenchymal stem cells (hMSCs), which are multipotent cells present in a number of tissues throughout the body, are among the most widely studied cell sources for bone tissue engineering [4]. For example, we recently seeded hMSCs in a gene-activated, methacrylated gelatin (GelMA)-based scaffold and achieved robust reparative capacity in cranial bone defects in mice [5].

To induce osteogenic differentiation of hMSCs, growth factors such as bone morphogenetic proteins (BMPs) are usually required. Among the different BMPs, BMP2 and BMP7 are both highly osteoinductive and have been used in commercially available medical products [6]. However, as is the case with other growth factors, BMP application in tissue engineering is generally hindered by its low stability, short half-life, high cost of production, and undesirable potential side effects, including carcinogenesis [7]. Therefore, growth factor-free approaches to bone tissue engineering has attracted increasing attention [8,9]. Previous studies by our group and others have shown that delivery of osteogenic genes to hMSCs could lead to a sustained production of osteogenic growth factors such as BMPs at physiologically relevant concentrations, resulting in enhanced osteogenic differentiation and mineralization of hMSCs in vitro and in vivo [5,10,11]. However, the safety of such gene delivery strategies warrants further investigation to reach clinical trials.

Discoveries and advances in nanomaterials and nanocomposites in recent decades has greatly advanced technological developments in tissue engineering and regenerative medicine [12,13]. The importance of the nano-dimension and -topography of the components of the native cellular niche provides the conceptual rationale driving these developments. For example, Reznikov et al. [14] identified the hierarchical assembly of bone minerals and collagen at the nanoscale. Recently, the application of graphene and graphene derivatives in tissue engineering has been enthusiastically pursued [15]. These two-dimensional (2D) nanomaterials possess remarkable electrical, mechanical, and thermal properties [16,17]; in addition, the large specific surface area makes graphene nanomaterials effective drug delivery carriers [18,19]. Nayak et al. [20] reported that hMSCs seeded on graphene-coated surfaces had accelerated osteogenic differentiation. Similar results were reported in another study, which demonstrated graphene's ability to preconcentrate β-glycerolphosphate and dexamethasone, two important osteogenic inducers [21]. Lu et al. fabricated free-standing, multi-layered graphene membranes, which facilitated in vivo bone formation via enhanced protein adsorption [22,23]. These studies suggest that graphene nanosheets can independently induce robust osteogenesis, thus overcoming the limitation of using osteogenic growth factors in bone tissue engineering. However, the mechanism governing the responses of cells/tissues to nano-dimensional materials such as graphene is incompletely understood. Future success in clinical applications of nanomaterials requires a more in-depth understanding of how nanomaterials influence cell behavior. In this study, we have characterized BMP signaling in hMSCs encapsulated in 3D nanocomposite scaffolds, in order to gain insights into the nature of biomolecular interactions between hMSCs and 2D nanomaterials.

Hydrogels, such as GelMA, represent an important type of scaffold for bone regeneration. With their high water content, hydrogels provide a tissue-like, biocompatible, 3D environment for cell culture. In particular, injectable and degradable hydrogels can conform to the shape of irregular bone defects and can be gradually replaced via natural biodegradation by the new, growing tissue. However, compared with commonly used biometals (e.g., Ti and Mg alloys) and bioceramics (e.g., tricalcium phosphate, hydroxyapatite), hydrogels usually have reduced osteoinductivity and substantially lower mechanical strengths [24,25]. With the incorporation of nanomaterials such as graphene, hydrogel-based nanocomposites can serve as a versatile bone tissue engineering platform that allows for the generation of 3D cell-laden constructs with tunable mechanical, structural, and biological characteristics [26]. For example, our previous study reported that graphene oxide (GO), a graphene derivative with high aqueous dispersity, increased the Young's modulus of a hydrogel-based cartilage scaffold [27]. GO incorporation in hydrogel scaffolds was also reported to enhance osteogenic differentiation of the encapsulated hMSCs [28,29]. However, the underlying mechanisms need to be further explored to pave the road for their future clinical applications.

In this study, we aimed to incorporate GO-based nanosheets to enhance hMSC osteogenesis within GelMA. We hypothesized that the nanosheet-functionalized hydrogels could induce robust osteogenesis of hMSCs without the supplementation of osteoinductive growth factors. To test this hypothesis, we first prepared 3D constructs consisting of GO-encapsulated, hMSC-laden hydrogel scaffolds, and osteogenesis was assessed after 28 days of culture in growth factor-free osteogenic medium (OM). Moreover, in light of the reported benefits of silicon in bone metabolism [30], we tested the potential of a novel GO derivative—silica-coated GO (SiGO)—in supporting hMSC osteogenesis, which has not been investigated previously. To explore the mechanism responsible for nanosheet-induced osteogenesis, we first assessed the presence of endogenous osteogenic growth factors, such as BMP2 and BMP7, and examined their interactions with the carbonaceous nanosheets. Next, the BMP antagonist, LDN-193189, was used to test the involvement of the BMP signaling pathway in the pro-osteogenesis effect of the nanosheets. Finally, a mouse subcutaneous implantation model was employed to assess the ability of constructs of hMSC-laden, GO- or SiGO-functionalized GelMA scaffolds for in vivo bone formation.

Section snippets

hMSC isolation and characterization

Bone marrow was obtained by flushing the femoral heads and trabecular bone collected from patients undergoing total hip arthroplasty (approved by Institutional Review Board, University of Washington and University of Pittsburgh). After several rounds of rinsing, the hMSCs were plated in T150 flasks (Corning Inc., corning, NY) and cultured in growth medium [GM, Dulbecco's Modified Eagle's Medium (DMEM; Gibco, Grand Island, NY) supplemented with 10% (v/v) fetal bovine serum (FBS; Gemini

Characterization of GO and SiGO nanosheets

Fig. 1A shows the schematic of converting GO nanosheets to SiGO. While GO displayed smooth, fabric-like morphology under TEM, SiGO appeared wavy and thicker (Fig. 1B and C). The topographical difference between GO and SiGO was clearly seen in the AFM micrographs (Fig. 1D and E). The height profile recorded from AFM imaging revealed a sheet thickness of ~1.3 nm for GO and ~46 nm for SiGO (Fig. 1D and E). Both D and G bands, characteristic of graphitic materials and indicative of structural

Discussion

Leveraging functional nanomaterials and their interactions with hMSCs represents a promising approach to growth factor-free bone tissue engineering. The results of in vitro and in vivo tests in this study show that matrix mineralization of 3D, hMSC-laden GelMA constructs was significantly enhanced with the incorporation of GO and SiGO nanosheets. Mechanistically, the introduction of the GO-based nanosheets increased the production, retention and function of endogenous, cell-secreted BMPs,

Conclusions

The incorporation of GO and SiGO nanosheets in 3D GelMA scaffolds markedly enhanced osteogenesis of encapsulated hMSCs both in vitro and in vivo. These 2D nanomaterials acted as “BMP reservoirs” that enabled effective adsorption and release of endogenous BMP2 and BMP7, which together acted to enhance osteogenesis accompanied by high activation of SMAD1/5. In addition, the beneficial osteoinductive effects of GO was effectively amplified by surface functionalization with a silica coating,

Credit author statement

Zhong Li: Conceptualization, Methodology, Investigation, Software, Data Curation, Writing - Original Draft, Writing - Review & Editing. Shiqi Xiang: Conceptualization, Methodology, Investigation, Validation, Writing - Review & Editing. Zixuan Lin: Methodology, Investigation, Data Curation. Eileen N. Li: Methodology, Investigation, Formal analysis. Haruyo Yagi: Methodology, Investigation. Guorui Cao: Methodology, Investigation. Lauren Yocum: Methodology, Investigation, Writing - Review &

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

Dr. Paul Manner (University of Washington) is gratefully acknowledged for providing the human tissue samples. We thank Dr. Jian Tan for isolating the human hMSCs, and Dr. Shuguang Bi for technical assistance with material preparation. This research was supported by funding from the National Institutes of Health (UG3/UH3TR002136) and Department of Orthopaedic Surgery at the University of Pittsburgh School of Medicine (to HL). The Center for Biological Imaging (CBI) at University of Pittsburgh

References (67)

  • T. Kihara et al.

    Three-dimensional visualization analysis of in vitro cultured bone fabricated by rat marrow mesenchymal stem cells

    Biochem. Biophys. Res. Commun.

    (2004)
  • A.S. Mao et al.

    Effects of substrate stiffness and cell-cell contact on mesenchymal stem cell differentiation

    Biomaterials

    (2016)
  • J.R. Henstock et al.

    Silicon: the evolution of its use in biomaterials

    Acta Biomater.

    (2015)
  • E.M. Carlisle

    In vivo requirement for silicon in articular cartilage and connective tissue formation in the chick

    J. Nutr.

    (1976)
  • J.R. Jones

    Review of bioactive glass: from Hench to hybrids

    Acta Biomater.

    (2013)
  • E.S. Thian et al.

    Silicon-substituted hydroxyapatite: the next generation of bioactive coatings

    Mater. Sci. Eng. C

    (2007)
  • H. Li et al.

    Silicate bioceramics enhanced vascularization and osteogenesis through stimulating interactions between endothelia cells and bone marrow stromal cells

    Biomaterials

    (2014)
  • Ž. Mladenović et al.

    Soluble silica inhibits osteoclast formation and bone resorption in vitro

    Acta Biomater.

    (2014)
  • D. Jiao et al.

    Bidirectional differentiation of BMSCs induced by a biomimetic procallus based on a gelatin-reduced graphene oxide reinforced hydrogel for rapid bone regeneration

    Bioactive Materials

    (2021)
  • C. Acevedo et al.

    Fatigue as the missing link between bone fragility and fracture

    Nat. Biomed. Eng

    (2018)
  • C. Laurencin et al.

    Bone graft substitutes

    Expet Rev. Med. Dev.

    (2006)
  • J.J. Li et al.

    Current approaches to bone tissue engineering: the interface between biology and engineering

    Adv. Healthc. Mater.

    (2018)
  • K. Sun et al.

    Injectable BMP-2 gene-activated scaffold for the repair of cranial bone defect in mice

    Stem Cells Transl. Med.

    (2020)
  • P.C. Bessa et al.

    Bone morphogenetic proteins in tissue engineering: the road from laboratory to clinic, part II (BMP delivery)

    J. Tissue Eng. Regen. Med.

    (2008)
  • B. Skovrlj et al.

    Association between BMP-2 and carcinogenicity

    Spine

    (2015)
  • Z.-K. Cui et al.

    Microporous methacrylated glycol chitosan-montmorillonite nanocomposite hydrogel for bone tissue engineering

    Nat. Commun.

    (2019)
  • J.R. Xavier et al.

    Bioactive nanoengineered hydrogels for bone tissue engineering: a growth-factor-free approach

    ACS Nano

    (2015)
  • H. Lin et al.

    Projection stereolithographic fabrication of BMP-2 gene-activated matrix for bone tissue engineering

    Sci. Rep.

    (2017)
  • M.M. Alvarez et al.

    Emerging trends in micro- and nanoscale Technologies in medicine: from basic discoveries to translation

    ACS Nano

    (2017)
  • N. Reznikov et al.

    Fractal-like hierarchical organization of bone begins at the nanoscale

    Science

    (2018)
  • C. Lee et al.

    Measurement of the elastic properties and intrinsic strength of monolayer graphene

    Science

    (2008)
  • K.S. Novoselov et al.

    Electric field effect in atomically thin carbon films

    Science

    (2004)
  • W.-G. La et al.

    Delivery of a therapeutic protein for bone regeneration from a substrate coated with graphene oxide

    Small

    (2013)
  • Cited by (43)

    • Graphene oxide/gallium nanoderivative as a multifunctional modulator of osteoblastogenesis and osteoclastogenesis for the synergistic therapy of implant-related bone infection

      2023, Bioactive Materials
      Citation Excerpt :

      The antimicrobial actions of GO nanosheets is mainly attributed to oxidative stress-associated membrane destruction [20]. In addition, GO nanosheets have clear osteogenic and angiogenic properties, leading to reliable anti-infective and osteoconductive capacities for local therapy of implant-related bone infection [21,22]. Gallium (Ga) is a semimetallic element that exhibits antitumor, antiresorptive, and anti-inflammatory properties, rendering it an ideal candidate for the treatment of Paget's disease, myeloma and malignancy with hypercalcemia [23,24].

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    2

    Current address: The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China. Email: [email protected]

    View full text