Elsevier

Biomaterials

Volume 27, Issue 32, November 2006, Pages 5535-5545
Biomaterials

Mineralization capacity of Runx2/Cbfa1-genetically engineered fibroblasts is scaffold dependent

https://doi.org/10.1016/j.biomaterials.2006.06.019Get rights and content

Abstract

Development of tissue-engineered constructs for skeletal regeneration of large critical-sized defects requires the identification of a sustained mineralizing cell source and careful optimization of scaffold architecture and surface properties. We have recently reported that Runx2-genetically engineered primary dermal fibroblasts express a mineralizing phenotype in monolayer culture, highlighting their potential as an autologous osteoblastic cell source which can be easily obtained in large quantities. The objective of the present study was to evaluate the osteogenic potential of Runx2-expressing fibroblasts when cultured in vitro on three commercially available scaffolds with divergent properties: fused deposition-modeled polycaprolactone (PCL), gas-foamed polylactide-co-glycolide (PLGA), and fibrous collagen disks. We demonstrate that the mineralization capacity of Runx2-engineered fibroblasts is scaffold dependent, with collagen foams exhibiting ten-fold higher mineral volume compared to PCL and PLGA matrices. Constructs were differentially colonized by genetically modified fibroblasts, but scaffold-directed changes in DNA content did not correlate with trends in mineral deposition. Sustained expression of Runx2 upregulated osteoblastic gene expression relative to unmodified control cells, and the magnitude of this expression was modulated by scaffold properties. Histological analyses revealed that matrix mineralization co-localized with cellular distribution, which was confined to the periphery of fibrous collagen and PLGA sponges and around the circumference of PCL microfilaments. Finally, FTIR spectroscopy verified that mineral deposits within all Runx2-engineered scaffolds displayed the chemical signature characteristic of carbonate-containing, poorly crystalline hydroxyapatite. These results highlight the important effect of scaffold properties on the capacity of Runx2-expressing primary dermal fibroblasts to differentiate into a mineralizing osteoblastic phenotype for bone tissue engineering applications.

Introduction

Conventional orthopedic grafting templates based on autogenic bone, allogenic bone, or synthetic materials are widely utilized for the clinical treatment of non-healing skeletal defects. Although successful in many cases, these grafts remain limited by inadequate osseo-integration, donor site morbidity, poor mechanical properties, and/or the risk of disease transmission [1], [2], [3], [4], [5], [6]. Bone tissue engineering has emerged as a promising strategy to overcome complications associated with these traditional skeletal repair therapies [7], [8], [9], [10]. Tissue-engineered bone substitutes have been successfully developed through the integration of osteoinductive growth factors and/or osteogenic cells into an osteoconductive scaffolding matrix. Notably, several groups have demonstrated in vitro and in vivo mineralization and repair of bone defects by combining marrow-derived and mesenchymal stem cells with three-dimensional (3-D) scaffolds [11], [12], [13], [14], [15], [16]. Despite these advances, the development of mechanically robust skeletal grafts which are immunoaccepted by the host and are capable of healing large, critical-sized defects has not been realized.

One significant barrier toward the clinical application of tissue-engineered bone grafts is the inadequate availability of a sustained mineralizing cell source. In order to address this limitation, genetic engineering strategies have been developed for the induction of osteoblastic differentiation in nonosteogenic cells [17], [18], [19], [20]. In particular, gene delivery of soluble factors, such as BMP-2 and BMP-7, or osteogenic transcription factors, such as Runx2/Cbfa1, has been investigated for the conversion of primary fibroblasts and fibroblastic cell lines into an osteoblastic phenotype [21], [22], [23], [24]. We have recently demonstrated that retroviral Runx2 overexpression induces significant levels of mineral deposition in primary dermal fibroblast monolayer cultures [25]. These genetically modified fibroblasts have considerable potential as a cell source for bone tissue engineering applications because they are easily obtained from autologous donors through minimally invasive skin biopsy and have a high capacity for in vitro expansion.

In addition to the identification of an autologous mineralizing cell source, the successful development of bone grafting templates requires careful optimization of scaffold architecture and surface properties. Biomaterial scaffolds typically function as a three-dimensional structural support, which facilitates tissue integration into the skeletal defect site and promotes cell attachment, proliferation, and differentiation into functional osteoblasts. Various classes of materials have been considered for bone grafting applications, including ceramics, natural and synthetic polymers, and their composites [26]. Among these, scaffolds based on naturally derived collagen and synthetic polycaprolactone (PCL) and polylactide-co-glycolide (PLGA) polymers were selected for investigation in this study because of their widespread use in tissue engineering applications, well-documented biodegradation profile, FDA-approval, and commercial availability [26], [27], [28], [29]. These scaffolds present a broad range of architectural and surface properties (e.g. topography, surface chemistry, roughness) that may potentially influence the biological response of seeded cells [30]. The objective of the present work was to evaluate the ability of three commonly utilized, commercially available scaffolds to support in vitro matrix mineralization when seeded with Runx2-expressing fibroblasts.

Section snippets

Cells and culture reagents

Primary fibroblasts were harvested from 8- to 16-week-old male Wistar rats by enzymatic digestion of the dermal skin layer [31]. Cells were expanded in growth media consisting of DMEM, 10% fetal bovine serum, and 1% penicillin–streptomycin. Antibiotics and cell culture media were obtained from Invitrogen (Carlsbad, CA), fetal bovine serum was purchased from Hyclone (Logan, UT), and all other cell culture supplements and reagents were acquired from Sigma (St. Louis, MO).

Retroviral transduction

The Runx2 retroviral

Cellular viability

Runx2-expressing and unmodified fibroblasts were seeded on collagen, PCL, and PLGA scaffolds at a density of 500,000 cells/construct and cultured in vitro under static conditions in osteogenic differentiation media. Scaffold colonization and cellular viability were assessed at 1, 21, and 42 days post-seeding by confocal microscopy and live/dead staining (Fig. 1). After 1 day in culture, cells displayed a fibroblastic morphology and were evenly distributed throughout all three scaffolds. Marked

Discussion

We demonstrate that the osteogenic potential of Runx2-expressing fibroblasts is highly dependent on the architecture and surface properties of polymeric scaffolds. Micro-CT imaging revealed that genetically modified fibroblasts deposit significantly higher levels of mineral on fibrous collagen disks relative to constructs based on PCL and PLGA. The chemical composition of the mineral phase on all three Runx2-engineered scaffolds was verified by FTIR spectroscopy to display the characteristic

Conclusions

We demonstrate that the osteogenic potential of Runx2-expressing fibroblasts is highly dependent on scaffold properties, with fibrous collagen disks exhibiting significantly higher mineral deposition than gas-foamed PLGA sponges and fused deposition modeled PCL. These results highlight the importance of scaffold optimization in the development of tissue-engineered bone constructs.

Acknowledgments

Collagen scaffolds were generously donated by Kensey Nash Corporation. The authors thank Angela S. Lin and Srinidhi Nagaraja for technical assistance with micro-CT and Tracey L. Couse for technical assistance with histology. This research was funded by the NIH (R01-EB003364), the Georgia Tech/Emory Engineering Research Center on the Engineering of Living Tissues (NSF EEC-9731643) and a National Science Foundation Graduate Research Fellowship to JEP.

References (81)

  • G.R. Sauer et al.

    Fourier transform infrared characterization of mineral phases formed during induction of mineralization by collagenase-released matrix vesicles in vitro

    J Biol Chem

    (1988)
  • B.D. Boyan et al.

    The importance of mineral in bone and mineral research

    Bone

    (2000)
  • P. Tengvall et al.

    Protein adsorption studies on model organic surfaces: an ellipsometric and infrared spectroscopic approach

    Biomaterials

    (1998)
  • M.A. Lan et al.

    Myoblast proliferation and differentiation on fibronectin-coated self assembled monolayers presenting different surface chemistries

    Biomaterials

    (2005)
  • B.G. Keselowsky et al.

    Surface chemistry modulates focal adhesion composition and signaling through changes in integrin binding

    Biomaterials

    (2004)
  • E. Tziampazis et al.

    PEG-variant biomaterials as selectively adhesive protein templates: model surfaces for controlled cell adhesion and migration

    Biomaterials

    (2000)
  • M.B. Gorbet et al.

    Leukocyte activation and leukocyte procoagulant activities after blood contact with polystyrene and polyethylene glycol-immobilized polystyrene beads

    J Lab Clin Med

    (2001)
  • B.S. Chang et al.

    Osteoconduction at porous hydroxyapatite with various pore configurations

    Biomaterials

    (2000)
  • Y.C. Wu et al.

    Bone tissue engineering evaluation based on rat calvaria stromal cells cultured on modified PLGA scaffolds

    Biomaterials

    (2006)
  • J. Glowacki et al.

    Perfusion enhances functions of bone marrow stromal cells in three-dimensional culture

    Cell Transplant

    (1998)
  • M.P. Lynch et al.

    The influence of type I collagen on the development and maintenance of the osteoblast phenotype in primary and passaged rat calvarial osteoblasts: modification of expression of genes supporting cell growth, adhesion, and extracellular matrix mineralization

    Exp Cell Res

    (1995)
  • G.Z. Xiao et al.

    Role of the alpha(2)-integrin in osteoblast-specific gene expression and activation of the Osf2 transcription factor

    J Biol Chem

    (1998)
  • R.W. Bucholz

    Nonallograft osteoconductive bone graft substitutes

    Clin Orthop Relat Res

    (2002)
  • C.G. Finkemeier

    Bone-grafting and bone-graft substitutes

    J Bone Joint Surg Am

    (2002)
  • J.M. Lane et al.

    Biosynthetic bone grafting

    Clin Orthop Relat Res

    (1999)
  • C.R. Perry

    Bone repair techniques, bone graft, and bone graft substitutes

    Clin Orthop Relat Res

    (1999)
  • A.R. Vaccaro

    The role of the osteoconductive scaffold in synthetic bone graft

    Orthopedics

    (2002)
  • S.P. Bruder et al.

    Tissue engineering of bone. Cell based strategies

    Clin Orthop Relat Res

    (1999)
  • A.S. Mistry et al.

    Tissue engineering strategies for bone regeneration

    Adv Biochem Eng Biotechnol

    (2005)
  • T.L. Arinzeh et al.

    Allogeneic mesenchymal stem cells regenerate bone in a critical-sized canine segmental defect

    J Bone Joint Surg Am

    (2003)
  • S.P. Bruder et al.

    Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy

    J Cell Biochem

    (1994)
  • J.R. Mauney et al.

    Role of adult mesenchymal stem cells in bone tissue engineering applications: current status and future prospects

    Tissue Eng

    (2005)
  • A. Kahn et al.

    Age-related bone loss. A hypothesis and initial assessment in mice

    Clin Orthop Relat Res

    (1995)
  • H. Ohgushi et al.

    Repair of bone defects with marrow cells and porous ceramic. Experiments in rats

    Acta Orthop Scand

    (1989)
  • R. Quarto et al.

    Repair of large bone defects with the use of autologous bone marrow stromal cells

    N Engl J Med

    (2001)
  • R.T. Franceschi et al.

    Gene therapy approaches for bone regeneration

    Cells Tissues Organs

    (2004)
  • R.B. Rutherford et al.

    Bone morphogenetic protein-transduced human fibroblasts convert to osteoblasts and form bone in vivo

    Tissue Eng

    (2002)
  • M. Zhao et al.

    Combinatorial gene therapy for bone regeneration: cooperative interactions between adenovirus vectors expressing bone morphogenetic proteins 2, 4, and 7

    J Cell Biochem

    (2005)
  • R.T. Franceschi et al.

    Gene therapy for bone formation: in vitro and in vivo osteogenic activity of an adenovirus expressing BMP7

    J Cell Biochem

    (2000)
  • P.H. Krebsbach et al.

    Gene therapy-directed osteogenesis: BMP-7-transduced human fibroblasts form bone in vivo

    Hum Gene Ther

    (2000)
  • Cited by (44)

    • Digital light processing porous TPMS structural HA & akermanite bioceramics with optimized performance for cancellous bone repair

      2022, Ceramics International
      Citation Excerpt :

      As bone implant substitutes, artificial bone scaffolds represent important components for bone tissue engineering. In addition, an ideal bone scaffold needs high porosity and excellent bioactivity and appropriate mechanical strength to promote bone repair faster and reduce patient suffering [5–7]. Combining material and structural design to regulate properties of bioceramic scaffolds has become a hot research topic [8–10].

    • Designing and validation of an automated ex-vivo bioreactor system for long term culture of bone

      2021, Bone Reports
      Citation Excerpt :

      From the live-dead fluorescent studies, we observed that the bone cores that were cultured in the static medium had more dead cells when compared with the bone cores cultured in the bioreactor group. It has been found that in static organ culture, the transfer of nutrients to the center of the tissue does not take place, leading to cell death (Phillips et al., 2006). We also found that the pH was stable in all groups at week 1 but with a slight decrease in value compared to the control medium's (7.32 ± 0.05) (Table 1).

    • In vitro proliferation and differentiation potential of bone marrow-derived mesenchymal stem cells from ovariectomized rats

      2014, Tissue and Cell
      Citation Excerpt :

      In the present study, we provided new evidence that different from the cellular senescence and disfunction in age-related osteoporosis, OVXBMMSCs are endowed with higher self-renewal, osteoblastic and adipogenic differentiation potential than ShamBMMSCs in culture. More specifically, this action may be partly attributed to the up-regulation of osteoblastic special markers Cbfa1, COL-I and Lrp5, as well as adipogenic special markers PPARγ, C/EBPα and aP2, that play hierarchically dominant roles in controlling the adipocyte/osteoblast relationship within the bone cavity (Backesjo et al., 2006; Phillips et al., 2006) and have been identified as well-established transduction mechanisms for various interventions acting on bone formation (Lin and Hankenson, 2011). These findings are not entirely in line with the observed bone loss in OVX rats.

    • The effects of the modulation of the fibronectin-binding capacity of fibrin by thrombin on osteoblast differentiation

      2012, Biomaterials
      Citation Excerpt :

      Based on these previous reports, we hypothesized that osteoblast differentiation is not caused by a direct cell response to thrombin via PAR1 but by a response caused by thrombin-induced alterations in fibrin structures. Runx2 has been widely accepted as an early osteogenic transcription factor that can switch on to osteoblast differentiation [22–26]. Runx2-knockout mice display complete bone loss due to arrested osteoblast maturation, and Runx2 plays a critical role in osteoblast marker gene expression including ALP, osteocalcin, and bone sialoprotein [22,23].

    • Engineered Proteins for Controlling Gene Expression

      2012, Handbook of Stem Cells, Second Edition: Volume 1-2
    View all citing articles on Scopus
    View full text