scispace - formally typeset
Search or ask a question

Showing papers in "European Cells & Materials in 2011"


Journal ArticleDOI
TL;DR: The properties of osteoinductive biomaterials are reviewed, in the light of the model and the conditions under which they were tested, to give an insight into the biological processes governing osteo Induction by biommaterials and the view on the future perspectives in this research field.
Abstract: In the past thirty years, a number of biomaterials have shown the ability to induce bone formation when implanted at heterotopic sites, an ability known as osteoinduction. Such biomaterials – osteoinductive biomaterials – hold great potential for the development of new therapies in bone regeneration. Although a variety of well characterised osteoinductive biomaterials have so far been reported in the literature, scientists still lack fundamental understanding of the biological mechanism underlying the phenomenon by which they induce bone formation. This is further complicated by the observations that larger animal models are required for research, since limited, if any, bone induction by biomaterials is observed in smaller animals, including particularly rodents. Besides interspecies variation, variations among individuals of the same species have been observed. Furthermore, comparing different studies and drawing general conclusions is challenging, as these usually differ not only in the physico-chemical and structural properties of the biomaterials, but also in animal model, implantation site and duration of the study. Despite these limitations, the knowledge of material properties relevant for osteoinduction to occur has tremendously increased in the past decades. Here we review the properties of osteoinductive biomaterials, in the light of the model and the conditions under which they were tested. Furthermore, we give an insight into the biological processes governing osteoinduction by biomaterials and our view on the future perspectives in this research field.

448 citations


Journal ArticleDOI
TL;DR: A thorough understanding of the similarities and particularly the marked differences in mechanisms of cartilage remodeling during development, osteoarthritis, and aging may lead to more effective strategies for preventing cartilage damage and promoting repair.
Abstract: Human cartilage is a complex tissue of matrix proteins that vary in amount and orientation from superficial to deep layers and from loaded to unloaded zones. A major challenge to efforts to repair cartilage by stem cell-based and other tissue engineering strategies is the inability of the resident chondrocytes to lay down new matrix with the same structural and resilient properties that it had upon its original formation. This is particularly true of the collagen network, which is susceptible to cleavage once proteoglycans are depleted. Thus, a thorough understanding of the similarities and particularly the marked differences in mechanisms of cartilage remodeling during development, osteoarthritis, and aging may lead to more effective strategies for preventing cartilage damage and promoting repair. To identify and characterize effectors or regulators of cartilage remodeling in these processes, we are using culture models of primary human and mouse chondrocytes and cell lines and mouse genetic models to manipulate gene expression programs leading to matrix remodeling and subsequent chondrocyte hypertrophic differentiation, pivotal processes which both go astray in OA disease. Matrix metalloproteinases (MMP)-13, the major type II collagen-degrading collagenase, is regulated by stress-, inflammation-, and differentiation-induced signals that not only contribute to irreversible joint damage (progression) in OA, but importantly, also to the initiation/onset phase, wherein chondrocytes in articular cartilage leave their natural growth- and differentiation-arrested state. Our work points to common mediators of these processes in human OA cartilage and in early through late stages of OA in surgical and genetic mouse models.

428 citations


Journal ArticleDOI
TL;DR: This study defines non-toxic conditions for photoencapsulation of human mesenchymal stem cells (hMSC) in PEGDA scaffolds using a visible light photoinitiator system composed of eosin Y, triethanolamine and 1-vinyl-2-pyrrolidinone.
Abstract: Biological activity can be added to synthetic scaffolds by incorporating functional peptide sequences that provide enzyme-mediated degradation sites, facilitate cellular adhesion or stimulate signaling pathways. Poly(ethylene glycol) diacrylate is a popular synthetic base for tissue engineering scaffolds because it creates a hydrophilic environment that can be chemically manipulated to add this biological functionality. Furthermore, the acrylate groups allow for encapsulation of cells using photopolymerization under physiological conditions. One complication with the addition of these peptides is that aromatic amino acids absorb light at 285 nm and compete with the ultraviolet (UV)-sensitive photoinitiators such as IrgacureTM 2959 (I2959), the most commonly used initiator for cytocompatible photoencapsulation of cells into synthetic scaffolds. In this study we define non-toxic conditions for photoencapsulation of human mesenchymal stem cells (hMSC) in PEGDA scaffolds using a visible light photoinitiator system composed of eosin Y, triethanolamine and 1-vinyl-2-pyrrolidinone. This visible light photoinitiator produced hydrogel scaffolds with an increased viability of encapsulated hMSCs and a more tightly crosslinked network in one-third the time of UV polymerization with I2959.

198 citations


Journal ArticleDOI
TL;DR: This study investigated the potential enhancing effect of surface shear, superimposed on cyclic axial compression, on chondrogenic differentiation of human bone marrow-derived stem cells and the results obtained may provide insight into post-operative care after cell therapy involving mesenchymal stromal cells.
Abstract: There is great interest in how bone marrow derived stem cells make fate decisions. Numerous studies have investigated the role of individual growth factors on mesenchymal stem cell differentiation, leading to protocols for cartilage, bone and adipose tissue. However, these protocols overlook the role of biomechanics on stem cell differentiation. There have been various studies that have applied mechanical stimulation to constructs containing mesenchymal stem cells, with varying degrees of success. One critical fate decision is that between cartilage and bone. Articular motion is a combination of compressive, tensile and shear deformations; therefore, one can presume that compression alone is unlikely to be a sufficient mechanical signal to generate a cartilage-like tissue in vitro. Within this study, we aimed to determine the role of shear on the fate of stem cell differentiation. Specifically, we investigated the potential enhancing effect of surface shear, superimposed on cyclic axial compression, on chondrogenic differentiation of human bone marrow-derived stem cells. Using a custom built loading device we applied compression, shear or a combination of both stimuli onto fibrin/polyurethane composites in which human mesenchymal stem cells were embedded, while no exogenous growth-factors were added to the culture medium. Both compression or shear alone was insufficient for the chondrogenic induction of human mesenchymal stem cells. However, the application of shear superimposed upon dynamic compression led to significant increases in chondrogenic gene expression. Histological analysis detected sulphated glycosaminoglycan and collagen II only in the compression and shear group. The results obtained may provide insight into post-operative care after cell therapy involving mesenchymal stromal cells.

176 citations


Journal ArticleDOI
TL;DR: It is suggested strongly that endothelial cells regulate the MSC activity in simulated in vivo conditions, by maintaining quiescence and facilitating niche exit via osteogenic differentiation following appropriate cues.
Abstract: An increasing body of data suggest that mesenchymal stem cells (MSCs) reside in a perivascular niche. To more closely mimic this in vivo microenvironment and for better understanding of its complexity, and the factors that regulate the MSC activity, human umbilical vein endothelial cells (HUVECs) were co-cultured with human bone marrow MSCs--using a novel three-dimensional (3D) spheroid co-culture system. Using confocal microscopy of fluorescently labelled cells, we observed HUVECs and MSCs to self-assemble and form organised structures with segregated cell-type partitioning. Under osteogenic conditions, the rate and extent of differentiation in MSC/HUVEC spheroids was significantly elevated compared to 3D co-cultures of MSCs and human dermal fibroblast controls as shown by alkaline phosphatase staining. Conversely, HUVECs inhibited adipogenic differentiation and the proliferation of MSCs in 3D co-cultures indicating that HUVECs suppressed MSC cycling and selectively promoted osteogenic differentiation in 3D. We have also shown that HUVECs enhanced activation of endogenous Wnt signalling and bone morphogenetic protein (BMP) signalling as shown by increased levels of active nuclear β-catenin and pSmad 1/5/8 immunopositivity respectively. These data suggest strongly that endothelial cells regulate the MSC activity in simulated in vivo conditions, by maintaining quiescence and facilitating niche exit via osteogenic differentiation following appropriate cues. Our findings also underline the importance of 3D heterotypic cell-cell interactions in the regulation of MSC behaviour, suggesting that multicellular cocktails and/or 3D-based delivery strategies may be beneficial for bone repair.

164 citations


Journal ArticleDOI
TL;DR: Osteoblast differentiation of foetal mouse calvarial cells was enhanced in the presence of bioactive glass particles containing 5 wt.% strontium, suggesting B75-Sr5 may represent a promising bone-grafting material for bone regeneration procedures.
Abstract: There is accumulating evidence that strontium-containing biomaterials have positive effects on bone tissue repair. We investigated the in vitro effect of a new Sr-doped bioactive glass manufactured by the sol-gel method on osteoblast viability and differentiation. Osteoblasts isolated from foetal mouse calvaria were cultured in the presence of bioactive glass particles; particles were undoped (B75) or Sr-doped with 1 wt.% (B75-Sr1) and 5 wt.% (B75-Sr5). Morphological analysis was carried out by contrast-phase microscopy and scanning electron microscopy (SEM). Cell viability was evaluated by the MTS assay at 24 h, 48 h and 72 h. At 24 h, day 6 and day 12, osteoblast differentiation was evaluated by assaying alkaline phosphatase (ALP) activity, osteocalcin (OC) secretion and gene expression of various bone markers, using Real-Time-PCR. Alizarin Red staining and ALP histoenzymatic localisation were performed on day 12. Microscopic observations and MTS showed an absence of cytotoxicity in the three investigated bioactive glasses. B75-Sr5 particles in cell cultures, in comparison with those of B75 and B75-Sr1, resulted in a significant up-regulation of Runx2, Osterix, Dlx5, collagen I, ALP, bone sialoprotein (BSP) and OC mRNA levels on day 12, which was associated with an increase of ALP activity on day 6 and OC secretion on day 12. In conclusion, osteoblast differentiation of foetal mouse calvarial cells was enhanced in the presence of bioactive glass particles containing 5 wt.% strontium. Thus, B75-Sr5 may represent a promising bone-grafting material for bone regeneration procedures.

160 citations


Journal ArticleDOI
TL;DR: Hypoxia and GDF5 may be suitable for directing MSCs towards the IVD-like phenotype and trends in gene expression regulation were confirmed on the protein level using immunohistochemistry.
Abstract: There is evidence that mesenchymal stem cells (MSCs) can differentiate towards an intervertebral disc (IVD)-like phenotype. We compared the standard chondrogenic protocol using transforming growth factor beta-1 (TGFs) to the effects of hypoxia, growth and differentiation factor-5 (GDF5), and coculture with bovine nucleus pulposus cells (bNPC). The efficacy of molecules recently discovered as possible nucleus pulposus (NP) markers to differentiate between chondrogenic and IVD-like differentiation was evaluated. MSCs were isolated from human bone marrow and encapsulated in alginate beads. Beads were cultured in DMEM (control) supplemented with TGFs or GDF5 or under indirect coculture with bNPC. All groups were incubated at low (2 %) or normal (20 %) oxygen tension for 28 days. Hypoxia increased aggrecan and collagen II gene expression in all groups. The hypoxic GDF5 and TGFs groups demonstrated most increased aggrecan and collagen II mRNA levels and glycosaminoglycan accumulation. Collagen I and X were most up-regulated in the TGFs groups. From the NP markers, cytokeratin-19 was expressed to highest extent in the hypoxic GDF5 groups; lowest expression was observed in the TGFs group. Levels of forkhead box F1 were down-regulated by TGFs and up-regulated by coculture with bNPC. Carbonic anhydrase 12 was also down-regulated in the TGFs group and showed highest expression in the GDF5 group cocultured with bNPC under hypoxia. Trends in gene expression regulation were confirmed on the protein level using immunohistochemistry. We conclude that hypoxia and GDF5 may be suitable for directing MSCs towards the IVD-like phenotype.

149 citations


Journal ArticleDOI
TL;DR: The dorsal skinfold chamber model does not only provide deep insights into the complex interactions of biomaterials with the surrounding soft tissues of the host but also represents an important tool for the future development of novel biommaterials aiming at an optimisation of their biofunctionality in clinical practice.
Abstract: The implantation of biomaterials into the human body has become an indispensable part of almost all fields of modern medicine. Accordingly, there is an increasing need for appropriate approaches, which can be used to evaluate the suitability of different biomaterials for distinct clinical indications. The dorsal skinfold chamber is a sophisticated experimental model, which has been proven to be extremely valuable for the systematic in vivo analysis of the dynamic interaction of small biomaterial implants with the surrounding host tissue in rats, hamsters and mice. By means of intravital fluorescence microscopy, this chronic model allows for repeated analyses of various cellular, molecular and microvascular mechanisms, which are involved in the early inflammatory and angiogenic host tissue response to biomaterials during the initial 2-3 weeks after implantation. Therefore, the dorsal skinfold chamber has been broadly used during the last two decades to assess the in vivo performance of prosthetic vascular grafts, metallic implants, surgical meshes, bone substitutes, scaffolds for tissue engineering, as well as for locally or systemically applied drug delivery systems. These studies have contributed to identify basic material properties determining the biocompatibility of the implants and vascular ingrowth into their surface or internal structures. Thus, the dorsal skinfold chamber model does not only provide deep insights into the complex interactions of biomaterials with the surrounding soft tissues of the host but also represents an important tool for the future development of novel biomaterials aiming at an optimisation of their biofunctionality in clinical practice.

142 citations


Journal ArticleDOI
TL;DR: It is demonstrated that genipin crosslinked fibrin gels show promise as a gap-filling adhesive biomaterial with tunable material properties, yet the slow cell proliferation suggests this biomaterial may be best suited as a sealant for small annulus fibrosus defects or as an adhesive to augment large annulus repairs.
Abstract: Treatment of damaged intervertebral discs is a significant clinical problem and, despite advances in the repair and replacement of the nucleus pulposus, there are few effective strategies to restore defects in the annulus fibrosus. An annular repair material should meet three specifications: have a modulus similar to the native annulus tissue, support the growth of disc cells, and maintain adhesion to tissue under physiological strain levels. We hypothesized that a genipin crosslinked fibrin gel could meet these requirements. Our mechanical results showed that genipin crosslinked fibrin gels could be created with a modulus in the range of native annular tissue. We also demonstrated that this material is compatible with the in vitro growth of human disc cells, when genipin:fibrin ratios were 0.25:1 or less, although cell proliferation was slower and cell morphology more rounded than for fibrin alone. Finally, lap tests were performed to evaluate adhesion between fibrin gels and pieces of annular tissue. Specimens created without genipin had poor handling properties and readily delaminated, while genipin crosslinked fibrin gels remained adhered to the tissue pieces at strains exceeding physiological levels and failed at 15-30%. This study demonstrated that genipin crosslinked fibrin gels show promise as a gap-filling adhesive biomaterial with tunable material properties, yet the slow cell proliferation suggests this biomaterial may be best suited as a sealant for small annulus fibrosus defects or as an adhesive to augment large annulus repairs. Future studies will evaluate degradation rate, fatigue behaviors, and long-term biocompatibility.

133 citations


Journal ArticleDOI
TL;DR: Overall, nanopores of 30 nm may promote early osteoblastic differentiation and, consequently, rapid osseointegration of titanium implants.
Abstract: Titanium implants are widely used in orthopaedic and dental surgery. Surface properties play a major role in cell and tissue interactions. The adhesion and differentiation of mesenchymal stem cells were studied as a function of nanostructures. Titanium surfaces with nanopores 30, 150 and 300 nm in diameter were prepared by physical vapour deposition. PCR arrays indicated that the expression of integrins was modulated by the nanopore size. Human Mesenchymal Stem Cells (hMSCs) exhibited more branched cell morphology on Ti30 than on other surfaces. Ti30 and Ti150 induced osteoblastic differentiation while Ti300 had a limited effect. Overall, nanopores of 30 nm may promote early osteoblastic differentiation and, consequently, rapid osseointegration of titanium implants.

121 citations


Journal ArticleDOI
TL;DR: It is demonstrated that an EF directed migration of BM-MSCs mainly to the anode, indicating that EFs are a powerful cue in directing migration of human MSCs in vitro.
Abstract: At sites of bone fracture, naturally-occurring electric fields (EFs) exist during healing and may guide cell migration. In this study, we investigated whether EFs could direct the migration of bone marrow mesenchymal stem cells (BM-MSCs), which are known to be key players in bone formation. Human BM-MSCs were cultured in direct current EFs of 10 to 600 mV/mm. Using time-lapse microscopy, we demonstrated that an EF directed migration of BM-MSCs mainly to the anode. Directional migration occurred at a low threshold and with a physiological EF of ~25 mV/mm. Increasing the EF enhanced the MSC migratory response. The migration speed peaked at 300 mV/mm, at a rate of 42 ±1 µm/h, around double the control (no EF) migration rate. MSCs showed sustained response to prolonged EF application in vitro up to at least 8 h. The electrotaxis of MSCs with either early (P3-P5) or late (P7-P10) passage was also investigated. Migration was passage-dependent with higher passage number showing reduced directed migration, within the range of passages examined. An EF of 200 mV/mm for 2 h did not affect cell senescence, phenotype, or osteogenic potential of MSCs, regardless of passage number within the range tested (P3-P10). Our findings indicate that EFs are a powerful cue in directing migration of human MSCs in vitro. An applied EF may be useful to control or enhance migration of MSCs during bone healing.

Journal ArticleDOI
TL;DR: Akermanite ceramic showed an enhanced effect on proliferation and osteogenic differentiation of hPDLCs, which might be attributed to the release of ions containing Ca, Mg and Si from the material.
Abstract: The purpose of this study was to investigate the effects of akermanite as compared to β-TCP on attachment, proliferation, and osteogenic differentiation of human periodontal ligament cells (hPDLCs). Scanning electron microscopy (SEM) and actin filament labeling were used to reveal attachment and growth of hPDLCs seeded on β-TCP and akermanite ceramic. Cell proliferation was tested by lactic acid production and MTT analysis, while osteogenic differentiation was assayed by alkaline phosphatase (ALP) expression and real-time polymerase chain reaction (PCR) analysis on markers of osteopontin (OPN), dentin matrix acidic phosphoprotein-1 (DMP-1), and osteocalcin (OCN), and further detected by enzyme-linked immunosorbent analysis (ELISA) analysis for OCN expression. Besides, the ions released from akermanite and their effect on hPDLCs was also measured by inductively coupled plasma atomic emission spectroscopy (ICP-AES), MTT analysis, ALP expression and real-time PCR analysis. hPDLCs attached well on both ceramics, but showed better spreading on akermanite. hPDLCs proliferated more rapidly on akermanite than β-TCP. Importantly, osteogenic differentiation of hPDLCs was enhanced on akermanite compared to β-TCP. Besides, Ca, Mg and Si ions were released from akermanite, while only Ca ions were released from β-TCP. Moreover, more pronounced proliferation and higher osteogenic gene expression for hPDLCs cultured with akermanite extract were detected as compared to cells cultured on akermanite. Therefore, akermanite ceramic showed an enhanced effect on proliferation and osteogenic differentiation of hPDLCs, which might be attributed to the release of ions containing Ca, Mg and Si from the material. It is suggested that akermanite ceramics may serve as a potential material for periodontal bone regeneration.

Journal ArticleDOI
TL;DR: 3D-fiber fleeces can be produced with controlled properties, being prerequisites for successful scaffolds in tissue engineering applications, and electrospinning parameters affect fiber diameter and alignment in an inverse relation.
Abstract: Degrapol ® and PLGA electrospun fiber fleeces were characterized with regard to fiber diameter, alignment, mechanical properties as well as scaffold porosity. The study showed that electrospinning parameters affect fiber diameter and alignment in an inverse relation: fiber diameter was increased with increased flow rate, with decrease in working distance and collector velocity, whereas fiber alignment increased with the working distance and collector velocity but decreased with increased flow rate. When Degrapol ® or PLGA-polymers were co-spun with increasing ratios of a water-soluble polymer that was subsequently removed; fibrous scaffolds with increased porosities were obtained. Mechanical properties correlated with fiber alignment rather than fiber diameter as aligned fiber scaffolds demonstrated strong mechanical anisotropy. For co-spun fibers the Young’s modulus correlated inversely with the amount of co-spun polymer. Cell proliferation was independent of the porosity of the scaffold, but different between the two polymers. Furthermore, fibrous scaffolds with different porosities were analyzed for cell infiltration suggesting that cell infiltration was enhanced with increased porosity and increasing time. These experiments indicate that 3D-fiber fleeces can be produced with controlled properties, being prerequisites for successful scaffolds in tissue engineering applications.

Journal ArticleDOI
TL;DR: In this article, the authors investigated cellular characteristics and the functional behaviour of mesenchymal stromal cells in response to mechanical loading, and they demonstrated a reduced expression of MSC surface markers CD73 (ecto-5'nucleotidase) and CD29 (integrin β1).
Abstract: The assumption that mesenchymal stromal cell (MSC)based-therapies are capable of augmenting physiological regeneration processes has fostered intensive basic and clinical research activities. However, to achieve sustained therapeutic success in vivo, not only the biological, but also the mechanical microenvironment of MSCs during these regeneration processes needs to be taken into account. This is especially important for e.g., bone fracture repair, since MSCs present at the fracture site undergo signifi cant biomechanical stimulation. This study has therefore investigated cellular characteristics and the functional behaviour of MSCs in response to mechanical loading. Our results demonstrated a reduced expression of MSC surface markers CD73 (ecto-5’-nucleotidase) and CD29 (integrin β1) after loading. On the functional level, loading led to a reduced migration of MSCs. Both effects persisted for a week after the removal of the loading stimulus. Specifi c inhibition of CD73/CD29 demonstrated their substrate dependent involvement in MSC migration after loading. These results were supported by scanning electron microscopy images and phalloidin staining of actin fi laments displaying less cell spreading, lamellipodia formation and actin accumulations. Moreover, focal adhesion kinase and Src-family kinases were identifi ed as candidate downstream targets of CD73/CD29 that might contribute to the mechanically induced decrease in MSC migration. These results suggest that MSC migration is controlled by CD73/CD29, which in turn are regulated by mechanical stimulation of cells. We therefore speculate that MSCs migrate into the fracture site, become mechanically entrapped, and thereby accumulate to fulfi l their regenerative functions.

Journal ArticleDOI
TL;DR: Gene expression profiling confirmed a key role for proliferation status for the bone forming ability suggesting that a rate limiting anabolism and open chromatin determined and predicted the therapeutic potency of culture-expanded MSC.
Abstract: Human mesenchymal stem cells (MSC) have attracted much attention for tissue regeneration including repair of non-healing bone defects. Heterogeneity of MSC cultures and considerable donor variability however, still preclude standardised production of MSC and point on functional deficits for some human MSC populations. We aimed to identify functional correlates of donor-dependency of bone formation in order to develop a potency assay predicting the therapeutic capacity of human MSC before clinical transplantation. MSC from 29 donors were characterised in vitro and results were correlated to bone formation potency in a beta-tricalcium-phosphate (β-TCP)-scaffold after subcutaneous implantation into immunocompromised mice. In contrast to osteogenic in vitro differentiation parameters, a doubling time below 43.23 hours allowed to predict ectopic bone formation at high sensitivity (81.8%) and specificity (100%). Enriched conditions adapted from embryonic stem cell expansion rescued bone formation of inferior MSC populations while growth arrest of potent MSC by mitomycin C abolished bone formation, establishing a causal relationship between neo-bone formation and growth. Gene expression profiling confirmed a key role for proliferation status for the bone forming ability suggesting that a rate limiting anabolism and open chromatin determined and predicted the therapeutic potency of culture-expanded MSC. Proliferation-based potency testing and switch to enriched expansion conditions may pave the way for standardised production of MSC for bone repair.

Journal ArticleDOI
TL;DR: Key parameters in an in vivo stem cell niche are introduced and recent work on DSCM for its rejuvenating or reprograming effect on various adult stem cells and chondrocytes is focused on.
Abstract: Cell-based therapy is a promising biological approach for the treatment of cartilage defects. Due to the small size of autologous cartilage samples available for cell transplantation in patients, cells need to be expanded to yield a sufficient cell number for cartilage repair. However, chondrocytes and adult stem cells tend to become replicatively senescent once they are expanded on conventional plastic flasks. Many studies demonstrate that the loss of cell properties is concomitant with the decreased cell proliferation capacity. This is a significant challenge for cartilage tissue engineering and regeneration. Despite much progress having been made in cell expansion, there are still concerns over expanded cell size and quality for cell transplantation applications. Recently, in vivo investigations in stem cell niches have suggested the importance of developing an in vitro stem cell microenvironment for cell expansion and tissue-specific differentiation. Our and other investigators' work indicates that a decellularized stem cell matrix (DSCM) may provide such an expansion system to yield large-quantity and high-quality cells for cartilage tissue engineering and regeneration. This review briefly introduces key parameters in an in vivo stem cell niche and focuses on our recent work on DSCM for its rejuvenating or reprograming effect on various adult stem cells and chondrocytes. Since research in DSCM is still in its infancy, we are only able to discuss some potential mechanisms of DSCM on cell proliferation and chondrogenic potential. Further investigations of the underlying mechanism and in vivo regeneration capacity will allow this approach to be used in clinics.

Journal ArticleDOI
TL;DR: The 3D co-culture system and the associated non-invasive analytical tools can be used as an advanced model to capture some aspects of the functional coupling of bone-like matrix deposition and resorption and could be exploited toward the engineering of multi-functional bone substitute implants.
Abstract: Three-dimensional (3D) organotypic culture models based on human cells may reduce the use of complex and costly animal models, while gaining clinical relevance. This study aimed at developing a 3D osteoblastic-osteoclastic-endothelial cell co-culture system, as an in vitro model to mimic the process of bone turnover. Osteoprogenitor and endothelial lineage cells were isolated from the stromal vascular fraction (SVF) of human adipose tissue, whereas CD14+ osteoclast progenitors were derived from human peripheral blood. Cells were co-cultured within 3D porous ceramic scaffolds using a perfusion-based bioreactor device, in the presence of typical osteoclastogenic factors. After 3 weeks, the scaffolds contained cells with endothelial (2.0±0.3%), pre/osteoclastic (14.0±1.4%) and mesenchymal/osteoblastic (44.0±8.4%) phenotypes, along with tartrate-resistant acid phosphatase-positive (TRAP+) osteoclastic cells in contact with deposited bone-like matrix. Supernatant analysis demonstrated sustained matrix deposition (by C-terminus procollagen-I propeptides), resorption (by N-terminus collagen-I telopeptides and phosphate levels) and osteoclastic activity (by TRAP-5b) only when SVF and CD14+ cells were co-cultured. Scanning electron microscopy and magnetic resonance imaging confirmed the pattern of matrix deposition and resorption. The effectiveness of Vitamin D in replacing osteoclastogenic factors indicated a functional osteoblast-osteoclast coupling in the system. The formation of human-origin bone-like tissue, blood vessels and osteoclasts upon ectopic implantation validated the functionality of the developed cell types. The 3D co-culture system and the associated non-invasive analytical tools can be used as an advanced model to capture some aspects of the functional coupling of bone-like matrix deposition and resorption and could be exploited toward the engineering of multi-functional bone substitute implants.

Journal ArticleDOI
TL;DR: In the present study, a cultivation regime was identified which firstly induces the differentiation of the human bone marrow stromal cells followed by the induction of osteoclastogenesis through the osteoblasts formed--without the external addition of the factors RANKL and M-CSF.
Abstract: The communication of bone-forming osteoblasts and bone-resorbing osteoclasts is a fundamental requirement for balanced bone remodelling. For biomaterial research, development of in vitro models is necessary to investigate this communication. In the present study human bone marrow stromal cells and human monocytes were cultivated in order to differentiate into osteoblasts and osteoclasts, respectively. Finally, a cultivation regime was identified which firstly induces the differentiation of the human bone marrow stromal cells followed by the induction of osteoclastogenesis through the osteoblasts formed--without the external addition of the factors RANKL and M-CSF. As a feedback on osteoblasts enhanced gene expression of BSP II was detected for modifications which facilitated the formation of large multinuclear osteoclasts. Phenotype characterization was performed by biochemical methods (DNA, LDH, ALP, TRAP 5b), gene expression analysis (ALP, BSP II, RANKL, IL-6, VTNR, CTSK, TRAP, OSCAR, CALCR) as well as light microscopy, confocal laser scanning microscopy, and scanning electron microscopy. After establishing this model on polystyrene, similar positive results were obtained for cultivation on a relevant bone substitution material--a composite xerogel of silica, collagen, and calcium phosphate.

Journal ArticleDOI
TL;DR: It is concluded that BMP-2 cDNA incorporated in alginate hydrogel appears to be a promising new strategy for minimal-invasive delivery of growth factors in bone regeneration.
Abstract: Bone regeneration is one of the major focus points in the field of regenerative medicine. A well-known stimulus of bone formation is bone morphogenetic protein-2 (BMP-2), which has already been extensively used in clinical applications. We investigated the possibility of achieving osteogenic differentiation both in vitro and in vivo as a result of prolonged presence of BMP-2 using plasmid DNA-based gene therapy. By delivering BMP-2 cDNA in an alginate hydrogel, a versatile formulation is developed. High transfection efficiencies of up to 95% were obtained in both human multipotent stromal cells (MSCs) and MG-63 cells using naked DNA in vitro. Over a period of 5 weeks, an increasing amount of biologically active BMP-2 was released from the cells and remained present in the gel. In vivo, transfected cells were found after both two and six weeks implantation in naked mice, even in groups without seeded cells, thus indicating in vivo transfection of endogenous cells. The protein levels were effective in inducing osteogenic differentiation in vitro, as seen by elevated alkaline phosphatase (ALP) production and in vivo, as demonstrated by the production of collagen I and osteocalcin in a mineralised alginate matrix. We conclude that BMP-2 cDNA incorporated in alginate hydrogel appears to be a promising new strategy for minimal-invasive delivery of growth factors in bone regeneration.

Journal ArticleDOI
TL;DR: MRI-based tracking of human Adipose Derived Stem Cells showed that hADSCs remain detectable until 28 d after implantation and could migrate from the scaffold and colonise the area around it, suggesting that this scaffold might behave as a cell carrier capable of both holding a cell fraction and delivering cells to the site of implantation.
Abstract: For bone tissue engineering, human Adipose Derived Stem Cells (hADSCs) are proposed to be associated with a scaffold for promoting bone regeneration. After implantation, cellularised scaffolds require a non-invasive method for monitoring their fate in vivo. The purpose of this study was to use Magnetic Resonance Imaging (MRI)-based tracking of these cells, labelled with magnetic agents for in vivo longitudinal assessment. hADSCs were isolated from adipose tissue and labelled with USPIO-rhodamine (Ultrasmall SuperParamagnetic Iron Oxide). USPIO internalisation, absence of toxicity towards hADSCs, and osteogenic differentiation of the labelled cells were evaluated in standard culture conditions. Labelled cells were then seeded within a 3D porous polysaccharide-based scaffold and imaged in vitro using fluorescence microscopy and MRI. Cellularised scaffolds were implanted subcutaneously in nude mice and MRI analyses were performed from 1 to 28 d after implantation. In vitro, no effect of USPIO labelling on cell viability and osteogenic differentiation was found. USPIO were efficiently internalised by hADSCs and generated a high T2* contrast. In vivo MRI revealed that hADSCs remain detectable until 28 d after implantation and could migrate from the scaffold and colonise the area around it. These data suggested that this scaffold might behave as a cell carrier capable of both holding a cell fraction and delivering cells to the site of implantation. In addition, the present findings evidenced that MRI is a reliable technique to validate cell-seeding procedures in 3D porous scaffolds, and to assess the fate of hADSCs transplanted in vivo.

Journal ArticleDOI
TL;DR: To establish a nascent vascular network in vitro within a CG scaffold as a first step towards that goal, Mesenchymal stem cells were chosen as primary vasculogenic candidate cells and a culture medium that promoted maximal network formation on Matrigel by these cells was selected.
Abstract: Collagen-glycosaminoglycan scaffolds that have been used clinically for skin regeneration have also shown significant promise for other applications in tissue engineering. However, regeneration of thicker tissues with the aid of implanted biomaterials is likely to depend on, or be accelerated by, the ability to establish rapid vascularisation of the implant. The present study aims to establish a nascent vascular network in vitro within a CG scaffold as a first step towards that goal. Mesenchymal stem cells (MSCs) were chosen as primary vasculogenic candidate cells and a culture medium that promoted maximal network formation on Matrigel by these cells was selected. MSCs seeded in the CG scaffold formed networks of cord-like structures after one to two weeks in the presence of the vasculogenic medium; similar structures were formed by aortic endothelial cells (ECs) cultured for comparison. Gene expression analysis suggested that the MSCs began to adopt an endothelial phenotype, with RNA for PECAM and VCAM rising while that for alpha-smooth muscle actin fell. However there was no increase in Tie-2 and vWF expression. Addition of smooth muscle cells (SMCs) as a potential perivascular stabilising component did not have a noticeable effect on MSC-derived networks, although it enhanced EC-derived structures.

Journal ArticleDOI
TL;DR: This study showed that RGD peptide sequences promoted cell spreading in OPF hydrogels and hence play a crucial role in cell survival during the early stage of culture, whereas CaP nanoparticles significantly enhanced cell-mediated hydrogel mineralization.
Abstract: The ultimate goal of this work was to develop a biocompatible and biomimetic in situ crosslinkable hydrogel scaffold with an instructive capacity for bone regenerative treatment. To this end, synthetic hydrogels were functionalized with two key components of the extracellular matrix of native bone tissue, i.e. the three-amino acid peptide sequence RGD (which is the principal integrin-binding domain responsible for cell adhesion and survival of anchorage-dependent cells) and calcium phosphate (CaP) nanoparticles in the form of hydroxyapatite (which are similar to the inorganic phase of bone tissue). Rat bone marrow osteoblast-like cells (OBLCs) were encapsulated in four different biomaterials (plain oligo(poly(ethylene glycol) fumarate) (OPF), RGD-modified OPF, OPF enriched with CaP nanoparticles and RGD-modified OPF enriched with CaP nanoparticles) and cell survival, cell spreading, proliferation and mineralized matrix formation were determined via cell viability assay, histology and biochemical analysis for alkaline phosphatase activity and calcium. This study showed that RGD peptide sequences promoted cell spreading in OPF hydrogels and hence play a crucial role in cell survival during the early stage of culture, whereas CaP nanoparticles significantly enhanced cell-mediated hydrogel mineralization. Although cell spreading and proliferation activity were inhibited, the combined effect of RGD peptide sequences and CaP nanoparticles within OPF hydrogel systems elicited a better biological response than that of the individual components. Specifically, both a sustained cell viability and mineralized matrix production mediated by encapsulated OBLCs were observed within these novel biomimetic composite systems.

Journal ArticleDOI
TL;DR: It is suggested that bone-derived scaffolds have good biocompatibility and show osteoinductive properties by increasing the osteogenic lineage commitment of BMSCs and enhancing osteogenic gene expressions, and ERK1/2 pathway plays an important role in microvibration-induced osteogenesis in BMSC cellular scaffolds.
Abstract: Although in vivo studies have shown that low-magnitude, high-frequency (LMHF) vibration (LM: < 1 ×g; HF: 20-90 Hz) exhibits anabolic effects on skeletal homeostasis, the underlying cellular/molecular regulation involved in bone adaptation to LMHF vibration is little known. In this report, we tested the effects of microvibration (magnitude: 0.3 ×g, frequency: 40 Hz, amplitude: ± 50 μm, 30 min/12 h) on proliferation and osteodifferentiation of bone marrow-derived mesenchymal stromal cells (BMSCs) seeded on human bone-derived scaffolds. The scaffolds were prepared by partial demineralisation and deproteinisation. BMSCs were allowed to attach to the scaffolds for 3 days. Morphological study showed that spindle-shaped BMSCs almost completely covered the surface of bone-derived scaffold and these cells expressed higher ALP activity than those cultured on plates. After microvibration treatment, BMSC proliferation was decreased on day 7 and 10; however, numbers of genes and proteins expressed during osteogenesis, including Cbfa1, ALP, collagen I and osteocalcin were greatly increased. ERK1/2 activation was involved in microvibration-induced BMSC osteogenesis. Taken together, this study suggests that bone-derived scaffolds have good biocompatibility and show osteoinductive properties. By increasing the osteogenic lineage commitment of BMSCs and enhancing osteogenic gene expressions, microvibration promotes BMSC differentiation and increase bone formation of BMSCs seeded on bone-derived scaffolds. Moreover, ERK1/2 pathway plays an important role in microvibration-induced osteogenesis in BMSC cellular scaffolds.

Journal ArticleDOI
TL;DR: The aim of this study was to investigate the impact of alginate type and of two bead diameters on mass transfers within beads and on the biological functions of encapsulated C3A cells.
Abstract: Liver-assist devices have been developed in the last few decades to support patients with liver failure on the road to recovery or transplantation. Fluidised bed bio-artificial livers--where liver cells are encapsulated within alginate beads--appear to be a valuable alternative to hollow fibre devices for improving mass transfers and enhancing treatment efficacy. This approach nevertheless deserves optimization in terms of bead production. The aim of this study was to investigate the impact of alginate type and of two bead diameters (1000 µm and 600 µm) on mass transfers within beads and on the biological functions of encapsulated C3A cells. After assessing the effect of the encapsulation process on bead quality, we investigated cell viability and metabolic activities (ammonia, albumin, alpha-fetoprotein synthesis and glucose consumption). They were successfully maintained over 48 h within fluidised bed bioreactors, independently of alginate type and bead diameter. Mass transfers were not significantly influenced by the latter parameters. Finally, suggestions are made for improving the entrapment process as a means of enhancing the treatment efficiency of the fluidised bed bioartificial liver.

Journal ArticleDOI
TL;DR: The release of implanted SDF-1 is supported as a promising method for enhancing progenitor cell responses and restoring perfusion to ischaemic tissues via neovascularisation.
Abstract: Although many regenerative cell therapies are being developed to replace or regenerate ischaemic muscle, the lack of vasculature and poor persistence of the therapeutic cells represent major limiting factors to successful tissue restoration. In response to ischaemia, stromal cell-derived factor-1 (SDF-1) is up-regulated by the affected tissue to stimulate stem cell-mediated regenerative responses. Therefore, we encapsulated SDF-1 into alginate microspheres and further incorporated these into an injectable collagen-based matrix in order to improve local delivery. Microsphere-matrix impregnation reduced the time for matrix thermogelation, and also increased the viscosity reached. This double-incorporation prolonged the release of SDF-1, which maintained adhesive and migratory bioactivity, attributed to chemotaxis in response to SDF-1. In vivo, treatment of ischaemic hindlimb muscle with microsphere-matrix led to increased mobilisation of bone marrow-derived progenitor cells, and also improved recruitment of angiogenic cells expressing the SDF-1 receptor (CXCR4) from bone marrow and local tissues. Both matrix and SDF-1-releasing matrix were successful at restoring perfusion, but SDF-1 treatment appeared to play an earlier role, as evidenced by arterioles that are phenotypically older and by increased angiogenic cytokine production, stimulating the generation of a qualitative microenvironment for a rapid and therefore more efficient regeneration. These results support the release of implanted SDF-1 as a promising method for enhancing progenitor cell responses and restoring perfusion to ischaemic tissues via neovascularisation.

Journal ArticleDOI
TL;DR: Results demonstrate the cytocompatibility of 58S-Zn0.4 bioglass and its capability to promote osteoblast differentiation.
Abstract: Bioactive glasses synthesized by the sol-gel technique possess many of the qualities associated with an ideal scaffold material for a bone graft substitute. In view of the potential clinical applications, we performed a detailed in vitro study of the biological reactivity of synthesized 58S bioactive glass containing-zinc, in terms of osteoblast morphology, proliferation, and deposition of a mineralized extracellular matrix (ECM). Human Sarcoma Osteoblast (SAOS-2) cells were used to i) assess cytotoxicity by lactate dehydrogenase (LDH) release and ii) evaluate the deposition of a calcified extracellular matrix by ELISA assay and quantitative RT-PCR (qRT-PCR). In comparison with pure silica and 58S, the 58S-Zn0.4 bioglass showed a significant increase in cellular proliferation and deposition of ECM components such as decorin, fibronectin, osteocalcin, osteonectin, osteopontin, type-I and -III collagens. Calcium deposition was significantly higher than on pure silica and 58S samples. Also Alkaline phosphatase (ALP) activity and its protein content was higher with respect to pure silica and 58S. qRT-PCR analysis revealed the up-regulation of type-I collagen, bone sialoprotein and osteopontin genes. All together these results demonstrate the cytocompatibility of 58S-Zn0.4 bioglass and its capability to promote osteoblast differentiation.

Journal ArticleDOI
TL;DR: Findings might indicate that, combined with findings of previous in vivo studies demonstrating that alkali-treated SBA Ti surfaces perform better in terms of osseointegration, a continuous and structured layer of blood components on the blood-facing surface supports later tissue integration of an endosseous implant.
Abstract: Titanium implants are most commonly used for bone augmentation and replacement due to their favorable osseointegration properties. Here, hyperhydrophilic sand-blasted and acid-etched (SBA) titanium surfaces were produced by alkali treatment and their responses to partially heparinized whole human blood were analyzed. Blood clot formation, platelet activation and activation of the complement system was analyzed revealing that exposure time between blood and the material surface is crucial as increasing exposure time results in higher amount of activated platelets, more blood clots formed and stronger complement activation. In contrast, the number of macrophages/monocytes found on alkali-treated surfaces was signifi cantly reduced as compared to untreated SBA Ti surfaces. Interestingly, when comparing untreated to modifi ed SBA Ti surfaces very different blood clots formed on their surfaces. On untreated Ti surfaces blood clots remain thin (below 15 mm), patchy and non-structured lacking large fi brin fi ber networks whereas blood clots on differentiated surfaces assemble in an organized and layered architecture of more than 30 mm thickness. Close to the material surface most nucleated cells adhere, above large amounts of non-nucleated platelets remain entrapped within a dense fi brin fi ber network providing a continuous cover of the entire surface. These fi ndings might indicate that, combined with fi ndings of previous in vivo studies demonstrating that alkali-treated SBA Ti surfaces perform better in terms of osseointegration, a continuous and structured layer of blood components on the blood-facing surface supports later tissue integration of an endosseous implant.

Journal ArticleDOI
TL;DR: A finite element model of a cell, with a tensegrity structure to model the cytoskeleton of actin filaments and microtubules, is used to explore the way cells sense the stiffness of the substrate and thereby adapt to it and suggests that F-actin reorganisation observed in mesenchymal stem cells (MSCs) in response to change of matrix elasticity is a structural-remodelling process that shifts the sensitivity peak towards the new value of Matrix elasticity.
Abstract: A eukaryotic cell attaches and spreads on substrates, whether it is the extracellular matrix naturally produced by the cell itself, or artificial materials, such as tissue-engineered scaffolds. Attachment and spreading require the cell to apply forces in the nN range to the substrate via adhesion sites, and these forces are balanced by the elastic response of the substrate. This mechanical interaction is one determinant of cell morphology and, ultimately, cell phenotype. In this paper we use a finite element model of a cell, with a tensegrity structure to model the cytoskeleton of actin filaments and microtubules, to explore the way cells sense the stiffness of the substrate and thereby adapt to it. To support the computational results, an analytical 1D model is developed for comparison. We find that (i) the tensegrity hypothesis of the cytoskeleton is sufficient to explain the matrix-elasticity sensing, (ii) cell sensitivity is not constant but has a bell-shaped distribution over the physiological matrix-elasticity range, and (iii) the position of the sensitivity peak over the matrix-elasticity range depends on the cytoskeletal structure and in particular on the F-actin organisation. Our model suggests that F-actin reorganisation observed in mesenchymal stem cells (MSCs) in response to change of matrix elasticity is a structural-remodelling process that shifts the sensitivity peak towards the new value of matrix elasticity. This finding discloses a potential regulatory role of scaffold stiffness for cell differentiation.

Journal ArticleDOI
TL;DR: It is demonstrated that COX-2 activity is a novel factor partaking in chondrocyte hypertrophy in the context of endochondral ossification and these observations provide a novel etiological perspective on the adverse effects of NSAIDs on bone fracture healing and have important implications for the use ofNSAIDs during endochondrals skeletal development.
Abstract: Skeletogenesis and bone fracture healing involve endochondral ossification, a process during which cartilaginous primordia are gradually replaced by bone tissue. In line with a role for cyclooxygenase-2 (COX-2) in the endochondral ossification process, non-steroidal anti-inflammatory drugs (NSAIDs) were reported to negatively affect bone fracture healing due to impaired osteogenesis. However, a role for COX-2 activity in the chondrogenic phase of endochondral ossification has not been addressed before. We show that COX-2 activity fulfils an important regulatory function in chondrocyte hypertrophic differentiation. Our data reveal essential cross-talk between COX-2 and bone morphogenic protein-2 (BMP-2) during chondrocyte hypertrophic differentiation. BMP-2 mediated chondrocyte hypertrophy is associated with increased COX-2 expression and pharmacological inhibition of COX-2 activity by NSAIDs (e.g., Celecoxib) decreases hypertrophic differentiation in various chondrogenic models in vitro and in vivo, while leaving early chondrogenic development unaltered. Our findings demonstrate that COX-2 activity is a novel factor partaking in chondrocyte hypertrophy in the context of endochondral ossification and these observations provide a novel etiological perspective on the adverse effects of NSAIDs on bone fracture healing and have important implications for the use of NSAIDs during endochondral skeletal development.

Journal ArticleDOI
TL;DR: The intrinsic osteogenic activity of poloxamines offers novel perspectives for bone regeneration using minimally invasive procedures (i.e., injectable scaffolds) and overcoming the safety and the cost/effectiveness concerns associated with large scale clinical use of recombinant growth factors.
Abstract: In situ gelling solutions for minimally invasive local application of bone growth factors are attracting increasing attention as efficient and patient-friendly alternative to bone grafts and solid scaffolds for repairing bone defects. Poloxamines, i.e., X-shaped poly(ethylene oxide)-poly(propylene oxide) block copolymers with an ethylenediamine core (Tetronic ® ), were evaluated both as an active osteogenic component and as a vehicle for rhBMP-2 injectable implants. After cytotoxicity screening of various poloxamine varieties, Tetronic 908, 1107, 1301 and 1307 solutions were chosen as the most cytocompatible and their sol-to-gel transitions were rheologically characterized. Viscoelastic gels, formed at 37 oC, sustained protein release under physiological-like conditions. Formulations of rhBMP-2 led to differentiation of mesenchymal stem cells to osteoblasts, quantifi ed as alkaline phosphatase activity with a maximum at day 7, and to mineralized nodules. Interestingly, poloxamine solely gels led to an initial proliferation of the mesenchymal stem cells (fi rst week), followed by differentiation to osteoblasts (second to third week). Histochemical analysis revealed that Tetronic 908 is only osteoinductive; Tetronic 1107 is mostly osteoinductive, although its use leads to a minor differentiation to adipocytes; Tetronic 1307, solely or loaded with rhBMP-2, causes differentiation of both osteoblasts and adipocytes. Enhanced expression levels of CBFA-1 and collagen type I were observed for Tetronic 908, 1107 and 1307, both solely and combined with rhBMP-2. The intrinsic osteogenic activity of poloxamines (not observed for Pluronic F127) offers novel perspectives for bone regeneration using minimally invasive procedures (i.e., injectable scaffolds) and overcoming the safety and the cost/ effectiveness concerns associated with large scale clinical use of recombinant growth factors.