87
Views
11
CrossRef citations to date
0
Altmetric
Review

Stem cell-mediated osteogenesis: therapeutic potential for bone tissue engineering

, , &
Pages 47-57 | Published online: 09 Mar 2012

Abstract

Intervertebral disc degeneration often requires bony spinal fusion for long-term relief. Current arthrodesis procedures use bone grafts from autogenous bone, allogenic backed bone, or synthetic materials. Autogenous bone grafts can result in donor site morbidity and pain at the donor site, while allogenic backed bone and synthetic materials have variable effectiveness. Given these limitations, researchers have focused on new treatments that will allow for safe and successful bone repair and regeneration. Mesenchymal stem cells have received attention for their ability to differentiate into osteoblasts, cells that synthesize new bone. With the recent advances in scaffold and biomaterial technology as well as stem cell manipulation and transplantation, stem cells and their scaffolds are uniquely positioned to bring about significant improvements in the treatment and outcomes of spinal fusion and other injuries.

Introduction

Intervertebral disc degeneration remains a pervasive and intractable disease arising from a combination of aging and stress on the bony and cartilaginous elements of the spinal column.Citation1 Some patients will have disease progression that, despite conservative and minimally invasive therapies, requires arthrodesis (fusion) of particular spinal segments as the direct treatment of pathological pain-generating motion, or in stabilization after the decompression of symptomatic neural elements. As spinal instrumentation has evolved over the last decade, the role that surgery plays in the treatment of degenerative spinal pathology has increased. Critical to achieving a durable repair of the spine, arthrodesis depends heavily on the quality and quantity of autograft bone or bone substitutes. The ideal bone graft substitute should provide the benefits afforded by autograft (successful physiological fusion), thus allowing surgical intervention to be successful without the risk of donor site morbidity from autograft harvesting.

Regenerative medicine attempts to repair, regenerate, or replace tissues damaged by factors such as injury or disease.Citation2 This growing field of medicine brings the promise of stem cells in the improvement of fusion options. In this pursuit, advancements in osteogenesis through the use of mesenchymal stem cells (MSCs) may provide the fundamental components that comprise autograft: osteoconductive scaffolds, osteoinductive signals, and osteogenic cells. Given the recent advances in minimally invasive spine surgery and the developing body of work on stem cell manipulation and transplantation, stem cells are uniquely poised to bring about large-scale improvements in treatment and outcomes of spinal fusion and other injuries. However, there are fundamental unresolved issues in the therapeutic use of stems cells; namely, the percentage and quantity of cells that are phenotypically MSCs remain to be discovered. These cells are most likely the “engine” that makes the bone heal. Essential to providing evidence toward the efficacy of MSC-mediated osteogenesis and bone healing is the evaluation of the gene expression profile and protein determination of MSCs and their cellular osteogenic progeny.

Natural bone healing cascade

Osteogenesis occurs throughout life, is involved in bone remodeling in adults, and is activated in injuries including, but not limited to, fractures of the bone.Citation3,Citation4 Bone formation depends on the cooperation of four factors: (1) specific cell types such as MSCs, osteoblasts, and osteoclasts; (2) the scaffold (hydroxyapatite, extracellular matrix molecules); (3) expression of soluble molecules (cytokines, growth factors, hormones, ions, vitamins);Citation3,Citation5,Citation6 and (4) various mechanical stimuli.Citation3,Citation7 Normal development of the skeleton during embryogenesis occurs by intramembranous and endochondral ossification.Citation3,Citation8

Osteoblasts secrete the growth factors and deposit the matrix necessary for osteogenesis, while osteoclasts function in bone remodeling.Citation9 Skeletal stability is reached by focal osteoclast-mediated degradation and osteoblast-mediated formation, while the overall architecture and anatomy of bones is maintained. Thus, the amount of new bone equals the amount resorbed, with no net change in the volume of bone.Citation10 As a consequence, new calcium phosphate crystals replace the less soluble and mature mineral crystalline component of bone.Citation10 Therefore, the molecular remodeling and composition of the adult skeleton is a dynamic process that changes as a new bone fills each resorbed site.Citation10 Moreover, the renewal of bone matrix is central for the essential role that bone has in mineral exchange and homeostasis.Citation10

The MSC component

In the adult stages, MSCs contribute to the maintenance of various tissues, particularly those of the bone.Citation3 MSCs can be isolated from bone marrow and adipose tissues in adult stages and from placenta and umbilical cord blood.Citation3,Citation11 MSCs can be induced in vitro and in vivo to differentiate into various mesenchymal tissues such as bone, cartilage, muscle, tendon, adipose tissue, and hematopoiesis-supporting stroma. 3 MSCs are selected by their capacity for adherence to plastic culture flasks and then expand via colony-forming unit fibroblasts after several weeks in vitro.Citation3,Citation11 However, this procedure does not permit characterization of the native form, whereas extensive work has described the in vitro-derived phenotype and multipotentiality of cultured MSCs.Citation3 Recent studies have clearly demonstrated that native-form MSCs are phenotypically and functionally different from cultured MSCs and similar to perivascular cells.Citation3 Under in vitro conditions, cultured MSCs can be characterized as nonhematopoietic cells (CD14-, CD34-, and CD45-) and express several surface markers such as CD44, CD106, CD146, and CD166.Citation3 Cultured MSCs are largely used in experimental bone reconstruction in vivo and in vitro.Citation3,Citation11,Citation12 The cells are generally cultured in basal medium such as Dulbecco’s modified Eagle’s medium with 10% fetal bovine serum.Citation11,Citation13 Researchers believe that serum components in fetal bovine serum play crucial roles in the attachment and proliferation of MSCs.Citation14 Osteogenic activation requires the presence of β-glycerol-phosphate, L-ascorbic acid-2 phosphate, dexamethasone, and fetal bovine serum.Citation13,Citation15 A study by Jaiswal et al found that optimal osteogenic differentiation was achieved with Dulbecco’s modif ied Eagle’s medium-base medium plus 100 nM dexamethasone, 0.05 mM L-ascorbic acid-2 phosphate, and 10 mM β-glycerol-phosphate.Citation15 Although few human clinical studies have investigated improvements in either bone defects or osteogenesis imperfecta disease, clinical studies, with related controls, are needed to confirm the potential of cultured MSCs, regardless of origin (allogeneic or autologous), for use in bone tissue engineering in the clinic.Citation3

MSC derivation

The limiting factor in exploiting stem cells for therapeutic use is obtaining well characterized cells for transplantation.Citation1 Several researchers have demonstrated that colonies derived from colony-forming unit fibroblast assays are heterogeneous in appearance and size as well as differentiation potential.Citation16Citation18 Studying the activity of MSCs is made difficult by the lack of unique identifying markers, and therefore an inconsistency in molecular expression.Citation2,Citation19Citation21 Consequently, directing the appropriate differentiation of both native-form MSCs and cultured MSCs is a complex molecular and cellular riddle that is dependent upon not only the inherent properties of cells but also the environment in which they are cultured.Citation1 The soluble growth factors, transforming growth factor beta and bone morphogenetic proteins (BMPs), are the necessary components of in vitro culture media used to induce chondrogenic and osteogenic differentiation of MSCs.Citation1 In fact, careful use of soluble factors in media can lead to chondrogenesis and osteogenesis with a genetic profile similar to intervertebral disc tissue, rather than articular cartilage.Citation1,Citation22

Another method exploits the MSCs’ microenvironment and triggers differentiation by coculturing with different cell populations to take advantage of cell–cell contact and activation.Citation1 Utilizing the autocrine and paracrine factors secreted by one cell type leads to the activation of cell surface receptors on MSCs.Citation1 Experiments culturing human neural progenitor cells and MSCs found that differentiation was reliant on cell–cell contact by looking at gene expression of Sox9, type II collagen, and aggrecan.Citation1,Citation23

The three-dimensional properties of the culture system have also been shown to exert substantial influence on the process of cell fate determination.Citation1 MSCs are pelleted down into a dense micromass in combination with transforming growth factor, BMP, and insulin-like growth factor growth factors,Citation9 before addition of soluble factors to recreate the in vivo state that leads to cartilage formation.Citation1 This structure helps direct the chondrogenic cascade of MSC differentiation from micromass into cartilage.Citation1 Mesenchymal condensation allows for extracellular signaling molecules, such as Wnt glycoproteins and N-cadherin, to form cadherin and connexin adhesion complexes for the beginning stages of extracellular matrix formation.Citation1 Cartilage then begins to form on this three-dimensional scaffold.Citation1 Plating density of MSCs prior to soluble factor addition also influences the efficiency of differentiation.Citation1 This is because plating density can change the cell morphology; specifically, wider spindle-shaped cells correspond with denser MSCs plating.Citation1 Wider cells also have an increased propensity to differentiate after exposure to soluble factors in vitro.Citation1,Citation24 In this way, employing density-dependent culturing techniques can produce cartilage formation from MSCs in vitro, and increase the efficiency of MSC differentiation.Citation1

It has been suggested that MSCs can be isolated from other tissues, but their similarity to those isolated from bone marrow in terms of potential for osteogenesis and chondrogenesis is not fully understood. Controversial cells from human umbilical cord blood and human derived-placenta cells have been extensively studied for MSC potential. An early study identified preterm cord blood as being rich in mesenchymal progenitor cells that displayed a fibroblast-like morphology and expressed several mesenchymal progenitor cell-related surface antigens.Citation25 Another group later isolated human MSC-like adherent cells from human umbilical cord blood that could differentiate into a variety of mesenchymal lineage cells, including osteoblasts and chondrocytes, under the appropriate conditions.Citation26 A related study identified MSC-like cells in human placenta that possessed multilineage differentiation potential similar to MSCs under specific conditions.Citation27 The less-controversial adipose tissue-derived MSCs have been previously shown to differentiate into bone, cartilage, fat, or muscle but have been found to have an inferior potential for both osteogenesis and chondrogenesis compared with bone marrow-derived MSCs.Citation28

Molecular mechanisms underlying bone healing

Osteoblasts are cells of mesenchymal origin that secrete bone-matrix proteins and promote mineralization.Citation29Citation31 The proliferation and differentiation of osteoblasts are under the control of a number of soluble factors and transcription factors such as runt-related transcription factor 2 (Runx2) and osterix (also known as Sp7).Citation29,Citation32,Citation33 Differentiated osteoblasts embedded in the bone matrix are called osteocytes and might have a specific, but currently unclear, role in mechanotransduction.Citation29,Citation30 Osteoclasts are cells of hematopoietic origin that decalcify and degrade the bone matrix by acid decalcification and proteolytic degradation, respectively.Citation29,Citation34 They are large, multinucleated cells formed by the fusion of precursor cells of the monocyte–macrophage lineage.Citation29

In vitro osteoclast differentiation is supported by mesenchymal cells (bone marrow stromal cells or osteoblasts) through cell–cell contact,Citation29,Citation35 although there has been little in vivo information about osteoclastogenesis-supporting cells.Citation29 Osteoclastogenic signals are mediated by receptor activator of nuclear kappa-B ligand (RANKL) and its costimulatory signals, in addition to macrophage colony-stimulating factor 5 ().Citation29,Citation36Citation38 The congenital lack of osteoclasts leads to osteopetrosis, which is characterized by a high bone mass and a defect in bone marrow formation.Citation29 Naturally occurring mutant mice or genetically modified mice with osteopetrosis have provided insights into the molecular mechanism of osteoclast differentiation and function.Citation29,Citation39 Macrophage colony-stimulating factor and the transcription factor PU.1 are crucial for the proliferation and survival of osteoclast precursor cells; transcription factors such as cFOS, microphthalmia-associated transcription factor, and nuclear factor kappa-B have been shown to be essential for osteoclast differentiation; and factors such as cSRC, VAV3, β3-integrin, chloride-channel family member ClC7, vacuolar adenosine triphosphatase, and cathepsin K are crucial for osteoclast function ().Citation29,Citation39

Figure 1 Osteoclast differentiation is induced by macrophage colony-stimulating factor (M-CSF), receptor activator of nuclear factor-κB [NF-κB] ligand (RANKL) and its costimulatory factor, immunoglobulin (Ig)-like receptor. (A) Precursor-cell stage. The binding of M-CSF to its receptor, cFMS, activates the proliferation and survival of osteoclast precursor cells of the monocyte–macrophage lineage that express receptor activator of nuclear factor-κB (RANK). The costimulatory receptors might be stimulated from early stages, although ligands of costimulatory receptors have yet to be identified. (B) Proximal RANK signals. RANKL binding to RANK results in the recruitment of tumor-necrosis-factor-receptor-associated factor 6 (TRAF6). At the same time, RANK activation results in the phosphorylation of the immunoreceptor tyrosine-based activation motif (ITAM) in DAP12 and Fc-receptor common γ-subunit (FcRγ), both of which are adaptor proteins associating with distinct Ig-like receptors. Ig-like receptor signals are called costimulatory signals for RANK. (C) Initial induction of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1). NFATc1, a key transcription factor for osteoclastogenesis, is initially induced by TRAF6-activated NF-κB and NFATc2 that is present in the cell before RANKL stimulation. Phosphorylation of the ITAM in DAP12 (or FcRγ) results in the recruitment of spleen tyrosine kinases (SYKs) that activate calcium signaling through phospholipase Cγ (PLCγ). (D) Auto amplification of NFATc1. Calcium signal–mediated persistent activation of NFATc1, as well as cooperation with activator protein 1 (AP1), is a prerequisite for the robust induction of NFATc1. AP1 activation is mediated by the induction and activation of cFOS by calcium/calmodulin-dependent protein kinase type IV (CaMKIV)-stimulated cyclic adenosine monophosphate responsive-element-binding protein (CREB) and cFMS. The NFATc1 promoter is epigenetically activated through histone acetylation and NFATc1 binds to an NFAT-binding site of its own promoter. (E) In the nucleus, NFATc1 works together with other transcription factors, such as AP1, PU.1, microphthalmia-associated transcription factor (MITF) and CREB, to induce various osteoclast-specific genes, including tartrate-resistant acid phosphatase, cathepsin K, and calcitonin receptor.

Note: Reprinted by permission from Macmillan Publishers Ltd: Nature Review Immunology, H Takayanagi, Osteoimmunology: shared mechanisms and crosstalk between the immune and bone systems, 7(4):292–304, copyright 2007.Citation29
Figure 1 Osteoclast differentiation is induced by macrophage colony-stimulating factor (M-CSF), receptor activator of nuclear factor-κB [NF-κB] ligand (RANKL) and its costimulatory factor, immunoglobulin (Ig)-like receptor. (A) Precursor-cell stage. The binding of M-CSF to its receptor, cFMS, activates the proliferation and survival of osteoclast precursor cells of the monocyte–macrophage lineage that express receptor activator of nuclear factor-κB (RANK). The costimulatory receptors might be stimulated from early stages, although ligands of costimulatory receptors have yet to be identified. (B) Proximal RANK signals. RANKL binding to RANK results in the recruitment of tumor-necrosis-factor-receptor-associated factor 6 (TRAF6). At the same time, RANK activation results in the phosphorylation of the immunoreceptor tyrosine-based activation motif (ITAM) in DAP12 and Fc-receptor common γ-subunit (FcRγ), both of which are adaptor proteins associating with distinct Ig-like receptors. Ig-like receptor signals are called costimulatory signals for RANK. (C) Initial induction of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1). NFATc1, a key transcription factor for osteoclastogenesis, is initially induced by TRAF6-activated NF-κB and NFATc2 that is present in the cell before RANKL stimulation. Phosphorylation of the ITAM in DAP12 (or FcRγ) results in the recruitment of spleen tyrosine kinases (SYKs) that activate calcium signaling through phospholipase Cγ (PLCγ). (D) Auto amplification of NFATc1. Calcium signal–mediated persistent activation of NFATc1, as well as cooperation with activator protein 1 (AP1), is a prerequisite for the robust induction of NFATc1. AP1 activation is mediated by the induction and activation of cFOS by calcium/calmodulin-dependent protein kinase type IV (CaMKIV)-stimulated cyclic adenosine monophosphate responsive-element-binding protein (CREB) and cFMS. The NFATc1 promoter is epigenetically activated through histone acetylation and NFATc1 binds to an NFAT-binding site of its own promoter. (E) In the nucleus, NFATc1 works together with other transcription factors, such as AP1, PU.1, microphthalmia-associated transcription factor (MITF) and CREB, to induce various osteoclast-specific genes, including tartrate-resistant acid phosphatase, cathepsin K, and calcitonin receptor.

Figure 2 The immune and skeletal systems share cytokines, receptors, signaling molecules, and transcription factors, all of which cooperatively regulate osteoclasts and osteoblasts as well as their interactions. Osteoblasts regulate osteoclastogenesis through receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-receptor activator of nuclear factor-κB (RANK) (and its decoy receptor osteoprotegerin [OPG]) interactions, macrophage colony-stimulating factor (M-CSF)–cFMS interactions and immunoglobulin (Ig)-like receptors associated with immunoreceptor tyrosine-based activation motif-harboring adaptor molecules (such as DAP12 and Fc-receptor common γ-subunit [FcRγ], the ligands of which are not well characterized). Although not depicted, semaphorin 6D, and its receptor plexin A1, and ephrin receptor B4 and ephrin B2 were newly identified as mediators of osteoblast–osteoclast interactions. There are extensive signaling pathways in osteoclasts. RANK and Ig-like receptors stimulate downstream signaling cascades (such as tumor necrosis factor [TNF] receptor-associated factor 6 [TRAF6], NF-κB, mitogen-activated protein kinases [MAPKs], activator protein 1 [AP1], calcineurin, and nuclear factor of activated T cells cytoplasmic 1 [NFATc1]), which are influenced by a number of immunoregulatory molecules including CD40 ligand (CD40L), interleukin-1 (IL-1), interferon-β (IFNβ), IFNγ, TNF, and lipopolysaccharide (LPS). Dendritic-cell-specific transmembrane protein (DC-STAMP) and ATP6V0D2 are necessary for the fusion of osteoclast precursor cells. Phosphoinositide 3-kinase (PI3K)-AKT and growth-factor-receptor-bound protein 2–extracellular-signal- regulated kinase (GRB2–ERK) pathways are important for the proliferation and survival of the osteoclast lineage, whereas VAV3, cSRC, and Casitas B-lineage lymphoma (cCBL) are included in the molecules required for cytoskeletal reorganization and bone-resorbing osteoclasts. Osteoclast activity is dependent on acidifying proton pump ATP6I and chloride channel 7 (ClC7), as well as matrix-degrading enzymes such as cathepsin K and matrix metalloproteinase 9 (MMP9). The following molecules are known to be involved in both the bone system and the immune system: NF-κB, RANKL, RANK, OPG, cFMS, M-CSF, Ig-like receptors, FcRγ, DAP12, TRAF6, MAPKs, AP1, calcineurin, NFATc1, CD40L, IL-1, IFNγ, IFNβ, TNF, LPS, DC-STAMP, PI3K, AKT, ERK, VAV3, cSRC, and cCBL.

Note: Reprinted by permission from Macmillan Publishers Ltd: Nature Review Immunology, H Takayanagi, Osteoimmunology: shared mechanisms and crosstalk between the immune and bone systems, 7(4):292–304, copyright 2007.Citation29
Abbreviations: CaMKIV, calcium/calmodulin-dependent protein kinase type IV; CREB, cyclic adenosine monophosphate responsive-element-binding protein; PLC, phospholipase C; MITF, microphthalmia-associated transcription factor; IKK, inhibitor of NF-κB (IκB) kinase; SYK, spleen tyrosine kinase.
Figure 2 The immune and skeletal systems share cytokines, receptors, signaling molecules, and transcription factors, all of which cooperatively regulate osteoclasts and osteoblasts as well as their interactions. Osteoblasts regulate osteoclastogenesis through receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-receptor activator of nuclear factor-κB (RANK) (and its decoy receptor osteoprotegerin [OPG]) interactions, macrophage colony-stimulating factor (M-CSF)–cFMS interactions and immunoglobulin (Ig)-like receptors associated with immunoreceptor tyrosine-based activation motif-harboring adaptor molecules (such as DAP12 and Fc-receptor common γ-subunit [FcRγ], the ligands of which are not well characterized). Although not depicted, semaphorin 6D, and its receptor plexin A1, and ephrin receptor B4 and ephrin B2 were newly identified as mediators of osteoblast–osteoclast interactions. There are extensive signaling pathways in osteoclasts. RANK and Ig-like receptors stimulate downstream signaling cascades (such as tumor necrosis factor [TNF] receptor-associated factor 6 [TRAF6], NF-κB, mitogen-activated protein kinases [MAPKs], activator protein 1 [AP1], calcineurin, and nuclear factor of activated T cells cytoplasmic 1 [NFATc1]), which are influenced by a number of immunoregulatory molecules including CD40 ligand (CD40L), interleukin-1 (IL-1), interferon-β (IFNβ), IFNγ, TNF, and lipopolysaccharide (LPS). Dendritic-cell-specific transmembrane protein (DC-STAMP) and ATP6V0D2 are necessary for the fusion of osteoclast precursor cells. Phosphoinositide 3-kinase (PI3K)-AKT and growth-factor-receptor-bound protein 2–extracellular-signal- regulated kinase (GRB2–ERK) pathways are important for the proliferation and survival of the osteoclast lineage, whereas VAV3, cSRC, and Casitas B-lineage lymphoma (cCBL) are included in the molecules required for cytoskeletal reorganization and bone-resorbing osteoclasts. Osteoclast activity is dependent on acidifying proton pump ATP6I and chloride channel 7 (ClC7), as well as matrix-degrading enzymes such as cathepsin K and matrix metalloproteinase 9 (MMP9). The following molecules are known to be involved in both the bone system and the immune system: NF-κB, RANKL, RANK, OPG, cFMS, M-CSF, Ig-like receptors, FcRγ, DAP12, TRAF6, MAPKs, AP1, calcineurin, NFATc1, CD40L, IL-1, IFNγ, IFNβ, TNF, LPS, DC-STAMP, PI3K, AKT, ERK, VAV3, cSRC, and cCBL.

Therefore, for bone remodeling after fracture, MSCs are recruited within lesions and induced to form new bone via both endochondral and intramembranous ossification. Citation3 Whether healing of a fracture or other injury occurs following an endochondral or intramembranous pathway is dependent on the sensitivity to the mechanical environment to which the injury is subjected.Citation2 During bone formation, Indian hedgehog (Ihh) acts at a very early stage to induce the expression of Runx2, which needs to be phosphorylated to be active.Citation3 BMPs are also necessary to lead osteoblastic commitment and to drive osteoblastic maturation, notably through Runx2 and distal-less homeobox 5 protein (Dlx5) induction.Citation3 Mitogen-activated protein kinase can phosphorylate Dlx5 and Runx2.Citation3 The osteochondroblastic progenitors can express Ihh, which induces secretion of parathyroid hormone (PTH)-related protein and acts on preosteoblastic cells (positive for collagen 1a1, alkaline phosphatase, and PTH receptor 1 (PTH-R1) to increase their maturation.Citation3 Msh homeobox 2 (Msx2) is a factor preferentially found within proliferative progenitors, whereas Dlx5 leads to maturation.Citation3 In addition, Dlx5 and Msx2 compete for DNA binding.Citation3 Therefore, the Dlx5:Msx2 content drives the maturation of cells.Citation3 In addition, Msx2 induces apoptosis in later stages of maturation.Citation3 Wnt proteins can induce the proliferation of osteochondroblastic progenitors and preosteoblasts.Citation3 When osteoblasts mature, they can express Wnt inhibitors such as Dickkopf-related protein 1 molecules.Citation3 Osteoclasts are capable of degrading mineralized bone and are hematopoietic-derived cells.Citation3 They are generated through RANKL and macrophage colony-stimulating factor cytokines secreted by activated T- and B-lymphocytes and by preosteoblasts.Citation3 Osteoclast activities can be regulated by interleukin-10 and by osteoprotegerin, a decoy receptor of RANKL.Citation3 Osteoprotegerin is preferentially expressed by mature osteoblasts (positive for osteocalcin; bone sialoprotein; PTH-R1; and osteonectin, also referred to as SPARC).Citation3 Finally, degradation of the bone matrix releases several cytokines and growth factors, such as BMPs and insulin-like growth factor, which in turn can activate immature cells ().Citation3

Figure 3 Bone remodeling after fracture. Mesenchymal stem cells are recruited within lesions and induced to form new bone following both endochondral and intramembranous pathways. During bone formation, Indian hedgehog (Ihh) acts at a very early stage to induce the expression of runt-related transcription factor 2 (Runx2), which needs to be phosphorylated to be active. Bone morphogenetic proteins (BMPs) are also necessary to lead osteoblastic commitment and to drive osteoblastic maturation, notably through Runx2 and distal-less homeobox 5 (Dlx5) induction. Mitogen-activated protein kinase (MAPK) can phosphorylate Dlx5 and Runx2. The osteochondroblastic progenitors can express Ihh, which induces secretion of parathyroid hormone-related protein (PTHrP) and also acts on preosteoblastic cells positive for collagen 1a1 (col1a1), alkaline phosphatase (ALP), and PTH receptor 1 (PTH-R1) to increase their maturation. Msh homeobox 2 (Msx2) is a factor preferentially found within proliferative progenitors, whereas Dlx5 leads to maturation. In addition, Dlx5 and Msx2 compete for DNA binding. Therefore, the Dlx5:Msx2 content drives the maturation of cells. In addition, Msx2 induces apoptosis in later stages of maturation. Wnt proteins can induce the proliferation of osteochondroblastic progenitors and preosteoblasts. When osteoblasts mature, they can express Wnt inhibitors such as Dickkopf-related protein 1 (Dkk1) molecules. Osteoclasts are capable of degrading mineralized bone and are hematopoietic-derived cells. They are generated through receptor activator of nuclear kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) cytokines secreted by activated T- and B-lymphocytes and by preosteoblasts. Osteoclast activities can be regulated by interleukin-10 (IL-10) and by osteoprotegerin (OPG), a decoy receptor of RANKL. OPG is preferentially expressed by mature osteoblasts (positive for osteocalcin [OSC], bone sialoprotein [BSP], PTH-R1, and osteonectin [SPARC]). Finally, degradation of the bone matrix releases several cytokines and growth factors, such as BMPs and insulin-like growth factor (IGF), which in turn can activate immature cells.

Note: Reprinted and modified by permission from Trends in Molecular Medicine, F Deschaseaux, L Sensebe, D Heymann, Mechanisms of bone repair and regeneration, Vol. 15, Issue 9, Pages 417–429, Copyright 1999, with permission from Elsevier.Citation3
Abbreviations: TRAP, tartrate-resistant acid phosphatase; Osx, osterix; Col10a1/2a1/1a1, collagen 10a1/2a1/1a1; GSK3β, glycogen synthase kinase 3β; TGFβ, transforming growth factor β; PPARg, peroxisome proliferator-activated receptor gamma; C/EBPa, CCAAT-enhancer-binding protein alpha.
Figure 3 Bone remodeling after fracture. Mesenchymal stem cells are recruited within lesions and induced to form new bone following both endochondral and intramembranous pathways. During bone formation, Indian hedgehog (Ihh) acts at a very early stage to induce the expression of runt-related transcription factor 2 (Runx2), which needs to be phosphorylated to be active. Bone morphogenetic proteins (BMPs) are also necessary to lead osteoblastic commitment and to drive osteoblastic maturation, notably through Runx2 and distal-less homeobox 5 (Dlx5) induction. Mitogen-activated protein kinase (MAPK) can phosphorylate Dlx5 and Runx2. The osteochondroblastic progenitors can express Ihh, which induces secretion of parathyroid hormone-related protein (PTHrP) and also acts on preosteoblastic cells positive for collagen 1a1 (col1a1), alkaline phosphatase (ALP), and PTH receptor 1 (PTH-R1) to increase their maturation. Msh homeobox 2 (Msx2) is a factor preferentially found within proliferative progenitors, whereas Dlx5 leads to maturation. In addition, Dlx5 and Msx2 compete for DNA binding. Therefore, the Dlx5:Msx2 content drives the maturation of cells. In addition, Msx2 induces apoptosis in later stages of maturation. Wnt proteins can induce the proliferation of osteochondroblastic progenitors and preosteoblasts. When osteoblasts mature, they can express Wnt inhibitors such as Dickkopf-related protein 1 (Dkk1) molecules. Osteoclasts are capable of degrading mineralized bone and are hematopoietic-derived cells. They are generated through receptor activator of nuclear kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) cytokines secreted by activated T- and B-lymphocytes and by preosteoblasts. Osteoclast activities can be regulated by interleukin-10 (IL-10) and by osteoprotegerin (OPG), a decoy receptor of RANKL. OPG is preferentially expressed by mature osteoblasts (positive for osteocalcin [OSC], bone sialoprotein [BSP], PTH-R1, and osteonectin [SPARC]). Finally, degradation of the bone matrix releases several cytokines and growth factors, such as BMPs and insulin-like growth factor (IGF), which in turn can activate immature cells.

Scaffolds

While supporting osteogenesis and osteoconduction, an ideal scaffold should also provide mechanical stability and support the necessary cell activity that leads to bone regrowth.Citation2 Important scaffold properties to consider include porosity, pore size, geometry, and material. Adequate porosity is essential for the infiltration of cells and the formation of blood vessels at the new bone.Citation40 Previous experiments found improved in vivo osteogenesis by using BMP-2-loaded hydroxyapatite scaffolds with high porosity, which allowed for cell recruitment and vascularization.Citation2,Citation41,Citation42

Scaffold geometry is another important factor for successful osteogenesis. It dictates cell adhesion, proliferation, and differentiation as well as nutrient and oxygen availability. An experiment by Kilian et al concluded that geometry is important for producing an osteogenic pathway.Citation43 They found that geometric features that increase actomyosin contractility promote osteogenesis through Wnt signaling and other pathways.Citation43 They also discovered that a stiff underlying matrix in MSC cultures led to enhanced osteogenesis, most likely because the environment closely resembled that of bone in vivo. The nature of the scaffold’s surface has important implications for bone tissue formation. The bone–implant interface has been shown to positively correlate with an increasing roughness of the implant surface.Citation44 Similarly, surface microcavities on a scaffold were shown to support a more vigorous osteogenic response from dental pulp-derived stromal stem cells.Citation45

When selecting the appropriate material for a scaffold, several elements need to be considered, such as the material’s composition, degradation rate, and the strength it affords the scaffold. Current options for materials are natural or synthetic polymers (ie, polytetrafluoroethylene), bioactive materials (ie, bioactive glass), and osteophilic materials such as hydroxyapatite ceramics or composites.Citation46 Animal studies have shown that the best procedure by which MSCs can induce bone formation is via an injection or implant with the use of a natural or synthetic scaffold.Citation9,Citation47,Citation48 The natural or synthetic polymers become encapsulated by fibrous tissue and their high molecular weight results in slower degradation and improved mechanical strength.Citation46,Citation47,Citation49,Citation50 The poly(lactic-co-glycolic acid) polymer is a popular scaffold choice as it is infrequently rejected and does not cause inflammation, therefore, it has excellent biocompatibility.Citation45 Bioactive materials chemically modify the surface of bone by forming a layer of hydroxyapatite and bind directly to the bone.Citation46,Citation51 For this reason, their use is limited to nonbearing applications. Osteophilic materials can directly apposition themselves to bone given their biocompatibility and high protein-binding affinity.Citation46 Both of these qualities make them good vehicles for drug delivery. However, their use is also restricted to non-load-bearing sites as they fail readily due to their brittleness.Citation46,Citation52 Collagen carriers used in conjunction with ceramics such as hydroxyapatite and β-tricalcium phosphate have been used to deliver BMP-7 in spinal fusion and have been shown to perform similarly to bone autografts.Citation46,Citation53

A scaffold’s degradation rate is determined by its chemical and structural properties as well as many in vivo factors including pH, temperature, ionic strength, and access to vasculature. However, degradation can be dangerous resulting in the release of acidic products and a decrease in local pH.Citation46 This can cause inflammation and/or tissue necrosis, and eventual tissue dysfunction.Citation46,Citation54

To provide structural support and mechanical stability, a scaffold must mimic the strength and stiffness of bone. A scaffold that is too weak can fail to support the skeleton, while one that is too stiff can lead to stress shielding.Citation46,Citation52 Stress shielding results when the scaffold absorbs the mechanical stress, but the host bone is not stimulated enough for bone formation to occur. Thus, bone resorption continues, resulting in potential lysis around the implant. The appropriate combination of soluble factors and biomaterials will result in the most efficient and effective tissue regeneration.

Clinical application of MSCs

Bone autografts have been used for centuries in reconstructive surgery, as well as in orthopedic surgery, as unvascularized free grafts. Also, they have been grafted with vessels that require microanastomosis to maintain cellular viability in the graft to facilitate osteogenesis. Bone marrow has been used in a similar way to autologous bone grafts, both containing osteoblast precursors.Citation46 Autologous marrow can be aspirated from the iliac crest and injected percutaneously into osseous defects.Citation46,Citation55 In 2005, Hernigou et al successfully used bone marrow cells that were isolated during surgery and concentrated them prior to implantation.Citation46,Citation56 Colony-forming unit fibroblasts are in vitro adherent bone marrow cells that can form bone in vivo.Citation46,Citation57 Although single adherent cells can form colonies containing multiple lineages, including chondrocytes, osteoblasts, adipocytes, and fibroblasts,Citation11,Citation46 these cells are not capable of extensive self-renewal and therefore are not bona fide MSCs.

In vitro osteogenesis has been thoroughly demonstrated in monolayer culture, three-dimensional scaffolds,Citation46,Citation58Citation62 research with animal and human cells, and has been reviewed elsewhere for a variety of audiences and clinical subspecialties.Citation20,Citation46,Citation63Citation66 A US Food and Drug Administration-approved cell-based product for bone regeneration in the clinic remains unavailable. Currently, the Food and Drug Administration has approved only one product under the autologous cellular therapy category (Carticel®; Genzyme Corporation, Cambridge, MA), which is designated to be used only for articular cartilage repair.Citation46

Therapeutic applications

MSCs show great promise for the treatment of disease and repair of injury. Their use has been explored in several physiological realms including cardiovascular repair, central nervous system or spinal cord injury, and bone and cartilage repair. Damaged myocardium has been an active experimental target for site-directed MSC therapy. In 2001, Orlic et al showed that locally delivered bone marrow cells can generate de novo myocardium, indicating a use for stem cells in treating coronary artery disease.Citation67 Similarly, the delivery of bone marrow cells into the infarct zone in patients following myocardial infarction resulted in a dramatic improvement in global heart function.Citation68 Following bone marrow transplantation, engraftment of bone marrow-derived cardiomyocytes was observed in the adult heart.Citation69 While MSCs and other bone marrow cells have shown improvements in myocardial function in many animal models of acute myocardial infarction, the mechanism of their effect is still unknown.

The in vivo use of stem cell-based therapy has been demonstrated for treatment of central nervous system or spinal cord injury. Two studies using a rat model of spinal cord injury have shown neurologic improvement upon site-directed application of MSCs.Citation70,Citation71 The MSCs are credited with the production of essential trophic factors, which not only promoted directed growth of new axons but also formed “guiding strands” that bridged the epicenter of injury.Citation16

The most successful application of MSCs has been site-directed administration for repair of bone or cartilage. It is estimated that 1,600,000 bone grafts are performed every year to regenerate bone lost to trauma and disease, 6% (96,000) of which are craniomaxillofacial in nature.Citation72 In a two-stage procedure, a composite MSC and a titanium/ hydroxyapatite scaffold was placed into a patient’s latissimus dorsi for 7 weeks before transplantation to replace a segment of mandible.Citation46 Although the procedure was associated with a number of technical problems, such as fracture of the titanium mesh, partial infection, and necrosis, islands of bone formation and increased bone density were documented within the transplant.Citation46,Citation73 Also, iliac crest MSCs injected around dental implants have been shown to result in new bone formation and osseointegration of the implants.Citation46,Citation74 Expanded periosteal cells were used in the reconstruction of an avulsed phalanx with porous hydroxyapatite.Citation46,Citation75 Follow-up at 28 months indicated retention of implant volume and structure, and evidence of lamellar bone comprising 5% of the construct volume.Citation46

There is some clinical experience of bone reconstruction with expanded human MSCs combined with scaffolds. MSCs implanted on hydroxyapatite/tricalcium phosphate scaffolds have accelerated bone formation when placed in craniofacial defects and critical long-bone defects in small and large animal systems.Citation76Citation78 Constructs of expanded autologous MSCs in macroporous hydroxyapatite were used in three patients with large segmental bone defects.Citation46,Citation79,Citation80 Fusion between the implant and host bone occurred 5–7 months after surgery, with restoration of pain-free function for 6–7 years.Citation46 A study by Horwitz et al demonstrated that allogenic bone marrow transplantation in children with osteogenesis imperfecta, a genetic bone disorder, resulted in engraftment of donor-derived MSCs and an increase in new bone formation.Citation81

Future directions

The basic and preclinical research literature clearly indicates that the use of MSCs for the reconstruction and repair of bone is feasible. Problem areas, however, include cell numbers, an overreliance on existing scaffold materials, optimum delivery of the required factors, and the control of transgene expression. Other matters that need further attention include the elimination of fetal calf serum, untoward effects of expanding cell numbers, the commitment to or reversibility of the differentiated state, the endurance of the cells in vivo, vascularization, integration with the recipient bone, and the capacity to form bone and marrow structures in vivo.

Concerns about the relevance of in vitro studies and studies in immunodeficient animals have dampened enthusiasm about the role of bone tissue engineering in bone regeneration. It remains unclear whether the cost and complexity of cell-based tissue engineering would hamper its potential application for bone tissue. Moreover, the relevance of troubling data from mouse studies needs to be addressed. In one study, after slow initial growth, mouse MSCs suddenly proliferated rapidly and became tumorigenic.Citation46,Citation82 In other studies, the host-derived sarcomas developed around implants with MSCs;Citation46,Citation83 this was thought to be a result of the stimulation of lymphocyte expansion by MSCs and suppression of the host’s antitumor immune response.Citation46 Paracrine secretions of MSCs, therefore, could represent a double-edged sword by both inhibiting allogeneic surveillance and by promoting tumor development. Of importance, however, is that, unlike most preparations of human MSCs, the mouse MSCs were prepared from whole marrow and included adherent blood cells and other differentiated cells. A reassessment of circulating osteoblast progenitor cells and perivascular cells, and their contribution to bone repair, raises attractive ideas for the capitalization of inherent regenerative potential without the use of exogenous cells.

What is needed now is a greater understanding of inadequate responses to tissue injury and the potential for mobilizing innate pathways. Current thinking about biologic, regenerative medicine reemphasizes the potential of “primed” materials for stimulating, enhancing, or controlling a tissue’s innate capacity for repair. Successful tissue regeneration will require the identification of tissues with the appropriate populations of cells, the best conditions for their ex vivo expansion, the optimal nature of the scaffolds and carriers used, and the development of suitable preclinical animal models. To optimize the therapeutic applications of MSCs, we must construct novel techniques that surpass the simple expansion and transplantation procedures currently in use.

Disclosure

The authors report no conflicts of interest in this work.

References

  • CiacciJHoAAmesCPJandialRStem cell horizons in intervertebral disc degenerationStem Cells Cloning200813139
  • SundelacruzSKaplanDLStem cell- and scaffold-based tissue engineering approaches to osteochondral regenerative medicineSemin Cell Dev Biol200920664665519508851
  • DeschaseauxFSensebeLHeymannDMechanisms of bone repair and regenerationTrends Mol Med200915941742919740701
  • DuplombLDagouassatMJourdonPHeymannDConcise review: embryonic stem cells: a new tool to study osteoblast and osteoclast differentiationStem Cells200725354455217095705
  • DuplombLBaud’huinMCharrierCInterleukin-6 inhibits receptor activator of nuclear factor kappaB ligand-induced osteoclastogenesis by diverting cells into the macrophage lineage: key role of Serine727 phosphorylation of signal transducer and activator of transcription 3Endocrinology200814973688369718403479
  • Kwan TatSPadrinesMTheoleyreSHeymannDFortunYIL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiologyCytokine Growth Factor Rev2004151496014746813
  • PapachroniKKKaratzasDNPapavassiliouKABasdraEKPapavassiliouAGMechanotransduction in osteoblast regulation and bone diseaseTrends Mol Med200915520821619362057
  • FergusonCAlpernEMiclauTHelmsJADoes adult fracture repair recapitulate embryonic skeletal formation?Mech Dev1999871–2576610495271
  • El TamerMKReisRLProgenitor and stem cells for bone and cartilage regenerationJ Tissue Eng Regen Med20093532733719418440
  • GlowackiJThe deceiving appearances of osteoclastsN Engl J Med20093601808219118309
  • PittengerMFMackayAMBeckSCMultilineage potential of adult human mesenchymal stem cellsScience1999284541114314710102814
  • MeijerGJde BruijnJDKooleRvan BlitterswijkCACell-based bone tissue engineeringPLoS Med200742e917311467
  • BarryFPMurphyJMMesenchymal stem cells: clinical applications and biological characterizationInt J Biochem Cell Biol200436456858415010324
  • LennonDPHaynesworthSEBruderSPJaiswalNCaplanAIHuman and animal mesenchymal progenitor cells from bone marrow: Identification of serum for optimal selection and proliferationIn Vitro Cell Dev Biol Anim19963210602611
  • JaiswalNHaynesworthSECaplanAIBruderSPOsteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitroJ Cell Biochem19976422953129027589
  • JavazonEHBeggsKJFlakeAWMesenchymal stem cells: paradoxes of passagingExp Hematol200432541442515145209
  • OwenMFriedensteinAJStromal stem cells: marrow-derived osteogenic precursorsCiba Found Symp198813642603068016
  • DigirolamoCMStokesDColterDPhinneyDGClassRProckopDJPropagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiateBr J Haematol1999107227528110583212
  • TogelFWestenfelderCAdult bone marrow-derived stem cells for organ regeneration and repairDev Dyn2007236123321333117685479
  • ArthurAZannettinoAGronthosSThe therapeutic applications of multipotential mesenchymal/stromal stem cells in skeletal tissue repairJ Cell Physiol2009218223724518792913
  • ChamberlainGFoxJAshtonBMiddletonJConcise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homingStem Cells200725112739274917656645
  • SteckEBertramHAbelRChenBWinterARichterWInduction of intervertebral disc-like cells from adult mesenchymal stem cellsStem Cells200523340341115749935
  • RichardsonSMCurranJMChenRThe differentiation of bone marrow mesenchymal stem cells into chondrocyte-like cells on poly-L-lactic acid (PLLA) scaffoldsBiomaterials200627224069407816569429
  • SekiyaILarsonBLSmithJRPochampallyRCuiJGProckopDJExpansion of human adult stem cells from bone marrow stroma: conditions that maximize the yields of early progenitors and evaluate their qualityStem Cells200220653054112456961
  • EricesACongetPMinguellJJMesenchymal progenitor cells in human umbilical cord bloodBr J Haematol2000109123524210848804
  • GangEJHongSHJeongJAIn vitro mesengenic potential of human umbilical cord blood-derived mesenchymal stem cellsBiochem Biophys Res Commun2004321110210815358221
  • FukuchiYNakajimaHSugiyamaDHiroseIKitamuraTTsujiKHuman placenta-derived cells have mesenchymal stem/progenitor cell potentialStem Cells200422564965815342929
  • ImGIShinYWLeeKBDo adipose tissue-derived mesenchymal stem cells have the same osteogenic and chondrogenic potential as bone marrow-derived cells?Osteoarthritis Cartilage2005131084585316129630
  • TakayanagiHOsteoimmunology: shared mechanisms and crosstalk between the immune and bone systemsNat Rev Immunol20077429230417380158
  • SeemanEDelmasPDBone quality – the material and structural basis of bone strength and fragilityN Engl J Med2006354212250226116723616
  • HaradaSRodanGAControl of osteoblast function and regulation of bone massNature2003423693734935512748654
  • KarsentyGWagnerEFReaching a genetic and molecular understanding of skeletal developmentDev Cell20022438940611970890
  • NakashimaKZhouXKunkelGThe novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formationCell20021081172911792318
  • BoyleWJSimonetWSLaceyDLOsteoclast differentiation and activationNature2003423693733734212748652
  • SudaTTakahashiNUdagawaNJimiEGillespieMTMartinTJModulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand familiesEndocr Rev199920334535710368775
  • TakayanagiHMechanistic insight into osteoclast differentiation in osteoimmunologyJ Mol Med (Berl)200583317017915776286
  • AsagiriMTakayanagiHThe molecular understanding of osteoclast differentiationBone200740225126417098490
  • TheillLEBoyleWJPenningerJMRANK-L and RANK: T cells, bone loss, and mammalian evolutionAnnu Rev Immunol20022079582311861618
  • TeitelbaumSLRossFPGenetic regulation of osteoclast development and functionNat Rev Genet20034863864912897775
  • BessaPCCasalMReisRLBone morphogenetic proteins in tissue engineering: the road from laboratory to clinic, part II (BMP delivery)J Tissue Eng Regen Med200822–3819618383454
  • RoyTDSimonJLRicciJLRekowEDThompsonVPParsonsJRPerformance of degradable composite bone repair products made via three-dimensional fabrication techniquesJ Biomed Mater Res A200366228329112888998
  • KarageorgiouVKaplanDPorosity of 3D biomaterial scaffolds and osteogenesisBiomaterials200526275474549115860204
  • KilianKABugarijaBLahnBTMrksichMGeometric cues for directing the differentiation of mesenchymal stem cellsProc Natl Acad Sci U S A2010107114872487720194780
  • BuserDSchenkRKSteinemannSFiorelliniJPFoxCHStichHInfluence of surface characteristics on bone integration of titanium implants. A histomorphometric study in miniature pigsJ Biomed Mater Res19912578899021918105
  • GrazianoAd’AquinoRCusella-De AngelisMGConcave pit-containing scaffold surfaces improve stem cell-derived osteoblast performance and lead to significant bone tissue formationPLoS One200726e49617551577
  • BuenoEMGlowackiJCell-free and cell-based approaches for bone regenerationNat Rev Rheumatol200951268569719901916
  • CanceddaRDozinBGiannoniPQuartoRTissue engineering and cell therapy of cartilage and boneMatrix Biol2003221819112714045
  • RichardsMHuibregtseBACaplanAIGouletJAGoldsteinSAMarrow-derived progenitor cell injections enhance new bone formation during distractionJ Orthop Res199917690090810632457
  • BrauneckerJBabaMMilroyGECameronREThe effects of molecular weight and porosity on the degradation and drug release from polyglycolideInt J Pharm20042821–2193415336379
  • ThomsonRCYaszemskiMJPowersJMMikosAGFabrication of biodegradable polymer scaffolds to engineer trabecular boneJ Biomater Sci Polym Ed19957123387662615
  • OonishiHHenchLLWilsonJQuantitative comparison of bone growth behavior in granules of Bioglass, A-W glass-ceramic, and hydroxyapatiteJ Biomed Mater Res2000511374610813743
  • KhanYYaszemskiMJMikosAGLaurencinCTTissue engineering of bone: material and matrix considerationsJ Bone Joint Surg Am200890Suppl 1364218292355
  • DelecrinJTakahashiSGouinFPassutiNA synthetic porous ceramic as a bone graft substitute in the surgical management of scoliosis: a prospective, randomized studySpine (Phila Pa 1976)200025556356910749632
  • TemenoffJSLuLMikosAGBone tissue engineering using synthetic biodegradable polymer scaffoldsDaviesJEBone EngineeringTorontoEM Squared Incorporated2000454461
  • ConnollyJFGuseRTiedemanJDehneRAutologous marrow injection for delayed unions of the tibia: a preliminary reportJ Orthop Trauma1989342762822600692
  • HernigouPPoignardAManicomOMathieuGRouardHThe use of percutaneous autologous bone marrow transplantation in nonunion and avascular necrosis of boneJ Bone Joint Surg Br200587789690215972899
  • FriedensteinAJPiatetzky-ShapiroIIPetrakovaKVOsteogenesis in transplants of bone marrow cellsJ Embryol Exp Morphol19661633813905336210
  • LisignoliGFiniMGiavaresiGNicoliANToneguzziSFacchiniAOsteogenesis of large segmental radius defects enhanced by basic fibroblast growth factor activated bone marrow stromal cells grown on non-woven hyaluronic acid-based polymer scaffoldBiomaterials20022341043105111791907
  • DattaNHoltorfHLSikavitsasVIJansenJAMikosAGEffect of bone extracellular matrix synthesized in vitro on the osteoblastic differentiation of marrow stromal cellsBiomaterials200526997197715369685
  • YamadaYUedaMNaikiTTakahashiMHataKNagasakaTAutogenous injectable bone for regeneration with mesenchymal stem cells and platelet-rich plasma: tissue-engineered bone regenerationTissue Eng2004105–695596415265313
  • SikavitsasVIBancroftGNHoltorfHLJansenJAMikosAGMineralized matrix deposition by marrow stromal osteoblasts in 3D perfusion culture increases with increasing fluid shear forcesProc Natl Acad Sci U S A200310025146831468814657343
  • LuHHEl-AminSFScottKDLaurencinCTThree-dimensional, bioactive, biodegradable, polymer-bioactive glass composite scaffolds with improved mechanical properties support collagen synthesis and mineralization of human osteoblast-like cells in vitroJ Biomed Mater Res A200364346547412579560
  • AbdallahBMKassemMThe use of mesenchymal (skeletal) stem cells for treatment of degenerative diseases: current status and future perspectivesJ Cell Physiol2009218191218726996
  • LeeKChanCKPatilNGoodmanSBCell therapy for bone regeneration– bench to bedsideJ Biomed Mater Res B Appl Biomater200989125226318777578
  • SterodimasADe FariaJCorreaWEPitanguyITissue engineering in plastic surgery: an up-to-date review of the current literatureAnn Plast Surg20096219710319131730
  • RobeyPGBiancoPThe use of adult stem cells in rebuilding the human faceJ Am Dent Assoc2006137796197216803822
  • OrlicDKajsturaJChimentiSBodineDMLeriAAnversaPBone marrow stem cells regenerate infarcted myocardiumPediatr Transplant20037Suppl 3868812603699
  • StammCWestphalBKleineHDAutologous bone-marrow stem-cell transplantation for myocardial regenerationLancet20033619351454612517467
  • DebAWangSSkeldingKAMillerDSimperDCapliceNMBone marrow-derived cardiomyocytes are present in adult human heart: a study of gender-mismatched bone marrow transplantation patientsCirculation200310791247124912628942
  • HofstetterCPSchwarzEJHessDMarrow stromal cells form guiding strands in the injured spinal cord and promote recoveryProc Natl Acad Sci U S A20029942199220411854516
  • ChoppMZhangXHLiYSpinal cord injury in rat: treatment with bone marrow stromal cell transplantationNeuroreport200011133001300511006983
  • EinhornTABasic science of bone graft substitutesAnnual Meeting of the Orthopaedic Trauma Association2003 Oct 8Salt Lake City, UT Available from: http://www.hwbf.org/ota/am/ota03/bssf/OTA03BG1.htmAccessed August 8, 2011
  • WarnkePHWiltfangJSpringerIMan as living bioreactor: fate of an exogenously prepared customized tissue-engineered mandibleBiomaterials200627173163316716504287
  • VacantiCABonassarLJVacantiMPShufflebargerJReplacement of an avulsed phalanx with tissue-engineered boneN Engl J Med2001344201511151411357154
  • UedaMYamadaYKagamiHHibiHInjectable bone applied for ridge augmentation and dental implant placement: human progress studyImplant Dent2008171829018332761
  • De KokIJPeterSJArchambaultMInvestigation of allogeneic mesenchymal stem cell-based alveolar bone formation: preliminary findingsClin Oral Implants Res200314448148912869011
  • MankaniMHKuznetsovSAFowlerBKingmanARobeyPGIn vivo bone formation by human bone marrow stromal cells: effect of carrier particle size and shapeBiotechnol Bioeng20017219610711084599
  • TsuchidaHHashimotoJCrawfordEManskePLouJEngineered allogeneic mesenchymal stem cells repair femoral segmental defect in ratsJ Orthop Res2003211445312507579
  • QuartoRMastrogiacomoMCanceddaRRepair of large bone defects with the use of autologous bone marrow stromal cellsN Engl J Med2001344538538611195802
  • MarcacciMKonEMoukhachevVStem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical studyTissue Eng200713594795517484701
  • HorwitzEMProckopDJFitzpatrickLATransplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfectaNat Med19995330931310086387
  • TolarJNautaAJOsbornMJSarcoma derived from cultured mesenchymal stem cellsStem Cells200725237137917038675
  • TassoRAugelloACaridaMDevelopment of sarcomas in mice implanted with mesenchymal stem cells seeded onto bioscaffoldsCarcinogenesis200930115015718849298