Skip to main content
Therapeutic Advances in Musculoskeletal Disease logoLink to Therapeutic Advances in Musculoskeletal Disease
. 2016 Apr 1;8(3):57–71. doi: 10.1177/1759720X16642372

Bone repair with skeletal stem cells: rationale, progress to date and clinical application

Elena A Jones 1,, Peter V Giannoudis 2, Dimitrios Kouroupis 3
PMCID: PMC4872172  PMID: 27247633

Abstract

Bone marrow (BM) contains stem cells for both hematopoietic and nonhematopoietic lineages. Hematopoietic stem cells enable hematopoiesis to occur in a controlled manner in order to accurately compensate for the loss of short- as well as long-lived mature blood cells. The physiological role of nonhematopoietic BM stem cells, often referred to as multipotential stromal cells or skeletal stem cells (SSCs), is less understood. According to an authoritative current opinion, the main function of SSCs is to give rise to cartilage, bone, marrow fat and hematopoiesis-supportive stroma, in a specific sequence during embryonic and postnatal development. This review outlines recent advances in the understanding of origins and homeostatic functions of SSCs in vivo and highlights current and future SSC-based treatments for skeletal and joint disorders.

Keywords: bone repair, hematopoietic stem cells, multipotential stromal cells, osteoarthritis, skeletal stem cells, systemic bone diseases

Introduction: bone marrow determined osteogenic progenitors

The discovery of multipotential stromal cells (MSCs) (‘determined osteogenic progenitors’) in the bone marrow (BM) emanates from seminal experiments by Friedenstein and colleagues, who demonstrated the presence in the BM of clonogenic and multipotential cells, able to generate mesenchymal tissues in vivo [Friedenstein et al. 1974]. Whereas an innate ability to form bony ‘ossicles’ in vivo has also been shown for MSCs resident in other tissues, such as periosteum [De Bari et al. 2006; Arnsdorf et al. 2009; Roberts et al. 2015] only BM-resident MSCs possessed an additional strong ability to support ectopic hematopoiesis [Miura et al. 2006; Sacchetti et al. 2007; Yamaza et al. 2009].

Whereas an ability to support hematopoiesis cannot be considered as an indispensable feature of all MSCs, its relevance to skeletal development and physiological live-long bone remodeling should not be underestimated. A well-known age-related phenomenon of red-to-yellow marrow conversion in human long bones clearly illustrates that BM MSC differentiation towards fat goes hand in hand with reduced hematopoiesis in the same bone [Bianco and Robey, 2015]. On the other hand, recent advances in the hematopoietic stem cell (HSC) field indicate that HSC stromal ‘niche’ cells, that is the same BM MSCs, may be involved in the progression of hematopoietic malignancies [Garcia-Garcia et al. 2015]. These unique features rightly justify positioning BM MSCs [or according to alternative terminology, skeletal stem cells (SSCs)] ‘above and beyond’ other tissue-resident MSCs, as they are critically important in the whole organism physiology, rather than purely in their host tissue maintenance [Oreffo et al. 2005; Kassem and Bianco, 2015]. This review article will summarize rationale, progress to date and clinical application of BM MSCs or SSCs in the field of bone regeneration and repair. While not attempting to describe the whole spectrum of tissue-resident MSCs, we will nevertheless give some pertinent examples of other MSC types (adult and fetal) that have been successfully used in preclinical and clinical investigations aimed at bone repair.

SSCs: one cell or many?

The fact that BM MSCs or SSCs should be able to produce four distinct lineages, cartilage, bone, fat and hematopoiesis-supportive stroma, has over the years posed a question of their potential heterogeneity and, if it existed, whether it was predetermined (i.e. developmentally regulated) or environmentally induced. Early studies have initially focused on tripotential features of BM MSCs [Muraglia et al. 2000] and attempted to assign certain specific markers to subpopulations of committed osteogenic and nonosteogenic progenitors [Satomura et al. 1998]. It was also recognized that standard cultivation leads to significant phenotypic changes in MSCs [Digirolamo et al. 1999] and that the presence of markers on certain MSC subsets in vitro does not warrant their presence in vivo [Buhring et al. 2007]. Cell-sorting studies using a number of surface marker combinations such as CD45/CD271 [Jones et al. 2002; Buhring et al. 2007; Battula et al. 2009; Churchman et al. 2012; Li et al. 2014] in humans, and platelet-derived growth factor (PDGFR)-α/Sca-1 in mice [Morikawa et al. 2009; Houlihan et al. 2012] have succeeded in isolating the whole fraction of BM MSCs, rather than its specific functional subsets. Interestingly, Tormin and colleagues have shown that two different phenotypes of BM MSCs were linked to their in vivo topography, rather than differentiation predisposition [Tormin et al. 2011]. The lin/CD271+/CD45/CD146–/low) subset characterized bone-lining MSCs located near the bone surfaces whereas the lin/CD271+/CD45/CD146+ subset characterized reticular cells located perivascularly, however up to now it remained unclear whether they had the same or different embryonic origins.

The neural crest (NC) is a transient embryonic tissue. During development, NC cells migrate to various locations and persist as undifferentiated cells at least until late gestation [Nagoshi et al. 2009]. According to earlier reports, NC stem cells were found to migrate through the bloodstream and reach the BM coinciding with the arrival of HSCs [Dzierzak and Speck, 2008; Nagoshi et al. 2008], where they persisted into adulthood and played a role in HSC maintenance. In more recent reports, NC stem cells were traced as travelling along developing nerves rather than via the bloodstream [Isern et al. 2014]. Significantly, only these NC stem cells (nestin+ MSCs) were located in neonatal BM and endowed with HSC support function while a separate population of mesoderm-derived cells (nestin MSCs, located in the appendicular skeleton) formed a pool of progenitors for endochondrogenesis [Isern et al. 2014]. Other recent articles provided additional evidence for developmental heterogeneity of mouse SSCs. For example, Wortley and colleagues recently demonstrated the existence of osteochondroreticular stem cells generating articular and growth plate cartilage during development, which were in late adulthood complemented by traditional ‘perisinusoidal’ adipogenesis-competent MSCs [Worthley et al. 2015]. Different phenotypic profiles of early postnatal and adult mouse SSCs were also reported in the studies of Ono and colleagues [Ono et al. 2014], Zhou and colleagues [Zhou et al. 2014] and Chan and colleagues [Chan et al. 2015]. Therefore, according to the most recent findings, SSCs do not in fact represent a ‘singularity’ [Bianco and Robey, 2015] but are rather a mixture of stem cells of different embryological origins [Komada et al. 2012; Isern et al. 2014], this being dependent on an anatomical location as well as a ante- and postnatal developmental stage [Kassem and Bianco, 2015].

Although therapeutic exploitation of the hematopoiesis-supporting subpopulation of SSCs is outside the scope of this article, it is worth noting that manipulating HSC-niche cells has broad implications for the treatment of leukemias and other BM diseases (reviewed by Garcia-Garcia and colleagues) [Garcia-Garcia et al. 2015]. Furthermore, recent evidence suggests that BM MSCs have a major role in ‘emergency hemopoiesis’, that is ‘on demand’ myeloid-lineage cell production in response to infection [Boettcher et al. 2012; Welner and Kincade, 2014; Ziegler et al. 2015]. This is because MSCs possess toll-like receptors [Liotta et al. 2008] and are able to respond to pathogens by producing soluble mediators that support the survival of innate immune cells [Cassatella et al. 2011] or skew HSC differentiation towards innate immune cell lineages [Welner and Kincade, 2014]. Furthermore, perisinusoidal MSCs are likely controlling the egress of hematopoietic cells into systemic circulation [Cariappa et al. 2005; Allende et al. 2009]. In some way, the ability of BM MSCs to orchestrate emergency hematopoiesis can be seen as one example of their native or in vivo immunoregulatory function, which is easily reconciled with their HSC niche support function.

The use of SSCs in orthopedics

Early reports have shown that standard culture expansion of BM MSCs or SSCs is permissive for the maintenance of osteo- and chondrogenic progenitors [Muraglia et al. 2000]. Early clinical studies utilized culture-expanded BM MSCs seeded on ceramic scaffolds to enable repair of large (segmental) bone defects [Quarto et al. 2001]. Several important clinical studies employing autologous cultured BM MSCs for large bone defect repair including avascular necrosis of the femoral head are outlined in Table 1. Avascular necrosis of the femoral head is a serious orthopedic condition often occurring in young individuals (Figure 1). Although limited in their level of evidence, a combination of these clinical case reports and retrospective studies suggests good clinical safety of cultured BM MSCs for a follow-up period of up to 7 years. Although local application of MSCs resulted in complete fusion/healing in cases of fractures and nonunions, and decreased necrotic lesions in cases of osteonecrosis (Table 1), it needs to be noted that larger-cohort, prospective, randomized studies are needed to confirm the observed beneficial effects.

Table 1.

Clinical studies using cultured autologous BM MSCs/SSCs for the treatment of orthopedic defects.

Type of study Cell/identity markers used Delivery Patient’s group Average follow up Outcome
Quarto et al. [2001] Marcacci et al. [2007] Prospective MSCs + scaffold, cell selection via specialized culture medium, NR Local implantation, 2.0 × 107 MSCs per ml mixed with scaffold Large long bone defectsN = 3, 4–7 cm segment from tibia, ulna, humerus 6–7 years No complications observed. Complete fusion between implant and host bone 5–7 months post surgery. At 6–7 years post surgery good integration was maintained and no late fractures observed
Kim et al. [2009] Randomized Osteogenically differentiated MSCs, cell selection via specialized culture medium/bone-specific collagen, alkaline phosphatase Local injection, 1.2 × 107 MSCs per 0.4 ml mixed with fibrin at 1:1 ratio Various long bone fractures N = 64 2 months No complications observed. Autologous osteoblast injection resulted in significant fracture healing acceleration
Zhao et al. [2012] Randomized MSCs, cell selection via specialized culture medium, NR Local injection, 2.0 × 106 MSCs in 2 ml of saline Osteonecrosis of femoral head N = 53 5 years No complications observed. At 5 years post surgery only 2 of the 53 BM MSC-treated femoral heads progressed and underwent vascularized bone grafting. Improved measures of femoral head function and decreased volume of the necrotic lesion
Giannotti et al. [2013] Retrospective Osteogenically differentiated MSCs + fibrin glue, cell selection via specialized culture medium/ISCT panel for MSCs,* alkaline phosphatase activity/von Kossa staining for osteoblasts Local implantation, 0.5–2.0 × 106 MSCs in 2 ml of fibrin clot Upper limb non-unions N = 8 6 years and 3 months No complications observed. All patients recovered limb function with no evidence of tissue overgrowth or tumour formation
*

According to ISCT minimal criteria, MSCs must express CD73, CD90, CD105 and lack expression of CD11β or CD14, CD19 or CD79α, CD34, CD45, and HLA-DR surface molecules [Dominici et al. 2006].

BM, bone marrow; MSC, multipotential stromal cell; NR, not reported; SSC, skeletal stem cell ; ISCT, international society of cellular therapies; HLA-DR: human leukocyte antigen-D related.

Figure 1.

Figure 1.

Anteroposterior radiograph of a 19-year-old man with post-traumatic late-stage avascular necrosis (a). Note the large cartilage defect discovered intraoperatively (b).

With the use of cultured MSCs, that remain undifferentiated, the main challenge remains how to insure their maximal osteogenic differentiation with minimal untoward commitment to other lineages. Recently proposed approaches for this include preselection of stage-specific osteogenic progenitor subsets [Twine et al. 2014; Loebel et al. 2015] or the use of ‘smart’ new-generation osteoconductive scaffolds with optimal pore sizes, calcium release and resorption profiles [Roberts et al. 2015] or precise nanosurface geometries or cell-material interfaces [Dalby et al. 2007; Crowder et al. 2016].

More recently, uncultured BM MSCs in the form of BM ‘concentrates’ or ‘viable allografts’ have emerged as a new way of bone defect treatment (Table 2). Bone marrow concentrates are normally prepared by minimal manipulation of a BM aspirate such as density centrifugation and volume reduction and their MSC content is normally assessed based on colony-forming unit-fibroblast (CFU-F) assay [Woodell-May et al. 2015]. Viable allografts are prepared from cadaveric cancellous bone after the removal of the immune cell component from the graft while preserving the multipotential nonimmune cells [Baboolal et al. 2014]. In their pioneering clinical studies, Hernigou and colleagues (studies described in Table 2) used BM concentrates for a wide range of orthopedic indications, such as avascular necrosis, fracture nonunion and rotator cuff repair. Overall, these and other studies showed the safety of local or systemic transplantation of uncultured BM MSCs with a follow up of up to 22 years. Furthermore, positive outcomes were found to be correlated with the number of transplanted MSCs (Table 2). It is noteworthy that although these treatments represented a mixture of cells, rather than a pure population of MSCs, some ‘contaminant’ cell fractions, for example monocytes, could contribute to bone repair, by virtue of being precursors of osteoclasts. Furthermore, concentrated injected platelets may also be highly beneficial as producers of high local levels of vascular endothelial growth factor, PDGFs and fibroblast growth factors [Dhillon et al. 2012], the molecules very important in bone repair [Giannoudis et al. 2007]. Although positive clinical outcomes were reported for both cultured and noncultured MSC preparations, the above evidence suggests that their mechanisms of action are likely to be very different and requiring further investigations in preclinical studies.

Table 2.

Clinical studies using uncultured BMAC or viable allografts for the treatment of orthopedic defects.

Type of study Cell Delivery Patient’s group Average follow up Outcome
Hernigou and Beaujean [2002] Prospective BMAC* Local injection25 × 103 MSCs per hip Hip osteonecrosisN = 116 (189 hips) 7 years (range 5–11) Extremely low incidence of complications. BMAC injection was effective in treating patients with earlier stages of osteonecrosis (stages I and II). Patients who had greater number of MSCs transplanted had better outcomes.
Hernigou et al. [2009] Prospective BMAC* Local injection24 × 103 MSCs per hip Hip osteonecrosisN = 342 (534 hips) 13 years (range 8–18) No complications observed. Sixty-nine hips with stage I osteonecrosis at the time of surgery showed total resolution of osteonecrosis post BMAC transplantation. For the other 371 hips the osteonecrosis volume was reduced from 26 to 12 cm3 post BMAC transplantation for a follow up of 12 years
Jäger et al. [2011] Prospective BMAC* + scaffolds (hydroxyapatite, collagen sponge) Local transplantationMSC dose: NR Various bone deficienciesN = 39 Minimal 6 monthsMaximal 2.5 years No complications observed. All patients showed new bone formation. BMAC combined with hydroxyapatite scaffold reduced the time of healing compared with BMAC combined with collagen sponge.
Gonshor et al. [2011] Retrospective Viable allograft** Local transplantation Sinus augmentation 5 months Viable allograft transplantation resulted in high percentage of vital bone content and relatively short healing phase
MSC dose: NR N = 21
Kerr et al. [2011] Retrospective Viable allograft** Local transplantation Extreme lateral interbody (XLIF) fusion 8–27 months No complications observed. Solid arthrodesis was achieved in 92.3% of patients
MSC dose: NR N = 52
Hollawell [2012] Prospective Viable allograft** Local transplantation Foot and ankle arthrodesis 6 months No complications observed. All patients showed arthrodesis with an average time to fusion of 13.5 weeks
MSC dose: NR N = 20
Tohmeh et al. [2012] Retrospective Viable allograft** Local transplantationMSC dose: NR XLIF 12 months No complications observed. At 12 months, complete interbody fusion was shown in 90.2% of XLIF levels
N = 40
Hernigou et al. [2012] Retrospective BMAC* Local injection, 119–195 × 103 MSCs per hip Hip osteonecrosis 17 years (range 15–20) Better reduction of pain, reduction with time in number of hips progressed to collapse, and delayed need for total hip replacement
N = 62, bilateral,one hip CD + BMAC(n = 62)
one hip CD alone(n = 62)
Hernigou et al. [2013] Retrospective BMAC* Local injection, 0.62 × 105–2.0 × 106 MSCs per patient Osteonecrosis 1089 12.5 years (range 5–22) No complications observed. No increased tumorigenesis risk at the site of BMAC transplantation or elsewhere for an average follow up of 12.5 years
Nonunion 523
General orthopedics 261
Scott and Hyer [2013] Retrospective Viable allograft** Local transplantation Foot and ankle arthrodesis 12 months No complications observed. The average interval to radiologic union ranged between 8 and 15 weeks. The interval to full weightbearing was 5–14 weeks
MSC dose: NR N = 20
Hernigou et al. [2014] Retrospective BMAC* Local injection, 2.6–7.6 × 104 MSCs per patient Rotator cuff repair N = 45 Minimum 10 years All BMAC-treated rotator cuffs healed 6 months post surgery whereas a substantial improvement in the tendon integrity was present for a follow up of 10 years
Matched controlN = 45
Hernigou et al. [2015] Retrospective BMAC* Local injection, 4.3–7.9 × 104 MSCs per patient Ankle nonunions 3–15 years Low number of complications observed. BMAC transplantation promoted nonunion healing in 70 of 86 patients with diabetes. Patients with diabetes treated with standard bone grafting showed lower healing rate and major complications
BMAC= 86
Standard bone grafting = 86
*

MSC presence confirmed by CFU-F assay.

**

MSC presence confirmed by manufacturer.

BMAC, bone marrow aspirate concentrate; CD, core decompression; CFU-F, colony-forming unit fibroblast; MSC, multipotential stromal cell; NR, not reported.

Despite encouraging clinical results, the dosages of implanted MSCs (either cultured or uncultured) remain in most cases empirical, and the fate of implanted cells remains to be elucidated. For the quantification of uncultured MSCs in BM aspirates, Cuthbert and colleagues developed a rapid (40 min) flow cytometry based assay that can be used to determine MSC dose intraoperatively [Cuthbert et al. 2012]. Another study from our laboratory [Baboolal et al. 2014] has established an average MSC dose in a viable allograft product, Osteocel (Nuvasive, Inc, San Diego, USA) (0.5 million MSCs/g of graft). In terms of in vivo tracking of implanted or injected MSCs, the most widely used noninvasive approach in animal models has been the green fluorescent protein (GFP) labeling [Guo et al. 2012] whereas magnetic resonance imaging (MRI) was applied to detect iron oxide nanoparticle or fluorine-19 agent labeled MSCs in other studies [Mathiasen et al. 2013; Gaudet et al. 2015].

Regarding the mechanisms of MSC action for their use in orthopedics, apart from their ‘classical’ roles as osteo- and chondroprogenitors (driving intramembranous and osteochondral repair processes, respectively) [Thompson et al. 2015], MSCs may also regulate later bone repair phases, including the remodeling of the transitional tissue and osteointegration via their immunomodulatory [Le Blanc and Davies, 2015], prosurvival and other ‘trophic’ functions [Caplan and Correa, 2011]. To retain both regenerative and immunomodulatory MSC subsets in therapeutic MSC preparations, and in order to capitalize on their combined functions, a better knowledge of their differential phenotypes [James et al. 2015], and methods for their prospective selection would be required. Finally, to achieve a broader understanding of the effectiveness of MSC delivery in orthopedics, particularly in terms of their survival and engraftment in hypoxic environments such as avascular necrosis lesions, more clinical studies with MRI-detectable labeled MSCs are definitely needed.

Exploiting SSCs for the conditions of systemic bone loss

Apart from bone damage due to injury, other conditions of bone loss are represented by systemic bone diseases such as osteoporosis (OP) and osteogenesis imperfecta (OI). In a pioneering study, Pereira and coworkers were the first to apply allogeneic BM MSC therapy in an OI mouse model, showing significant increase in collagen and mineral content 2.5 months after local transplantation [Pereira et al. 1998]. In other studies, systemic infusion of allogeneic BM MSCs in OI mouse models showed MSC engraftment in recipient BM, trabecular bone and cortical bones where they directly differentiated into osteoblasts and synthesized type I collagen [Wang et al. 2006a; Li et al. 2007].

The first human clinical trial to treat OI has been performed by Horwitz and colleagues [Horwitz et al. 2002] who systemically infused allogeneic BM MSCs to treat six children with severe OI. Although four out of five children showed increased bone mineral density and accelerated linear growth at 18–36 months of follow up, only 1% of infused BM MSCs could be detected in bone, skin and other tissues. Low engraftment of systemically infused MSCs in humans could be explained by their entrapment in capillaries of various tissues, including lungs. Because of the observed low osteopoietic cell engraftment, another study from the same group explored an ‘indirect’, collagen mutation-independent mechanism of MSC action in OI where they demonstrated that infused MSCs induced ‘the production of a soluble mediator by a second tissue that activates the proliferation of chondrocytes in the growth plate’ [Otsuru et al. 2012]. These two complementary mechanisms of MSC action in OI (direct and indirect) were also described in another study, although the researchers used fetal chorionic stem cells, not BM MSCs, in their preclinical investigation [Guillot et al. 2008].

Allogeneic fetal MSCs (fMSCs) rather than BM MSCs, have been also transplanted in two separate patients with OI (one with OI type III and one with OI type IV) at 32 and 31 weeks of gestation [Le Blanc et al. 2005]. Neither patient showed alloreactivity towards donor fMSCs or manifested any evidence of toxicity. Postnatal boosting with fMSCs resulted in low-level engraftment in bone but improved linear growth, mobility, and fracture incidence, possibly as a result of an indirect mechanism of MSC action [Gotherstrom et al. 2014].

Regarding OP treatment, local transplantation of autologous BM MSCs has been first assessed in an ovariectomized OP rabbit model. Four to eight weeks after transplantation, newly formed osteoids and enhanced trabecular thickness were evident [Wang et al. 2006b]. In allogeneic settings, when BM MSCs were transduced with receptor activator of nuclear factor κB Fc or bone morphogenetic protein 2 and locally transplanted in ovariectomized OP models, increased bone mineral density and mature bone formation were observed [Kim et al. 2006; Tang et al. 2008]. These preclinical studies indicated the potential of autologous or allogeneic BM MSCs to engraft in recipients’ bones contributing to improved bone mineral density and biomechanical stiffness.

Most recent preclinical studies for OP and OI cell therapies are outlined in Table 3. Some of these therapies use MSCs as vehicles to target miRNAs (noncoding single-stranded RNAs) involved in post-transcriptional expression regulation of bone-related genes. For example, Li and colleagues generated baculovirus-engineered rat BM MSCs expressing microRNA sponges to antagonize microRNA-140 and microRNA-214. Local transplantation of engineered MSCs in osteoporotic mice not only resulted in healing of critical sized bone defects but also in amelioration of bone quality [Li et al. 2016].

Table 3.

Recent preclinical studies using allogeneic BM MSCs/SSCs for the treatment of systemic bone diseases.

Cell/identity markers used Delivery Animal model Average follow up Outcome
Panaroni et al. [2009] Allogeneic GFP-labeled mouse MSCs/CD117, Sca-1, Lin- phenotype In utero Transplantation, 5 × 106 mouse MSCs per fetus Osteogenesis imperfecta BrtlIV+/– mouse model 2 months MSCs differentiated to osteoblasts in trabecular and cortical bones. MSCs synthesized up to 20% of all type I collagen in host bone. At 2 months, bone geometric parameters and biomechanical strength were significantly improved
Cho et al. [2009] Allogeneic RANK-Fc and CXC chemokine receptor 4 (CXCR4) cotransduced mouse MSCs/NR Systemic infusion, 6–7 × 105 mouse MSCs per mouse Osteoporotic model 8 weeks CXCR4 transduction promoted MSCs engraftment to bone. Increased bone mineral density and attenuated bone loss
Ovariectomized mice
Li et al. [2010] Allogeneic GFP-labeled mouse MSCs + type I collagen/CD11β, CD13, CD34, CD44, CD45, CD73, CD90, CD117, CD105, CD146 markers Local injection, 1 × 106 mouse MSCs per mouse Osteogenesis imperfecta oim–/– mouse model 2–6 weeks New bone formation. GFP-labeled MSCs were located close to osteoblasts. Injected bones were biomechanically stronger than noninjected bones
Ocarino Nde et al. [2010] Allogeneic rat MSCs/CD45, CD54, CD73, CD90 markers Local injection, 0.75 × 106 rat MSCs per rat Osteoporotic model 2 months Intrabone marrow injection of rat MSCs improved femur bone mass
Ovariectomized rats
Otsuru et al. [2012] Allogeneic GFP-labeled mouse MSCs/CD45, TER119, CD3, B220, CD11b, Gr-1, CD29, CD49e, CD90, CD105, Sca-1 markers Local injection, 1.0 × 106 mouse MSCs per mouse Osteogenesis imperfecta oim–/– mouse model 4 weeks MSCs did not substantially engraft in bone, but secreted a soluble mediator that indirectly stimulated bone growth
Pauley et al. [2014] Allogeneic Col2.3GFP-labeled mouse MSCs/NR Local transplantation, 5 × 106 Col2.3GFP-labeled mouse MSCs per mouse Osteogenesis imperfecta oim–/– mouse model of osteogenesis imperfecta type III 4 weeks MSC differentiation to Col2.3GFP+ osteoblasts was evident in trabecular and cortical bone. New bone formation observed close to Col2.3GFP+ osteoblasts. New bone had more organized collagen structure and type Iα2 collagen expression
Ma et al. [2015] Human MSCs/CD11β, CD14, CD35, CD45, CD73, CD90, CD105, CD146 markers Systemic infusion, 1 × 105 human MSCs per 10 g of mouse body weight Osteoporotic model 4 weeks Human MSCs improved functionally impaired recipient mouse MSCs via interleukin-17 suppression. Maintained regular positive bone metabolism by induction of osteoblasts and suppression of osteoclasts
MRL/lpr mice
Li et al. [2016] Allogeneic rat MSCs expressing microRNA sponges/NR Local implantation, 2 × 106 rat MSCs per rat Osteoporotic model 4 weeks MSCs expressing microRNA sponges healed critical size bone defect and ameliorated bone quality
Ovariectomized rats

BM, bone marrow; MSC, multipotential stromal cell; NR, not reported; RANK-Fc, receptor activator of nuclear factor κB Fc; SSC, skeletal stem cell.

In parallel to the development of cell-based therapies for the treatment of OP and OI, new-generation treatments are aimed at targeting native MSCs or their progeny in these diseases. As mentioned already, the enhancement of MSC anabolic (i.e. bone forming) ability can be achieved via in vivo delivery of miRNAs driving MSC differentiation to osteoblasts or by using miRNA ‘sponges’ to sequester and neutralize miRNAs controlling MSC differentiation to other lineages [Taipaleenmaki et al. 2011]. Other pharmacological interventions for OP treatment include targeting DLK1/Pref-1, a protein expressed by BM MSCs and involved in bone-fat homeostasis, or targeting other mediators such as Wnt pathway molecules in OP MSCs [Janeczek et al. 2015]. Monoclonal antibodies to molecular targets, such as sclerostin, primarily produced by osteocytes (terminally differentiated bone cells derived from MSCs) have also been investigated for the treatment of postmenopausal OP. In preclinical studies, a sclerostin neutralizing antibody increased bone formation, bone mineral density and bone strength [Li et al. 2009; Ominsky et al. 2010, 2011; Suen et al. 2015]. Two humanized antisclerostin monoclonal antibodies, romosozumab and blosozumab, have been shown to possess osteoanabolic properties with the potential to improve clinical outcomes in patients with OP in phase II clinical trials [McClung et al. 2014; Recknor et al. 2015]. With increased knowledge on the in vivo abnormalities in OP MSCs, new pharmacological agents (for example, Wnt pathway modifiers) or biological agents, similar to antisclerostin antibodies, to ‘correct’ their altered function in OP could be developed.

Targeting SSCs to reverse sclerotic bone formation and restore cartilage in osteoarthritis

Some musculoskeletal conditions are characterized by deranged bone formation or resorption processes, one example of which is osteoarthritis (OA). OA involves complex pathologies, which depend in many ways on the type of joint involved (hip, knee, etc.). Nevertheless, the loss of cartilage and subchondral bone abnormalities including bone marrow lesions (BMLs), represent the most common features of all types of OA. Bone marrow lesions represent mesenchymal tissue abnormalities within OA bone and are associated with OA structural progression and pain [Xu et al. 2012; Wenham and Conaghan, 2013]. Although the OA primary lesion may be indeed located in cartilage, abnormal joint loading and shock absorbance are most certainly transduced to the underlying bone, leading to excessive remodeling within the bone tissue and the subsequent formation of BMLs and osteophytes.

The initial appreciation of these processes in OA bone has firstly led to the proposed use of antiresorptive drugs such as bisphosphonates for OA treatment, with a potential to combine those with bone anabolic compounds such as calcitonin, strontium ranelate, parathyroid hormone, estrogens, or selective estrogen receptor modifiers [Kwan Tat et al. 2010; Karsdal et al. 2014]. Being the precursors of both bone and cartilage, MSCs have also attracted a considerable interest as cell and gene therapy modalities for OA (reviewed by Coleman and colleagues) [Coleman et al. 2010]. More recently, it has been recognized that OA MSCs may in fact contribute to abnormal remodeling processes in OA bone [Zhen et al. 2013]. Zhen and colleagues clearly demonstrated that the numbers of MSCs were dramatically increased in OA subchondral bone. Furthermore, the authors proposed that the altered biomechanical pressures on the OA joint have led to increased numbers of osteoclasts and transforming growth factor (TGF)-β1 release from the bone matrix, leading to the TGF-β1-induced MSC recruitment to the subchondral bone contributing to aberrant bone formation and disease progression [Zhen et al. 2013]. In agreement, we have recently found increased numbers and altered transcriptional profiles of CD271+ MSCs in BML areas of hip OA bone and showed their accumulation at the sites of damage [Campbell et al. 2016]. Based on these studies, new OA disease-modifying treatments to target abnormally activated signaling pathways in OA MSCs have been proposed [Zhen and Cao, 2014]. The timing and the mode of delivery of such future therapies remain to be established.

Conclusion

MSCs have been isolated from various tissues of the musculoskeletal system, including periosteum [De Bari et al. 2008], synovium [De Bari et al. 2001], tendon [Lui, 2013], joint fat pad [Wickham et al. 2003; English et al. 2007] and others. Although periosteal MSCs seeded on osteoconductive scaffolds have been shown to contain BM elements following implantation in vivo, only BM-resident MSCs possess homeostatic HSC-supporting niche properties without additional stimulation and in steady-state physiological conditions. This unique feature sets them apart from other tissue-resident MSCs making the use of a specific term, SSCs, justifiable. Not only have their stem cell nature and self-renewal ability been proven in vivo [Sacchetti et al. 2007], their native phenotypes and developmental origins [Bianco, 2011; Isern et al. 2014; Worthley et al. 2015] are now mapped much better compared with other types of MSCs.

There have been many reports on the potential use of adipose-derived MSCs for bone repair applications [Dragoo et al. 2003; Khan et al. 2009; Mesimaki et al. 2009]. However, it does appear more ‘natural’ to capitalize on native abilities of BM MSCs/SSCs to regulate bone formation and remodeling when considering the therapeutic use of MSCs for bone repair. Autologous bone grafting with uncultured SSCs surrounded by their native extracellular matrix (ECM) and other supportive cell populations remains the gold standard for repairing bone in orthopedics. Ex vivo expansion and implantation of cultured SSCs is safe and efficacious, however it may now be surpassed by newer, less laborious and less expensive treatments based on uncultured SSCs such as BM concentrates. Although encouraging clinical results have been obtained by transplanting either uncultured SSCs or cultured SSCs, the exact dosage and route of application remain to be optimized and the fate of transplanted cells and their mechanisms of action need to be better monitored in larger clinical trials. For the treatment of systemic bone diseases, the implantation of cultured or noncultured SSCs appears efficacious and newer approaches could involve targeting of SSCs in situ using pharmacological compounds and biologic treatments. Better understanding of the pivotal functional roles of SSCs in health, ageing and disease will provide novel and advanced therapeutic strategies for skeletal and joint disorders.

Acknowledgments

EJ and PVG are partly supported by NIHR-Leeds Musculoskeletal Biomedical Research Unit (LMBRU).

Footnotes

Funding: This review article received no grant from any funding body in the public, commercial, or non-profit sectors.

Conflict of interest statement: The authors declare that there is no conflict of interest.

Contributor Information

Elena A. Jones, Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James’s University Hospital, Room 5.24 Clinical Sciences Building, Leeds, West Yorkshire LS9 7TF, UK.

Peter V. Giannoudis, Academic Department of Trauma & Orthopaedic Surgery, University of Leeds, Leeds General Infirmary, Leeds, UK NIHR Leeds Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK

Dimitrios Kouroupis, Department of Biomedical Research, Foundation for Research and Technology-Hellas, Institute of Molecular Biology and Biotechnology, University Campus of Ioannina, Ioannina, Greece.

References

  1. Allende M., Tuymetova G., Lee B., Bonifacino E., Wu Y., Proia R. (2009) S1P1 receptor directs the release of immature B cells from bone marrow into blood. J Exp Med 207: 1113–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Arnsdorf E., Jones L., Carter D., Jacobs C. (2009) The periosteum as a cellular source for functional tissue engineering. Tissue Eng Part A 15: 2637–2642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baboolal T., Boxall S., El-Sherbiny Y., Moseley T., Cuthbert R., Giannoudis P., et al. (2014) Multipotential stromal cell abundance in cellular bone allograft: comparison with fresh age-matched iliac crest bone and bone marrow aspirate. Regenerative Med 9: 593–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Battula V., Treml S., Bareiss P., Gieseke F., Roelofs H., De Zwart P., et al. (2009) Isolation of functionally distinct mesenchymal stem cell subsets using antibodies against CD56, CD271, and mesenchymal stem cell antigen-1. Haematologica 94: 173–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bianco P. (2011) Back to the future: moving beyond ‘mesenchymal stem cells’. J Cell Biochem 112: 1713–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bianco P., Robey P. (2015) Skeletal stem cells. Development 142: 1023–1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Boettcher S., Ziegler P., Schmid M., Takizawa H., Van Rooijen N., Kopf M., et al. (2012) Cutting edge: LPS-induced emergency myelopoiesis depends on TLR4-expressing nonhematopoietic cells. J Immunol 188: 5824–5828. [DOI] [PubMed] [Google Scholar]
  8. Buhring H., Battula V., Treml S., Schewe B., Kanz L., Vogel W. (2007) Novel markers for the prospective isolation of human MSC. Ann N Y Acad Sci 1106: 262–271. [DOI] [PubMed] [Google Scholar]
  9. Campbell T., Churchman S., Gomez A., McGonagle D., Conaghan P., Ponchel F., et al. (2016) Mesenchymal stem cell alterations in bone marrow lesions in hip osteoarthritis. Arthritis Rheumatol 11 February (Epub ahead of print). [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Caplan A., Correa D. (2011) The MSC: an injury drugstore. Cell Stem Cell 9: 11–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cariappa A., Mazo I., Chase C., Shi H., Liu H., Li Q., et al. (2005) Perisinusoidal B cells in the bone marrow participate in T-independent responses to blood-borne microbes. Immunity 23: 397–407. [DOI] [PubMed] [Google Scholar]
  12. Cassatella M., Mosna F., Micheletti A., Lisi V., Tamassia N., Cont C., et al. (2011) Toll-like receptor-3-activated human mesenchymal stromal cells significantly prolong the survival and function of neutrophils. Stem Cells 29: 1001–1011. [DOI] [PubMed] [Google Scholar]
  13. Chan C., Seo E., Chen J., Lo D., Mcardle A., Sinha R., et al. (2015) Identification and specification of the mouse skeletal stem cell. Cell 160: 285–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cho S., Sun H., Yang J., Jung J., An J., Cho H., et al. (2009) Transplantation of mesenchymal stem cells overexpressing RANK-Fc or CXCR4 prevents bone loss in ovariectomized mice. Mol Ther 17: 1979–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Churchman S., Ponchel F., Boxall S., Cuthbert R., Kouroupis D., Roshdy T., et al. (2012) Transcriptional profile of native CD271+ multipotential stromal cells: evidence for multiple fates, with prominent osteogenic and Wnt pathway signaling activity. Arthritis Rheum 64: 2632–2643. [DOI] [PubMed] [Google Scholar]
  16. Coleman C., Curtin C., Barry F., O’Flatharta C., Murphy J. (2010) Mesenchymal stem cells and osteoarthritis: remedy or accomplice? Hum Gene Ther 21: 1239–1250. [DOI] [PubMed] [Google Scholar]
  17. Crowder S., Leonardo V., Whittaker T., Papathanasiou P., Stevens M. (2016) Material cues as potent regulators of epigenetics and stem cell function. Cell Stem Cell 18: 39–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cuthbert R., Boxall S., Tan H., Giannoudis P., McGonagle D., Jones E. (2012) Single-platform quality control assay to quantify multipotential stromal cells in bone marrow aspirates prior to bulk manufacture or direct therapeutic use. Cytotherapy 14: 431–440. [DOI] [PubMed] [Google Scholar]
  19. Dalby M., Gadegaard N., Tare R., Andar A., Riehle M., Herzyk P., et al. (2007) The control of human mesenchymal cell differentiation using nanoscale symmetry and disorder. Nat Mater 6: 997–1003. [DOI] [PubMed] [Google Scholar]
  20. De Bari C., Dell’Accio F., Karystinou A., Guillot P., Fisk N., Jones E., et al. (2008) A biomarker-based mathematical model to predict bone-forming potency of human synovial and periosteal mesenchymal stem cells. Arthritis Rheum 58: 240–250. [DOI] [PubMed] [Google Scholar]
  21. De Bari C., Dell’Accio F., Tylzanowski P., Luyten F. (2001) Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum 44: 1928. [DOI] [PubMed] [Google Scholar]
  22. De Bari C., Dell’Accio F., Vanlauwe J., Eyckmans J., Khan I., Archer C., et al. (2006) Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum 54: 1209–1221. [DOI] [PubMed] [Google Scholar]
  23. Dhillon R., Schwarz E., Maloney M. (2012) Platelet-rich plasma therapy – future or trend? Arthritis Res Ther 14: 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Digirolamo C., Stokes D., Colter D., Phinney D., Class R., Prockop D. (1999) Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. Br J Haematol 107: 275–281. [DOI] [PubMed] [Google Scholar]
  25. Dominici M., Le Blanc K., Mueller I., Slaper-Cortenbach I., Marini F., Krause D., et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 8: 315–317. [DOI] [PubMed] [Google Scholar]
  26. Dragoo J., Choi J., Lieberman J., Huang J., Zuk P., Zhang J., et al. (2003) Bone induction by BMP-2 transduced stem cells derived from human fat. J Orthop Res 21: 622–629. [DOI] [PubMed] [Google Scholar]
  27. Dzierzak E., Speck N. (2008) Of lineage and legacy – the development of mammalian hematopoietic stem cells. Nat Immunol 9: 129–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. English A., Jones E., Corscadden D., Henshaw K., Chapman T., Emery P., et al. (2007) A comparative assessment of cartilage and joint fat pad as a potential source of cells for autologous therapy development in knee osteoarthritis. Rheumatology 46: 1676–1683. [DOI] [PubMed] [Google Scholar]
  29. Friedenstein A., Chailakhyan R., Latsinik N., Panasyuk A., Keiliss-Borok I. (1974) Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation 17: 331–340. [DOI] [PubMed] [Google Scholar]
  30. Garcia-Garcia A., De Castillejo C., Mendez-Ferrer S. (2015) BMSCs and hematopoiesis. Immunol Lett 168: 129–135. [DOI] [PubMed] [Google Scholar]
  31. Gaudet J., Ribot E., Chen Y., Gilbert K., Foster P. (2015) Tracking the fate of stem cell implants with fluorine-19 MRI. PLoS ONE 10: e0118544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Giannotti S., Trombi L., Bottai V., Ghilardi M., D’Alessandro D., Danti S., et al. (2013) Use of Autologous Human Mesenchymal Stromal Cell/Fibrin Clot Constructs in Upper Limb Non-Unions: Long-Term Assessment. PLoS ONE 8: e73893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Giannoudis P., Einhorn T., Marsh D. (2007) Fracture healing: the diamond concept. Injury 38(Suppl. 4): S3–S6. [DOI] [PubMed] [Google Scholar]
  34. Gonshor A., McAllister B., Wallace S., Prasad H. (2011) Histologic and histomorphometric evaluation of an allograft stem cell-based matrix sinus augmentation procedure. Int J Oral Maxillofacial Implants 26: 123–131. [PubMed] [Google Scholar]
  35. Gotherstrom C., Westgren M., Shaw S., Astrom E., Biswas A., Byers P., et al. (2014) Pre- and postnatal transplantation of fetal mesenchymal stem cells in osteogenesis imperfecta: a two-center experience. Stem Cells Transl Med 3: 255–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Guillot P., Abass O., Bassett J., Shefelbine S., Bou-Gharios G., Chan J., et al. (2008) Intrauterine transplantation of human fetal mesenchymal stem cells from first-trimester blood repairs bone and reduces fractures in osteogenesis impertecta mice. Blood 111: 1717–1725. [DOI] [PubMed] [Google Scholar]
  37. Guo Y., Su L., Wu J., Zhang D., Zhang X., Zhang G., et al. (2012) Assessment of the green florescence protein labeling method for tracking implanted mesenchymal stem cells. Cytotechnology 64: 391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Hernigou P., Beaujean F. (2002) Treatment of osteonecrosis with autologous bone marrow grafting. Clin Orthop Relat Res (405): 14–23. [DOI] [PubMed] [Google Scholar]
  39. Hernigou P., Flouzat Lachaniette C., Delambre J., Zilber S., Duffiet P., Chevallier N., et al. (2014) Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. Int Orthop 38: 1811–1818. [DOI] [PubMed] [Google Scholar]
  40. Hernigou P., Guissou I., Homma Y., Poignard A., Chevallier N., Rouard H., et al. (2015) Percutaneous injection of bone marrow mesenchymal stem cells for ankle non-unions decreases complications in patients with diabetes. Int Orthop 39: 1639–1643. [DOI] [PubMed] [Google Scholar]
  41. Hernigou P., Homma Y., Flouzat-Lachaniette C., Poignard A., Chevallier N., Rouard H. (2013) Cancer risk is not increased in patients treated for orthopaedic diseases with autologous bone marrow cell concentrate. J Bone Joint Surg 95: 2215–2221. [DOI] [PubMed] [Google Scholar]
  42. Hernigou P., Poignard A., Lachaniette C. (2012) Treatment of corticosteroid hip osteonecrosis with stem cells. J Bone Joint Surg 94-B: 68. [Google Scholar]
  43. Hernigou P., Poignard A., Zilber S., Rouard H. (2009) Cell therapy of hip osteonecrosis with autologous bone marrow grafting. Ind J Orthop 43: 40–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Hollawell S. (2012) Allograft cellular bone matrix as an alternative to autograft in hindfoot and ankle fusion procedures. J Foot Ankle Surg 51: 222–225. [DOI] [PubMed] [Google Scholar]
  45. Horwitz E., Gordon P., Koo W., Marx J., Neel M., McNall R., et al. (2002) Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl Acad Sci USA 99: 8932–8937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Houlihan D., Mabuchi Y., Morikawa S., Niibe K., Araki D., Suzuki S., et al. (2012) Isolation of mouse mesenchymal stem cells on the basis of expression of SCA-1 and PDGFR-α. Nat Prot 7: 2103–2111. [DOI] [PubMed] [Google Scholar]
  47. Isern J., Garcia-Garcia A., Martin A., Arranz L., Martin-Perez D., Torroja C., et al. (2014) The neural crest is a source of mesenchymal stem cells with specialized hematopoietic stem cell niche function. eLife 3: e03696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Jäger M., Herten M., Fochtmann U., Fischer J., Hernigou P., Zilkens C., et al. (2011) Bridging the gap: bone marrow aspiration concentrate reduces autologous bone grafting in osseous defects. J Orthop Res 29: 173–180. [DOI] [PubMed] [Google Scholar]
  49. James S., Fox J., Afsari F., Lee J., Clough S., Knight C., et al. (2015) Multiparameter analysis of human bone marrow stromal cells identifies distinct immunomodulatory and differentiation-competent subtypes. Stem Cell Rep 4: 1004–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Janeczek A., Tare R., Scarpa E., Moreno-Jimenez I., Rowland C., Jenner D., et al. (2015) Transient canonical Wnt stimulation enriches human bone marrow mononuclear cell isolates for osteoprogenitors. Stem Cells 34: 418–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Jones E., Kinsey S., English A., Jones R., Straszynski L., Meredith D., et al. (2002) Isolation and characterization of bone marrow multipotential mesenchymal progenitor cells. Arthritis Rheum 46: 3349–3360. [DOI] [PubMed] [Google Scholar]
  52. Karsdal M., Bay-Jensen A., Lories R., Abramson S., Spector T., Pastoureau P., et al. (2014) The coupling of bone and cartilage turnover in osteoarthritis: opportunities for bone antiresorptives and anabolics as potential treatments? Ann Rheum Dis 73: 336–348. [DOI] [PubMed] [Google Scholar]
  53. Kassem M., Bianco P. (2015) Skeletal stem cells in space and time. Cell 160: 17–19. [DOI] [PubMed] [Google Scholar]
  54. Kerr E., 3rd, Jawahar A., Wooten T., Kay S., Cavanaugh D., Nunley P. (2011) The use of osteo-conductive stem-cells allograft in lumbar interbody fusion procedures: an alternative to recombinant human bone morphogenetic protein. J Surg Orthop Adv 20: 193–197. [PubMed] [Google Scholar]
  55. Khan W., Adesida A., Tew S., Andrew J., Hardingham T. (2009) The epitope characterisation and the osteogenic differentiation potential of human fat pad-derived stem cells is maintained with ageing in later life. Injury 40: 150–157. [DOI] [PubMed] [Google Scholar]
  56. Kim D., Cho S., Her S., Yang J., Kim S., Kim S., et al. (2006) Retrovirus-mediated gene transfer of receptor activator of nuclear factor-kB-Fc prevents bone loss in ovariectomized mice. Stem Cells 24: 1798–1805. [DOI] [PubMed] [Google Scholar]
  57. Kim S., Shin Y., Yang K., Kim S., Yoo M., Han S., et al. (2009) A multi-center, randomized, clinical study to compare the effect and safety of autologous cultured osteoblast (Ossrontm) injection to treat fractures. BMC Musculoskel Disord 10: 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Komada Y., Yamane T., Kadota D., Isono K., Takakura N., Hayashi S., et al. (2012) Origins and properties of dental, thymic, and bone marrow mesenchymal cells and their stem cells. PLoS One 7: e46436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Kwan Tat S., Lajeunesse D., Pelletier J., Martel-Pelletier J. (2010) Targeting subchondral bone for treating osteoarthritis: what is the evidence? Best Pract Res Clin Rheumatol 24: 51–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Le Blanc K., Davies L. (2015) Mesenchymal stromal cells and the innate immune response. Immunol Lett 168: 140–146. [DOI] [PubMed] [Google Scholar]
  61. Le Blanc K., Gotherstrom C., Ringden O., Hassan M., McMahon R., Horwitz E., et al. (2005) Fetal mesenchymal stem-cell engraftment in bone after in utero transplantation in a patient with severe osteogenesis imperfecta. Transplantation 79: 1607–1614. [DOI] [PubMed] [Google Scholar]
  62. Li F., Wang X., Niyibizi C. (2007) Distribution of single-cell expanded marrow derived progenitors in a developing mouse model of osteogenesis imperfecta following systemic transplantation. Stem Cells 25: 3183–3193. [DOI] [PubMed] [Google Scholar]
  63. Li F., Wang X., Niyibizi C. (2010) Bone marrow stromal cells contribute to bone formation following infusion into femoral cavities of a mouse model of osteogenesis imperfecta. Bone 47: 546–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Li H., Ghazanfari R., Zacharaki D., Ditzel N., Isern J., Ekblom M., et al. (2014) Low/negative expression of PDGFR-α identifies the candidate primary mesenchymal stromal cells in adult human bone marrow. Stem Cell Reports 3: 965–974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Li K., Chang Y., Yeh C., Hu Y. (2016) Healing of osteoporotic bone defects by baculovirus-engineered bone marrow-derived MSCs expressing MicroRNA sponges. Biomaterials 74: 155–166. [DOI] [PubMed] [Google Scholar]
  66. Li X., Ominsky M., Warmington K., Morony S., Gong J., Cao J., et al. (2009) Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J Bone Min Res 24: 578–588. [DOI] [PubMed] [Google Scholar]
  67. Liotta F., Angeli R., Cosmi L., Fili L., Manuelli C., Frosali F., et al. (2008) Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing notch signaling. Stem Cells 26: 279–289. [DOI] [PubMed] [Google Scholar]
  68. Loebel C., Czekanska E., Bruderer M., Salzmann G., Alini M., Stoddart M. (2015) In vitro osteogenic potential of human mesenchymal stem cells is predicted by Runx2/Sox9 ratio. Tissue Eng Part A 21: 115–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Lui P. (2013) Identity of tendon stem cells – how much do we know? J Cell Mol Med 17: 55–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ma L., Aijima R., Hoshino Y., Yamaza H., Tomoda E., Tanaka Y., et al. (2015) Transplantation of mesenchymal stem cells ameliorates secondary osteoporosis through interleukin-17-impaired functions of recipient bone marrow mesenchymal stem cells in MRL/lpr mice. Stem Cell Res Ther 6: 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Marcacci M., Kon E., Moukhachev V., Lavroukov A., Kutepov S., Quarto R., et al. (2007) Stem cells associated with macroporous bioceramics for long bone repair: 6-to 7-year outcome of a pilot clinical study. Tissue Eng 13: 947–955. [DOI] [PubMed] [Google Scholar]
  72. Mathiasen A., Hansen L., Friis T., Thomsen C., Bhakoo K., Kastrup J. (2013) Optimal labeling dose, labeling time, and magnetic resonance imaging detection limits of ultrasmall superparamagnetic iron-oxide nanoparticle labeled mesenchymal stromal cells. Stem Cells Int 2013: 353105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. McClung M., Grauer A., Boonen S., Bolognese M., Brown J., Diez-Perez A., et al. (2014) Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med 370: 412–420. [DOI] [PubMed] [Google Scholar]
  74. Mesimaki K., Lindroos B., Tornwall J., Mauno J., Lindqvist C., Kontio R., et al. (2009) Novel maxillary reconstruction with ectopic bone formation by GMP adipose stem cells. Int J Oral Maxillofac Surg 38: 201–209. [DOI] [PubMed] [Google Scholar]
  75. Miura Y., Gao Z., Miura M., Seo B., Sonoyama W., Chen W., et al. (2006) Mesenchymal stem cell-organized bone marrow elements: an alternative hematopoietic progenitor resource. Stem Cells 24: 2428–2436. [DOI] [PubMed] [Google Scholar]
  76. Morikawa S., Mabuchi Y., Kubota Y., Nagai Y., Niibe K., Hiratsu E., et al. (2009) Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med 206: 2483–2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Muraglia A., Cancedda R., Quarto R. (2000) Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci 113: 1161–1166. [DOI] [PubMed] [Google Scholar]
  78. Nagoshi N., Shibata S., Kubota Y., Nakamura M., Nagai Y., Satoh E., et al. (2008) Ontogeny and multipotency of neural crest-derived stem cells in mouse bone marrow, dorsal root ganglia, and whisker pad. Cell Stem Cell 2: 392–403. [DOI] [PubMed] [Google Scholar]
  79. Nagoshi N., Shibata S., Nakamura M., Matsuzaki Y., Toyama Y., Okano H. (2009) Neural crest-derived stem cells display a wide variety of characteristics. J Cell Biochem 107: 1046–1052. [DOI] [PubMed] [Google Scholar]
  80. Ocarino Nde M., Boeloni J., Jorgetti V., Gomes D., Goes A., Serakides R. (2010) Intra-bone marrow injection of mesenchymal stem cells improves the femur bone mass of osteoporotic female rats. Connect Tissue Res 51: 426–433. [DOI] [PubMed] [Google Scholar]
  81. Ominsky M., Li C., Li X., Tan H., Lee E., Barrero M., et al. (2011) Inhibition of sclerostin by monoclonal antibody enhances bone healing and improves bone density and strength of nonfractured bones. J Bone Miner Res 26: 1012–1021. [DOI] [PubMed] [Google Scholar]
  82. Ominsky M., Vlasseros F., Jolette J., Smith S., Stouch B., Doellgast G., et al. (2010) Two doses of sclerostin antibody in cynomolgus monkeys increases bone formation, bone mineral density, and bone strength. J Bone Miner Res 25: 948–959. [DOI] [PubMed] [Google Scholar]
  83. Ono N., Ono W., Nagasawa T., Kronenberg H. (2014) A subset of chondrogenic cells provides early mesenchymal progenitors in growing bones. Nat Cell Biol 16: 1157–1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Oreffo R., Cooper C., Mason C., Clements M. (2005) Mesenchymal stem cells. Stem Cell Rev 1: 169–178. [DOI] [PubMed] [Google Scholar]
  85. Otsuru S., Gordon P., Shimono K., Jethva R., Marino R., Phillips C., et al. (2012) Transplanted bone marrow mononuclear cells and MSCs impart clinical benefit to children with osteogenesis imperfecta through different mechanisms. Blood 120: 1933–1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Panaroni C., Gioia R., Lupi A., Besio R., Goldstein S., Kreider J., et al. (2009) In utero transplantation of adult bone marrow decreases perinatal lethality and rescues the bone phenotype in the knockin murine model for classical, dominant osteogenesis imperfecta. Blood 114: 459–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Pauley P., Matthews B., Wang L., Dyment N., Matic I., Rowe D., et al. (2014) Local transplantation is an effective method for cell delivery in the osteogenesis imperfecta murine model. Int Orthop 38: 1955–1962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Pereira R., O’Hara M., Laptev A., Halford K., Pollard M., Class R., et al. (1998) Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta. Proc Natl Acad Sci USA 95: 1142–1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Quarto R., Mastrogiacomo M., Cancedda R., Kutepov S., Mukhachev V., Lavroukov A., et al. (2001) Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med 344: 385–386. [DOI] [PubMed] [Google Scholar]
  90. Recknor C., Recker R., Benson C., Robins D., Chiang A., Alam J., et al. (2015) The effect of discontinuing treatment with blosozumab: follow-up results of a phase 2 randomized clinical trial in postmenopausal women with low bone mineral density. J Bone Miner Res 30: 1717–1725. [DOI] [PubMed] [Google Scholar]
  91. Roberts S., Van Gastel N., Carmeliet G., Luyten F. (2015) Uncovering the periosteum for skeletal regeneration: the stem cell that lies beneath. Bone 70: 10–18. [DOI] [PubMed] [Google Scholar]
  92. Sacchetti B., Funari A., Michienzi S., Di Cesare S., Piersanti S., Saggio I., et al. (2007) Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 131: 324–326. [DOI] [PubMed] [Google Scholar]
  93. Satomura K., Derubeis A., Fedarko N., Ibaraki-O’Connor K., Kuznetsov S., Rowe D., et al. (1998) Receptor tyrosine kinase expression in human bone marrow stromal cells. J Cell Physiol 177: 426–438. [DOI] [PubMed] [Google Scholar]
  94. Scott R., Hyer C. (2013) Role of cellular allograft containing mesenchymal stem cells in high-risk foot and ankle reconstructions. J Foot Ankle Surg 52: 32–35. [DOI] [PubMed] [Google Scholar]
  95. Suen P., Zhu T., Chow D., Huang L., Zheng L., Qin L. (2015) Sclerostin antibody treatment increases bone formation, bone mass, and bone strength of intact bones in adult male rats. Sci Rep 5: 15632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Taipaleenmaki H., Bjerre Hokland L., Chen L., Kauppinen S., Kassem M. (2011) Mechanisms in endocrinology: micro-RNAs: targets for enhancing osteoblast differentiation and bone formation. Eur J Endocrinol 166: 359–371. [DOI] [PubMed] [Google Scholar]
  97. Tang Y., Tang W., Lin Y., Long J., Wang H., Liu L., et al. (2008) Combination of bone tissue engineering and bmp-2 gene transfection promotes bone healing in osteoporotic rats. Cell Biol Int 32: 1150–1157. [DOI] [PubMed] [Google Scholar]
  98. Thompson E., Matsiko A., Farrell E., Kelly D., O’Brien F. (2015) Recapitulating endochondral ossification: a promising route to in vivo bone regeneration. J Tissue Eng Regen Med 9: 889–902. [DOI] [PubMed] [Google Scholar]
  99. Tohmeh A., Watson B., Tohmeh M., Zielinski X. (2012) Allograft cellular bone matrix in extreme lateral interbody fusion: preliminary radiographic and clinical outcomes. Sci World J 2012: 263637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Tormin A., Li O., Brune J., Walsh S., Schutz B., Ehinger M., et al. (2011) CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization. Blood 117: 5067–5077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Twine N., Chen L., Pang C., Wilkins M., Kassem M. (2014) Identification of differentiation-stage specific markers that define the ex vivo osteoblastic phenotype. Bone 67: 23–32. [DOI] [PubMed] [Google Scholar]
  102. Wang X., Li F., Niyibizi C. (2006a) Progenitors systemically transplanted into neonatal mice localize to areas of active bone formation in vivo: implications of cell therapy for skeletal diseases. Stem Cells 24: 1869–1878. [DOI] [PubMed] [Google Scholar]
  103. Wang Z., Goh J., Das De S., Ge Z., Ouyang H., Chong J., et al. (2006b) Efficacy of bone marrow-derived stem cells in strengthening osteoporotic bone in a rabbit model. Tissue Eng 12: 1753–1761. [DOI] [PubMed] [Google Scholar]
  104. Welner R., Kincade P. (2014) 9-1-1: HSCs respond to emergency calls. Cell Stem Cell 14: 415–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Wenham C., Conaghan P. (2013) New horizons in osteoarthritis. Age Ageing 42: 272–278. [DOI] [PubMed] [Google Scholar]
  106. Wickham M., Erickson G., Gimble J., Vail T., Guilak F. (2003) Multipotent stromal cells derived from the infrapatellar fat pad of the knee. Clin Orthop Rel Res 412: 196–212. [DOI] [PubMed] [Google Scholar]
  107. Woodell-May J., Tan M., King W., Swift M., Welch Z., Murphy M., et al. (2015) Characterization of the cellular output of a point-of-care device and the implications for addressing critical limb ischemia. BioRes Open Access 4: 417–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Worthley D., Churchill M., Compton J., Tailor Y., Rao M., Si Y., et al. (2015) Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential. Cell 160: 269–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Xu L., Hayashi D., Roemer F., Felson D., Guermazi A. (2012) Magnetic resonance imaging of subchondral bone marrow lesions in association with osteoarthritis. Semin Arthritis Rheum 42: 105–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Yamaza T., Miura Y., Akiyama K., Bi Y., Sonoyama W., Gronthos S., et al. (2009) Mesenchymal stem cell-mediated ectopic hematopoiesis alleviates aging-related phenotype in immunocompromised mice. Blood 113: 2595–2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Zhao D., Cui D., Wang B., Tian F., Guo L., Yang L., et al. (2012) Treatment of early stage osteonecrosis of the femoral head with autologous implantation of bone marrow-derived and cultured mesenchymal stem cells. Bone 50: 325–330. [DOI] [PubMed] [Google Scholar]
  112. Zhen G., Cao X. (2014) Targeting TGFβ signaling in subchondral bone and articular cartilage homeostasis. Trends Pharmacol Sci 35: 227–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Zhen G., Wen C., Jia X., Li Y., Crane J., Mears S., et al. (2013) Inhibition of TGFβ signaling in subchondral bone mesenchymal stem cells attenuates osteoarthritis. Nat Med 19: 704–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Zhou B., Yue R., Murphy M., Peyer J., Morrison S. (2014) Leptin receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell 15: 154–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Ziegler P., Boettcher S., Takizawa H., Manz M., Brummendorf T. (2015) LPS-stimulated human bone marrow stroma cells support myeloid cell development and progenitor cell maintenance. Ann Hematol 95: 173–178. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Musculoskeletal Disease are provided here courtesy of SAGE Publications

RESOURCES