Next Article in Journal
Novel Small Molecule Inhibitors That Prevent the Neuroparalysis of Tetanus Neurotoxin
Next Article in Special Issue
Adipose Tissue-Derived Mesenchymal Stem Cells as a Potential Restorative Treatment for Cartilage Defects: A PRISMA Review and Meta-Analysis
Previous Article in Journal
Thymoquinone Potentiates the Effect of Phenytoin against Electroshock-Induced Convulsions in Rats by Reducing the Hyperactivation of m-TOR Pathway and Neuroinflammation: Evidence from In Vivo, In Vitro and Computational Studies
Previous Article in Special Issue
Human Salivary Histatin-1-Functionalized Gelatin Methacrylate Hydrogels Promote the Regeneration of Cartilage and Subchondral Bone in Temporomandibular Joints
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Osteogenic and Chondrogenic Potential of Periosteum-Derived Mesenchymal Stromal Cells: Do They Hold the Key to the Future?

by
Madhan Jeyaraman
1,2,3,
Sathish Muthu
2,3,4,*,
Prakash Gangadaran
5,6,
Rajni Ranjan
1,
Naveen Jeyaraman
7,
Gollahalli Shivashankar Prajwal
8,
Prabhu Chandra Mishra
3,
Ramya Lakshmi Rajendran
6,* and
Byeong-Cheol Ahn
5,6,*
1
Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201306, Uttar Pradesh, India
2
Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
3
International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India
4
Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, Tamil Nadu, India
5
BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
6
Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
7
Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli 620002, Tamil Nadu, India
8
Department of Orthopaedics, Mallika Spine Centre, Guntur 522001, Andhra Pradesh, India
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(11), 1133; https://doi.org/10.3390/ph14111133
Submission received: 12 October 2021 / Revised: 2 November 2021 / Accepted: 4 November 2021 / Published: 8 November 2021
(This article belongs to the Special Issue Cell-Based Therapies for Bone and Cartilage Regeneration)

Abstract

:
The periosteum, with its outer fibrous and inner cambium layer, lies in a dynamic environment with a niche of pluripotent stem cells for their reparative needs. The inner cambium layer is rich in mesenchymal progenitors, osteogenic progenitors, osteoblasts, and fibroblasts in a scant collagen matrix environment. Their role in union and remodeling of fracture is well known. However, the periosteum as a source of mesenchymal stem cells has not been explored in detail. Moreover, with the continuous expansion of techniques, newer insights have been acquired into the roles and regulation of these periosteal cells. From a therapeutic standpoint, the periosteum as a source of tissue engineering has gained much attraction. Apart from its role in bone repair, analysis of the bone-forming potential of periosteum-derived stem cells is lacking. Hence, this article elucidates the role of the periosteum as a potential source of mesenchymal stem cells along with their capacity for osteogenic and chondrogenic differentiation for therapeutic application in the future.

1. Introduction

The periosteum is a thin fibrous layer, which forms an outer covering of the bone surface. It contains the outer fibrous layer and inner cambium layer, which possess significant osteogenic potential [1,2]. In 1867, Ollier claimed that the cambium layer is responsible for appositional bone growth [3]. The periosteum lies in a dynamic mechanically loaded environment and provides a niche for pluripotent stem cells [2,4]. The outer fibrous layer contains a superficial inelastic hypocellular layer with high content of the collagenous matrix, which contributes a rich vascular supply to the bone and skeletal muscle and provides a network of neural fibers, whereas the deep elastic hypocellular hypovascular layer features a high-collagen matrix, as shown in Figure 1 [5]. The inner cambium layer is hypercellular with mesenchymal progenitors, osteogenic progenitors, osteoblasts, and fibroblasts in fewer quantities in the collagen matrix [6]. The cambium layer is rich in vascularity and neural networks. The presence of pericytes in the cambium layer confers the periosteum with more osteoblastic potential.1 The regenerative potential of the periosteum differs with age and bony location [6].

2. Periosteum as MSC Source

The cambium layer of the periosteum contains mesenchymal progenitor cells which can be extrapolated as periosteum-derived MSCs (P-MSCs) [7,8,9]. Various studies have stated that the periosteum is the best cellular therapeutic agent for bone regeneration due to the multipotent nature at the single-cell level, higher proliferation and differentiation rate, and retention of differentiation ability in vitro and in vivo [10,11]. P-MSCs from load-bearing sites possess more osteogenic potential than flat bones [12]. In cases of fractures, quiescent P-MSCs induce chondrogenesis and osteogenesis, which helps in long-term integration with native bone [13,14].
Molecular analysis revealed the periostin gene, which is responsible for enhanced tissue response to injury of periosteum [15]. Duchamp de Lageneste et al. demonstrated the higher regenerative potential of periosteal cells than bone marrow-derived MSCs (BM-MSCs). They observed that P-MSCs integrate into callus and cartilage by day 10 [16]. Lineage analysis of P-MSCs demonstrated that P-MSCs are derived from the Prx-1 mesenchymal lineage, which contributes to cartilage and bone within the callus [16].

3. Characterization and Isolation of P-MSCs

P-MSCs exhibit multilineage differentiation potential both in vitro and in vivo. P-MSCs exert the regenerative potential by possessing the effects of paracrine signaling, direct cell–cell interactions, and extracellular vehicles [17,18,19]. They possess surface antigens such as MSC markers (CD-73, -90, and -105, MSCA-1, CD-9 and -13, STRO-1, SSEA-4, ScaI, SOX-2, Oct-4, and Nanog) [20,21,22], integrin markers (CD-29 and -49e) [23,24], adhesion molecules (CD-31, -44, -166, -54, and -146) [25,26], and MHC class markers (HLA-ABC) [27], whereas they lack HSC markers (CD-14, -33, -34, 45, and -133, and HLA-DR) [28].
Frey et al. demonstrated the immunohistochemical and molecular characterization of P-MSCs in periosteal tissue samples [29]. They identified the presence of osteoblasts (ALP, M-CSF, Cbfa-1/Runx2, RANK-L, and Osterix), osteoclasts (TRAP, CTR, and cathepsin K), chondrocytes (SOX-9), and dendritic cells (MHC-II) in the periosteum. Yang et al. identified discoidal domain receptor-2 (DDR2), a novel marker for isolating osteoblasts and osteoblast progenitors of the periosteum [30]. Deveza et al. identified positive markers (Mx1Cre, ROSATomato, and OsteocalcinGFP) and negative markers (Mx1+Ocn−) of periosteal progenitor cells in mice [31].
Mx1 and α-smooth muscle actin are potent markers of quiescent skeletal stem cells in adult periosteal tissues [32]. Gao et al. evaluated the potentialities of periosteal progenitor cells and BM-MSCs using FACS Nestin+ PDGFR-α+ CD45 Ter119 CD31 LepR+ markers. They demonstrated that both Nestin+ PDGFR-α+ and LepR+ periosteal progenitor cells formed more colony-forming unit fibroblasts (CFU-Fs) than BM-MSCs [33]. Craniofacial-derived periosteal progenitor cells show a similar growth curve to tibial-derived periosteal progenitor cells. Craniofacial human periosteum-derived cells (hPDCs) are positive for skeletal stem and progenitor cell markers CD73, CD164, and podoplanin and negative for CD146, HSC, and endothelial lineage markers [34].
Olbrich et al. obtained human periosteal samples from the maxilla and mandible bones [35]. Those samples were subjected to type 9 collagenase for 90 min and were plated onto 75 cm2 culture flasks. Periosteal tissues were cultured in a DMEM/F-12 medium containing antimicrobials. The proliferated periosteal tissues from the fifth to seventh passages were used for further experiments. Hence, magnetic separation (MACS) is a more suitable separation method to isolate osteoprogenitors from the entire jaw periosteal cell population.
From human donors, a 1 cm2 proximal medial tibial segment was removed, and the periosteum was harvested. Periosteal tissues were plated onto the serum-free α-MEM medium. Further tissue digestion was performed using collagenase D enzyme for 1 h and resuspended in α-MEM medium containing antimicrobial solutions. Cells were allowed to grow to 80% confluence, collected from the second and third passages for a lineage differentiation assay, and evaluated for stem-cell surface markers. De Bari et al. stated that, regardless of donor age, the adult human periosteum contains cells that, upon enzymatic release and culture expansion, are multipotent MSCs at the single-cell level [36].

4. Intracellular Signaling in Osteogenic Differentiation

Osteogenesis plays a major role in bone formation and turnover, fracture healing, and osseointegration of implants. Osteogenesis is an interplay among osteoprogenitor cells, osteoblasts, osteocytes, and osteoclasts. Osteogenic differentiation is regulated by various signaling pathways governed by osteoprogenitor markers (ALP, collagen type-1 and -2, decorin, Runx-2/CBFA-1, osterix, CD-146/M-CAM, and matrix extracellular phosphoglycoprotein (MEPE)), osteoblast markers (ALP, BAP-1 and -31, collagen type 1, fibronectin, osteocalcin, osterix, and SPARC), and osteocyte markers (biglycan, DMP-1, FGF-23, fibronectin, MEPE, podoplanin, sclerostin, and SPARC).
Temporary hypoxia exposure to MSCs leads to downregulation of type 1 collagen and Runx-2, and upregulation of OPN, which limits the in vivo osteogenic potential of MSCs [37]. More than 48 h of exposure to hypoxia in vitro exhibited MSC apoptosis when transplanted in vivo. Hence, the duration of in vitro hypoxia plays a major role in osteogenesis and osteoblastic differentiation when transplanted in vivo. Hypoxia-induced MSC-bound osteogenesis is primarily due to the presence of hypoxia-inducible factor 1α (HIF1α) with downregulation of Cbf-α1 [38]. Hypoxia upregulates phosphorylation of STAT-3 and expression of VEGF in MSCs. Downregulation of STAT-3 signaling results in dysfunctional osteogenesis in vitro [39].
Cheng et al. demonstrated osteogenic enhancement and adipogenic suppression of mesenchymal progenitors by Msx-2 [40]. KIAA1199 is a cell migration-inducing protein (CMIP), which helps in migration and mobilization of osteoprogenitor cells in vivo. KIAA1199 interacts with the wingless type (Wnt) signaling pathway, which induces osteogenesis and bone remodeling [41]. Wu et al. observed that overexpression of IGF-binding protein 4 (IGFBP-4) inhibits osteogenic differentiation in rat MSCs [42]. Upregulation of retinoic acid signaling inhibits the activity of ALP, leads to mineralization of dental pulp MSCs, and decreases the expression of factors inducing osteogenesis [43]. Extracellular matrix proteins possess varying affinity (fibronectin > type 1 and 4 collagen ≥ vitronectin > laminin-1) for the osteogenic differentiation of MSCs [44].
Suehiro et al. found that zinc finger and homeobox-3 (ZHX-3) helps in the switching of undifferentiated MSCs to in vitro osteogenic differentiation [45]. A forkhead transcription factor (FOXO) provides bidirectional regulation of MSC differentiation toward osteogenic cells, with positive regulators being Runx-2, ALP, PCN, and ATF-4 and negative regulators being β-catenin and OCN [46]. Differential expression of miRNAs is implicated in proliferation, differentiation, oncogenesis, and stemness of MSCs. The miR-31 exosome is the key regulator of osterix, a bone-specific transcription factor, which modulates osteogenesis and osteobiology [47].
Homeobox genes are well documented in the literature for intracellular signaling for osteogenesis. Homeobox C10 (HOX-C10) downregulates in vitro osteogenic differentiation [22,48], whereas distal less homeobox (DLX)-2 [49] and -5 [50] and HOX-B7 [51] upregulate osteogenic differentiation. The trimethylation of lysine 4 of histone H3 (H3K4Me3) correlates with osteogenic differentiation [52]. WD repeat-containing protein 63 (WDR63) enhances ALP activity, mineralization of osteoid, and the expression of BSP, OSX, and RUNX2 in vitro, as well as osteogenesis in nude mice [52].
Long noncoding RNAs (lncRNAs), considered “transcriptional noise”, emerged as key genomic regulators in cellular and tissue engineering. lncRNAs regulate osteogenic differentiation by binding to transcription factors and chromatin modification. The various mechanisms of osteogenesis by lncRNAs are as follows: (a) maternally expressed gene-3 (MEG-3) dissociates SOX-2 and promotes MSC-bound osteogenesis by enhancing BMP-4 expression, (b) phosphorylation of IκBα and activation of NF-κB inhibits osteogenesis, (c) downregulation of HOXA-AS3 enhances osteogenesis by targeting EZH2, and (d) upregulation of HIF1α-AS1 and AK141205 promotes MSC-bound osteogenesis and osteoblastic differentiation, respectively [53].
Extracellular vehicles (containing miRNAs) have the potential for osteogenic differentiation. miR-675 promotes osteogenesis by inhibiting the recruitment of histone deacetylases to Runx-2-bound DNA sequences. miR-675 targets H19, which further downregulates the β-catenin pathway and results in downregulation of osteogenesis. Upregulation of HOTAIR (inhibits miR-17-5p) [54] and MEG-3 (inhibits miR-133a-3p) [55] inhibits osteogenic differentiation by downregulating Runx-2, ALP, and COL1A1.
To enhance cellular therapeutic options in regenerative medicine, electromagnetic fields act as an auxiliary modality in tissue regeneration. Kang et al. observed both positive and negative osteogenic differentiation of AD-MSCs when applied using appropriate (30/45 Hz at 1mT) electromagnetic fields [56]. This hypothesis could form a basis for the acceleration of tissue expansion in vitro on 3D scaffolds when exposed to an electromagnetic field [56]. Enhanced osteogenic differentiation was observed in BM-MSCs when exposed to modified titanium surface (hydrophilic sandblasted and acid-etched) [57].

5. Osteogenicity of P-MSCs

P-MSCs sorted with CD-90+ demonstrate higher osteogenic potential than unsorted P-MSCs whether in vitro or in vivo [58]. Hence, CD-90+ sorted P-MSCs represent the ideal cell source with higher osteogenic potential for bone regeneration. Upregulation of fibroblast growth factor (FGF)-2, -5, and -6 leads to early callus formation and maintains periosteal osteogenesis [59]. Upregulation of FGFR-1 and -2 was observed in proliferating periosteal mesenchyme [59]. Upregulated expression of Jag-1 and Notch-2 genes was observed in MSCs of the healing periosteum [60].
The lineage differentiation potential of MSCs is mainly governed by PPARγ and Runx2 genes. Runx2 is the key regulator for pro-osteogenesis, whereas PPARγ is responsible for anti-osteoblastogenic effects [61]. Runx-2 and -3 are responsible for osteogenesis and chondrogenesis of MSCs. Along with Runx-2, TGF-β1, BMP, Wnt, Hedgehog (HH), and (Nel)-like protein type 1 (NELL-1) activate and regulate osteogenic responses, as shown in Figure 2 and Figure 3 [62,63].
A dysfunctional β-catenin pathway results in impaired osteoblastic maturation and mineralization [64]. The Wnt/β-catenin signaling pathway leads to downregulated osteoclastogenesis and bone resorption [65]. Antagonists of Wnt signaling molecules such as anti-sclerostin (SOST) and anti-dickkopf-1 enhance MSC-dependent osteogenesis and upregulate bone mineral density [66,67].
Macrophage lineage progenitors recruit P-MSCs for cortical osteogenesis. Colony-stimulating factor knockout results in dysregulated recruitment of macrophage progenitors, leading to impaired periosteal osteogenesis [33]. TRAP+ macrophages induce the expression of periostin and P-MSC recruitment to the periosteal surface through platelet-derived growth factor-BB (PDGF-BB). In the mice model, cortical bone osteogenesis was exhibited by P-MSC-derived Nestin+ and LepR+ CD45 Ter119 CD31 cells. Hence, macrophage lineage progenitors play a significant role in periosteal homeostasis [33]. The upregulation of periostin by PDGF-BB was induced by PDGFR-β, PI3K, AKT, and CREB phosphorylation [15,68]. Periostin, along with autophosphorylated PDGFR-β, regulates and maintains periosteal osteogenesis [15].
Intraosseous 5 μg BMP-2 injection in a mouse model of osteogenesis imperfecta resulted in periosteal ossification and enhanced biomechanical strength and thickness of cortical bone [69]. Endosteal and periosteal osteogenesis was ascertained in chronic ovariectomized female cynomolgus monkeys with an intraosseous injection of recombinant BMP-2 loaded into calcium phosphate matrix into the femoral neck [70]. In a mouse model of spondyloarthropathy, IL-17A deficiency attenuated inflammatory cytokines and bony erosions and promoted periosteal osteogenesis [71]. IL-17A inhibited osteoblastic differentiation in inflammatory periosteum by inducing Wnt antagonist secreted frizzled-related protein (sFRP)-1 and suppressing sFRP-3 expression [72,73,74]. Beta tricalcium phosphate block induced periosteal osteogenesis and acted as a space maker in the soft tissue defect [75].

6. Engineered Osteogenesis by P-MSCs

The key challenge in tissue engineering is the formation of new blood vessels (neovasculogenesis) at the transplanted site. Van Gastel et al. observed increased proangiogenic potential when murine P-MSCs were cotransplanted with a collagen calcium phosphate scaffold and endothelial cells in vivo [76]. As a result, they exhibited pericyte-like cells which induced hematopoietic stroma with neovasculogenesis [76]. Zheng et al. observed enhanced osteogenesis in the form of increased expression of osteocalcin, osteonectin, and type 1 collagen in 3D culture when P-MSCs were loaded onto a poly(lactic-co-glycolic acid) scaffold with allogenic serum [77]. Enzymatically harvested and culture-expanded periosteal cells from human rib periosteum exhibited in vitro osteogenesis and chondrogenesis [78].
Cell-specific COX-2 gene deletion leads to inhibition of BMP-2-mediated differentiation lineages of osteogenesis, chondrogenesis, and vasculogenesis in P-MSCs. Gene profiling exhibited the downregulation of genes responsible for osteogenesis and chondrogenesis such as SOX-9, MMP-9, Osx, Runx-2, and RANKL. COX-2-deficient cells displayed downregulation of HIF-1, PI3K-AKT, and Wnt pathways [79]. Sostdc-1 gene expression was observed in the periosteum, which serves as an important factor in fracture remodeling. Dysregulation of the Sostdc-1 gene resulted in acceleration of fracture healing by promoting the expansion of P-MSCs [80]. They enhanced and maintained quiescent MSCs of the periosteum [80,81].
Administration of a prostaglandin E1 receptor antagonist in wild mice enhanced the production of CFUs of periosteal cells, osteoblastic CFUs, and osteoblastic differentiation of P-MSCs. This enhanced the callus formation in fracture by 10 days [82]. Bravo et al. emphasized that inhibition of plasminogen activator by epsilon aminocaproic acid (EACA)-treated P-MSCs promoted osteogenesis during hard callus formation in appendicular skeleton fractures [83]. The evaluation of callus treated with EACA exhibited increased Wnt and BMP signaling and reduced TGF-β-signaling. EACA treatment enabled a robust switching from chondrogenesis to enhanced osteogenesis, which changed the fate of osteoprogenitor cells of the periosteum [83]. Fibrin admixed with P-MSCs enhanced the proliferation of osteogenic precursors. Tagging of tranexamic acid along with fibrin constructs maintained the integrity of the osteogenic differentiation of P-MSCs [84].
In hypoxic conditions, the enhanced level of H3K27me3 in the promoter region of BMP-2, in combination with decreased KDM6B activity, resulted in inhibition of osteogenic progenitors of P-MSCs [85]. Dysregulation of the Lin28a gene, an RNA-binding protein, resulted in the suppression of osteogenesis and mitochondrial activity of P-MSCs [86]. Jumonji domain-containing 3 (Jmjd3), a histone demethylase, induces osteoblastic differentiation and regulates the expressions of BSP and OCN via transcription factors Runx2 and osterix [87]. Jmjd3 maintains the stemness of MSCs in the periosteum [87,88]. Jagged-1, a Notch ligand, downregulates the osteoprogenitor pool and periosteal expansion along with homotypic notch signals and upregulates trabecular bone formation [89].
Enhanced osteogenesis was observed when MSCs were seeded with an ECM sheet containing calcium phosphate nanoparticles and growth factors such as ANG-1, TGF-β1, bFGF, and VEGF. Such engineered MSC populations act as a biomimetic periosteum that can be used in critical bone defects [90]. Integration of vascular bone graft by an engineered periosteal sheet and β-TCP scaffold mimicked the cellular configuration of the periosteum and, hence, enhanced neo-osteogenesis and neovasculogenesis in bone tissue regeneration [91]. Injectable periosteal ECM activated macrophage (M2) polarization and enhances MSC differentiation into osteogenic cells in calvaria defects in rats [92].

7. Intracellular Signaling in Chondrogenic Differentiation

Various researchers have demonstrated the chondrogenic differentiation of MSCs in vitro with the addition of external biological micro-molecules such as growth factors, bone morphogenetic proteins (BMPs), Hedgehog, SOX signaling molecules, transcription factors, matrix proteins, and Wnt glycoproteins.
Co-expression of SOX-9 with SOX-5 and -6 activates chondrogenic differentiation. Knockout of SOX-9 results in chondrocyte hypertrophy. Repression of chondrocyte hypertrophy by SOX-9 is due to expression of VEGF antagonist and COL10A1 in hypertrophied chondrocyte and inhibition of Runx-2 activation.
Wnt (Wnt5a and Wnt5b) signal regulation of chondrogenesis results in controlled longitudinal growth of long bones by regulating cyclin D1, p130, and chondrocyte-specific COL2A1 expression. Yang et al. stated that Wnt5a and Wnt5b signals control the proliferation of chondrocytes in different zones of cartilage tissues [93]. Wnt5a upregulates and Wnt11 downregulates chondrogenesis via the exposure of chondrocytes to IL-1β. Such transcription results in the inhibition of collagen type 2 expression in chondrocytes [94]. Through BMP-dependent signaling, Twist-1, transcription suppressor gene, inhibits chondrogenesis and chondrocyte gene expression [95].
Fibroblast growth factor (FGF) is an important factor in endochondral and intramembranous bone development, where it regulates proliferation and hypertrophy of chondrocytes. FGF-R1 and -R2 exhibit a chondrogenesis lineage, whereas FGF-R1 is expressed in higher quantities in hypertrophied chondrocytes and FGF-R2 downregulates proliferating chondrocytes. FGF-R3 upregulates chondrocyte proliferation and downregulates hypertrophic chondrocytes. FGF-R9 promotes chondrocyte proliferation and hypertrophy in early stages, whereas it regulates neoangiogenesis in later stages. Indian Hedgehog (IHH) gene and PTHrP signaling work simultaneously in chondrogenesis, as shown in Figure 2 and Figure 3. The IHH gene promotes chondrocyte proliferation and controls PTHrP expression. Hyperexpression of PTHrP exhibits delayed chondrocyte differentiation.
Sustained activation of NF-κB releases nitric oxide, which induces catabolism of cartilage and results in cartilage degeneration [96]. Changes in the oxygen tension have profound effects on chondrocyte differentiation, gene expression, and morphology, along with their production and response to cytokines in the environment. Although hypoxia is a necessary factor in their development, low oxygen tension levels inhibit glycolysis, induce negative Pasteur effects, and inhibit matrix production due to the alteration in the levels of reactive oxygen species (ROS) [97]. Cartilage oligomeric matrix protein (COMP) inhibits BMP-2-induced MSC-bound osteogenesis as evidenced by the evaluation of COL1A1, Runx-2, OPN, and bone Gla protein, while it enhances BMP-2-induced MSC bound chondrogenesis as evidenced by the evaluation of COL2A1, SOX-9, and aggrecan [98].
Guerit et al. exhibited that the upregulation of FOXO3A and downregulation of miR-29a are essential for MSC differentiation into chondrocytes and in vivo osteochondral formation [99]. Upregulation of Wnt signaling promotes the transcription of miR-140-5p in MSC-bound chondrogenic lineage and inhibits cartilage degradation and inflammation [100]. Enhanced expression of miR-145 negatively regulates chondrogenesis by decreasing mRNA levels of chondrogenic markers such as COL2A1, aggrecan, COMP, COL9A2, and COL11A1 in murine MSCs induced by TGF-β3 [101]. miR-194 downregulation enhances AD-MSC-mediated chondrogenesis by targeting the Sox-5 gene [102]. miR-199a, a BMP-2 encoded miRNA, enhances MSC-mediated chondrogenesis by targeting the Smad-1 gene [70,103].
Enhancement of early chondrogenesis and inhibition of hypertrophic chondrocyte differentiation is promoted by low oxygen tension. Portron et al. emphasized that hypoxia prevents calcification of MSC-bound chondrogenesis [104]. Under chondrogenic mediators (hypoxia) influences, MSCs were cultured in a fibrin glue scaffold to observe the proliferation of rounded chondrocyte-like cells with the chondral phenotype; it was concluded that hypoxia is the better stimulant for chondrogenesis [105].
In the collagen-induced arthritis model, small hyaluronic acid fragment activity inhibition and A2A adenosine receptor pathway stimulation limited apoptosis and reduced cartilage damage [106]. MSCs complexed with nonviral vector polyethyleneimine (PEI), mediated by SOX-5, -6, and -9 and loaded onto poly(lactic-co-glycolic acid) (PGLA) nanoparticles, resulted in enhanced chondrogenesis of human MSCs in vitro in culture media [107]. Codelivery of chondrogenic mediators such as SOX-9 and anti-Cbfa-1 siRNA loaded onto PGLA nanoparticles exhibited MSC-bound enhanced chondrogenesis [108].

8. Chondrogenicity of P-MSCs

Ito et al. studied the chondrocyte precursors in the periosteum, and their observations were as follows: (a) the cambium layer of the periosteum caters to chondrocyte precursors, (b) appositional neochondrogenesis displaces the fibrous layer away from already formed cartilaginous tissue, and (c) chondrogenesis commences from the juxtaosseous to juxtafibrous region of the cambium layer [14].
Periosteal progenitors possess chondrogenic potential in the presence of chondrogenic-dependent transcriptional factors and signaling pathways. In the presence of TGF-β3, periosteal progenitors differentiate into chondrocytes along with atelocollagen, which was further evaluated by immunohistochemical staining for type 2 collagen [109].
Downregulation of neural cell adhesion molecule (NCAM) expression during periosteal cell commitment during secondary chondrogenesis provides an alternate pathway for periosteal chondrogenesis [110]. O’Driscoll et al. exhibited that the chondrogenic potential for periosteal MSCs declined with aging in a 12 month old rabbit model by 87% [111]. P-MSCs from the rat model exhibited chondrogenesis due to the induction of BMP-2, whereas terminal chondrocyte differentiation was modulated by TGF-β1 [112].
Higher expression of FGF-16 and -18 was observed in periosteal chondrogenesis of callus formation [59]. Downregulation of Notch-2 expression was observed during chondrogenesis of P-MSCs [60]. Subperiosteal injections of TGF-β1 either alone or in combination with IGF-1 rejuvenated aged periosteum in the rabbit model by increasing the cellular count in the cambium layer and via in vitro chondrogenesis [113].
Shreds of evidence prove that Wnt signaling inhibitors enhance early chondrogenesis, which was demonstrated by a glycosaminoglycan assay, as well as SOX-9, and COL2A1 gene expression. In long-term culture and later in chondrogenesis, Wnt signaling does not play any role in the cartilage tissue engineering of MSCs [114].

9. Engineered Chondrogenesis of P-MSCs

Various studies have emphasized that the periosteum predominantly gives rise to chondrocytes [115,116]. Brittberg et al. demonstrated that the periosteum possessed a higher degree of clonogenicity when chondrocytes were cocultured with periosteal cells and agarose [116]. Such cocultured chondrocytes expressed higher levels of IL-6 and -8, TGF-β3, and GM-CSF. They concluded that TGF-β3 can promote periosteal chondrogenesis. P-MSC chondrogenesis and MSC differentiation were observed when TGF-β3 was injected subperiosteally [117,118,119]. Periosteal cells from adults and the elderly population retain proliferation and multilineage differentiation capacity. Subperiosteal injection of TGF-β1 and IGF-1 increased the cellular count and phenotypic stability in the cambium layer of periosteum in aged rabbits, as well as in vitro cartilage regeneration [117]. In vitro cultured rabbit periosteal explants exhibited chondrogenic potential when exposed to TGF-β1 [120]. Cultured chondrocytes showed increased expression of type 2 collagen.
Digoxin and ATP increase collagen content in neocartilage by around 110%. The tensile strength of newly formed cartilage was increased by digoxin (280%) and ATP (180%). Digoxin and ATP increased Ca2+ oscillations in monolayer cultured chondrocytes. Hence, calcium modulators enhance neochondrogenesis [121]. Evidence has shown that the supplementation of ECM materials such as COMP and aggrecan in a biogel formulation (low-melting-point agarose) mimicked the composition of the cartilage tissue when injected in between bone and the periosteum of the superomedial aspect of rabbit tibia. Regenerated subperiosteal chondrogenic tissue was analyzed for glycosaminoglycan and DNA content, as well as ALP activity. ALP activity was grossly decreased with an increase in GAG and DNA content in regenerated cartilage. This model created a suppressive environment for the chondrocyte hypertrophy niche with chondrogenic progenitors for cartilage tissue engineering in in vitro bioreactors [122]. Hence, this milieu provides a broad-based platform for cartilage grafting in cartilage defects.
In the presence of TGF-β3 in in vitro cultures of periosteal cells, aggrecan induces the chondrogenic potential of periosteal cells and regenerates in vitro bioreactor cartilaginous tissue [123,124]. Such neocartilaginous tissues were able to exhibit GAG with increased TGF-β3 signals [125]. These models suppressed chondrocyte hypertrophy via NKX3-2/Bapx-1 expression [126]. Addition of anti-osteogenic reagents such as fulvestrant and IL-1β to the vascularized periosteum enhanced cartilage regeneration by downregulating osteogenesis [127]. P-MSCs mediated chondrogenesis when administered a cocktail of thermoreversible gelation polymer (TGP) (poly(N-isopropyl acrylamide)), polyethylene oxide, and TGF-β3. The combination of P-MSCs and TGP injection provides a better biocompatible complex for injured cartilage as a minimally invasive procedure [128]. The enhanced expression of TGF-β1 and COL1A1 genes in periosteal paracrine cocultures and the enhanced release of TGF-β1 induced chondrogenesis in an in vitro model of P-MSC-based autologous chondrocyte implantation [129].
In a rib fracture mouse model, Li et al. demonstrated the early stages of chondrogenesis and osteogenesis of periosteal cells. They demonstrated the potential of P-MSCs from osteoblastic differentiation into a chondrogenic lineage and the involvement in osteocytes in osteochondral regeneration by P-MSCs [130]. Downregulation of α1 integrin and upregulation of α3 integrin by threefold, of α5 integrin by threefold, and of β1 by fourfold were observed during extracellular matrix synthesis by periosteal cells. These markers were confirmed in periosteal chondrocytes by immunohistochemistry methods [131]. Injection of TGF-β1 using poly-ε-caprolactone nanofiber scaffolds seeded in vivo with P-MSCs resulted in chondrogenesis in 6 month old rabbits. The chondrocyte yield from P-MSCs was significantly increased by subperiosteal TGF-β1 injections [132].

10. Future Perspectives

Having explored the comparative osteogenic, chondrogenic, and adipogenic potential of P-MSCs in vitro in comparison to other potential sources such as bone marrow MSCs [133], clinical trials on similar grounds are needed to validate the results of the in vitro studies. Apart from periostin, other potential key mediators in the cellular molecular mechanism involved in the mechanosensing and intrinsic repair capabilities of P-MSCs need identification to probe the molecular mechanisms underlying their function. P-MSCs were identified to form an osteoid complex via an intramembranous route, which provides a cellular divergence between the developmental pathways of bone formation [134].
The concept of tissue-engineered periosteum (TEP) is being evaluated for scenarios where the native periosteum is damaged with very few P-MSCs to orchestrate the repair mechanisms. Currently, few studies have been done to characterize the mechanical properties of TEP, and they are being used successfully in oral applications rather than a high-demand site such as long bones of the body [4]. This tissue engineering approach with 3D scaffolds as a periosteal mimetic for cellular attachment, migration, and proliferation needs further validation for practical applicability toward routine clinical use. Furthermore, the identification of factors that regulate stem-cell quiescence and activation is needed. Identification markers of P-MSCs from the resident cambium layer are also needed.

11. Conclusions

The role of P-MSCs in the anabolic pathways of osteochondral tissue repair was highlighted. With a deeper understanding of the mechanosensing and key mediators driving the differentiation pathways, TEP could be used as a substitute in post-traumatic and degenerative conditions of the bone. P-MSCs possess both osteogenic and chondrogenic potential, which is of high therapeutic value. However, clinical validation studies on their utility in various scenarios need to be undertaken to mark their comparative benefit across the various common sources of MSCs.

Author Contributions

Conceptualization—M.J., S.M. and N.J.; Methodology—R.R.; Software—S.M.; Validation—M.J. and S.M.; Data curation—M.J., S.M. and P.G.; Writing—original draft preparation—M.J., S.M., P.G., R.R., N.J., G.S.P., P.C.M., R.L.R., B.-C.A.; Review and editing—R.L.R. and B.-C.A.; Project administration—M.J., S.M. and G.S.P.; Funding acquisition—P.G. and R.L.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2019R1I1A1A01061296 and NRF-2021R1I1A1A01040732).

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dwek, J.R. The periosteum: What is it, where is it, and what mimics it in its absence? Skelet. Radiol. 2010, 39, 319–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Alencar, C.H.M.F.; Silveira, C.R.S.; Cavalcante, M.M.; Vieira, C.G.M.; Teixeira, M.J.D.; Neto, F.A.; de Abreu, A.; Chhabra, A. Periosteum: An imaging review. Eur. J. Radiol. Open 2020, 7, 100249. [Google Scholar] [CrossRef]
  3. Egawa, S.; Miura, S.; Yokoyama, H.; Endo, T.; Tamura, K. Growth and differentiation of a long bone in limb development, repair and regeneration. Dev. Growth Differ. 2014, 56, 410–424. [Google Scholar] [CrossRef] [PubMed]
  4. Evans, S.F.; Chang, H.; Knothe Tate, M.L. Elucidating multiscale periosteal mechanobiology: A key to unlocking the smart properties and regenerative capacity of the periosteum? Tissue Eng. Part B Rev. 2013, 19, 147–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mahajan, A. Periosteum: A Highly Underrated Tool in Dentistry. Int. J. Dent. 2011, 2012, e717816. [Google Scholar] [CrossRef] [PubMed]
  6. Ferretti, C.; Mattioli-Belmonte, M. Periosteum derived stem cells for regenerative medicine proposals: Boosting current knowledge. World J. Stem Cells 2014, 6, 266–277. [Google Scholar] [CrossRef]
  7. Hutmacher, D.W.; Sittinger, M. Periosteal cells in bone tissue engineering. Tissue Eng. 2003, 9 (Suppl. 1), 45–64. [Google Scholar] [CrossRef]
  8. Warnke, P.H.; Douglas, T.; Sivananthan, S.; Wiltfang, J.; Springer, I.; Becker, S.T. Tissue engineering of periosteal cell membranes in vitro. Clin. Oral Implants Res. 2009, 20, 761–766. [Google Scholar] [CrossRef]
  9. Arnsdorf, E.J.; Jones, L.M.; Carter, D.R.; Jacobs, C.R. The periosteum as a cellular source for functional tissue engineering. Tissue Eng. Part A 2009, 15, 2637–2642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Colnot, C.; Zhang, X.; Knothe Tate, M.L. Current insights on the regenerative potential of the periosteum: Molecular, cellular, and endogenous engineering approaches. J. Orthop. Res. 2012, 30, 1869–1878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Chang, H.; Knothe Tate, M.L. Concise review: The periosteum: Tapping into a reservoir of clinically useful progenitor cells. Stem Cells Transl. Med. 2012, 1, 480–491. [Google Scholar] [CrossRef]
  12. Moore, E.R.; Zhu, Y.X.; Ryu, H.S.; Jacobs, C.R. Periosteal progenitors contribute to load-induced bone formation in adult mice and require primary cilia to sense mechanical stimulation. Stem Cell Res. Ther. 2018, 9, 190. [Google Scholar] [CrossRef] [Green Version]
  13. Lin, Z.; Fateh, A.; Salem, D.M.; Intini, G. Periosteum. J. Dent. Res. 2014, 93, 109–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ito, Y.; Fitzsimmons, J.S.; Sanyal, A.; Mello, M.A.; Mukherjee, N.; O’Driscoll, S.W. Localization of chondrocyte precursors in periosteum. Osteoarthr. Cartil. 2001, 9, 215–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Conway, S.J.; Izuhara, K.; Kudo, Y.; Litvin, J.; Markwald, R.; Ouyang, G.; Arron, J.R.; Holweg, C.T.J.; Kudo, A. The role of periostin in tissue remodeling across health and disease. Cell. Mol. Life Sci. 2014, 71, 1279–1288. [Google Scholar] [CrossRef] [Green Version]
  16. Xiao, H.; Wang, L.; Zhang, T.; Chen, C.; Chen, H.; Li, S.; Hu, J.; Lu, H. Periosteum progenitors could stimulate bone regeneration in aged murine bone defect model. J. Cell. Mol. Med. 2020, 24, 12199–12210. [Google Scholar] [CrossRef]
  17. Gnecchi, M.; Zhang, Z.; Ni, A.; Dzau, V.J. Paracrine mechanisms in adult stem cell signaling and therapy. Circ. Res. 2008, 103, 1204–1219. [Google Scholar] [CrossRef] [PubMed]
  18. Baraniak, P.R.; McDevitt, T.C. Stem cell paracrine actions and tissue regeneration. Regen. Med. 2010, 5, 121–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Pittenger, M.F.; Discher, D.E.; Péault, B.M.; Phinney, D.G.; Hare, J.M.; Caplan, A.I. Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen. Med. 2019, 4, 761–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Maleki, M.; Ghanbarvand, F.; Reza Behvarz, M.; Ejtemaei, M.; Ghadirkhomi, E. Comparison of Mesenchymal Stem Cell Markers in Multiple Human Adult Stem Cells. Int. J. Stem Cells 2014, 7, 118–126. [Google Scholar] [CrossRef] [Green Version]
  21. Lv, F.-J.; Tuan, R.S.; Cheung, K.M.C.; Leung, V.Y.L. Concise Review: The Surface Markers and Identity of Human Mesenchymal Stem Cells. Stem Cells 2014, 32, 1408–1419. [Google Scholar] [CrossRef] [PubMed]
  22. Lin, C.-S.; Xin, Z.-C.; Dai, J.; Lue, T.F. Commonly used mesenchymal stem cell markers and tracking labels: Limitations and challenges. Histol. Histopathol. 2013, 28, 1109–1116. [Google Scholar] [CrossRef]
  23. Olivares-Navarrete, R.; Rodil, S.E.; Hyzy, S.L.; Dunn, G.R.; Almaguer-Flores, A.; Schwartz, Z.; Boyan, B.D. Role of Integrin Subunits in Mesenchymal Stem Cell Differentiation and Osteoblast Maturation on Graphitic Carbon-coated Microstructured Surfaces. Biomaterials 2015, 51, 69–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Krebsbach, P.H.; Villa-Diaz, L.G. The Role of Integrin α6 (CD49f) in Stem Cells: More than a Conserved Biomarker. Stem Cells Dev. 2017, 26, 1090–1099. [Google Scholar] [CrossRef] [PubMed]
  25. Ren, G.; Roberts, A.I.; Shi, Y. Adhesion molecules: Key players in Mesenchymal stem cell-mediated immunosuppression. Cell Adhes. Migr. 2011, 5, 20–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Benvenuto, F.; Voci, A.; Carminati, E.; Gualandi, F.; Mancardi, G.; Uccelli, A.; Vergani, L. Human mesenchymal stem cells target adhesion molecules and receptors involved in T cell extravasation. Stem Cell Res. Ther. 2015, 6, 245. [Google Scholar] [CrossRef] [Green Version]
  27. Machado, C.D.V.; Telles, P.D.D.S.; Nascimento, I.L.O. Immunological characteristics of mesenchymal stem cells. Rev. Bras. Hematol. E Hemoter. 2013, 35, 62–67. [Google Scholar] [CrossRef] [PubMed]
  28. Horwitz, E.M.; Andreef, M.; Frassoni, F. Mesenchymal Stromal Cells. Curr. Opin. Hematol. 2006, 13, 419–425. [Google Scholar] [CrossRef] [Green Version]
  29. Frey, S.P.; Jansen, H.; Doht, S.; Filgueira, L.; Zellweger, R. Immunohistochemical and molecular characterization of the human periosteum. Sci. World J. 2013, 2013, 341078. [Google Scholar] [CrossRef]
  30. Yang, H.; Sun, L.; Cai, W.; Gu, J.; Xu, D.; Deb, A.; Duan, J. DDR2, a discoidin domain receptor, is a marker of periosteal osteoblast and osteoblast progenitors. J. Bone Miner. Metab. 2020, 38, 670–677. [Google Scholar] [CrossRef]
  31. Deveza, L.; Ortinau, L.; Lei, K.; Park, D. Comparative analysis of gene expression identifies distinct molecular signatures of bone marrow- and periosteal-skeletal stem/progenitor cells. PLoS ONE 2018, 13, e0190909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ortinau, L.C.; Wang, H.; Lei, K.; Deveza, L.; Jeong, Y.; Hara, Y.; Grafe, I.; Rosenfeld, S.B.; Lee, D.; Lee, B.; et al. Identification of Functionally Distinct Mx1+αSMA+ Periosteal Skeletal Stem Cells. Cell Stem Cell 2019, 25, 784–796. [Google Scholar] [CrossRef] [PubMed]
  33. Gao, B.; Deng, R.; Chai, Y.; Chen, H.; Hu, B.; Wang, X.; Zhu, S.; Cao, Y.; Ni, S.; Wan, M.; et al. Macrophage-lineage TRAP+ cells recruit periosteum-derived cells for periosteal osteogenesis and regeneration. J. Clin. Investig. 2019, 129, 2578–2594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Groeneveldt, L.C.; Herpelinck, T.; Maréchal, M.; Politis, C.; van IJcken, W.F.J.; Huylebroeck, D.; Geris, L.; Mulugeta, E.; Luyten, F.P. The Bone-Forming Properties of Periosteum-Derived Cells Differ Between Harvest Sites. Front. Cell Dev. Biol. 2020, 8, 554984. [Google Scholar] [CrossRef]
  35. Olbrich, M.; Rieger, M.; Reinert, S.; Alexander, D. Isolation of Osteoprogenitors from Human Jaw Periosteal Cells: A Comparison of Two Magnetic Separation Methods. PLoS ONE 2012, 7, e47176. [Google Scholar] [CrossRef] [Green Version]
  36. De Bari, C.; Dell’Accio, F.; Vanlauwe, J.; Eyckmans, J.; Khan, I.M.; Archer, C.W.; Jones, E.A.; McGonagle, D.; Mitsiadis, T.A.; Pitzalis, C.; et al. Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum. 2006, 54, 1209–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Potier, E.; Ferreira, E.; Andriamanalijaona, R.; Pujol, J.-P.; Oudina, K.; Logeart-Avramoglou, D.; Petite, H. Hypoxia affects mesenchymal stromal cell osteogenic differentiation and angiogenic factor expression. Bone 2007, 40, 1078–1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Huang, J.; Deng, F.; Wang, L.; Xiang, X.-R.; Zhou, W.-W.; Hu, N.; Xu, L. Hypoxia induces osteogenesis-related activities and expression of core binding factor α1 in mesenchymal stem cells. Tohoku J. Exp. Med. 2011, 224, 7–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yu, X.; Wan, Q.; Ye, X.; Cheng, Y.; Pathak, J.L.; Li, Z. Cellular hypoxia promotes osteogenic differentiation of mesenchymal stem cells and bone defect healing via STAT3 signaling. Cell. Mol. Biol. Lett. 2019, 24, 64. [Google Scholar] [CrossRef]
  40. Cheng, S.-L.; Shao, J.-S.; Charlton-Kachigian, N.; Loewy, A.P.; Towler, D.A. Msx2 Promotes Osteogenesis and Suppresses Adipogenic Differentiation of Multipotent Mesenchymal Progenitors. J. Biol. Chem. 2003, 278, 45969–45977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Chen, L.; Shi, K.; Andersen, T.L.; Qiu, W.; Kassem, M. KIAA1199 is a secreted molecule that enhances osteoblastic stem cell migration and recruitment. Cell Death Dis. 2019, 10, 126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Wu, J.; Wang, C.; Miao, X.; Wu, Y.; Yuan, J.; Ding, M.; Li, J.; Shi, Z. Age-Related Insulin-Like Growth Factor Binding Protein-4 Overexpression Inhibits Osteogenic Differentiation of Rat Mesenchymal Stem Cells. Cell. Physiol. Biochem. 2017, 42, 640–650. [Google Scholar] [CrossRef]
  43. Wang, J.; Li, G.; Hu, L.; Yan, F.; Zhao, B.; Wu, X.; Zhang, C.; Wang, J.; Du, J.; Wang, S. Retinoic Acid Signal Negatively Regulates Osteo/Odontogenic Differentiation of Dental Pulp Stem Cells. Stem Cells Int. 2020, 2020, 5891783. [Google Scholar] [CrossRef]
  44. Salasznyk, R.M.; Williams, W.A.; Boskey, A.; Batorsky, A.; Plopper, G.E. Adhesion to Vitronectin and Collagen I Promotes Osteogenic Differentiation of Human Mesenchymal Stem Cells. J. Biomed. Biotechnol. 2004, 2004, 24–34. [Google Scholar] [CrossRef]
  45. Suehiro, F.; Nishimura, M.; Kawamoto, T.; Kanawa, M.; Yoshizawa, Y.; Murata, H.; Kato, Y. Impact of zinc fingers and homeoboxes 3 on the regulation of mesenchymal stem cell osteogenic differentiation. Stem Cells Dev. 2011, 20, 1539–1547. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, D.; Gong, Y.; Xu, L.; Zhou, M.; Li, J.; Song, J. Bidirectional regulation of osteogenic differentiation by the FOXO subfamily of Forkhead transcription factors in mammalian MSCs. Cell Prolif. 2018, 52, e12540. [Google Scholar] [CrossRef] [Green Version]
  47. Baglìo, S.R.; Devescovi, V.; Granchi, D.; Baldini, N. MicroRNA expression profiling of human bone marrow mesenchymal stem cells during osteogenic differentiation reveals Osterix regulation by miR-31. Gene 2013, 527, 321–331. [Google Scholar] [CrossRef]
  48. Li, G.; Han, N.; Yang, H.; Wang, L.; Lin, X.; Diao, S.; Du, J.; Dong, R.; Wang, S.; Fan, Z. Homeobox C10 inhibits the osteogenic differentiation potential of mesenchymal stem cells. Connect. Tissue Res. 2018, 59, 201–211. [Google Scholar] [CrossRef] [PubMed]
  49. Qu, B.; Liu, O.; Fang, X.; Zhang, H.; Wang, Y.; Quan, H.; Zhang, J.; Zhou, J.; Zuo, J.; Tang, J.; et al. Distal-less homeobox 2 promotes the osteogenic differentiation potential of stem cells from apical papilla. Cell Tissue Res. 2014, 357, 133–143. [Google Scholar] [CrossRef]
  50. Yang, H.; Fan, J.; Cao, Y.; Gao, R.; Fan, Z. Distal-less homeobox 5 promotes the osteo-/dentinogenic differentiation potential of stem cells from apical papilla by activating histone demethylase KDM4B through a positive feedback mechanism. Exp. Cell Res. 2019, 374, 221–230. [Google Scholar] [CrossRef]
  51. Gao, R.-T.; Zhan, L.-P.; Meng, C.; Zhang, N.; Chang, S.-M.; Yao, R.; Li, C. Homeobox B7 promotes the osteogenic differentiation potential of mesenchymal stem cells by activating RUNX2 and transcript of BSP. Int. J. Clin. Exp. Med. 2015, 8, 10459–10470. [Google Scholar]
  52. Diao, S.; Yang, D.-M.; Dong, R.; Wang, L.-P.; Wang, J.-S.; Du, J.; Wang, S.-L.; Fan, Z. Enriched trimethylation of lysine 4 of histone H3 of WDR63 enhanced osteogenic differentiation potentials of stem cells from apical papilla. J. Endod. 2015, 41, 205–211. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, Q.; Jia, L.; Li, X.; Guo, R.; Huang, Y.; Zheng, Y.; Li, W. Long Noncoding RNAs: New Players in the Osteogenic Differentiation of Bone Marrow- and Adipose-Derived Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2018, 14, 297–308. [Google Scholar] [CrossRef] [PubMed]
  54. Wei, B.; Wei, W.; Zhao, B.; Guo, X.; Liu, S. Long non-coding RNA HOTAIR inhibits miR-17-5p to regulate osteogenic differentiation and proliferation in non-traumatic osteonecrosis of femoral head. PLoS ONE 2017, 12, e0169097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wang, Q.; Li, Y.; Zhang, Y.; Ma, L.; Lin, L.; Meng, J.; Jiang, L.; Wang, L.; Zhou, P.; Zhang, Y. LncRNA MEG3 inhibited osteogenic differentiation of bone marrow mesenchymal stem cells from postmenopausal osteoporosis by targeting miR-133a-3p. Biomed. Pharmacother. 2017, 89, 1178–1186. [Google Scholar] [CrossRef] [PubMed]
  56. Kang, K.S.; Hong, J.M.; Kang, J.A.; Rhie, J.-W.; Jeong, Y.H.; Cho, D.-W. Regulation of osteogenic differentiation of human adipose-derived stem cells by controlling electromagnetic field conditions. Exp. Mol. Med. 2013, 45, e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wall, I.; Donos, N.; Carlqvist, K.; Jones, F.; Brett, P. Modified titanium surfaces promote accelerated osteogenic differentiation of mesenchymal stromal cells in vitro. Bone 2009, 45, 17–26. [Google Scholar] [CrossRef]
  58. Kim, Y.-K.; Nakata, H.; Yamamoto, M.; Miyasaka, M.; Kasugai, S.; Kuroda, S. Osteogenic Potential of Mouse Periosteum-Derived Cells Sorted for CD90 In Vitro and In Vivo. Stem Cells Transl. Med. 2016, 5, 227–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Du, X.; Xie, Y.; Xian, C.J.; Chen, L. Role of FGFs/FGFRs in skeletal development and bone regeneration. J. Cell. Physiol. 2012, 227, 3731–3743. [Google Scholar] [CrossRef]
  60. Dishowitz, M.I.; Terkhorn, S.P.; Bostic, S.A.; Hankenson, K.D. Notch signaling components are upregulated during both endochondral and intramembranous bone regeneration. J. Orthop. Res. 2012, 30, 296–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. James, A.W. Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation. Scientifica 2013, 2013, e684736. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, K.S.; Kim, H.J.; Li, Q.L.; Chi, X.Z.; Ueta, C.; Komori, T.; Wozney, J.M.; Kim, E.G.; Choi, J.Y.; Ryoo, H.M.; et al. Runx2 is a common target of transforming growth factor beta1 and bone morphogenetic protein 2, and cooperation between Runx2 and Smad5 induces osteoblast-specific gene expression in the pluripotent mesenchymal precursor cell line C2C12. Mol. Cell. Biol. 2000, 20, 8783–8792. [Google Scholar] [CrossRef] [Green Version]
  63. Pratap, J.; Wixted, J.J.; Gaur, T.; Zaidi, S.K.; Dobson, J.; Gokul, K.D.; Hussain, S.; van Wijnen, A.J.; Stein, J.L.; Stein, G.S.; et al. Runx2 transcriptional activation of Indian Hedgehog and a downstream bone metastatic pathway in breast cancer cells. Cancer Res. 2008, 68, 7795–7802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Glass, D.A.; Bialek, P.; Ahn, J.D.; Starbuck, M.; Patel, M.S.; Clevers, H.; Taketo, M.M.; Long, F.; McMahon, A.P.; Lang, R.A.; et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev. Cell 2005, 8, 751–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Takahashi, N.; Maeda, K.; Ishihara, A.; Uehara, S.; Kobayashi, Y. Regulatory mechanism of osteoclastogenesis by RANKL and Wnt signals. Front. Biosci 2011, 16, 21–30. [Google Scholar] [CrossRef] [Green Version]
  66. Papapoulos, S.E. Targeting sclerostin as potential treatment of osteoporosis. Ann. Rheum. Dis. 2011, 70 (Suppl. 1), i119–i122. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, J.H.; Liu, X.; Wang, J.; Chen, X.; Zhang, H.; Kim, S.H.; Cui, J.; Li, R.; Zhang, W.; Kong, Y.; et al. Wnt signaling in bone formation and its therapeutic potential for bone diseases. Ther. Adv. Musculoskelet. Dis. 2013, 5, 13–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Wang, H.; Yin, Y.; Li, W.; Zhao, X.; Yu, Y.; Zhu, J.; Qin, Z.; Wang, Q.; Wang, K.; Lu, W.; et al. Over-expression of PDGFR-β promotes PDGF-induced proliferation, migration, and angiogenesis of EPCs through PI3K/Akt signaling pathway. PLoS ONE 2012, 7, e30503. [Google Scholar] [CrossRef] [Green Version]
  69. Cheng, T.L.; Cantrill, L.C.; Schindeler, A.; Little, D.G. Induction of periosteal bone formation by intraosseous BMP-2 injection in a mouse model of osteogenesis imperfecta. J. Child. Orthop. 2019, 13, 543–550. [Google Scholar] [CrossRef]
  70. Seeherman, H.J.; Li, X.J.; Smith, E.; Parkington, J.; Li, R.; Wozney, J.M. Intraosseous injection of rhBMP-2/calcium phosphate matrix improves bone structure and strength in the proximal aspect of the femur in chronic ovariectomized nonhuman primates. J. Bone Joint Surg. 2013, 95, 36–47. [Google Scholar] [CrossRef]
  71. Shaw, A.T.; Maeda, Y.; Gravallese, E.M. IL-17A deficiency promotes periosteal bone formation in a model of inflammatory arthritis. Arthritis Res. Ther. 2016, 18, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Claudel, M.; Jouzeau, J.-Y.; Cailotto, F. Secreted Frizzled-related proteins (sFRPs) in osteo-articular diseases: Much more than simple antagonists of Wnt signaling? FEBS J. 2019, 286, 4832–4851. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, F.-S.; Lin, C.-L.; Chen, Y.-J.; Wang, C.-J.; Yang, K.D.; Huang, Y.-T.; Sun, Y.-C.; Huang, H.-C. Secreted frizzled-related protein 1 modulates glucocorticoid attenuation of osteogenic activities and bone mass. Endocrinology 2005, 146, 2415–2423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Katagiri, W.; Osugi, M.; Kawai, T.; Hibi, H. Secreted Frizzled-Related Protein Promotes Bone Regeneration by Human Bone Marrow-Derived Mesenchymal Stem Cells. Int. J. Mol. Sci. 2015, 16, 23250–23258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Yamauchi, K.; Takahashi, T.; Funaki, K.; Yamashita, Y. Periosteal Expansion Osteogenesis Using Highly Purified Beta-Tricalcium Phosphate Blocks: A Pilot Study in Dogs. J. Periodontol. 2008, 79, 999–1005. [Google Scholar] [CrossRef] [PubMed]
  76. van Gastel, N.; Torrekens, S.; Roberts, S.J.; Moermans, K.; Schrooten, J.; Carmeliet, P.; Luttun, A.; Luyten, F.P.; Carmeliet, G. Engineering vascularized bone: Osteogenic and proangiogenic potential of murine periosteal cells. Stem Cells 2012, 30, 2460–2471. [Google Scholar] [CrossRef] [Green Version]
  77. Zheng, Y.; Ringe, J.; Liang, Z.; Loch, A.; Chen, L.; Sittinger, M. Osteogenic potential of human periosteum-derived progenitor cells in PLGA scaffold using allogeneic serum. J. Zhejiang Univ. Sci. B 2006, 7, 817–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Nakahara, H.; Goldberg, V.M.; Caplan, A.I. Culture-expanded human periosteal-derived cells exhibit osteochondral potential in vivo. J. Orthop. Res. 1991, 9, 465–476. [Google Scholar] [CrossRef] [PubMed]
  79. Huang, C.; Xue, M.; Chen, H.; Jiao, J.; Herschman, H.R.; O’Keefe, R.J.; Zhang, X. The Spatiotemporal Role of COX-2 in Osteogenic and Chondrogenic Differentiation of Periosteum-Derived Mesenchymal Progenitors in Fracture Repair. PLoS ONE 2014, 9, e100079. [Google Scholar] [CrossRef] [Green Version]
  80. Collette, N.M.; Yee, C.S.; Hum, N.R.; Murugesh, D.K.; Christiansen, B.A.; Xie, L.; Economides, A.N.; Manilay, J.O.; Robling, A.G.; Loots, G.G. Sostdc1 deficiency accelerates fracture healing by promoting the expansion of periosteal mesenchymal stem cells. Bone 2016, 88, 20–30. [Google Scholar] [CrossRef] [Green Version]
  81. Kurenkova, A.D.; Medvedeva, E.V.; Newton, P.T.; Chagin, A.S. Niches for Skeletal Stem Cells of Mesenchymal Origin. Front. Cell Dev. Biol. 2020, 8, 592. [Google Scholar] [CrossRef] [PubMed]
  82. Feigenson, M.; Jonason, J.H.; Shen, J.; Loiselle, A.E.; Awad, H.A.; O’Keefe, R.J. Inhibition of the Prostaglandin EP-1 Receptor in Periosteum Progenitor Cells Enhances Osteoblast Differentiation and Fracture Repair. Ann. Biomed. Eng. 2020, 48, 927–939. [Google Scholar] [CrossRef] [PubMed]
  83. Bravo, D.; Josephson, A.; Bradaschia-Correa, V.; Wong, M.; Yim, N.; Neibart, S.; Lee, S.; Huo, J.; Coughlin, T.; Mizrahi, M.; et al. Temporary inhibition of the plasminogen activator inhibits periosteal chondrogenesis and promotes periosteal osteogenesis during appendicular bone fracture healing. Bone 2018, 112, 97–106. [Google Scholar] [CrossRef] [PubMed]
  84. Demol, J.; Eyckmans, J.; Roberts, S.J.; Luyten, F.P.; Van Oosterwyck, H. Does tranexamic acid stabilised fibrin support the osteogenic differentiation of human periosteum derived cells? Eur. Cells Mater. 2011, 21, 272–285. [Google Scholar] [CrossRef]
  85. Yoon, D.-K.; Park, J.-S.; Rho, G.-J.; Lee, H.-J.; Sung, I.-Y.; Son, J.-H.; Park, B.-W.; Kang, Y.-H.; Byun, S.-H.; Hwang, S.-C.; et al. The involvement of histone methylation in osteoblastic differentiation of human periosteum-derived cells cultured in vitro under hypoxic conditions. Cell Biochem. Funct. 2017, 35, 441–452. [Google Scholar] [CrossRef]
  86. Park, J.-H.; Park, B.-W.; Kang, Y.-H.; Byun, S.-H.; Hwang, S.-C.; Kim, D.R.; Woo, D.K.; Byun, J.-H. Lin28a enhances in vitro osteoblastic differentiation of human periosteum-derived cells. Cell Biochem. Funct. 2017, 35, 497–509. [Google Scholar] [CrossRef] [PubMed]
  87. Yang, D.; Okamura, H.; Nakashima, Y.; Haneji, T. Histone demethylase Jmjd3 regulates osteoblast differentiation via transcription factors Runx2 and osterix. J. Biol. Chem. 2013, 288, 33530–33541. [Google Scholar] [CrossRef] [Green Version]
  88. Agger, K.; Cloos, P.A.C.; Christensen, J.; Pasini, D.; Rose, S.; Rappsilber, J.; Issaeva, I.; Canaani, E.; Salcini, A.E.; Helin, K. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature 2007, 449, 731–734. [Google Scholar] [CrossRef] [PubMed]
  89. Youngstrom, D.W.; Dishowitz, M.I.; Bales, C.B.; Carr, E.; Mutyaba, P.L.; Kozloff, K.M.; Shitaye, H.; Hankenson, K.D.; Loomes, K.M. Jagged1 expression by osteoblast-lineage cells regulates trabecular bone mass and periosteal expansion in mice. Bone 2016, 91, 64–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Xing, Q.; Qian, Z.; Kannan, B.; Tahtinen, M.; Zhao, F. Osteogenic Differentiation Evaluation of an Engineered Extracellular Matrix Based Tissue Sheet for Potential Periosteum Replacement. ACS Appl. Mater. Interfaces 2015, 7, 23239–23247. [Google Scholar] [CrossRef]
  91. Kang, Y.; Ren, L.; Yang, Y. Engineering vascularized bone grafts by integrating a biomimetic periosteum and β-TCP scaffold. ACS Appl. Mater. Interfaces 2014, 6, 9622–9633. [Google Scholar] [CrossRef] [PubMed]
  92. Qiu, P.; Li, M.; Chen, K.; Fang, B.; Chen, P.; Tang, Z.; Lin, X.; Fan, S. Periosteal matrix-derived hydrogel promotes bone repair through an early immune regulation coupled with enhanced angio-and osteogenesis. Biomaterials 2020, 227, 119552. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, Y.; Topol, L.; Lee, H.; Wu, J. Wnt5a and Wnt5b exhibit distinct activities in coordinating chondrocyte proliferation and differentiation. Dev. Camb. Engl. 2003, 130, 1003–1015. [Google Scholar] [CrossRef] [Green Version]
  94. Ryu, J.-H.; Chun, J.-S. Opposing roles of WNT-5A and WNT-11 in interleukin-1beta regulation of type II collagen expression in articular chondrocytes. J. Biol. Chem. 2006, 281, 22039–22047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Reinhold, M.I.; Kapadia, R.M.; Liao, Z.; Naski, M.C. The Wnt-inducible transcription factor Twist1 inhibits chondrogenesis. J. Biol. Chem. 2006, 281, 1381–1388. [Google Scholar] [CrossRef] [Green Version]
  96. Clancy, R.M.; Gomez, P.F.; Abramson, S.B. Nitric oxide sustains nuclear factor kappaB activation in cytokine-stimulated chondrocytes. Osteoarthr. Cartil. 2004, 12, 552–558. [Google Scholar] [CrossRef] [Green Version]
  97. Gibson, J.S.; Milner, P.I.; White, R.; Fairfax, T.P.A.; Wilkins, R.J. Oxygen and reactive oxygen species in articular cartilage: Modulators of ionic homeostasis. Pflüg. Arch.-Eur. J. Physiol. 2008, 455, 563–573. [Google Scholar] [CrossRef] [PubMed]
  98. Guo, P.; Shi, Z.; Liu, A.; Lin, T.; Bi, F.; Shi, M.; Yan, S.-G. Effects of cartilage oligomeric matrix protein on bone morphogenetic protein-2-induced differentiation of mesenchymal stem cells. Orthop. Surg. 2014, 6, 280–287. [Google Scholar] [CrossRef]
  99. Guérit, D.; Brondello, J.-M.; Chuchana, P.; Philipot, D.; Toupet, K.; Bony, C.; Jorgensen, C.; Noël, D. FOXO3A regulation by miRNA-29a Controls chondrogenic differentiation of mesenchymal stem cells and cartilage formation. Stem Cells Dev. 2014, 23, 1195–1205. [Google Scholar] [CrossRef]
  100. Duan, L.; Liang, Y.; Xu, X.; Xiao, Y.; Wang, D. Recent progress on the role of miR-140 in cartilage matrix remodelling and its implications for osteoarthritis treatment. Arthritis Res. Ther. 2020, 22, 194. [Google Scholar] [CrossRef]
  101. Yang, B.; Guo, H.; Zhang, Y.; Chen, L.; Ying, D.; Dong, S. MicroRNA-145 regulates chondrogenic differentiation of mesenchymal stem cells by targeting Sox9. PLoS ONE 2011, 6, e21679. [Google Scholar] [CrossRef]
  102. Xu, J.; Kang, Y.; Liao, W.; Yu, L. MiR-194 Regulates Chondrogenic Differentiation of Human Adipose-Derived Stem Cells by Targeting Sox5. PLoS ONE 2012, 7, e31861. [Google Scholar] [CrossRef] [Green Version]
  103. Lin, E.A.; Kong, L.; Bai, X.-H.; Luan, Y.; Liu, C.-J. miR-199a, a bone morphogenic protein 2-responsive MicroRNA, regulates chondrogenesis via direct targeting to Smad1. J. Biol. Chem. 2009, 284, 11326–11335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Portron, S.; Hivernaud, V.; Merceron, C.; Lesoeur, J.; Masson, M.; Gauthier, O.; Vinatier, C.; Beck, L.; Guicheux, J. Inverse regulation of early and late chondrogenic differentiation by oxygen tension provides cues for stem cell-based cartilage tissue engineering. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2015, 35, 841–857. [Google Scholar] [CrossRef] [PubMed]
  105. Baumgartner, L.; Arnhold, S.; Brixius, K.; Addicks, K.; Bloch, W. Human mesenchymal stem cells: Influence of oxygen pressure on proliferation and chondrogenic differentiation in fibrin glue in vitro. J. Biomed. Mater. Res. Part A 2010, 93, 930–940. [Google Scholar] [CrossRef] [PubMed]
  106. Campo, G.M.; Micali, A.; Avenoso, A.; D’Ascola, A.; Scuruchi, M.; Pisani, A.; Bruschetta, A.; Calatroni, A.; Puzzolo, D.; Campo, S. Inhibition of small HA fragment activity and stimulation of A2A adenosine receptor pathway limit apoptosis and reduce cartilage damage in experimental arthritis. Histochem. Cell Biol. 2015, 143. [Google Scholar] [CrossRef] [PubMed]
  107. Park, J.S.; Yang, H.N.; Woo, D.G.; Jeon, S.Y.; Do, H.-J.; Lim, H.-Y.; Kim, J.-H.; Park, K.-H. Chondrogenesis of human mesenchymal stem cells mediated by the combination of SOX trio SOX5, 6, and 9 genes complexed with PEI-modified PLGA nanoparticles. Biomaterials 2011, 32, 3679–3688. [Google Scholar] [CrossRef] [PubMed]
  108. Jeon, S.Y.; Park, J.S.; Yang, H.N.; Woo, D.G.; Park, K.-H. Co-delivery of SOX9 genes and anti-Cbfa-1 siRNA coated onto PLGA nanoparticles for chondrogenesis of human MSCs. Biomaterials 2012, 33, 4413–4423. [Google Scholar] [CrossRef]
  109. Choi, Y.-S.; Lim, S.-M.; Shin, H.-C.; Lee, C.-W.; Kim, S.-L.; Kim, D.-I. Chondrogenesis of human periosteum-derived progenitor cells in atelocollagen. Biotechnol. Lett. 2007, 29, 323–329. [Google Scholar] [CrossRef] [PubMed]
  110. Fang, J.; Hall, B.K. Chondrogenic cell differentiation from membrane bone periostea. Anat. Embryol. 1997, 196, 349–362. [Google Scholar] [CrossRef]
  111. O’Driscoll, S.W.; Saris, D.B.; Ito, Y.; Fitzimmons, J.S. The chondrogenic potential of periosteum decreases with age. J. Orthop. Res. 2001, 19, 95–103. [Google Scholar] [CrossRef]
  112. Hanada, K.; Solchaga, L.A.; Caplan, A.I.; Hering, T.M.; Goldberg, V.M.; Yoo, J.U.; Johnstone, B. BMP-2 induction and TGF-β1 modulation of rat periosteal cell chondrogenesis. J. Cell. Biochem. 2001, 81, 284–294. [Google Scholar] [CrossRef]
  113. Reinholz, G.G.; Fitzsimmons, J.S.; Casper, M.E.; Ruesink, T.J.; Chung, H.W.; Schagemann, J.C.; O’Driscoll, S.W. Rejuvenation of periosteal chondrogenesis using local growth factor injection. Osteoarthr. Cartil. 2009, 17, 723–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Im, G.-I.; Quan, Z. The effects of Wnt inhibitors on the chondrogenesis of human mesenchymal stem cells. Tissue Eng. Part A 2010, 16, 2405–2413. [Google Scholar] [CrossRef]
  115. Gooding, C.R.; Bartlett, W.; Bentley, G.; Skinner, J.A.; Carrington, R.; Flanagan, A. A prospective, randomised study comparing two techniques of autologous chondrocyte implantation for osteochondral defects in the knee: Periosteum covered versus type I/III collagen covered. Knee 2006, 13, 203–210. [Google Scholar] [CrossRef]
  116. Brittberg, M.; Sjögren-Jansson, E.; Thornemo, M.; Faber, B.; Tarkowski, A.; Peterson, L.; Lindahl, A. Clonal growth of human articular cartilage and the functional role of the periosteum in chondrogenesis. Osteoarthr. Cartil. 2005, 13, 146–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. De Bari, C.; Dell’Accio, F.; Luyten, F.P. Human periosteum-derived cells maintain phenotypic stability and chondrogenic potential throughout expansion regardless of donor age. Arthritis Rheum. 2001, 44, 85–95. [Google Scholar] [CrossRef]
  118. Bian, L.; Zhai, D.Y.; Tous, E.; Rai, R.; Mauck, R.L.; Burdick, J.A. Enhanced MSC chondrogenesis following delivery of TGF-β3 from alginate microspheres within hyaluronic acid hydrogels in vitro and in vivo. Biomaterials 2011, 32, 6425–6434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Koga, H.; Muneta, T.; Nagase, T.; Nimura, A.; Ju, Y.-J.; Mochizuki, T.; Sekiya, I. Comparison of mesenchymal tissues-derived stem cells for in vivo chondrogenesis: Suitable conditions for cell therapy of cartilage defects in rabbit. Cell Tissue Res. 2008, 333, 207–215. [Google Scholar] [CrossRef]
  120. O’Driscoll, S.W.; Recklies, A.D.; Poole, A.R. Chondrogenesis in periosteal explants. An organ culture model for in vitro study. JBJS 1994, 76, 1042–1051. [Google Scholar] [CrossRef]
  121. Makris, E.A.; Huang, B.J.; Hu, J.C.; Chen-Izu, Y.; Athanasiou, K.A. Digoxin and adenosine triphosphate enhance the functional properties of tissue-engineered cartilage. Tissue Eng. Part A 2015, 21, 884–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Caron, M.M.J.; Janssen, M.P.F.; Peeters, L.; Haudenschild, D.R.; Cremers, A.; Surtel, D.A.M.; van Rhijn, L.W.; Emans, P.J.; Welting, T.J.M. Aggrecan and COMP Improve Periosteal Chondrogenesis by Delaying Chondrocyte Hypertrophic Maturation. Front. Bioeng. Biotechnol. 2020, 8, 1036. [Google Scholar] [CrossRef] [PubMed]
  123. Kock, L.; van Donkelaar, C.C.; Ito, K. Tissue engineering of functional articular cartilage: The current status. Cell Tissue Res. 2012, 347, 613–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Chen, F.H.; Rousche, K.T.; Tuan, R.S. Technology Insight: Adult stem cells in cartilage regeneration and tissue engineering. Nat. Clin. Pract. Rheumatol. 2006, 2, 373–382. [Google Scholar] [CrossRef] [PubMed]
  125. Dahlin, R.L.; Ni, M.; Meretoja, V.V.; Kasper, F.K.; Mikos, A.G. TGF-β3-induced chondrogenesis in co-cultures of chondrocytes and mesenchymal stem cells on biodegradable scaffolds. Biomaterials 2014, 35, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Rainbow, R.S.; Won, H.K.; Zeng, L. The role of Nkx3.2 in chondrogenesis. Front. Biol. 2014, 9, 376–381. [Google Scholar] [CrossRef] [Green Version]
  127. Hsiao, H.-Y.; Cheng, C.-M.; Kao, S.-W.; Liu, J.-W.; Chang, C.-S.; Harhaus, L.; Huang, J.-J. The effect of bone inhibitors on periosteum-guided cartilage regeneration. Sci. Rep. 2020, 10, 8372. [Google Scholar] [CrossRef]
  128. Choi, Y.S.; Lim, S.M.; Shin, H.C.; Lee, C.W.; Kim, D.-I. Chondrogenic properties of human periosteum-derived progenitor cells (PDPCs) embedded in a thermoreversible gelation polymer (TGP). Biotechnol. Bioprocess Eng. 2006, 11, 550–552. [Google Scholar] [CrossRef]
  129. Grässel, S.; Rickert, M.; Opolka, A.; Bosserhoff, A.; Angele, P.; Grifka, J.; Anders, S. Coculture between periosteal explants and articular chondrocytes induces expression of TGF-beta1 and collagen I. Rheumatol. Oxf. Engl. 2010, 49, 218–230. [Google Scholar] [CrossRef] [Green Version]
  130. Li, M.; Amizuka, N.; Oda, K.; Tokunaga, K.; Ito, T.; Takeuchi, K.; Takagi, R.; Maeda, T. Histochemical evidence of the initial chondrogenesis and osteogenesis in the periosteum of a rib fractured model: Implications of osteocyte involvement in periosteal chondrogenesis. Microsc. Res. Tech. 2004, 64, 330–342. [Google Scholar] [CrossRef]
  131. Fukumoto, T.; Sanyal, A.; Fitzsimmons, J.S.; O’Driscoll, S.W. Expression of beta1 integrins during periosteal chondrogenesis. Osteoarthr. Cartil. 2002, 10, 135–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Casper, M.E.; Fitzsimmons, J.S.; Stone, J.J.; Meza, A.O.; Huang, Y.; Ruesink, T.J.; O’Driscoll, S.W.; Reinholz, G.G. Tissue engineering of cartilage using poly-ε-caprolactone nanofiber scaffolds seeded in vivo with periosteal cells. Osteoarthr. Cartil. 2010, 18, 981–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Duchamp de Lageneste, O.; Julien, A.; Abou-Khalil, R.; Frangi, G.; Carvalho, C.; Cagnard, N.; Cordier, C.; Conway, S.J.; Colnot, C. Periosteum contains skeletal stem cells with high bone regenerative potential controlled by Periostin. Nat. Commun. 2018, 9, 773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Debnath, S.; Yallowitz, A.R.; McCormick, J.; Lalani, S.; Zhang, T.; Xu, R.; Li, N.; Liu, Y.; Yang, Y.S.; Eiseman, M.; et al. Discovery of a periosteal stem cell mediating intramembranous bone formation. Nature 2018, 562, 133–139. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Anatomy of periosteum. Created with BioRender.com.
Figure 1. Anatomy of periosteum. Created with BioRender.com.
Pharmaceuticals 14 01133 g001
Figure 2. Osteogenic and chondrogenic differentiation of periosteal mesenchymal stem cells. Created with BioRender.com.
Figure 2. Osteogenic and chondrogenic differentiation of periosteal mesenchymal stem cells. Created with BioRender.com.
Pharmaceuticals 14 01133 g002
Figure 3. Intracellular signaling of osteogenic and chondrogenic differentiation of periosteal mesenchymal stem cells. Created with BioRender.com.
Figure 3. Intracellular signaling of osteogenic and chondrogenic differentiation of periosteal mesenchymal stem cells. Created with BioRender.com.
Pharmaceuticals 14 01133 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jeyaraman, M.; Muthu, S.; Gangadaran, P.; Ranjan, R.; Jeyaraman, N.; Prajwal, G.S.; Mishra, P.C.; Rajendran, R.L.; Ahn, B.-C. Osteogenic and Chondrogenic Potential of Periosteum-Derived Mesenchymal Stromal Cells: Do They Hold the Key to the Future? Pharmaceuticals 2021, 14, 1133. https://doi.org/10.3390/ph14111133

AMA Style

Jeyaraman M, Muthu S, Gangadaran P, Ranjan R, Jeyaraman N, Prajwal GS, Mishra PC, Rajendran RL, Ahn B-C. Osteogenic and Chondrogenic Potential of Periosteum-Derived Mesenchymal Stromal Cells: Do They Hold the Key to the Future? Pharmaceuticals. 2021; 14(11):1133. https://doi.org/10.3390/ph14111133

Chicago/Turabian Style

Jeyaraman, Madhan, Sathish Muthu, Prakash Gangadaran, Rajni Ranjan, Naveen Jeyaraman, Gollahalli Shivashankar Prajwal, Prabhu Chandra Mishra, Ramya Lakshmi Rajendran, and Byeong-Cheol Ahn. 2021. "Osteogenic and Chondrogenic Potential of Periosteum-Derived Mesenchymal Stromal Cells: Do They Hold the Key to the Future?" Pharmaceuticals 14, no. 11: 1133. https://doi.org/10.3390/ph14111133

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop