Skip to main content

Using mesenchymal stem cells as a therapy for bone regeneration and repairing

Abstract

Bone is a unique tissue which could regenerate completely after injury rather than heal itself with a scar. Compared with other tissues the difference is that, during bone repairing and regeneration, after the inflammatory phase the mesenchymal stem cells (MSCs) are recruited to the injury site and differentiate into either chondroblasts or osteoblasts precursors, leading to bone repairing and regeneration. Besides these two precursors, the MSCs can also differentiate into adipocyte precursors, skeletal muscle precursors and some other mesodermal cells. With this multilineage potentiality, the MSCs are probably used to cure bone injury and other woundings in the near future. Here we will introduce the recent developments in understanding the mechanism of MSCs action in bone regeneration and repairing.

Introduction

In 1964 Till et al. gave us the idea and definition of stem cell-self-renewal and differentiation [1]. Nowadays clinically it is a common understanding that the regenerative medicine is dependent on the properties of stem cells [2]. Mesenchymal stem cells (MSCs) are thought to be essential for bone-healing because of its ability to differentiate in vitro into both chondrocytes and osteocytes, and provide great potential for tissue engineering.

Tissue engineering is to replace tissues through the usage of specific cell types when the tissue function or structure is lost because of trauma or disease. Usually a 3D scaffold with various growth factors, cytokines, hormones and other biological molecules will be utilized during the engineering, in order to facilitate the generation of a critical mass of specific precursor cells and their differentiation into the required cell type.

As the key process in tissue engineering is to generate enough amount of a certain cell type, stem cells are thought to be the proper starting cells in clinical practice. They could be collected from various tissues, for example bone marrow, adipose, amniotic fluid, umbilical cord blood and even breast milk [36]. Among these origins, bone marrow-derived MSCs are the most commonly utilized stem cells for bone repairing, both in research and clinical practice [7]. There are several reports indicating that the transplantation of bone marrow-derived MSCs promotes bone formation at sites of bone defects [810]. In this review we will give a brief introduction of MSCs and importance of its usage in tissue repairing.

Characteristics of MSCs

Self-renewal is a hallmark of all stem cells [1]. The maintenance of self-renewal and pluripotency of stem cells occurs in the stem cell niche, where stem cells are able to receive signals from the stroma or other cells via contact or secreted molecules within this niche [11, 12]. Adult MSCs also share this kind of ability and could differentiate not only into osteoblasts, adipocytes, and chondrocytes, but also into cells of non-mesodermal lineages including hepatocytes, neuron-like cells, and pancreatic cells [1317].

MSCs could be easily distinguished from other bone marrow cells by their rapid adherence to plastic and their fibroblast-like morphology [18]. For decades cultured plastic-adherent bone marrow cells are the main source of MSCs, but now MSCs could also be derived from other tissues, such as placenta, adipose tissue, peripheral blood, umbilical blood and so on [1922]. In vitro bone marrow-derived MSCs express the surface markers CD105, CD90 and CD73, but lack CD45, CD34, CD14, CD11b, CD7a and CD19 [2326]. There are also some MSCs expressing markers, such as CD271 [23]. However, it is lack of evidences showing all the human MSCs have those markers. MSCs derived from different sources also vary in their differentiation capacity. As bone marrow-derived MSCs have better chondrogenic differentiation potential, they are considered to be a proper source for the therapy which facilitates bone repairing and regeneration [27, 28].

Besides the differentiation potential, MSCs also have immune-suppressive activity, which together with their paracrine function plays an essential role in bone regeneration and repairing [28]. This issue will be discussed in the section “Mechanism of MSCs action”.

Differentiation capacity of MSCs

Bone marrow-derived MSCs for Bone Engineering

MSCs exist in most of the postnatal tissues, and they all have differentiation capacity and the potential to be utilized in tissue engineering. Theoretically all the MSCs have the ability to differentiate towards osteogenic, chondrogenic and adipogenic lineages. They could be successfully expanded in vitro and induced to different types of subsets when the optimal cultivation protocols were used [29, 30]. As bone marrow-derived MSCs have the strongest chondrogenic differentiation potential, they are thought to be a better choice for bone engineering than the other MSCs sources [27].

Chondrogenic and osteogenic potential of MSCs in vitro

Even though MSCs with different origins have differences in differentiation capacity, all of them could be derived into osteoblasts and chondrocytes, which are essential in bone repairing. The tendency to differentiate into a certain cell lineage is usually dependent on the culture conditions. Bone marrow-derived MSCs have the highest potential to generate chondrocytes and osteoblasts. And the standard protocol for chondrogenesis was first established for bone marrow-derived MSCs. In this standard protocol growth factors from transforming growth factor β (TGF-β) superfamily are the key factors which facilitate chondrogenic differentiation of MSCs [31, 32]. Previous studies showed that these factors regulate MSCs differentiation via several signaling pathways, such as Smad, extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase pathways [3336].

MSCs derived from other tissues may also be used in clinic because of their advantages. For example, adipose-derived MSCs are easy to obtain, and umbilical cord-derived MSCs have high differentiation capacity and can be stored after birth and used as autologous implantation or tissue engineering. In cultivation with some certain growth factors such as TGF-β and bone morphogenetic protein-6 (BMP-6), adipose-derived MSCs can express cartilage-specific proteins such as type II collagen, but they do not express hypertrophic chondrocyte markers such as type X collagen [3739]. In 2010 a standard protocol for isolation of adipose-derived MSCs and induction to chondrogenesis was published in Nature, which is very useful in practice [38]. However, in comparison with bone marrow-derived MSCs, the chondrogenic potential of adipose-derived MSCs is quite limited.

Apart from the prominent advantages of umbilical cord-derived MSCs, such as a painless collection procedure and fast self-renewal, they also have a very high differentiation capacity including chondrogenic potential. Not like bone marrow-derived MSCs, umbilical cord-derived MSCs show a gene expression pattern more similar to embryonic stem cells (ESCs), and with proper expansion they could be used as a therapy not only for bone injury but also for other tissues repairing, even for neural injury. The most appreciable advantage of umbilical cord-derived MSCs is that the collection procedure is noninvasive and ethically acceptable [4042]. Maybe in the future with a proper inducing method, umbilical cord-derived MSCs could be a better choice for bone engineering.

Influence of oxygen and mechanical stimulation on MSCs differentiation

Besides the culture and growth factors, O2 partial pressure and mechanical stimulation are also important factors influencing MSCs differentiation. Usually cells are cultured under normoxic (21 % O2) conditions. However, most of the cells in vivo are not under such a high O2 concentration. For example, there is no vascular tissue in cartilage, thus the microenvironment in cartilage is hypoxic, where the O2 concentration is about 1–5 % [43]. Bone marrow is also under hypoxic condition in vivo and the O2 concentration there is about 1–7 % [44]. Though adipose tissue is a vascularized tissue, the O2 concentration there is only about 10–15 % [45]. It has been showed that hypoxia promotes chondrogenesis rather than osteogenesis [4649]. When bone marrow-derived MSCs are expanded under low O2 concentration (5 %), the subsequent osteogenesis would be enhanced [50]. In practice, it is a simple way to control the differentiation of MSCs using O2. Furthermore, the cell fate can be changed without adding some expensive biological factors.

Mechanical forces, which are generated within the cell in response to its extracellular environment and extrinsic mechanical signals, play a central role in determining MSC fate. Though the exact mechanism of mechanical sensitivity and respond is still not clear, it is believed that integrins and ion channels participate in this mechanotransduction progress [51]. It has been demonstrated that MSCs respond to oscillatory fluid flow with intracellular Ca2+ transient increasing, and then increase the proliferation rate, up-regulate osteoblastic gene expression and down-regulate alkaline phosphatase activity [52]. The fact that oscillatory fluid flow could up-regulate the expression of Runx2, Sox9 and PPARγ in MSCs, suggested that this kind of mechanical stimuli could regulate gene expression of some transcription factors which participate in MSCs differentiation [53, 54].

Mechanisms of MSCs action in bone regeneration and repairing

MSCs contribute to bone regeneration and repairing

With the achievements in the identification, isolation, and cultivation of MSCs, it is not difficult to develop improved technologies for defining therapeutic MSCs and MSC-based bone repairing. In 1998 a study demonstrated that implantation of a bone marrow-derived MSCs supplemented scaffold leaded to bone regeneration in bone fracture. On the other hand, implanted scaffold without MSCs had no such function [55]. Further studies indicated that pre-differentiated osteogenetic MSCs supplemented scaffold had superior healing effects [56, 57]. More interestingly, delivery of MSCs into clinical models of diabetes resulted in higher fracture healing activity than those did not received MSCs [58].

It is believed that MSCs can differentiate into skeletal progenitor cells which generate skeletal tissues in vivo. Transplanted CD45CD146+ human bone marrow-derived MSCs have the capacity to generate ectopic bones and hematopoietic microenvironment in vivo [59]. Further studies by Omatsu et al. demonstrated that perivascular MSCs which express CXCL12 were essential for hematopoietic stem cells (HSCs) proliferation [60], and in this way play a role in angiogenesis. As we know, osteogenesis is strongly associated with angiogenesis, it is not difficult to speculate that angiogenesis also contributes to bone regeneration and repairing.

In the early days bone marrow-derived MSCs were thought to achieve their functions via replacing the cells in damaged tissues, but recently it was found that MSCs could also provide paracrine signals to repair vascular injury or modulate pathological immune responses [61]. This issue will be discussed later.

MSCs homing to the injury site

The way MSCs home to the injury site is still not clear, anyway chemoattractant molecules released at the bone injury site must play an essential role in MSCs attracting. It has been known that MSCs express at least 19 chemokine receptors [62]. Stromal cell-derived factor 1 (SDF-1) expressed by stromal niche is the primary attractant for CXCR4-expressing MSCs and during trauma CXCR4 is up-regulated by MSCs [63, 64]. Many other chemotactic factors such as RANTES, macrophage inflammatory protein-1α (MIP-1α), monocyte chemotactic protein 1 (MCP-1) and so on, also work on MSCs. All these indicate that MSCs homing is attractant/receptor dependent [65]. However, the negative side of the homing property of MSCs is that they may home to other tissues even developing tumors [66, 67] or undergo necrosis/apoptosis, which is very harmful.

Direct and indirect contributions of MSCs

MSCs display a broad differentiation capacity in vitro, it was originally hypothesized MSCs transplantation would induce tissue repairing by replacing cells in the damaged host tissue. During bone repairing, the progenitor cells will migrate to the injury site and differentiate into osteoblasts and chondrocytes, and then lead to bone remodeling [68, 69].

Despite the long-lasting therapeutic efficacy of MSCs in many in vivo models (such as bone and cartilage repairing, cardiovascular and neurological diseases), the incidence of MSCs engraftment remained surprisingly low [70, 71]. This unexpected low engraftment efficacy implied a major challenge to explain the beneficial effects of the MSCs. Accumulating evidences indicated that the general therapeutic effects of MSCs are due to their ability to modify the host micro-environment rather than their capacity to differentiate and incorporate into the host tissue.

In mice it has been observed that transplanted MSCs migrate to the site of fractures, integrate into the callus and secrete BMP-2 which acts as a disulfide-linked homodimer and induces bone and cartilage formation. It is a candidate as a retinoid mediator, playing a key role in osteoblast differentiation [72]. Adding vascular endothelial growth factor (VEGF) together with BMPs could facilitate both osteogenesis and angiogenesis, leading to higher bone formation activity and mechanical strength [73]. Transplanted osteoblastic progenitors repair the fracture via intramembranous ossification, while less differentiated MSCs lead to endochondral ossification, a process reminiscent of embryonic skeletal development. This indicates that bone repairing function of MSCs is based on its maturation state [74].

It is commonly accepted that osteogenesis is associated with angiogenesis. Studies by De Luca et al. demonstrated that the secretion of angiogenic factors by MSCs could be increased when trauma happened. Under chemokines and hypoxic conditions, TGF-α induces the secretion of a range of growth factors such as VEGF, hepatocyte growth factor (HGF), interleukin 6 (IL-6), IL-8, platelet-derived growth factor subunit B (PDGF-BB), angiopoietin-2 (Ang-2) and so on in bone marrow-derived MSCs via MEK/MAPK and PI3 K/AKT pathways [75]. The angiogenesis process is very complicated involving endothelial cell (EC) survival, proliferation, migration, tube formation and maturation. With the help of MSCs paracrine signal, angiogenesis could be enhanced. In turn this angiogenic function changes the micro-environment of the damaged tissue and then benefits osteogenesis.

The ability to regulate immune response is another important contribution of MSCs during bone repairing. It was reported that MSCs transplantation leads to higher survival rates in graft-versus-host disease than usual [76]. MSCs modulate the inflammatory micro-environment at the trauma site and decrease the levels of IL-1β, IL-6 and tumor necrosis factor-α (TNF-α) [72]. As TNF-α is up-regulated in the injury or fracture sites [77], the function of MSCs results in a limited inflammation reaction which facilitates osteogenesis and angiogenesis and in turn contributes to bone repairing. It is now clear that TNF-α recruits osteoclasts and MSCs to the trauma site and causes apoptosis of hypertrophic chondrocytes in order to facilitate endochondral ossification [78]. Because of the osteoclastic activity, high level of TNF-α can inhibit bone repairing [79]. Thus transplantation of MSCs could regulate exaggerated immune responses and then facilitates fracture repair.

Another study demonstrated that factors released by bone marrow derived MSCs could recruit macrophages and endothelial lineage cells into the wound thus enhancing wound healing [80]. Together, the transplanted MSCs could modify the local micro-environment such as angiogenesis, and suppress exaggerated immune reaction, and therefore lead to bone repairing. Even though the detailed mechanism of the exogenously implanted MSCs therapy is still not clear, its potential is quite promising.

Now there are also evidences indicating that MSCs could possibly transfer mitochondria or vesicular components which contain mRNA, miRNA or proteins [81]. Generally speaking, the basic mechanisms of MSCs action on bone regeneration and repairing are as following (Fig. 1): replacing the damaged cells by proliferation and differentiation; the modulation of the immune system; the secretion of factors that induce tissue repair; recruitment of endogenous MSCs or progenitor cells to the injury site; possible transfer of mitochondria or vesicular components containing mRNA, miRNA and proteins.

Fig. 1
figure 1

Proposed mechanisms of action of MSCs transplantation. Paracrine effects of MSCs include the stimulation of angiogenesis, protection of other cells from apoptosis, recruitment of host MSCs or other progenitor cells and stimulation of their proliferation and differentiation [82, 83]. The immunomodulatory effects of MSC consist of inhibiting the proliferation and activity of neutrophils, NK cells, B cells, CD4+ cells and CD8+ T cells, preventing the maturation of monocytes into dendritic cells, suppressing plasma cell immunoglobulin production but stimulating proliferation of regulatory T cells [84]. In some physiological settings, MSCs are able to differentiate into multiple cell types and transfer vesicles containing mRNA, miRNA, proteins and perhaps mitochondria to the host cells [81, 84, 85]

Discussion

Although MSCs were studied because of their differentiation capacity, there is now accumulating evidences suggesting that immunomodulatory and paracrine actions predominantly contribute to their therapeutic efficacy. Some molecules such as VEGF, BMP-2 and MCP-1 play essential roles in the mechanism of MSCs induced tissue regeneration.

Since 1995 more than 3000 patients have received autologous culture-expanded MSCs implantation as a therapy, no serious adverse reaction was reported [86, 87]. However, some unsolved issues still prevent these promising cells to become a realistic treatment option for the future, for example, the absence of a unique marker for MSCs, unstable MSCs isolation and expansion, and recent pre-clinical reports raise the concern that MSCs may cause harmful ectopic bone formation [88]. In order to solve these problems, more preclinical researches about their differentiation capacity and potential effect on the microenvironment need to be done.

In conclusion, over the last decade, numerous studies have been done on MSCs. This is evidenced by the successful application in a wide variety of in vivo wound healing, resulting in an increasing optimism of both basic scientists and clinicians. Besides bone regeneration and repairing, MSCs could be engineered for anti-cancer treatments, designed as carriers for certain factors or genes [89]. Though it is now still impossible to let MSCs become an everyday therapeutic option, routine clinical application of MSCs for a wide variety of pathologies associated with limited angiogenesis, osteogenesis and chondrogenesis is an exciting prospect.

Abbreviations

BMP-2:

bone morphogenetic protein 2

CFU-F:

colony-forming unit fibroblast

ESCs:

embryonic stem cells

HSCs:

hematopoietic stem cells

MSCs:

mesenchymal stem cells

PF-4:

platelet factor-4

SDF-1:

stromal cell-derived factor 1

TGF-α:

transformation growth factor α

VEGF:

vascular endothelial growth factor

References

  1. Till JE, McCulloch EA, Siminovitch L. A Stochastic MODEL OF stem cell proliferation, based on the growth of spleen colony-forming cells. Proc Natl Acad Sci USA. 1964;51:29–36.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Andrews PW. Toward safer regenerative medicine. Nat Biotechnol. 2011;29:803–5.

    Article  CAS  PubMed  Google Scholar 

  3. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA. 2000;97:13625–30.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109:235–42.

    Article  CAS  PubMed  Google Scholar 

  6. Patki S, Kadam S, Chandra V, Bhonde R. Human breast milk is a rich source of multipotent mesenchymal stem cells. Hum Cell. 2010;23:35–40.

    Article  CAS  PubMed  Google Scholar 

  7. Colnot C. Cell sources for bone tissue engineering: insights from basic science. Tissue Eng Part B Rev. 2011;17:449–57.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. An SH, Matsumoto T, Miyajima H, Nakahira A, Kim KH, Imazato S. Porous zirconia/hydroxyapatite scaffolds for bone reconstruction. Dent Mater. 2012;28:1221–31.

    Article  CAS  PubMed  Google Scholar 

  9. Vulcano E, Murena L, Falvo DA, Baj A, Toniolo A, Cherubino P. Bone marrow aspirate and bone allograft to treat acetabular bone defects in revision total hip arthroplasty: preliminary report. Eur Rev Med Pharmacol Sci. 2013;17(16):2240–9.

    CAS  PubMed  Google Scholar 

  10. Chen KY, Dong GC, Hsu CY, Chen YS, Yao CH. Autologous bone marrow stromal cells loaded onto porous gelatin scaffolds containing Drynaria fortunei extract for bone repair. J Biomed Mater Res A. 2013;101:954–62.

    Article  PubMed  Google Scholar 

  11. Bianco P. Minireview: the stem cell next door: skeletal and hematopoietic stem cell “niches” in bone. Endocrinology. 2011;152:2957–62.

    Article  CAS  PubMed  Google Scholar 

  12. Jones DL, Wagers AJ. No place like home: anatomy and function of the stem cell niche. Nat Rev Mol Cell Biol. 2008;9:11–21.

    Article  CAS  PubMed  Google Scholar 

  13. Aurich H, Sgodda M, Kaltwasser P, Vetter M, Weise A, Liehr T, et al. Hepatocyte differentiation of mesenchymal stem cells from human adipose tissue in vitro promotes hepatic ntegration in vivo. Gut. 2009;58:570–81.

    Article  CAS  PubMed  Google Scholar 

  14. Pino AM, Rosen CJ, Rodríguez JP. In osteoporosis, differentiation of mesenchymal stem cells (MSCs) improves bone marrow adipogenesis. Biol Res. 2012;45(3):279–87.

    Article  CAS  PubMed  Google Scholar 

  15. Liang J, Wu S, Zhao H, Li SL, Liu ZX, Wu J, et al. Human umbilical cord mesenchymal stem cells derived from Wharton’s jelly differentiate into cholinergic-like neurons in vitro. Neurosci Lett. 2013;532:59–63.

    Article  CAS  PubMed  Google Scholar 

  16. Li D, Chai J, Shen C, Han Y, Sun T. Human umbilical cord-derived mesenchymal stem cells differentiate into epidermal-like cells using a novel co-culture technique. Cytotechnology. 2014;66:699–708.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Jin P, Zhang X, Wu Y, Li L, Yin Q, Zheng L, et al. Streptozotocin-induced diabetic rat-derived bone marrow mesenchymal stem cells have impaired abilities in proliferation, paracrine, antiapoptosis, and myogenic differentiation. Transplant Proc. 2010;42:2745–52.

    Article  CAS  PubMed  Google Scholar 

  18. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3:393–403.

    CAS  PubMed  Google Scholar 

  19. Puglisi MA, Saulnier N, Piscaglia AC, Tondi P, Agnes S, Gasbarrini A. Adipose tissue-derived mesenchymal stem cells and hepatic differentiation: old concepts and futureperspectives. Eur Rev Med Pharmacol Sci. 2011;15(4):355–64.

    CAS  PubMed  Google Scholar 

  20. Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, et al. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol. 2008;214:413–21.

    Article  CAS  PubMed  Google Scholar 

  21. Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, Moss J, Burger JA, et al. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res. 2000;2:477–88.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  22. Jin HJ, Nam HY, Bae YK, Kim SY, Im IR, Oh W, et al. GD2 expression is closely associated with neuronal differentiation of human umbilical cord blood-derived mesenchymal stem cells. Cell Mol Life Sci. 2010;67:1845–58.

    Article  CAS  PubMed  Google Scholar 

  23. Alvarez-Viejo M, Menendez-Menendez Y, Otero-Hernandez J. CD271 as a marker to identify mesenchymal stem cells from diverse sources before culture. World J Stem Cells. 2015;7:470–6.

    Article  PubMed Central  PubMed  Google Scholar 

  24. Pogozhykh O, Pogozhykh D, Neehus AL, Hoffmann A, Blasczyk R, Muller T. Molecular and cellular characteristics of human and non-human primate multipotent stromal cells from the amnion and bone marrow during long term culture. Stem Cell Res Ther. 2015;6:150.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Harichandan A, Buhring HJ. Prospective isolation of human MSC. Best Pract Res Clin Haematol. 2011;24:25–36.

    Article  CAS  PubMed  Google Scholar 

  26. Buhring HJ, Battula VL, Treml S, Schewe B, Kanz L, Vogel W. Novel markers for the prospective isolation of human MSC. Ann N Y Acad Sci. 2007;1106:262–71.

    Article  PubMed  Google Scholar 

  27. Singh J, Onimowo JO, Khan WS. Bone marrow derived stem cells in trauma and orthopaedics: a review of the current trend. Curr Stem Cell Res Ther. 2014;10:37–42.

    Article  PubMed  Google Scholar 

  28. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31:890–6.

    Article  PubMed  Google Scholar 

  29. Dasari VR, Veeravalli KK, Dinh DH. Mesenchymal stem cells in the treatment of spinal cord injuries: a review. World J Stem Cells. 2014;6:120–33.

    Article  PubMed Central  PubMed  Google Scholar 

  30. Hatlapatka T, Moretti P, Lavrentieva A, Hass R, Marquardt N, Jacobs R, et al. Optimization of culture conditions for the expansion of umbilical cord-derived mesenchymal stem or stromal cell-like cells using xeno-free culture conditions. Tissue Eng Part C Methods. 2011;17:485–93.

    Article  PubMed  Google Scholar 

  31. Brunner D, Frank J, Appl H, Schoffl H, Pfaller W, Gstraunthaler G. Serum-free cell culture: the serum-free media interactive online database. ALTEX. 2010;27:53–62.

    PubMed  Google Scholar 

  32. Mannello F, Tonti GA. Concise review: no breakthroughs for human mesenchymal and embryonic stem cell culture: conditioned medium, feeder layer, or feeder-free; medium with fetal calf serum, human serum, or enriched plasma; serum-free, serum replacement nonconditioned medium, or ad hoc formula? All that glitters is not gold! Stem Cells. 2007;25:1603–9.

    Article  CAS  PubMed  Google Scholar 

  33. Wang L, Yang Y, Zhu Y, Ma X, Liu T, Zhang G, et al. Characterization of placenta-derived mesenchymal stem cells cultured in autologous human cord blood serum. Mol Med Rep. 2012;6:760–6.

    CAS  PubMed  Google Scholar 

  34. Montoya F, Martínez F, García-Robles M, Balmaceda-Aguilera C, Koch X, Rodríguez F, et al. Clinical and experimental approaches to knee cartilage lesion repair and mesenchymal stem cell chondrocyte differentiation. Biol Res. 2013;46(4):441–51.

    Article  PubMed  Google Scholar 

  35. Doucet C, Ernou I, Zhang Y, Llense JR, Begot L, Holy X, et al. Platelet lysates promote mesenchymal stem cell expansion: a safety substitute for animal serum in cell-based therapy applications. J Cell Physiol. 2005;205:228–36.

    Article  CAS  PubMed  Google Scholar 

  36. Rodriguez IA, Growney KEA, Bowlin GL, Sell SA. Platelet-rich plasma in bone regeneration: engineering the delivery for improved clinical efficacy. Biomed Res Int. 2014;2014:392398.

    PubMed Central  PubMed  Google Scholar 

  37. Londin ER, Hatzimichael E, Loher P, Edelstein L, Shaw C, Delgrosso K, et al. The human platelet: strong transcriptome correlations among individuals associate weakly with the platelet proteome. Biol Direct. 2014;9:3.

    Article  PubMed Central  PubMed  Google Scholar 

  38. Lange C, Cakiroglu F, Spiess AN, Cappallo-Obermann H, Dierlamm J, Zander AR. Accelerated and safe expansion of human mesenchymal stromal cells in animal serum-free medium for transplantation and regenerative medicine. J Cell Physiol. 2007;213:18–26.

    Article  CAS  PubMed  Google Scholar 

  39. Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO, Pittenger MF. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng. 1998;4:415–28.

    Article  CAS  PubMed  Google Scholar 

  40. Jaing TH. Umbilical cord blood: a trustworthy source of multipotent stem cells for regenerative medicine. Cell Transplant. 2014;23:493–6.

    Article  PubMed  Google Scholar 

  41. Peters R, Wolf MJ, van den Broek M, Nuvolone M, Dannenmann S, Stieger B, et al. Efficient generation of multipotent mesenchymal stem cells from umbilical cord blood in stroma-free liquid culture. PLoS One. 2010;5:e15689.

    Article  PubMed Central  PubMed  Google Scholar 

  42. Nagamura-Inoue T, He H. Umbilical cord-derived mesenchymal stem cells: their advantages and potential clinical utility. World J Stem Cells. 2014;6:195–202.

    Article  PubMed Central  PubMed  Google Scholar 

  43. Lafont JE. Lack of oxygen in articular cartilage: consequences for chondrocyte biology. Int J Exp Pathol. 2010;91:99–106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Kofoed H, Sjontoft E, Siemssen SO, Olesen HP. Bone marrow circulation after osteotomy. Blood flow, pO2, pCO2, and pressure studied in dogs. Acta Orthop Scand. 1985;56:400–3.

    Article  CAS  PubMed  Google Scholar 

  45. Bizzarri A, Koehler H, Cajlakovic M, Pasic A, Schaupp L, Klimant I, et al. Continuous oxygen monitoring in subcutaneous adipose tissue using microdialysis. Anal Chim Acta. 2006;573–574:48–56.

    Article  PubMed  Google Scholar 

  46. Hirao M, Tamai N, Tsumaki N, Yoshikawa H, Myoui A. Oxygen tension regulates chondrocyte differentiation and function during endochondral ossification. J Biol Chem. 2006;281:31079–92.

    Article  CAS  PubMed  Google Scholar 

  47. Scherer K, Schunke M, Sellckau R, Hassenpflug J, Kurz B. The influence of oxygen and hydrostatic pressure on articular chondrocytes and adherent bone marrow cells in vitro. Biorheology. 2004;41:323–33.

    CAS  PubMed  Google Scholar 

  48. Krinner A, Zscharnack M, Bader A, Drasdo D, Galle J. Impact of oxygen environment on mesenchymal stem cell expansion and chondrogenic differentiation. Cell Prolif. 2009;42:471–84.

    Article  CAS  PubMed  Google Scholar 

  49. Robins JC, Akeno N, Mukherjee A, Dalal RR, Aronow BJ, Koopman P, et al. Hypoxia induces chondrocyte-specific gene expression in mesenchymal cells in association with transcriptional activation of Sox9. Bone. 2005;37:313–22.

    Article  CAS  PubMed  Google Scholar 

  50. Sheehy EJ, Buckley CT, Kelly DJ. Oxygen tension regulates the osteogenic, chondrogenic and endochondral phenotype of bone marrow derived mesenchymal stem cells. Biochem Biophys Res Commun. 2012;417:305–10.

    Article  CAS  PubMed  Google Scholar 

  51. Liu L, Yuan W, Wang J. Mechanisms for osteogenic differentiation of human mesenchymal stem cells induced by fluid shear stress. Biomech Model Mechanobiol. 2010;9:659–70.

    Article  PubMed  Google Scholar 

  52. Li YJ, Batra NN, You L, Meier SC, Coe IA, Yellowley CE, et al. Oscillatory fluid flow affects human marrow stromal cell proliferation and differentiation. J Orthop Res. 2004;22:1283–9.

    Article  CAS  PubMed  Google Scholar 

  53. Arnsdorf EJ, Tummala P, Kwon RY, Jacobs CR. Mechanically induced osteogenic differentiation–the role of RhoA, ROCKII and cytoskeletal dynamics. J Cell Sci. 2009;122:546–53.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Arnsdorf EJ, Tummala P, Castillo AB, Zhang F, Jacobs CR. The epigenetic mechanism of mechanically induced osteogenic differentiation. J Biomech. 2010;43:2881–6.

    Article  PubMed Central  PubMed  Google Scholar 

  55. Agacayak S, Gulsun B, Ucan MC, Karaoz E, Nergiz Y. Effects of mesenchymal stem cells in critical size bone defect. Eur Rev Med Pharmacol Sci. 2012;16(5):679–86.

    CAS  PubMed  Google Scholar 

  56. Petite H, Viateau V, Bensaid W, Meunier A, de Pollak C, Bourguignon M, et al. Tissue-engineered bone regeneration. Nat Biotechnol. 2000;18:959–63.

    Article  CAS  PubMed  Google Scholar 

  57. Yamada Y, Ueda M, Naiki T, Nagasaka T. Tissue-engineered injectable bone regeneration for osseointegrated dental implants. Clin Oral Implants Res. 2004;15:589–97.

    Article  PubMed  Google Scholar 

  58. Breitbart EA, Meade S, Azad V, Yeh S, Al-Zube L, Lee YS, et al. Mesenchymal stem cells accelerate bone allograft incorporation in the presence of diabetes mellitus. J Orthop Res. 2010;28:942–9.

    PubMed  Google Scholar 

  59. Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007;131:324–36.

    Article  CAS  PubMed  Google Scholar 

  60. Omatsu Y, Sugiyama T, Kohara H, Kondoh G, Fujii N, Kohno K, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity. 2010;33:387–99.

    Article  CAS  PubMed  Google Scholar 

  61. Li F, Whyte N, Niyibizi C. Differentiating multipotent mesenchymal stromal cells generate factors that exert paracrine activities on exogenous MSCs: implications for paracrine activities in bone regeneration. Biochem Biophys Res Commun. 2012;426:475–9.

    Article  CAS  PubMed  Google Scholar 

  62. Wu Y, Zhao RC. The role of chemokines in mesenchymal stem cell homing to myocardium. Stem Cell Rev. 2012;8:243–50.

    Article  CAS  PubMed  Google Scholar 

  63. Kitaori T, Ito H, Schwarz EM, Tsutsumi R, Yoshitomi H, Oishi S, Nakano M, et al. Stromal cell-derived factor 1/CXCR4 signaling is critical for the recruitment of mesenchymal stem cells to the fracture site during skeletal repair in a mouse model. Arthritis Rheum. 2009;60:813–23.

    Article  CAS  PubMed  Google Scholar 

  64. Liu H, Liu S, Li Y, Wang X, Xue W, Ge G, et al. The role of SDF-1-CXCR4/CXCR7 axis in the therapeutic effects of hypoxia-preconditioned mesenchymal stem cells for renal ischemia/reperfusion injury. PLoS One. 2012;7:e34608.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  65. Ito H. Chemokines in mesenchymal stem cell therapy for bone repair: a novel concept of recruiting mesenchymal stem cells and the possible cell sources. Mod Rheumatol. 2011;21:113–21.

    Article  CAS  PubMed  Google Scholar 

  66. Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J, et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65:3307–18.

    CAS  PubMed  Google Scholar 

  67. Dwyer RM, Khan S, Barry FP, O’Brien T, Kerin MJ. Advances in mesenchymal stem cell-mediated gene therapy for cancer. Stem Cell Res Ther. 2010;1:25.

    Article  PubMed Central  PubMed  Google Scholar 

  68. Yu DA, Han J, Kim BS. Stimulation of chondrogenic differentiation of mesenchymal stem cells. Int J Stem Cells. 2012;5:16–22.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7.

    Article  CAS  PubMed  Google Scholar 

  70. Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, et al. 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. 2002;99:8932–7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  71. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25:2648–59.

    Article  CAS  PubMed  Google Scholar 

  72. Granero-Molto F, Weis JA, Miga MI, Landis B, Myers TJ, O’Rear L, et al. Regenerative effects of transplanted mesenchymal stem cells in fracture healing. Stem Cells. 2009;27:1887–98.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  73. Ren G, Chen X, Dong F, Li W, Ren X, Zhang Y, et al. Concise review: mesenchymal stem cells and translational medicine: emerging issues. Stem Cells Transl Med. 2012;1:51–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  74. Tortelli F, Tasso R, Loiacono F, Cancedda R. The development of tissue-engineered bone of different origin through endochondral and intramembranous ossification following the implantation of mesenchymal stem cells and osteoblasts in a murine model. Biomaterials. 2010;31:242–9.

    Article  CAS  PubMed  Google Scholar 

  75. De Luca A, Gallo M, Aldinucci D, Ribatti D, Lamura L, D’Alessio A, et al. Role of the EGFR ligand/receptor system in the secretion of angiogenic factors in mesenchymal stem cells. J Cell Physiol. 2011;226:2131–8.

    Article  PubMed  Google Scholar 

  76. Li ZY, Wang CQ, Lu G, Pan XY, Xu KL. Effects of bone marrow mesenchymal stem cells on hematopoietic recovery and acute graft-versus-host disease in murine allogeneic umbilical cord blood transplantation model. Cell Biochem Biophys. 2014;70:115–22.

    Article  CAS  PubMed  Google Scholar 

  77. Scotti C, Piccinini E, Takizawa H, Todorov A, Bourgine P, Papadimitropoulos A, et al. Engineering of a functional bone organ through endochondral ossification. Proc Natl Acad Sci USA. 2013;110:3997–4002.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  78. Gerstenfeld LC, Cho TJ, Kon T, Aizawa T, Tsay A, Fitch J, et al. Impaired fracture healing in the absence of TNF-alpha signaling: the role of TNF-alpha in endochondral cartilage resorption. J Bone Miner Res. 2003;18:1584–92.

    Article  CAS  PubMed  Google Scholar 

  79. Alblowi J, Kayal RA, Siqueira M, McKenzie E, Krothapalli N, McLean J, et al. High levels of tumor necrosis factor-alpha contribute to accelerated loss of cartilage in diabetic fracture healing. Am J Pathol. 2009;175:1574–85.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  80. Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3:e1886.

    Article  PubMed Central  PubMed  Google Scholar 

  81. Spees JL, Olson SD, Whitney MJ, Prockop DJ. Mitochondrial transfer between cells can rescue aerobic respiration. Proc Natl Acad Sci USA. 2006;103:1283–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  82. Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 2009;20:419–27.

    Article  PubMed  Google Scholar 

  83. Hocking AM, Gibran NS. Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res. 2010;316:2213–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Ben-Ami E, Berrih-Aknin S, Miller A. Mesenchymal stem cells as an immunomodulatory therapeutic strategy for autoimmune diseases. Autoimmun Rev. 2011;10:410–5.

    Article  CAS  PubMed  Google Scholar 

  85. Bieback K, Wuchter P, Besser D, Franke W, Becker M, Ott M, et al. Mesenchymal stromal cells (MSCs): science and f(r)iction. J Mol Med (Berl). 2012;90:773–82.

    Article  Google Scholar 

  86. Lazarus HM, Haynesworth SE, Gerson SL, Rosenthal NS, Caplan AI. Ex vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): implications for therapeutic use. Bone Marrow Transplant. 1995;16:557–64.

    CAS  PubMed  Google Scholar 

  87. Quarto R, Mastrogiacomo M, Cancedda R, Kutepov SM, Mukhachev V, Lavroukov A, et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med. 2001;344:385–6.

    Article  CAS  PubMed  Google Scholar 

  88. Tasso R, Ulivi V, Reverberi D, Lo SC, Descalzi F, Cancedda R. In vivo implanted bone marrow-derived mesenchymal stem cells trigger a cascade of cellular events leading to the formation of an ectopic bone regenerative niche. Stem Cells Dev. 2013;22:3178–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  89. Bronckaers A, Hilkens P, Martens W, Gervois P, Ratajczak J, Struys T, et al. Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate angiogenesis. Pharmacol Ther. 2014;143:181–96.

    Article  CAS  PubMed  Google Scholar 

Download references

Authors’ contributions

TY and WZ, the corresponding authors conceived of the study, participated in its design and coordination, and have critically examined and corrected the manuscript. JS and WZ carried out literature search, data analysis, participated in the design of the study and drafted the manuscript, and contributed equally to this work. All authors read and approved the final manuscript.

Acknowledgements

This work was supported by National Natural Science Foundation of China (81201367), Funding Scheme for Training Young Teachers in Shanghai Colleges (ZZjdyx12062), Key Discipline and Specialty Foundation of Shanghai Municipal Commission of Health and Family Planning, Seed Fund Research Training Program of Renji Hospital (RJZZ12-012), Key Discipline Construction Project of Pudong Health Bureau of Shanghai (PWZx2014-09), Shanghai Municipal Natural Science Foundation (10ZR1427700).

Competing interests

The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Weiwei Zhang or Tieyi Yang.

Additional information

Jin Shao and Weiwei Zhang contributed equally to this work

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shao, J., Zhang, W. & Yang, T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res 48, 62 (2015). https://doi.org/10.1186/s40659-015-0053-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40659-015-0053-4

Keywords