Skip to main content

Mesenchymal stem cells in tissue repairing and regeneration: Progress and future

Abstract

The presence of mesenchymal progenitor cells within bone marrow has been known since the late nineteenth century. To date, mesenchymal stem cells (MSCs) have been isolated from several different connective tissues, such as adipose tissue, muscle, placenta, umbilical cord matrix, blood, liver, and dental pulp. Bone marrow, however, is still one of the major sources of MSCs for preclinical and clinical research. MSCs were first evaluated for regenerative applications and have since been shown to directly influence the immune system and to promote neovascularization of ischemic tissues. These observations have prompted a new era of MSC transplantation as a treatment for various diseases. In this review, we summarize the important studies that have investigated the use of MSCs as a therapeutic agent for regenerative medicine, immune disorders, cancer, and gene therapy. Furthermore, we discuss the mechanisms involved in MSC-based therapies and clinical-grade MSC manufacturing.

Introduction

Knowledge of the presence of mesenchymal progenitor cells within bone marrow can be traced back to the late nineteenth century to the work of Goujon[1] and Biakow[2] on the heterotopic transplantation of rabbit marrow. The definitive evidence that bone marrow includes some non-hematopoietic, plastic-adherent precursor cells was originally provided by Friedenstein and his colleagues, who grew whole bone marrow cells in a medium supplemented with 10% selected fetal bovine serum to demonstrate the ability of a rare population of plastic-adherent cells (approximately 1 in 10,000 nucleated cells in the bone marrow) to form spindlelike and round-shaped colonies. Friedenstein defined these fibroblast precursors as fibroblast colony-forming units (CFU-F), analogous to hematopoietic colony-forming units.[3] As other groups subsequently extended these initial observations, the CFU-F progenitors were accepted to be multipotent and able to differentiate into adipocytes, osteoblasts, and chondrocytes. Arnold Caplan first used the term mesenchymal stem cells[4] based on the notion of a stromal stem cell proposed by Maureen Owen.[5] To date, mesenchymal stem cells (MSCs) have been reported to have been isolated from several different connective tissues, such as adipose tissue, muscle, placenta, umbilical cord matrix, blood, liver, and dental pulp. Bone marrow, however, is still one of the major sources for MSCs used in preclinical and clinical research.[6]

Table 1

Since the initial use of the term “MSC,” the stem cell identity of MSCs has been the subject of considerable debate. In 2000, a workshop at the Annual Meeting of the International Society for Cellular Therapy (ISCT) concluded that there was a lack of solid data in support of the ‘stemness’ of unfractionated plastic-adherent cells from bone marrow.[7] In 2005, the ISCT proposed “multipotent mesenchymal stromal cells” as a replacement term for “mesenchymal stem cells,” although the two terms shared the abbreviation “MSC”.[8] The ISCT then suggested minimal criteria for the definition of human MSCs: (1) MSCs must be plastic-adherent when maintained in standard culture conditions; (2) MSCs must express CD105, CD73, and CD90, while lacking expression of CD45, CD34, CD14 or CD11b, CD79a or CD19, and HLA-DR surface molecules; and (3) MSCs must be capable of differentiating into osteoblasts, adipocytes, and chondroblasts in vitro.[9] All criteria must be satisfied, as no single characteristic is sufficient for identifying MSCs. The bone marrow-derived plastic-adherent cell population also contains endothelial cells, fibroblasts, and macrophages. Contamination by hematopoietic and endothelial cells can be ruled out by the combination of cell surface markers. However, no specific markers currently exist that can reliably discriminate between passaged MSCs and fibroblasts. Furthermore, colony-forming capacity and differentiation potential are important specific properties that distinguish MSCs from fibroblasts.[10] Recently, it was reported that the level of expression of CD166 was significantly higher in MSCs than in fibroblasts, while the expression level of CD9 was significantly lower. CD146 was found to be exclusively expressed in MSCs; however, CD146 was downregulated and CD9 was upregulated with the passage of MSCs. The expression levels of all other markers were unchanged.[11]

Initially heralded as stem cells, MSCs were first evaluated for regenerative applications. MSCs have since been shown to directly influence the immune system[12] and to promote the neovascularization of ischemic tissues.[13,14] These observations have prompted MSC transplantation as a treatment for various diseases. In this review, we summarize the important studies of MSCs that describe the potential use of these cells as a therapeutic agent for regenerative medicine, immune disorders, cancer, and gene therapy. Furthermore, we discuss the mechanisms involved in MSC therapy as well as clinical-grade cell manufacturing of MSCs.

Identification of mesenchymal stem cells

Although MSCs have been isolated from many postnatal organs and tissues, bone marrow stroma is still the most recurrent tissue source utilized in cultivating MSCs. Most MSC populations have been isolated using methods similar to those originally used by Friedenstein and his colleagues.[15] In general, low-density mononuclear cells from normal human donors are plated in a basal medium supplemented with selected batches of fetal bovine serum, and the cells that readily adhere to plastic culture dishes and form large CFU-F clones are considered to be primary MSCs. Although endothelial cells, macrophages, lymphocytes, and differentiated smooth muscle cells can also adhere to plastic and contaminate the MSC culture, these cells are not successfully passaged and expanded in the specialized culture medium. After a few passages, the MSC cultures display a rather homogenous population of fibroblast-like cells. These cells lack CD11 and CD14 (monocyte and macrophage markers), CD34 (primitive HSCs and endothelial cells), CD45 (pan-leukocytes), CD19 (B cells), CD3 (T-cell receptor), CD31 (endothelial cells), and HLA-DR, but they show expression of CD29, CD144, CD166, CD105, and CD90. All of these markers are used retrospectively to identify MSCs via the elimination of endothelial and hematopoietic cell contaminants.[16] Immunofluorescence and immunohistochemical staining have also demonstrated that bone marrow (BM)-MSCs were positive for myofibroblastic markers, such as α-SMA, vimentin, fibronectin, and N-cadherin, but negative for epithelial markers, such as CK18 and E-cadherin.

Until now, the best marker for prospectively identifying MSCs has remained unclear. A few papers have reported the isolation of MSCs using surface markers such as Nestin,[17] stage specific embryonic antigen-1 (SSEA-1),[18] SSEA-4,[19] and Stro-1. Nestin-positive cells in human bone marrow represent true mesenchymal stem cells in that they show a close physical association with hematopoietic stem cells (HSCs) and can express a high level of core HSC maintenance genes. However, whether this putative MSC marker can be utilized to isolate MSCs from other tissues remains to be determined.

In addition to the morphologic and phenotypic characteristics of MSCs, one of the hallmarks for the identification of MSCs is their multipotent ability to differentiate into various lineages, including adipose, bone, cartilage, and myogenic cells.[9] To induce adipogenic differentiation, MSCs were cultured in regular medium supplemented with dexamethasone, insulin, isobutyl methyl xanthine, and indomethacin. Adipogenic differentiation was verified by the accumulation of lipid vacuoles, which were detected by Oil Red O staining. These differentiated cells also expressed adipogenic lineage-specific genes, such as peroxisome proliferation-activated receptor γ2 (PPARγ2), lipoprotein lipase (LPL), fatty acid-binding protein (aP2), adipsin, and leptin. To promote the differentiation of MSCs into osteoblasts, confluent monolayers of MSCs were incubated with ascorbic acid, ß-glycerol phosphate, and dexamethasone; in addition, bone morphogenic proteins (BMPs), particularly BMP-2 and BMP-6, were sometimes added to the induction system and strongly promoted osteogenesis in MSCs. Early osteogenic differentiation was evaluated by alkaline phosphatase staining, and late osteogenic differentiation was assessed by von Kossa staining for calcium mineralization detection. Meanwhile, specific gene expression of osteocalcin (OCN), osteopontin (OPN), Runx2, and alkaline phosphatase (ALP) was determined to confirm osteoblastic differentiation. For chondrogenic differentiation, MSCs were usually centrifuged gently to form a micromass and then cultured in serum-free High Glucose Dulbecco’s Modified Eagle Media (HG-DMEM) and supplemented with dexamethasone, Insulin-Transferrin- Selenium (ITS), ascorbic acid, sodium pyruvate, proline, and transforming growth factor-ß3. The development and accumulation of cartilage matrix was shown by staining proteoglycans with toluidine blue, alcian blue, and safranin O. Differentiated MSCs also express chondrogenic-specific genes, such as type II collagen and aggrecan.[20] Further studies have shown that MSCs can also differentiate into bone marrow stromal cells, tendon-ligament fibroblasts, myocytes, endothelial cells, neurons, and tenocytes.[21]

Mesenchymal stem cells for regenerative medicine

It is known that MSCs can be used for the treatment of injured myocardium, skin, pancreas, and bone. Although the exact mechanisms of action have not been fully elucidated, studies show that MSCs can act on several levels of endogenous repair to bring about treatment of disease. MSCs have been shown to directly promote tissue repair.[22,23] When administered to treat animals undergoing acute renal failure, MSCs promoted the proliferation of renal-tubule epithelial cells.[24] MSCs can directly protect and repair damaged ß-islets from autoimmune attacks [Figure 1].[25] MSCs can differentiate into osteoblasts and chondrocytes. The osteoblastic potential of MSCs may be exploited for treating long bone defect.[26] Although MSCs are effective for long bone reconstruction, MSCs are not effective for jaw reconstruction[27] because MSCs from the bone marrow cannot differentiate into osteoblasts when implanted in the jaw[28] These data emphasize the importance of the origin of MSCs used for tissue repair.

Figure 1:
figure 1

Characteristics of MSCs from adult healthy bone marrow (Modified from Yan et al.[20]) (a) Representative morphology of MSCs derived from healthy adult bone marrow. (b) Flow cytometry analysis of BM-MSC showed that BM-MSC is positive for mesenchymal markers and negative for endothelial and hematopoietic markers. (c) After specific adipogenic induction, BM-MSCs showed many lipid vacuoles that were verified by Oil Red O staining. (d) After being cultured in an osteogenic medium for 4 weeks, BM-MSCs showed early osteogenic differentiation capacity through ALP staining. Late calcium deposits in the extracellular matrix were verified by von Kossa staining. (e) BM-MSCs demonstrated chondrogenic differentiation potential as shown by toluidine blue staining, Alcian blue staining, Safranin O staining and H&E staining (cited from Fiorina et al).

Evidence for the actual and potential clinical applications of MSCs for cardiovascular diseases comes from the findings that MSCs can spontaneously differentiate into beating cardiomyocytes that respond to physiological and pharmacological stimuli.[29] Many clinical trials have been performed that primarily focused on treating heart damage.[30] The treatment was found to be safe and resulted in improved cardiac performance of the MSC-injected group and is thus very encouraging.

It was shown that MSCs could improve the healing of skin defects in humans. Recent data suggest that autologous and allogeneic MSCs appear to be equally effective for wound repair.[31,32] A shortcoming of using MSCs for wound repair is the heterogeneity of methods used for MSC culture as well as the lack of functional characterization of MSCs.

Finally, MSCs have also been proposed for the treatment of spinal cord injury,[33] stroke,[34] amyotrophic lateral sclerosis[35] and neurometabolic diseases.[36]

Mesenchymal stem cells for treating immune disorders

MSCs demonstrate a profound inhibitory effect on T, B, dendritic cells and natural killer cell proliferation in vitro and in vivo. These results suggested that MSCs could be used to cure immune disorders. Bartholomew et al.,[37] first demonstrated that the administration of MSCs could prolong skin graft survival. Currently, more than 12 preclinical animal models have been developed to demonstrate the capabilities of MSCs in modulating the immune system.[38] Le Blanc and colleagues have shown that MSCs can successfully cure severe, treatment-resistant graft-versus-host disease (GvHD).[39,40] However, the use of MSCs to prevent the rejection of allogeneic organ transplantation is still limited to animal models, and the obtained results have been conflicting.[41]

MSCs have also been proposed as a treatment for autoimmune diseases.[42] The intravenous injection of MSCs into diabetic nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice resulted in an increased number of pancreatic islets and insulin-producing ß cells.[43] Indeed, MSCs have been used for the treatment of rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and multiple sclerosis (MS).

Studies have been performed to investigate the effect of MSCs on experimental autoimmune encephalomyelitis (EAE).[44] More recently, it was shown that the intravenous infusion of MSCs also suppresses pathogenic B cell responses in vivo.[45] A decrease in neuronal loss was observed in EAE mice treated with MSCs, suggesting a protective effect on damaged tissues.[45]

MSCs can also provide support for the growth and differentiation of haemopoietic progenitor cells and can promote the engraftment of haemopoietic stem cells. It was shown that the proliferation of activated lymphocytes were suppressed by MSCs in a non-human leukocyte antigen (HLA) restricted manner.[46] A clinical trial has been in progress that investigates the use of MSCs in treating hematologic malignancy by co-transplanting MSCs and HSCs from HLA-identical siblings of the study participants.[47] It was exciting that, in 2008, the US Food and Drug Administration (FDA) gave the green light to clinical trials for the application of MSCs in MS and articular cartilage repair.

Mesenchymal stem cells for anticancer treatment

human MSCs (hMSCs) engraft and remain detectable at the injured sites and can co-localize with the sites of tumor development.[48] Thus, MSCs could be used as drug carriers for the treatment of tumors. The homing potential of hMSCs was demonstrated in vitro by hMSC coculture with glioma cells or glioma-conditioned medium that increased hMSC invasiveness.[49] In vivo, hMSCs could efficiently home to brain tumors and human glioma xenografts.[50] Unfortunately, culture-expanded hMSCs lose CXCR4, which is a key receptor responsible for hematopoietic stem cell homing.[51] However, in vitro 3-D culturing of hMSCs as spheroids increases stromal cell-derived factor 1 (SDF1) signaling, restoring the functional expression of CXCR4 that is crucial for the therapeutic application of MSCs.[51] Many cytokines and growth factors also enhance hMSC migration,[50,52] and many of those factors are secreted by tumor cells.[50] The migration of MSCs toward tumor cells is accompanied by the expression of various proteins.[52] This is similar to the in vivo myocardial injury recruitment of MSCs by the establishment of an SDF1 gradient toward the heart.[53]

It was shown that in vitro hMSC chondrogenic predifferentiation increases engraftment during in vivo cartilage repair.[54] Moreover, both the sustained delivery of growth factors to injected MSCs and the cotransplantation of MSCs with healthy tissue parts[55] can improve the MSC engraftment.[54]

Because of the ability of hMSCs to migrate to distant tumors, strategies of using them in anticancer therapies are being developed. The limited lifespan of hMSCs can be prolonged by gene modification.[56] The tumor suppressors are not downregulated in transduced MSCs, suggesting that the growth of these cells is still normal.[56] It will be very exciting to see if genetically modified MSCs can directly act as a drug vehicle that causes the complete, irreversible elimination of tumors. The treatment of human colon cancer with MSCs expressing a suicidal tyrosine kinase gene resulted in tumor elimination.[57] Therefore, gene-modified MSCs with homing potential could be used to treat tumors that cannot be cured with conventional drugs.

Mesenchymal stem cells for gene therapy

Genetic engineering of cells has become a mainstay in cell and molecular biology, and MSCs offer a unique opportunity to establish transplantation schemes to correct genetic diseases. These cells may be easily obtained, manipulated genetically, and expanded in number before reintroduction. This eliminates the limitations and risks associated with the delivery of genetic repair material directly to the patient via pathogen-associated vectors. Furthermore, there is far less concern of inappropriate differentiation than in human embryonic stem cells.

If a short-lived effect, such as bone regeneration, is the goal, transient transduction would be the desired outcome and utilized methods might include electroporation, chemical methods, plasmids, and viral constructs such as adenovirus. For the treatment of recessive diseases, permanent transduction is required, which has depended on the use of adeno-associated viruses, retroviruses, and lentiviruses.

However, some hurdles exist that must be overcome before this technology will become practical. The first hurdle is the optimization of ex vivo transfection; the second hurdle relates to the durability of the desired gene expression because, in most instances, it has been reported that expression decreases with time. The efficacy of gene therapy using MSCs will depend on the efficiency with which the reagents are incorporated into MSCs and the selection of specific targets.

Potential mechanisms of mesenchymal stem cell therapy

It was once believed that MSCs repair damaged tissues by robustly replacing the damaged cells. However, researchers have shown that in response to injury, MSCs secrete large quantities of bioactive molecules; this constitutes the most biologically significant role of MSCs under injury conditions.[58] We now know that the paracrine effect of MSCs and cell replacement by MSC-derived cells are both important. Understanding this intricate activity as well as the properties of MSCs in vivo is central to harnessing their clinical potential [Figure 2].[59]

Figure 2:
figure 2

Paracrine effects of cultured MSCs (Cited from da Silva Meirelles et al.[59]).

The secretion of bioactive molecules can be divided into six main categories: immunomodulation, anti-apoptosis, angiogenesis, support of the growth and differentiation of local stem and progenitor cells, anti-scarring and chemoattraction.[59] The number of molecules known to mediate the paracrine action of MSCs increases every day. The immunomodulatory effects of MSCs consist of the inhibition of immune cell proliferation, the suppression of immunoglobulin production, the inhibition of dentric cell maturation and the stimulation of regulatory T-cell proliferation. MSCs also limit apoptosis and stimulate local angiogenesis through the secretion of extracellular matrix molecules. A group of at least 15 chemokines produced by MSCs can elicit leukocyte migration to the injured area.[56]

Clinical-grade cell manufacture

During clinical applications of MSCs, the culture conditions must be tested and quality controls must be adapted. The release criteria of the cell batch must be strict and must take into account the effectiveness of the cellular product and the safety of the patient. Currently, the MSCs of many clinical research protocols are being generated in hematopoietic cell transplantation (HCT) processing laboratories or similar facilities. Other protocols employ current Good Manufacturing Practice (cGMP) standards, in which the cells are manufactured under the highest standards of sterility, quality control and documentation. A number of factors in the cell manufacturing process influence the nature and function of the MSCs.

Although the ISCT has established the definition of MSCs,[8,9] a major challenge in establishing release criteria is the lack of an accepted functional assay. However, given the wide range of the potential clinical effects of MSCs, any such assays will need to be specific to the particular indication or clinical trial. There may also be significant differences among MSC products from different tissue sources. Researchers have compared adipose-derived MSCs to MSCs from bone marrow,[60] and the source of MSCs may influence the ability of the cells to differentiate. Cell-manufacturing protocols must therefore take into account the variability in the characteristics of the MSCs. To mitigate the malignant transformation of human MSCs, the MSCs to be used for clinical treatment must have undergone fewer than 25–30 cell doublings.[61] The strategy to make the MSC product available quickly is to rapidly grow the MSCs in bioreactors.

Future perspective

Although perhaps several thousand patients have been treated with MSCs to date, no infusional toxicity or immediate adverse outcomes have been reported, suggesting MSC infusion to be safe. However, MSC therapies have potential risks to patients. The collection of data on patients treated with MSCs and the evaluation of long-term patient outcomes can provide an excellent infrastructure that can avoid publication bias. A registry specific for novel cellular therapies has already been established in the European Group for Blood and Marrow Transplantation (EBMT).[62]

Although many studies have demonstrated the safety and effectiveness of MSC treatment for various diseases, there are still many questions to be answered. The first is to confirm the therapeutic efficacy of transplanted MSCs. The second is to deeply understand the mechanisms of engraftment, homing and in vivo differentiation. The third is the availability of validated in vitro methods for the large-scale production of MSCs for clinical applications. The final question is the need for novel engineered devices for the tissue-specific delivery of MSCs. As these areas are addressed, the clinical significance of MSCs will be tremendously promoted.

References

  1. Goujon E. Recherches expérimentales sur les propriétés physiologiques de la moelle des os. J Anat Physiol 1869;6:399–412.

    Google Scholar 

  2. Baikow A. Ueber Transplantation von Knochenmark. Centralbl fd Med Wiss 1870;8:371–3.

    Google Scholar 

  3. 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 

  4. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9:641–50.

    Article  CAS  PubMed  Google Scholar 

  5. Owen ME, Cave J, Joyner CJ. Clonal analysis in vitro of osteogenic differentiation of marrow CFU-F. J Cell Sci 1987;87:731–8.

    PubMed  Google Scholar 

  6. Salem HK, Thiemermann C. Mesenchymal stromal cells: Current understanding and clinical status. Stem Cells 2010;28:585–96.

    CAS  PubMed  Google Scholar 

  7. Horwitz EM, Keating A. Nonhematopoietic mesenchymal stem cells: What are they? Cytotherapy 2000;2:387–8.

    Article  CAS  PubMed  Google Scholar 

  8. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, et al. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 2005;7:393–5.

    Article  CAS  PubMed  Google Scholar 

  9. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–7.

    Article  CAS  PubMed  Google Scholar 

  10. Alt E, Yan Y, Gehmert S, Song YH, Altman A, Gehmert S, et al. Fibroblasts share mesenchymal phenotypes with stem cells, but lack their differentiation and colony-forming potential. Biol Cell 2011;103:197–208.

    Article  CAS  PubMed  Google Scholar 

  11. Halfon S, Abramov N, Grinblat B, Ginis I. Markers distinguishing mesenchymal stem cells from fibroblasts are downregulated with passaging. Stem Cells Dev 2011;20:53–66.

    Article  CAS  PubMed  Google Scholar 

  12. Aggarwal S, Pittenger MF Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005;105:1815–22.

    Article  CAS  PubMed  Google Scholar 

  13. Roura S, Bagó JR, Soler-Botija C, Pujal JM, Gálvez-Montón C, Prat-Vidal C, et al. Human umbilical cord blood-derived mesenchymal stem cells promote vascular growth in vivo. PLoS One 2012;7:e49447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lee HC, An SG, Lee HW, Park JS, Cha KS, Hong TJ, et al. Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia. Circ J 2012;76:1750–60.

    Article  CAS  PubMed  Google Scholar 

  15. Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk AF, Rudakowa SF, Luriá EA, et al. Precursors for fibroblasts in different populations of hematopoietic cells as detected by the in vitro colony assay method. Exp Hematol 1974;2:83–92.

    CAS  PubMed  Google Scholar 

  16. 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 

  17. Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010;466:829–34.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Anjos-Afonso F, Bonnet D. Nonhematopoietic/endothelial SSEA−1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood 2007;109:1298–306.

    Article  CAS  PubMed  Google Scholar 

  19. Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC. SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood 2007;109:1743–51.

    Article  CAS  PubMed  Google Scholar 

  20. Yan XL, Fu CJ, Chen L, Qin JH, Zeng Q, Yuan HF, et al. Mesenchymal stem cells from primary breast cancer tissue promote cancer proliferation and enhance mammosphere formation partially via EGF/EGFR/Akt pathway. Breast Cancer Res Treat 2012;132:153–64.

    Article  CAS  PubMed  Google Scholar 

  21. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: Mesenchymal stem cells: Their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 2007;25:2739–49.

    Article  CAS  PubMed  Google Scholar 

  22. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A 2003;100:8407–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Caplan AI, Correa D. The MSC: An injury drugstore. Cell Stem Cell 2011;9:11–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Togel F, Weiss K, Yang Y, Hu Z, Zhang P, Westenfelder C. Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to the recovery from acute kidney injury. Am J Physiol Renal Physiol 2007;292:F1626–35.

    Article  CAS  PubMed  Google Scholar 

  25. Fiorina P, Jurewicz M, Augello A, Vergani A, Dada S, La Rosa S, et al. Immunomodulatory function of bone marrow-derived mesenchymal stem cells in experimental autoimmune type 1 diabetes. J Immunol 2009;183:993–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. Meijer GJ, de Bruijn JD, Koole R, van Blitterswijk CA. Cell based bone tissue engineering in jaw defects. Biomaterials 2008;29:3053–61.

    Article  CAS  PubMed  Google Scholar 

  28. Leucht P, Kim JB, Amasha R, James AW, Girod S, Helms JA. Embryonic origin and Hox status determine progenitor cell fate during adult bone regeneration. Development 2008;135:2845–54.

    Article  CAS  PubMed  Google Scholar 

  29. Planat-Bénard V, Menard C, André M, Puceat M, Perez A, Garcia-Verdugo JM, et al. Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circ Res 2004;94:223–9.

    Article  PubMed  Google Scholar 

  30. Burt RK, Loh Y, Pearce W, Beohar N, Barr WG, Craig R, et al. Clinical applications of blood-derived and marrow-derived stem cells for nonmalignant diseases. JAMA 2008;299:925–36.

    Article  CAS  PubMed  Google Scholar 

  31. Kim SS, Song CK, Shon SK, Lee KY, Kim CH, Lee MJ, et al. Effects of human amniotic membrane grafts combined with marrow mesenchymal stem cells on healing of full-thickness skin defects in rabbits. Cell Tissue Res 2009;336:59–66.

    Article  PubMed  Google Scholar 

  32. Luo G, Cheng W, He W, Wang X, Tan J, Fitzgerald M, et al. Promotion of cutaneous wound healing by local application of mesenchymal stem cells derived from human umbilical cord blood. Wound Repair Regen 2010;18:506–13.

    Article  PubMed  Google Scholar 

  33. Hofstetter CP, Schwarz EJ, Hess D, Widenfalk J, El Manira A, Prockop DJ, et al. Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proc Natl Acad Sci U S A 2002;99:2199–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chopp M, Li Y Treatment of neural injury with marrow stromal cells. Lancet Neurol 2002;1:92–100.

    Article  PubMed  Google Scholar 

  35. Mazzini L, Mareschi K, Ferrero I, Vassallo E, Oliveri G, Boccaletti R, et al. Autologous mesenchymal stem cells: Clinical applications in amyotrophic lateral sclerosis. Neurol Res 2006;28:523–6.

    Article  PubMed  Google Scholar 

  36. Müller I, Kustermann-Kuhn B, Holzwarth C, Isensee G, Vaegler M, Harzer K, et al. In vitro analysis of multipotent mesenchymal stromal cells as potential cellular therapeutics in neurometabolic diseases in pediatric patients. Exp Hematol 2006;34:1413–9.

    Article  PubMed  Google Scholar 

  37. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 2002;30:42–8.

    Article  PubMed  Google Scholar 

  38. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008;8:726–36.

    Article  CAS  PubMed  Google Scholar 

  39. Le Blanc K, Rasmusson I, Sundberg B, Götherström C, Hassan M, Uzunel M, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004;363:1439–41.

    Article  PubMed  Google Scholar 

  40. Ringdén O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, Lönnies H, et al. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation 2006;81:1390–7.

    Article  PubMed  Google Scholar 

  41. Crop M, Baan C, Weimar W, Hoogduijn M. Potential of mesenchymal stem cells as immune therapy in solid-organ transplantation. Transpl Int 2009;22:365–76.

    Article  CAS  PubMed  Google Scholar 

  42. van Laar JM, Tyndall A. Adult stem cells in the treatment of autoimmune diseases. Rheumatology 2006;45:1187–93.

    Article  PubMed  Google Scholar 

  43. Lee RH, Seo MJ, Reger RL, Spees JL, Pulin AA, Olson SD, et al. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc Natl Acad Sci U S A 2006;103:17438–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E, et al. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood 2005;106:1755–61.

    Article  CAS  PubMed  Google Scholar 

  45. Gerdoni E, Gallo B, Casazza S, Musio S, Bonanni I, Pedemonte E, et al. Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol 2007;61:219–27.

    Article  CAS  PubMed  Google Scholar 

  46. Ning H, Yang F, Jiang M, Hu L, Feng K, Zhang J, et al. The correlation between cotransplantation of mesenchymal stem cells and higher recurrence rate in hematologic malignancy patients: Outcome of a pilot clinical study. Leukemia 2008;22:593–9.

    Article  CAS  PubMed  Google Scholar 

  47. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: A phase II study. Lancet 2008;371:1579–86.

    Article  PubMed  Google Scholar 

  48. Kidd S, Spaeth E, Dembinski JL, Dietrich M, Watson K, Klopp A, et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 2009;27:2614–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Schichor C, Birnbaum T, Etminan N, Schnell O, Grau S, Miebach S, et al. Vascular endothelial growth factor A contributes to glioma-induced migration of human marrow stromal cells (hMSC). Exp Neurol 2006;199:301–10.

    Article  CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  51. Potapova IA, Brink PR, Cohen IS, Doronin SV Culturing of human mesenchymal stem cells as three-dimensional aggregates induces functional expression of CXCR4 that regulates adhesion to endothelial cells. J Biol Chem 2008;283:13100–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P. MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: Differential regulation by inflammatory cytokines. Blood 2007;109:4055–63.

    Article  CAS  PubMed  Google Scholar 

  53. Lee BC, Hsu HC, Tseng WY, Chen CY, Lin HJ, Ho YL, et al. Cell therapy generates a favourable chemokine gradient for stem cell recruitment into the infarcted heart in rabbits. Eur J Heart Fail 2009;11:238–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Moioli EK, Hong L, Guardado J, Clark PA, Mao JJ. Sustained release of TGF beta3 from PLGA microspheres and its effect on early osteogenic differentiation of human mesenchymal stem cells. Tissue Eng 2006;12:537–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Figliuzzi M, Cornolti R, Perico N, Rota C, Morigi M, Remuzzi G, et al. Bone marrow-derived mesenchymal stem cells improve islet graft function in diabetic rats. Transplant Proc 2009;41:1797–800.

    Article  CAS  PubMed  Google Scholar 

  56. Böker W, Yin Z, Drosse I, Haasters F, Rossmann O, Wierer M, et al. Introducing a single-cell-derived human mesenchymal stem cell line expressing hTERT after lentiviral gene transfer. J Cell Mol Med 2008;12:1347–59.

    Article  Google Scholar 

  57. Mathis JM, Williams BJ, Sibley DA, Carroll JL, Li J, Odaka Y et al. Cancer-specific targeting of an adenovirus-delivered herpes simplex virus thymidine kinase suicide gene using translational control. J Gene Med 2006;8:1105–20.

    Article  CAS  PubMed  Google Scholar 

  58. Prockop DJ, Olson SD. Clinical trials with adult stem/ progenitor cells for tissue repair: Let’s not overlook some essential precautions. Blood 2007;109:3147–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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  CAS  Google Scholar 

  60. Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006;24:1294–301.

    Article  CAS  PubMed  Google Scholar 

  61. Prockop DJ, Brenner M, Fibbe WE, Horwitz E, Le Blanc K, Phinney DG, et al. Defining the risks of mesenchymal stromal cell therapy. Cytotherapy 2010;12:576–8.

    Article  PubMed  Google Scholar 

  62. Le Blanc K, Fibbe W. A new cell therapy registry coordinated by the European Group for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant 2008;41:319.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xuetao Pei.

Additional information

How to cite this article: Xi J, Yan X, Zhou J, Yue W, Pei X. Mesenchymal stem cells in tissue repairing and regeneration: Progress and future. Burn Trauma 2013;1:13–20.

Source of Support: This work was supported by the National High Technology Research and Development Program of China (Grant No: 2011AA020109). Conflict of Interest: None declared.

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 use, duplication, adaptation, distribution, and reproduction in any medium or format, as long as 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

Reprints and permissions

About this article

Cite this article

Xi, J., Yan, X., Zhou, J. et al. Mesenchymal stem cells in tissue repairing and regeneration: Progress and future. Burn Trauma 1, 13–20 (2013). https://doi.org/10.4103/2321-3868.113330

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.4103/2321-3868.113330

Key words