Abstract
Multipotent mesenchymal stem/stromal cells (MSCs) have been extensively used as a transplantable cell source for regenerative medicine and immunomodulatory therapy. Specifically in allogeneic hematopoietic stem cell transplantation (HSCT), co-transplantation or post-transplant infusion of MSCs derived from bone marrow (BM) of non-self donors has been implicated in accelerating hematopoietic recovery, ameliorating graft-vs.-host disease, and promoting tissue regeneration. However, irrespective of the use of MSC co-administration, post-transplant chimerism of BM-derived MSCs after allogeneic HSCT has been reported to remain of host origin, suggesting that the infused donor MSCs are immunologically rejected or not capable of long-term engraftment in the host microenvironment. Also, hematopoietic cell allografts currently used for HSCT do not seem to contain sufficient amount of MSCs or their precursors to reconstitute host BM microenvironment. Since the toxic conditioning employed in allo-HSCT may impair the function of host MSCs to maintain hematopoietic/regenerative stem cell niches and to provide a local immunomodulatory milieu, we propose that new directions for enhancing immunohematopoietic reconstitution and tissue repair after allogeneic HSCT include the development of strategies to support functional replenishment of residual host MSCs or to support more efficient engraftment of infused donor MSCs. Future areas of research should include in vivo tracking of infused MSCs and detection of their microchimeric presence in extra-marrow sites as well as in BM.
Keywords: mesenchymal stem/stromal cells, hematopoietic cell transplantation, stem cell niches, hematopoietic reconstitution, graft-versus-host disease
Introduction
Multipotent mesenchymal stem/stromal cells (MSCs) are originally isolated from human bone marrow (BM) as adherent, fibroblast-like shaped cells with an ability to differentiate into the mesenchymal lineage cells of mesoderm such as osteoblasts, adipocytes, and chondrocytes.1,2 In addition to such multi-differentiation capacity, MSCs are shown to have protean immunomodulatory properties to ameliorate immune dysfunctions caused by pathologic effector cells. In the last decade, substantial efforts have been made to develop regenerative or anti-inflammatory cellular therapy using culture-expanded autologous or allogeneic MSCs.3,4 Specifically in the field of allogeneic hematopoietic stem cell transplantation (HSCT), various clinical trials have been performed with the aim of accelerating engraftment or ameliorating graft-vs.-host disease (GVHD) by infusion of MSCs obtained from the hematopoietic cell donors or third-party donors. However, in the allogeneic setting, most of infused MSCs are considered short-lived in the host and the mechanisms by which MSC-based therapy can work in clinics remain mostly unclarified. In this short review, we critically look back on the biological basis of cellular therapy using culture-expanded allogeneic MSCs in the context of allogeneic HSCT. We also propose that a new area of translational research should include functional replenishment of host-type MSCs by pharmacologic agents or chimerism enhancement of donor-type MSCs by their in situ infusions.
Biological Characteristics of Human Mesenchymal Stem/Stromal Cells (MSCs)
In a current understanding of the biology of human MSCs, they are characterized by the following in vitro features3,5: (1) their ability to adhere to plastic plate and to form colony forming unit-fibroblastic (CFU-F); (2) their ability to differentiate into osteoblasts, adipocytes and chondrocytes; (3) their positive surface expression of CD105 (endoglin), CD73, and CD90 (Thy-1) in the absence of pan-leukocyte (CD45), endothelial/primitive hematopoietic (CD34), and hematopoietic lineage markers as well as in the absence of surface human leukocyte antigen (HLA)-class II molecules. MSCs or MSC-like cells appear to be widely distributed in the human body, because the cells with similar biological characteristics to BM-derived MSCs have been demonstrated to be isolated from a variety of adult organs or tissues including adipose tissues, cartilages, fetal liver, and fetal lungs, although their mutual identities remain elusive.3
Whether umbilical cord blood (CB) and peripheral blood (PB) contain MSCs or not has been a great interest since, as well as BM, CB and cytokine-mobilized PB have been successfully used as stem cell sources for allogeneic HSCT. Recent studies have clearly demonstrated that CB contains MSCs, but its frequency is estimated to be extremely low around one in 1 × 108 mononuclear cells compared with that in BM around one in 1 × 106 to 1 × 104 mononuclear cells. However, CB-derived MSCs show tremendous proliferation to generate much more progeny than BM-derived MSCs and can be expanded to the order of 108 cells, which are deemed sufficient for clinical studies.6,7 CB-derived MSCs differ from BM-derived MSCs in their variable expression levels of CD146, a marker for multipotency and differentiation potential: CB-derived MSCs show robust potential for chondrogenic differentiation, while their potential for adipogenic differentiation is not obvious. Therefore, the use of an appropriate origin or unit of MSCs is important for achieving satisfactory effects according to the planned clinical application.
On the other hand, the presence of MSCs in steady-state PB is a matter of controversy, although several investigators have reported that MSCs or MSC-like cells can be isolated in postnatal PB or in PB samples obtained from individuals receiving granulocyte-colony stimulating factor,8,9 patients with breast cancer,10 and patients with osteoporosis.11 These findings might reflect one of the potential features of MSCs that they might be occasionally supplied into the blood circulation from the MSC niche in response to systemic or local hormones, cytokines, and growth factors to migrate toward organs or tissues in need.12,13 However, further studies are necessary to confirm if MSC-like multipotent cells can be reproducibly obtained from the steady-state circulating blood.14
Immunomodulatory Properties of MSCs and Treatment of Immunologic Diseases by Culture-Expanded MSCs
Lines of evidence demonstrate that MSCs have immunomodulatory properties with anti-inflammatory, anti-proliferative and immunosuppressive capacities. The possible mechanisms underlying such immunomodulatory effects of MSCs are thought to be mediated by a combination of soluble molecules in the microenvironment where MSCs can directly interact with immune cells. MSCs are initially demonstrated to exhibit anti-proliferative effects on T cells,15 but these effects are observed on other immune cells including B cells, NK cells, and dendritic cells.16,17 Mature mesenchymal cells such as chondrocytes can also show anti-proliferative effects on immune cells.18 However, they do not exert the immunomodulatory effects mediated by MSCs,19 implying the unique property of MSCs in the immune system. Effective immunosuppression by MSCs is supposed to be achieved in local microenvironment where MSCs reside. In mouse experiments, MSCs become activated by proinflammatory cytokines interferon-γ, tumor necrosis factor-α, interleukin (IL)-1-α or IL-1-β and express T-cell-specific chemokines CXCL-9 and CXCL-10 to attract T cells into close proximity with MSCs.20,21 Conversely, MSCs express chemokine receptors that are involved in migration of MSCs to the sites where they participate in immune responses.22-24 In such a milieu, MSCs exert their inhibitory effects on T cell proliferation through the secretion of soluble factors such as nitric oxide,25 indoleamine 2,3-dioxygenase,26-28 heme oxygenase-1,29 prostaglandin E2,30,31 transforming growth factor-β1,22 and leukemia inhibitory factor.32 Some of these soluble molecules are also demonstrated to be involved in the suppression of proliferation and cytotoxicity of NK cells or maturation and function of dendritic cells.28,33-35 However, MSC-mediated immunosuppression is thought to be a “combined” effect that cannot be fully explained by any one of those molecules released from MSCs. Furthermore, direct cell-to-cell interactions might also be associated with the MSC-mediated immunosuppression. Although activating NK cells are capable of killing MSCs with low levels of HLA class I molecules expressed on their surface, upon simulation of MSCs with interferon-γ, the expression of HLA class I molecules is upregulated,36 resulting in inhibition of NK cell-mediated lysis of MSCs.37
Given such protean immunomodulatory properties, infusions of allogeneic MSCs have been suggested to be beneficial for the treatment of diseases in which the immune system is dysregulated such as multiple sclerosis, type 1 diabetes mellitus, Crohn’s disease, systemic lupus erythematosus, and rheumatoid arthritis/Sjögren syndrome.3 Le Blanc et al. was the first to report successful treatment of steroid-resistant severe acute GVHD of the gut and liver by intravenous infusion of culture-expanded BM-derived MSCs from a haploidentical mother in a boy who had previously received unrelated HLA-matched HSCT for his acute lymphoblastic leukemia.38 In this report, they demonstrated the partial female donor epithelium chimerism (4%) in the biopsy specimens of colon by X and Y chromosomes fluorescence in situ hybridization analysis, which might imply the contribution of donor MSCs to the improvement of gut GVHD. A large study from the same group showed that infusion of MSCs from multiple donor sources conferred overall response rate of 69% including complete response for the treatment of steroid-resistant acute GVHD.39 These studies support the experimental and preclinical observations that the infused MSCs can directly or indirectly exert immunomodulatory effects possibly through the production of soluble factors and/or direct cell-to-cell interactions. However, intriguingly, many of these immunomodulatory effects of infused MSCs can be exerted without their long-term persistence in the host.40,41
Effects of Ex Vivo Expanded MSCs on Hematopoietic reconstitution after Hematopoietic Cell Transplantation
Clinical application of MSCs for tissue/organ regeneration has also been extensively considered based on their capability of secreting trophic factors, homing to sites of damage, and multi-differentiation. One of the important roles of BM-derived MSCs is to provide the microenvironment of hematopoietic stem cell (HSC) niche that supports durable and effective hematopoiesis. In animal models, nestin-positive cells are shown to be MSCs that comprise the HSC niche in BM.42 Osteoblasts, CD146-expressing sub-endothelial stromal cells, and adipo-osteogenic progenitors expressing CXCL12 also are demonstrated to be cellular constituents of HSC niche;43,44 these cells are derived from MSCs or have similar characteristics to MSCs. Conversely, aberrant MSC-derived osteoprogenitor cells have been demonstrated be associated with disease pathophysiology in animal models.45-48 Given the results of studies demonstrating the importance of MSCs or MSC-derived stromal cells for the maintenance of hematopoiesis, the Lazarus group intravenously administered autologous culture-expanded MSCs in patients with hematologic malignancies without any infusion-related adverse reactions.49 Their group also reported the ability of MSCs to enhance hematopoietic recovery by co-infusion of autologous PB progenitor cells and culture-expanded MSCs in patients with breast cancer receiving high-dose chemotherapy.50
Upon allogenic HSCT especially when using HLA-mismatched grafts, primary graft failure is one of the early and very serious complications. Co-administration of MSCs with HSCs could be a promising strategy to overcome this shortcoming. In myeloablative allogeneic HSCT using BM or PB progenitor cells from HLA-identical siblings, co-transplantation of culture-expanded BM-derived MSCs was safe without immediate or late toxicities but showed no apparent acceleration of engraftment.51 Importantly, at 6 and 18 mo after transplantation, microsatellite-based chimerism analysis of MSCs in BM aspirates revealed that loss or very rare presence of infused donor-type MSCs.51 In a situation of graft failure or graft rejection after the first allogenic HSCT, a pilot study showed the utility of co-infusion of MSCs in the subsequent salvage HSCT to improve engraftment of the second graft.52 In haploidentical allogenic HSCT using cytokine-mobilized CD34 positive progenitor cells, co-transplantation of haploidentical MSCs did not confer acceleration of either neutrophil or platelet engraftment but prevented graft rejection.53 In this study, donor-type chimerism of MSCs evaluated by PCR analysis using donor/recipient polymorphisms was only transiently detected at very low levels in 3 out of 14 patients at 3 mo after transplantation. In unrelated HLA-mismatched CB transplantation for children with high risk hematological malignancies, co-transplantation of haploidentical parental MSCs conferred relatively rapid hematopoietic recovery.54 More recently, de Lima et al. reported that ex vivo coculture of CB cells with MSCs derived from BM of haploidentical family members or third-party donors is feasible and effectively expanded CD34+ cells to accelerate hematopoietic recovery when infused with the second unexpanded CB unit.55 Although these pilot studies suggest the utility of MSCs on hematopoietic recovery after allogeneic HSCT, further studies are needed to elucidate the appropriate dose of MSCs and timing of their infusions for achieving optimal outcomes.
Chimerism of Donor-Derived MSCs after Allogeneic Hematopoietic Cell Transplantation
Although there is experimental evidence suggesting the presence of a common mesoderm cell as origin of both hematopoietic and mesenchymal progenitor cells in an animal model,56 it is still controversial if durable engraftment of native donor-derived MSCs without ex vivo treatment can occur in the recipient of allogeneic HSCT.57-63 In this context, clinical studies of culture-expanded high-dose MSCs transplantation for children with severe osteogenesis imperfecta (OI) are very informative.64,65 Horwitz et al. reported the engraftment of donor-type MSCs in children who underwent allogeneic marrow transplantation and subsequent infusion of culture-expanded booster MSCs as treatment for severe OI; the infused MSCs were derived from the BM of the original donors and gene-marked for tracking.66 Despite high doses of infused MSCs, the fraction of donor cells at any samples was less than 1% when PCR-based chimerism analyses of osteoblasts, fibroblasts, and marrow stromal cells of these children were performed between 18 and 34 mo after the MSC infusions.
Similarly to gene-marking, recent development of noninvasive imaging techniques has enabled us to have a better insight into in vivo cell tracking of transplanted MSCs in a real-time manner.66 These experimental imagings have revealed that intravenously transplanted MSCs are mainly distributed in the lungs and liver, while their distribution to the other tissues such as BM is barely detectable.67 Also, in conventional BM transplantation where MSCs are being transferred in their native form without an ex vivo expansion phase, it is believed that only a small proportion of MSCs can reach to the marrow because most of them are trapped in the microvasculatures of lungs, particularly when MSCs are intravenously administered. By contrast, in a rat model of myocardial infarction, Barbash et al. reported that direct left ventricular infusion of MSCs could enhance their migration and colonization to the area of ischemic myocardium as compared with systemic intravenous infusion.68 Similarly, there is accumulating evidence that intra-BM transplantation of hematopoietic cell graft relatively rich in MSCs may be a promising procedure to facilitate the sustained engraftment of donor MSCs in a series of experiments using large animal models.69 To get a better understanding of the fate of donor-derived MSCs after allogeneic HSCT or other MSC-based therapy, development of reliable imaging methods to track infused MSCs is highly required. Furthermore, it is warranted to develop a technique for detecting microchimeric presence of infused MSCs in extra-marrow sites as well as in BM.
Future Prospects
One caveat in allogeneic HSCT using current protocols might be very low chimerism of donor-type MSCs at least in the hematopoietic microenvironment. Although it is well known that recipients of allogeneic HSCT successfully recover normal hematopoietic function despite lack of engraftment of donor-derived MSCs in the majority of cases, host-type MSCs that survived after sublethal conditioning for HSCT can harbor genetic abnormalities that may compromise crucial functions of MSCs to support effective hematopoiesis and tissue regeneration.
Although little information exists regarding the function of MSCs residing in the recipient of allogeneic HSCT, BM-derived MSCs from a long-term survivor after myeloablative transplant can have complex chromosome abnormalities (TI, unpublished observation). Since various cytokines including erythropoietin and parathyroid hormone are reported to have activities to enhance MSC-mediated bone formation, hematopoiesis, and tissue regeneration,70-72 we propose that future direction of MSC-based therapy should include pharmacologic upregulation of their functions. Another promising approach will be to improve the engraftment of donor-type MSCs by their in situ administration into the BM cavity of the recipient. Attempts to reconstitute abnormal microenvironment with allogeneic normal MSCs and to achieve appropriate donor MSC chimerism could be a novel therapeutic approach for the treatment of intractable hematologic and MSC-associated disorders.
Acknowledgments
This work was supported in part by grants from the ministry of Education, Culture, Sports, Science, and Technology of Japan (#22591061 to TI) and grants from the ministry of Health, Labour and Welfare of Japan (TI).
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Footnotes
Previously published online: www.landesbioscience.com/journals/chimerism/article/25609
References
- 1.Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation. 1968;6:230–47. doi: 10.1097/00007890-196803000-00009. [DOI] [PubMed] [Google Scholar]
- 2.Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641–50. doi: 10.1002/jor.1100090504. [DOI] [PubMed] [Google Scholar]
- 3.Frenette PS, Pinho S, Lucas D, Scheiermann C. Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu Rev Immunol. 2013;31:285–316. doi: 10.1146/annurev-immunol-032712-095919. [DOI] [PubMed] [Google Scholar]
- 4.Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–36. doi: 10.1038/nri2395. [DOI] [PubMed] [Google Scholar]
- 5.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. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
- 6.Bieback K, Kern S, Klüter H, Eichler H. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells. 2004;22:625–34. doi: 10.1634/stemcells.22-4-625. [DOI] [PubMed] [Google Scholar]
- 7.Zhang X, Hirai M, Cantero S, Ciubotariu R, Dobrila L, Hirsh A, et al. Isolation and characterization of mesenchymal stem cells from human umbilical cord blood: reevaluation of critical factors for successful isolation and high ability to proliferate and differentiate to chondrocytes as compared to mesenchymal stem cells from bone marrow and adipose tissue. J Cell Biochem. 2011;112:1206–18. doi: 10.1002/jcb.23042. [DOI] [PubMed] [Google Scholar]
- 8.Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R, Massy M, et al. Mesenchymal stem cells derived from CD133-positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity. Stem Cells. 2005;23:1105–12. doi: 10.1634/stemcells.2004-0330. [DOI] [PubMed] [Google Scholar]
- 9.Cesselli D, Beltrami AP, Rigo S, Bergamin N, D’Aurizio F, Verardo R, et al. Multipotent progenitor cells are present in human peripheral blood. Circ Res. 2009;104:1225–34. doi: 10.1161/CIRCRESAHA.109.195859. [DOI] [PubMed] [Google Scholar]
- 10.Fernández M, Simon V, Herrera G, Cao C, Del Favero H, Minguell JJ. Detection of stromal cells in peripheral blood progenitor cell collections from breast cancer patients. Bone Marrow Transplant. 1997;20:265–71. doi: 10.1038/sj.bmt.1700890. [DOI] [PubMed] [Google Scholar]
- 11.Dalle Carbonare L, Valenti MT, Zanatta M, Donatelli L, Lo Cascio V. Circulating mesenchymal stem cells with abnormal osteogenic differentiation in patients with osteoporosis. Arthritis Rheum. 2009;60:3356–65. doi: 10.1002/art.24884. [DOI] [PubMed] [Google Scholar]
- 12.Huang GT, Gronthos S, Shi S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res. 2009;88:792–806. doi: 10.1177/0022034509340867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res. 2003;18:696–704. doi: 10.1359/jbmr.2003.18.4.696. [DOI] [PubMed] [Google Scholar]
- 14.Eghbali-Fatourechi GZ, Lamsam J, Fraser D, Nagel D, Riggs BL, Khosla S. Circulating osteoblast-lineage cells in humans. N Engl J Med. 2005;352:1959–66. doi: 10.1056/NEJMoa044264. [DOI] [PubMed] [Google Scholar]
- 15.Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838–43. doi: 10.1182/blood.V99.10.3838. [DOI] [PubMed] [Google Scholar]
- 16.Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107:367–72. doi: 10.1182/blood-2005-07-2657. [DOI] [PubMed] [Google Scholar]
- 17.Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22. doi: 10.1182/blood-2004-04-1559. [DOI] [PubMed] [Google Scholar]
- 18.Jones S, Horwood N, Cope A, Dazzi F. The antiproliferative effect of mesenchymal stem cells is a fundamental property shared by all stromal cells. J Immunol. 2007;179:2824–31. doi: 10.4049/jimmunol.179.5.2824. [DOI] [PubMed] [Google Scholar]
- 19.Németh K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15:42–9. doi: 10.1038/nm.1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Groh ME, Maitra B, Szekely E, Koç ON. Human mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells. Exp Hematol. 2005;33:928–34. doi: 10.1016/j.exphem.2005.05.002. [DOI] [PubMed] [Google Scholar]
- 21.Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–50. doi: 10.1016/j.stem.2007.11.014. [DOI] [PubMed] [Google Scholar]
- 22.Kortesidis A, Zannettino A, Isenmann S, Shi S, Lapidot T, Gronthos S. Stromal-derived factor-1 promotes the growth, survival, and development of human bone marrow stromal stem cells. Blood. 2005;105:3793–801. doi: 10.1182/blood-2004-11-4349. [DOI] [PubMed] [Google Scholar]
- 23.Ji JF, He BP, Dheen ST, Tay SS. Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem Cells. 2004;22:415–27. doi: 10.1634/stemcells.22-3-415. [DOI] [PubMed] [Google Scholar]
- 24.Wynn RF, Hart CA, Corradi-Perini C, O’Neill L, Evans CA, Wraith JE, et al. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone marrow. Blood. 2004;104:2643–5. doi: 10.1182/blood-2004-02-0526. [DOI] [PubMed] [Google Scholar]
- 25.Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, Nagai T, et al. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 2007;109:228–34. doi: 10.1182/blood-2006-02-002246. [DOI] [PubMed] [Google Scholar]
- 26.Meisel R, Zibert A, Laryea M, Göbel U, Däubener W, Dilloo D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004;103:4619–21. doi: 10.1182/blood-2003-11-3909. [DOI] [PubMed] [Google Scholar]
- 27.Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells. 2006;24:386–98. doi: 10.1634/stemcells.2005-0008. [DOI] [PubMed] [Google Scholar]
- 28.Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008;111:1327–33. doi: 10.1182/blood-2007-02-074997. [DOI] [PubMed] [Google Scholar]
- 29.Chabannes D, Hill M, Merieau E, Rossignol J, Brion R, Soulillou JP, et al. A role for heme oxygenase-1 in the immunosuppressive effect of adult rat and human mesenchymal stem cells. Blood. 2007;110:3691–4. doi: 10.1182/blood-2007-02-075481. [DOI] [PubMed] [Google Scholar]
- 30.Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22. doi: 10.1182/blood-2004-04-1559. [DOI] [PubMed] [Google Scholar]
- 31.Jarvinen L, Badri L, Wettlaufer S, Ohtsuka T, Standiford TJ, Toews GB, et al. Lung resident mesenchymal stem cells isolated from human lung allografts inhibit T cell proliferation via a soluble mediator. J Immunol. 2008;181:4389–96. doi: 10.4049/jimmunol.181.6.4389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Nasef A, Mazurier C, Bouchet S, François S, Chapel A, Thierry D, et al. Leukemia inhibitory factor: Role in human mesenchymal stem cells mediated immunosuppression. Cell Immunol. 2008;253:16–22. doi: 10.1016/j.cellimm.2008.06.002. [DOI] [PubMed] [Google Scholar]
- 33.Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Interactions between human mesenchymal stem cells and natural killer cells Stem Cells. 2006;24:74–85. doi: 10.1634/stemcells.2004-0359. [DOI] [PubMed] [Google Scholar]
- 34.Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol. 2006;177:2080–7. doi: 10.4049/jimmunol.177.4.2080. [DOI] [PubMed] [Google Scholar]
- 35.Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L. MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: central role of MSC-derived prostaglandin E2. Blood. 2009;113:6576–83. doi: 10.1182/blood-2009-02-203943. [DOI] [PubMed] [Google Scholar]
- 36.Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringdén O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31:890–6. doi: 10.1016/S0301-472X(03)00110-3. [DOI] [PubMed] [Google Scholar]
- 37.Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood. 2006;107:1484–90. doi: 10.1182/blood-2005-07-2775. [DOI] [PubMed] [Google Scholar]
- 38.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. doi: 10.1016/S0140-6736(04)16104-7. [DOI] [PubMed] [Google Scholar]
- 39.Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, et al. Developmental Committee of the European Group for Blood and Marrow Transplantation Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579–86. doi: 10.1016/S0140-6736(08)60690-X. [DOI] [PubMed] [Google Scholar]
- 40.Prockop DJ. Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther. 2009;17:939–46. doi: 10.1038/mt.2009.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Prockop DJ, Kota DJ, Bazhanov N, Reger RL. Evolving paradigms for repair of tissues by adult stem/progenitor cells (MSCs) J Cell Mol Med. 2010;14:2190–9. doi: 10.1111/j.1582-4934.2010.01151.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Méndez-Ferrer S, Michiurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA,, et al. Mesenchymal and hematopoietic stem cells from a unique bone marrow niche. Nature. 2012;466:829–34. doi: 10.1038/nature09262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.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. doi: 10.1016/j.cell.2007.08.025. [DOI] [PubMed] [Google Scholar]
- 44.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. doi: 10.1016/j.immuni.2010.08.017. [DOI] [PubMed] [Google Scholar]
- 45.Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010;464:852–7. doi: 10.1038/nature08851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Walkley CR, Olsen GH, Dworkin S, Fabb SA, Swann J, McArthur GA, et al. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell. 2007;129:1097–110. doi: 10.1016/j.cell.2007.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Walkley CR, Shea JM, Sims NA, Purton LE, Orkin SH. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell. 2007;129:1081–95. doi: 10.1016/j.cell.2007.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Miura M, Chen XD, Allen MR, Bi Y, Gronthos S, Seo BM, et al. A crucial role of caspase-3 in osteogenic differentiation of bone marrow stromal stem cells. J Clin Invest. 2004;114:1704–13. doi: 10.1172/JCI20427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.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. [PubMed] [Google Scholar]
- 50.Koç ON, Gerson SL, Cooper BW, Dyhouse SM, Haynesworth SE, Caplan AI, et al. Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J Clin Oncol. 2000;18:307–16. doi: 10.1200/JCO.2000.18.2.307. [DOI] [PubMed] [Google Scholar]
- 51.Lazarus HM, Koc ON, Devine SM, Curtin P, Maziarz RT, Holland HK, et al. Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol Blood Marrow Transplant. 2005;11:389–98. doi: 10.1016/j.bbmt.2005.02.001. [DOI] [PubMed] [Google Scholar]
- 52.Le Blanc K, Samuelsson H, Gustafsson B, Remberger M, Sundberg B, Arvidson J, et al. Transplantation of mesenchymal stem cells to enhance engraftment of hematopoietic stem cells. Leukemia. 2007;21:1733–8. doi: 10.1038/sj.leu.2404777. [DOI] [PubMed] [Google Scholar]
- 53.Ball LM, Bernardo ME, Roelofs H, Lankester A, Cometa A, Egeler RM, et al. Cotransplantation of ex vivo expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood. 2007;110:2764–7. doi: 10.1182/blood-2007-04-087056. [DOI] [PubMed] [Google Scholar]
- 54.Macmillan ML, Blazar BR, DeFor TE, Wagner JE. Transplantation of ex-vivo culture-expanded parental haploidentical mesenchymal stem cells to promote engraftment in pediatric recipients of unrelated donor umbilical cord blood: results of a phase I-II clinical trial. Bone Marrow Transplant. 2009;43:447–54. doi: 10.1038/bmt.2008.348. [DOI] [PubMed] [Google Scholar]
- 55.de Lima M, McNiece I, Robinson SN, Munsell M, Eapen M, Horowitz M, et al. Cord-blood engraftment with ex vivo mesenchymal-cell coculture. N Engl J Med. 2012;367:2305–15. doi: 10.1056/NEJMoa1207285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lange C, Kaltz C, Thalmeier K, Kolb HJ, Huss R. Hematopoietic reconstitution of syngeneic mice with a peripheral blood-derived, monoclonal CD34-, Sca-1+, Thy-1(low), c-kit+ stem cell line. J Hematother Stem Cell Res. 1999;8:335–42. doi: 10.1089/152581699320090. [DOI] [PubMed] [Google Scholar]
- 57.Bartsch K, Al-Ali H, Reinhardt A, Franke C, Hudecek M, Kamprad M, et al. Mesenchymal stem cells remain host-derived independent of the source of the stem-cell graft and conditioning regimen used. Transplantation. 2009;87:217–21. doi: 10.1097/TP.0b013e3181938998. [DOI] [PubMed] [Google Scholar]
- 58.García-Castro J, Balas A, Ramírez M, Pérez-Martínez A, Madero L, González-Vicent M, et al. Mesenchymal stem cells are of recipient origin in pediatric transplantations using umbilical cord blood, peripheral blood, or bone marrow. J Pediatr Hematol Oncol. 2007;29:388–92. doi: 10.1097/MPH.0b013e3180645186. [DOI] [PubMed] [Google Scholar]
- 59.Pozzi S, Lisini D, Podestà M, Bernardo ME, Sessarego N, Piaggio G, et al. Donor multipotent mesenchymal stromal cells may engraft in pediatric patients given either cord blood or bone marrow transplantation. Exp Hematol. 2006;34:934–42. doi: 10.1016/j.exphem.2006.03.007. [DOI] [PubMed] [Google Scholar]
- 60.Poloni A, Leoni P, Buscemi L, Balducci F, Pasquini R, Masia MC, et al. Engraftment capacity of mesenchymal cells following hematopoietic stem cell transplantation in patients receiving reduced-intensity conditioning regimen. Leukemia. 2006;20:329–35. doi: 10.1038/sj.leu.2404018. [DOI] [PubMed] [Google Scholar]
- 61.Dickhut A, Schwerdtfeger R, Kuklick L, Ritter M, Thiede C, Neubauer A, et al. Mesenchymal stem cells obtained after bone marrow transplantation or peripheral blood stem cell transplantation originate from host tissue. Ann Hematol. 2005;84:722–7. doi: 10.1007/s00277-005-1067-8. [DOI] [PubMed] [Google Scholar]
- 62.Rieger K, Marinets O, Fietz T, Körper S, Sommer D, Mücke C, et al. Mesenchymal stem cells remain of host origin even a long time after allogeneic peripheral blood stem cell or bone marrow transplantation. Exp Hematol. 2005;33:605–11. doi: 10.1016/j.exphem.2005.02.004. [DOI] [PubMed] [Google Scholar]
- 63.Villaron EM, Almeida J, López-Holgado N, Alcoceba M, Sánchez-Abarca LI, Sanchez-Guijo FM, et al. Mesenchymal stem cells are present in peripheral blood and can engraft after allogeneic hematopoietic stem cell transplantation. Haematologica. 2004;89:1421–7. [PubMed] [Google Scholar]
- 64.Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med. 1999;5:309–13. doi: 10.1038/6529. [DOI] [PubMed] [Google Scholar]
- 65.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 U S A. 2002;99:8932–7. doi: 10.1073/pnas.132252399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Li SC, Tachiki LM, Luo J, Dethlefs BA, Chen Z, Loudon WG. A biological global positioning system: considerations for tracking stem cell behaviors in the whole body. Stem Cell Rev. 2010;6:317–33. doi: 10.1007/s12015-010-9130-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Allers C, Sierralta WD, Neubauer S, Rivera F, Minguell JJ, Conget PA. Dynamic of distribution of human bone marrow-derived mesenchymal stem cells after transplantation into adult unconditioned mice. Transplantation. 2004;78:503–8. doi: 10.1097/01.TP.0000128334.93343.B3. [DOI] [PubMed] [Google Scholar]
- 68.Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: feasibility, cell migration, and body distribution. Circulation. 2003;108:863–8. doi: 10.1161/01.CIR.0000084828.50310.6A. [DOI] [PubMed] [Google Scholar]
- 69.Ikehara S. A novel BMT technique for treatment of various currently intractable diseases. Best Pract Res Clin Haematol. 2011;24:477–83. doi: 10.1016/j.beha.2011.04.003. [DOI] [PubMed] [Google Scholar]
- 70.Yamaza T, Miura Y, Bi Y, Liu Y, Akiyama K, Sonoyama W, et al. Pharmacologic stem cell based intervention as a new approach to osteoporosis treatment in rodents. PLoS One. 2008;3:e2615. doi: 10.1371/journal.pone.0002615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yamaza T, Miura Y, Akiyama K, Bi Y, Sonoyama W, Gronthos S, et al. Mesenchymal stem cell-mediated ectopic hematopoiesis alleviates aging-related phenotype in immunocompromised mice. Blood. 2009;113:2595–604. doi: 10.1182/blood-2008-10-182246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Shi S, Gronthos S, Chen S, Reddi A, Counter CM, Robey PG, et al. Bone formation by human postnatal bone marrow stromal stem cells is enhanced by telomerase expression. Nat Biotechnol. 2002;20:587–91. doi: 10.1038/nbt0602-587. [DOI] [PubMed] [Google Scholar]