Mesenchymal stem cells (MSCs) are multipotential nonhematopoietic progenitor cells capable of differentiating into multiple lineages of the mesenchyme. MSCs have emerged as a promising therapeutic modality for tissue regeneration and repair. Further clinical interest has been raised by the observation that MSCs are immunoprivileged and, more importantly, display immunomodulatory capacities. Although the mechanisms underlying the immunosuppressive effects of MSCs have not been clearly defined, their immunosuppressive properties have already been exploited in the clinical setting. The aim of this review is to critically discuss the immunogenicity and immunomodulatory properties of MSCs, both in vitro and in vivo, the possible underlying mechanisms, the potential clinical use of MSCs as modulators of immune responses in vivo, and to indicate clinical safety concerns and recommendations for future research.

Bone marrow (BM) stromal cells were first identified by Friedenstein, who described an adherent fibroblast-like population able to differentiate into bone that he referred to as osteogenic precursor cells.1  Subsequent studies demonstrated that these cells have the ability to differentiate into various other mesodermal cell lineages, including chondrocytes, tenocytes, and myoblasts (reviewed in Prockop2 ). Based on this multilineage differentiation capacity, Caplan introduced the term mesenchymal stem cells (MSCs),3  although many other terms have been introduced to describe a nonhomogenous population of multipotent cells. Although MSCs at a population level fulfill stem-cell criteria (ie, self renewal and multilineage differentiation capacity), it remains questionable whether the qualification “stem cell” is legitimate for MSCs at the single cell level. It was therefore recently proposed to use the term multipotent mesenchymal stromal cells (with the acronym MSCs) to describe fibroblast-like plastic-adherent cells.4  Recently, Bonnet et al demonstrated that single cell–derived populations of murine BM-derived MSCs characterized by stage-specific embryonic antigen-1 expression, were capable of differentiation in vivo,5  thus showing their true stem-cell properties. In this review, we will refer to the multipotent mesenchymal stromal cells with the acronym MSCs.

Although MSCs originally were isolated from BM,6  similar populations have been isolated from other tissues, including adipose tissue,7  placenta,8  amniotic fluid,9  and fetal tissues such as fetal lung and blood.10  In addition, umbilical cord blood (UCB) has been identified as a source of MSCs.11,12  Probably as a result of their low frequency in UCB, conflicting reports initially have been published on the presence of MSCs in UCB. It has now become clear that the volume and storage time of the cord blood are important parameters for successful isolation of MSCs from UCB.11 

At present no specific marker or combination of markers has been identified that specifically defines MSCs. Phenotypically, ex vivo expanded MSCs express a number of nonspecific markers, including CD105 (SH2 or endoglin), CD73 (SH3 or SH4), CD90, CD166, CD44, and CD29.6,13  MSCs are devoid of hematopoietic and endothelial markers, such as CD11b, CD14, CD31, and CD45.6 

The capacity to differentiate into multiple mesenchymal lineages, including bone, fat, and cartilage, is being used as a functional criterion to define MSCs.2  Recent studies indicated the identification of pluripotent cells that not only differentiate into cells of the mesoderm lineage, but also into endoderm and neuroectoderm lineages, including neurons,14  hepatocytes,15  and endothelia.16  Such pluripotent stem cells have been identified in BM and referred to as multipotent adult progenitor cells (MAPCs),17  human BM-derived multipotent stem cells (hBMSCs),18  marrow-isolated adult multilineage inducible (MIAMIs) cells,19  or very small embryonic-like stem cells (VSELs).20  Similar pluripotent cell types have been found in UCB (unrestricted somatic stem cells; USSCs),12  in adipose tissue,21  and recently in amniotic fluid.22  These primitive cell types require specific and stringent culture conditions, including embryonic stem cell–specific fetal calf serum (FCS), coated culture dishes (a.o. fibronectin), medium with specific growth factor requirements, specific type or culture dish, and prolonged culture duration at low cell density. Culturing these cells at higher cell density promotes differentiation toward a mesenchymal progenitor cell with restricted differentiation potential.23  It has not been possible to prospectively isolate these cells from freshly obtained tissues, blood, or BM. Therefore, it is still unclear to what extent they are primary cells that play a physiological role or are the result of prolonged culture expansion under specific culture conditions.

The identification of MSCs in vivo and the prospective isolation of MSCs from primary tissues is hampered by the availability of a specific MSC marker. To date, MSC isolation still relies on their adherence to plastic, resulting in a heterogeneous population of adherent cells, popularly defined as MSCs. Currently, direct proof that these multipotent stem cells play a physiological role in vivo is lacking. Specific markers to prospectively isolate these cells could help to further resolve this issue.

Several cell surface antigens have been reported to enrich for MSCs. For human cells, selection of Stro-1–positive cells has been demonstrated to result in a 10- to 20-fold enrichment of CFU-F relative to their incidence in unseparated human BM.24  Further enrichment of mesenchymal progenitor cells with capacity for differentiation into multiple lineages was obtained by combination of Stro-1 with CD106 (vascular adhesion molecule-1; VCAM-1).25  Caplan and colleagues described additional antibodies to identify human BM cells, including CD105 (SH2) and CD73 (SH3/SH4).26  The low-affinity nerve growth factor receptor (LNGFR; CD271) also has been shown to enrich for marrow stromal cells, and MSCs selected by CD271 expression were shown to have a 10- to 1000-fold higher proliferative capacity in comparison to MSCs isolated by plastic adherence.27  Recently, Buhring et al developed new antibodies for prospective isolation of MSCs, including W8B2 and frizzled-9 (FZD9), for the isolation of human MSCs from BM and placenta, respectively.28 

Immunosuppressive properties of MSCs

An emerging body of data indicate that MSCs possess immunomodulatory properties29,30  and may play specific roles as immunomodulators in maintenance of peripheral tolerance, transplantation tolerance, autoimmunity, tumor evasion, as well as fetal-maternal tolerance.

MSC-mediated immunosuppression in vitro

The interaction between MSCs and T cells.

The first indications for the immunosuppressive nature of MSCs were derived from studies with human,31-34  baboon35  and murine MSCs36,37  that demonstrated that MSCs were able to suppress T lymphocyte activation and proliferation in vitro. This inhibition affects the proliferation of T cells induced by alloantigens,32-34  mitogens31  as well as activation of T cells by CD3 and CD28 antibodies.34,37  MSCs have been reported to inhibit the cytotoxic effects of antigen-primed cytotoxic T cells (CTLs),33  that might be due to suppression of the proliferation of CTLs, rather than to a direct inhibition of cytolytic activity.38,39  Suppression of T cell proliferation by MSCs has no immunological restriction, similar suppressive effects being observed with cells that were autologous or allogeneic to the responder cells.32,37  Most studies agree that soluble factors are involved because separation of MSCs and PBMCs by a semi permeable membrane (transwell) does not prevent inhibition of proliferation.31,34,39  Supernatants from human and mouse MSCs cultures show no inhibitory effect,33,41,42  unless MSCs have been cocultured with lymphocytes,36  suggesting that the suppressive factor(s) are not constitutively secreted by MSCs, but require a dynamic cross talk between MSCs and T-lymphocytes. A role for TGF-β and hepatocyte growth factor (HGF) as mediators for suppression of T-cell proliferation in a mixed lymphocyte reaction has been suggested by Di Nicola et al, who found that neutralizing antibodies against TGF-β and HGF restored the proliferative response of T cells.31  Others demonstrated that these factors do not play a role in the suppressive effect by MSCs on T cells stimulated with mitogens42,43  and suggest that different mechanisms are involved depending on the stimuli. Prostaglandin E2 (PGE2) represents another candidate molecule. MSCs constitutively produce PGE2, a process that is enhanced upon coculture with PBMCs.34  Although inhibition of PGE2 synthesis was demonstrated to mitigate MSC-mediated suppression of T-cell proliferation and cytokine production by T cells,44  other studies contradict these findings.34,43  More recently, the tryptophan catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been suggested to play a role in the suppression of T-cell proliferation by MSCs.45  Upon stimulation with IFN-γ, MSCs express IDO activity that degrades essential tryptophan and results in kynurenine breakdown products, resulting in reduced lymphocyte proliferation. However, the role of IDO in MSC-mediated suppression is not very clear.31,34,44  While one recent study indicated that MSCs induce apoptosis of T cells due to the conversion of tryptophan to kynurenine,46  several other studies however found no effect of addition of MSC on apoptosis of T cells.29,32,42  MSC-induced T-cell anergy has been proposed as another potential mechanism of immune suppression. MSCs lack surface expression of costimulatory molecules, such as CD80 (B7-1) and CD86 (B7-2), and it is believed that MSCs can render T cells anergic. Several studies have shown that the unresponsiveness of T cells in the presence of MSCs was transient and could be restored after removal of MSCs,31,37  whereas others have demonstrated in murine models that T-cell tolerance was induced.47  Alternatively, MSCs have been demonstrated to induce a condition of anergy due to divisional arrest in T cells (ie, following removal of MSCs); IFN-γ production but not proliferation of murine PBMCs was restored, despite addition of exogenous IL-2.48  The discrepancy between these studies may be due to different experimental conditions or the origin of the MSCs.

Another level at which MSCs may modulate immune responses is through the induction of regulatory T cells (Treg). MSCs have been reported to induce formation of CD8+ regulatory T cells that were responsible for inhibition of allogeneic lymphocyte proliferation.36  Furthermore, an increase in the population of CD4+CD25+ cells, displaying a regulatory phenotype (ie, Fox P3 positivity) has been demonstrated in mitogen-stimulated PBMC cultures in the presence of MSC,38,44  although the functional properties (ie, the suppression of T-cell proliferation) of these cells have not yet been demonstrated. In contrast, depletion of CD4+CD25+ regulatory T cells had no effect on the inhibition of T-cell proliferation by MSCs.37 

Although these data are generally interpreted to indicate an immunosuppressive role of MSCs, the current evidence shows that MSCs merely suppress T-cell proliferation in vitro and thereby primarily affects the effector arm of T-cell immune response. In support of the hypothesis that the “immunosuppressive” effect of MSCs might be ascribed to a nonspecific antiproliferative effect is a recent study by Ramasamy et al, who described that MSCs also inhibited the proliferation of malignant cells of different lineages.40 

Antigen-presenting cells are directed toward a regulatory phenotype by MSCs.

Dendritic cells (DCs) play a key role in the induction of immunity and tolerance, depending on the activation and maturation stage and, as recently proposed, the cytokine milieu at sites of inflammation.49  MSCs have been demonstrated to interfere with DC differentiation, maturation and function. Addition of MSCs results in inhibition of differentiation of both monocytes and CD34+ progenitors into CD1a+-DCs, skewing their differentiation toward cells with features of macrophages. DCs generated in the presence of MSCs were impaired in their response to maturation signals and exhibited no expression of CD83 or up-regulation of HLA-DR and costimulatory molecules.50-52  Consistent with these findings, immature DCs generated in the presence of MSCs were strongly hampered in their ability to induce activation of T cells. In addition, an altered cytokine production pattern, ie decreased production of proinflammatory cytokines tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and interleukin (IL)-12 and increased production of the anti-inflammatory cytokine IL-10 in MSC/monocyte culture, was also observed.44,50,53  Taken together, these results suggest that MSCs suppress the differentiation of dendritic cells, resulting in the formation of immature DCs that exhibit a suppressor or inhibitory phenotype.

Transwell experiments have indicated that the suppressive effect of MSCs on DC differentiation is mediated by soluble factors.51  The production of IL-6 and M-CSF by MSCs may contribute to the inhibitory effect of MSCs on DC differentiation, although blocking studies indicate that these factors are not solely responsible for the inhibitory effect. Alternatively, PGE2 might be an intriguing candidate factor. Inhibition of PGE2 synthesis restored the secretion of TNF-α and IFN-γ by DCs cultured in the presence of MSCs.44  The increased production of IL-10 by DCs upon coculture with MSCs may also contribute to the suppressive effects of MSCs. Neutralizing antibodies to IL-10 indeed restored T-cell proliferation,53  although not completely. In addition to direct suppression of T-cell proliferation, the induction of regulatory antigen-presenting cells (APCs) might thus be a key mechanism by which MSCs indirectly suppress proliferation of T cells.

MSCs modulate B-cell functions.

In murine studies, MSCs have been reported to inhibit the proliferation of B cells, stimulated with anti-CD40L and IL-4,48  or with pokeweed mitogen.41  Allogeneic MSCs have been shown to inhibit the proliferation, activation and IgG secretion of B cells from BXSB mice that are used as an experimental model for human systemic lupus erythematosus.54  Consistent with the murine studies, human MSCs have been shown to inhibit proliferation of B cells activated with anti-Ig antibodies, soluble CD40 ligand and cytokines.55  In addition, differentiation, antibody production and chemotactic behavior of B cells was affected by MSCs.55  Krampera et al showed that MSCs only reduced the proliferation of B cells in the presence of IFN-γ. The suppressive effect of IFN-γ was possibly related to its ability to stimulate the production of IDO by MSCs, which in turn suppresses the proliferative response of effector cells through the tryptophan pathway.56  Although the mechanisms involved in these activities are not yet fully understood, transwell experiments indicated that soluble factors released by MSCs were sufficient to inhibit proliferation of B cells.55  In contrast, culture supernatant from MSCs had no effect, suggesting that the release of inhibitory factors requires paracrine signals from B cells.

Interaction between MSCs and natural killer cells.

Natural killer (NK) cells exhibit spontaneous cytolytic activity that mainly targets cells that lack expression of HLA class I molecules. Killing by NK cells is regulated by a balance of signals transmitted by activating and inhibitory receptors interacting with HLA molecules on target cells. However, NK cells are also able to lyse autologous tumor cells regulated by their activating receptors.57  It has been suggested that MSCs suppress IL-2 or IL-15 driven NK-cell proliferation and IFN-γ production.38,39,44,58  MSCs do not inhibit the lysis of freshly isolated NK cells,39  whereas NK cells cultured for 4 to 5 days with IL-2 in the presence of MSCs have a reduced cytotoxic potential against K562 target cells.56  Furthermore, Sotiropoulou et al demonstrated that short term culture with MSCs only affect NK-cell cytotoxicity against HLA class I-positive tumor cells but not against HLA class I-negative targets.58  These data indicate that MSCs exert an inhibitory effect on the NK-cell cytotoxicity against HLA class I-positive targets that are less susceptible to NK-mediated lysis than HLA class I-negative cells.

Experiments with transwell culture systems have indicated that MSCs are able to suppress the proliferation and cytokine production of IL-15 stimulated NK cells via soluble factors. In contrast, the inhibitory effect of MSCs on NK-cell cytotoxicity required cell-cell contact, suggesting the existence of different mechanisms for MSC-mediated NK-cell suppression.58  PGE2 secretion by MSCs was demonstrated to partially affect NK-cell proliferation, CD56 expression and cytotoxicity, but did not interfere with cytokine production or expression of activating receptors.58  Inhibition of TGF-β partially restores NK-cell proliferation, whereas blocking both PGE2 and TGF-β completely restored the proliferation capacity of NK cells, indicating that these factors suppress NK-cell activity by different mechanisms.

Until recently, MSCs were considered immunoprivileged and previous studies reported that MSCs were not lysed by freshly isolated NK cells.39,59  However, recent data indicate that activated NK cell are capable of effectively lysing MSCs.58,60  Although MSCs express normal levels of MHC class I that should protect against NK-mediated killing, MSCs express different ligands that are recognized by activating NK receptors that trigger NK alloreactivity.60  Treatment of MSCs with IFN-γ decreased their susceptibility to NK cell–mediated lysis due to up-regulation of HLA class I molecules.60 

Taken together, numerous studies convincingly demonstrate that MSCs are able to modulate the function of different immune cells in vitro, particularly involving the suppression of T-cell proliferation and the inhibition of DC differentiation (Figure 1). The mechanisms underlying the immunosuppressive effects of MSCS are still unclear and several different, sometimes contradictionary results have been proposed. Finally, the in vivo biological relevance of these in vitro observations has yet to be shown.

Figure 1

Immunomodulatory effects of MSCs. CTL indicates cytotoxic T cell; HGF, hepatocyte growth factor; IDO, indoleamine 2,3-dioxygenase; PGE2, prostaglandin E2; and TGF-β, transforming growth factor β. Illustration by Paulette Dennis.

Figure 1

Immunomodulatory effects of MSCs. CTL indicates cytotoxic T cell; HGF, hepatocyte growth factor; IDO, indoleamine 2,3-dioxygenase; PGE2, prostaglandin E2; and TGF-β, transforming growth factor β. Illustration by Paulette Dennis.

Close modal

Immunosuppressive properties of MSCs in vivo

Animal models.

The immunomodulatory effects of MSCs have been examined in a variety of animal models related to alloreactive immunity (organ and stem cell transplantation), autoimmunity or tumor immunity (Table 1). One of the first in vivo studies demonstrated that systemic infusion of allogeneic MSCs derived from the BM of baboons prolonged the survival of allogeneic skin grafts to 11 days compared with 7 days in animals not receiving MSCs.35  In addition, we have recently demonstrated that infusion of syngeneic host-derived MSCs resulted in decreased rejection of allogeneic stem cell grafts in a murine allogeneic BM transplantation model,61  although we did not address possible immunological mechanisms underlying these observations.

Table 1

In vivo immunosuppressive effects of MSCs

Animal, modelMSCsOutcomeReference no.
Mouse   36  
    Melanoma Allogeneic IV and SC infusion Promotion of tumor growth — 
    GVHD Allogeneic multiple IV infusions Prevention of GVHD 62  
    GVHD Allogeneic single IV infusion No effect on development of GVHD 63  
    Graft rejection Syngeneic Decreased graft rejection 61  
    EAE Allogeneic IV infusion Prevention of EAE development 47  
    CIA Allogeneic IV infusion No effect 65  
Baboon: Skin graft transplantation Allogeneic IV infusion Prolonged skin graft survival 35  
Rat: Ischemia/reperfusion injury Allogeneic IV infusion Protection against renal ischemia/reperfusion injury 66  
Human    
    Acute myeloid leukemia Haploidentical IV infusion HSC engraftment with no GVHD 91  
    Severe acute GVHD Haploidentical IV infusion Resolution of grade IV acute GVHD 71, 73  
    Leukemia Haploidentical IV infusion Rapid platelet engraftment, low incidence of GVHD 70  
Animal, modelMSCsOutcomeReference no.
Mouse   36  
    Melanoma Allogeneic IV and SC infusion Promotion of tumor growth — 
    GVHD Allogeneic multiple IV infusions Prevention of GVHD 62  
    GVHD Allogeneic single IV infusion No effect on development of GVHD 63  
    Graft rejection Syngeneic Decreased graft rejection 61  
    EAE Allogeneic IV infusion Prevention of EAE development 47  
    CIA Allogeneic IV infusion No effect 65  
Baboon: Skin graft transplantation Allogeneic IV infusion Prolonged skin graft survival 35  
Rat: Ischemia/reperfusion injury Allogeneic IV infusion Protection against renal ischemia/reperfusion injury 66  
Human    
    Acute myeloid leukemia Haploidentical IV infusion HSC engraftment with no GVHD 91  
    Severe acute GVHD Haploidentical IV infusion Resolution of grade IV acute GVHD 71, 73  
    Leukemia Haploidentical IV infusion Rapid platelet engraftment, low incidence of GVHD 70  

EAE indicates experimental acute encephalomyelitis; IV, intravenous; SC, subcutaneous; and —, not applicable.

One of the most impressive in vivo effects of MSCs has been observed in the treatment of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation. Systemical infusion of ex-vivo expanded MSCs derived from adipose tissue was able to control lethal GVHD in mice transplanted with haploidentical hematopoietic stem cells grafts.62  This study demonstrated that only infusions of MSCs early after transplantation were effective in controlling GVHD. Moreover, it was suggested that repeated infusions of MSCs are required to ameliorate GVHD and this might explain a recent observation that the infusion of a single dose of MSCs at the time of an allogeneic BM transplantation did not affect the incidence and severity of GVHD in mice.63 

Modulation of autoimmunity is considered a potential novel target for MSC treatment. Recently, 3 reports on the effects of MSCs in animal models of autoimmunity have appeared. Murine MSCs have been demonstrated to ameliorate experimental autoimmune encephalomyelitis (EAE), a model of human multiple sclerosis, through the induction of peripheral T cell tolerance against the pathogenic antigen.47,64  The infusion of MSCs was only effective at disease onset and at the peak of the disease, but not after disease stabilization. In contrast, infusion of MSCs had no beneficial effects on collagen-induced arthritis (CIA) as tested in a murine model of rheumatoid arthritis (RA).65 

Djouad et al demonstrated that MSCs prevented the rejection of allogeneic tumor cells in immunocompetent mice. MSCs infused systemically or adjacent to subcutaneously implanted B16 melanoma cells resulted in enhanced tumor formation, whereas melanoma cells injected alone were eliminated by the host immune system.36  MSCs have also been demonstrated to provide tissue protective effects in a rat kidney model of ischemia/reperfusion injury, possibly mediated by the secretion of soluble immunomodulating factors.66  Moreover, infusion of rat MSCs in an experimental rat model of glomerulonephritis was shown to accelerate glomerular healing, probably related to the release of growth factors.67  In line with this are recent observations demonstrating that MSCs preferentially engraft at sites of tissue damage or tumor growth.68 

Clinical experience.

The immunosuppressive capacities of MSCs have generated clinical interest in the field of solid organ of hematopoietic stem cell (HSC) transplantation in order to prevent graft rejection and to prevent or control graft-versus-host disease following HSC transplantation. Due to their immunosuppressive properties, MSCs are considered a potential cellular therapy for prevention of graft rejection and GVHD. The first clinical studies were performed to assess the safety of MSC infusion. A clinical study in breast cancer patients69  showed that the infusion of MSCs was safe and resulted in a rapid hematopoietic recovery. In a multicenter clinical trial, culture-expanded MSCs derived from the bone marrow of HLA-identical siblings were coinfused with HLA-identical hematopoietic stem-cells in 46 patients undergoing allogeneic stem-cell transplantation after a myeloablative conditioning regimen.70  MSCs were infused 4 hours before infusion of the stem cell graft without any infusion-related adverse events, ectopic tissue formation, or increase in the incidence or severity of GVHD. In comparison with historical controls, no acceleration of hematopoietic engraftment or prevention of graft rejection was observed.

In an European phase I-II study, 13 children received a haploidentical stem-cell graft in combination with expanded MSCs derived from the marrow of the stem-cell donor.71  No immediate adverse effects were observed after infusion of MSCs, while the incidence of graft failure or rejection in a control cohort of 52 patients was 20%, all patients engrafted and the preliminary data demonstrated an accelerated leukocyte recovery, although platelet and neutrophil engraftment kinetics were similar. Further support for the possible clinical benefit of MSCs has been presented by several case reports. The results from such case reports suggests that MSCs may not only exert preventive effects on the development of GVHD, but also exhibit therapeutic effects in established GVHD of the gut after allogeneic stem-cell transplantation.72  At present, additional patients with acute and chronic GVHD have been treated with MSCs. No side effects were seen after MSC infusions. Among the 40 patients treated for severe acute GVHD, 19 had complete responses, 9 patients showed improvements, 7 patients did not respond, 4 had stable disease, and 1 patient was not evaluated due to short follow-up. Twenty-one patients are alive between 6 weeks up to 3.5 years after transplantation. Of these patients 9 developed extensive chronic GVHD.73  Two prospective randomized European phase III studies recently have been launched to further explore the therapeutic usefulness of MSCs for the treatment or prevention of acute GVHD following allogeneic stem- cell transplantation.

The mechanisms underlying the possible in vivo immunomodulatory effects of MSCs remain a critical and unresolved question. It has been difficult to recover MSCs from BM of recipients who received transplants. Therefore it is conceivable that MSCs home to other tissues or organs to mediate immune suppression. In line with this hypothesis are recent observations in animal studies demonstrating that MSCs migrate to lymphoid organs47  and engraft at sites of tissue damage or tumor growth.68  Several studies indicated that after systemic administration, MSCs lodge nonspecifically in the capillary beds of various tissues, mainly the lungs.74  Interestingly, a murine study demonstrated that ex vivo expansion of MSCs dramatically reduced their homing and engraftment capacity.75 

The ability of MSCs to prevent or reverse GVHD may be via secretion of soluble factors or direct cell-cell contact on alloreactive T cells or by suppressing DC function. Alternatively or additionally, MSCs might increase the healing of wounded tissue by providing soluble factors, transdifferentiation, or cell fusion.76 

Safety concerns

Little is know regarding the in vivo survival of MSCs, and there are no clinical studies reporting whether MSCs remain present after transplantation. Although the few clinical studies performed to date confirm safety of infusion of MSCs, the lack of adverse effects might be due to the limited survival of MSCs. Therefore, concerns remain over the potential of systemic immune suppression, ectopic tissue formation, malignant transformation, and immunogenicity. Further controlled studies are required to address these concerns.

Systemic administration

The disadvantage of current immunosuppressive drugs is that they do not distinguish between pathological immune responses and protective immune responses. Systemic immunosuppression may therefore also depress host immune responses against infections caused by bacteria, fungi, and viruses. This is particularly important in allogeneic stem cell transplant or solid organ transplant recipients and for patients with autoimmune diseases, since in these patients the immune system is already compromised. It is therefore of importance to critically examine the effects of immunosuppression induced by MSCs and to compare with immunosuppressive agents currently used in the clinic. MSCs have been demonstrated to home to sites of injury and may therefore provide site-specific and local immunosuppression. This may serve as a potential mechanism to induce organ or tissue specificity. In contrast, a recent animal study demonstrated that local as well as systemic infusion of MSCs systemically suppressed the host antitumor immune response, thereby favoring allogeneic tumor formation.36  The immunosuppressive effects of MSCs are antigen nonspecific and independent of MHC expression: similar inhibitory effects being reported with MSCs that were autologous, allogeneic, or xenogeneic to the responder cells.35,36,38  Infusion of MSCs might therefore provide nonspecific systemic immunosuppression.

Ectopic tissue formation.

MSCs have the ability to differentiate into several mesenchymal lineages and therefore are considered attractive candidates for tissue repair. Horwitz et al have reported the use of MSCs for the repair of bone in patients with osteogenesis imperfecta (OI).78  It is assumed that the differentiation of MSCs toward a particular tissue lineage is primarily driven by the tissue-specific microenvironment. If true, this may serve as a mechanism protecting against cross-differentiation toward other mesenchymal tissue lineages. Recently, however, calcifications were observed in the infarcted hearts at mice that received local MSC treatment.77  In view of the paucity of available clinical data, ectopic tissue formation after MSC treatment therefore remains an important clinical safety concern.

Malignant transformation of MSCs.

Although MSCs have emerged as a promising tool for clinical applications due to the relatively simple requirements of ex vivo expansion without loss of their differentiation potential, culture expansion may (negatively) alter the functional in vivo characteristics. No immortalization and transformation has been observed after expansion of human MSCs, although it has been recently demonstrated that adipose tissue-derived MSCs undergo spontaneous transformation upon prolonged ex vivo expansion under stressful conditions.79  Murine cells are described to be more susceptible to chromosomal aberration under in vitro cultivation. Furthermore, the growth characteristics of murine MSCs differ from human MSCs in that they exhibit a significantly prolonged lag phase. The exponential growth observed after this phase might be due to the proliferation of transformed cells with a potential growth advantage. Emerging evidence in mice suggest that tumors may originate from spontaneous mutations of mesenchymal stem cells.80  In line with these observations are results from our own group demonstrating that even short-term culture was sufficient for the transformation of murine MSCs into a cell population with autonomous growth and biological characteristics of osteosarcoma.81  While no in vivo transformation or tumor formation has been observed in patients, these observations underscore the requirement for further studies on the genetic stability of expanded MSCs. It seems appropriate to test expanded MSC products for the presence of a normal karyotype before administration.

Immunogenicity of MSCs.

MSCs are considered to be hypoimmunogenic, displaying low expression levels of human leukocyte antigen (HLA) major histocompatibility complex (MHC) class I and, importantly, no expression of costimulatory molecules.34  An emerging body of data indicate that MSCs do not elicit a proliferative response by allogeneic lymphocytes.33-35,37  However, MSCs directly suppress T-cell proliferation in an antigen-independent fashion, and therefore it is not appropriate to use proliferation as a read-out for immunogenicity. In vivo studies demonstrated that MSCs avoid normal alloresponses.82  These characteristics support the possibility of exploiting universal donor MSCs for therapeutic applications. However, recent evidence indicates that MSCs can function as APCs and activate immune responses under appropriate conditions.83,84  MSCs are able to take up antigens, and after stimulation with IFNγ, to induce T-cell responses to recall antigens.83  Stagg et al reported that IFNγ-stimulated MSCs induced antigen-specific responses of primary ovalbumin (OVA)–specific transgenic T cells.84  Moreover, when mice were immunized with OVA-loaded, IFNγ-stimulated MSCs, they developed antigen-specific cytotoxic CD8+ T cells and rejected OVA-expressing tumor cells. We have demonstrated that infusion of allogeneic MSCs can prime naive T cells in immunocompetent mice.61  Furthermore, in a recent study it was demonstrated that subcutaneously implanted allogeneic MSCs were rejected in nonimmunosuppressed recipient mice. Splenocytes isolated from mice that had been implanted with allogeneic MSCs displayed a significant IFN-γ response against allogeneic MSCs in vitro.85  Xenotransplantation studies with human MSCs suggest that MSCs are not intrinsically immunoprivileged. Intracoronary injection of adult human MSCs in rat myocardium was associated with rejection and significant infiltration of mainly macrophages, whereas persistent engraftment of adult human MSCs was observed in immunological incompetent rats.86  These studies support the notion that allogeneic MSCs can engraft in immunocompromised hosts or at immunoprivileged sites but trigger an immune response in hosts with an intact immune system. Although further studies of the immunogenicity of MSCs are needed, rejection of MSCs and its clinical consequences should therefore be carefully considered in clinical trials. On the other hand, rejection of allogeneic MSCs might be profitable: in this way MSCs only temporary suppress the immune system, thereby reducing the risk of infection, malignant transformation, or suppression of a graft-versus-tumor effect.

Physiological role for MSCs in immunosuppression

The current knowledge about MSCs is primarily derived from studies performed on ex vivo expanded cells. There is a general consensus that MSCs are residents of the microenvironment and play a role in supporting hematopoiesis. However, whether the immunosuppressive properties of MSCs play a physiological role in maintaining immune homeostasis has not been established.

Recent observations suggested that MSCs from patients with autoimmune diseases are affected. MSCs derived from the BM of patients with severe aplastic anemia are deficient in their ability to suppress T-cell proliferation and cytokine release.87  Whether these defects are relevant for the pathogenesis of aplastic anemia remains to be shown. Both stromal and endothelial progenitors in patients with systemic sclerosis also have been reported to be functionally impaired, showing reduced proliferation and differentiation capacity. It has been suggested that the functional impairment of the BM microenvironment might play a role in the impaired vasculogenesis in scleroderma.88  It may therefore be hypothesized that the immunosuppressive capacities of MSCs might play a role in the BM microenvironment to create an immunoprivileged site that protects primitive stem cells against bystander effects of local immune responses.

The observation that human MSCs can be isolated from decidua,8  amniotic fluid,9  fetal blood,10  and umbilical cord blood11,12  may indicate a role for MSCs in fetal tolerance. Fetal immune responses to paternal antigens are suppressed by a phenomenon called “immune privilege.”89  The emerging data on the mechanisms contributing to immune privilege in the pregnant uterus show striking similarity to the immunosuppressive effects of MSCs, including the production of IDO,45  and support the hypothesis that MSCs are involved in fetal tolerance.90  Further studies are required to dissect the potential of MSCs in providing immune privilege in the pregnant uterus and to gain more insight in the mechanisms of immune inhibition.

The current body of data on the immunosuppressive properties of MSCs holds great promise for treating immune-mediated disorders. Despite the fact that relatively little is known about their in vivo biology, MSCs already have been introduced in the clinic. Preliminary clinical results are encouraging, and randomized studies are under way. However, it is important not to overestimate the potential therapeutic effects of MSCs, and many questions need to be addressed before the putative therapeutic promise of these cells can be realized. At present, MSCs are extensively characterized in a culture-expanded state, and relatively little is known of their biological properties in an unmanipulated state. Culture expansion of MSCs may alter their fundamental biological properties and changes may occur, including the accumulation of molecular alterations. In addition, the use of different isolation methods and culture conditions has led to multiple populations described as “MSCs” with different, sometimes conflicting, characteristics. There is an obvious need for standardization, and although several markers have been examined to prospectively identify and isolate human MSCs, there is yet no defined universal marker.

Directions for future research include (i) standardization and validation of the isolation and culture expansion method of MSCs used in animal and clinical studies, to facilitate comparisons between cell products generated at different sites; (ii) identification of cell-surface specific markers in order to dissect the hierarchy within MSC populations and facilitate the generation of homogenous cell populations; (iii) animal studies to unravel the mechanism underlying the immunosuppressive effects of MSCs in order to optimize the potential therapeutic application; (iv) in vivo tracking studies to examine the in vivo survival and homing of MSCs; and (v) multicenter randomized clinical trials to further assess safety and efficacy. Finally, in vivo tracking of MSCs in patients will allow directed diagnostic interventions to further study their local effects that may explain their biological activities. A better understanding of this fascinating cell population might realize a novel therapeutic strategy to modulate immune responses in a variety of immune-mediated diseases.

This manuscript was supported by research grant 03-3014 from the Dutch Cancer Society, the Netherlands, and EuroCord Nederland Foundation.

Contribution: A.J.N. and W.E.F. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Willem E. Fibbe, Department of Immunohematology and Blood Transfusion, E3-Q, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; e-mail:w.e.fibbe@lumc.nl.

1
Friedenstein
 
AJ
Gorskaja
 
JF
Kulagina
 
NN
Fibroblast precursors in normal and irradiated mouse hematopoietic organs.
Exp Hematol
1976
, vol. 
4
 (pg. 
267
-
274
)
2
Prockop
 
DJ
Marrow stromal cells as stem cells for nonhematopoietic tissues.
Science
1997
, vol. 
276
 (pg. 
71
-
74
)
3
Caplan
 
AI
Mesenchymal stem cells.
J Orthop Res
1991
, vol. 
9
 (pg. 
641
-
650
)
4
Horwitz
 
EM
Le Blanc
 
K
Dominici
 
M
, et al. 
Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement.
Cytotherapy
2005
, vol. 
7
 (pg. 
393
-
395
)
5
Anjos-Afonso
 
F
Bonnet
 
D
Non-hematopoietic/endothelial SSEA-1pos cells defines the most primitive progenitors in the adult murine bone marrow mesenchymal compartment.
Blood
2007
, vol. 
109
 (pg. 
1298
-
1306
)
6
Pittenger
 
MF
Mackay
 
AM
Beck
 
SC
, et al. 
Multilineage potential of adult human mesenchymal stem cells.
Science
1999
, vol. 
284
 (pg. 
143
-
147
)
7
Zuk
 
PA
Zhu
 
M
Mizuno
 
H
, et al. 
Multilineage cells from human adipose tissue: implications for cell-based therapies.
Tissue Eng
2001
, vol. 
7
 (pg. 
211
-
228
)
8
In 't Anker
 
PS
Scherjon
 
SA
Kleijburg-van der Keur
 
C
, et al. 
Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta.
Stem Cells
2004
, vol. 
22
 (pg. 
1338
-
1345
)
9
In 't Anker
 
PS
Scherjon
 
SA
Kleijburg-van der Keur
 
C
, et al. 
Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation.
Blood
2003
, vol. 
102
 (pg. 
1548
-
1549
)
10
In 't Anker
 
PS
Noort
 
WA
Scherjon
 
SA
, et al. 
Mesenchymal stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential.
Haematologica
2003
, vol. 
88
 (pg. 
845
-
852
)
11
Bieback
 
K
Kern
 
S
Kluter
 
H
Eichler
 
H
Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood.
Stem Cells
2004
, vol. 
22
 (pg. 
625
-
634
)
12
Kogler
 
G
Sensken
 
S
Airey
 
JA
, et al. 
A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential.
J Exp Med
2004
, vol. 
200
 (pg. 
123
-
135
)
13
Deans
 
RJ
Moseley
 
AB
Mesenchymal stem cells: biology and potential clinical uses.
Exp Hematol
2000
, vol. 
28
 (pg. 
875
-
884
)
14
Sanchez-Ramos
 
J
Song
 
S
Cardozo-Pelaez
 
F
, et al. 
Adult bone marrow stromal cells differentiate into neural cells in vitro.
Exp Neurol
2000
, vol. 
164
 (pg. 
247
-
256
)
15
Schwartz
 
RE
Reyes
 
M
Koodie
 
L
, et al. 
Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells.
J Clin Invest
2002
, vol. 
109
 (pg. 
1291
-
1302
)
16
Caplan
 
AI
Bruder
 
SP
Mesenchymal stem cells: building blocks for molecular medicine in the 21st century.
Trends Mol Med
2001
, vol. 
7
 (pg. 
259
-
264
)
17
Jiang
 
Y
Jahagirdar
 
BN
Reinhardt
 
RL
, et al. 
Pluripotency of mesenchymal stem cells derived from adult marrow.
Nature
2002
, vol. 
418
 (pg. 
41
-
49
)
18
Yoon
 
YS
Wecker
 
A
Heyd
 
L
, et al. 
Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction.
J Clin Invest
2005
, vol. 
115
 (pg. 
326
-
338
)
19
D'Ippolito
 
G
Diabira
 
S
Howard
 
GA
, et al. 
Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential.
J Cell Sci
2004
, vol. 
117
 (pg. 
2971
-
2981
)
20
Kucia
 
M
Reca
 
R
Campbell
 
FR
, et al. 
A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow.
Leukemia
2006
, vol. 
20
 (pg. 
857
-
869
)
21
Rodriguez
 
AM
Elabd
 
C
Delteil
 
F
, et al. 
Adipocyte differentiation of multipotent cells established from human adipose tissue.
Biochem Biophys Res Commun
2004
, vol. 
315
 (pg. 
255
-
263
)
22
De
 
CP
Bartsch
 
G
Siddiqui
 
MM
, et al. 
Isolation of amniotic stem cell lines with potential for therapy.
Nat Biotechnol
2007
, vol. 
25
 (pg. 
100
-
106
)
23
Verfaillie
 
CM
Pera
 
MF
Lansdorp
 
PM
Stem cells: hype and reality.
Hematology Am Soc Hematol Educ Program
2002
(pg. 
369
-
391
)
24
Simmons
 
PJ
Torok-Storb
 
B
Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1.
Blood
1991
, vol. 
78
 (pg. 
55
-
62
)
25
Gronthos
 
S
Zannettino
 
AC
Hay
 
SJ
, et al. 
Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow.
J Cell Sci
2003
, vol. 
116
 (pg. 
1827
-
1835
)
26
Haynesworth
 
SE
Baber
 
MA
Caplan
 
AI
Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies.
Bone
1992
, vol. 
13
 (pg. 
69
-
80
)
27
Quirici
 
N
Soligo
 
D
Bossolasco
 
P
, et al. 
Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies.
Exp Hematol
2002
, vol. 
30
 (pg. 
783
-
791
)
28
Buhring
 
HJ
Battula
 
VL
Treml
 
S
Kanz
 
L
Vogel
 
W
Novel markers for the isolation of primary bone marrow derived MSC with multi-lineage differentiation capacity. [abstract].
Blood
2006
, vol. 
108
  
Abstract, 2573
29
Rasmusson
 
I
Immune modulation by mesenchymal stem cells.
Exp Cell Res
2006
, vol. 
312
 (pg. 
2169
-
2179
)
30
Uccelli
 
A
Moretta
 
L
Pistoia
 
V
Immunoregulatory function of mesenchymal stem cells.
Eur J Immunol
2006
, vol. 
36
 (pg. 
2566
-
2573
)
31
Di Nicola
 
M
Carlo-Stella
 
C
Magni
 
M
, et al. 
Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli.
Blood
2002
, vol. 
99
 (pg. 
3838
-
3843
)
32
Le Blanc
 
K
Tammik
 
L
Sundberg
 
B
Haynesworth
 
SE
Ringden
 
O
Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex.
Scand J Immunol
2003
, vol. 
57
 (pg. 
11
-
20
)
33
Potian
 
JA
Aviv
 
H
Ponzio
 
NM
Harrison
 
JS
Rameshwar
 
P
Veto-like activity of mesenchymal stem cells: functional discrimination between cellular responses to alloantigens and recall antigens.
J Immunol
2003
, vol. 
171
 (pg. 
3426
-
3434
)
34
Tse
 
WT
Pendleton
 
JD
Beyer
 
WM
Egalka
 
MC
Guinan
 
EC
Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation.
Transplantation
2003
, vol. 
75
 (pg. 
389
-
397
)
35
Bartholomew
 
A
Sturgeon
 
C
Siatskas
 
M
, et al. 
Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo.
Exp Hematol
2002
, vol. 
30
 (pg. 
42
-
48
)
36
Djouad
 
F
Plence
 
P
Bony
 
C
, et al. 
Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals.
Blood
2003
, vol. 
102
 (pg. 
3837
-
3844
)
37
Krampera
 
M
Glennie
 
S
Dyson
 
J
, et al. 
Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide.
Blood
2003
, vol. 
101
 (pg. 
3722
-
3729
)
38
Maccario
 
R
Podesta
 
M
Moretta
 
A
, et al. 
Interaction of human mesenchymal stem cells with cells involved in alloantigen-specific immune response favors the differentiation of CD4+ T-cell subsets expressing a regulatory/suppressive phenotype.
Haematologica
2005
, vol. 
90
 (pg. 
516
-
525
)
39
Rasmusson
 
I
Ringden
 
O
Sundberg
 
B
Le Blanc
 
K
Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells.
Transplantation
2003
, vol. 
76
 (pg. 
1208
-
1213
)
40
Ramasamy
 
R
Lam
 
EW
Soeiro
 
I
, et al. 
Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth.
Leukemia
2006
, vol. 
21
 (pg. 
304
-
310
)
41
Augello
 
A
Tasso
 
R
Negrini
 
SM
, et al. 
Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway.
Eur J Immunol
2005
, vol. 
35
 (pg. 
1482
-
1490
)
42
Le Blanc
 
K
Rasmusson
 
I
Gotherstrom
 
C
, et al. 
Mesenchymal stem cells inhibit the expression of CD25 (interleukin-2 receptor) and CD38 on phytohaemagglutinin-activated lymphocytes.
Scand J Immunol
2004
, vol. 
60
 (pg. 
307
-
315
)
43
Rasmusson
 
I
Ringden
 
O
Sundberg
 
B
Le Blanc
 
K
Mesenchymal stem cells inhibit lymphocyte proliferation by mitogens and alloantigens by different mechanisms.
Exp Cell Res
2005
, vol. 
305
 (pg. 
33
-
41
)
44
Aggarwal
 
S
Pittenger
 
MF
Human mesenchymal stem cells modulate allogeneic immune cell responses.
Blood
2005
, vol. 
105
 (pg. 
1815
-
1822
)
45
Meisel
 
R
Zibert
 
A
Laryea
 
M
, et al. 
Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation.
Blood
2004
, vol. 
103
 (pg. 
4619
-
4621
)
46
Plumas
 
J
Chaperot
 
L
Richard
 
MJ
, et al. 
Mesenchymal stem cells induce apoptosis of activated T cells.
Leukemia
2005
, vol. 
19
 (pg. 
1597
-
1604
)
47
Zappia
 
E
Casazza
 
S
Pedemonte
 
E
, et al. 
Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy.
Blood
2005
, vol. 
106
 (pg. 
1755
-
1761
)
48
Glennie
 
S
Soeiro
 
I
Dyson
 
PJ
Lam
 
EW
Dazzi
 
F
Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells.
Blood
2005
, vol. 
105
 (pg. 
2821
-
2827
)
49
Rutella
 
S
Danese
 
S
Leone
 
G
Tolerogenic dendritic cells: cytokine modulation comes of age.
Blood
2006
, vol. 
108
 (pg. 
1435
-
1440
)
50
Jiang
 
XX
Zhang
 
Y
Liu
 
B
, et al. 
Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells.
Blood
2005
, vol. 
105
 (pg. 
4120
-
4126
)
51
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
, vol. 
177
 (pg. 
2080
-
2087
)
52
Zhang
 
W
Ge
 
W
Li
 
C
, et al. 
Effects of mesenchymal stem cells on differentiation, maturation, and function of human monocyte-derived dendritic cells.
Stem Cells Dev
2004
, vol. 
13
 (pg. 
263
-
271
)
53
Beyth
 
S
Borovsky
 
Z
Mevorach
 
D
, et al. 
Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness.
Blood
2005
, vol. 
105
 (pg. 
2214
-
2219
)
54
Deng
 
W
Han
 
Q
Liao
 
L
, et al. 
Effects of allogeneic bone marrow-derived mesenchymal stem cells on T and B lymphocytes from BXSB mice.
DNA Cell Biol
2005
, vol. 
24
 (pg. 
458
-
463
)
55
Corcione
 
A
Benvenuto
 
F
Ferretti
 
E
, et al. 
Human mesenchymal stem cells modulate B-cell functions.
Blood
2006
, vol. 
107
 (pg. 
367
-
372
)
56
Krampera
 
M
Cosmi
 
L
Angeli
 
R
, et al. 
Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells.
Stem Cells
2006
, vol. 
24
 (pg. 
386
-
398
)
57
Smyth
 
MJ
Hayakawa
 
Y
Takeda
 
K
Yagita
 
H
New aspects of natural-killer-cell surveillance and therapy of cancer.
Nat Rev Cancer
2002
, vol. 
2
 (pg. 
850
-
861
)
58
Sotiropoulou
 
PA
Perez
 
SA
Gritzapis
 
AD
Baxevanis
 
CN
Papamichail
 
M
Interactions between human mesenchymal stem cells and natural killer cells.
Stem Cells
2006
, vol. 
24
 (pg. 
74
-
85
)
59
Le Blanc
 
K
Immunomodulatory effects of fetal and adult mesenchymal stem cells.
Cytotherapy
2003
, vol. 
5
 (pg. 
485
-
489
)
60
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
, vol. 
107
 (pg. 
1484
-
1490
)
61
Nauta
 
AJ
Westerhuis
 
G
Kruisselbrink
 
AB
, et al. 
Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting.
Blood
2006
, vol. 
108
 (pg. 
2114
-
2120
)
62
Yanez
 
R
Lamana
 
ML
Garcia-Castro
 
J
, et al. 
Adipose tissue-derived mesenchymal stem cells (AD-MSC) have in vivo immunosuppressive properties applicable for the control of graft-versus-host disease (GVHD).
Stem Cells
2006
, vol. 
24
 (pg. 
2582
-
2591
)
63
Sudres
 
M
Norol
 
F
Trenado
 
A
, et al. 
Bone marrow mesenchymal stem cells suppress lymphocyte proliferation in vitro but fail to prevent graft-versus-host disease in mice.
J Immunol
2006
, vol. 
176
 (pg. 
7761
-
7767
)
64
Zhang
 
J
Li
 
Y
Chen
 
J
, et al. 
Human bone marrow stromal cell treatment improves neurological functional recovery in EAE mice.
Exp Neurol
2005
, vol. 
195
 (pg. 
16
-
26
)
65
Djouad
 
F
Fritz
 
V
Apparailly
 
F
, et al. 
Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor alpha in collagen-induced arthritis.
Arthritis Rheum
2005
, vol. 
52
 (pg. 
1595
-
1603
)
66
Togel
 
F
Hu
 
Z
Weiss
 
K
, et al. 
Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms.
Am J Physiol Renal Physiol
2005
, vol. 
289
 (pg. 
F31
-
F42
)
67
Kunter
 
U
Rong
 
S
Djuric
 
Z
, et al. 
Transplanted mesenchymal stem cells accelerate glomerular healing in experimental glomerulonephritis.
J Am Soc Nephrol
2006
, vol. 
17
 (pg. 
2202
-
2212
)
68
Studeny
 
M
Marini
 
FC
Champlin
 
RE
, et al. 
Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors.
Cancer Res
2002
, vol. 
62
 (pg. 
3603
-
3608
)
69
Koc
 
ON
Gerson
 
SL
Cooper
 
BW
, 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
, vol. 
18
 (pg. 
307
-
316
)
70
Lazarus
 
HM
Koc
 
ON
Devine
 
SM
, 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
, vol. 
11
 (pg. 
389
-
398
)
71
Ball
 
LM
Bernardo
 
ME
Locatelli
 
F
, et al. 
Co-transplantation of haploidentical bone marrow derived mesenchymal stem cells overcomes graft dysfunction and improves hematological and lymphocyte recovery in haploidentical stem cell transplantation [abstract].
Blood
2006
, vol. 
108
  
Abstract, 3118
72
Le Blanc
 
K
Rasmusson
 
I
Sundberg
 
B
, et al. 
Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells.
Lancet
2004
, vol. 
363
 (pg. 
1439
-
1441
)
73
Le Blanc
 
K
Frassoni
 
F
Ball
 
LM
, et al. 
Mesenchymal stem cells for treatment of severe acute graft-versus-host disease [abstract].
Blood
2006
, vol. 
108
 pg. 
753
 
74
Gao
 
J
Dennis
 
JE
Muzic
 
RF
Lundberg
 
M
Caplan
 
AI
The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion.
Cells Tissues Organs
2001
, vol. 
169
 (pg. 
12
-
20
)
75
Rombouts
 
WJ
Ploemacher
 
RE
Primary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture.
Leukemia
2003
, vol. 
17
 (pg. 
160
-
170
)
76
Prockop
 
DJ
Gregory
 
CA
Spees
 
JL
One strategy for cell and gene therapy: harnessing the power of adult stem cells to repair tissues.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
Suppl 1
(pg. 
11917
-
11923
)
77
Breitbach
 
M
Bostani
 
T
Roell
 
W
, et al. 
Potential risks of bone marrow cell transplantation into infarcted hearts.
Blood
2007
, vol. 
110
 (pg. 
1362
-
1369
)
78
Horwitz
 
EM
Gordon
 
PL
Koo
 
WK
, 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
, vol. 
99
 (pg. 
8932
-
8937
)
79
Rubio
 
D
Garcia-Castro
 
J
Martin
 
MC
, et al. 
Spontaneous human adult stem cell transformation.
Cancer Res
2005
, vol. 
65
 (pg. 
3035
-
3039
)
80
Miura
 
M
Miura
 
Y
Padilla-Nash
 
HM
, et al. 
Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation.
Stem Cells
2006
, vol. 
24
 (pg. 
1095
-
1103
)
81
Tolar
 
J
Nauta
 
AJ
Osborn
 
MJ
, et al. 
Sarcoma derived from cultured mesenchymal stem cells.
Stem Cells
2006
, vol. 
25
 (pg. 
371
-
379
)
82
Koc
 
ON
Day
 
J
Nieder
 
M
, et al. 
Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH).
Bone Marrow Transplant
2002
, vol. 
30
 (pg. 
215
-
222
)
83
Chan
 
JL
Tang
 
KC
Patel
 
AP
, et al. 
Antigen-presenting property of mesenchymal stem cells occurs during a narrow window at low levels of interferon-gamma.
Blood
2006
, vol. 
107
 (pg. 
4817
-
4824
)
84
Stagg
 
J
Pommey
 
S
Eliopoulos
 
N
Galipeau
 
J
Interferon-gamma-stimulated marrow stromal cells: a new type of nonhematopoietic antigen-presenting cell.
Blood
2006
, vol. 
107
 (pg. 
2570
-
2577
)
85
Eliopoulos
 
N
Stagg
 
J
Lejeune
 
L
Pommey
 
S
Galipeau
 
J
Allogeneic marrow stromal cells are immune rejected by MHC class I and II mismatched recipient mice.
Blood
2005
, vol. 
106
 (pg. 
4057
-
4065
)
86
Grinnemo
 
KH
Mansson
 
A
Dellgren
 
G
, et al. 
Xenoreactivity and engraftment of human mesenchymal stem cells transplanted into infarcted rat myocardium.
J Thorac Cardiovasc Surg
2004
, vol. 
127
 (pg. 
1293
-
1300
)
87
Bacigalupo
 
A
Valle
 
M
Podesta
 
M
, et al. 
T-cell suppression mediated by mesenchymal stem cells is deficient in patients with severe aplastic anemia.
Exp Hematol
2005
, vol. 
33
 (pg. 
819
-
827
)
88
Del Papa
 
N
Quirici
 
N
Soligo
 
D
, et al. 
Bone marrow endothelial progenitors are defective in systemic sclerosis.
Arthritis Rheum
2006
, vol. 
54
 (pg. 
2605
-
2615
)
89
Niederkorn
 
JY
See no evil, hear no evil, do no evil: the lessons of immune privilege.
Nat Immunol
2006
, vol. 
7
 (pg. 
354
-
359
)
90
Barry
 
FP
Murphy
 
JM
English
 
K
Mahon
 
BP
Immunogenicity of adult mesenchymal stem cells: lessons from the fetal allograft.
Stem Cells Dev
2005
, vol. 
14
 (pg. 
252
-
265
)
91
Lee
 
ST
Jang
 
JH
Cheong
 
JW
, et al. 
Treatment of high-risk acute myelogenous leukaemia by myeloablative chemoradiotherapy followed by co-infusion of T cell-depleted haematopoietic stem cells and culture-expanded marrow mesenchymal stem cells from a related donor with one fully mismatched human leucocyte antigen haplotype.
Br J Haematol
2002
, vol. 
118
 (pg. 
1128
-
1131
)
Sign in via your Institution