Open access

Role of Bone Marrow Derived Mesenchymal Stem Cells in Management of Graft Versus Host Disease

Written By

Aisha Nasef

Submitted: 18 May 2012 Published: 08 May 2013

DOI: 10.5772/55013

From the Edited Volume

Stem Cell Biology in Normal Life and Diseases

Edited by Kamran Alimoghaddam

Chapter metrics overview

2,357 Chapter Downloads

View Full Metrics

1. Introduction

Human bone marrow stromal cells, referred as mesenchymal stem cells (MSC) are multipotent unspecialized cells localized in the medullary stroma [1,2]. They have a capacity for self-renewal and differentiation into multiple cell lineages [3-6].

Mesenchymal and tissue stem cell committee of the International society for cellular therapy proposes minimal criteria to define human MSCs. First, MSCs must be plastic-adherent when maintained in standard culture conditions. Second, MSCs must express CD105, CD73 and CD90, and lack the expression of CD45, CD34, CD14 or CD11b, CD79alpha or CD19 and HLA-DR surface molecules. Third, MSCs must be capable to differentiate into osteoblasts, adipocytes and chondroblasts in vitro [7,8]. In the hand of Delorme B and Charbord P, MSCs can be defined phenotypically with a minimal set of markers as CD31-, CD34-, and CD45-negative cells and CD13, CD29, CD73, CD90, CD105, and CD166 positive cells [9]. MSCs have been used in cell-based therapy in various disease conditions. Experimental evidence and preliminary clinical studies have demonstrated that MSCs, have an important immunomodulatory function in the management of allogeneic hematopoietic stem cell (HSC) transplantation [10,11]. These immunomodulatory effects have been demonstrated in various species, including humans, rodents, and primates and show clinical promise for the treatment of graft versus host disease (GVHD) and graft failure management. In this chapter we will discuss current research finding.

Advertisement

2. General background

2.1. MSC administration

2.1.1. Safety and therapeutic methods of MSC administration

The extensive capacity for expansion in vitro at clinical scale has recently facilitated the development of clinical trials designed to assess safety, feasibility, and efficacy of transplanting MSCs for a variety of diseases [11]. There was no toxicities related to the infusion of expanded autologus MSCs into patients with advanced breast cancer or with haematological malignancy, into hurler syndrome patients and into patients of metachromatic leukodystrophy [12,13].

MSC could be delivered systematically and locally. Systemic delivery circumvent the problems such as tissue damage and unsuitability of delivering multiple doses. Site specific delivery has the advantage of delivering large numbers of cell directly to the required sites. Majority of studies showed that MSC infused systematically homed to injured tissues [14,15].

Successful systemic delivery of MSC is dependent upon efficient homing of the cells to the required sites. In this respect, migration of MSC from the circulation into damaged tissues leading to therapeutic effect s has been documented [16]. Ability of MSC to home to bone marrow (BM) can be affected by in vitro culture, which could be due to decrease of adhesion molecules and Chemokine receptors [17].

2.2. Mechanism of homing of MSC

Homing of leukocytes to inflammatory sites involve selectin, chemokines, integrins and other adhesion molecules [18]. Hemopoietic stem cells (HSC) recruited from blood vessels on similar process to that of leucocytes [19]. MSC recruited to damaged tissues. It is a fair assumption to suppose that they utilize comparable mechanisms of recruitment of MSC. P-selectin play an important role in the trafficking of MSC [20]. MSC roll upon endothelial cells as the first stage in their recruitment. Chemokine receptors are expressed on the cell surface of MSC [21] and their stimulation has been shown to induce cell migration and directing MSC.

2.3. Transplant ability, engraftment and tracking of MSC

Numerous studies have demonstrated migration and multiorgan engraftment of MSCs both in animal models and in human clinical trials. [22-24].

Direct injection of human marrow stromal cells into the corpus striatum of rat brain showed engraftment of 20% of the infused cells [25]. Rat bone marrow stromal cells infused into briefly distally occluded ascending aorta migrated after 8 weeks in the scar and periscar tissue [26]. MSCs injected intravenously into irradiated primate had the ability of engraftment in different injured tissues as the bone marrow, skin, digestive tract and muscle [9,11]. In rat model, MSC engrafted in multiple organs such as lung, liver, kidneys and spleen.

Human MSCs engrafted and demonstrated site-specific differentiation in sheep [27,28] and in murine models [29-31]. The capacity of engrafment of MSCs was not influenced neither by the route of administration nor by the difference in conditioning protocols [32]. Clinically, both autologous or allogeneic MSCs were given to patients [33, 34]. Allogeneic HLA-mismatched male fetal MSCs injected into HLA-incompatible female fetus with osteogenesis imperfecta (OI) engrafted and differentiated into bone [35]. Haploidentical MSCs had a low level of engrafment in a patient with aplastic anemia, however there was a partial restoration of bone marrow microenvironement [36]. In contrast, Infused allogeneic MSCs did not expand significantly in patients and they originate from the host [34, 37]. It is of interest to know into which organs MSCs home. Allogeneic and autologus MSCs distributed to a wide range of tissues in baboons [22].

2.4. Transformation of MSCs

MSCs transformed in vitro. The transformation of MSCs is associated with chromosomal abnormalities, increased levels of telomerase activity and c-myc expression [38]. Human MSCs isolated from adipose tissue undergo spontaneous transformation after long-term culture (4-5months) [39]. Others found that short-term culture was sufficient for the transformation of murine MSCs into a cell population with autonomous growth and biological characteristics of osteosarcoma [40]. In contrast, even long term cultures could not induce MSC transformation [41]. Previous study reported that gastric cancer could originate from BM-derived cells, presumably MSCs [42]. Human BM-MSCs cultured extensively, with a high Telomerase activity is capable of forming solid tumors in multiple organs in mice [43].

MSCs could migrate towards primary tumors and metastatic sites. Chemokines secreted by MSCs have been shown to enhance the emergence of pulmonary metastases and such secretion has a strong interaction between breast cancer cells and MSCs. In addition, MSCs have also been found to play a role in drug resistance in various cancer cells. MSCs protect chronic lymphocytic leukemic cells from fludarabine-, dexamethosone-, and cyclophosphamide-induced apoptosis. MSC non-selectively protected chronic myeloid leukemia cells from imatinib-induced apoptosis. [44].

Indeed, the transformation potential in cultured MSCs must be documented before considering infusion of these cells into patients. However, this issue remains controversial, as other studies did not observe transformation of human BM-MSCs.

In conclusion, it is possible that the way MSCs are expanded and long term culture lead to transformation. The safety of using MSCs in humans remains open. The use of MSCs in patients should definitely require precise and limited procedures of expansion to avoid the risk of injecting transformed cells. These finding emphasize the need for accurate studies aimed at investigating the bio safety of these cells

2.5. MSCs and metastasis

So far, very few studies have addressed the question of the effects of MSCs on metastasis. A few studies have supported that MSCs may suppress tumor growth while others believe that MSCs may contribute to tumor protection via antiapoptotic effect, tumor progression, metastasis, and drug-resistance of cancer cells.

Human MSCs played a dual role in tumor cell growth in vitro and in vivo. It was found that human MSCs inhibited the proliferation of cancer cell lines and caused G1 phase cell cycle arrest and apoptosis in vitro. However they enhance tumor formation and growth in vivo. MSCs have also been found to prevent apoptosis of acute myeloid leukemia cells by up-regulation of antiapoptotic proteins [44].

The main adverse role of MSCs was its pro-invasive potential [45]. It is likely that other molecules participate to the enhancement of metastasis by MSCs and this will be the challenge of future studies.

To use MSCs in anti-cancer therapies, it will be essential to identify the factors produced by MSCs cells responsible for the inhibition or the enhancement of tumor growth and those governing the response of tumor cells.

Evaluation of the potential use of MSCs in cell-based anti-cancer therapies is just starting. These cells have shown some promise as several studies have reported that a portion (which remains to be defined) of MSCs is able to migrate to the tumor site. However, this homing of MSCs is not selective and it will be important to evaluate possible side effects in organs, which are not affected by the disease. Overall, MSCs represent great hope for cancer therapies, but a thorough evaluation of their potential risk will be pre-required step [46].

Advertisement

3. Immunological characteristics of MSCs

Studies in animal models and in humans demonstrated that co-transplantation of HSCs along with MSCs obtained by ex-vivo expansion enhance hematopoietic reconstitution [47,48]. Experimental and clinical studies demonstrated the safety and immunosuppressive role of MSCs infusion [49, 50].

3.1. Immunological characteristics of MSCs in vivo

It has been demonstrated since 1984 that reconstitution of irradiated host with T-cell depleted bone marrow containing both host (syngeneic) and donor (allogeneic or Xenogeneic) components leads to long-term survival of the reconstituted animals and specific prolongation of subsequent skin grafts of donor type. However these animals are fully reactive to third-party allograft and do not appear to manifest signs of graft-versus-host disease (GVHD) [51]. The possibility of the presence of immunomodulatory cells in bone has been noticed after donor specific long-term hypo responsive status obtained by transplantation of bone and HSCs in murine models [52, 53]. MSCs constitute the stromal scaffold which is close to the endostenum and interact tightly with HSCs [54]. They contribute to the formation of HSCs niche, support HSCs engraftment and survival of blood cells in vivo [11]. This indicate the presence of underlying immunomodulatory effect of MSCs.

3.1.1. MSCs immunosuppressive effect in experimental animal models

Various animal models have been used to study the immunomodulatory effects of MSCs in treatment of GVHD, autoimmune disorders and tumor immunity. It seems to be that early and repeated injection of MSCs following HSCs transplantation is primordial to control GVHD [55, 56]. In autoimmunity model only early and re-injection of MSCs at the peak of the disease were effective as compared to injection after disease stabilization [57]. MSCs do not elicit immunological reaction in recipients [49, 56]. MSCs play tolerogeneic effect in recipients [58-60]. However, MSCs had been rejected by mismatched recipients [61] and failed to prevent GVHD [56].

3.1.2. MSCs immunosuppressive effect in humans

In the field of HSC transplantation, there are two applications of MSCs:

  • Improvement of stem cell engrafting and the acceleration of hematopoietic reconstitution based on the hematopoesis-supporting ability.

  • Treatment of severe GVHD based on the immunomodulatory ability.

3.1.2.1. Role of MSCs in support of hematopoiesis

MSCs constitute the functional and structural support of medullary hematopoiesis by providing growth factors and extracellular matrix [62, 63]. MSCs have a positive impact on hematopoesis and results in rapid hematopoietic recovery [64].

Co transplantation of HSCs and haplotype-mismatched MSCs to patients with high-risk acute myelogenous leukemia result in rapid engraftment [65]. Co transplantation of allogeneic MSCs and multidonor umbilical cord blood (UCB) correlated with a higher overall level of engraftment into NOD/SCID mice [66]. In European phase I-II study co-infusion of haploidentical HSCs and MSCs accelerated leukocyte recovery as compared to control group [67]. In multicentre clinical trial, co transplantation of MSCs with an HLA-identical sibling HSCs after a myeloablative conditioning regimen induced hematopoietic recovery of peripheral mono nuclear cells (MNC) and platelets [68, 69] and resulted in fast engraftment of absolute neutrophils count and 100% donor chimerism [69]. Transplantation of MSCs into immunosupressed patients generated neither alloantibody against MSC nor against fetal calf serum (FCS) [70].

3.1.2.2. Role of MSCs in management of graft versus host disease (GVHD)

Injection of MSCs could cure severe graft versus host disease (GVHD) and promote hematopoietic recovery.

MSC-mediated inhibition of immune response is a complex mechanism involving changes in the maturation of antigen-presenting cells and in cytokine secretion profiles as well as the suppression of monocyte differentiation into dendritic cells [71]. They exert profound immunosuppression by inhibiting T-cell proliferation in response to various stimuli in vitro. They induce regulatory immunosuppressive lymphocytes and CD8 apoptosis. MSCs inhibit cell cycle progression and CD4 allo-proliferation. This immunosuppressive effect of MSCs is mediated through several inducible soluble factors [71].

3.1.2.3 Clinical trials of GVHD management by MSCs

Patients treated with MSCs had less GVHD and less toxicity in a retrospective study comparing allograft of geno-identical HSCs with and without MSCs [72]. No acute side effects occurred after the infusion of haploidentical and mismatched MSCs from unrelated donors into patients suffering from GVHD [50, 73]. Haploidentical MSCs was used to treat grade IV GVHD of the gut and liver that was resistant to conventional treatment. The aim was to use the tissue repair effect shown in vivo in animal models, and the immunomodulatory effects seen in vitro on human lymphocytes. The clinical response was striking with normalization of stool and bilirubin on two occasions. They leads to healing of damaged bowel epithelia [50]. The patient was highly immunosupressed. However, his lymphocytes continued to proliferate in response to lymphocytes of MSCs donor in vitro in several occasions. This could indicate an immunosuppressive effect of MSCs rather than tolerance induction. We would be able to prevent GVHD after MSCs infusion and maintaining in the same time the response of host lymphocytes against alloantigens. Infusion of MSCs into 8 patients with steroid refractory GVHD cause dramatical disappearance of all symptoms and repaired gut in six patients and liver in one. The survival curve was better than that of 16 patients with steroid-resistant biopsy-proven gastrointestinal GVHD, not treated with MSCs [245]. MSCs were used to treat 40 patients with acute and chronic GVHD. More than forty-seven (47.5 %) showed complete response, 22.5 % showed improvement, 10 % had stable disease and 17.5 % had no response. Between 6 weeks up to 3.5 years after transplantation more than half of patients are alive [73].

At the same time of hematopoietic stem cell transplantation (HSCT), 46 patients received culture-expanded MSCs from their HLA-identical sibling donors [74]. Moderate to severe acute GVHD was observed in 13 (28%) of 46 patients. Chronic GVHD was observed in 22 (61%) of 36 patients and 2-year survival was 53%. No MSC-associated toxicities were seen. Stromal cell chimerism was demonstrated in 2 of 19 examined patients at 6 and 18 months after transplantation. MSCs are safe to give, but are difficult to detect after infusion, even in immunocompromised patients who have undergone HSCT [75].

Ten patients undergoing HSCT were treated with MSCs due to tissue toxicity. In five patients, severe hemorrhagic cystitis cleared after MSC infusion. Gross hematuria disappeared after a median of 3 days. Two patients with grade five hemorrhagic cystitis had reduced transfusion requirements after MSC infusions, but both died of multi-organ failure. MSC donor DNA was demonstrated in the urinary bladder in one of them. Two patients were treated for pneumomediastinum, which disappeared after MSC infusion. A patient with steroid-resistant GVHD of the gut experienced perforated diverticulitis and peritonitis that was dramatically reversed twice after infusion of MSCs[76].

In Europe, 55 patients have been treated for steroid-resistant acute GVHD with an overall response rate of 69%. Non responders have died of progressive GVHD and several responders have died from infections with an overall survival of 23 of 55 (42%) from 2 months to 5 years. Although the experience is limited, MSCs seems a promising treatment for severe steroid-resistant acute GVHD [73].

In a phase I/II clinical trial, ten percent of patients treated for acute refractory GVHD obtained a complete response, 60 % had a partial response and 30 % did not respond. They found that 50% of patients with chronic refractory GVHD did not respond, 12.5 % had complete remission and 37.5% had partial response [77]. Transplantation of MSCs into 15 patients with haematological malignancies was safe and induced complete remission [78].

These preliminary data suggest that MSCs may also play a role in repairing severe tissue toxicity. It seems to be that there is beneficial effect of MSCs infusion in humans. However, The optimal MSCs dose and frequency of administration needed to be evaluated to control GVHD.

The underlying molecular mechanisms of immunosuppression in vivo are unknown. It has been demonstrated that MSCs engrafted to injured tissues rich on inflammatory cytokines [107]. MSCs inhibitory effect is inducible [79] and presence of MSCs in such media could explain partially the beneficial effect of MSCs infusion in the recipients, despite non detectable MSCs in recipients BM.

3.2. Suggested mechanisms for MSC immunomodulatory effect

Co-culture of MSCs with allogeneic lymphocytes failed to stimulate their proliferation, indicating that these cells are innately not immunogeneic. Recent reports suggest that MSCs have immunomodulatory properties and can inhibit lymphocyte antigen presenting cells, natural killer cells, and cytotoxic lymphocyte proliferation in mixed-lymphocyte reactions (MLR). MSCs inhibit CD2, CD4 and CD8 subsets of T lymphocytes. The immunosuppressive effect of human MSCs was higher in cultures with cell contact than in cultures without contact (transwell), and the difference was statistically significant [80, 81]. MSCs produce a variety of growth factors that are likely to play a role in immunomodulation. Human and murine MSCs do inhibit the proliferation of lymphocytes in MLR by soluble factors [80-83]. Indoleamine 2,3 dioxygenase (IDO ) inhibit the alloreactivity induced by antigen presenting cells (APC) [84]. The inhibitory effect of MSCs on alloreactivity seems to be due to other mechanisms rather than apoptosis of lymphocytes. Several studies demonstrated reversibility of MSCs inhibitory effect. [81, 85, 86].

Cell anergy is a state of immune unresponsiveness, defines the inability of an immune cell to mount a complete response against its target. MSCs induce anergy due to divisional arrest of T cells. IFN-γ production but not proliferation of murine cells was restored [87]. Restoration of CD4+ cell proliferation but not CD8+ cells after the removal of human MSCs was observed. For the time being we could assume that MSCs do induce anergy [88]. MSCs induced regulatory cells [88]. Regulatory cells have been involved in the regulation of immune response.

Despite the expression of human leukocyte antigen (HLA) by MSCs, they were well tolerated without side effects in allogeneic hosts. Major Histocomaptability Complex (MHC) had no role in MSCs inhibitory effect as autologus and allogenic human [81], murine [89] and baboon [49]. MSCs were capable of inhibiting the proliferation of lymphocytes. In addition, Xenogeneic murine MSCs inhibited the proliferation of human cells [79]. However, one study showed that transplantation of allogeneic MSCs resulted in rejection by class I and class II mismatched recipients in murine model [61].

The major effectors of the innate immunity are natural killer (NK) cells. They eliminate malignant and virus infected cells. MSCs alter the phenotype of NK cells and suppress proliferation and cytotoxicity against HLA-class I-expressing targets in time and dose dependent manner in cell contact or transwell cultures [90, 91].

Veto activity was defined by Miller [92] as the ability to induce specific suppression of cytotoxic T lymphocyte precursors (CTL-Ps) against antigens present on veto cell surface, but not against those on third-party allogeneic cells. MSCs had veto activity [93]. This is contradictory to all the present data where MSCs were able to inhibit the allo-response to allogeneic lymphocytes in MLR in different species [79, 81]. Even more, veto property contrasts with the proliferation of allogeneic lymphocytes observed in the presence of MSCs by the same team [93].

There was drastic reduction of the recipient's CTL response against injected class I antigens due to veto phenomenon [94]. These findings still keep on with Miller’s definition [92]. Veto mechanism could play a role in low immunogenicity of MSCs, but certainly doesn’t explain the suppression of allogenic lymphocytes by MSCs [71]. MSCs have no effect on the lymphocyte response to recall antigen [93].

Human MSC inhibited the pro--inflammatory Th1 cytokines (IFN-γ, TNF- α, IL-1α and IL-β) whereas the anti-inflammatory Th2 cytokines IL-3, IL-5, IL-10, IL-4 and IL-13 and the Th2 Chemokine I-309 (a chemo attractant for regulatory T cells) were increased [71, 95]. This could indicate a shift from the prominence of pro-inflammatory Th1 cells towards an increase in anti-inflammatory TH2 cells and support MSC inhibitory properties. MSCs can skew the dendritic cells (DCs) function, thus biasing the immune system toward Th2 and away from Th1 responses [96]. MSC had no effect on neutrophils phagocytosis, expression of adhesion molecules and chemotaxis [97]. This is an interesting finding, as MSCs could not interfere with neutrophils function. Summary of MSCs inhibitory effect is illustrated in figure-1. For further reading, about detailed description of possible MSCs inhibitory mechanisms, refer to this article [10].

3.2.1. Inhibitory effect of autogenic and allogenic MSCs

Allogeneic MSCs have stronger immunosuppressive effects than autologus MSCs [88]. Others found comparable and significant inhibition was elicited by autologus or allogeneic MSCs [81,85]. This could help in management of GVHD in cases where non-matched HSCs are used

Advertisement

4. What starts the first step in induction of inhibitory effect in MSC/Mixed lymphocyte reaction MLR?

It is not known for the time being whether MSCs or lymphocytes do start the first signals to activate the other one to induce the immunosuppressive effect. MSC supernatant failed to inhibit T-cell activation and, on the contrary, MSC supernatant had a stimulatory effect on MLR. Surprisingly, cell-free supernatant obtained from MLR had an inhibitory effect similar to that seen with the addition of MSCs [58]. In agreement Djouad et al. found that only MSC conditioned supernatant by lymphocytes was capable to inhibit secondary MLR but less efficiently than the co-culture of cell partners, in indicating that the immunosuppressive properties of MSCs are mediated by inducible soluble factors and the interaction between the MSCs and lymphocytes is a pre-requirement for MSC mediated inhibitory effect [79].

In addition, an enhancement of the MSC immunosuppressive effect was observed after addition of irradiated third party MNC to MSC culture, indicating that the physical interaction between MSC and PBMC increase the suppressive activity [98]. Human MSCs do not constitutively express IDO. But activation by IFN-γ secreted by allogeneic lymphocytes in a dose dependent manner is required for induction of functional IDO activity [99].

It has been demonstrated that auto reactive cells may induce the transdifferentiation of MSCs to neural stem cells. This phenomenon could be due to stimulation of cytokine production and generation of suitable environment that results in differentiation into neural stem cells [100]. All these data could confirm the interaction between MSCs and lymphocytes. Physical interaction between MSCs and T-cells increases the suppressive activity as there was an increased expression of IL-10 and TGF-β genes [80] as compared to non contact cultures. All these data could complicate the issue. Is MSC mediated inhibitory effect is a consequence of MSC activation by IFN-γ secreted by two allogeneic populations of lymphocytes in MLR?. Why there is no stimulation of single allogeneic PBL by allogeneic MSCs?

Are there two separate effector mechanisms, one to escape the immunological recognition and the other to inhibit the alloreactivity? Is the constitutive expression of TGF-β and HLA-G by the MSCs play a role in immunological escape?

Maccario et al. had demonstrated the absence of regulatory cell in co-culture of MSCs with single allogeneic PBMCs as compared to the presence of MSCs in MLR [88]. This could indicate that allogeneic lymphocytes initiate the first signals to stimulate the production of molecules that induce the production of regulatory cells.

It has been found that CD14+ monocyte are the PBMC subpopulation, being responsible for MSC activation through IL-B secretion [101]. MSC inhibitory effect was mediated through CD8+ in human and murine model [97, 88]. CD8+ cells are the executive cells for MSC inhibitory effect rather than being the inducers of MSC inhibitory effect. It has been demonstrated that MSC inhibitory mechanisms differ depending on nature of stimulus [102]. This could indicate that alloantigens or mitogen are responsible for the first stimulating signals in MLR, or at least they are responsible for later on modification of these signals. It has been found that lymphocytes and MSCs are mutually inhibitory on their respective proliferation and indicate the bi-directional interaction and cross talk between lymphocytes and MSCs [103, 104]. More recently, it has been found that IFNγ and concomitant presence of TNFα or IL-1α or IL-1β induce the expression of several chemokine and inducible Nitric oxide synthase by MSCs [105].

This could indicate that lymphocytes start the first signals for MSCs activation, however cross talk between both of them are essential to have the full immunological effects.

In conclusion, MSC must be handled with extreme caution before a large scale clinical trial is performed. it has been found in a pilot clinical study, that co transplantation of MSC and HSC prevent GVHD, but caused a higher relapse rate in hematologic malignancy patients as compared to control [106].

Figure 1.

Mechanisms of action of MSCs upon dendrietic, lymphocytes T, lymphocytes B and natural killer cells.

MSCs inhibit monocytes (MO) differentiation into immature DCs (IDC) and skewed them toward macrophage. MSCs reduced percentage of CD34(+) derived IDC and increased plasmacytoid DC (pDC). They inhibit maturation IDC into mature DCs. They inhibit the regulation of HLA-DR, CD80, CD86, CD1a, CD40, CD14 and CD83 antigen on DCs surface through IL-6, M-CSF, PGE2, IL-10 and TGF- b secretion. MSCs increased production of IL-10 and decreased production of Il-12, TNF-a and INF-g by DCs. MSCs induced DC that exhibit a suppressor phenotype (supp APC) and generated alloantigen-specific regulatory cells (Reg cells). MSCs inhibit NK proliferation and change cytokines secretion pattern through IDO, TGF-b, PGE2. MSCs inhibit lymphocytes B Proliferation & differentiation to plasma cells through IDO. MSCs inhibit lymphocytes T proliferation and cytotoxicity through IDO, PGE2, HLA-G, TGF-b, HGF, Il-10 and IL-1 b.

References

  1. 1. A. JFriedensteinR. KChailakhyanN. VLatsinikA. FPanasyukand I. VKeiliss-borokStromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation 171974197433140
  2. 2. A. JFriedensteinPrecursor cells of mechanocytes. Int Rev Cytol 471976197632759
  3. 3. A. JFriedensteinK. VPetrakovaA. IKurolesovaand G. PFrolovaHeterotopic of bone marrow.Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 61968196823047
  4. 4. A. JFriedensteinR. KChailakhyanand U. VGerasimovBone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet 201987198726372
  5. 5. MOwenand A. JFriedensteinStromal stem cells: marrow-derived osteogenic precursors. Ciba Found Symp 136198819884260
  6. 6. D. JProckopMarrow stromal cells as stem cells for nonhematopoietic tissues. Science 27619971997714
  7. 7. E. MHorwitzKLeBlancMDominiciIMuellerISlaper-cortenbachF. CMariniR. JDeansD. SKrause, and A. Keating, Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 7200520053935
  8. 8. MDominiciKLeBlancIMuellerISlaper-cortenbachFMariniDKrauseRDeansAKeatingDProckop, and E. Horwitz, Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8200620063157
  9. 9. BDelormeand PCharbordCulture and characterization of human bone marrow mesenchymal stem cells. Methods Mol Med 140200720076781
  10. 10. H. MLazarusO. NKocS. MDevinePCurtinR. TMaziarzH. KHollandE. JShpallPMccarthyKAtkinsonB. WCooperS. LGersonM. JLaughlinF. RLoberizaJrA. BMoseleyand ABacigalupo, Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol Blood Marrow Transplant 112005200538998
  11. 11. ANasefNAshammakhiLFouillardImmunomodulatory Effect of Mesenchymal Stem Cells: Possible mechanisms. Regenerative medicine (2008Review article..
  12. 12. O. NKocS. LGersonB. WCooperS. MDyhouseS. EHaynesworthA. ICaplanand H. MLazarusRapid 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 182000200030716
  13. 13. O. NKocJDayMNiederS. LGersonH. MLazarusand WKrivitAllogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH). Bone Marrow Transplant 302002200221522
  14. 14. RRamasamyE. WLamISoeiroVTisatoDBonnetand FDazziMesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth. Leukemia 212007200730410
  15. 15. SFrancoisMBensidhoumMMouiseddineCMazurierBAllenetASemontJFrickASacheSBouchetDThierryPGourmelonN. CGorinand AChapelLocal irradiation not only induces homing of human mesenchymal stem cells at exposed sites but promotes their widespread engraftment to multiple organs: a study of their quantitative distribution after irradiation damage. Stem Cells 242006200610209
  16. 16. JChenYLiLWangMLuXZhangand MChoppTherapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci 189200120014957
  17. 17. W. JRomboutsand R. EPloemacherPrimary murine MSC show highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia 172003200316070
  18. 18. T. ASpringerTraffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 761994199430114
  19. 19. J. PChuteStem cell homing. Curr Opin Hematol 1320062006399406
  20. 20. BRusterSGottigR. JLudwigRBistrianSMullerESeifriedJGilleand RHenschlerMesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood 10820062006393844
  21. 21. J. MFoxGChamberlainB. AAshtonand JMiddletonRecent advances into the understanding of mesenchymal stem cell trafficking. Br J Haematol 13720072007491502
  22. 22. S. MDevineCCobbsMJenningsABartholomewand RHoffmanMesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 1012003200329993001
  23. 23. YLiHHishaMInabaZLianCYuMKawamuraYYamamotoNNishioJTokiHFanand SIkeharaEvidence for migration of donor bone marrow stromal cells into recipient thymus after bone marrow transplantation plus bone grafts: A role of stromal cells in positive selection. Exp Hematol 282000200095060
  24. 24. TSaitoJ. QKuangC. CLinand R. CChiuTranscoronary implantation of bone marrow stromal cells ameliorates cardiac function after myocardial infarction. J Thorac Cardiovasc Surg 1262003200311423
  25. 25. D. GPhinneyMBaddooMDutreilDGauppW. TLaiand I. AIsakovaMurine mesenchymal stem cells transplanted to the central nervous system of neonatal versus adult mice exhibit distinct engraftment kinetics and express receptors that guide neuronal cell migration. Stem Cells Dev 152006200643747
  26. 26. YLiHHishaMInabaZLianCYuMKawamuraYYamamotoNNishioJTokiHFanand SIkeharaEvidence for migration of donor bone marrow stromal cells into recipient thymus after bone marrow transplantation plus bone grafts: A role of stromal cells in positive selection. Exp Hematol 282000200095060
  27. 27. K.W. Liechty, T.C. MacKenzie, A.F. Shaaban, A. Radu, A.M. Moseley, R. Deans, D.R. Marshak, and A.W. Flake, Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med 6 (2000) 1282-6.
  28. 28. J. AAireyGAlmeida-poradaE. JCollettiC. DPoradaJChamberlainMMovsesianJ. LSutkoand E. DZanjaniHuman mesenchymal stem cells form Purkinje fibers in fetal sheep heart. Circulation 1092004200414017
  29. 29. M. BHerreraBBussolatiSBrunoVFonsatoG. MRomanazziand GCamussiMesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury. Int J Mol Med 1420042004103541
  30. 30. L. AOrtizFGambelliCMcbrideDGauppMBaddooNKaminskiand D. GPhinneyMesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A 10020032003840711
  31. 31. A. AMangiNNoiseuxDKongHHeMRezvaniJ. SIngwalland V. JDzauMesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med 9200320031195201
  32. 32. NMahmudWPangCCobbsPAlurJBornemanRDoddsMArchambaultSDevineJTurianABartholomewPVanguriAMackayRYoungand RHoffmanStudies of the route of administration and role of conditioning with radiation on unrelated allogeneic mismatched mesenchymal stem cell engraftment in a nonhuman primate model. Exp Hematol 3220042004494501
  33. 33. H. MLazarusS. EHaynesworthS. LGersonN. SRosenthaland A. ICaplanEx vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): implications for therapeutic use. Bone Marrow Transplant 161995199555764
  34. 34. JWangKLiuand D. PLuMesenchymal stem cells in stem cell transplant recipients are damaged and remain of host origin. Int J Hematol 82200520051528
  35. 35. KLeBlancCGotherstromORingdenMHassanRMcmahonEHorwitzGAnnerenOAxelssonJNunnUEwaldSNorden-lindebergMJanssonADaltonEAstrom, and M. Westgren, Fetal mesenchymal stem-cell engraftment in bone after in utero transplantation in a patient with severe osteogenesis imperfecta. Transplantation 7920052005160714
  36. 36. LFouillardMBensidhoumDBoriesHBonteMLopezA. MMoseleyASmithSLesageFBeaujeanDThierryPGourmelonANajmanand N. CGorinEngraftment of allogeneic mesenchymal stem cells in the bone marrow of a patient with severe idiopathic aplastic anemia improves stroma. Leukemia 17200320034746
  37. 37. ADickhutRSchwerdtfegerLKuklickMRitterCThiedeANeubauerand CBrendelMesenchymal stem cells obtained after bone marrow transplantation or peripheral blood stem cell transplantation originate from host tissue. Ann Hematol 84200520057227
  38. 38. DRubioSGarciaM. FPazTDe La CuevaL. ALopez-fernandezA. CLloydJGarcia-castroand ABernadMolecular characterization of spontaneous mesenchymal stem cell transformation. PLoS ONE 3 (2008e1398.
  39. 39. DRubioJGarcia-castroM. CMartinRDe La FuenteJ. CCigudosaA. CLloydand ABernadSpontaneous human adult stem cell transformation. Cancer Res 652005200530359
  40. 40. JTolarA. JNautaM. JOsbornAPanoskaltsisMortariR. TMcelmurrySBellLXiaNZhouMRiddleT. MSchroederJ. JWestendorfR. SMcivorP. CHogendoornKSzuhaiLOsethBHirschS. RYantM. AKayAPeisterD. JProckopW. EFibbe, and B.R. Blazar, Sarcoma derived from cultured mesenchymal stem cells. Stem Cells 25200720073719
  41. 41. M. EBernardoNZaffaroniFNovaraA. MCometaM. AAvanziniAMorettaDMontagnaRMaccarioRVillaM. GDaidoneOZuffardiand FLocatelliHuman bone marrow derived mesenchymal stem cells do not undergo transformation after long-term in vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res 672007200791429
  42. 42. JHoughtonCStoicovSNomuraA. BRogersJCarlsonHLiXCaiJ. GFoxJ. RGoldenringand T. CWangGastric cancer originating from bone marrow-derived cells. Science 30620042004156871
  43. 43. MMiuraYMiuraH. MPadilla-nashA. AMolinoloBFuVPatelB. MSeoWSonoyamaJ. JZhengC. CBakerWChenTRiedand SShiAccumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 24200620061095103
  44. 44. RebeccaS. YWong. Review Articl. Mesenchymal StemCells: Angels or Demons? Journal of Biomedicine and Biotechnology. Volume, Article ID 459510, 8 pages
  45. 45. A. EKarnoubA. BDashA. PVoASullivanM. WBrooksG. WBellA. LRichardsonKPolyakRTuboand R. AWeinbergMesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 4492007200755763
  46. 46. GLazennecand CJorgensenAdult Multipotent Stromal Cells and Cancer: Risk or Benefit? Stem Cells (2008
  47. 47. W. ANoortA. BKruisselbrinkP. SIntAnkerMKrugerR. LVan BezooijenR. ADe PausM. HHeemskerkC. WLowikJ. HFalkenburgRWillemzeand W. EFibbeMesenchymal stem cells promote engraftment of human umbilical cord blood-derived CD34(+) cells in NOD/SCID mice. Exp Hematol 30200220028708
  48. 48. P. Anklesaria, K. Kase, J. Glowacki, C.A. Holland, M.A. Sakakeeny, J.A. Wright, T.J. FitzGerald, C.Y. Lee, and J.S. Greenberger, Engraftment of a clonal bone marrow stromal cell line in vivo stimulates hematopoietic recovery from total body irradiation. Proc Natl Acad Sci U S A 84 (1987) 7681-5.
  49. 49. ABartholomewCSturgeonMSiatskasKFerrerKMcintoshSPatilWHardySDevineDUckerRDeansAMoseleyand RHoffmanMesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 3020022002428
  50. 50. KLeBlancIRasmussonBSundbergCGotherstromMHassanMUzunel, and O. Ringden, Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 36320042004143941
  51. 51. S. TIldstadand D. HSachsReconstitution with syngeneic plus allogeneic or xenogeneic bone marrow leads to specific acceptance of allografts or xenografts. Nature 3071984198416870
  52. 52. OGurevitchT. BPrigozhinaTPugatschand SSlavinTransplantation of allogeneic or xenogeneic bone marrow within the donor stromal microenvironment. Transplantation 681999199913628
  53. 53. A. WBingamanS. YWaitzeD. ZAlexanderH. RChoALinCTucker-burdenS. RCowanT. CPearsonand C. PLarsenTransplantation of the bone marrow microenvironment leads to hematopoietic chimerism without cytoreductive conditioning. Transplantation 692000200024916
  54. 54. YSakaguchiISekiyaKYagishitaSIchinoseKShinomiyaand TMunetaSuspended cells from trabecular bone by collagenase digestion become virtually identical to mesenchymal stem cells obtained from marrow aspirates. Blood 10420042004272835
  55. 55. RYanezM. LLamanaJGarcia-castroIColmeneroMRamirezand J. ABuerenAdipose tissue-derived mesenchymal stem cells have in vivo immunosuppressive properties applicable for the control of the graft-versus-host disease. Stem Cells 2420062006258291
  56. 56. MSudresFNorolATrenadoSGregoireFCharlotteBLevacherJ. JLatailladePBourinXHolyJ. PVernantDKlatzmannand J. LCohenBone marrow mesenchymal stem cells suppress lymphocyte proliferation in vitro but fail to prevent graft-versus-host disease in mice. J Immunol 1762006200677617
  57. 57. FDjouadVFritzFApparaillyPLouis-plenceCBonyJSanyCJorgensenand DNoelReversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor alpha in collagen-induced arthritis. Arthritis Rheum 52200520051595603
  58. 58. BMaitraESzekelyKGjiniM. JLaughlinJDennisS. EHaynesworthand O. NKocHuman mesenchymal stem cells support unrelated donor hematopoietic stem cells and suppress T-cell activation. Bone Marrow Transplant 3320042004597604
  59. 59. N. GChungD. CJeongS. JParkB. OChoiBChoH. KKimC. SChunJ. HWonand C. WHanCotransplantation of marrow stromal cells may prevent lethal graft-versus-host disease in major histocompatibility complex mismatched murine hematopoietic stem cell transplantation. Int J Hematol 80200420043706
  60. 60. GXuLZhangGRenZYuanYZhangR. CZhaoand YShiImmunosuppressive properties of cloned bone marrow mesenchymal stem cells. Cell Res 17200720072408
  61. 61. NEliopoulosJStaggLLejeuneSPommeyand JGalipeauAllogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood 10620052005405765
  62. 62. C. MVerfaillieAdhesion receptors as regulators of the hematopoietic process. Blood 9219981998260912
  63. 63. A. GArroyoJ. TYangHRayburnand R. OHynesAlpha4 integrins regulate the proliferation/differentiation balance of multilineage hematopoietic progenitors in vivo. Immunity 111999199955566
  64. 64. O. NKocS. LGersonB. WCooperS. MDyhouseS. EHaynesworthA. ICaplanand H. MLazarusRapid 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 182000200030716
  65. 65. S. TLeeJ. HJangJ. WCheongJ. SKimH. YMaemgJ. SHahnY. WKoand Y. HMinTreatment 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 11820022002112831
  66. 66. D. WKimY. JChungT. GKimY. LKimand I. HOhCotransplantation of third-party mesenchymal stromal cells can alleviate single-donor predominance and increase engraftment from double cord transplantation. Blood 1032004200419418
  67. 67. BallL. MBernardoM. ERoelofsHLankesterACometaAEgelerMGiorgianiGLocatelliFand F. W. ECo-transplantationof HLA-Haploidentical, Bone Marrow Derived Mesenchymal Stem Cells Prevents Graft Failure and Improves Hematological Recovery in T-Cell Depleted Haploidentical Stem Cell Transplantation. Blood 108 (2006
  68. 68. LFouillardAChapelDBoriesSBouchetJ. MCostaHRouardPHervePGourmelonDThierryMLopezand N. CGorinInfusion of allogeneic-related HLA mismatched mesenchymal stem cells for the treatment of incomplete engraftment following autologous haematopoietic stem cell transplantation. Leukemia (2007
  69. 69. KLeBlancHSamuelssonBGustafssonMRembergerBSundbergJArvidsonPLjungmanHLonniesSNava, and O. Ringden, Transplantation of mesenchymal stem cells to enhance engraftment of hematopoietic stem cells. Leukemia 212007200717338
  70. 70. MSundinORingdenBSundbergSNavaCGotherstromand K. Le Blanc, No alloantibodies against mesenchymal stromal cells, but presence of anti-fetal calf serum antibodies, after transplantation in allogeneic hematopoietic stem cell recipients. Haematologica 9220072007120815
  71. 71. SAggarwaland M. FPittengerHuman mesenchymal stem cells modulate allogeneic immune cell responses. Blood 10520052005181522
  72. 72. l. MFrassoniFBacigalupoA, et al., Expanded mesenchymql stem cells (MSC) co-infused with HLA-identical hematopoietic stem cells transplants, reduce acute and chronic graft-vs-host disease: a matched pair analysis.. Bone Marrow Transplant 29 (2002
  73. 73. Le Blanc KFrassoni F, Ball LM, Lanino E, Sundberg B, Lonnies L, Roelofs H, Dini G, Bacigalupo A, Locatelli F, Fibbe WF, and O. Ringden, Mesenchymal Stem Cells for Treatment of Severe Acute Graft-Versus-Host Disease.. Blood 108 (2006
  74. 74. H. MLazarusO. NKocS. MDevinePCurtinR. TMaziarzH. KHollandE. JShpallPMccarthyKAtkinsonB. WCooperS. LGersonM. JLaughlinF. RLoberizaJrA. BMoseleyand ABacigalupo, Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol Blood Marrow Transplant 112005200538998
  75. 75. ORingdenMUzunelIRasmussonMRembergerBSundbergHLonniesH. UMarschallADlugoszASzakosZHassanBOmazicJAschanLBarkholtand K. Le Blanc, Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation 812006200613907
  76. 76. ORingdenMUzunelBSundbergLLonniesSNavaJGustafssonLHenningsohnand K. Le Blanc, Tissue repair using allogeneic mesenchymal stem cells for hemorrhagic cystitis, pneumomediastinum and perforated colon. Leukemia 212007200722716
  77. 77. Pérez-simonJ. ALópez-villarOAndreuE. JRifónJMuntionSCampeloM. DSánchez-guijoF. MMartinezCValcarcelDand del zo C. Mesenchymal stem cellsexpanded in vitro with human serum for the treatment of acuteand chronic graft-versus-host disease: results phase I/II clinical trial. Haematologica 2011960710721076
  78. 78. H. MLazarusS. EHaynesworthS. LGersonN. SRosenthaland A. ICaplanEx vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): implications for therapeutic use. Bone Marrow Transplant 161995199555764
  79. 79. FDjouadPPlenceCBonyPTropelFApparaillyJSanyDNoeland CJorgensenImmunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood 10220032003383744
  80. 80. ANasefAChapelCMazurierSBouchetMLopezNMathieuLSensebeYZhangN. CGorinDThierryand LFouillardIdentification of IL-10 and TGF-beta transcripts involved in the inhibition of T-lymphocyte proliferation during cell contact with human mesenchymal stem cells. Gene Expr 132007200721726
  81. 81. MDiNicolaCCarlo-stellaMMagniMMilanesiP. DLongoniPMatteucciSGrisanti, and A.M. Gianni, Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 9920022002383843
  82. 82. A. ICaplanand J. EDennisMesenchymal stem cells as trophic mediators. J Cell Biochem 9820062006107684
  83. 83. D. HKimK. HYooK. SChoiJChoiS. YChoiS. EYangY. SYangH. JImK. HKimH. LJungK. WSungand H. HKooGene expression profile of cytokine and growth factor during differentiation of bone marrow-derived mesenchymal stem cell. Cytokine 312005200511926
  84. 84. N. KSteckelUKuhnD. WBeelenand A. HElmaagacliIndoleamine 2,3-dioxygenase expression in patients with acute graft-versus-host disease after allogeneic stem cell transplantation and in pregnant women: association with the induction of allogeneic immune tolerance? Scand J Immunol 572003200318591
  85. 85. KLeBlancLTammikBSundbergS. EHaynesworth, and O. Ringden, Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand J Immunol 57200320031120
  86. 86. JPlumasLChaperotM. JRichardJ. PMolensJ. CBensaand M. CFavrotMesenchymal stem cells induce apoptosis of activated T cells. Leukemia 19200520051597604
  87. 87. SGlennieISoeiroP. JDysonE. WLamand FDazziBone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 1052005200528217
  88. 88. RMaccarioMPodestaAMorettaACometaPComoliDMontagnaLDaudtAIbaticiGPiaggioSPozziFFrassoniand FLocatelliInteraction 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 902005200551625
  89. 89. MKramperaSGlennieJDysonDScottRLaylorESimpsonand FDazziBone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood 1012003200337229
  90. 90. DAngoulvantAClercSBenchalalCGalambrunAFarreYBertrandand AEljaafariHuman mesenchymal stem cells suppress induction of cytotoxic response to alloantigens. Biorheology 412004200446976
  91. 91. P. ASotiropoulouS. APerezA. DGritzapisC. NBaxevanisand MPapamichailInteractions between human mesenchymal stem cells and natural killer cells. Stem Cells 24200620067485
  92. 92. R. GMillerSMuraokaM. HClaessonJReimannand PBenvenisteThe veto phenomenon in T-cell regulation. Ann N Y Acad Sci 532198819881706
  93. 93. J. APotianHAvivN. MPonzioJ. SHarrisonand PRameshwarVeto-like activity of mesenchymal stem cells: functional discrimination between cellular responses to alloantigens and recall antigens. J Immunol 17120032003342634
  94. 94. H. GRammenseeP. JFinkand M. JBevanFunctional clonal deletion of class I-specific cytotoxic T lymphocytes by veto cells that express antigen. J Immunol 1331984198423906
  95. 95. PBattenPSarathchandraJ. WAntoniwS. STayM. WLowdellP. MTaylorand M. HYacoubHuman mesenchymal stem cells induce T cell anergy and downregulate T cell allo-responses via the TH2 pathway: relevance to tissue engineering human heart valves. Tissue Eng 1220062006226373
  96. 96. LChenWZhangHYueQHanBChenMShiJLiBLiSYouYShiand R. CZhaoEffects of human mesenchymal stem cells on the differentiation of dendritic cells from CD34+ cells. Stem Cells Dev 162007200771931
  97. 97. LRaffaghelloGBianchiMBertolottoFMontecuccoABuscaFDallegriLOttonelloand VPistoiaHuman mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells 262008200815162
  98. 98. W. TTseJ. DPendletonW. MBeyerM. CEgalkaand E. CGuinanSuppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation 752003200338997
  99. 99. RMeiselAZibertMLaryeaUGobelWDaubenerand DDillooHuman bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood 10320042004461921
  100. 100. G. AMovigliaGVarelaC. AGaetaJ. ABrizuelaFBastosand JSaslavskyAutoreactive T cells induce in vitro BM mesenchymal stem cell transdifferentiation to neural stem cells. Cytotherapy 820062006196201
  101. 101. M. EGrohBMaitraESzekelyand O. NKocHuman mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells. Exp Hematol 332005200592834
  102. 102. IRasmussonORingdenBSundbergand K. Le Blanc, Mesenchymal stem cells inhibit lymphocyte proliferation by mitogens and alloantigens by different mechanisms. Exp Cell Res 305200520053341
  103. 103. APoggiCPrevostoA. MMassaroSNegriniSUrbaniIPierriRSaccardiMGobbiand M. RZocchiInteraction between human NK cells and bone marrow stromal cells induces NK cell triggering: role of NKp30 and NKG2D receptors. J Immunol 17520052005635260
  104. 104. G. MSpaggiariACapobiancoHAbdelrazikFBecchettiM. CMingariand LMorettaMesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 11120082008132733
  105. 105. GRenLZhangXZhaoGXuYZhangA. IRobertsR. CZhaoand YShiMesenchymal Stem Cell-Mediated Immunosuppression Occurs via Concerted Action of Chemokines and Nitric Oxide. Cell Stem Cell 220082008141150
  106. 106. HNingFYangMJiangLHuKFengJZhangzYuBLiCXuYLiJWangJHuXLouand HChenThe correlation between cotransplantation of mesenchymal stem cells and higher recurrence rate in hematologic malignancy patients: outcome of apilot clinical study. Leukemia 22 (3) (2008

Written By

Aisha Nasef

Submitted: 18 May 2012 Published: 08 May 2013