Abstract
Umbilical cord blood (UCB) is a source of primitive hematopoietic stem (HSC) and progenitor cells, that served as an alternative to bone marrow (BM) for effective transplantation therapy. Success of HSC transplantation (HSCT) is limited in part by graft-versus-host disease (GVHD), graft rejection and delayed immune reconstitution, which all relate to immunological complications. GVHD after UCB transplantation is lower compared to that of BM HSCT. This may relate to the tolerogenic nature of T cells, mononuclear cells (MNCs) and especially immune regulatory cells existing in UCB. UCB contains limiting numbers of HSC or CD34+ cell dose for adult patients resulting in delayed engraftment after UCB transplantation (UCBT). This needs to be improved for optimal transplantation outcomes. Approaches have been undertaken to promote HSC engraftment, including co-infusion of multiple units of UCB cells. These new methods however added additional immunological complications. Herein, we describe current knowledge on features of UCB immune cells, including regulatory T cells (Tregs) and mesenchymal stem/stromal cells (MSCs) and their potential future usage to reduce GVHD.
Keywords: Cord blood Transplantation, Transplantation Tolerance, T-lymphocytes, Dendritic Cells, Regulatory T cells, Mesenchymal Stromal/Stem Cells, Natural Killer Cells
1. Introduction
UCB offers an effective transplantation therapy to reconstitute the hematopoietic system in patients with malignant and non-malignant disorders. UCB HSCs cryopreserved for many years are functional [1] and ready to use. In general, UCB immune cells are less aggressive against allogeneic responses mainly due to their immaturity. In addition, UCB contains regulatory T cells (Tregs) and mesenchymal stem cells (MSCs) with potent functions for suppressing allogeneic immune responses. It is likely that such UCB immune regulatory cells contribute to less frequent GVHD in patients transplanted with UCB compared to the patients transplanted with BM. Conversely, UCBT often bears the disadvantage of insufficient HSC numbers in UCB for timely immune reconstitution particularly in adult patients and development of antigen-specific cellular immunity is affected by numerical and qualitative deficits of UCB T cells [2-4]. As a result, infections that afflict transplant patients occur frequently within the first 3-4 months after UCBT [5-6]. To overcome this limiting numbers of cells, double-unit UCBT has been used, but with additional immunological concerns [7].
This review focuses mainly on characteristics of immune regulatory cells, Tregs and MSCs in UCB, their potential modulating roles in GVHD and immune reconstitution, particularly after double-unit UCB transplantation for adult patients.
2. Umbilical Cord Blood Hematopoietic Stem Cell Transplantation
The first UBC-HSCT from an HLA-identical sibling was performed for a 6-year-old boy with Fanconi anemia over 20 years ago [8-9]. The first clinical efforts were based on laborattory studies suggesting the presence of HSCs in UCB [10]. The ability of UCB to reconstitute hematopoiesis after myeloablative conditioning was soon proven in large number of patients, being facilitated by numerous UCB banks worldwide. Prospective, multi-center studies of unrelated UCB-HSCT for pediatric patients clearly demonstrated the efficacy of UCB-HSCT [11].
2.1 UCB-HSCT allows a higher degree of HLA disparity
GVHD is a major cause of morbidity and mortality after allogeneic HSCT and donor lymphocyte infusions [7]. The main risk factor for development of acute GVHD is human leukocyte antigen (HLA) disparity of donor T cells with reactivity against recipient histocompatibility antigens [7]. The number of mismatched HLA class I antigens correlates with instance of GVHD and poor engraftment kinetics [12]. UCBT is characterized by lower incidence of acute and chronic GVHD compared to BMT [13-15]. In a matched-pair analysis, GVHD and survival of patients receiving unrelated UCB-HSCT with up to 3 HLA mismatches were comparable to those receiving HLA-matched unrelated donors-HSCT with 6/6 HLA matches [13,16]. Mechanisms of reduced acute GVHD in patients receiving unrelated donor UCB still remain poorly understood. T cells are primary cells responsible for GVHD. T cells from UCB have greater immune tolerance to HLA mismatch [13], and impaired allogeneic activation [17-18]. It is generally accepted that immaturity of UCB T cells is a major reason for permitting use of an HLA mismatched UCB unit as a graft for transplantation. Following in vitro priming with alloantigen, cytotoxicity of CD8+ T cells from UCB is significantly diminished compared to that from adult peripheral blood (APB) and BM, suggesting that CD8+ T cell cytotoxic defect may, in part, be related to the low incidence of GVHD thus far noted in human CB transplants [19-20]. Reduced incidence and severity of GVHD after allogeneic UCBT may also be due to a defect in activation of specific transcription factors and an impaired production of certain cytokines, although this has not been extensively proven. More recent studies indicate that immune regulatory cells, such as Tregs could be dominant factors in control of GVHD [21]. In this section, we describe the characteristic properties of UCB T cells and antigen presenting cells and their therapeutic implications for GVHD and graft rejection.
2.2. Defective Th1 responses of UCB T cells
In contrast to adult PB, UCB T cells are largely naïve (CD45RA+). Thus, the primary reason for CB T cells being immunologically less responsive to alloantigens is an overall prevalent immaturity of immune cells [22]. This may be due to relatively low exposure of the fetus to environmental pathogens and vaccines as compared to the adult. As a result, T cells in UCB are activated significantly less readily than APB T cells in allogeneic response, producing less interferon-γ (IFN-γ) compared with APB CD4+ T cells after allogeneic stimulation [18]. Donor CD4+ T cell interactions with host antigen presenting cells (APCs) lead to activation of donor T cells and their differentiation into T helper (Th) cells. However, the role of Th1, Th2, and Th17 cells in acute GVHD pathogenesis is still controversial. In general, Th1 cells play a critical role in mediating acute GVHD pathogenesis [23-24], and Th2 cells ameliorate GVHD mediated by Th1 cells [25-26]. Defective type Th1 immune responses by UCB T cells in comparison with APB T cells may be attributed to impaired expression and activation of transcription factors [27]. Expression of T-bet, a Th1-specific transcription factor, is low in UCB compared to APB T cells, whereas expression of GATA-3, a transcription factor correlating with expression of Th2 cytokines, is similar. Freshly purified UCB naive CD4+ T cells show markedly lower expression of STAT4 at mRNA and protein levels than APB naive CD4+ T cells. Since STAT4 plays a critical role in Th1 differentiation [28], low STAT4 expression might contribute to reduced IFN-γ production by UCB CD4+ T cells after allogeneic stimulation. UCB naive CD4+ T cells demonstrate substantially decreased expression of tyrosine phosphorylated-STAT4, an activated form of STAT4, after IL-12 treatment compared with similarly IL-12-treated APB naive CD4+ T cells [18]. Transcription factors involved in the nuclear factor of activated T-cells (NFAT) pathway, including C/EBPβ and AP-1 are expressed at reduced levels in UCB compared with APB during primary stimulation. Reduced global expression of NFAT-associated genes, as well as Th1-related transcription factors in UCB CD4+ T cells may contribute to the decreased GVHD observed after UCB transplantation [29].
Unexpectedly, donor T cells deficient in IFN-γ, a Th1 cytokine, induce exacerbated acute GVHD [30-31]. It was proposed that tissue damage mediated by IFN-γ−/− donor T cells was associated with the lack of an IFN-γ inducible protective mechanism [32]. More recently, the protective role of IFN-γ was identified. IFN-γ induces expression of a coinhibitory molecule, B7-H1 on host tissues [23]. B7-H1 plays an important role in inducing anergy and apoptosis of allogeneic T cells through programmed death-1 (PD-1) and contributes to protect tissue damage in recipients [33]. Th17 cells were shown to augment GVHD [34-35]. In contrast, it was shown that Th17 cells ameliorate acute GVHD in allogeneic recipients, and neutralizing IL-17 augments acute GVHD [24]. These seemingly contradictory reports remain to be resolved.
There are also conflicting reports regarding the ability of UCB T cells to produce IL-4, a Th2 cytokine. A majority of reports demonstrate that UCB T cells are more prevalent for skewing Th2 cytokine production compared to APB T cells in response to allogeneic stimulation. However, some investigators reported that UCB T cells are not different from ABL CD4+ T cells in their ability to produce IL-4 in vitro [18]. IL-10 is an important anti-inflammatory cytokine and IL-10 produced by donor T cells has been known to reduce severity of acute GVHD [36] and to promote graft tolerance [37]. In contrast to tumor necrosis factor-α (TNF-α) and IFN-γ, IL-10 is readily produced by UCB T cells during T cell receptor (TCR) stimulation, while IL-10 is secreted by adult T cells only after repeated stimulation. Thus, the potential of UCB T cells to secrete IL-10 in abundance may contribute to reduced incidence of GVHD associated with UCBT [38], possibly through production of Treg cell populations [37].
2.3. Impaired activity of UCB-derived dendritic cells (DCs) in allogeneic T cell responses
Although the allogeneic response of donor T cells is associated with GVHD after HSCT, the antigen-presenting function of donor DCs may be also involved in chronic GVHD [39-40]. It has been speculated that the antigen presenting activity of DCs derived from UCB may be immunologically defective because UCB DCs are immature [41-42]. This hypothesis was validated by findings that UCB DCs were weakly stimulatory to T cells in a mixed lymphocyte reaction when compared to APB counterparts [40,43-45].
DCs can be developed from CD14+ monocytes isolated from either APB or UCB by culture in vitro in the presence of GM-CSF and IL-4. GM-CSF/IL-4 derived immature DCs are converted to the mature form of DCs by various stimuli from pathogen related products such as bacterial lipopolysaccharides (LPS) [46], proinflammatory cytokines such as TNF-α and IL-1β [47], as well as CD40 ligation [48]. Studies performed with isolated CD14+ monocytes, or in vitro-generated CD14+ monocyte-derived DCs indicate that mononuclear cells (MNCs) and UCB DC are less susceptible to complete maturation compared to their counterparts in APB [49]. DCs generated from UCB CD14+ monocytes produced lower amounts of TNF-α compared to DCs derived from APB [49]. UCB derived DC are characterized by a defect in IL-12 production, which is responsible for an impaired ability to elicit IFN-γ production by allogeneic T cells [50-51]. The low production of these pro-inflammatory cytokines does not result from low expression levels of LPS receptors, CD14 and toll-like receptor (TLR)-4 [49], suggesting that UCB MNCs have an intrinsic limitation in their differentiation into fully mature DCs [52]. Consistent with this, surface expression of HLA-DR, CD40, CD86 and CD83 was significantly lower in UCB DCs compared to APB DC after LPS stimulation [49]. These characteristics of UCB DC can delay activation of naive T cells, especially that of Th1 cell development. The defective APC activity of CB DCs can be restored with upregulation of CD86 and HLA-DR after prolonged stimulation with irradiated allogeneic T cells, suggesting that UCB APC activity can be gradually increased after transplantation and can contribute to delayed GVHD [40].
Human DCs consist of two subsets, CD11c+ CD123- myeloid (mDC) and CD11c− CD123+ plasmacytoid DCs (pDCs). pDCs tend to induce Th2 cell responses, involving the induction and maintenance of tolerance, whereas mDCs stimulate Th1 cell responses [53]. UCB DCs are characterized by a higher ratio of CD11c− CD123+ pDCs to CD11c+ CD123− mDCs than APB DCs [49,54]. In addition, UCB pDCs display low expression of costimulatory molecules and TNF-α compared to APB counterpart DCs, eliciting impaired responses to LPS and unmethylated cytosine-phosphate-guanosine (CpG) [49]. Taken together, weakly immunogenic UCB DCs to allogeneic T cells may be an additional important factor responsible for the relatively low incidence of severity of GVHD in UCB transplants.
2.4. Tolerogenic properties of UCB DCs
Immature DC are known to act as immune suppressor cells inducing antigen specific T-cell tolerance [55-58]. Allograft rejection has been partly prevented through tolerance inducing immature DCs in the periphery [59-60]. Immature DCs also induce regulatory properties to effector T cells [61-62] and expansion of naturally occurring CD25+ Tregs [63]. In support of this, administration of DCs at an immature state into healthy volunteers in the absence of inflammatory signals induced Tregs capable of suppressing alloantigen-specific T cell responses [64]. The immunological features of DCs are affected by MSCs; DCs cultured in the presence of MSCs show increased production of IL-10 and reduced production of IL-12 [65]. Hence, MSCs in umbilical cord (UC) may induce DCs to enter an immature or a tolerogenic state, which might be an indirect mechanism responsible for inducing T-cell hyporesponsiveness.
Higher serum levels of macrophage colony-stimulating factor (M-CSF) have been detected in UCB than in APB [66], and pregnancy is associated with a skewed Th2 cell cytokine profile [67]. Thus, UCB CD14+ monocytes might have a greater chance to be exposed to M-CSF and IL-4. We previously demonstrated that UCB CD14+ monocytes cultured with M-CSF and IL-4 differentiated into DCs expressing high and low levels of IL-10 and IL-12, respectively with tolerogenic potential in allogeneic mixed lymphocyte reaction [68]. This suggests that M-CSF rich in UCB may play a role in maintaining or inducing immature tolerogenic DCs from UCB. Therefore, UCB DCs may be characterized by the lack of antigen presenting capacity but by the potential to induce and expand Tregs [56,61-63].
3. Immune modulator cells
UCB and umbilical cord (UC) are a source of both Tregs and MSCs. These cells have been scruinized intensively as potential cell mediators to control GVHD. Advances in Treg and MSC biology may be helpful for designing safer and more effective HSCT in treatment of a variety of malignant and non-malignant diseases [69]. The following discusses charateristics of Tregs and MSC in UCB, and the significance of these immune regulatory cells in terms of clinical implications in GVHD after UCBT. Also described are recent methods to expand these immune regulatory cells.
3.1. Tregs
Sakaguchi et al. [70] first reported that CD25+-depleted CD4+ T cells transferred into nude mice resulted in autoimmune disease, events reversed by adding back CD25+ T cells. Since then, CD25+CD4+ T cells have been termed Tregs by their functional characteristics of suppressing T cell immune response [71]. Foxp3, a helix-loop-helix transcription factor, is considered to be one specific marker for Treg cells [72]. Treg cells are characterized as cells with constitutive expression of CD25 and Foxp3, and represent 5–10% of peripheral CD4+ T cells. Tregs express other surface markers, including CTLA-4 (CD152) [73], glucocorticoid-induced TNF receptor (GITR) [74-75] and OX-40 (CD134) [76]. As Tregs modulate immune responses through selective migration and accumulative retention at tissue specific sites, they express integrin molecule CD62L (L-selectin) [77-78] and chemokine receptor CCR7 [79], crucial for homing to secondary lymphoid tissues. Treg cells also specifically express chemokine receptors CCR4 and CCR8 which may guide Treg cells to inflamed areas to attenuate T cell activation [80].
Transwell experiments demonstrate that the suppressive function of Tregs partially depend on cell–cell contact. Surface transforming growth factor-β (TGF-β) [81] and CTLA-4 might be involved in Treg mediated suppression [82]. TGF-β secreted by Tregs may further convert naive CD4+ CD25− cells to CD25+ Foxp3+ Tregs referred to as induced Tregs (iTregs) [83]. Secretion of IL-10 and possibly other unknown cytokines action in a paracrine fashion may also be responsible for the suppressive activity of Tregs. In the presence of IL-6 and TGF-β, naive T cells differentiate into Th17 cells, whereas in the absence of IL-6, same cells are induced to become Tregs [84]. Thus, IL-6 occupies a unique position at the crossroads of Treg and proinflammatory Th17 cells. IL-6 is not indispensable for Th17 cell development because IL-21 may induce Th17 differentiation even in the absence of IL-6 [85].
Mammalian target of rapamycin (mTOR), a downstream target of the PI3K/AKT signaling pathway, acts as a negative regulator for Tregs. Rapamycin, an inhibitor of mTOR pathway promotes expression of Foxp3 and proliferation of Tregs, while it promotes activation-induced cell death of effector T cells [86-87]. In vivo, rapamycin administration reduces IL-6 production and promotes alloantigen-specific Treg cell activity as well as long-term, donor-specific heart graft survival in immunocompetent hosts [88]. These findings suggest that mTOR inhibition in Tregs may be an effective approach to control GVHD and promote long-term organ graft survival. It remains to be seen whether UCB Tregs are distinct from those from other tissue sources in the mTOR pathway or IL-6 production.
3.2. Human vs. mouse Treg
For more than two decades, immune suppressive properties of Tregs have been well-described mainly in mice [70,89-91]. Compared to mouse Tregs, human Tregs appeared to be difficult to study due to lack of specific surface markers. Initially, all CD25+ cells in CD4+ T cell populations was considered to represent Tregs in laboratory rodents [92], but CD25 is also expressed on recently activated T cells [93]. Twenty–60% of CD4+ T cells in human APB express CD25 [94], and a large proportion of these human T cells represent activated or memory cells generated as a result of previous encounter with foreign antigens [95]. Thus, the CD25 molecule is not a reliable marker for natural Tregs particularly in humans. CD4+ CD25+ T cells existing in human APB consist of two populations: CD25int and CD25bright cells. Only the 25bright population in human CD4+25+ cells, which consist of only 1-2% of CD4+ T cells, are largely FoxP3+ Treg cells, and these populations may be relevant to murine CD25+ CD4+ T cells with regulatory ability [96-97].
Although Foxp3 is considered to be a reliable marker for murine Treg, in humans Foxp3 is also expressed in activated CD4+ T cells [72,98-99]. Thus, Foxp3 expression detected in human CD4+ CD25+ T cells may not fully reflect the regulatory capacity of these cells. Other reliable markers have been sought to further specify human Tregs and to improve isolation and purification procedures. CD4+ CD25+ cells with low levels of CD127 (IL-7Ra chain) denote human Treg cells with increased suppressive activity. The CD127lo phenotype highly correlates with expression of FoxP3 within the CD4+CD25+ population [100]. Expression of CD45RA has also been reported to distinguish Tregs from activated T cells [101], but CD127 and CD45RA does not absolutely designate human Tregs. Booth et al. [102] report that the majority (approximately 80%) of natural Tregs in adults were CD45RO+. The proportion of CD45RO+ to CD45RA+ Tregs increased significantly with age. CD4+CD25+CD27+ Tregs have been identified as a potent regulatory subset in UCB and in a large-scale in vitro expansion system [103]. Combined expression of CD25 and CD27 allow the differentiation of highly suppressive FoxP3+ regulatory T cells from activated effector T cells[104]. Rapamycin fosters dominance of CD27+Tregs over CD27− Tregs after expansion of the CD4+CD25+ Treg pool upon allogeneic activation[104]. New discriminatory cell surface antigens have yet to be discovered to further specify human Tregs from CD4+CD25+ population.
3.3. Characteristics of UCB Tregs
UCB contains CD4+CD25+ cells which manifest similar suppressive activity to APB[97,105-106], but it remains controversial as to whether UCB possesses distinctive Treg functions compared to APB. It has been reported that the CD4+CD25bright subset exists at relatively higher frequency in UCB compared to APB [97,106]. Consistent with this, UCB CD4+ CD25+ T cells have been shown to contain greater Foxp3 expression than their APB counterparts, suggesting the greater abundance of Tregs in UCB than APB [106]. Another study, however, showed that the expression of Foxp3 protein on naïve UCB CD4+ T cells was lower compared with those of APB [18], which argues against increased Treg activity as a mechanism for decreased Th1 differentiation of UCB CD4+ T cells. UCB CD25+ cells suppress polyclonal T cell activation, but do not suppress antigen-specific responses [107]. It remains unclear what the causes are of the inconsistency among reports in respect to suppressive activity of CB Tregs. The inconsistent results on suppressive activities of CB Tregs may result from distinct sources of target CD4+ T cells tested. UCB CD4+ CD25+ T cells show suppressive effects on APB CD4+ T cells, but not UCB CD4+ T cells, suggesting that target CD4+ T cells from UCB are largely immature and therefore not as susceptible as target CD4+ T cells from APB.
UCB CD25+ cells are mainly naive [107]. Thus, a significant number of UCB Tregs remain as precursor cells and become capable of potent suppressor function after maturation [103]. It is likely that UCB CD25+ CD4+ T cells may require a certain type of stimulation to fully gain suppressive activity [108]. For example, UCB CD4+CD25+ cells function as Tregs after polyclonal stimulation [107]. IL-2 and IL-15 are known to increase expression of Foxp3, CTLA-4, GITR and membrane bound TGF-β, necessary elements for Treg, in UCB CD4+CD25+ T cells [106]. The number and activity of Treg cells in UCB have been correlated with degrees of antigenic exposures of mothers during pregnancy [109]. Therefore, the inconsistency of UCB Treg activities reported may be rooted in the different isolation procedures used and degree of stimulation during Treg cell preparation.
3.4. UCB Tregs for clinical applications
Numerous studies from mice administered allogeneic HSCs demonstrated that donor or host Tregs are able to ameliorate GVHD [21,77,110-115]. In contrast, removal of Tregs from the donor allograft accelerated GVHD in recipients [111,115-117]. Although accumulated evidence suggests that donor Treg cell infusion is beneficial for allogeneic transplantation in mouse models, minimal data are reported on the function of UCB Tregs transplanted in humans. Further studies are needed to validate putative roles of UCB Tregs in preventing GVHD in humans. In fact, clinical trials of donor Treg infusions in humans have lagged behind mice mainly due to the difficulty in isolating definitive human Treg populations. The main difficulty in preparing adequate numbers of Tregs with high quality stems from the fact that unlike mouse Tregs of which separation is readily achievable, human Tregs are not clearly defined by surface expression of CD25. Although Foxp3 is the most reliable marker for Tregs, isolation of live Foxp3+ population is not feasible, because intracellular staining of FoxP3+ cells requires permeabilization of T cells, which is incompatible with their viability. Thus, establishment of a protocol to reliably and consistently identify and isolate human Tregs is necessary to fully exploit its therapeutic potential for controlling GVHD after UCB-HSCT. Tregs need to be at high frequency and present in the right microenvironment to exert sufficient suppressive function-dosing and localization. This requirement may render Treg therapy challenging. The suppressive effects of Tregs may be temporary and may be overridden by certain proinflammatory conditions and activating cell signaling [118]. Further research efforts are needed to ensure that infused Tregs retain immune suppressive activity and the capacity to home to local inflammatory sites and secondary lymphoid organs where GVHD is initiated. Since IL-2 is required for Treg cell function, paradoxically, immunosuppressive reagents such as steroids and calcineurin inhibitors, which are intended to block IL-2 production by preventing alogeneic T cell immune responses should be avoided when Tregs are infused to patients to suppress GVHD [119].
3.5. Ex vivo expansion of UCB Tregs
Tregs can be purified from UCB with a less cumbersome approach than from APB, because CD25+ CD4+ T cells in UCB do not contain activated or memory T cell populations [97]. Treg cells have been purified from frozen UCB units by positive selection using directly conjugated anti-CD25 magnetic microbeads [120]. Although CD25bright Tregs are more readily purified from UCB units than APB [103,121], only ~3–7.5 × 106 Tregs can be isolated from a single UCB unit [69,103]. The numbers of Tregs may not be enough for the patients who need multiple infusions for adoptive immunotherapy in GVHD prevention [69]. Therefore, many expansion procedures have been explored. Treg cells are cultured with beads coated with anti-CD3/28 monoclonal antibody (mAb) and artificial antigen-presenting cells (aAPCs) preloaded with anti-CD3/28 mAbs plus exogenous IL-2 and rapamycin for 18 to 21 days and cell cultures split every 2 to 3 days [120]. Tregs are successfully expanded approximately by ~200- to 1000-fold in <3 weeks under the conditions. These expanded CD4+CD25+ cells express Foxp3 mRNA at a level about 100-fold higher than that in isolated CD25− cells and can suppress allogeneic mixed lymphocyte culture by >80% (effector cells: CD4+CD25+ cells = 2:1) [122].
Costimulation promotes Treg cell proliferation as seen in conventional T cells. When additional costimulation with OX40 or 4-1BB, which belong to the tumor necrosis factor receptor family, is added to that with CD28, UCB Tregs expand to a significantly greater extent, reaching mean expansion levels exceeding 1250-fold. Expanded UCB Treg cells (routinely ≥50% CD4+25++Foxp3+) preserve suppressive potency and increase TGF-β secretion, as tested in a xenogeneic mouse GVHD lethality model [120]. IL-2 is important for expanding Treg cells [103,121]. As Tregs do not produce sufficient amounts of IL-2, exogenous IL-2 is critical for their expansion [123]. IL-15, whose receptor shares IL-2 receptor β and γ chains, is comparable or superior to IL-2 for optimal expansion of CD3/CD28-stimulated UCB Tregs [106], as well as APB derived Treg cells [124]. A combination of IL-15 and IL-2 enhances survival of Treg cells [123]. mTOR negatively regulates development of Treg [88]. Rapamycin, an mTOR inhibitor enhances Treg cell expansion and potency, while suppressing outgrowth of contaminating activated or memory T cells [125]. Improved techniques to expand Tregs ex vivo in high quality without losing their regulatory function are yet to be established for clinical applications.
3.6. In vitro development of Treg cells from UCB CD34+ cells
CD4+CD25+ CD127loFoxP3+ Treg cells can be developed in vitro from UCB CD34+ cells by co-culture on Notch ligand, Delta-like 1 (DL1) expressing OP9 stromal cells [126]. These CD34+ cell-derived Tregs are comparable in suppressive activity with natural Tregs existing in UCB, as tested in vitro. If this culture system can generate functional Tregs in sufficient numbers, UCB CD34+ cell-derived Tregs may provide an alternative means to prepare Tregs for cell-based therapy to induce transplantation tolerance and treatment of GVHD. Recently, it has been reported that Tregs can be derived from human embryonic stem cells in vitro [127]. However, generation of Treg cells from UCB CD34+ cells may be more efficient and less controversial for future clinical use than those from human embryonic stem cells.
4. Mesenchymal stem cells (MSCs)
Mesenchymal stem cells, also called mesenchymal stroma cells, were identified as undifferentiated stromal cells [128-129] originally from BM, and then from various other tissue sources [130]. MSCs can differentiate into several types of cells, including osteocytes, chondrocytes, adipocytes, cardiomyocytes and neurons [128-129,131]. They have been suggested as potential sources of cells for tissue engineering and cell-based therapy [128,132-134]. MSCs are able to self-renew with high proliferative capacity [135]. MSCs contain heterogeneous cell populations with multiple phenotypes [134,136], and thus, identity and functional complexity of MSCs in vivo remain to be better evaluated.
4.1. MSCs as immunomodulator cells
Importantly, MSCs are considered to have low immunogenicity and an ability to suppress immune responses [60,137-138], primarily suppressing T cell proliferation and cytokine production [139-147]. Because of their immune regulatory activity, MSCs are implicated in roles for controlling GVHD [148]. MSCs have been shown to exhibit immunosuppressive activity [60,138], MSCs do not express MHC class II molecules or costimulatory molecules (B7 and CD40) required for T cell activation [149]. Thus, MSCs do not initiate allogeneic responses in vitro [149], and may not be rapidly rejected in vivo. It is however unclear how long transplanted MSCs can be maintained in vivo and exactly where they home and reside.
Although MSC are able to prevent expansion of allogeneic T cells [144,149], exact mechanisms of MSC-mediated immunosuppression are largely unknown. Various immune suppressive factors such as IL-10 [150], TGF-β [151], nitric oxide [146], indoleamine 2,3-dioxygenase (IDO) [152], and prostaglandin (PG) E2 [144,153] have been suggested to be involved in MSC-mediated immunosuppression. In vitro addition of TNF-α, a proinflammatory cytokine, is sufficient to reverse the immunosuppressive effect of MSCs on T cell proliferation [154]. Interestingly, IFN-γ and inducible nitric oxide synthase (iNOS) have been shown to be involved in MSC-mediated prevention of GVHD in mice, suggesting that proinflammatory cytokines are required to induce immunosuppression by MSCs [138]. In mice, BM MSCs inhibit proliferation of T and B lymphocytes via engagement of the inhibitory molecule PD-1 [143]. In addition to the suppressive effect on T cell function, MSCs induce de novo generation of antigen-specific CD4+CD25+Foxp3+ Treg cells with the capacity to suppress effector T cell responses [155]. However, the clinical potential of these immunosuppressive properties is still a matter of speculation, because most data has been obtained from in vitro studies. If MSCs are immunosuppressive in controlled in vivo settings, then transplantation of MSCs together with allogeneic HSCs might facilitate acceptance of stem cell-based therapies, including possibly ES cell derivatives [60].
4.2. Umbilical cord (UC) and UCB MSCs
MSCs have been isolated from UCB [137,156-159], but their numbers are low and there is great variability in numbers of these cells in different UCB collections [160-163]. UCB-MSCs do not express costimulatory molecules, CD40, CD80, CD86 and MHC class II molecules commonly expressed on antigen presenting cells [137,164]. Parameters involved in MSC purification, including time from collection to isolation (less than 15 hours), a net volume (more than 33 ml), and MNC count (more than 1 × 108 MNCs) seem important for isolating MSCs from UCB [159]. More recently, instead of UCB, umbilical cord (UC) has been recognized as a source of MSCs [135,160,163,165-169]. In situ analysis showed that cells expressing mesenchymal markers CD44, CD105, CD73, and CD90 were enriched in UC. Jelly-like matrix and surrounding connective tissue, including vessels called Wharton’s Jelly (WJ), in UC are tissue sources to obtain high numbers of MSCs [135,168-170]. Secco et al.[160] reported that they were able to isolate large amounts of multipotent MSCs from all UC samples, whereas MSCs were obtained from only 1 of the 10 UCB samples. MSCs isolated from WJ (UC-MSC) express MSC markers, including matrix receptors (CD44, CD105) and integrin markers (CD29, CD51), but not hematopoietic lineage markers (CD34, CD45) [164]. Unlike traditional MSCs derived from adult BM, small populations of UC-MSCs express endoglin (CD105) and CD49e. UC-MSCs are negative for CD14, CD33, CD56, CD31, CD34, CD45, and HLA-DR [170-171].
Perivascular niches are major sources of MSCs in several organs [168,172-173]. MSC-like cells have been obtained from the vasculature of the UC, called human umbilical cord perivascular cells (HUCPVCs) [174]. HUCPVCs have the same capacity as other MSCs to differentiate into an osteogenic phenotype with high proliferative potential. HUCPVCs have been considered as possible extra-embryonic MSC sources for cell-based therapies [175].
MSCs isolated either from UCB or UC are similarly multipotent for differentiation into various cell types and can undergo long-term expansion in culture [164,169,175]. However, UC- and UCB-MSCs appear to differ in gene expression profiles [167]. Genes related to cell adhesion, morphogenesis, secretion, angiogenesis and neurogenesis are expressed more dominantly in UC-MSCs, whereas genes related to osteogenesis and the immune system are preferentially expressed in UCB-MSCs [167]. These tissue-specific MSC gene expression patterns may reflect functional activities influenced by distinct niches. These findings highlight the consideration that banking UC as well as UCB may be of future clinical utility.
4.3. UC(B)- versus BM-MSC
UC-MSCs are different from BM MSCs in cytokine expression profile [171,176]. UC-MSCs secrete significantly higher levels of G-CSF, GM-CSF, HGF, LIF, IL-1α, IL-6, IL-8, and IL-11 compared with BM-MSCs, whereas BM-MSCs produce more VEGF and SDF-1β than UC-MSCs [177]. UC-MSCs act to significantly enhance numbers of granulocyte macrophage (CFU-GM), but not erythroid (BFU-E) or multi-potential (CFU-GEMM) progenitor cells from UCB CD34+ cells [177]. This UCB-MSC effect in relation to BM-MSCs has not been addressed. Recently, pluripotency of MSC was further extended by reprogramming into embryonic stem cell-like, induced pluripotent stem cells (iPS) [178]. Also, iPS cells have been generated from immature subpopulations of UCB, but the potential of these cells for clinical applicability is not clear [179].
MSCs derived from UCB and BM exhibit similar multilineage progenitor potential and cytokine expression profiles [180-182] and differentiate equally well to osteocytes and adipocytes [130,159,183-184]. However, there are studies indicating that UC-MSCs are more committed to angiogenesis, whereas BM-MSCs are more committed to osteogenesis [176]. Also, although MSCs derived from all three sources are morphologically and immunophenotypically similar, some studies suggest that UCB-derived MSCs produce less adipogenic differentiation with fewer and smaller lipid vacuoles, in contrast to those of BM- and adipose tissue (AT)-derived MSCs [185]. It was shown that although there are no obvious significant differences concerning morphology and immune phenotype of the MSCs derived from UCB, BM and AT, the success rate for isolating MSCs from UCB was low due to the low colony frequency compared to cells from BM and AT [130]. However, MSCs isolated from UCB have higher self-renewal capacity than those from BM [130,156]. UCB-MSC can be cultured for longer with a higher proliferation capacity compared with MSC derived from BM and AT [130,156-158,185-186]. This may reflect a superior “stemness” of UCB-MSC as newborn cells [187-190]. Of note, differentiation potential and maximum life span of human BM-derived MSC significantly decline with age of donor [191-193].
HUCPVCs are capable of osteogenic, chondrogenic, and adipogenic differentiation and show a higher proliferative potential than BM MSCs. Like UC-MSCs [176], HUCPVCs proceed to osteogenic differentiation more rapidly than BM MSCs [175]. HUCPVCs show a higher rate of growth relative to BM MSCs. BM MSCs tend to experience contact-inhibited growth, whereas HUCPVCs continue to grow in a multilayer form [175].
4.4. Preclinical possibilities of UCB-MSCs
MSCs have been suggested as potential clinically useful cells to prevent or treat autoimmune diseases, such as experimental autoimmune encephalomyelitis [194], and collagen-induced arthritis [154]. An important aspects of MSCs for clinical application is low immunogenicity and immunosuppressive activity [137]. The immunosuppressive property of MSCs is also attractive tool for modulating GVHD and HSC engraftment [140,195-196]. Most information on beneficial effects of UCB-MSCs on GVHD is available from mouse models. The ability of UCB-MSC to prevent and treat GVHD has been tested using sublethally irradiatiated non-obese diabetic/severe combined immunodeficient (NOD/SCID) mice which were injected with human peripheral blood mononuclear cells (PBMCs) [148,197]. These mice exhibited pathologic signs of GVHD such as massive infiltrates of human T cells in mesenteric lymph node, liver, lung and peritoneal lavage [148]. Multiple systemic administration of UCB-MSCs to this NOD/SCID mouse model significantly reduced human T cell proliferation and tissue damage, improving survival of the animals [148]. No therapeutic effect was observed when UCB-MSCs were administered at the onset of GVHD, suggesting the clinical use of MSCs in HSCT as a prophylaxic treatment [148]. Multiple administration of UCB-MSC administration were required to prevent GVHD, suggesting that effects of MSCs may be transient. Rats that were not immune-suppressed have been used to test human UC-MSCs for therapeutic value. Transplantation of UC-MSCs into brains of hemiparkinsonian rats ameliorated the pathological symptoms without host immune rejection response [170]. This opens a possibility, far from proven, that UC-MSCs could be a cell source to treat neurodegenerative diseases.
Discordant reports have been reported on the efficacy of MSCs to treat GVHD in mouse models. Sudres et al. [198] demonstrated that infusion of MSCs was not sufficient to prevent the development of GVHD in mice, with infused MSCs being rapidly eliminated by an immune response from the recipient [199-200]. Chung et al. [201] showed that MSCs could prevent GVHD only when given to irradiated recipient mice. There have been numerous reports on preclinical human trials using MSCs isolated from BM to treat GVHD [202-204], but controversial results from preclinical models remain to be clarified. Thus far, the efficacy of MSCs on GVHD in preclinical settings, while encouraging, are not without controversy. Besides immune regulatory activity, in vivo administration of MSCs was found to be effective for promoting the engraftment of CD34+ hematopoietic stem/progenitor and embryonic stem (ES) cells [196,205].
5. Natural killer (NK) cells as a regulatory cell in UCB
Recently, the impact of donor NK cells on autologous donor alloreactive T cells in GVHD induction has been investigated in a mouse model [206]. Animals receiving donor NK cells in addition to T cells showed improved survival and decreased GVHD compared to controls receiving donor T cells alone. NK cells exert a direct cytotoxic effect on autoantigen-specific, encephalitogenic T cells [207]. In another mouse model, NK cells recognize and kill syngeneic CD4+ and CD8+ T cells activated by APCs. Lysis required perforin and NKG2D in the NK cells [208]. These studies suggest that NK cells may play regulatory roles for down-modulating alloreactive T cells in GVHD by reducing donor T cell proliferation and increasing T cell apoptosis.
Human natural killer (NK) cells are defined as being membrane CD3−, CD16+, and/or CD56+lymphocytes. Based on the surface expression of CD56 and CD16, NK cells can be subdivided into four subsets on the basis of CD56 and CD16 expression with distinct functions which are associated with expression of NKG2A, NKG2D, CD95 and the intracellular granzyme B and perforin. It was shown that the CD56−CD16+ subset is particularly abundant in UCB compared to APB and BM [209]. UCB CD56−CD16+ NK cells express equivalent levels of perforin and granzyme B to CD56+CD16+ NK cells [209-212]. Among different NK subsets, expression of NKG2A, an inhibitory receptor, is lower on CD56−CD16+ subset than the other NK cell subsets, while expression NKG2D, an activating receptor, is indistinguishably high on all the NK subsets [210]. CD56−CD16+ NKs have also been identified after allo-HCT using UCB, but not after BM or PB transplants [213]. CD56−CD16+ NKs are found at higher frequencies in immune compromised hosts, including those with chronic viral infections [214-216]. Culture of the UCB-derived CD56−CD16+ NK cells with IL-2 and/or IL-15 resulted in CD56 acquisition, possibly suggesting that CD56−CD16+ NK cells may be NK progenitors. These results suggest that UCB CD56+CD16+ NK cells are functionally immature but have potential for cell lysis. Whether alloreactive T cells are particularly susceptible to the CD56−CD16+ NK cell-mediated lysis remains unclear.
6. Summary
In summary, UCB is an alternative source of primitive HSC and progenitor cells to BM or G-CSF-mobilized APB stem cells. UCBT results in less incidence of GVHD compared to BM stem cell transplantation. Although reduced GVHD in patients receiving unrelated donor UCB transplantation is generally thought to be attributed to greater immune tolerance to HLA mismatch, the exact mechanisms remain unclear. In this review, we described the nature of immune regulatory cells in UCB. Tregs and MSCs, may eventually be of use for adaptive cellular therapy to suppress alloreactivity for GVHD. In light of the tolerogeneic nature of Tregs and MSCs, how to effectively expand these immune regulatory cells in UCB is a key issue for clinical usage of these cells to treat GVHD in the near future. Delayed immune reconstitution is problematic in UCBT, largely due to the limited number of HSC infused, lack of prior priming against pathogens, and relative bias away from a protective Th1/Tc1 cytokine profile that in combination may delay emergence of protective antiviral immunity. Double unit UCBT [217-220] and inhibition of CD26 [221-223] may be feasible approaches to increasing the efficiency of immune reconstitution in UCBT.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Broxmeyer HE, Srour EF, Hangoc G, et al. High-efficiency recovery of functional hematopoietic progenitor and stem cells from human cord blood cryopreserved for 15 years. Proc Natl Acad Sci U S A. 2003;100:645–50. doi: 10.1073/pnas.0237086100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Komanduri KV, St. John LS, De Lima M, et al. Delayed immune reconstitution after cord blood transplantation is characterized by impaired thymopoiesis and late memory T-cell skewing. Blood. 2007;110:4543–51. doi: 10.1182/blood-2007-05-092130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Szabolcs P, Cairo MS. Unrelated umbilical cord blood transplantation and immune reconstitution. Semin Hematol. 2010;47:22–36. doi: 10.1053/j.seminhematol.2009.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Szabolcs P, Niedzwiecki D. Immune reconstitution after unrelated cord blood transplantation. Cytotherapy. 2007;9:111–22. doi: 10.1016/j.bbmt.2007.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Parody R, Martino R, Rovira M, et al. Severe infections after unrelated donor allogeneic hematopoietic stem cell transplantation in adults: comparison of cord blood transplantation with peripheral blood and bone marrow transplantation. Biol Blood Marrow Transplant. 2006;12:734–48. doi: 10.1016/j.bbmt.2006.03.007. [DOI] [PubMed] [Google Scholar]
- [6].Szabolcs P, Niedzwiecki D. Immune reconstitution in children after unrelated cord blood transplantation. Biol Blood Marrow Transplant. 2008;14:66–72. doi: 10.1016/j.bbmt.2007.10.016. [DOI] [PubMed] [Google Scholar]
- [7].Macmillan ML, Weisdorf DJ, Brunstein CG, et al. Acute graft-versus-host disease after unrelated donor umbilical cord blood transplantation: analysis of risk factors. Blood. 2009;113:2410–5. doi: 10.1182/blood-2008-07-163238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Gluckman E, Devergie A, Bourdeau-Esperou H, et al. Transplantation of umbilical cord blood in Fanconi’s anemia. Nouv Rev Fr Hematol. 1990;32:423–5. [PubMed] [Google Scholar]
- [9].Gluckman E, Broxmeyer HE, Auerbach AD, et al. Hematopoietic reconstitution in a patient with Fanconi’s anemia by means of umbilical-cord blood from an HLA-identical sibling. N Engl J Med. 1989;321:1174–8. doi: 10.1056/NEJM198910263211707. [DOI] [PubMed] [Google Scholar]
- [10].Broxmeyer HE, Douglas GW, Hangoc G, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A. 1989;86:3828–32. doi: 10.1073/pnas.86.10.3828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Broxmeyer HE, Smith F. Cord blood hematopoietic cell transplantation. In: Appelbaum FR, Forman SJ, Negrin RS, et al., editors. Thomas’ hematopoietic cell transplantation. 4th Edition 2009. pp. 559–76. [Google Scholar]
- [12].Matsuno N, Wake A, Uchida N, et al. Impact of HLA disparity in the graft-versus-host direction on engraftment in adult patients receiving reduced-intensity cord blood transplantation. Blood. 2009;114:1689–95. doi: 10.1182/blood-2008-12-194696. [DOI] [PubMed] [Google Scholar]
- [13].Barker JN, Davies SM, Defor T, et al. Survival after transplantation of unrelated donor umbilical cord blood is comparable to that of human leukocyte antigen-matched unrelated donor bone marrow: results of a matched-pair analysis. Blood. 2001;97:2957–61. doi: 10.1182/blood.v97.10.2957. [DOI] [PubMed] [Google Scholar]
- [14].Rocha V, Wagner JE, Sobocinski KA, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. N Engl J Med. 2000;342:1846–54. doi: 10.1056/NEJM200006223422501. [DOI] [PubMed] [Google Scholar]
- [15].Rocha V, Labopin M, Sanz G, et al. Transplants of umbilical-cord blood or bone marrow from unrelated donors in adults with acute leukemia. N Engl J Med. 2004;351:2276–85. doi: 10.1056/NEJMoa041469. [DOI] [PubMed] [Google Scholar]
- [16].Leung AYH, Kwong Y-L. Haematopoietic stem cell transplantation: current concepts and novel therapeutic strategies. Br Med Bull. 2009:ldp040. doi: 10.1093/bmb/ldp040. [DOI] [PubMed] [Google Scholar]
- [17].Canto E, Rodriguez-Sanchez JL, Vidal S. Distinctive response of naive lymphocytes from cord blood to primary activation via TCR. J Leukoc Biol. 2003;74:998–1007. doi: 10.1189/jlb.0303098. [DOI] [PubMed] [Google Scholar]
- [18].Chen L, Cohen AC, Lewis DB. Impaired allogeneic activation and T-helper 1 differentiation of human cord blood naive CD4 T cells. Biol Blood Marrow Transplant. 2006;12:160–71. doi: 10.1016/j.bbmt.2005.10.027. [DOI] [PubMed] [Google Scholar]
- [19].Risdon G, Gaddy J, Broxmeyer HE. Allogeneic responses of human umbilical cord blood. Blood Cells. 1994;20:566–70. [PubMed] [Google Scholar]
- [20].Risdon G, Gaddy J, Stehman FB, et al. Proliferative and cytotoxic responses of human cord blood T lymphocytes following allogeneic stimulation. Cell Immunol. 1994;154:14–24. doi: 10.1006/cimm.1994.1053. [DOI] [PubMed] [Google Scholar]
- [21].Trenado A, Charlotte F, Fisson S, et al. Recipient-type specific CD4+CD25+ regulatory T cells favor immune reconstitution and control graft-versus-host disease while maintaining graft-versus-leukemia. J Clin Invest. 2003;112:1688–96. doi: 10.1172/JCI17702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Reen DJ. Activation and functional capacity of human neonatal CD4 T-cells. Vaccine. 1998;16:1401–08. doi: 10.1016/s0264-410x(98)00099-1. [DOI] [PubMed] [Google Scholar]
- [23].Yi T, Chen Y, Wang L, et al. Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease. Blood. 2009;114:3101–12. doi: 10.1182/blood-2009-05-219402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Yi T, Zhao D, Lin CL, et al. Absence of donor Th17 leads to augmented Th1 differentiation and exacerbated acute graft-versus-host disease. Blood. 2008;112:2101–10. doi: 10.1182/blood-2007-12-126987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Fowler D, Kurasawa K, Smith R, et al. Donor CD4-enriched cells of Th2 cytokine phenotype regulate graft- versus-host disease without impairing allogeneic engraftment in sublethally irradiated mice. Blood. 1994;84:3540–49. [PubMed] [Google Scholar]
- [26].Krenger W, Ferrara JL. Graft-versus-host disease and the Th1/Th2 paradigm. Immunol Res. 1996;15:50–73. doi: 10.1007/BF02918284. [DOI] [PubMed] [Google Scholar]
- [27].Nitsche A, Zhang M, Clauss T, et al. Cytokine profiles of cord and adult blood leukocytes: differences in expression are due to differences in expression and activation of transcription factors. BMC Immunol. 2007;8:18. doi: 10.1186/1471-2172-8-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Szabo SJ, Sullivan BM, Peng SL, et al. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21:713–58. doi: 10.1146/annurev.immunol.21.120601.140942. [DOI] [PubMed] [Google Scholar]
- [29].Kaminski BA, Kadereit S, Miller RE, et al. Reduced expression of NFAT-associated genes in UCB versus adult CD4+ T lymphocytes during primary stimulation. Blood. 2003;102:4608–17. doi: 10.1182/blood-2003-05-1732. [DOI] [PubMed] [Google Scholar]
- [30].Yang YG, Dey BR, Sergio JJ, et al. Donor-derived interferon γ is required for inhibition of acute graft-versus-host disease by interleukin 12. J Clin Invest. 1998;102:2126–35. doi: 10.1172/JCI4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Murphy WJ, Welniak LA, Taub DD, et al. Differential effects of the absence of interferon-γ and IL-4 in acute graft-versus-host disease after allogeneic bone marrow transplantation in mice. J Clin Invest. 1998;102:1742–8. doi: 10.1172/JCI3906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Burman AC, Banovic T, Kuns RD, et al. IFNγ differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract. Blood. 2007;110:1064–72. doi: 10.1182/blood-2006-12-063982. [DOI] [PubMed] [Google Scholar]
- [33].Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Kappel LW, Goldberg GL, King CG, et al. IL-17 contributes to CD4-mediated graft-versus-host disease. Blood. 2009;113:945–52. doi: 10.1182/blood-2008-08-172155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Carlson MJ, West ML, Coghill JM, et al. In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations. Blood. 2009;113:1365–74. doi: 10.1182/blood-2008-06-162420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Blazar BR, Taylor PA, Panoskaltsis-Mortari A, et al. Interleukin-10 dose-dependent regulation of CD4+ and CD8+ T cell-mediated graft-versus-host disease 1. Transplantation. 1998;66:1220–29. doi: 10.1097/00007890-199811150-00018. [DOI] [PubMed] [Google Scholar]
- [37].Groux H, O’garra A, Bigler M, et al. A CD4+T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature. 1997;389:737–42. doi: 10.1038/39614. [DOI] [PubMed] [Google Scholar]
- [38].Rainsford E, Reen DJ. Interleukin 10, produced in abundance by human newborn T cells, may be the regulator of increased tolerance associated with cord blood stem cell transplantation. Br J Haematol. 2002;116:702–09. doi: 10.1046/j.0007-1048.2001.03321.x. [DOI] [PubMed] [Google Scholar]
- [39].Chakraverty R, Sykes M. The role of antigen-presenting cells in triggering graft-versus-host disease and graft-versus-leukemia. Blood. 2007;110:9–17. doi: 10.1182/blood-2006-12-022038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Chunduri S, Mahmud D, Abbasian J, et al. Cord blood nucleated cells induce delayed T cell alloreactivity. Biol Blood Marrow Transplant. 2008;14:872–9. doi: 10.1016/j.bbmt.2008.05.009. [DOI] [PubMed] [Google Scholar]
- [41].Sorg RV, Kogler G, Wernet P. Identification of cord blood dendritic cells as an immature CD11c− population. Blood. 1999;93:2302–07. [PubMed] [Google Scholar]
- [42].Borràs FE, Matthews NC, Lowdell MW, et al. Identification of both myeloid CD11c+ and lymphoid CD11c− dendritic cell subsets in cord blood. Br J Haematol. 2001;113:925–31. doi: 10.1046/j.1365-2141.2001.02840.x. [DOI] [PubMed] [Google Scholar]
- [43].Kollmann TR, Crabtree J, Rein-Weston A, et al. Neonatal innate TLR-mediated responses are distinct from those of adults. J Immunol. 2009;183:7150–60. doi: 10.4049/jimmunol.0901481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Sandeep C, Dolores M, Javaneh A, et al. Cord blood nucleated cells induce delayed T cell alloreactivity. biology of blood and marrow transplantation. Journal of the American Society for Blood and Marrow Transplantation. 2008;14:872–79. doi: 10.1016/j.bbmt.2008.05.009. [DOI] [PubMed] [Google Scholar]
- [45].Liu E, Tu W, Law HKW, et al. Decreased yield, phenotypic expression and function of immature monocyte-derived dendritic cells in cord blood. Br J Haematol. 2001;113:240–46. doi: 10.1046/j.1365-2141.2001.02720.x. [DOI] [PubMed] [Google Scholar]
- [46].Rescigno M, Martino M, Sutherland CL, et al. Dendritic cell survival and maturation are regulated by different signaling pathways. J Exp Med. 1998;188:2175–80. doi: 10.1084/jem.188.11.2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811. doi: 10.1146/annurev.immunol.18.1.767. [DOI] [PubMed] [Google Scholar]
- [48].Martin-Fontecha A, Baumjohann D, Guarda G, et al. CD40L+ CD4+ memory T cells migrate in a CD62P-dependent fashion into reactive lymph nodes and license dendritic cells for T cell priming. J Exp Med. 2008;205:2561–74. doi: 10.1084/jem.20081212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Drohan L, Harding JJ, Holm B, et al. Selective developmental defects of cord blood antigen-presenting cell subsets. Human Immunology. 2004;65:1356–69. doi: 10.1016/j.humimm.2004.09.011. [DOI] [PubMed] [Google Scholar]
- [50].Goriely S, Vincart B, Stordeur P, et al. Deficient IL-12(p35) gene expression by dendritic cells derived from neonatal monocytes. J Immunol. 2001;166:2141–46. doi: 10.4049/jimmunol.166.3.2141. [DOI] [PubMed] [Google Scholar]
- [51].Langrish CL, Buddle JC, Thrasher AJ, et al. Neonatal dendritic cells are intrinsically biased against Th-1 immune responses. Clinical & Experimental Immunology. 2002;128:118–23. doi: 10.1046/j.1365-2249.2002.01817.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Naderi N, Pourfathollah AA, Alimoghaddam K, et al. Cord blood dendritic cells prevent the differentiation of naive T-helper cells towards Th1 irrespective of their subtype. Clin Exp Med. 2009;9:29–36. doi: 10.1007/s10238-008-0020-2. [DOI] [PubMed] [Google Scholar]
- [53].Arpinati M, Chirumbolo G, Urbini B, et al. Role of plasmacytoid dendritic cells in immunity and tolerance after allogeneic hematopoietic stem cell transplantation. Transplant Immunology. 11:345–56. doi: 10.1016/S0966-3274(03)00055-8. [DOI] [PubMed] [Google Scholar]
- [54].Crespo I, Paiva A, Couceiro A, et al. Immunophenotypic and functional characterization of cord blood dendritic cells. Stem Cells Dev. 2004;13:63–70. doi: 10.1089/154732804773099263. [DOI] [PubMed] [Google Scholar]
- [55].Mahnke K, Schmitt E, Bonifaz L, et al. Immature, but not inactive: the tolerogenic function of immature dendritic cells. Immunol Cell Biol. 2002;80:477–83. doi: 10.1046/j.1440-1711.2002.01115.x. [DOI] [PubMed] [Google Scholar]
- [56].Steinbrink K, Mahnke K, Grabbe S, et al. Myeloid dendritic cell: From sentinel of immunity to key player of peripheral tolerance? Human Immunology. 2009;70:289–93. doi: 10.1016/j.humimm.2009.02.003. [DOI] [PubMed] [Google Scholar]
- [57].Toubai T, Malter C, Tawara I, et al. Immunization with host-type CD8α+ dendritic cells reduces experimental acute GVHD in an IL-10-dependent manner. Blood. 2010;115:724–35. doi: 10.1182/blood-2009-06-229708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Kubach J, Becker C, Schmitt E, et al. Dendritic cells: Sentinels of immunity and tolerance. Int J Hematol. 2005;81:197–203. doi: 10.1532/IJH97.04165. [DOI] [PubMed] [Google Scholar]
- [59].Rutella S, Danese S, Leone G. Tolerogenic dendritic cells: cytokine modulation comes of age. Blood. 2006;108:1435–40. doi: 10.1182/blood-2006-03-006403. [DOI] [PubMed] [Google Scholar]
- [60].Chidgey AP, Layton D, Trounson A, et al. Tolerance strategies for stem-cell-based therapies. Nature. 2008;453:330–37. doi: 10.1038/nature07041. [DOI] [PubMed] [Google Scholar]
- [61].Roncarolo MG, Levings MK, Traversari C. Differentiation of T regulatory cells by immature dendritic cells. J Exp Med. 2001;193:F5–9. doi: 10.1084/jem.193.2.f5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Jonuleit H, Schmitt E, Schuler G, et al. Induction of interleukin 10-producing, nonproliferating CD4+ T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J Exp Med. 2000;192:1213–22. doi: 10.1084/jem.192.9.1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Yamazaki S, Inaba K, Tarbell KV, et al. Dendritic cells expand antigen-specific Foxp3+CD25+CD4+ regulatory T cells including suppressors of alloreactivity. Immunol Rev. 2006;212:314–29. doi: 10.1111/j.0105-2896.2006.00422.x. [DOI] [PubMed] [Google Scholar]
- [64].Dhodapkar MV, Steinman RM, Krasovsky J, et al. Antigen-Specific Inhibition of Effector T Cell Function in Humans after Injection of Immature Dendritic Cells. J Exp Med. 2001;193:233–38. doi: 10.1084/jem.193.2.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Jiang X-X, Zhang Y, Liu B, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105:4120–26. doi: 10.1182/blood-2004-02-0586. [DOI] [PubMed] [Google Scholar]
- [66].Saito S, Motoyoshi K, Ichijo M, et al. High serum human macrophage colony-stimulating factor level during pregnancy. Int J Hematol. 1992;55:219–25. [PubMed] [Google Scholar]
- [67].Mellor AL, Munn DH. Immunology at the maternal-fetal interface: lessons for T cell tolerance and suppression. Annu Rev Immunol. 2000;18:367–91. doi: 10.1146/annurev.immunol.18.1.367. [DOI] [PubMed] [Google Scholar]
- [68].Li G, Kim YJ, Broxmeyer HE. Macrophage colony-stimulating factor drives cord blood monocyte differentiation into IL-10highIL-12absent dendritic cells with tolerogenic potential. J Immunol. 2005;174:4706–17. doi: 10.4049/jimmunol.174.8.4706. [DOI] [PubMed] [Google Scholar]
- [69].Tolar J, Hippen KL, Blazar BR. Immune regulatory cells in umbilical cord blood: T regulatory cells and mesenchymal stromal cells. Br J Haematol. 2009;147:200–6. doi: 10.1111/j.1365-2141.2009.07781.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Sakaguchi S, Sakaguchi N, Asano M, et al. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor α-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–64. [PubMed] [Google Scholar]
- [71].Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–62. doi: 10.1146/annurev.immunol.21.120601.141122. [DOI] [PubMed] [Google Scholar]
- [72].Fontenot JD, Rasmussen JP, Williams LM, et al. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity. 2005;22:329–41. doi: 10.1016/j.immuni.2005.01.016. [DOI] [PubMed] [Google Scholar]
- [73].Dieckmann D, Plottner H, Berchtold S, et al. Ex vivo isolation and characterization of CD4+CD25+ T cells with regulatory properties from human blood. J Exp Med. 2001;193:1303–10. doi: 10.1084/jem.193.11.1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Shimizu J, Yamazaki S, Takahashi T, et al. Stimulation of CD25+CD4+ regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol. 2002;3:135–42. doi: 10.1038/ni759. [DOI] [PubMed] [Google Scholar]
- [75].Mchugh RS, Whitters MJ, Piccirillo CA, et al. CD4+CD25+ immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity. 2002;16:311–23. doi: 10.1016/s1074-7613(02)00280-7. [DOI] [PubMed] [Google Scholar]
- [76].Valzasina B, Guiducci C, Dislich H, et al. Triggering of OX40 (CD134) on CD4+CD25+ T cells blocks their inhibitory activity: a novel regulatory role for OX40 and its comparison with GITR. Blood. 2005;105:2845–51. doi: 10.1182/blood-2004-07-2959. [DOI] [PubMed] [Google Scholar]
- [77].Taylor PA, Panoskaltsis-Mortari A, Swedin Jm, et al. L-Selectinhi but not the L-selectinlo CD4+25+ T-regulatory cells are potent inhibitors of GVHD and BM graft rejection. Blood. 2004;104:3804–12. doi: 10.1182/blood-2004-05-1850. [DOI] [PubMed] [Google Scholar]
- [78].Ermann J, Hoffmann P, Edinger M, et al. Only the CD62L+ subpopulation of CD4+CD25+ regulatory T cells protects from lethal acute GVHD. Blood. 2005;105:2220–6. doi: 10.1182/blood-2004-05-2044. [DOI] [PubMed] [Google Scholar]
- [79].Wei S, Kryczek I, Zou W. Regulatory T-cell compartmentalization and trafficking. Blood. 2006;108:426–31. doi: 10.1182/blood-2006-01-0177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Iellem A, Mariani M, Lang R, et al. Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4+CD25+ regulatory T Cells. J Exp Med. 2001;194:847–54. doi: 10.1084/jem.194.6.847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Nakamura K, Kitani A, Fuss I, et al. TGF-β 1 plays an important role in the mechanism of CD4+CD25+ regulatory T cell activity in both humans and mice. J Immunol. 2004;172:834–42. doi: 10.4049/jimmunol.172.2.834. [DOI] [PubMed] [Google Scholar]
- [82].Oida T, Xu L, Weiner HL, et al. TGF-β-mediated suppression by CD4+CD25+ T cells is facilitated by CTLA-4 signaling. J Immunol. 2006;177:2331–9. doi: 10.4049/jimmunol.177.4.2331. [DOI] [PubMed] [Google Scholar]
- [83].Zheng SG, Wang J, Wang P, et al. IL-2 is essential for TGF-β to convert naive CD4+CD25− cells to CD25+Foxp3+ regulatory T cells and for expansion of these cells. J Immunol. 2007;178:2018–27. doi: 10.4049/jimmunol.178.4.2018. [DOI] [PubMed] [Google Scholar]
- [84].Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–38. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- [85].Korn T, Bettelli E, Gao W, et al. IL-21 initiates an alternative pathway to induce proinflammatory TH17 cells. Nature. 2007;448:484–7. doi: 10.1038/nature05970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Gao W, Lu Y, Essawy BE, et al. Contrasting Effects of cyclosporine and rapamycin in de novo generation of alloantigen-specific regulatory T cells. American Journal of Transplantation. 2007;7:1722–32. doi: 10.1111/j.1600-6143.2007.01842.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Sauer S, Bruno L, Hertweck A, et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc Natl Acad Sci U S A. 2008;105:7797–802. doi: 10.1073/pnas.0800928105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Raimondi G, Sumpter TL, Matta BM, et al. Mammalian target of rapamycin inhibition and alloantigen-specific regulatory T cells synergize to promote long-term graft survival in immunocompetent recipients. J Immunol. 2010;184:624–36. doi: 10.4049/jimmunol.0900936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Asano M, Toda M, Sakaguchi N, et al. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med. 1996;184:387–96. doi: 10.1084/jem.184.2.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Suri-Payer E, Amar AZ, Thornton AM, et al. CD4+CD25+ T Cells Inhibit both the induction and effector function of autoreactive T cells and represent a unique lineage of immunoregulatory cells. J Immunol. 1998;160:1212–18. [PubMed] [Google Scholar]
- [91].Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med. 1998;188:287–96. doi: 10.1084/jem.188.2.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Thornton AM, Shevach EM. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol. 2000;164:183–90. doi: 10.4049/jimmunol.164.1.183. [DOI] [PubMed] [Google Scholar]
- [93].Waldmann TA. The interleukin-2 receptor. J Biol Chem. 1991;266:2681–84. [PubMed] [Google Scholar]
- [94].Zola H, Fusco M, Macardle PJ, et al. Expression of cytokine receptors by human cord blood lymphocytes: comparison with adult blood lymphocytes. Pediatr Res. 1995;38:397–403. doi: 10.1203/00006450-199509000-00021. [DOI] [PubMed] [Google Scholar]
- [95].Taga K, Kasahara Y, Yachie A, et al. Preferential expression of IL-2 receptor subunits on memory populations within CD4+ and CD8+ T cells. Immunology. 1991;72:15–9. [PMC free article] [PubMed] [Google Scholar]
- [96].Baecher-Allan C, Brown JA, Freeman GJ, et al. CD4+CD25high regulatory cells in human peripheral blood. J Immunol. 2001;167:1245–53. doi: 10.4049/jimmunol.167.3.1245. [DOI] [PubMed] [Google Scholar]
- [97].Wing K, Ekmark A, Karlsson H, et al. Characterization of human CD25+CD4+ T cells in thymus,cord and adult blood. Immunology. 2002;106:190–99. doi: 10.1046/j.1365-2567.2002.01412.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
- [99].Roncador G, Brown PJ, Maestre L, et al. Analysis of FOXP3 protein expression in human CD4+CD25+ regulatory T cells at the single-cell level. Eur J Immunol. 2005;35:1681–91. doi: 10.1002/eji.200526189. [DOI] [PubMed] [Google Scholar]
- [100].Seddiki N, Santner-Nanan B, Martinson J, et al. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med. 2006;203:1693–700. doi: 10.1084/jem.20060468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Hoffmann P, Eder R, Boeld TJ, et al. Only the CD45RA+ subpopulation of CD4+CD25high T cells gives rise to homogeneous regulatory T-cell lines upon in vitro expansion. Blood. 2006;108:4260–7. doi: 10.1182/blood-2006-06-027409. [DOI] [PubMed] [Google Scholar]
- [102].Booth NJ, Mcquaid AJ, Sobande T, et al. Different proliferative potential and migratory characteristics of human CD4+ regulatory T cells that express either CD45RA or CD45RO. J Immunol. 2010;184:4317–26. doi: 10.4049/jimmunol.0903781. [DOI] [PubMed] [Google Scholar]
- [103].Godfrey WR, Spoden DJ, Ge YG, et al. Cord blood CD4+CD25+-derived T regulatory cell lines express FoxP3 protein and manifest potent suppressor function. Blood. 2005;105:750–8. doi: 10.1182/blood-2004-06-2467. [DOI] [PubMed] [Google Scholar]
- [104].Coenen JJA, Koenen HJPM, Van Rijssen E, et al. Rapamycin, and not cyclosporin A, preserves the highly suppressive CD27+ subset of human CD4+CD25+ regulatory T cells. Blood. 2006;107:1018–23. doi: 10.1182/blood-2005-07-3032. [DOI] [PubMed] [Google Scholar]
- [105].Ng WF, Duggan PJ, Ponchel F, et al. Human CD4+CD25+ cells: a naturally occurring population of regulatory T cells. Blood. 2001;98:2736–44. doi: 10.1182/blood.v98.9.2736. [DOI] [PubMed] [Google Scholar]
- [106].Lee C-C, Lin S-J, Cheng P-J, et al. The regulatory function of umbilical cord blood CD4+CD25+ T cells stimulated with anti-CD3/anti-CD28 and exogenous interleukin (IL)-2 or IL-15. Pediatric Allergy and Immunology. 2009;20:624–32. doi: 10.1111/j.1399-3038.2008.00843.x. [DOI] [PubMed] [Google Scholar]
- [107].Wing K, Lindgren S, Kollberg G, et al. CD4 T cell activation by myelin oligodendrocyte glycoprotein is suppressed by adult but not cord blood CD25+ T cells. Eur J Immunol. 2003;33:579–87. doi: 10.1002/eji.200323701. [DOI] [PubMed] [Google Scholar]
- [108].Chang CC, Satwani P, Oberfield N, et al. Increased induction of allogeneic-specific cord blood CD4+CD25+ regulatory T (Treg) cells: a comparative study of naive and antigenic-specific cord blood Treg cells. Exp Hematol. 2005;33:1508–20. doi: 10.1016/j.exphem.2005.09.002. [DOI] [PubMed] [Google Scholar]
- [109].Schaub B, Liu J, Hoppler S, et al. Maternal farm exposure modulates neonatal immune mechanisms through regulatory T cells. J Allergy Clin Immunol. 2009;123:774–82. e5. doi: 10.1016/j.jaci.2009.01.056. [DOI] [PubMed] [Google Scholar]
- [110].Wood KJ, Sakaguchi S. Regulatory T cells in transplantation tolerance. Nat Rev Immunol. 2003;3:199–210. doi: 10.1038/nri1027. [DOI] [PubMed] [Google Scholar]
- [111].Anderson BE, Mcniff JM, Matte C, et al. Recipient CD4+ T cells that survive irradiation regulate chronic graft-versus-host disease. Blood. 2004;104:1565–73. doi: 10.1182/blood-2004-01-0328. [DOI] [PubMed] [Google Scholar]
- [112].Zhao D, Zhang C, Yi T, et al. In vivo-activated CD103+CD4+ regulatory T cells ameliorate ongoing chronic graft-versus-host disease. Blood. 2008;112:2129–38. doi: 10.1182/blood-2008-02-140277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Sakaguchi S, Sakaguchi N, Shimizu J, et al. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev. 2001;182:18–32. doi: 10.1034/j.1600-065x.2001.1820102.x. [DOI] [PubMed] [Google Scholar]
- [114].Hanash AM, Levy RB. Donor CD4+CD25+ T cells promote engraftment and tolerance following MHC-mismatched hematopoietic cell transplantation. Blood. 2005;105:1828–36. doi: 10.1182/blood-2004-08-3213. [DOI] [PubMed] [Google Scholar]
- [115].Cohen JL, Trenado A, Vasey D, et al. CD4+CD25+ Immunoregulatory T cells: New therapeutics for graft-versus-host disease. J Exp Med. 2002;196:401–06. doi: 10.1084/jem.20020090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Taylor PA, Lees CJ, Blazar BR. The infusion of ex vivo activated and expanded CD4+CD25+ immune regulatory cells inhibits graft-versus-host disease lethality. Blood. 2002;99:3493–99. doi: 10.1182/blood.v99.10.3493. [DOI] [PubMed] [Google Scholar]
- [117].Chen X, Vodanovic-Jankovic S, Johnson B, et al. Absence of regulatory T-cell control of TH1 and TH17 cells is responsible for the autoimmune-mediated pathology in chronic graft-versus-host disease. Blood. 2007;110:3804–13. doi: 10.1182/blood-2007-05-091074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Oldenhove G, Bouladoux N, Wohlfert EA, et al. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity. 2009;31:772–86. doi: 10.1016/j.immuni.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Zeiser R, Nguyen VH, Beilhack A, et al. Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production. Blood. 2006;108:390–9. doi: 10.1182/blood-2006-01-0329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Hippen KL, Harker-Murray P, Porter SB, et al. Umbilical cord blood regulatory T-cell expansion and functional effects of tumor necrosis factor receptor family members OX40 and 4-1BB expressed on artificial antigen-presenting cells. Blood. 2008;112:2847–57. doi: 10.1182/blood-2008-01-132951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Godfrey WR, Ge YG, Spoden DJ, et al. In vitro-expanded human CD4+CD25+ T-regulatory cells can markedly inhibit allogeneic dendritic cell-stimulated MLR cultures. Blood. 2004;104:453–61. doi: 10.1182/blood-2004-01-0151. [DOI] [PubMed] [Google Scholar]
- [122].Junji T, Junichi S, Naoko K, et al. Expansion of natural killer cell receptor (CD94/NKG2A)–expressing cytolytic CD8 T cells and CD4+CD25+ regulatory T cells from the same cord blood unit. Exp Hematol. 2007;35:1562–66. doi: 10.1016/j.exphem.2007.06.006. [DOI] [PubMed] [Google Scholar]
- [123].Bacchetta R, Sartirana C, Levings MK, et al. Growth and expansion of human T regulatory type 1 cells are independent from TCR activation but require exogenous cytokines. Eur J Immunol. 2002;32:2237–45. doi: 10.1002/1521-4141(200208)32:8<2237::AID-IMMU2237>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
- [124].Koenen HJ, Fasse E, Joosten I. IL-15 and cognate antigen successfully expand de novo-induced human antigen-specific regulatory CD4+ T cells that require antigen-specific activation for suppression. J Immunol. 2003;171:6431–41. doi: 10.4049/jimmunol.171.12.6431. [DOI] [PubMed] [Google Scholar]
- [125].Zeiser R, Leveson-Gower DB, Zambricki EA, et al. Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells. Blood. 2008;111:453–62. doi: 10.1182/blood-2007-06-094482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Hutton JF, Gargett T, Sadlon TJ, et al. Development of CD4+CD25+FoxP3+ regulatory T cells from cord blood hematopoietic progenitor cells. J Leukoc Biol. 2009;85:445–51. doi: 10.1189/jlb.1008620. [DOI] [PubMed] [Google Scholar]
- [127].Martin CH, Woll PS, Ni Z, et al. Differences in lymphocyte developmental potential between human embryonic stem cell and umbilical cord blood-derived hematopoietic progenitor cells. Blood. 2008;112:2730–37. doi: 10.1182/blood-2008-01-133801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Abdallah BM, Kassem M. Human mesenchymal stem cells: from basic biology to clinical applications. Gene Ther. 2007;15:109–16. doi: 10.1038/sj.gt.3303067. [DOI] [PubMed] [Google Scholar]
- [129].Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell. 2008;2:313–9. doi: 10.1016/j.stem.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Kern S, Eichler H, Stoeve J, et al. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord Blood, or adipose tissue. Stem Cells. 2006;24:1294–301. doi: 10.1634/stemcells.2005-0342. [DOI] [PubMed] [Google Scholar]
- [131].Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
- [132].Cetrulo CLJ. Cord-blood mesenchymal stem cells and tissue engineering. Stem Cell Rev. 2006;2:163–8. doi: 10.1007/s12015-006-0023-x. [DOI] [PubMed] [Google Scholar]
- [133].Caplan A. Why are MSCs therapeutic? New data: new insight. J Pathol. 2009;217:318–24. doi: 10.1002/path.2469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Phinney DG, Prockop DJ. Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair. Stem Cells. 2007;25:2896–902. doi: 10.1634/stemcells.2007-0637. [DOI] [PubMed] [Google Scholar]
- [135].Schugar RC, Chirieleison SM, Wescoe KE, et al. High harvest yield, high expansion, and phenotype stability of CD146 mesenchymal stromal cells from whole primitive human umbilical cord tissue. J Biomed Biotechnol. 2009;2009:789526. doi: 10.1155/2009/789526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Prockop DJ. Repair of tissues by adult stem/progenitor cells (MSCs): Controversies, myths, and changing paradigms. Mol Ther. 2009;17:939–46. doi: 10.1038/mt.2009.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Wang M, Yang Y, Yang D, et al. The immunomodulatory activity of human umbilical cord blood-derived mesenchymal stem cells in vitro. Immunology. 2009;126:220–32. doi: 10.1111/j.1365-2567.2008.02891.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Ren G, Zhang L, Zhao X, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–50. doi: 10.1016/j.stem.2007.11.014. [DOI] [PubMed] [Google Scholar]
- [139].Glennie S, Soeiro I, Dyson PJ, et al. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood. 2005;105:2821–27. doi: 10.1182/blood-2004-09-3696. [DOI] [PubMed] [Google Scholar]
- [140].Maitra B, Szekely E, Gjini K, et al. Human mesenchymal stem cells support unrelated donor hematopoietic stem cells and suppress T-cell activation. Bone Marrow Transplant. 2004;33:597–604. doi: 10.1038/sj.bmt.1704400. [DOI] [PubMed] [Google Scholar]
- [141].Keating A. Mesenchymal stromal cells. Curr Opin Hematol. 2006;13:419–25. doi: 10.1097/01.moh.0000245697.54887.6f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Rasmusson I, Ringdén O, Sundberg B, et al. Mesenchymal stem cells inhibit lymphocyte proliferation by mitogens and alloantigens by different mechanisms. Exp Cell Res. 2005;305:33–41. doi: 10.1016/j.yexcr.2004.12.013. [DOI] [PubMed] [Google Scholar]
- [143].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;35:1482–90. doi: 10.1002/eji.200425405. [DOI] [PubMed] [Google Scholar]
- [144].Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22. doi: 10.1182/blood-2004-04-1559. [DOI] [PubMed] [Google Scholar]
- [145].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;30:42–8. doi: 10.1016/s0301-472x(01)00769-x. [DOI] [PubMed] [Google Scholar]
- [146].Sato K, Ozaki K, Oh I, et al. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 2007;109:228–34. doi: 10.1182/blood-2006-02-002246. [DOI] [PubMed] [Google Scholar]
- [147].Beyth S, Borovsky Z, Mevorach D, et al. Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood. 2005;105:2214–19. doi: 10.1182/blood-2004-07-2921. [DOI] [PubMed] [Google Scholar]
- [148].Tisato V, Naresh K, Girdlestone J, et al. Mesenchymal stem cells of cord blood origin are effective at preventing but not treating graft-versus-host disease. Leukemia. 2007;21:1992–99. doi: 10.1038/sj.leu.2404847. [DOI] [PubMed] [Google Scholar]
- [149].Romieu-Mourez R, Francois M, Boivin M-N, et al. Regulation of MHC class II expression and antigen processing in murine and human mesenchymal stromal cells by IFN-γ, TGF-β, and cell density. J Immunol. 2007;179:1549–58. doi: 10.4049/jimmunol.179.3.1549. [DOI] [PubMed] [Google Scholar]
- [150].Batten P, Sarathchandra P, Antoniw Jw, et al. Human 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. 2006;12:2263–73. doi: 10.1089/ten.2006.12.2263. [DOI] [PubMed] [Google Scholar]
- [151].Groh ME, Maitra B, Szekely E, et al. Human mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells. Exp Hematol. 2005;33:928–34. doi: 10.1016/j.exphem.2005.05.002. [DOI] [PubMed] [Google Scholar]
- [152].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;103:4619–21. doi: 10.1182/blood-2003-11-3909. [DOI] [PubMed] [Google Scholar]
- [153].Chen K, Wang D, Du WT, et al. Human umbilical cord mesenchymal stem cells hUC-MSCs exert immunosuppressive activities through a PGE2-dependent mechanism. Clin Immunol. 2010 doi: 10.1016/j.clim.2010.01.015. [DOI] [PubMed] [Google Scholar]
- [154].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 & Rheumatism. 2005;52:1595–603. doi: 10.1002/art.21012. [DOI] [PubMed] [Google Scholar]
- [155].Gonzalez MA, Gonzalez-Rey E, Rico L, et al. Treatment of experimental arthritis by inducing immune tolerance with human adipose-derived mesenchymal stem cells. Arthritis Rheum. 2009;60:1006–19. doi: 10.1002/art.24405. [DOI] [PubMed] [Google Scholar]
- [156].Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109:235–42. doi: 10.1046/j.1365-2141.2000.01986.x. [DOI] [PubMed] [Google Scholar]
- [157].Lee Ok, Kuo TK, Chen W-M, et al. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103:1669–75. doi: 10.1182/blood-2003-05-1670. [DOI] [PubMed] [Google Scholar]
- [158].Goodwin HS, Bicknese AR, Chien SN, et al. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow Transplant. 2001;7:581–8. doi: 10.1053/bbmt.2001.v7.pm11760145. [DOI] [PubMed] [Google Scholar]
- [159].Bieback K, Kern S, Klüter H, et al. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord bood. Stem Cells. 2004;22:625–34. doi: 10.1634/stemcells.22-4-625. [DOI] [PubMed] [Google Scholar]
- [160].Secco M, Zucconi E, Vieira NM, et al. Multipotent stem cells from umbilical cord: Cord is richer than blood. Stem Cells. 2008;26:146–50. doi: 10.1634/stemcells.2007-0381. [DOI] [PubMed] [Google Scholar]
- [161].Wexler SA, Donaldson C, Denning-Kendall P, et al. Adult bone marrow is a rich source of human mesenchymal ‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol. 2003;121:368–74. doi: 10.1046/j.1365-2141.2003.04284.x. [DOI] [PubMed] [Google Scholar]
- [162].Mareschi K, Biasin E, Piacibello W, et al. Isolation of human mesenchymal stem cells: bone marrow versus umbilical cord blood. Haematologica. 2001;86:1099–100. [PubMed] [Google Scholar]
- [163].Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: Candidate MSC-like cells from umbilical cord. Stem Cells. 2003;21:105–10. doi: 10.1634/stemcells.21-1-105. [DOI] [PubMed] [Google Scholar]
- [164].Wang H-S, Hung S-C, Peng S-T, et al. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells. 2004;22:1330–37. doi: 10.1634/stemcells.2004-0013. [DOI] [PubMed] [Google Scholar]
- [165].Lu LL, Liu YJ, Yang SG, et al. Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis-supportive function and other potentials. Haematologica. 2006;91:1017–26. [PubMed] [Google Scholar]
- [166].Boissel L, Tuncer HH, Betancur M, et al. Umbilical cord mesenchymal stem cells increase expansion of cord blood natural killer cells. Biol Blood Marrow Transplant. 2008;14:1031–8. doi: 10.1016/j.bbmt.2008.06.016. [DOI] [PubMed] [Google Scholar]
- [167].Secco M, Moreira YB, Zucconi E, et al. Gene expression profile of mesenchymal stem cells from paired umbilical cord units: cord is different from blood. Stem Cell Rev. 2009;5:387–401. doi: 10.1007/s12015-009-9098-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–13. doi: 10.1016/j.stem.2008.07.003. [DOI] [PubMed] [Google Scholar]
- [169].Can A, Karahuseyinoglu S. Concise review: Human umbilical cord stroma with regard to the source of fetus-derived stem cells. Stem Cells. 2007;25:2886–95. doi: 10.1634/stemcells.2007-0417. [DOI] [PubMed] [Google Scholar]
- [170].Weiss ML, Medicetty S, Bledsoe AR, et al. Human umbilical cord matrix stem cells: preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells. 2006;24:781–92. doi: 10.1634/stemcells.2005-0330. [DOI] [PubMed] [Google Scholar]
- [171].Weiss ML, Troyer DL. Stem cells in the umbilical cord. Stem Cell Rev. 2006;2:155–62. doi: 10.1007/s12015-006-0022-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Covas DT, Panepucci RA, Fontes Am, et al. Multipotent mesenchymal stromal cells obtained from diverse human tissues share functional properties and gene-expression profile with CD146+ perivascular cells and fibroblasts. Exp Hematol. 2008;36:642–54. doi: 10.1016/j.exphem.2007.12.015. [DOI] [PubMed] [Google Scholar]
- [173].Meirelles LDS, Caplan AI, Nardi NB. In search of the in vivo identity of mesenchymal stem cells. Stem Cells. 2008;26:2287–99. doi: 10.1634/stemcells.2007-1122. [DOI] [PubMed] [Google Scholar]
- [174].Sarugaser R, Lickorish D, Baksh D, et al. Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells. 2005;23:220–9. doi: 10.1634/stemcells.2004-0166. [DOI] [PubMed] [Google Scholar]
- [175].Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells. 2007;25:1384–92. doi: 10.1634/stemcells.2006-0709. [DOI] [PubMed] [Google Scholar]
- [176].Panepucci RA, Siufi JLC, Silva WA, et al. Comparison of gene expression of umbilical cord vein and bone marrow-derived mesenchymal stem cells. Stem Cells. 2004;22:1263–78. doi: 10.1634/stemcells.2004-0024. [DOI] [PubMed] [Google Scholar]
- [177].Friedman R, Betancur M, Boissel L, et al. Umbilical cord mesenchymal stem cells: adjuvants for human cell transplantation. Biol Blood Marrow Transplant. 2007;13:1477–86. doi: 10.1016/j.bbmt.2007.08.048. [DOI] [PubMed] [Google Scholar]
- [178].Park IH, Arora N, Huo H, et al. Disease-specific induced pluripotent stem cells. Cell. 2008;134:877–86. doi: 10.1016/j.cell.2008.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Broxmeyer HE. Will iPS cells enhance therapeutic applicability of cord blood cells and banking? Cell Stem Cell. 2010;6:21–4. doi: 10.1016/j.stem.2009.12.008. [DOI] [PubMed] [Google Scholar]
- [180].Feldmann REJ, Bieback K, Maurer MH, et al. Stem cell proteomes: A profile of human mesenchymal stem cells derived from umbilical cord blood. Electrophoresis. 2005;26:2749–58. doi: 10.1002/elps.200410406. [DOI] [PubMed] [Google Scholar]
- [181].Liu C-H, Hwang S-M. Cytokine interactions in mesenchymal stem cells from cord blood. Cytokine. 2005;32:270–79. doi: 10.1016/j.cyto.2005.11.003. [DOI] [PubMed] [Google Scholar]
- [182].Yoo KH, Jang IK, Lee MW, et al. Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues. Cell Immunol. 2009;259:150–6. doi: 10.1016/j.cellimm.2009.06.010. [DOI] [PubMed] [Google Scholar]
- [183].Bieback K, Kluter H. Mesenchymal stromal cells from umbilical cord blood. Curr Stem Cell Res Ther. 2007;2:310–23. doi: 10.2174/157488807782793763. [DOI] [PubMed] [Google Scholar]
- [184].Bieback K, Kern S, Kocaomer A, et al. Comparing mesenchymal stromal cells from different human tissues: bone marrow, adipose tissue and umbilical cord blood. Biomed Mater Eng. 2008;18:S71–6. [PubMed] [Google Scholar]
- [185].Rebelatto CK, Aguiar AM, Moretao MP, et al. Dissimilar differentiation of mesenchymal stem cells from bone marrow, umbilical cord blood, and adipose tissue. Exp. Biol. Med. 2008;233:901–13. doi: 10.3181/0712-RM-356. [DOI] [PubMed] [Google Scholar]
- [186].Kakinuma S, Tanaka Y, Chinzei R, et al. Human umbilical cord blood as a source of transplantable hepatic progenitor cells. Stem Cells. 2003;21:217–27. doi: 10.1634/stemcells.21-2-217. [DOI] [PubMed] [Google Scholar]
- [187].Kögler 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;200:123–35. doi: 10.1084/jem.20040440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Kogler G, Sensken S, Wernet P. Comparative generation and characterization of pluripotent unrestricted somatic stem cells with mesenchymal stem cells from human cord blood. Exp Hematol. 2006;34:1589–95. doi: 10.1016/j.exphem.2006.07.011. [DOI] [PubMed] [Google Scholar]
- [189].Mcguckin CP, Forraz N, Baradez M-O, et al. Production of stem cells with embryonic characteristics from human umbilical cord blood. Cell Proliferation. 2005;38:245–55. doi: 10.1111/j.1365-2184.2005.00346.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Kucia M, Halasa M, Wysoczynski M, et al. Morphological and molecular characterization of novel population of CXCR4+ SSEA-4+ Oct-4+ very small embryonic-like cells purified from human cord blood - preliminary report. Leukemia. 2006;21:297–303. doi: 10.1038/sj.leu.2404470. [DOI] [PubMed] [Google Scholar]
- [191].Mueller SM, Glowacki J. Age-related decline in the osteogenic potential of human bone marrow cells cultured in three-dimensional collagen sponges. J Cell Biochem. 2001;82:583–90. doi: 10.1002/jcb.1174. [DOI] [PubMed] [Google Scholar]
- [192].Stenderup K, Justesen J, Clausen C, et al. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone. 2003;33:919–26. doi: 10.1016/j.bone.2003.07.005. [DOI] [PubMed] [Google Scholar]
- [193].Rao MS, Mattson MP. Stem cells and aging: expanding the possibilities. Mechanisms of Ageing and Development. 2001;122:713–34. doi: 10.1016/s0047-6374(01)00224-x. [DOI] [PubMed] [Google Scholar]
- [194].Zappia E, Casazza S, Pedemonte E, et al. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood. 2005;106:1755–61. doi: 10.1182/blood-2005-04-1496. [DOI] [PubMed] [Google Scholar]
- [195].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;11:389–98. doi: 10.1016/j.bbmt.2005.02.001. [DOI] [PubMed] [Google Scholar]
- [196].Ringden O, Uzunel M, Rasmusson I, et al. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation. 2006;81:1390–97. doi: 10.1097/01.tp.0000214462.63943.14. [DOI] [PubMed] [Google Scholar]
- [197].Sandhu JS, Gorczynski R, Shpitz B, et al. A human model of xenogeneic graft-versus-host disease in SCID mice engrafted with human peripheral blood lymphocytes. Transplantation. 1995;60:179–84. [PubMed] [Google Scholar]
- [198].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;176:7761–67. doi: 10.4049/jimmunol.176.12.7761. [DOI] [PubMed] [Google Scholar]
- [199].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;108:2114–20. doi: 10.1182/blood-2005-11-011650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Eliopoulos N, Stagg J, Lejeune L, et al. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood. 2005;106:4057–65. doi: 10.1182/blood-2005-03-1004. [DOI] [PubMed] [Google Scholar]
- [201].Chung NG, Jeong DC, Park SJ, et al. Cotransplantation of marrow stromal cells may prevent lethal graft-versus-host disease in major histocompatibility complex mismatched murine hematopoietic stem cell transplantation. Int J Hematol. 2004;80:370–6. doi: 10.1532/ijh97.a30409. [DOI] [PubMed] [Google Scholar]
- [202].Von Bonin M, Stolzel F, Goedecke A, et al. Treatment of refractory acute GVHD with third-party MSC expanded in platelet lysate-containing medium. Bone Marrow Transplant. 2008;43:245–51. doi: 10.1038/bmt.2008.316. [DOI] [PubMed] [Google Scholar]
- [203].Zhou H, Guo M, Bian C, et al. Efficacy of bone marrow-derived mesenchymal stem cells in the treatment of sclerodermatous chronic graft-versus-host disease: Clinical Report. Biology of Blood and Marrow Transplantation. 2010;16:403–12. doi: 10.1016/j.bbmt.2009.11.006. [DOI] [PubMed] [Google Scholar]
- [204].Gonzalo-Daganzo R, Regidor C, Martin-Donaire T, et al. Results of a pilot study on the use of third-party donor mesenchymal stromal cells in cord blood transplantation in adults. Cytotherapy. 2009;11:278–88. doi: 10.1080/14653240902807018. [DOI] [PubMed] [Google Scholar]
- [205].Le Blanc K, Ringdén O. Mesenchymal stem cells: properties and role in clinical bone marrow transplantation. Curr Opin Immunol. 2006;18:586–91. doi: 10.1016/j.coi.2006.07.004. [DOI] [PubMed] [Google Scholar]
- [206].Olson JA, Leveson-Gower DB, Gill S, et al. NK cells mediate reduction of GVHD by inhibiting activated, alloreactive T cells while retaining GVT effects. Blood. 2010 doi: 10.1182/blood-2009-05-222190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Xu W, Fazekas G, Hara H, et al. Mechanism of natural killer (NK) cell regulatory role in experimental autoimmune encephalomyelitis. J Neuroimmunol. 2005;163:24–30. doi: 10.1016/j.jneuroim.2005.02.011. [DOI] [PubMed] [Google Scholar]
- [208].Rabinovich BA, Li J, Shannon J, et al. Activated, but not resting, T cells can be recognized and killed by syngeneic NK cells. J Immunol. 2003;170:3572–76. doi: 10.4049/jimmunol.170.7.3572. [DOI] [PubMed] [Google Scholar]
- [209].Gaddy J, Broxmeyer HE. Cord blood CD16+56− cells with low lytic activity are possible precursors of mature natural killer cells. Cell Immunol. 1997;180:132–42. doi: 10.1006/cimm.1997.1175. [DOI] [PubMed] [Google Scholar]
- [210].Fan YY, Yang BY, Wu CY. Phenotypic and functional heterogeneity of natural killer cells from umbilical cord blood mononuclear cells. Immunol Invest. 2008;37:79–96. doi: 10.1080/10826080701690714. [DOI] [PubMed] [Google Scholar]
- [211].Dalle J-H, Menezes J, Wagner E, et al. Characterization of cord blood natural killer cells: Implications for transplantation and neonatal infections. Pediatric Research. 2005;57:649–55. doi: 10.1203/01.PDR.0000156501.55431.20. [DOI] [PubMed] [Google Scholar]
- [212].Wang Y, Xu H, Zheng X, et al. High expression of NKG2A/CD94 and low expression of granzyme B are associated with reduced cord blood NK cell activity. Cell Mol Immunol. 2007;4:377–82. [PubMed] [Google Scholar]
- [213].Lu X, Kondo Y, Takamatsu H, et al. CD16+ CD56− NK cells in the peripheral blood of cord blood transplant recipients: a unique subset of NK cells possibly associated with graft-versus-leukemia effect. Eur J Haematol. 2008;81:18–25. doi: 10.1111/j.1600-0609.2008.01073.x. [DOI] [PubMed] [Google Scholar]
- [214].Mavilio D, Lombardo G, Benjamin J, et al. Characterization of CD56−/CD16+ natural killer (NK) cells: A highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc Natl Acad Sci U S A. 2005;102:2886–91. doi: 10.1073/pnas.0409872102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Gonzalez VD, Falconer K, Michaëlsson J, et al. Expansion of CD56− NK cells in chronic HCV/HIV-1 co-infection: Reversion by antiviral treatment with pegylated IFNα and ribavirin. Clin Immunol. 2008;128:46–56. doi: 10.1016/j.clim.2008.03.521. [DOI] [PubMed] [Google Scholar]
- [216].Zarife MASA, Reis EAG, Carmo TMA, et al. Increased frequency of CD56BrightNK−cells, CD3− CD16+CD56− NK−cells and activated CD4+T-cells or B-cells in parallel with CD4+CDC25High T-cells control potentially viremia in blood donors with HCV. J Med Virol. 2009;81:49–59. doi: 10.1002/jmv.21340. [DOI] [PubMed] [Google Scholar]
- [217].Barker JN, Weisdorf DJ, Defor TE, et al. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005;105:1343–47. doi: 10.1182/blood-2004-07-2717. [DOI] [PubMed] [Google Scholar]
- [218].Majhail NS, Brunstein CG, Wagner JE. Double umbilical cord blood transplantation. Curr Opin Immunol. 2006;18:571–75. doi: 10.1016/j.coi.2006.07.015. [DOI] [PubMed] [Google Scholar]
- [219].Brunstein CG, Barker JN, Weisdorf DJ, et al. Umbilical cord blood transplantation after nonmyeloablative conditioning: impact on transplantation outcomes in 110 adults with hematologic disease. Blood. 2007;110:3064–70. doi: 10.1182/blood-2007-04-067215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [220].Ballen KK, Spitzer TR, Yeap BY, et al. Double unrelated reduced-intensity umbilical cord blood transplantation in adults. Biol Blood Marrow Transplant. 2007;13:82–9. doi: 10.1016/j.bbmt.2006.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Christopherson KW, Hangoc G, Mantel CR, et al. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science. 2004;305:1000–3. doi: 10.1126/science.1097071. [DOI] [PubMed] [Google Scholar]
- [222].Campbell TB, Hangoc G, Liu Y, et al. Inhibition of CD26 in human cord blood CD34+ cells enhances their engraftment of nonobese diabetic/severe combined immunodeficiency mice. Stem Cells Dev. 2007;16:347–54. doi: 10.1089/scd.2007.9995. [DOI] [PubMed] [Google Scholar]
- [223].Campbell TB, Broxmeyer HE. CD26 inhibition and hematopoiesis: a novel approach to enhance transplantation. Front Biosci. 2008;13:1795–805. doi: 10.2741/2800. [DOI] [PubMed] [Google Scholar]