Cell Transplantation, Vol. 24, pp. 133–149, 2015 Printed in the USA. All rights reserved. Copyright © 2015 Cognizant Comm. Corp.

0963-6897/15 $90.00 + .00 DOI: http://dx.doi.org/10.3727/096368913X675728 E-ISSN 1555-3892 www.cognizantcommunication.com

Review Recent Advances in Mesenchymal Stem Cell Immunomodulation: The Role of Microvesicles Alessandra Fierabracci,* Andrea Del Fattore,* Rosa Luciano,* Marta Muraca,† Anna Teti,‡ and Maurizio Muraca§ *Research Laboratories, Bambino Gesù Children’s Hospital, Rome, Italy †School of Medicine, University “Sapienza”, Rome, Italy ‡Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy §Department of Women's and Children's Health, University of Padua, Padua, Italy

Mesenchymal stem cells are the most widely used cell phenotype for therapeutic applications, the main reasons being their well-established abilities to promote regeneration of injured tissues and to modulate immune responses. Efficacy was reported in the treatment of several animal models of inflammatory and autoimmune diseases and, in clinical settings, for the management of disorders such as GVHD, systemic lupus erythematosus, multiple sclerosis, and inflammatory bowel disease. The effects of mesenchymal stem cells are believed to be largely mediated by paracrine signals, and several secreted molecules have been identified as contributors to the net biological effect. Recently, it has been recognized that bioactive molecules can be shuttled from cell to cell packed in microvesicles, tiny portions of cytoplasm surrounded by a membrane. Coding and noncoding RNAs are also carried in such microvesicles, transferring relevant biological activity to target cells. Several reports indicate that the regenerative effect of mesenchymal stem cells can be reproduced by microvesicles isolated from their culture medium. More recent evidence suggests that the immunomodulatory effects of mesenchymal stem cells are also at least partially mediated by secreted microvesicles. These findings allow better understanding of the mechanisms involved in cell-to-cell interaction and may have interesting implications for the development of novel therapeutic tools in place of the parent cells. Key words: Microvesicles; Exosomes; Mesenchymal stem cells; Immunity

INTRODUCTION Bone marrow is a complex organ where different resident lineages of hematopoietic and stromal cells support hematopoiesis (50). Mesenchymal stem (or stromal) cells (MSCs) represent a subpopulation of undifferentiated cells with the hallmark properties of self-renewal and differentiation. This population, first described in the 1960s as bone-forming cells (59), is one of the most widely investigated with still unexplored properties and potential for clinical application. The proper definition of MSCs has been extremely challenging. Initially identified as a subgroup of cells with osteogenic potential, they were subsequently recognized as fundamental players in the process of hematopoiesis (57). Researchers were attracted by their ability of differentiating toward the mesodermal lineage in adipogenic, chondrogenic, and osteogenic cells

emphasizing their potential in tissue regeneration, although extensive debate persists regarding their multipotential capacity in vitro (57). A particular challenge has been the absence of specific markers to define this plastic-adherent population with fibroblast-like morphology, despite the large number of proposed determinants (84), that is, cell surface expression of cluster of differentiation 105 (CD105; endoglin, SH2), CD73 (ecto-5¢-nucleotidase), and CD90 (thymocyate antigen 1; Thy1) and the absence of hematopoietic markers CD45, CD19 or CD79, CD14 or CD11b, and HLA (human leucocyte antigen)-DR. The International Society for Cellular Therapy has established the minimal criteria for defining multipotent MSCs (49). Of note, MSCs could also be derived from adipose tissue (102,188), placental tissue (80), skin (148), dental pulp (64), umbilical cord blood (54), amniotic fluid (120), umbilical cord

Received July 11, 2013; final acceptance November 20, 2013. Online prepub date: November 21, 2013. Address correspondence to Alessandra Fierabracci, Ph.D., Research Laboratories, Children’s Hospital Bambino Gesù, Piazza Sant’Onofrio 4, 00165 Rome, Italy. Tel: +39 06 6859 2656; Fax: +39 06 6859 2904; E-mail: [email protected]

133

134

perivascular cells (144), Wharton’s jelly (174), synovial tissue (45), and breast milk (70,125). MSCs IN REGENERATIVE MEDICINE Currently, 167 actively recruiting, MSC-based interventional clinical studies are registered at the NIH site (http://clinicaltrials.gov/). The present review will address more in detail the use of MSCs in the treatment of immune disorders and provide a concise overview of MSC-based cell therapy in tissue regeneration. For a more complete discussion of the latter topic, the reader is referred to excellent recent reviews [see, e.g., (84,139)]. However, the above distinction is largely artificial, since consolidated experimental evidence indicates that the therapeutic effects result from an interplay of different mechanisms including immunomodulation, protection from injury, and regenerative stimuli. Moreover, a better understanding of the interactions between administered MSCs and local microenvironments is a prerequisite for the development of more successful treatments. In immune disorders, the therapeutic efficacy of MSCs probably depends on the nature of different diseases because of the distinct inflammatory environments. Even for one specific disease, the variety of microenvironments in different tissues may result in different levels of efficacy. For instance, a recent trial on graft-versus-host disease (GVHD) showed significant effects of MSC treatment only on gastrointestinal and hepatic involvement (112). Clinical trials of MSCs in tissue regeneration have largely addressed heart and limb ischemia, bone defect and trauma repair, and neurodegenerative diseases. In a fundamental study, MSCs were administered after acute myocardial infarction in 53 patients, showing the safety of the procedure and providing preliminary efficacy data (68), including a significant reduction of ventricular arrhythmias and improvement in pulmonary function. Moreover, improved left ventricular function was achieved in a subset of patients receiving MSCs compared with the placebo group. These results provided the basis for further phase II–III clinical trials. Several issues are being addressed in designing present and forthcoming clinical trials using MSCs for heart repair, including timing of MSC administration, route of delivery, patient selection, and (last but not least) the efficacy index (37), since improvement in ventricular ejection fraction may not correlate with clinical outcome. MSCs need to be combined with a suitable scaffold in order to provide efficient bone regeneration, especially with large bone defects. Such a tissue-engineering approach provides abundant callus formation and good integration of the implants in long-term follow-up (133). A combination with scaffolds was also used in clinical trials for the treatment of osteoarthritis with MSCs, resulting in symptomatic relief and promotion of cartilage repair after intra-articular injection (3,44,74,82,173). Various MSC-based clinical studies have been carried out with patients suffering from

FIERABRACCI ET AL.

neurological disorders. Preliminary encouraging results have been reported in stroke (73), multiple sclerosis, and amyotrophic lateral sclerosis (33,42,83). Chronic skin ulcers represent a common pathology where effective cell therapy is much waited for, due to insufficient results of current conventional treatments, also involving significant nurse care time. In August 2013, a press release announced the results of a randomized, controlled clinical trial in patients with chronic diabetic foot ulcers comparing the safety and effectiveness of a human cellular repair matrix including living MSCs (Grafix®) to standard of care. For the primary endpoint, 62% of patients receiving Grafix had complete wound closure compared to 21% (p < 0.0001) of patients who received conventional treatment. The interim analysis conducted on the first 97 enrolled patients met the prespecified stopping rules for proven efficacy as determined by the data monitoring committee (4). IMMUNOMODULATION BY MSCs In Vitro Studies MSCs are immunoprivileged due to the low expression of class II major histocompatibility complex (MHC II) and costimulatory molecules CD40, CD40L, CD80, CD86 on the cell surface, thus escaping T-cell recognition (24). They also interfere with different pathways both through cell–cell interaction and secretion of soluble factors. MSCs can alter the response of both the adaptive and the innate immune systems by suppressing T-cell and dendritic cell maturation, affecting B-cell activation and proliferation, inhibiting proliferation and cytotoxicity of natural killer (NK) cells, and inducing regulatory T-cells (Tregs) (84). MSCs were shown to modulate T-cell proliferation and activation (181,187) in different experimental protocols. Di Nicola et al. (47) found that both autologous and allogeneic bone marrow stromal cells (BMSCs) reduced T-cell proliferation in mixed lymphocyte reactions (MLRs) with allogeneic dendritic cells (DCs) or peripheral blood lymphocytes (PBLs). When human MSCs were added in cocultures with purified subpopulations of immune cells, the cytokine secretion profile of DCs, naive and effector T helper 1 (Th1), Th2, and NK cells was altered, resulting in a more anti-inflammatory phenotype, while increasing the proportion of Tregs (2). Furthermore, MSCs inhibited the division of stimulated T-cells avoiding their entry into the S phase of the cell cycle and inducing irreversible G0/ G1 phase arrest (62). Work by Beyth et al. (19) supports an immunoregulatory mechanism wherein MSCs inhibit T-cells indirectly by contact-dependent induction of regulatory antigen-presenting cells (APCs) with T-cell-suppressive properties. Additional studies suggest that T-cell inhibition is not antigen specific, but acting through HLA both on primary and secondary responses (90,181). There is increasing interest in the effect of MSCs on B lymphocytes, since these cells are increasingly recognized

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

to play an important role in autoimmune disorders and in transplant rejection. The immunomodulatory effect of MSCs on B-cells is still controversial, and the mechanisms involved are unclear. The role of soluble factors is generally recognized, and it was even found to be more relevant in mediating the effect on B than on T lymphocytes (12). When cocultured with purified B-cells, MSCs were reported to have different and even opposite effects in different experimental conditions. MSCs inhibited Bcell proliferation and antibody production in vitro in the presence of cytosine–phosphate–guanosine (CpG), CD40L, anti-immunoglobulin, interleukin (IL)-2, and IL-4 (43), while they had no effect after B-cell stimulation by a combination of allogeneic T-cell-depleted peripheral blood mononuclear cells (PBMCs) and CpG (90). In the latter work, B-cells became susceptible to the suppressive activity of MSCs in the presence of exogenously added interferon-g (IFN-g). On the other hand, in the work of Traggiai et al. (164), MSCs promoted proliferation and differentiation into immunoglobulin-secreting cells of transitional and naive B-cells stimulated with CpG, in the absence of B-cell receptor triggering. Using bacterial/viral antigen stimulation of PBMCs, Rasmusson et al. (136) demonstrated MSC-dependent induction or inhibition of antibody secretion in B-cells depending on the strength of the baseline stimulus, inhibition being associated with a stronger stimulation. A perhaps more consistent MSC-mediated inhibition of B lymphocyte function was observed where T-helper effect on B-cell function is maintained such as in the work of Comoli et al. (41) or in PBMC cocultures (22,141). Collectively, these findings suggest that MSC-mediated inhibition of B lymphocyte function involves T-helper suppression rather than a direct effect on B lymphocytes, given also the observation that Ig production is not inhibited by MSCs when CD40 engagement is obtained by agonist anti-CD40L antibody rather than by the direct help of T-cells (41). MSCs induce inhibition of differentiation of monocytes to immature DCs (121,181). MSCs may also affect NK cell function by downregulating IFN-g secretion (2). Contrasting data were reported by Rasmusson et al. (137), suggesting that MSCs do not affect NK cell or cytotoxic T lymphocyte (CTL) lysis. Such discrepancies may result from the different experimental settings, since it seems that MSCs can inhibit T-cells in the early activation phase, but not in the effector phase of allograft rejection (137). Similarly, the contrasting results on NK cell lysis may be related to the timing of the experiment, since the late lytic phase of NK cells may be mediated by cell surface molecules that are not affected by MSCs. Of note, MSCs significantly inhibited IL2-stimulated proliferation on resting NK cells (149), while the proliferation of activated NK cells was only partially affected. Soluble mediators of MSC immunomodulatory effects have been identified in several studies. MSCs inhibit T-cell

135

proliferation by decreasing both IFN-g and tumor necrosis factor-a (TNF-a) production, while increasing IL-10 secretion (23,181). Also, in MLRs, addition of MSCs stimulates indoleamine 2,3-dioxygenase (IDO) activity, resulting in the degradation of tryptophan and inhibition of T-cell proliferation. Concerted actions of chemokines and nitric oxide (NO) were shown to mediate the immunosuppressive effect on T-cell proliferation and cytokine production (145). Species differences in the relative contribution of inducible nitric oxide synthase (iNOS) and IDO in suppressing T-cell function have been described in vitro with mouse and human MSCs (140). Soluble factors are believed to mediate the inhibition of B-cell proliferation and differentiation by MSCs, but these mechanisms have been less characterized (12). MSCs secrete prostaglandin E2 (PGE2), thus inhibiting NK cell proliferation and cytotoxicity (2). Impaired monocyte and DC maturation was associated with decreased secretion of IL-1, CD40, and TNF-a together with production of PGE2 by MSCs (91). Relevant Animal Models for MSC Immunomodulation The immunomodulatory effects of MSCs in vivo were first observed in a baboon model, where expanded donor or third-party bone marrow-derived MSCs delayed rejection of incompatible skin grafts (18). Afterwards, MSC immunomodulation was tested in several animal models of immune disorders. MSCs were shown to alleviate GVHD, a much-feared complication following allogeneic bone marrow transplantation (30,63,81,88,142). Animal models might help predict efficacy of different cell sources, since different therapeutic outcomes were observed in a GVHD rat model when using bone marrow-derived MSCs than conventional short-term cultures or cloned MSCs, the latter being ineffective (88). In experimental allergic encephalomyelitis (EAE), MSCs induce a strong immunosuppression on effector T-cells at the level of secondary lymphoid organs, leading to IL-2reversible T-cell anergy (183), and decrease central nervous system injury while reducing infiltration of T-cells (14). Contrasting results were obtained in experimental collagen-induced arthritis (CIA), where injected MSCs were either beneficial, with decreased serum TNF-a and induced hyporesponsiveness of T lymphocytes (13) or detrimental, with accentuation of the Th1 response (48). Interestingly, in the latter study, an immortalized MSC C3 line was used, further emphasizing the importance of cell source in determining the outcome of treatment. Contrasting data were also reported in murine models of systemic lupus erythematosus (SLE) (25,52,65,151,177, 182,186). When Murphy Roths Large/Fas (TNF receptor superfamily 6) lymphoproliferation mutation (MRL/lpr) mice were treated with human MSCs from healthy donors, T-cell proliferation was affected in a dose-dependent manner with control of autoimmune progression (186), reduction

136

of circulating anti-double-stranded DNA (dsDNA) antibodies, and of proteinuria. Transplantation of MSCs from human exfoliated deciduous teeth was also effective in treating SLE-like MRL/lpr mice, by increasing the ratio of Tregs mediated by inhibition of Th17 cells (177). Effective treatment of lupus nephritis was also obtained by transplantation of human umbilical cord blood-derived MSCs in New Zealand Black/White F1 (NZB/WF1) mice (25). In contrast, Youd et al. observed that allogeneic MSCs exacerbated lupus disease in NZBxNZW F1 mice (182). Such discrepancies might be due to differences in cell source and in animal models and highlight the complexity of exogenous MSC–host tissue interactions possibly leading to unpredictability of treatment outcome (see later). The successful use of MSCs in animal models of insulindependent diabetes mellitus (type 1 diabetes) has received much interest (31). MSCs were found to induce the regeneration of recipient-derived pancreatic insulin-secreting cells, inhibit T-cell-mediated immune responses against newly formed b-cells, and ameliorate hyperglycemia and microalbuminuria (16,32,56,58,69,78,96,99,111,166). Moreover, coencapsulation with MSCs improved engraftment and survival of transplanted islets (86,134,170). However, the mechanisms underlying the observed effects of MSCs in vivo are largely unknown. Available data suggest limited engraftment and even poorer persistence of transplanted MSCs at the injury site. When male MSCs were injected directly at the border of the infarcted site in syngeneic female rats, the percentage of donor cells in the heart decreased rapidly from 34–80% of injected cells immediately after transplantation to less than 5% at 3 weeks (118). In a similar rat model using allogeneic transplantation, neither iron oxide-labeled nor unlabeled cells survived in the infarcted myocardium after 4 weeks (9). In a more recent work on a mouse model with hepatic ischemia– reperfusion injury (52), labeled MSCs were found only in the lungs 1 h after intravenous (IV) infusion. The induction of hepatic injury did not trigger the migration of viable MSCs to the liver, and viable labeled cells completely disappeared from the body within 24 h. The authors conclude that long-term immunomodulatory and regenerative effects of infused MSCs must therefore be mediated by indirect mechanisms or via other cell types. Little information is available in humans. After intracoronary transplantation of radiolabeled unfractionated autologous bone marrow-derived mononuclear cells in a patient with myocardial infarction, the estimated radioactivity uptake by the heart was 5% of the injected radioactivity at 2 h and only 1% at 18 h after transplantation (127). In conclusion, the disproportion between the observed functional effects of MSC administration and the low rates of cell engraftment and persistence supports an indirect primary mechanism

FIERABRACCI ET AL.

other than structural integration of transplanted cells at injured tissue. Clinical Applications Studies on biological properties and therapeutic applications of MSCs have been extensively propelled within the past 5 years, including efforts to define this cell population by the use of appropriate markers and extensive use of animal models. Nowadays, there is the urgency to optimize protocols for clinical applications through the collaborative effort of clinicians and laboratory scientists. As mentioned above, GVHD is the area where MSCbased therapy provided the most significant results. This serious complication occurs following hematopoietic stem cell transplantation when donor T-cells react against the immunocompromised recipient (53,57). The first experience was reported by Le Blanc and colleagues in a patient with severe steroid-refractory acute GVHD, who showed complete response after two infusions of ex vivo expanded haplo-identical human MSCs (95). A phase II trial was subsequently published by the same group (94) in 55 patients with steroid-resistant acute GVHD treated using MSCs from haplo-identical donors (16 cases) or from third-party HLA-mismatched donors (69 cases). No adverse reactions were reported either during the treatment or after MSC infusion. Results were encouraging, since a complete response was observed in 30 patients while 9 patients showed a partial response. Patients with complete response also had longer survival. Similar outcomes were reported in another phase II trial (85). A larger phase III double-blind, placebo-controlled clinical trial using commercial allogeneic MSCs (Prochymal®) consisted of two different protocols (104,112). One protocol evaluated the safety and efficacy of Prochymal in conjunction with steroid therapy in 192 patients with newly diagnosed acute GVHD. The primary endpoint of this study was not met, since there was no statistical difference in the proportion of patients surviving at least 90 days that achieved a complete response when Prochymal was added to steroid therapy compared to those receiving steroids alone. The other protocol evaluated the safety and efficacy of Prochymal in conjunction with standard of care for the treatment of patients who had failed to respond to corticosteroid treatment for acute GVHD. The primary endpoint was a durable complete response defined as complete resolution of GVHD for a duration of at least 28 days. This endpoint was not met in the whole group of patients, but Prochymal significantly improved response rates in the subgroups of patients with steroid-refractory hepatic and gastrointestinal GVHD. Importantly, Prochymal showed a stronger trend of improvement in response rates in pediatric patients (86% vs. 57%, p = 0.094, n = 28). No adverse events were registered. Despite the mixed results

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

of the trial, on May 2012, the sponsor company received market authorization from Canada Health Authorities for the treatment of steroid-resistant GVHD in pediatric patients, making Prochymal the world’s first approved drug having stem cells as its active ingredient. This trial highlights the importance of well-defined patient populations in determining the outcome of MSCbased therapy. Moreover, several issues were raised regarding protocols used in current clinical trials for GVHD, including the need to optimize the timing, dose, and frequency of MSC administration. Usually the number of cells varies from 1 × 106 to 2 × 108 per kg of body weight, administered in one to six infusions. An additional concern regards the purity of MSC preparations (128,181), where cells are isolated using the conventional density-gradient centrifugation approach. It is widely recognized that MSCs expanded in culture consist of a mixture of different cells with various degrees of stemness, only a minority of them showing multilineage potential and self-renewal (98,119, 138). A clonal allogeneic MSC preparation was recently developed by Lim et al. (100) and successfully used for the treatment of two patients with steroid-resistant acute GVHD. If safety and efficacy of clonal MSCs are confirmed, they might represent a more reproducible therapeutic tool with respect to nonclonal preparations. Another major area of interest is the potential application of MSCs in treating autoimmune diseases, due to their immunomodulating and anti-inflammatory properties (165). Crohn’s disease is a disorder of unknown etiology characterized by transmural inflammation of the gastrointestinal tract, resulting in fibrosis causing intestinal obstruction or giving rise to perforations and fistulae. In a pilot study, nine patients with refractory Crohn’s disease were treated with autologous bone marrow-derived MSCs (51). Two IV infusions of 1–2 × 106 cells per kilogram of patient body weight were administered 7 days apart. Three patients showed clinical response, while three patients required surgery due to disease worsening. A large company-sponsored phase III study using allogeneic MSC infusion in patients with refractory Crohn’s disease was halted in March 2009 because of negative preliminary results (15). As an alternative therapeutic approach, autologous adipose tissue-derived MSCs in fibrin glue were inoculated into the fistulas in patients with Crohn’s disease, in an attempt to achieve local stimulation of ulcer healing. A preliminary phase I study in five patients showed a fistula healing rate of 75% and no adverse events with a follow-up of 12–30 months (60). These encouraging results were confirmed by a phase II trial in 24 patients (61). Nevertheless, only a low proportion of the stem cell-treated patients with closure after the procedure remained free of recurrence after more than 3 years of follow-up (66). Moreover, a multicenter phase III clinical trial led by the same group

137

and including 200 patients showed comparable healing rates (about 50% at 1 year) in patients treated with MSCs, MSCs in fibrin glue, or fibrin glue alone (72). Thus, despite the above-reported positive results in patients with steroidrefractory gastrointestinal GVHD, data on the treatment of chronic inflammatory bowel disease with MSCs have been inconclusive so far, highlighting the unpredictability of response to cell therapy in complex disorders, where unknown individual patient characteristics may play a critical role. In recent years, encouraging results were reported from clinical trials evaluating the treatment of SLE (52) with MSCs in patients refractory to conventional therapy (57). Sun et al. (151) used MSCs for treating four patients with active SLE refractory to prolonged therapy with IV cyclophosphamide and oral prednisone. MSCs improved the disease activity index without side effects in a prolonged follow-up to 12 months and induced expansion of Tregs in the peripheral blood of patients at 3-month follow-up. No complications were developed within 12–18 months of follow-up. Based on these encouraging results, a larger phase I trial in 15 patients with refractory disease was performed by the same group (152). Allogeneic MSCs were obtained from non-HLA-matched family members. Clinical amelioration of disease was achieved during a follow-up period of 3–36 months without side effects. Parameters such as anti-dsDNA antibodies and proteinuria decreased significantly, while the percentage of Tregs was significantly restored from the low levels observed before MSC administration. The same authors (152) also explored the use of umbilical cord blood-derived MSCs in 16 severe SLE patients with improvement of SLEDAI (Systemic Lupus Erythematosus Disease Activity Index) score. With this successful background, the European League Against Rheumatism (EULAR) Stromal Cell group is at present conducting a multicenter trial on lupus nephritis treated with allogeneic MSCs (165). MSC therapy was also applied to systemic sclerosis (57), a disease characterized by thickening of the skin caused by accumulation of collagen and by injuries to the smallest arteries, based on the principle that MSCs are able to differentiate into endothelial cells, thus contributing to angiogenesis in adult tissues (103). Following the successful treatment of the first two patients (34,65), Akiyama et al. (5) treated five patients with allogeneic MSCs. The cell administration induced T-cell apoptosis, lymphopenia, and Treg expansion. Clinical improvement of the skin score and of the autoantibody titer was reported. The use of allogeneic versus autologous cells represents an unresolved issue in MSC clinical application. Studies in vitro and in vivo showed similar efficacy of the two cell sources in suppressing immune response (18,41,47). As mentioned above, no serious adverse events were reported

138

in clinical trials with allogeneic MSCs. The latter cell source would be available “off the shelf,” also allowing better standardization and reducing costs of production. This would increase the number of patients taking advantage of such novel therapies. Moreover, autologous cells are not always suitable for efficient ex vivo expansion. There is however a need of reliable specific potency assays in vitro in order to predict the clinical efficacy of different cell preparations. Two major considerations can be put forward when collectively evaluating the results of the above studies. First, the enormous progress of molecular and cell biology that allowed the isolation, characterization, and production of stem cells as therapeutic tools was not paralleled by a similar improvement in our understanding of tissue biology (157). Thus, interactions between administered cells and host tissue remain largely unknown. Second, on the clinical side, appropriate patient selection is an often overlooked factor, which can critically influence the success of the study. Indeed, especially in designing companysponsored clinical trials, the need to reach a sufficient number of patients in a limited time frame leads to increased variability, both including several participating centers from different countries, and by enrolling patients with different characteristics. Such an attitude can increase the risk of missing primary endpoints and can also limit the chances to understand the underlying mechanisms by including patients with a wide spectrum of pathophysiology. Despite such limitations, significant progress has been made over the past decade, also testified by the raising interest of Big Pharma in cell therapy (146). MICROVESICLES: A PATHWAY OF CELL–CELL COMMUNICATION As described above, MSCs—like several other cell types—exert many of their effects via paracrine signaling. The traditional view of cell–cell communication consists in the release of bioactive molecules, resulting in a complex mixture whose local precise composition is largely undetermined. However, it has been known for many years that cells also secrete membrane vesicles, composed of a lipid bilayer including transmembrane proteins and enclosing cytoplasmic components. Such vesicles can transmit signals by interacting at the cell surface, by internalization into endocytic compartments, or by fusion with plasma membranes (7,105) (Fig. 1). Cell-derived membrane vesicles are generally classified according to their size and intracellular origin. Exosomes are 40–100 nm in size, derived from multivesicular bodies (MVBs), a late endosomal compartment, and are secreted via fusion of multivesicular bodies with the plasma membrane. Exosome production is generally regarded as a constitutive membrane vesicle pathway, although it can increase upon stimulation.

FIERABRACCI ET AL.

Shedding vesicles, also known as microvesicles, are a heterogeneous population of membrane vesicles up to 1 mm in size directly derived from the cell membrane of activated cells through the disruption of cortical cytoskeleton. The process is initiated by increased intracellular Ca2+ levels, inhibiting scramblase and floppase to return phospholipids into the inner membrane surface and resulting in altered phospholipid balance of the cell membrane. Interestingly, the targeting of proteins into shedding vesicles seems to be selective (38,117,169). Apoptotic signals also promote the release of membrane particles via the initiation of actin–myosin sliding, resulting in budding and detachment of the membrane from the cytoskeleton. It is a challenge to experimentally distinguish between exosomes and shedding vesicles because of their overlapping biophysical characteristics and the lack of discriminating markers (38,169). Therefore, in this review, the term “microvesicles” (MVs) is used to refer to a mixed population of vesicles. MVs are commonly isolated by differential centrifugation. Such a procedure is, however, cumbersome and unfeasible to produce the large quantities needed for extensive in vivo investigations or for clinical applications. Methods based on immunoaffinity and immunomagnetic isolation by antibodies directed against surface proteins have been developed (28,35). While this approach may prove feasible for MV quantification (although no specific marker has been identified), it cannot be used for functional studies, since particles cannot be retrieved following binding. Ultrafiltration is another widely used isolation method, and it was shown to result in higher MV recovery when compared to ultracentrifugation (93,108). MVs can be approximately quantified by total protein determination (158), although one should consider possible contamination by proteins present, for example, in culture media. Flow cytometry has been assayed by several investigators, although the size of most particles is below the detection limit of commonly used instruments (168). Recently, Nolte-’t Hoen et al. developed a flow cytometric method to analyze single vesicles (124) based on forward scatter wide-angle detection of fluorescentlabeled MVs, reportedly allowing for both quantitative and qualitative analysis of nanosized vesicles such as exosomes. Composition of MVs More than 4,000 different proteins and 2,400 different RNAs are described in the exosome database ExoCarta (106). Interestingly, phosphatidylserine (PS) has been described as a canonical MV lipid present on the surface, as PS is an important docking site for proteins involved in signaling and fusogenesis, and it may contribute to the selective MV enrichment in proteins involved in signaling and fusion with target cells (150). PS could also be recognized

Figure 1. Immunomodulatory effects of MSC-derived MVs on lymphocytes. MVs can transfer signals to target cells by endocytosis, fusion with cell membrane, and delivery of content into the cell or by ligand–receptor interaction. The effects of MSC-derived MVs on lymphocytes are described in the text. CD4 +CD25+FoxP3+ Treg cells: cluster of differentiation 4-positive cluster of differentiation 25-positive forkhead box P3-positive regulatory T-cells.

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES 139

140

FIERABRACCI ET AL.

as a signal inducing capture by target cells, as is the case for apoptotic cells (8). MVs of all cellular origins consistently bear adhesion molecules, which could favor their capture by recipient cells, such as intercellular adhesion molecule 1 (ICAM1), lysosomal-associated membrane protein 2 (LAMP2), tetraspanins [such as CD9, CD151, and tetraspanin 8 (Tspan8)], and integrins [integrin a 3 (CD49C), a 3 subunit of very late activation protein 3 (VLA-3) receptor and integrin a 4 (CD49D), a 4 subunit of VLA-4 receptor] (17,122,123,185). Lactadherin is bound to PS exposed at the surface of exosomes (171). When bound to apoptotic cells, milk fat globule-epidermal growth factor (EGF) factor VIII protein (MFGE8)/lactadherin promotes their phagocytosis by macrophages expressing avb3 and avb5 integrins (67); it could thus play a similar role for capture of MVs. Heat shock proteins, cytoskeletal proteins, MV proteins involved in membrane trafficking [“Ras-related in brain” proteins (rabs) and annexins], scaffolding (tetraspanins), transmembrane transport (solute carriers), and translation (ribosomal proteins) are also commonly found in MVs (107). An excellent recent overview provides exhaustive proteomics data from ExoCarta (106). ROLE OF MVs IN IMMUNE RESPONSES The role of MVs in immune responses has been reviewed extensively by the group of Théry (27,160) and by others (79,143,155). Stimulation of Immune Response Under several circumstances, secreted vesicles can have immuno-activatory properties. Macrophages infected by various pathogens (Mycobacterium and Toxoplasma) release MVs containing pathogen-derived proinflammatory molecular determinants inducing the secretion of proinflammatory cytokines by recipient macrophages (20). Cultured cells infected by Mycoplasma also release proinflammatory exosomes that induce polyclonal activation of B- and T-cells (132). MVs isolated from body fluids could exacerbate autoimmune diseases. In rheumatoid arthritis (RA) patients, fibroblasts obtained from synovial fluid secrete exosomes bearing active membrane-bound TNF-a, which binds to T lymphocytes and renders them resistant to activation-induced cell death (184). Qazi et al. (131) found abundant exosomes in bronchoalveolar fluid from sarcoidosis patients, displaying proinflammatory activity such as increased secretion of IFN-g and IL-13 by PBMCs, and IL-8 by epithelial cells. Exosomes from mature DCs pulsed with male antigen peptide speed up male skin graft rejection by female mice, showing in vivo priming and effector differentiation of activated CD4+ T-cells (147). Exosomes secreted by DCs can also induce humoral responses against the antigens that were fed to DCs before exosome purification (6,35, 130), leading, in the case of the parasite Toxoplasma gondii, to strong protection against acute or congenital

infection (6). In fact, feeding DCs with intact antigen (rather than peptide) allows them to secrete more efficient exosomes, able to stimulate both T- and B-cells in vivo, leading to both memory Th1 and immunoglobulin responses (130). Thus exosomes can induce effector immune responses in vivo. In addition, careful evaluation of immune responses in the first cancer patients treated with exosomes from their own DCs (55) recently suggested a promoting effect of these exosomes on the patient’s NK cell activity (172). Exosomes carry both antigenic material and MHC– peptide complexes required for the initiation of immune responses by APCs. In addition to whole or partially processed antigens, secreted vesicles also expose MHC–peptide complexes, which can be presented to T lymphocytes. Exosomes are not only capable of presenting antigens directly but are also able to transfer both the MHC II molecule and the antigen to neighboring DCs that can in turn present the antigen to T-cells (159). Tumor-derived membrane vesicles stimulate the immune response by transferring tumor antigens to DCs (176) as well as MHC–peptide complexes, leading to antigen-specific T-cell activation in vitro. Exosomes secreted by virtually any cell type bear MHC class I molecules that could potentially induce CD8 T-cell activation and seem to carry antigens, which have to be processed by the recipient APCs. With respect to exosomes secreted by APCs, several groups have reported that DC-derived exosomes could induce activation of CD8+ CTL clones, either by themselves (55,167), or when incubated with DCs expressing allogeneic MHC class I, showing that exosomes bear functional preformed MHC I–peptide complexes (26,76). In addition, more efficient T-cell activation was obtained with exosomes purified from mature rather than immature DCs, suggesting that costimulatory molecules present in exosomes participate in costimulation of T-cells (1). APC-derived exosomes also bear large amounts of MHC II molecules (1). Exosomes bearing specific MHC II–peptide complexes can be obtained by loading APCs with antigen (135) or peptide (115,147,159). Such exosomes can activate cognate clones (135), T-cell lines (147), or activated CD4+ T-cells (159) by themselves, but need to be captured by recipient DCs to activate naive CD4+ T-cells (147,159). Recipient DCs may also use MHC–peptide complexes from exosomes as a source of peptides to load on their own MHC molecules (115). Inhibition of Immune Responses As mentioned earlier, tumor-derived MVs can stimulate DCs to initiate the immune response, but are, on the other hand, able to exert immunosuppressive properties (36,129) resulting in tumor cell escape from immune surveillance. MVs or exosomes obtained from tumor cell lines or tumor-bearing patients were shown in vitro to

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

induce T-cell apoptosis via Fas ligand (FasL) (10,77) and galectin-9 (89). In addition, tumor-derived exosomes can inhibit IL-2-induced T-cell proliferation (158), and/or promote differentiation into Tregs (153), reduce CD8+ T-cell proliferation (175), decrease NK cell cytotoxicity by displaying NK cell receptor D (NKG2D) ligands leading to downregulation of the receptor (11,101), impair myeloid precursor differentiation into DCs (178), and induce myeloidsuppressor cells (39). Vesicles secreted by immune cells can also display immunosuppressive properties. Activated T-cells secrete exosomes bearing FasL, which induce apoptosis of bystander T-cells (114). Exosomes in plasma of pregnant women bear FasL and reduce CD3z expression by T-cells (154). Another mechanism of immunosuppression has been observed for exosomes secreted by placental explants: they bear NKG2D ligands, and induce reduced cytotoxicity of NK and CD8+ T-cells in vitro (71). Thus, placental MVs seem to contribute to fetomaternal tolerance (154). Membrane vesicles secreted by immature DCs also induce tolerogenic, rather than effector, immune responses (126). Such exosomes promote graft survival (126) and reduce inflammation in a model of arthritis (87) or of inflammatory bowel disease (179). Exosomes secreted by normal immature DCs also reduce inflammation in a model of septic shock (109), promoting phagocytosis of sepsis-induced apoptotic cells by macrophages and reducing the release of proinflammatory cytokines (110). ROLE OF MSC–MVs IN REGENERATION The regenerative stimulus induced by MSCs has been described in several tissues (75,84). Stimulation of cell proliferation, inhibition of apoptosis, and stimulation of angiogenesis were reported following MSC transplantation into a variety of injured tissues. Initially, these findings were interpreted as the capacity of MSCs to “transdifferentiate” and replace the cells in the host tissue. More recently, however, it has been recognized that the underlying mechanisms can be better characterized as paracrine effects. Cardiac regeneration induced by MSCs has been extensively studied (92,162,163). It was shown that infusion of media conditioned by MSCs reduces ischemia/ reperfusion (I/R) injury and improves cardiac repair by reducing inflammation, oxidative stress, and apoptosis, and by stimulating angiogenesis (162,163). Lai et al. (92) showed that the cardioprotective factor was present in the MV fraction isolated from MSC supernatant. Purified MSC–MVs strongly reduced infarct size in a mouse model of myocardial I/R injury even at low concentrations, suggesting that the regenerative effect of MSCs on injured myocardium was mainly mediated by secreted MVs. In experimental models of acute renal injury, human MSC–MVs are as effective as MSCs in accelerating recovery from glycerol-induced acute kidney injury in severe combined immunodeficient

141

(SCID) mice in vivo, stimulate proliferation, and inhibit apoptosis of tubular epithelial cells in vitro, effects that are abolished by RNase treatment of the MVs (21,40). The latter finding suggests that the regenerative response is mediated through the transfer of mRNA. In a murine model of hypoxic pulmonary hypertension, IV delivery of MSC–MVs suppressed the hypoxic pulmonary influx of macrophages and the induction of proinflammatory and proproliferative mediators, including monocyte chemoattractant protein-1 and hypoxia-inducible mitogenic factor, inhibiting vascular remodeling and pulmonary hypertension, while fibroblast-derived exosomes had no effect. Moreover, MSC–MVs inhibited signal transducer and activator of transcription 3 (STAT3)-mediated hyperproliferative signaling in isolated human pulmonary artery endothelial cells, pointing to a direct effect on hypoxic vascular cells (97). ROLE OF MSC–MVs IN IMMUNOMODULATION Some recent studies suggest that the immunomodulatory activity of MSCs could be at least partially mediated by their ability to release MVs. We observed that the inhibitory effects of MSCs on B-cell proliferation and differentiation in a CpG-stimulated PBMC coculture system could be fully reproduced by MVs isolated from MSC culture supernatants in a dose-dependent fashion (22) (Fig. 1). A dose-dependent inhibitory activity of MSC–MVs was also observed for immunoglobulin M (IgM), IgG, and IgA production. Moreover, in the same coculture system, 7-aminoactinomycin D (7-AAD)-negative and annexinpositive MSC–MVs isolated from MSCs were internalized in a subset of CD86/CD19-positive cells corresponding to activated B lymphocytes. The effect of MVs on T-cells was investigated by Mokarizadeh et al. (113). These authors showed that MVs isolated from murine BMSCs inhibited the proliferation of both syngenic and allogeneic T lymphocytes. Additionally, they demonstrated that these microparticles were able to induce apoptosis in activated T-cells. Interestingly, this inhibition was associated with an increased proportion of regulatory T CD4+-CD25+-forkhead box P3-positive (FoxP3+) cells (Fig. 1). Moreover, an increased secretion of IL-10 and transforming growth factor-b1 (TGF-b1) by cultured splenic cells added with MSC–MVs was observed. These results suggest that MSC–MVs can induce tolerogenic signaling. We recently observed similar results in human PBMC cultures treated with human T-cell activator CD3/CD28 beads (unpublished). In vitro results are also supported by in vivo observations in an animal model of inflammatory bowel disease (46) induced by dextran sulfate sodium (DSS). Mice injected daily with MSC–MVs showed less weight loss, improved disease activity index, and a less severe reduction in colon length when compared to DSS/vehicle-treated controls. Real-time RT-PCR analysis performed on RNA extracted from colon tissue revealed a strong inhibition of the

142

induction of inflammatory cytokines with respect to untreated animals. Collectively, these data suggest that MVs isolated from MSCs could reproduce the immunomodulatory effect of MSCs. IMPLICATIONS FOR THERAPY Results and considerations reported above suggest that MSC–MVs could offer several advantages as therapeutic tools with respect to MSCs. MSCs can exhibit either an anti-inflammatory or proinflammatory phenotype depending on the balance between the cytokines/chemokines released into the surrounding microenvironment (84). For instance, as reported above, MSCs were shown to have both inhibitory (41,43) and stimulatory (136,164) effects on B-cell proliferation, differentiation, and antibody production. The effect of MSCs on B-cells appears to be indirect, that is, mediated by other cell types, thus again strongly dependent on the inflammatory environment (41). As reported above, we confirmed MSC-mediated inhibition of B-cell function in PBMCs (22), but we also observed MSC-mediated stimulation of both proliferation and differentiation when using purified B-cells, in agreement with the work of Traggiai et al. (164). Thus, we confirmed that MSCs can exert opposite effects on B-cell function depending on the environment. However, we observed that MSC–MVs suppress B-cell function both in PBMCs and in purified B-cell coculture (Muraca and Fierabracci, unpublished). This preliminary observation is in agreement with the hypothesis that MSC–MVs could provide simpler interactions with the target cells by a onehit stimulus (156), that is, not mediated by other cell types. This is also consistent with our observations that MSC–MVs are taken up specifically by activated B-cells (22). Another possible advantage of using MVs rather than MSCs could be to avoid possible long-term maldifferentiation of engrafted cells or tumor generation (161). Cell products could also be easier to characterize and standardize with respect to the cells themselves. Phase I trials involving MVs derived from DCs for immunotherapy of advanced cancer (55,116) showed that large-scale, clinical-grade production, isolation, and infusion of MVs is feasible and safe. A critical issue will be defining the effective therapeutic dosage of MSC–MVs. In our experience, administration of MVs isolated from the supernatant of 2 × 106 MSC cultures results in significant clinical and biochemical improvements in a mouse model of inflammatory bowel disease (IBD) (46). In a 70-kg human, this therapeutic dosage would theoretically correspond to the isolate from the supernatant of about 6–8 × 109 MSCs. A similar cell mass can be easily cultured in bioreactors. Immortalization of MSCs by Myc (29) has been proposed in order to ensure production of MSC–MVs in sufficient quantity to support clinical applications. It was also shown that MSC lines expressing high levels of CD81, a protein belonging to the

FIERABRACCI ET AL.

tetraspanin family, release more MVs in culture medium, an observation that could help select optimal cell phenotype for MSC–MV production (180). Clinical-grade isolation and purification of MVs from large volumes (1–4 L) of supernatant has been described (93). Collectively, these considerations support the feasibility of future therapeutic applications of MSC–MVs. However, additional studies are needed to explore the role and mechanisms of MSC–MVs as modulators of immune response before they can be proposed for clinical use in place of the parent cells. CONCLUSIVE REMARKS Increasing experimental evidence points to the possibility of using MSC-derived MVs both as a stimulus for the regeneration of injured tissues and for immunomodulation, two closely related processes. Clearly, these encouraging results deserve additional investigations before any claim for the use of MV–MSCs as a novel therapeutic approach can be addressed. However, this line of investigation should be pursued, since MVs could represent a safer and more reproducible therapeutic tool than their parent cells. ACKNOWLEDGMENTS: We thank Massimo Dominici (University of Modena and Reggio Emilia, Modena, Italy) for helpful discussion. The financial contribution of CryoSave (Zutphen, Netherlands) is gratefully acknowledged. The authors declare no conflicts of interest.

REFERENCES 1. Admyre, C.; Grunewald, J.; Thyberg, J.; Gripenback, S.; Tornling, G.; Eklund, A.; Scheynius, A.; Gabrielsson, S. Exosomes with major histocompatibility complex class II and co-stimulatory molecules are present in human BAL fluid. Eur. Respir. J. 22(4):578–583; 2003. 2. Aggarwal, S.; Pittenger, M. F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105(4):1815–1822; 2005. 3. Agung, M.; Ochi, M.; Yanada, S.; Adachi, N.; Izuta, Y.; Yamasaki, T.; Toda, K. Mobilization of bone marrowderived mesenchymal stem cells into the injured tissues after intraarticular injection and their contribution to tissue regeneration. Knee Surg. Sports Traumatol. Arthrosc. 14(12): 1307–1314; 2006. 4. Ahmad, A. Grafix® demonstrates overwhelming efficacy in landmark stem cell study for the treatment of diabetic foot ulcers; 2013. Available from http://www.osiris.com/ 2013-08-13%20Grafix%20DFU%20Trial%20Results.pdf 5. Akiyama, K.; Chen, C.; Wang, D.; Xu, X.; Qu, C.; Yamaza, T.; Cai, T.; Chen, W.; Sun, L.; Shi, S. Mesenchymalstem-cell-induced immunoregulation involves FAS-ligand-/ FAS-mediated T cell apoptosis. Cell Stem Cell 10(5):544– 555; 2012. 6. Aline, F.; Bout, D.; Amigorena, S.; Roingeard, P.; DimierPoisson, I. Toxoplasma gondii antigen-pulsed-dendritic cellderived exosomes induce a protective immune response against T. gondii infection. Infect. Immun. 72(7):4127–4137; 2004. 7. Al-Nedawi, K.; Meehan, B.; Kerbel, R. S.; Allison, A. C.; Rak, J. Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

8.

9.

10.

11.

12.

13.

14.

15. 16. 17.

18.

19.

20.

21.

EGFR. Proc. Natl. Acad. Sci. USA 106(10):3794–3799; 2009. Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 10(5):619–624; 2008. Amsalem, Y.; Mardor, Y.; Feinberg, M. S.; Landa, N.; Miller, L.; Daniels, D.; Ocherashvilli, A.; Holbova, R.; Yosef, O.; Barbash, I. M.; Leor, J. Iron-oxide labeling and outcome of transplanted mesenchymal stem cells in the infarcted myocardium. Circulation 116(11 Suppl):38–45; 2007. Andreola, G.; Rivoltini, L.; Castelli, C.; Huber, V.; Perego, P.; Deho, P.; Squarcina, P.; Accornero, P.; Lozupone, F.; Lugini, L.; Stringaro, A.; Molinari, A.; Arancia, G.; Gentile, M.; Parmiani, G.; Fais, S. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J. Exp. Med. 195(10):1303–1316; 2002. Ashiru, O.; Boutet, P.; Fernandez-Messina, L.; AgueraGonzalez, S.; Skepper, J. N.; Vales-Gomez, M.; Reyburn, H. T. Natural killer cell cytotoxicity is suppressed by exposure to the human NKG2D ligand MICA*008 that is shed by tumor cells in exosomes. Cancer Res. 70(2):481–489; 2010. Augello, A.; Tasso, R.; Negrini, S. M.; Amateis, A.; Indiveri, F.; Cancedda, R.; Pennesi, G. Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur. J. Immunol. 35(5):1482–1490; 2005. Augello, A.; Tasso, R.; Negrini, S. M.; Cancedda, R.; Pennesi, G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 56(4):1175–1186; 2007. Bai, L.; Lennon, D. P.; Eaton, V.; Maier, K.; Caplan, A. I.; Miller, S. D.; Miller, R. H. Human bone marrow-derived mesenchymal stem cells induce Th2-polarized immune response and promote endogenous repair in animal models of multiple sclerosis. Glia 57(11):1192–1203; 2009. Baker, M. Stem-cell drug fails crucial trials. Nature http:// www.nature.com/news/2009/090909/full/news.2009.894. htm; 2009. Banerjee, M.; Kumar, A.; Bhonde, R. R. Reversal of experimental diabetes by multiple bone marrow transplantation. Biochem. Biophys. Res. Commun. 328(1):318–325; 2005. Barres, C.; Blanc, L.; Bette-Bobillo, P.; Andre, S.; Mamoun, R.; Gabius, H. J.; Vidal, M. Galectin-5 is bound onto the surface of rat reticulocyte exosomes and modulates vesicle uptake by macrophages. Blood 115(3):696–705; 2010. Bartholomew, A.; Sturgeon, C.; Siatskas, M.; Ferrer, K.; McIntosh, K.; Patil, S.; Hardy, W.; Devine, S.; Ucker, D.; Deans, R.; Moseley, A.; Hoffman, R. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp. Hematol. 30(1): 42–48; 2002. Beyth, S.; Borovsky, Z.; Mevorach, D.; Liebergall, M.; Gazit, Z.; Aslan, H.; Galun, E.; Rachmilewitz, J. Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood 105(5): 2214–2219; 2005. Bhatnagar, S.; Schorey, J. S. Exosomes released from infected macrophages contain Mycobacterium avium glycopeptidolipids and are proinflammatory. J. Biol. Chem. 282(35): 25779–25789; 2007. Bruno, S.; Grange, C.; Deregibus, M. C.; Calogero, R. A.; Saviozzi, S.; Collino, F.; Morando, L.; Busca, A.; Falda, M.; Bussolati, B.; Tetta, C.; Camussi, G. Mesenchymal

143

22.

23. 24.

25.

26.

27. 28.

29.

30.

31. 32.

33.

34.

stem cell-derived microvesicles protect against acute tubular injury. J. Am. Soc. Nephrol. 20(5):1053–1067; 2009. Budoni, M.; Fierabracci, A.; Luciano, R.; Petrini, S.; Di Ciommo, V.; Muraca, M. The immunosuppressive effect of mesenchymal stromal cells on B lymphocytes is mediated by membrane vesicles. Cell Transplant. 22(2):369– 379; 2013. Caplan, A. I. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J. Cell. Physiol. 213(2):341–347; 2007. Chamberlain, G.; Fox, J.; Ashton, B.; Middleton, J. Concise review: Mesenchymal stem cells: Their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 25(11):2739–2749; 2007. Chang, J. W.; Hung, S. P.; Wu, H. H.; Wu, W. M.; Yang, A. H.; Tsai, H. L.; Yang, L. Y.; Lee, O. K. Therapeutic effects of umbilical cord blood-derived mesenchymal stem cell transplantation in experimental lupus nephritis. Cell Transplant. 20(2):245–257; 2011. Chaput, N.; Schartz, N. E. C.; Andre, F.; Taieb, J.; Novault, S.; Bonnaventure, P.; Aubert, N.; Bernard, J.; Lemonnier, F.; Merad, M.; Adema, G.; Adams, M.; Ferrantini, M.; Carpentier, A. F.; Escudier, B.; Tursz, T.; Angevin, E.; Zitvogel, L. Exosomes as potent cell-free peptide-based vaccine. II. Exosomes in CpG adjuvants efficiently prime naive Tc1 lymphocytes leading to tumor rejection. J. Immunol. 172(4):2137–2146; 2004. Chaput, N.; Thery, C. Exosomes: Immune properties and potential clinical implementations. Semin. Immunopathol. 33(5):419–440; 2011. Chen, C.; Skog, J.; Hsu, C. H.; Lessard, R. T.; Balaj, L.; Wurdinger, T.; Carter, B. S.; Breakefield, X. O.; Toner, M.; Irimia, D. Microfluidic isolation and transcriptome analysis of serum microvesicles. Lab. Chip 10(4):505–511; 2010. Chen, T. S.; Arslan, F.; Yin, Y.; Tan, S. S.; Lai, R. C.; Choo, A. B. H.; Padmanabhan, J.; Lee, C. N.; de Kleijn, D. P. V.; Lim, S. K. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J. Transl. Med. 9:47; 2011. Chen, W.; Li, M.; Li, Z.; Yan, Z.; Cheng, H.; Pan, B.; Cao, J.; Chen, C.; Zeng, L.; Xu, K. CXCR4-transduced mesenchymal stem cells protect mice against graft-versushost disease. Immunol. Lett. 143(2):161–169; 2012. Chhabra, P.; Brayman, K. L. Stem cell therapy to cure type 1 diabetes: From hype to hope. Stem Cells Transl. Med. 2(5):328–336; 2013. Choi, J. B.; Uchino, H.; Azuma, K.; Iwashita, N.; Tanaka, Y.; Mochizuki, H.; Migita, M.; Shimada, T.; Kawamori, R.; Watada, H. Little evidence of transdifferentiation of bone marrow-derived cells into pancreatic beta cells. Diabetologia 46(10):1366–1374; 2003. Choi, M. R.; Kim, H. Y.; Park, J. Y.; Lee, T. Y.; Baik, C. S.; Chai, Y. G.; Jung, K. H.; Park, K. S.; Roh, W.; Kim, K. S.; Kim, S. H. Selection of optimal passage of bone marrow-derived mesenchymal stem cells for stem cell therapy in patients with amyotrophic lateral sclerosis. Neurosci. Lett. 472(2):94–98; 2010. Christopeit, M.; Schendel, M.; Foll, J.; Muller, L. P.; Keysser, G.; Behre, G. Marked improvement of severe progressive systemic sclerosis after transplantation of mesenchymal stem cells from an allogeneic haploidenticalrelated donor mediated by ligation of CD137L. Leukemia 22(5):1062–1064; 2008.

144

35. Clayton, A.; Court, J.; Navabi, H.; Adams, M.; Mason, M. D.; Hobot, J. A.; Newman, G. R.; Jasani, B. Analysis of antigen presenting cell derived exosomes, based on immuno-magnetic isolation and flow cytometry. J. Immunol. Methods 247(1–2):163–174; 2001. 36. Clayton, A.; Mitchell, J. P.; Court, J.; Mason, M. D.; Tabi, Z. Human tumor-derived exosomes selectively impair lymphocyte responses to interleukin-2. Cancer Res. 67(15): 7458–7466; 2007. 37. Clifford, D. M.; Fisher, S. A.; Brunskill, S. J.; Doree, C.; Mathur, A.; Clarke, M. J.; Watt, S. M.; Martin-Rendon, E. Long-term effects of autologous bone marrow stem cell treatment in acute myocardial infarction: Factors that may influence outcomes. PLoS One 7(5):e37373; 2012. 38. Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: Artefacts no more. Trends Cell Biol. 19(2): 43–51; 2009. 39. Cocucci, E.; Racchetti, G.; Podini, P.; Meldolesi, J. Enlargeosome traffic: Exocytosis triggered by various signals is followed by endocytosis, membrane shedding or both. Traffic 8(6):742–757; 2007. 40. Collino, F.; Deregibus, M. C.; Bruno, S.; Sterpone, L.; Aghemo, G.; Viltono, L.; Tetta, C.; Camussi, G. Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs. PLoS One 5(7):e11803; 2010. 41. Comoli, P.; Ginevri, F.; Maccario, R.; Avanzini, M. A.; Marconi, M.; Groff, A.; Cometa, A.; Cioni, M.; Porretti, L.; Barberi, W.; Frassoni, F.; Locatelli, F. Human mesenchymal stem cells inhibit antibody production induced in vitro by allostimulation. Nephrol. Dial. Transplant. 23(4): 1196–1202; 2008. 42. Connick, P.; Kolappan, M.; Patani, R.; Scott, M. A.; Crawley, C.; He, X.-L.; Richardson, K.; Barber, K.; Webber, D. J.; Wheeler-Kingshott, C. A. M.; Tozer, D. J.; Samson, R. S.; Thomas, D. L.; Du, M.-Q.; Luan, S. L.; Michell, A. W.; Altmann, D. R.; Thompson, A. J.; Miller, D. H.; Compston, A.; Chandran, S. The mesenchymal stem cells in multiple sclerosis (MSCIMS) trial protocol and baseline cohort characteristics: An open-label pre-test: Post-test study with blinded outcome assessments. Trials 12:62; 2011. 43. Corcione, A.; Benvenuto, F.; Ferretti, E.; Giunti, D.; Cappiello, V.; Cazzanti, F.; Risso, M.; Gualandi, F.; Mancardi, G. L.; Pistoia, V.; Uccelli, A. Human mesenchymal stem cells modulate B-cell functions. Blood 107(1): 367–372; 2006. 44. Davatchi, F.; Abdollahi, B. S.; Mohyeddin, M.; Shahram, F.; Nikbin, B. Mesenchymal stem cell therapy for knee osteoarthritis. Preliminary report of four patients. Int. J. Rheum. Dis. 14(2):211–215; 2011. 45. De Bari, C.; Dell’Accio, F.; Tylzanowski, P.; Luyten, F. P. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum. 44(8):1928–1942; 2001. 46. Del Fattore, A.; Luciano, R.; Fierabracci, A.; Muraca, M. Microvesicles derived from mesenchymal stem cells show anti-inflammatory activity in an animal model of ulcerative colitis. 5th Meeting of the Forum of Italian Researchers on Mesenchymal and Stromal Stem Cells. Milan, Italy; 2013. 47. Di Nicola, M.; Carlo-Stella, C.; Magni, M.; Milanesi, M.; Longoni, P. D.; Matteucci, P.; Grisanti, S.; Gianni, A. M. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 99(10):3838–3843; 2002.

FIERABRACCI ET AL.

48. Djouad, F.; Fritz, V.; Apparailly, F.; Louis-Plence, P.; Bony, C.; Sany, J.; Jorgensen, C.; Noel, D. Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor alpha in collagen-induced arthritis. Arthritis Rheum. 52(5):1595–1603; 2005. 49. Dominici, M.; Le Blanc, K.; Mueller, I.; SlaperCortenbach, I.; Marini, F. C.; Krause, D. S.; Deans, R. J.; Keating, A.; Prockop, D.; Em, H. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315– 317; 2006. 50. Dorshkind, K. Regulation of hemopoiesis by bone marrow stromal cells and their products. Annu. Rev. Immunol. 8: 111–137; 1990. 51. Duijvestein, M.; Vos, A. C. W.; Roelofs, H.; Wildenberg, M. E.; Wendrich, B. B.; Verspaget, H. W.; Kooy-Winkelaar, E. M. C.; Koning, F.; Zwaginga, J. J.; Fidder, H. H.; Verhaar, A. P.; Fibbe, W. E.; van den Brink, G. R.; Hommes, D. W. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: Results of a phase I study. Gut 59(12):1662–1669; 2010. 52. Eggenhofer, E.; Benseler, V.; Kroemer, A.; Popp, F. C.; Geissler, E. K.; Schlitt, H. J.; Baan, C. C.; Dahlke, M. H.; Hoogduijn, M. J. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front. Immunol. 3:297; 2012. 53. English, K.; French, A.; Wood, K. J. Mesenchymal stromal cells: Facilitators of successful transplantation? Cell Stem Cell 7(4):431–442; 2010. 54. Erices, A.; Conget, P.; Minguell, J. J. Mesenchymal progenitor cells in human umbilical cord blood. Br. J. Haematol. 109(1):235–242; 2000. 55. Escudier, B.; Dorval, T.; Chaput, N.; Andre, F.; Caby, M. P.; Novault, S.; Flament, C.; Leboulaire, C.; Borg, C.; Amigorena, S.; Boccaccio, C.; Bonnerot, C.; Dhellin, O.; Movassagh, M.; Piperno, S.; Robert, C.; Serra, V.; Valente, N.; Le Pecq, J. B.; Spatz, A.; Lantz, O.; Tursz, T.; Angevin, E.; Zitvogel, L. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of the first phase I clinical trial. J. Transl. Med. 3(1):10; 2005. 56. Ezquer, F. E.; Ezquer, M. E.; Parrau, D. B.; Carpio, D.; Yanez, A. J.; Conget, P. A. Systemic administration of multipotent mesenchymal stromal cells reverts hyperglycemia and prevents nephropathy in type 1 diabetic mice. Biol. Blood Marrow Transplant. 14(6):631–640; 2008. 57. Figueroa, F. E.; Carrión, F.; Villanueva, S.; Khoury, M. Mesenchymal stem cell treatment for autoimmune diseases: A critical review. Biol. Res. 45:269–277; 2012. 58. Fiorina, P.; Jurewicz, M.; Augello, A.; Vergani, A.; Dada, S.; La Rosa, S.; Selig, M.; Godwin, J.; Law, K.; Placidi, C.; Smith, R. N.; Capella, C.; Rodig, S.; Adra, C. N.; Atkinson, M.; Sayegh, M. H.; Abdi, R. Immunomodulatory function of bone marrow-derived mesenchymal stem cells in experimental autoimmune type 1 diabetes. J. Immunol. 183(2):993–1004; 2009. 59. Friedenstein, A. J.; Piatetzky-Shapiro, I. I.; Petrakova, K. Osteogenesis in transplants of bone marrow cells. J. Embryol. Exp. Morphol. 16(3):381–390; 1996. 60. Garcia-Olmo, D.; Garcia-Arranz, M.; Herreros, D.; Pascual, I.; Peiro, C.; Rodriguez-Montes, J. A. A phase I clinical trial of the treatment of Crohn’s fistula by adipose mesenchymal stem cell transplantation. Dis. Colon Rectum 48(7):1416–1423; 2005.

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

61. Garcia-Olmo, D.; Herreros, D.; Pascual, I.; Pascual, J. A.; Del-Valle, E.; Zorrilla, J.; De-La-Quintana, P.; GarciaArranz, M.; Pascual, M. Expanded adipose-derived stem cells for the treatment of complex perianal fistula: A phase II clinical trial. Dis. Colon Rectum 52(1):79–86; 2009. 62. Glennie, S.; Soeiro, I.; Dyson, P. J.; Lam, E. W. F.; Dazzi, F. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 105(7):2821–2827; 2005. 63. Gregoire-Gauthier, J.; Selleri, S.; Fontaine, F.; Dieng, M. M.; Patey, N.; Despars, G.; Beausejour, C. M.; Haddad, E. Therapeutic efficacy of cord blood-derived mesenchymal stromal cells for the prevention of acute graft-versus-host disease in a xenogenic mouse model. Stem Cells Dev. 21(10):1616–1626; 2012. 64. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P. G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 97(25):13625– 13630; 2000. 65. Gu, Z.; Akiyama, K.; Ma, X.; Zhang, H.; Feng, X.; Yao, G.; Hou, Y.; Lu, L.; Gilkeson, G. S.; Silver, R. M.; Zeng, X.; Shi, S.; Sun, L. Transplantation of umbilical cord mesenchymal stem cells alleviates lupus nephritis in MRL/lpr mice. Lupus 19(13):1502–1514; 2010. 66. Guadalajara, H.; Herreros, D.; De-La-Quintana, P.; Trebol, J.; Garcia-Arranz, M.; Garcia-Olmo, D. Long-term follow-up of patients undergoing adipose-derived adult stem cell administration to treat complex perianal fistulas. Int. J. Colorectal Dis. 27(5):595–600; 2012. 67. Hanayama, R.; Tanaka, M.; Miwa, K.; Shinohara, A.; Iwamatsu, A.; Nagata, S. Identification of a factor that links apoptotic cells to phagocytes. Nature 417(6885):182–187; 2002. 68. Hare, J. M.; Traverse, J. H.; Henry, T. D.; Dib, N.; Strumpf, R. K.; Schulman, S. P.; Gerstenblith, G.; DeMaria, A. N.; Denktas, A. E.; Gammon, R. S.; Hermiller, J. B.; Reisman, M. A.; Schaer, G. L.; Sherman, W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J. Am. Coll. Cardiol. 54(24):2277–2286; 2009. 69. Hasegawa, Y.; Ogihara, T.; Yamada, T.; Ishigaki, Y.; Imai, J.; Uno, K.; Gao, J.; Kaneko, K.; Ishihara, H.; Sasano, H.; Nakauchi, H.; Oka, Y.; Katagiri, H. Bone marrow (BM) transplantation promotes beta-cell regeneration after acute injury through BM cell mobilization. Endocrinology 148(5):2006–2015; 2007. 70. Hass, R.; Kasper, C.; Bohm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 9:12; 2011. 71. Hedlund, M.; Stenqvist, A. C.; Nagaeva, O.; Kjellberg, L.; Wulff, M.; Baranov, V.; Mincheva-Nilsson, L. Human placenta expresses and secretes NKG2D ligands via exosomes that down-modulate the cognate receptor expression: Evidence for immunosuppressive function. J. Immunol. 183(1):340–351; 2009. 72. Herreros, M. D.; Garcia-Arranz, M.; Guadalajara, H.; De-La-Quintana, P.; Garcia-Olmo, D. Autologous expanded adipose-derived stem cells for the treatment of complex cryptoglandular perianal fistulas: A phase III randomized clinical trial (FATT 1: fistula Advanced Therapy Trial 1) and long-term evaluation. Dis. Colon Rectum 55(7):762– 772; 2012.

145

73. Honmou, O.; Houkin, K.; Matsunaga, T.; Niitsu, Y.; Ishiai, S.; Onodera, R.; Waxman, S. G.; Kocsis, J. D. Intravenous administration of auto serum-expanded autologous mesenchymal stem cells in stroke. Brain 134(Pt 6): 1790–1807; 2011. 74. Horie, M.; Sekiya, I.; Muneta, T.; Ichinose, S.; Matsumoto, K.; Saito, H.; Murakami, T.; Kobayashi, E. Intra-articular injected synovial stem cells differentiate into meniscal cells directly and promote meniscal regeneration without mobilization to distant organs in rat massive meniscal defect. Stem Cells 27(4):878–887; 2009. 75. Horwitz, E. M.; Dominici, M. How do mesenchymal stromal cells exert their therapeutic benefit? Cytotherapy 10(8):771–774; 2008. 76. Hsu, D. H.; Paz, P.; Villaflor, G.; Rivas, A.; MehtaDamani, A.; Angevin, E.; Zitvogel, L.; Le Pecq, J. B. Exosomes as a tumor vaccine: Enhancing potency through direct loading of antigenic peptides. J. Immunother. 26(5): 440–450; 2003. 77. Huber, V.; Fais, S.; Iero, M.; Lugini, L.; Canese, P.; Squarcina, P.; Zaccheddu, A.; Colone, M.; Arancia, G.; Gentile, M.; Seregni, E.; Valenti, R.; Ballabio, G.; Belli, F.; Leo, E.; Parmiani, G.; Rivoltini, L. Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: Role in immune escape. Gastroenterology 128(7):1796–1804; 2005. 78. Ianus, A.; Holz, G. G.; Theise, N. D.; Hussain, M. A. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J. Clin. Invest. 111(6):843–850; 2003. 79. Iero, M.; Valenti, R.; Huber, V.; Filipazzi, P.; Parmiani, G.; Fais, S.; Rivoltini, L. Tumour-released exosomes and their implications in cancer immunity. Cell Death Differ. 15(1):80–88; 2008. 80. In ’t Anker, P. S.; Scherjon, S. A.; Kleijburg-van der Keur, C.; de Groot-Swings, G. M. J. S.; Claas, F. H. J.; Fibbe, W. E.; Kanhai, H. H. H. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 22(7):1338–1345; 2004. 81. Jang, M. J.; Kim, H.-S.; Lee, H.-G.; Kim, G. J.; Jeon, H. G.; Shin, H.-S.; Chang, S.-K.; Hur, G.-H.; Chong, S. Y.; Oh, D.; Chung, H.-M. Placenta-derived mesenchymal stem cells have an immunomodulatory effect that can control acute graft-versus-host disease in mice. Acta Haematol. 129(4):197–206; 2013. 82. Johnson, K.; Zhu, S.; Tremblay, M. S.; Payette, J. N.; Wang, J.; Bouchez, L. C.; Meeusen, S.; Althage, A.; Cho, C. Y.; Wu, X.; Schultz, P. G. A stem cell-based approach to cartilage repair. Science 336(6082):717–721; 2012. 83. Karussis, D.; Karageorgiou, C.; Vaknin-Dembinsky, A.; Gowda-Kurkalli, B.; Gomori, J. M.; Kassis, I.; Bulte, J. W. M.; Petrou, P.; Ben-Hur, T.; Abramsky, O.; Slavin, S. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch. Neurol. 67(10):1187– 1194; 2010. 84. Keating, A. Mesenchymal stromal cells: New directions. Cell Stem Cell 10(6):709–716; 2012. 85. Kebriaei, P.; Isola, L.; Bahceci, E.; Holland, K.; Rowley, S.; McGuirk, J.; Devetten, M.; Jansen, J.; Herzig, R.; Schuster, M.; Monroy, R.; Uberti, J. Adult human mesenchymal stem cells added to corticosteroid therapy for the treatment of acute graft-versus-host disease. Biol. Blood Marrow Transplant. 15(7):804–811; 2009.

146

86. Kerby, A.; Jones, E. S.; Jones, P. M.; King, A. J. Co-transplantation of islets with mesenchymal stem cells in microcapsules demonstrates graft outcome can be improved in an isolated-graft model of islet transplantation in mice. Cytotherapy 15(2):192–200; 2013. 87. Kim, S.-H.; Lechman, E. R.; Bianco, N.; Menon, R.; Keravala, A.; Nash, J.; Mi, Z.; Watkins, S. C.; Gambotto, A.; Robbins, P. D. Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collageninduced arthritis. J. Immunol. 174(10):6440–6448; 2005. 88. Kitazawa, Y.; Li, X.-K.; Xie, L.; Zhu, P.; Kimura, H.; Takahara, S. Bone marrow-derived conventional, but not cloned, mesenchymal stem cells suppress lymphocyte proliferation and prevent graft-versus-host disease in rats. Cell Transplant. 21(2-3):581–590; 2012. 89. Klibi, J.; Niki, T.; Riedel, A.; Pioche-Durieu, C.; Souquere, S.; Rubinstein, E.; Le Moulec, S.; Moulec, S. L. E.; Guigay, J.; Hirashima, M.; Guemira, F.; Adhikary, D.; Mautner, J.; Busson, P. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by EpsteinBarr virus-infected nasopharyngeal carcinoma cells. Blood 113(9):1957–1966; 2009. 90. Krampera, M.; Cosmi, L.; Angeli, R.; Pasini, A.; Liotta, F.; Andreini, A.; Santarlasci, V.; Mazzinghi, B.; Pizzolo, G.; Vinante, F.; Romagnani, P.; Maggi, E.; Romagnani, S.; Annunziato, F. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 24(2):386–398; 2006. 91. Kronsteiner, B.; Peterbauer-Scherb, A.; Grillari-Voglauer, R.; Redl, H.; Gabriel, C.; van Griensven, M.; Wolbank, S. Human mesenchymal stem cells and renal tubular epithelial cells differentially influence monocyte-derived dendritic cell differentiation and maturation. Cell. Immunol. 267(1):30–38; 2011. 92. Lai, R. C.; Arslan, F.; Lee, M. M.; Sze, N. S. K.; Choo, A.; Chen, T. S.; Salto-Tellez, M.; Timmers, L.; Lee, C. N.; El Oakley, R. M.; Pasterkamp, G.; de Kleijn, D. P. V.; Lim, S. K. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 4(3):214–222; 2010. 93. Lamparski, H. G.; Metha-Damani, A.; Yao, J.-Y.; Patel, S.; Hsu, D.-H.; Ruegg, C.; Le Pecq, J.-B. Production and characterization of clinical grade exosomes derived from dendritic cells. J. Immunol. Methods 270(2):211–226; 2002. 94. Le Blanc, K.; Frassoni, F.; Ball, L.; Locatelli, F.; Roelofs, H.; Lewis, I.; Lanino, E.; Sundberg, B.; Bernardo, M. E.; Remberger, M.; Pasterkamp, G.; de Kleijn, D. P. V.; Lim, S. K. Mesenchymal stem cells for treatment of steroidresistant, severe, acute graft-versus-host disease: A phase II study. Lancet 371(9624):1579–1586; 2008. 95. Le Blanc, K.; Rasmusson, I.; Sundberg, B.; Gotherstrom, C.; Hassan, M.; Uzunel, M.; Ringden, O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 363(9419): 1439–1441; 2004. 96. Lechner, A.; Yang, Y.-G.; Blacken, R. A.; Wang, L.; Nolan, A. L.; Habener, J. F. No evidence for significant transdifferentiation of bone marrow into pancreatic beta-cells in vivo. Diabetes 53(3):616–623; 2004. 97. Lee, C.; Mitsialis, S. A.; Aslam, M.; Vitali, S. H.; Vergadi, E.; Konstantinou, G.; Sdrimas, K.; Fernandez-Gonzalez, A.; Kourembanas, S. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation 126(22):2601–2611; 2012.

FIERABRACCI ET AL.

98. Lee, C. C.; Christensen, J. E.; Yoder, M. C.; Tarantal, A. F. Clonal analysis and hierarchy of human bone marrow mesenchymal stem and progenitor cells. Exp. Hematol. 38(1):46–54; 2010. 99. Lee, R. H.; Seo, M. J.; Reger, R. L.; Spees, J. L.; Pulin, A. A.; Olson, S. D.; Prockop, D. J. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc. Natl. Acad. Sci. USA 103(46):17438–17443; 2006. 100. Lim, J. H.; Lee, M. H.; Yi, H. G.; Kim, C. S.; Kim, J. H.; Song, S. U. Mesenchymal stromal cells for steroid-refractory acute graft-versus-host disease: A report of two cases. Int. J. Hematol. 92(1):204–207; 2010. 101. Liu, C.; Yu, S.; Zinn, K.; Wang, J.; Zhang, L.; Jia, Y.; Kappes, J. C.; Barnes, S.; Kimberly, R. P.; Grizzle, W. E.; Zhang, H.-G. Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. J. Immunol. 176(3):1375–1385; 2006. 102. Maiorana, A.; Fierabracci, A.; Cianfarani, S. Isolation and characterization of omental adipose progenitor cells in children: A potential tool to unravel the pathogenesis of metabolic syndrome. Horm. Res. 72(6):348–358; 2009. 103. Martens, T. P.; See, F.; Schuster, M. D.; Sondermeijer, H. P.; Hefti, M. M.; Zannettino, A.; Gronthos, S.; Seki, T.; Itescu, S. Mesenchymal lineage precursor cells induce vascular network formation in ischemic myocardium. Nat. Clin. Pract. Cardiovasc. Med. 3(Suppl 1):18–22; 2006. 104. Martin, P. J.; Uberti, J. P.; Soiffer, R. J.; Klingemann, H.; Waller, E. K.; Daly, A. S.; Herrmann, R. P.; Kebriaei, P. Prochymal improves response rates in patients with steroid-refractory acute graft versus host disease (SR-GVHD) involving the liver and gut: Results of a randomized, placebo-controlled, multicenter phase III trial in GVHD. Biol. Blood Marrow Transplant. 16(2):S169–S170; 2010. 105. Mathivanan, S.; Ji, H.; Simpson, R. J. Exosomes: Extracellular organelles important in intercellular communication. J. Proteomics 73(10):1907–1920; 2010. 106. Mathivanan, S.; Simpson, R. J. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics 9(21):4997– 5000; 2009. 107. Meckes, D. G. Jr.; Raab-Traub, N. Microvesicles and viral infection. J. Virol. 85(24):12844–12854; 2011. 108. Merchant, M. L.; Powell, D. W.; Wilkey, D. W.; Cummins, T. D.; Deegens, J. K.; Rood, I. M.; McAfee, K. J.; Fleischer, C.; Klein, E.; Klein, J. B. Microfiltration isolation of human urinary exosomes for characterization by MS. Proteomics Clin. Appl. 4(1):84–96; 2010. 109. Miksa, M.; Wu, R.; Dong, W.; Das, P.; Yang, D.; Wang, P. Dendritic cell-derived exosomes containing milk fat globule epidermal growth factor-factor VIII attenuate proinflammatory responses in sepsis. Shock 25(6):586–593; 2006. 110. Miksa, M.; Wu, R.; Dong, W.; Komura, H.; Amin, D.; Ji, Y.; Wang, Z.; Wang, H.; Ravikumar, T. S.; Tracey, K. J.; Wang, P. Immature dendritic cell-derived exosomes rescue septic animals via milk fat globule epidermal growth factor-factor VIII. J. Immunol. 183(9):5983–5990; 2009. 111. Milanesi, A.; Lee, J.-W.; Li, Z.; Da Sacco, S.; Villani, V.; Cervantes, V.; Perin, L.; Yu, J. S. b-Cell regeneration mediated by human bone marrow mesenchymal stem cells. PLoS One 7(8):e42177; 2012. 112. Mills, C. Osiris Therapeutics announces preliminary results for Prochymal phase III GvHD trials; 2009. Available

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

113.

114.

115.

116.

117. 118.

119.

120. 121.

122.

123.

124.

125.

from http://investor.osiris.com/releasedetail.cfm?ReleaseID= 407404 Mokarizadeh, A.; Delirezh, N.; Morshedi, A.; Mosayebi, G.; Farshid, A.-A.; Mardani, K. Microvesicles derived from mesenchymal stem cells: Potent organelles for induction of tolerogenic signaling. Immunol. Lett. 147(1–2):47– 54; 2012. Monleon, I.; Martinez-Lorenzo, M. J.; Monteagudo, L.; Lasierra, P.; Taules, M.; Iturralde, M.; Pineiro, A.; Larrad, L.; Alava, M. A.; Naval, J.; Anel, A. Differential secretion of Fas ligand- or APO2 ligand/TNF-related apoptosisinducing ligand-carrying microvesicles during activationinduced death of human T cells. J. Immunol. 167(12): 6736–6744; 2001. Montecalvo, A.; Shufesky, W. J.; Stolz, D. B.; Sullivan, M. G.; Wang, Z.; Divito, S. J.; Papworth, G. D.; Watkins, S. C.; Robbins, P. D.; Larregina, A. T.; Morelli, A. E. Exosomes as a short-range mechanism to spread alloantigen between dendritic cells during T cell allorecognition. J. Immunol. 180(5):3081–3090; 2008. Morse, M. A.; Garst, J.; Osada, T.; Khan, S.; Hobeika, A.; Clay, T. M.; Valente, N.; Shreeniwas, R.; Sutton, M. A.; Delcayre, A.; Hsu, D. H.; Le Pecq, J. B.; Lyerly, H. K. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J. Transl. Med. 3(1): 9; 2005. Moskovich, O.; Fishelson, Z. Live cell imaging of outward and inward vesiculation induced by the complement c5b-9 complex. J. Biol. Chem. 282(41):29977–29986; 2007. Muller-Ehmsen, J.; Krausgrill, B.; Burst, V.; Schenk, K.; Neisen, U. C.; Fries, J. W. U.; Fleischmann, B. K.; Hescheler, J.; Schwinger, R. H. G. Effective engraftment but poor mid-term persistence of mononuclear and mesenchymal bone marrow cells in acute and chronic rat myocardial infarction. J. Mol. Cell. Cardiol. 41(5):876–884; 2006. Muraglia, A.; Cancedda, R.; Quarto, R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J. Cell Sci. 113(Pt 7):1161–1166; 2000. Nadri, S.; Soleimani, M. Comparative analysis of mesenchymal stromal cells from murine bone marrow and amniotic fluid. Cytotherapy 9(8):729–737; 2007. Nauta, A. J.; Kruisselbrink, A. B.; Lurvink, E.; Willemze, R.; Fibbe, W. E. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocytederived dendritic cells. J. Immunol. 177(4):2080–2087; 2006. Nazarenko, I.; Rana, S.; Baumann, A.; McAlear, J.; Hellwig, A.; Trendelenburg, M.; Lochnit, G.; Preissner, K. T.; Zoller, M. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 70(4):1668–1678; 2010. Nolte-’t Hoen, E. N. M.; Buschow, S. I.; Anderton, S. M.; Stoorvogel, W.; Wauben, M. H. M. Activated T cells recruit exosomes secreted by dendritic cells via LFA-1. Blood 113(9):1977–1981; 2009. Nolte-’t Hoen, E. N. M.; van der Vlist, E. J.; Aalberts, M.; Mertens, H. C. H.; Bosch, B. J.; Bartelink, W.; Mastrobattista, E.; van Gaal, E. V. B.; Stoorvogel, W.; Arkesteijn, G. J. A.; Wauben, M. H. M. Quantitative and qualitative flow cytometric analysis of nanosized cell-derived membrane vesicles. Nanomedicine 8(5):712–720; 2012. Patki, S.; Kadam, S.; Chandra, V.; Bhonde, R. Human breast milk is a rich source of multipotent mesenchymal stem cells. Hum. Cell 23(2):35–40; 2010.

147

126. Peche, H.; Heslan, M.; Usal, C.; Amigorena, S.; Cuturi, M. C. Presentation of donor major histocompatibility complex antigens by bone marrow dendritic cell-derived exosomes modulates allograft rejection. Transplantation 76(10):1503– 1510; 2003. 127. Penicka, M.; Widimsky, P.; Kobylka, P.; Kozak, T.; Lang, O. Images in cardiovascular medicine. Early tissue distribution of bone marrow mononuclear cells after transcoronary transplantation in a patient with acute myocardial infarction. Circulation 112(4):63–65; 2005. 128. Phinney, D. G. Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy. J. Cell. Biochem. 113(9):2806–2812; 2012. 129. Poutsiaka, D. D.; Schroder, E. W.; Taylor, D. D.; Levy, E. M.; Black, P. H. Membrane vesicles shed by murine melanoma cells selectively inhibit the expression of Ia antigen by macrophages. J. Immunol. 134(1):138–144; 1985. 130. Qazi, K. R.; Gehrmann, U.; Domange Jordo, E.; Karlsson, M. C. I.; Gabrielsson, S. Antigen-loaded exosomes alone induce Th1-type memory through a B-cell-dependent mechanism. Blood 113(12):2673–2683; 2009. 131. Qazi, K. R.; Torregrosa Paredes, P.; Dahlberg, B.; Grunewald, J.; Eklund, A.; Gabrielsson, S. Proinflammatory exosomes in bronchoalveolar lavage fluid of patients with sarcoidosis. Thorax 65(11):1016–1024; 2010. 132. Quah, B. J. C.; O’Neill, H. C. Mycoplasma contaminants present in exosome preparations induce polyclonal B cell responses. J. Leukoc. Biol. 82(5):1070–1082; 2007. 133. Quarto, R.; Mastrogiacomo, M.; Cancedda, R.; Kutepov, S. M.; Mukhachev, V.; Lavroukov, A.; Kon, E.; Marcacci, M. Repair of large bone defects with the use of autologous bone marrow stromal cells. N. Engl. J. Med. 344(5): 385–386; 2001. 134. Rackham, C. L.; Dhadda, P. K.; Chagastelles, P. C.; Simpson, S. J. S.; Dattani, A. A.; Bowe, J. E.; Jones, P. M.; King, A. J. F. Pre-culturing islets with mesenchymal stromal cells using a direct contact configuration is beneficial for transplantation outcome in diabetic mice. Cytotherapy 15(4):449–459; 2013. 135. Raposo, G.; Nijman, H. W.; Stoorvogel, W.; Liejendekker, R.; Harding, C. V.; Melief, C. J.; Geuze, H. J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 183(3):1161–1172; 1996. 136. Rasmusson, I.; Le Blanc, K.; Sundberg, B.; Ringden, O. Mesenchymal stem cells stimulate antibody secretion in human B cells. Scand. J. Immunol. 65(4):336–343; 2007. 137. Rasmusson, I.; Ringden, O.; Sundberg, B.; Le Blanc, K. Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation 76(8):1208–1213; 2003. 138. Raynaud, C. M.; Rafii, A. The necessity of a systematic approach for the use of MSCs in the clinical setting. Stem Cells Int. 2013:892340; 2013. 139. Ren, G.; Chen, X.; Dong, F.; Li, W.; Ren, X.; Zhang, Y.; Shi, Y. Concise review: Mesenchymal stem cells and translational medicine: Emerging issues. Stem. Cells Transl. Med. 1(1):51–58; 2012. 140. Ren, G.; Su, J.; Zhang, L.; Zhao, X.; Ling, W.; L’Huillie, A.; Zhang, J.; Lu, Y.; Roberts, A. I.; Ji, W.; Zhang, H.; Rabson, A. B.; Shi, Y. Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells 27(8):1954–1962; 2009. 141. Ren, G.; Zhang, L.; Zhao, X.; Xu, G.; Zhang, Y.; Roberts, A. I.; Zhao, R. C.; Shi, Y. Mesenchymal stem cell-mediated

148

142. 143. 144.

145.

146. 147.

148.

149.

150. 151.

152.

153.

154. 155. 156. 157. 158.

FIERABRACCI ET AL.

immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell 2(2):141–150; 2008. Ringden, O.; Keating, A. Mesenchymal stromal cells as treatment for chronic GVHD. Bone Marrow Transplant. 46(2):163–164; 2011. Sadallah, S.; Eken, C.; Schifferli, J. A. Ectosomes as modulators of inflammation and immunity. Clin. Exp. Immunol. 163(1):26–32; 2011. Sarugaser, R.; Lickorish, D.; Baksh, D.; Hosseini, M. M.; Davies, J. E. Human umbilical cord perivascular (HUCPV) cells: A source of mesenchymal progenitors. Stem Cells 23(2):220–229; 2005. Sato, K.; Ozaki, K.; Oh, I.; Meguro, A.; Hatanaka, K.; Nagai, T.; Muroi, K.; Ozawa, K. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 109(1):228–234; 2007. Scudellari, M. The little cell that could. Scientist 26(7): 67–69; 2012. Segura, E.; Nicco, C.; Lombard, B.; Veron, P.; Raposo, G.; Batteux, F.; Amigorena, S.; Thery, C. ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood 106(1):216–223; 2005. Shih, D. T.; Lee, D. C.; Chen, S. C.; Tsai, R. Y.; Huang, C. T.; Tsai, C. C.; Shen, E. Y.; Chiu, W. T. Isolation and characterization of neurogenic mesenchymal stem cells in human scalp tissue. Stem Cells 23(7):1012–1020; 2005. Spaggiari, G. M.; Capobianco, A.; Becchetti, S.; Mingari, M. C.; Moretta, L. Mesenchymal stem cell-natural killer cell interactions: Evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood 107(4):1484–1490; 2006. Subra, C.; Laulagnier, K.; Perret, B.; Record, M. Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Biochimie 89(2):205–212; 2007. Sun, L.; Akiyama, K.; Zhang, H.; Yamaza, T.; Hou, Y.; Zhao, S.; Xu, T.; Le, A.; Shi, S. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells 27(6): 1421–1432; 2009. Sun, L.; Wang, D.; Liang, J.; Zhang, H.; Feng, X.; Wang, H.; Hua, B.; Liu, B.; Ye, S.; Hu, X.; Xu, W.; Zeng, X.; Hou, Y.; Gilkeson, G. S.; Silver, R. M.; Lu, L.; Shi, S. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum. 62(8):2467–2475; 2010. Szajnik, M.; Czystowska, M.; Szczepanski, M. J.; Mandapathil, M.; Whiteside, T. L. Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg). PLoS One 5(7): e11469; 2010. Taylor, D. D.; Akyol, S.; Gercel-Taylor, C. Pregnancyassociated exosomes and their modulation of T cell signaling. J. Immunol. 176(3):1534–1542; 2006. Taylor, D. D.; Gercel-Taylor, C. Exosomes/microvesicles: Mediators of cancer-associated immunosuppressive microenvironments. Semin. Immunopathol. 33(5):441–454; 2011. Tetta, C.; Bruno, S.; Fonsato, V.; Deregibus, M. C.; Camussi, G. The role of microvesicles in tissue repair. Organogenesis 7(2):105–115; 2011. Theise, N. D. Stem cell research: Elephants in the room. Mayo Clin. Proc. 78(8):1004–1009; 2003. Thery, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and characterization of exosomes from cell culture

159.

160. 161. 162.

163.

164.

165. 166.

167.

168.

169.

170.

171.

supernatants and biological fluids. Curr. Protoc. Cell Biol. 30:3.22.1–3.22.29; 2006. Thery, C.; Duban, L.; Segura, E.; Veron, P.; Lantz, O.; Amigorena, S. Indirect activation of naive CD4+ T cells by dendritic cell-derived exosomes. Nat. Immunol. 3(12): 1156–1162; 2002. Thery, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 9(8):581–593; 2009. Thirabanjasak, D.; Tantiwongse, K.; Thorner, P. S. Angiomyeloproliferative lesions following autologous stem cell therapy. J. Am. Soc. Nephrol. 21(7):1218–1222; 2010. Timmers, L.; Lim, S. K.; Arslan, F.; Armstrong, J. S.; Hoefer, I. E.; Doevendans, P. A.; Piek, J. J.; El Oakley, R. M.; Choo, A.; Lee, C. N.; Pasterkamp, G.; de Kleijn, D. P. V. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 1(2):129–137; 2007. Timmers, L.; Lim, S. K.; Hoefer, I. E.; Arslan, F.; Lai, R. C.; van Oorschot, A. A. M.; Goumans, M. J.; Strijder, C.; Sze, S. K.; Choo, A.; Piek, J. J.; Doevendans, P. A.; Pasterkamp, G.; de Kleijn, D. P. V. Human mesenchymal stem cell-conditioned medium improves cardiac function following myocardial infarction. Stem Cell Res. 6(3): 206–214; 2011. Traggiai, E.; Volpi, S.; Schena, F.; Gattorno, M.; Ferlito, F.; Moretta, L.; Martini, A. Bone marrow-derived mesenchymal stem cells induce both polyclonal expansion and differentiation of B cells isolated from healthy donors and systemic lupus erythematosus patients. Stem Cells 26(2): 562–569; 2008. Tyndall, A. Successes and failures of stem cell transplantation in autoimmune diseases. Hematology Am. Soc. Hematol. Educ. Program 2011:280–284; 2011. Urban, V. S.; Kiss, J.; Kovacs, J.; Gocza, E.; Vas, V.; Monostori, E.; Uher, F. Mesenchymal stem cells cooperate with bone marrow cells in therapy of diabetes. Stem Cells 26(1):244–253; 2008. Utsugi-Kobukai, S.; Fujimaki, H.; Hotta, C.; Nakazawa, M.; Minami, M. MHC class I-mediated exogenous antigen presentation by exosomes secreted from immature and mature bone marrow derived dendritic cells. Immunol. Lett. 89(2–3):125–131; 2003. van der Pol, E.; Hoekstra, A. G.; Sturk, A.; Otto, C.; van Leeuwen, T. G.; Nieuwland, R. Optical and non-optical methods for detection and characterization of microparticles and exosomes. J. Thromb. Haemost. 8(12):2596–2607; 2010. van Dommelen, S. M.; Vader, P.; Lakhal, S.; Kooijmans, S. A. A.; van Solinge, W. W.; Wood, M. J. A.; Schiffelers, R. M. Microvesicles and exosomes: Opportunities for cell-derived membrane vesicles in drug delivery. J. Control. Release 161(2):635–644; 2012. Veriter, S.; Gianello, P.; Igarashi, Y.; Beaurin, G.; Ghyselinck, A.; Aouassar, N.; Jordan, B.; Gallez, B.; Dufrane, D. Improvement of subcutaneous bioartificial pancreas vascularization and function by co-encapsulation of pig islets and mesenchymal stem cells in primates. Cell Transplant. 23(11):1349–1364; 2014. Veron, P.; Segura, E.; Sugano, G.; Amigorena, S.; Thery, C. Accumulation of MFG-E8/lactadherin on exosomes from immature dendritic cells. Blood Cells Mol. Dis. 35(2): 81–88; 2005.

IMMUNOMODULATION BY MSC-DERIVED MICROVESICLES

172. Viaud, S.; Terme, M.; Flament, C.; Taieb, J.; Andre, F.; Novault, S.; Escudier, B.; Robert, C.; Caillat-Zucman, S.; Tursz, T.; Zitvogel, L.; Chaput, N. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: A role for NKG2D ligands and IL-15R alpha. PLoS One 4(3):e4942; 2009. 173. Wakitani, S.; Imoto, K.; Yamamoto, T.; Saito, M.; Murata, N.; Yoneda, M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage 10(3):199–206; 2002. 174. Wang, H. S.; Hung, S. C.; Peng, S. T.; Huang, C. C.; Wei, H. M.; Guo, Y. J.; Fu, Y. S.; Lai, M. C.; Chen, C. C. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells 22(7):1330–1337; 2004. 175. Wieckowski, E. U.; Visus, C.; Szajnik, M.; Szczepanski, M. J.; Storkus, W. J.; Whiteside, T. L. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J. Immunol. 183(6):3720–3730; 2009. 176. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Thery, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; Angevin, E.; Amigorena, S.; Zitvogel, L. Tumorderived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 7(3):297–303; 2001. 177. Yamaza, T.; Kentaro, A.; Chen, C.; Liu, Y.; Shi, Y.; Gronthos, S.; Wang, S.; Shi, S. Immunomodulatory properties of stem cells from human exfoliated deciduous teeth. Stem Cell Res. Ther. 1(1):5; 2010. 178. Yang, C.; Robbins, P. D. The roles of tumor-derived exosomes in cancer pathogenesis. Clin. Dev. Immunol. 2011:842849; 2011. 179. Yang, X.; Meng, S.; Jiang, H.; Chen, T.; Wu, W. Exosomes derived from interleukin-10-treated dendritic cells can inhibit trinitrobenzene sulfonic acid-induced rat colitis. Scand. J. Gastroenterol. 45(10):1168–1177; 2010. 180. Yeo, R. W.; Lai, R. C.; Zhang, B.; Tan, S. S.; Yin, Y.; Teh, B. J.; Lim, S. K. Mesenchymal stem cell: An efficient

149

181. 182.

183.

184.

185.

186.

187.

188.

mass producer of exosomes for drug delivery. Adv. Drug Deliv. Rev. 65(3):336–341; 2013. Yi, T.; Song, S. U. Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications. Arch. Pharm. Res. 35(2):213–221; 2012. Youd, M.; Blickarz, C.; Woodworth, L.; Touzjian, T.; Edling, A.; Tedstone, J.; Ruzek, M.; Tubo, R.; Kaplan, J.; Lodie, T. Allogeneic mesenchymal stem cells do not protect NZBxNZW F1 mice from developing lupus disease. Clin. Exp. Immunol. 161(1):176–186; 2010. Zappia, E.; Casazza, S.; Pedemonte, E.; Benvenuto, F.; Bonanni, I.; Gerdoni, E.; Giunti, D.; Ceravolo, A.; Cazzanti, F.; Frassoni, F.; Mancardi, G.; Uccelli, A. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood 106(5): 1755–1761; 2005. Zhang, H. G.; Liu, C.; Su, K.; Su, K.; Yu, S.; Zhang, L.; Zhang, S.; Wang, J.; Cao, X.; Grizzle, W.; Kimberly, R. P. A membrane form of TNF-alpha presented by exosomes delays T cell activation-induced cell death. J. Immunol. 176(12):7385–7393; 2006. Zheng, M.; Fang, H.; Tsuruoka, T.; Tsuji, T.; Sasaki, T.; Hakomori, S. Regulatory role of GM3 ganglioside in alpha 5 beta 1 integrin receptor for fibronectin-mediated adhesion of FUA169 cells. J. Biol. Chem. 268(3):2217–2222; 1993. Zhou, K.; Zhang, H.; Jin, O.; Feng, X.; Yao, G.; Hou, Y.; Sun, L. Transplantation of human bone marrow mesenchymal stem cell ameliorates the autoimmune pathogenesis in MRL/lpr mice. Cell Mol. Immunol. 5(6):417–424; 2008. Zhou, Y.; Day, A.; Haykal, S.; Keating, A.; Waddell, T. K. Mesenchymal stromal cells augment CD4+ and CD8+ T-cell proliferation through a CCL2 pathway. Cytotherapy 15(10):1195–1207; 2013. Zuk, P.; M, M. Z.; P, P. A.; Ugarte, D. D.; Huang, J.; Mizuno, H.; Alfonso, Z.; Fraser, J.; Benhaim, P.; Hedrick, M. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell 13:4279–4295; 2002.

Recent advances in mesenchymal stem cell immunomodulation: the role of microvesicles.

Mesenchymal stem cells are the most widely used cell phenotype for therapeutic applications, the main reasons being their well-established abilities t...
324KB Sizes 0 Downloads 0 Views