Abstract
Mesenchymal stem cells (MSCs) are a group of fibroblast-like multipotent mesenchymal stromal cells that have the ability to differentiate into osteoblasts, adipocytes, and chondrocytes. Recent studies have demonstrated that MSCs possess a unique ability to exert suppressive and regulatory effects on both adaptive and innate immunity in an autologous and allogeneic manner. A vital step in stem cell transplantation is overcoming the potential graft-versus-host disease, which is a limiting factor to transplantation success. Given that MSCs attain powerful differentiation capabilities and also present immunosuppressive properties, which enable them to survive host immune rejection, MSCs are of great interest. Due to their ability to differentiate into different cell types and to suppress and modulate the immune system, MSCs are being developed for treating a plethora of diseases, including immune disorders. Moreover, in recent years, MSCs have been genetically engineered to treat and sometimes even cure some diseases, and the use of MSCs for cell therapy presents new perspectives for overcoming tissue rejection. In this review, we discuss the potential extrinsic and intrinsic mechanisms that underlie MSCs’ unique ability to modulate inflammation, and both innate and adaptive immunity.
Keywords: graft versus host disease, immunity, inflammation, mesenchymal stem cells, stem cell transplantation
I. INTRODUCTION
Stem cells (SCs) were first used as a form of cell therapy in 1968 when bone marrow stem cells (BMSCs) were used to treat lymphoma. The transplanted bone marrow contained SCs, which were able to produce all cell components of the blood and supporting matrix of the bone marrow. Today, BMSCs are routinely transplanted to treat a variety of blood and bone marrow diseases, blood cancers, immune disorders, and lysosomal storage diseases. Recently, peripheral blood SCs and umbilical cord blood SCs have also been used to treat some of the same blood-based diseases. These treatments involve the use of versatile hematopoietic SCs that can readily differentiate into all blood cell types and efficiently repopulate the recipient's bone marrow, making SCs an excellent tool for cell therapy.
Over the past decade, much attention has been given to mesenchymal stem cells (MSCs), a group of fibroblast-like multipotent mesenchymal stromal cells.1,2 MSCs were originally identified as multipotent stromal precursor cells in bone marrow by Friedenstein and coworkers in the 1970s.3-5 These cells present the in vitro ability to differentiate into osteoblasts, adipocytes, and chondroblasts. Currently recognized markers for MSCs are negativity for the cell surface markers CD45, CD34, CD14, CD11b, CD79α, CD19 and HLA-DR, and positivity for CD44, CD90, CD105, CD79, and CD29. MSCs possess a unique ability to exert suppressive and regulatory effects on both adaptive and innate immune cells in an autologous and allogeneic manner.6
The MSC cell surface receptors involved in immune regulation are listed in Table 1. MSCs can be isolated from a variety of tissues, such as bone marrow (BM), umbilical cord (U),7 adipose tissue (AT),8 dental pulp (DP),9 hair follicle (HF), and corneal stroma,10,11 making them readily available. MSCs isolated from BM, AT, DP, and HF have the advantage that they can be used for autologous transplantation. Moreover, DPSCs and HFSCs may be obtained as a by-product after removal of molar teeth and by noninvasive surgery, respectively, making them very attractive sources of SCs.
Table 1.
Cell surface receptors involved in MSC inflammatory cell cross-talk
| Surface receptor | Role |
|---|---|
| • MSCs | |
| HLA class II | Present antigens to T-cells161 |
| CXCR4 | Stimulate cell migration162,163 |
| CD44 | Cell-cell communication164-166 |
| Integrin | Cell-cell communication167,168; α5β1169 |
| Notch | Epithelial-to-mesenchymal transition (EMT)170 |
| ICAM-1/2 | Homing through migration and adhesion171 |
| VCAM-1 (CD106) | Mobilization of granulocyte-macrophage colony-forming units172,173 |
| ALCAM (CD166) | Regulation of tumor cell invasion174,175 |
| CD90 | Mechanical stimuli receptor176; immunosuppressive activity177 |
| CD105 | Allogenic modulation of immune cells176 |
| CD45 | Antigen receptor signal transduction and lymphocyte development178 |
| CD34 | Decrease cell adhesion179 |
| CD14/ CD11b | LPS receptor for promoting TLR4 endocytosis and interferon expression180-182 |
| • T-cells | |
| CCR10 | Migration of T cells,183 chemokine receptor184 |
II. IMMUNOSUPPRESSIVE PROPERTIES OF MESENCHYMAL STEM CELLS
An important step in regenerative medicine is to engineer cells or tissues which are then transplanted to restore normal tissue function. However, graft-versus-host disease limits transplantation success. MSCs in addition to powerful differentiation capabilities also present immunosuppressive properties, which aid them in surviving host immune rejection. Therefore, the use of MSCs for cell therapy presents new perspectives for overcoming tissue rejection.
The first report to describe the immunosuppressive properties of MSCs was by Le Blanc, who found that transplantation of allogeneic MSC infusions, at the time of mismatched skin grafts in baboons, suppressed immune cells, preventing the graft-versus-host attack.12 Since then, evidence has mounted with regard to how MSCs modulate immune cells, and over the past 10 years, MSCs have been shown to alter the cytokine secretion profile of subpopulations of inflammatory cells into a more anti-inflammatory phenotype,13 to stimulate macrophages into attaining higher phagocytic activity,14,15 to impair proliferation and differentiation of B cells, and to inhibit natural killer function.16
MSCs have been shown to modulate the innate immune system. They actively regulate macrophages,17,18 neutrophils,19,20 dendritic cells,21-23 and natural killer cells,24,25 thereby modulating the first line of defense. MSCs also regulate the adaptive immune system, i.e., B and T lymphocytes.26 In the case of T cells, MSCs primarily promote the activation of T regulatory cells.27,28 In the case of B cells, MSCs inhibit B-cell proliferation, plasma-cell differentiation, and antibody production, all in a T-cell-dependent manner.29 To date, MSCs have been shown to regulate the innate and adaptive immune responses primarily in a cytokine-dependent manner through the secretion of transforming growth factor-beta (TGFβ), interferon gamma (IFNγ), prostaglandin E2 (PGE2), interleukin (IL)-10 and tumor necrosis factor-alpha (TNFα). (See Table 2.) However, we have recently shown that UMSCs have unique immunosuppressive properties that can prevent xenograft rejection, making these cells extremely attractive for cell therapy.27 Our study demonstrated that UMSCs actively regulate immune cells, inhibiting M1 polarization and promoting M2 polarization in an extracellular matrix-dependent manner.
Table 2.
Immunoregulatory cytokines secreted by MSCs
| Secreted cytokines | Role |
|---|---|
| HLA | Present antigens to T-cells185; inhibitory interactions and NK cell activation186.187 |
| TGF-β | Induce epithelial to mesenchymal transition (EMT),188 pluripotency maintenance189 |
| IL-10/Galectin-3 | Immunosuppression190-194 |
| Leukemia inhibitory factor (LIF) | Transplantation tolerance37,195 |
| Semaphorin-3A | Endogenous antiangiogenic agent196,197 |
| TNF-α/INF-γ | Immunomodulation13,198 |
Macrophages present different functional states, which dictate their involvement in either promoting the immune response (recruiting and activating inflammatory cells) or resolving the immune response (secreting anti-inflammatory cytokines and having increased apoptotic potential).30-32 The M1 and M2 macrophage phenotypes were first coined to differentiate between these two macrophage functional states. During the early stages of inflammation, M1 macrophages are essential for development of the acute response; however, failure to then downregulate this initial response and activate M2 macrophages, primarily involved in tissue repair, leads to chronic pathogenesis. Therefore, the ability of MSCs to stimulate M2 polarization and inhibit M1 polarization is of great pharmaceutical significance. However, MSCs have also been shown to secrete a plethora of cytokines that directly regulate immune cells both in the surrounding tissue and in the circulation. Therefore, it has been well established that MSCs directly modulate immune cells, both through the rich environment they produce (extrinsic mechanisms) and by means of the extensive cytokine panels they have been shown to secrete (intrinsic mechanisms). In the following sections, we will discuss the different intrinsic and extrinsic mechanisms described to date for MSCs, which directly modulate immune cells.
A. Intrinsic Mechanisms
Depending on the environment that MSCs are exposed to, they may adopt either an immunosuppressive or pro-inflammatory phenotype.33 Pro-inflammatory cytokines activate MSCs via the Toll-like receptor 3 (TLR3), which leads to an immunosuppressive phenotype: suppression of proliferation, activation of dendritic cells, macrophages, B cells, T cells, NK cells, NKT cells, and neutrophils. Thus, in a pro-inflammatory environment, MSCs respond by secreting anti-inflammatory cytokines, such as TGF-β, IL-10, leukemia inhibitory factor (LIF), galectin-1, galectin-3, and semaphorin-3A (Figure 1).34-39 All of these factors have been shown to suppress T cell proliferation and are upregulated following MSC stimulation with TNF-α and IFN-γ.40 On the other hand, TLR4 activation may lead to a pro-inflammatory MSC phenotype, where MSCs secrete pro-inflammatory factors, such as IL-6 and IL-8, and promote neutrophil and T cell activation enhancing the immune response.33,41
Figure 1.
Schematic of the intrinsic mechanisms by which MSCs modulate the inflammatory response. MSCs respond differently to pro-inflammatory and anti-inflammatory environments. In an anti-inflammatory environment (low levels of IFN-γ) TLR4 activation in MSCs leads to MHC-II expression and the presentation of antigens to T cells, which in turn secrete IFN-γ creating a pro-inflammatory environment. In a pro-inflammatory environment TLR3 activation leads to the secretion of anti-inflammatory cytokines such as IL-10, TGF-β, Sema3A and galectin-1 and -3 thereby creating an anti-inflammatory environment and inhibiting T-cell, neutrophil and macrophage (Mφ) recruitment. Thus, the delicate balance of IFN-γ in the MSC environment dictates MSC phenotype
IFN-γ levels have been shown to coordinate the pro- and anti-inflammatory properties of MSCs through a feedback loop (Figure 1).42 In addition, a delicate balance between MSCs, T-regulatory cells, and IFN-γ levels regulates the immune stimulatory and inhibitory properties of MSCs through this feedback loop.43 In the presence of low levels of IFN-γ, MSCs express MHC-II, which enables them to present antigens upon insult, leading to the activation of T-regulatory cells. The activated T-regulatory cells express IFN-γ, which then supresses MHC-II presentation by MSCs, thereby hampering the inflammatory process and concluding the feedback loop.
The immunosuppressive effects of BMSCs have been shown to be dependent on Notch-RBP-J (recombination signal binding protein-Jκ) signaling, which regulates the production of IL-6 and PGE2, using a murine lethal acute graft-versus-host disease model.44 Secretion of high levels of IL-6 by MSCs has been shown to downregulate the expression of MHC II, CD40, and CD86 on mature dendritic cells and reduce T-cell proliferation,45 while PGE2 inhibits both T-cell proliferation and activation.46,47 MSCs secreting PGE2 have been shown to be a potential effector for T-cell suppression.48 Interestingly, PGE2-sensitive T cells have been characterized by low production of IL-2 and IL-4, while PGE2-resistant T cells secrete high levels of IL-2, IL-4 or both.49 MSCs upregulate PGE2 upon IFN-γ and TNFα treatment, suggesting that T-cell regulation depends on the inflammatory microenvironment. MSCs have also been shown to induce macrophage reprogramming by PGE2 secretion, and the presence of either LPS or TNFα in the microenvironment is essential.50
MSCs have also been shown to inhibit the maturation of dendritic cells through the stimulation of IL-10 secretion, and by activating the JAK1 and STAT3 signalling pathway.51 BMSCs can differentiate mature dendritic cells into a distinct regulatory dendritic cell population that has lower CD1a, CD80, CD86 and CD40 expression, higher CD11b expression and does not stimulate T-cell proliferation.52 The nitric oxide pathway has also been shown to play a role in regulating T-cell activation. IFNγ-stimulated MSCs express high levels of nitric oxide synthase, which in turn supresses T-cell responsiveness, and MSCs have been shown to prevent graft-versus-host disease in both an IFNγ- and nitric oxide-dependent manner.53 MSCs have also been shown to regulate follicular helper T cells through the secretion of indoleamine 2,3-dioxygenase.54
UMSCs have been shown to be the MSCs with the greatest immune suppressive properties, and this has been correlated to the high levels of HLA-G (non-classical HLA with strong immune-inhibitory properties) that these cells express.55 Interestingly, UMSCs upregulate HLA-G expression when treated with IFNγ.55 Human UMSCs (hUMSCs) express an immunosuppressive isoform of HLA-I, but not HLA-DR (major histocompatibility complex [MHC] class II cell surface receptor), indicating that these cells have low immunogenicity.56 Furthermore, the expression of immune response-related surface antigens, such as CD40, CD40 ligand, CD80, and CD86 is absent on hUMSCs, allowing hUMSCs to escape the host immune attack.56,57 Similar to hUMSCs, hBMSCs and hATSCs are negative for immunologically relevant surface markers.58
MSCs have been shown to modulate inflammatory processes not only through the secretion of cytokines, but also by secreting systemic factors that can neutralize the activity of inflammatory cytokines.59 This study reveals that MSCs secrete systemic agents that neutralize pro-inflammatory mediators, which in turn attenuate generalized inflammation. MSCs secrete a soluble form of TNF receptor 1, which neutralizes TNFα, and again the presence of LPS in the microenvironment enhances the expression of TNF receptor 1 by MSCs.59
The intrinsic properties of MSCs for modulating inflammation are not restricted to cytokine secretion. We have also shown that umbilical cord mesenchymal stem cells (UMSCs) secrete exosomes.60 Recently, other groups have shown that these exosomes secreted by MSCs can modulate the inflammatory response (Figure 1). MSC-derived exosomes improve functional recovery after traumatic brain injury by reducing inflammation and by promoting both endogenous angiogenesis and neurogenesis.61 MSC-derived exosomes have also been shown to be active effectors of immune regulation, stimulating T-cell proliferation, inducing B cell-mediated tumor suppression, inducing apoptosis in activated cytotoxic T cells, promoting differentiation of monocytes into dendritic cells, and stimulating T regulatory and myeloid-suppressive cells.62-65 Infusion of immunologically active MSC-derived exosomes enhances the survival of allogeneic skin grafts in mice, inducing high levels of anti-inflammatory IL-10 and TGF-β1.66 These exosomes induce an attenuated pro-inflammatory cytokine response and enhance anti-inflammatory IL-10 expression, reminiscent of an M2 macrophage profile, which are capable of promoting tissue repair while limiting injury. Additionally, Peche et al found that treatment of heart allograft recipients with intravenously administered donor-type dendritic cell-derived exosomes delays acute allograft rejection and induces significant prolongation of allograft survival in a rat model.67 They also observed a significant decrease in both IFN-γ mRNA expression and graft-infiltrating leukocytes in the exosome-treated rats, suggesting immunomodulation of allograft rejection. MSC-derived exosomes have also been shown to inhibit the proliferation of Concanavalin A-activated lymphocytes.68 Currently, MSC-derived exosomes are also being developed as drug delivery agents.
In a proteomic study, analysis of the content of BM-MSC-derived exosomes from control and multiple myeloma patients revealed that BM-MSC-derived exosomes from multiple myeloma patients contained higher expression of oncogenic proteins, cytokines, and protein kinases, such as IL-5, MCP-1, junction plakoglobin and fibronectin, than control patients.69 MSC-like cells isolated from islet of Langerhans release highly immunostimulatory exosomes that activate autoreactive B and T cells that have been endogenously primed in a non-obese diabetic mouse model.70 Both the serum levels of exosomes and exosome-induced IFN-γ production have been positively correlated with disease progression in prediabetic mice, suggesting that the exosomes could be autoantigen carriers acting as an autoimmune trigger.70 The immunomodulatory properties of MSC-derived exosomes have also been previously tested in a model of stimulated T cells, revealing that exosomes exert an inhibitory effect on the activation, proliferation, and differentiation of T cells reducing IFN-γ release.71
Recently, gene ontology analysis has been used to predict micro-RNA (miRNA) targets in MSCs and MSC-derived exosomes that could be related to development and cell survival.72 MSC-derived exosome miRNAs were mostly associated with immune system regulation. These exosome-derived miRNAs suppressed specific targets when transferred to recipient cells, demonstrating the direct functionality of these mediators in cell-to-cell communication.73 Moreover, multipotent MSC-derived exosome-mediated transfer of miR-133b to neural cells has been shown to contribute towards neurite outgrowth.74
B. Extrinsic Mechanisms
Over the past decade, extensive evidence has demonstrated that there is significant cross-talk between MSCs and inflammatory cells, and that pro- and anti-inflammatory microenvironments significantly alter the expression profile of MSCs. Many cytokines have been implicated in this cross-talk between MSCs and inflammatory cells; however, the precise mechanism by which MSCs supress inflammatory cells, and also the mechanism by which MSCs sense pro- or anti-inflammatory environments, remain to be fully elucidated. Interestingly, over the past couple of years a new layer has been added to this MSC and inflammatory cell cross-talk: the extracellular matrix (ECM). MSCs have been shown to secrete a rich environment, which also plays a fundamental role in differentiation and immune suppression. We have recently demonstrated that UMSCs secrete a specific ECM that is composed of hyaluronan (HA), heavy chains (HCs), tumor necrosis factor-inducible gene 6 protein (TSG-6), and pentraxin 3 (PTX3/TSG14).27 (See Figure 2.) This specific glycocalyx inhibits the maturation of M1 macrophages and promotes the maturation of both M2 macrophages and T-regulatory cells.
Figure 2.
Schematic of the extrinsic mechanisms by which MSCs modulate the inflammatory response. MSCs express a rich glycocalyx composed of HA/HCs/TSG-6/PTX3 which actively suppresses inflammatory cells. A soluble form of HA/HCs/TSG-6/PTX3 inhibits maturation of M1 macrophages and promotes the maturation of M2 macrophages. The HA cables produced sequester macrophages maintaining them in a non-polarized state (M0).
Recently, Kota et al demonstrated that BM-MSCs secrete a HA-rich glycocalyx (extensive pericellular HA coat) when treated with FGF-2, and proposed that this pericellular HA forms a protective niche.75 This group also demonstrated that MSCs secrete HA cable-like structures which sequester/trap inflammatory cells.75 This group activated MSCs with Poly I:C in the presence of serum containing Inter-α–Inhibitor (IαI) and MSC-secreted TSG-6, which cross-linked these HA chains and also catalyzed the transfer of heavy chains onto the HA, enabling the formation of the cable-like structures, as previously shown by de la Motte et al.76 The formation of this HC/HA/TSG-6 matrix has also recently been shown to be essential for the settlement and differentiation of MSCs in muscles.77 In this case, TSG6, HA and IαI were crucial factors for MSCs to create a microenvironment that was critical for transplantation success.
Oh et al have shown that solely injecting recombinant human TSG6 into the corneas of mice following chemical burns ameliorates the inflammatory response and improves corneal opacity.78 This same group was able to reduce the rejection of mouse allogeneic corneal transplants by intravenous administration of MSCs, which were shown to be trapped primarily in the lungs and to increase TSG-6 levels.79 Dyer et al have recently shown that free TSG-6 impairs both the binding of CXCL8 to cell surface glycosaminoglycans (GAGs) and its transport across the endothelium, thereby impeding formation of the CXCL8 gradient required for neutrophil chemotaxis and extravasation.80 Also, BM-MSCs treated with pro-inflammatory macrophages lead to an increase in GAG expression.81
The fact that MSCs secrete a rich ECM microenvironment, which is essential for the settlement, differentiation, and immunosuppressive properties of MSCs, comes as no surprise. The ECM is crucial for directing cellular processes and maintaining tissue integrity.82-87 Moreover, the severe developmental phenotypes shown by knock-out mice for heparan sulfate, an important ECM component, is a testament to the importance of the ECM for creating microenvironments.88,89 Therefore, more studies should be directed toward unveiling the intricate ECM secreted by MSCs, which enables them to perform their vital tasks-- immune suppression and tissue repair. Moreover, given the importance the ECM plays on the tissue microenvironment, many groups are developing HA-based tissue scaffolds to use as a means of MSC delivery, which increase transplantation success.90,91 Therefore, the development of HA-based cell-delivery vehicles for regenerative therapies is an exciting, growing field with prospects of increasing transplantation efficiency. TSG-6 has recently been suggested as a biomarker for predicting the efficacy of MSCs for treating sterile inflammatory models of corneal injury.92
III. Therapeutic Potential of Mesenchymal Stem Cells
A. Treatment of Ocular Surface Diseases
1. Anterior Segment
The therapeutic potential of MSCs for treating the ocular surface has been widely explored, primarily because the cornea is easily accessible and is an immune-privileged tissue. MSCs have the ability to differentiate into epithelial cells93-95 and keratocytes.96 Our previous work has shown that UMSCs can be used to successfully treat the congenital lysosomal storage disorder mucopolysaccharidosis VII (MPS VII).60 This study shows that UMSCs transplanted into the corneal stroma of MPS VII mice secrete exosomes containing the deficient enzyme, which are taken up by host keratocytes enabling GAG catabolism, thereby reducing GAG accumulation in both the ECM and keratocytes.60 Moreover, one month after transplantation of UMSCs into mouse corneal stroma, these cells assume keratocyte-like morphology and express a keratocyte cell marker (Figure 3).
Figure 3.
UMSCs in vitro and in vivo. (A) UMSCs in culture prior to transplantation into mouse corneal stroma. (B) 1 month after transplantation UMSCs assume keratocyte-like morphology and express a keratocyte cell marker in the corneal stroma.
Great strides have been made in applying MSCs to treatment of corneal disorders, such as experimental limbal stem cell deficiency (LSCD).97 BMSCs and limbal epithelial stem cells are similarly efficacious for treating limbal cell deficiency after alkali burn.98 The ability of BMSCs to treat LSCD is attributed not only to transdifferentiation, but also to suppression of local inflammation and secretion of growth factors, thereby supporting the growth of residual corneal epithelial cells.98 The use of MSCs for treating corneal diseases has recently been reviewed by our group, and the discussion below focuses primarily on the use of MSCs for treating inflammatory response in the ocular surface (Figure 4).99
Figure 4.
Schematic of MSC therapeutic developments for treating inflammatory disorders of the ocular surface. Various clinical applications have been developed for treating inflammatory disorders associated with the corneal epithelium (listed below the image) and stroma (listed above the image). However MSC treatments for curing endothelial defects remain to be developed. Each line represents a study with the type of MSC used listed in bold, followed by the condition being treated and finally the mechanism by which the MSCs resolve the condition listed in parenthesis.
Systemically administered MSCs have been shown to home into the inflamed cornea.100,101 We have recently demonstrated the efficacy of human UMSCs to limit inflammation after alkali burn using the mouse model.27 The use of BMSCs was further demonstrated for limiting damage after alkali burn.92,102 Both human MSCs and human MSC-conditioned media were shown to improve cell survival after ethanol injury to corneal epithelial progenitor cells.103 Human MSC infusions have also been shown to prevent mouse allogenic corneal transplant rejection by suppressing inflammation.78
The success of these studies involving grafting human MSCs into animal models resides in the fact that MSCs actively modulate immune cells, enabling them to survive xenograft rejection. Both mouse and rabbit BMSCs transplanted onto the ocular surface have been shown to suppress local inflammation.104,105 Moreover, our recent work has demonstrated that UMSCs inhibit the polarization of M1 macrophages and lead to the maturation of M2 macrophages.
Great expectations reside in generating treatments that increase the levels of “therapeutic cells,” such as M2 macrophages, as a means of resolving inflammation as opposed to simply using immunosuppressors to hamper the whole immune system. Current findings indicate that MSCs show great improvement over current treatments, since they actively modulate immune cells into restorative phenotypes rather than simply supressing the immune system. The use of MSCs has recently been widely explored for preventing host-versus-graft disease. For these studies, MSCs are administered due to their immune-suppressive properties for preventing the rejection of transplanted tissues. BMSCs have been shown to promote the survival of a fully allo-MHC-mismatched corneal allograft by supressing both local and systemic inflammation.106
2. Posterior Segment
The use of MSCs to treat the posterior chamber of the eye has also been explored. Vitreoretinal and chorioretinal diseases are chronic, progressive conditions related to refractory retinal degeneration due to pathophysiological conditions. Some stem cell therapy strategies involving human embryonic stem cells (hESC) have been successfully used to treat age-related macular degeneration (AMD) and Stargardt's macular dystrophy107,108; however, MSC targeting as a choice of delivery is still under development and has not yet reached clinical trial level.
Recently, delivery of MSCs labelled with magnetic iron oxide nanoparticles has been investigated for targeting dystrophic retinas.109 These particles were well tolerated, and the MSCs remained viable, retaining their differentiation abilities after intravitreal and/or intravenous administration. The use of MSCs for treating non-neovascular dry AMD has also been explored. Dry AMD is primarily caused by the accumulation of reactive oxygen free radicals and lipid peroxide in retinal pigment epithelium (RPE) cells, leading to the activation of chronic inflammation and apoptosis, damaging outer nuclear layer photoreceptors.110 MSCs have the ability to cross-differentiate into RPE-like cells in vivo with similar morphological and phagocytic capabilities.111,112 This has also been accomplished by altering MSC gene expression prior to transplantation.113,114
Currently, cells are transplanted as subretinal masses, thereby limiting the number of cells injected and consequently the levels of photoreceptors that can be rescued in areas surrounding injection sites. This emphasizes the need for in-depth studies regarding the delivery method115,116 and the need/efficacy of gene manipulation of MSCs prior to treatment.
Another posterior segment malady is retinitis pigmentosa, which affects mainly photoreceptors, making it a more difficult target for regeneration. Recent studies have tried to access invasive117-121 and non-invasive122 delivery therapies, with MSCs being capable of surviving primarily in the outer layer of the retina. These cells were able to differentiate into RPE cells and photoreceptors, but whether there was any functional recovery of the retina remains to be investigated.
B. Treatment of Other Diseases
MSCs are still being explored for their great potential in cell replacement therapy, but their ability to alter the immune response is being vastly exploited for therapeutic purposes. Currently, clinical trials are underway that explore the use of MSCs for the treatment of steroid-refractory graft-versus-host disease.123 Moreover, MSC pre-clinical studies have also been carried out showing that MSCs are effective for treating ischemic heart disease, ischemic kidney injury, type 2 diabetes mellitus, and, more recently, several models of sepsis.54,124-126 All these studies in one way or another have taken advantage of the valuable immunosuppressive properties of MSCs.
MSCs have also been suggested as a new alternative for repairing skin wounds. Topical administration of UMSCs together with skin microparticles after degree III deep burn wounds leads to the development of newborn skin and its appendages. New methods of delivering MSCs and supporting matrices are also currently being explored. Laminin-PAMs have been used to support MSCs in an in vivo model of transient stroke in order to increase MSC survival. This strategy achieved prolonged vascular endothelial growth factor (VEGF) release, increasing angiogenesis around the implantation site and supporting the migration of immature neurons towards the ischemic site.127
It is of particular interest that MSCs present tumor-homing properties and can therefore be used in tumor-targeting treatments. Qiao et al showed that MSCs transfected with adenoviruses carrying the OPG gene migrate to tumor sites and express OPG protein in mice bearing osteosarcoma, reducing tumor growth and inhibiting bone destruction.128 MSCs have also been shown to be useful for the delivery of therapeutic genes in a colon cancer liver metastasis mouse model.129 There are many other examples of MSCs being a potential vehicle for therapeutic delivery to tumors, including to brain tumors,130 lung tumors,131 hepatocellular carcinoma,132 and prostate cancer.133 These studies describe the use of MSCs for delivering not only therapeutic genes, but also therapeutic drugs and theranostic agents.
The characteristic of MSCs for suppressing the activity of several T-lymphocyte populations both in vivo and in vitro has been explored for therapeutic applications in treating T-cell mediated diseases,134-138 such as, graft-versus-host disease,123,139 and Crohn's disease,140 and to prevent rejection after organ transplantation.141
IV. Future Directions of Mesenchymal Stem Cell Research
Advances in MSC biology have raised expectations that diseases, injuries, and the need for an organ transplant can be ameliorated by transplanting MSCs. Recent advances in MSC research have broken the stem cell therapeutic paradigm with several groups, revealing that the therapeutic benefits of MSCs do not derive exclusively from extensive cell engraftment. Furthermore, studies have shown that solely soluble factors secreted by MSCs are sufficient for therapeutic benefits; for example, TSG-6 secreted by MSCs trapped in the lungs decreases early and excessive neutrophil infiltration into the heart of a mouse myocardial infarction model.79 Moreover, recently, MSC-derived exosomes have been successfully used to suppress the immune response.62,142-146 The ability of MSC-derived exosomes to suppress immune cells has attracted much attention for the development of MSC-derived exosomes for alternative treatments. MSC-derived exosomes are being tailored for development as vehicles for drug delivery; for example, siRNA has been delivered to the mouse brain by systemic injection of exosome-packaged siRNA.147 Furthermore, solely extracellular vesicles released by MSCs have been shown to promote angiogenesis following myocardial infarction and protect the cardiac tissue from ischemic injury.148
In recent years, within the field of MSCs, much attention has been dedicated to isolating active components from MSCs as therapeutic agents as opposed to using the MSCs themselves, of which TSG-6 is an excellent prototype. Many in vivo studies have shown that injecting TSG-6 alone reproduces many of the beneficial effects of MSCs. Further studies have demonstrated that administration of solely enriched MSC-secreted soluble compounds can suppress inflammation and lead to regeneration.149,150 Studies have also started to isolate active components from MSCs for promoting cell differentiation; for example, active components isolated from UMSCs can induce osteogenic differentiation of BM-MSCs. These studies therefore suggest that active components can be extracted from MSCs for treating chronic inflammatory disorders and promoting differentiation.130
Currently, there has been a growing trend toward developing MSCs for therapy due to their ability to actively suppress inflammation and not for their capacity to differentiate into bone, cartilage, or adipose tissue. MSC research avenues are now dedicated to exploring the immunomodulatory capability of these cells, and many preclinical and clinical studies are using MSCs as therapeutic agents for the treatment of autoimmune or degenerative diseases. Moreover, due to the self-renewal capacity of MSCs and their ability to differentiate into tissues of mesodermal origin, MSCs show great potential for being developed into a cell-based therapy for immune disorders or degenerative diseases. Adipose-derived MSCs have been effectively used to treat a murine model of Crohn's disease by downregulating pro-inflammatory cytokines, such as TNF-α, IL-12 and VEGF, and upregulating anti-inflammatory cytokine IL-10.151 Adipose-derived MSCs have also been used for ameliorating radiation-induced pulmonary fibrosis in Sprague-Dawley rats, whereby hepatocyte growth factor (HGF) and PGE2 levels increased and TNF-α and TGF-β1 levels decreased after MSC infusion.152 Another example is the use of BM-MSCs to treat chronic obstructive pulmonary disease, where the BM-MSCs alleviate airway inflammation and emphysema by promoting the down-regulation of cyclooxygenase-2 (COX-2) and COX-2-mediated PGE2 production in alveolar macrophages through p38 and ERK.153
MSCs provide therapeutic potential beyond immunosuppression and cell therapy. BM-MSCs have been used to treat a non-immune cuprizone model of multiple sclerosis, where intravenously transfused BM-MSCs were able to migrate to demyelinating sites, engraft and promote remyelination and reduce demyelination by oligodendrocytes. This study suggests that MSCs induce oligo/neuroprotection, demonstrating that MSCs can also support functional improvement of resident cells.154 BM-MSCs have also been shown to promote functional improvement following myocardial infarction,155 and the outcome can be further improved by using genetically modified or treated MSCs, such as HGF gene-modified BMMSCs,146 heat shock protein 27 (Hsp27) gene-modified MSCs,156 receptor activity-modifying protein 1 (RAMP1) gene-modified MSCs,157 or MSCs previously treated with IL-1β and TNF-α.158 Moreover, solely the supernatant of MSCs transfected with adenovirus carrying human heme oxygenase-1 gene has been shown to improve cardiac function suggesting an effect of paracrine-acting mediators secreted by MSCs.159
MSCs are attracting a great deal of attention from pharmaceutical companies, and commercial BM-MSC cell lines are now available, e.g., stempeucel®, which are adult allogeneic mesenchymal stem cells that are being used in a number of phase II clinical trials with clinical trials planned for 2015/2016. Stempeucel® have recently been used in a phase I/II study for treating humans after acute myocardial infarction with intravenous transplantation.160 These preliminary studies with commercial BM-MSCs have demonstrated that they are safe and well tolerated when administered intravenously.
V. Conclusion
A recent paradigm shift in MSC research has generated interest in using MSCs primarily for their immunosuppressive properties and secreted components, and not solely for use as cell replacement therapy. Moreover, interest is growing in isolating the active components from MSCs for therapeutic purposes, as opposed to using MSC cell engraftment. These new therapeutic avenues have generated great enthusiasm for MSC research.
Acknowledgments
The review was in part supported by NIH/NEI grant 1R01EY021768, Research to Prevent Blindness, Ohio Lions Eye Research Foundation, Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq) and Coordenação de Aperfeiçoamento de Pessoal de Nível (CAPES).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
The authors have no commercial or proprietary interest in any concept or product discussed in this article.
References
- 1.Horwitz EM, Le Blanc K, Dominici M, et al. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy. 2005;7:393–5. doi: 10.1080/14653240500319234. [DOI] [PubMed] [Google Scholar]
- 2.Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–36. doi: 10.1038/nri2395. [DOI] [PubMed] [Google Scholar]
- 3.Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3:393–403. doi: 10.1111/j.1365-2184.1970.tb00347.x. [DOI] [PubMed] [Google Scholar]
- 4.Friedenstein AJ, Chailakhyan RK, Latsinik NV, et al. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation. 1974;17:331–40. doi: 10.1097/00007890-197404000-00001. [DOI] [PubMed] [Google Scholar]
- 5.Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol. 1976;4:267–74. [PubMed] [Google Scholar]
- 6.Marigo I, Dazzi F. The immunomodulatory properties of mesenchymal stem cells. Semin Immunopathol. 2011;33:593–602. doi: 10.1007/s00281-011-0267-7. [DOI] [PubMed] [Google Scholar]
- 7.Wang JF, Wang LJ, Wu YF, et al. Mesenchymal stem/progenitor cells in human umbilical cord blood as support for ex vivo expansion of CD34(+) hematopoietic stem cells and for chondrogenic differentiation. Haematologica. 2004;89:837–44. [PubMed] [Google Scholar]
- 8.Rodriguez AM, Elabd C, Amri EZ, et al. The human adipose tissue is a source of multipotent stem cells. Biochimie. 2005;87:125–8. doi: 10.1016/j.biochi.2004.11.007. [DOI] [PubMed] [Google Scholar]
- 9.Pierdomenico L, Bonsi L, Calvitti M, et al. Multipotent mesenchymal stem cells with immunosuppressive activity can be easily isolated from dental pulp. Transplantation. 2005;80:836–42. doi: 10.1097/01.tp.0000173794.72151.88. [DOI] [PubMed] [Google Scholar]
- 10.Du Y, Funderburgh ML, Mann MM, et al. Multipotent stem cells in human corneal stroma. Stem Cells. 2005;23:1266–75. doi: 10.1634/stemcells.2004-0256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Branch MJ, Hashmani K, Dhillon P, et al. Mesenchymal stem cells in the human corneal limbal stroma. Invest Ophthalmol Vis Sci. 2012;53:5109–16. doi: 10.1167/iovs.11-8673. [DOI] [PubMed] [Google Scholar]
- 12.Le Blanc K. Immunomodulatory effects of fetal and adult mesenchymal stem cells. Cytotherapy. 2003;5:485–9. doi: 10.1080/14653240310003611. [DOI] [PubMed] [Google Scholar]
- 13.Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22. doi: 10.1182/blood-2004-04-1559. [DOI] [PubMed] [Google Scholar]
- 14.Kim J, Hematti P. Mesenchymal stem cell-educated macrophages: a novel type of alternatively activated macrophages. Exp Hematol. 2009;37:1445–53. doi: 10.1016/j.exphem.2009.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Krasnodembskaya A, Samarani G, Song Y, et al. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. Am J Physiol Lung Cell Mol Physiol. 2012;302:L1003–13. doi: 10.1152/ajplung.00180.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Corcione A, Benvenuto F, Ferretti E, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107:367–72. doi: 10.1182/blood-2005-07-2657. [DOI] [PubMed] [Google Scholar]
- 17.Song X, Xie S, Lu K, Wang C. Mesenchymal stem cells alleviate experimental asthma by inducing polarization of alveolar macrophages. Inflammation. 2015;38:485–92. doi: 10.1007/s10753-014-9954-6. [DOI] [PubMed] [Google Scholar]
- 18.Zanier ER, Marchesi F, Ortolano F, et al. Fractalkine receptor deficiency is associated with early protection but late worsening of outcome following brain trauma in mice. J Neurotrauma. 2015;32:1–13. doi: 10.1089/neu.2015.4041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Khan I, Zhang L, Mohammed M, et al. Effects of Wharton's jelly-derived mesenchymal stem cells on neonatal neutrophils. J Inflamm Res. 2015;8:1–8. doi: 10.2147/JIR.S71987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Teo GS, Yang Z, Carman CV, et al. Intravital imaging of mesenchymal stem cell trafficking and association with platelets and neutrophils. Stem Cells. 2015;33:265–77. doi: 10.1002/stem.1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mohammadpour H, Pourfathollah AA, Zarif MN, Tahoori MT. TNF-alpha modulates the immunosuppressive effects of MSCs on dendritic cells and T cells. Int Immunopharmacol. 2015;28:1009–17. doi: 10.1016/j.intimp.2015.07.045. [DOI] [PubMed] [Google Scholar]
- 22.Consentius C, Akyuz L, Schmidt-Lucke JA, et al. Mesenchymal stromal cells prevent allostimulation in vivo and control checkpoints of Th1 priming: Migration of human DC to lymph nodes and NK cell activation. Stem Cells. 2015;33:3087–99. doi: 10.1002/stem.2104. [DOI] [PubMed] [Google Scholar]
- 23.Abomaray FM, Al Jumah MA, Kalionis B, et al. Human chorionic villous mesenchymal stem cells modify the functions of human dendritic cells, and induce an anti-inflammatory phenotype in CD1+ dendritic cells. Stem Cell Rev. 2015;11:423–41. doi: 10.1007/s12015-014-9562-8. [DOI] [PubMed] [Google Scholar]
- 24.Qu M, Cui J, Zhu J, et al. Bone marrow-derived mesenchymal stem cells suppress NK cell recruitment and activation in PolyI:C-induced liver injury. Biochem Biophys Res Commun. 2015;466:173–9. doi: 10.1016/j.bbrc.2015.08.125. [DOI] [PubMed] [Google Scholar]
- 25.Lu Y, Liu J, Liu Y, et al. TLR4 plays a crucial role in MSC-induced inhibition of NK cell function. Biochem Biophys Res Commun. 2015;464:541–7. doi: 10.1016/j.bbrc.2015.07.002. [DOI] [PubMed] [Google Scholar]
- 26.Uccelli A, de Rosbo NK. The immunomodulatory function of mesenchymal stem cells: mode of action and pathways. Ann N Y Acad Sci. 2015:114–26. doi: 10.1111/nyas.12815. [DOI] [PubMed] [Google Scholar]
- 27.Coulson-Thomas VJ, Gesteira TF, Hascall V, Kao W. Umbilical cord mesenchymal stem cells suppress host rejection: the role of the glycocalyx. J Biol Chem. 2014;289:23465–81. doi: 10.1074/jbc.M114.557447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ghannam S, Pene J, Moquet-Torcy G, et al. Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol. 2010;185:302–12. doi: 10.4049/jimmunol.0902007. [DOI] [PubMed] [Google Scholar]
- 29.Rosado MM, Bernardo ME, Scarsella M, et al. Inhibition of B-cell proliferation and antibody production by mesenchymal stromal cells is mediated by T cells. Stem Cells Devel. 2015;24:93–103. doi: 10.1089/scd.2014.0155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–69. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Murray PJ, Allen JE, Biswas SK, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41:14–20. doi: 10.1016/j.immuni.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Xue J, Schmidt SV, Sander J, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40:274–88. doi: 10.1016/j.immuni.2014.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Waterman RS, Tomchuck SL, Henkle SL, Betancourt AM. A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an immunosuppressive MSC2 phenotype. PloS One. 2010;5:e10088. doi: 10.1371/journal.pone.0010088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bright JJ, Kerr LD, Sriram S. TGF-beta inhibits IL-2-induced tyrosine phosphorylation and activation of Jak-1 and Stat 5 in T lymphocytes. J Immunol. 1997;159:175–83. [PubMed] [Google Scholar]
- 35.Kalinski P. Regulation of immune responses by prostaglandin E2. J Immunol. 2012;188:21–8. doi: 10.4049/jimmunol.1101029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rasmusson I, Ringden O, Sundberg B, Le Blanc K. Mesenchymal stem cells inhibit lymphocyte proliferation by mitogens and alloantigens by different mechanisms. Exp Cell Res. 2005;305:33–41. doi: 10.1016/j.yexcr.2004.12.013. [DOI] [PubMed] [Google Scholar]
- 37.Nasef A, Mazurier C, Bouchet S, et al. Leukemia inhibitory factor: Role in human mesenchymal stem cells mediated immunosuppression. Cell Immunol. 2008;253:16–22. doi: 10.1016/j.cellimm.2008.06.002. [DOI] [PubMed] [Google Scholar]
- 38.Lepelletier Y, Lecourt S, Renand A, et al. Galectin-1 and semaphorin-3A are two soluble factors conferring T-cell immunosuppression to bone marrow mesenchymal stem cell. Stem Cells Devel. 2010;19:1075–9. doi: 10.1089/scd.2009.0212. [DOI] [PubMed] [Google Scholar]
- 39.Sioud M, Mobergslien A, Boudabous A, Floisand Y. Evidence for the involvement of galectin-3 in mesenchymal stem cell suppression of allogeneic T-cell proliferation. Scand J Immunol. 2010;71:267–74. doi: 10.1111/j.1365-3083.2010.02378.x. [DOI] [PubMed] [Google Scholar]
- 40.Sivanathan KN, Gronthos S, Rojas-Canales D, et al. Interferon-gamma modification of mesenchymal stem cells: implications of autologous and allogeneic mesenchymal stem cell therapy in allotransplantation. Stem Cell Rev. 2014;10:351–75. doi: 10.1007/s12015-014-9495-2. [DOI] [PubMed] [Google Scholar]
- 41.Yan H, Wu M, Yuan Y, et al. Priming of Toll-like receptor 4 pathway in mesenchymal stem cells increases expression of B cell activating factor. Biochem Biophys Res Commun. 2014;448:212–7. doi: 10.1016/j.bbrc.2014.04.097. [DOI] [PubMed] [Google Scholar]
- 42.Chan JL, Tang KC, Patel AP, et al. Antigen-presenting property of mesenchymal stem cells occurs during a narrow window at low levels of interferon-gamma. Blood. 2006;107:4817–24. doi: 10.1182/blood-2006-01-0057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sheng H, Wang Y, Jin Y, et al. A critical role of IFN gamma in priming MSC-mediated suppression of T cell proliferation through up-regulation of B7-H1. Cell Res. 2008;18:846–57. doi: 10.1038/cr.2008.80. [DOI] [PubMed] [Google Scholar]
- 44.Wang YC, Wang SH, Wei YN, et al. Notch-RBP-J signaling is required by bone marrow stromal cells for the treatment of acute graft versus host disease. Stem Cell Res. 2013;11:721–35. doi: 10.1016/j.scr.2013.04.009. [DOI] [PubMed] [Google Scholar]
- 45.Djouad F, Charbonnier LM, Bouffi C, et al. Mesenchymal stem cells inhibit the differentiation of dendritic cells through an interleukin-6-dependent mechanism. Stem Cells. 2007;25:2025–32. doi: 10.1634/stemcells.2006-0548. [DOI] [PubMed] [Google Scholar]
- 46.Hendricks A, Leibold W, Kaever V, Schuberth HJ. Prostaglandin E2 is variably induced by bacterial superantigens in bovine mononuclear cells and has a regulatory role for the T cell proliferative response. Immunobiology. 2000;201:493–505. doi: 10.1016/S0171-2985(00)80069-8. [DOI] [PubMed] [Google Scholar]
- 47.Leung KH, Mihich E. Prostaglandin modulation of development of cell-mediated immunity in culture. Nature. 1980;288:597–600. doi: 10.1038/288597a0. [DOI] [PubMed] [Google Scholar]
- 48.English K, Ryan JM, Tobin L, et al. Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4+CD25(High) forkhead box P3+ regulatory T cells. Clin Exp Immunol. 2009;156:149–60. doi: 10.1111/j.1365-2249.2009.03874.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.He X, Stuart JM. Prostaglandin E2 selectively inhibits human CD4+ T cells secreting low amounts of both IL-2 and IL-4. J Immunol. 1999;163:6173–9. [PubMed] [Google Scholar]
- 50.Nemeth K, Leelahavanichkul A, Yuen PS, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15:42–9. doi: 10.1038/nm.1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu WH, Liu JJ, Wu J, et al. Novel mechanism of inhibition of dendritic cells maturation by mesenchymal stem cells via interleukin-10 and the JAK1/STAT3 signaling pathway. PloS One. 2013;8:e55487. doi: 10.1371/journal.pone.0055487. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 52.Zhao ZG, Xu W, Sun L, et al. Immunomodulatory function of regulatory dendritic cells induced by mesenchymal stem cells. Immunol Invest. 2012;41:183–98. doi: 10.3109/08820139.2011.607877. [DOI] [PubMed] [Google Scholar]
- 53.Ren G, Zhang L, Zhao X, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–50. doi: 10.1016/j.stem.2007.11.014. [DOI] [PubMed] [Google Scholar]
- 54.Shi S, Jia S, Liu J, Chen G. Accelerated regeneration of skin injury by co-transplantation of mesenchymal stem cells from Wharton's jelly of the human umbilical cord mixed with microparticles. Cell Biochem Biophys. 2015;71:951–6. doi: 10.1007/s12013-014-0292-y. [DOI] [PubMed] [Google Scholar]
- 55.Ding DC, Chou HL, Chang YH, et al. Characterization of HLA-G and related immunosuppressive effects in human umbilical cord stroma derived stem cells. Cell Transplant. 2015;1263:321–36. doi: 10.3727/096368915X688182. [DOI] [PubMed] [Google Scholar]
- 56.Wang H, Qiu X, Ni P, et al. Immunological characteristics of human umbilical cord mesenchymal stem cells and the therapeutic effects of their transplantion on hyperglycemia in diabetic rats. Int J Molec Med. 2014;33:263–70. doi: 10.3892/ijmm.2013.1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang M, Yang Y, Yang D, et al. The immunomodulatory activity of human umbilical cord blood-derived mesenchymal stem cells in vitro. Immunology. 2009;126:220–32. doi: 10.1111/j.1365-2567.2008.02891.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Niemeyer P, Kornacker M, Mehlhorn A, et al. Comparison of immunological properties of bone marrow stromal cells and adipose tissue-derived stem cells before and after osteogenic differentiation in vitro. Tissue Eng. 2007;13:111–21. doi: 10.1089/ten.2006.0114. [DOI] [PubMed] [Google Scholar]
- 59.Yagi H, Soto-Gutierrez A, Navarro-Alvarez N, et al. Reactive bone marrow stromal cells attenuate systemic inflammation via sTNFR1. Mol Ther. 2010;18:1857–64. doi: 10.1038/mt.2010.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Coulson-Thomas VJ, Caterson B, Kao WW. Transplantation of human umbilical mesenchymal stem cells cures the corneal defects of mucopolysaccharidosis VII mice. Stem Cells. 2013;31:2116–26. doi: 10.1002/stem.1481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Joyce N, Annett G, Wirthlin L, et al. Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen Med. 2010;5:933–46. doi: 10.2217/rme.10.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Taylor DD, Gercel-Taylor C. Exosomes/microvesicles: mediators of cancer-associated immunosuppressive microenvironments. Semin Immunopathol. 2011;33:441–54. doi: 10.1007/s00281-010-0234-8. [DOI] [PubMed] [Google Scholar]
- 63.Gyorgy B, Szabo TG, Pasztoi M, et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci. 2011;68:2667–88. doi: 10.1007/s00018-011-0689-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chaput N, Thery C. Exosomes: immune properties and potential clinical implementations. Semin Immunopathol. 2011;33:419–40. doi: 10.1007/s00281-010-0233-9. [DOI] [PubMed] [Google Scholar]
- 65.Bobrie A, Colombo M, Raposo G, Thery C. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic. 2011;12:1659–68. doi: 10.1111/j.1600-0854.2011.01225.x. [DOI] [PubMed] [Google Scholar]
- 66.Zhang B, Yin Y, Lai RC. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Devel. 2014;23:1233–44. doi: 10.1089/scd.2013.0479. [DOI] [PubMed] [Google Scholar]
- 67.Peche H, Heslan M, Usal C, et al. Presentation of donor major histocompatibility complex antigens by bone marrow dendritic cell-derived exosomes modulates allograft rejection. Transplantation. 2003;76:1503–10. doi: 10.1097/01.TP.0000092494.75313.38. [DOI] [PubMed] [Google Scholar]
- 68.Yeo RW, Lai RC, Zhang B, et al. Mesenchymal stem cell: an efficient mass producer of exosomes for drug delivery. Adv Drug Del Rev. 2013;65:336–41. doi: 10.1016/j.addr.2012.07.001. [DOI] [PubMed] [Google Scholar]
- 69.Roccaro AM, Sacco A, Maiso P, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 2013;123:1542–55. doi: 10.1172/JCI66517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Rahman MJ, Regn D, Bashratyan R, Dai YD. Exosomes released by islet-derived mesenchymal stem cells trigger autoimmune responses in NOD mice. Diabetes. 2014;63:1008–20. doi: 10.2337/db13-0859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Blazquez R, Sanchez-Margallo FM, de la Rosa O, et al. Immunomodulatory potential of human adipose mesenchymal stem cells derived exosomes on in vitro stimulated T cells. Front Immunol. 2014;5:556. doi: 10.3389/fimmu.2014.00556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Corrado C, Raimondo S, Chiesi A, et al. Exosomes as intercellular signaling organelles involved in health and disease: basic science and clinical applications. Int J Mol Sci. 2013;14:5338–66. doi: 10.3390/ijms14035338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol. 2012;3:359. doi: 10.3389/fphys.2012.00359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Xin H, Li Y, Buller B, et al. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012;30:1556–64. doi: 10.1002/stem.1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kota DJ, Prabhakara KS, Cox CS, Olson SD. MSCs and hyaluronan: sticking together for new therapeutic potential? Int J Biochem Cell Biol. 2014;55:1–10. doi: 10.1016/j.biocel.2014.07.022. [DOI] [PubMed] [Google Scholar]
- 76.de la Motte CA, Hascall VC, Drazba J, et al. Mononuclear leukocytes bind to specific hyaluronan structures on colon mucosal smooth muscle cells treated with polyinosinic acid:polycytidylic acid: inter-alpha-trypsin inhibitor is crucial to structure and function. Am J Pathol. 2003;163:121–33. doi: 10.1016/s0002-9440(10)63636-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Torihashi S, Ho M, Kawakubo Y, et al. Acute and temporal expression of TNF-alpha-stimulated gene 6 product,TSG-6,in mesenchymal stem cells creates microenvironments required for their successul transplantation into the muscle tissue. J Biol Chem. 2015;290:22771–81. doi: 10.1074/jbc.M114.629774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Oh JY, Lee RH, Yu JM, et al. Intravenous mesenchymal stem cells prevented rejection of allogeneic corneal transplants by aborting the early inflammatory response. Mol Ther. 2012;20:2143–52. doi: 10.1038/mt.2012.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lee TH, Lee GW, Ziff EB, Vilcek J. Isolation and characterization of eight tumor necrosis factor-induced gene sequences from human fibroblasts. Mol Cell Biol. 1990;10:1982–8. doi: 10.1128/mcb.10.5.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dyer DP, Thomson JM, Hermant A, et al. TSG-6 inhibits neutrophil migration via direct interaction with the chemokine CXCL8. J Immunol. 2014;192:2177–85. doi: 10.4049/jimmunol.1300194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sesia SB, Duhr R, Medeiros da Cunha C, Todorov A, et al. Anti-inflammatory/tissue repair macrophages enhance the cartilage-forming capacity of human bone marrow-derived mesenchymal stromal cells. J Cell Physiol. 2015;230:1258–69. doi: 10.1002/jcp.24861. [DOI] [PubMed] [Google Scholar]
- 82.Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci. 2010;123:4195–200. doi: 10.1242/jcs.023820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Coulson-Thomas VJ, Gesteira TF, Coulson-Thomas YM, et al. Fibroblast and prostate tumor cell cross-talk: fibroblast differentiation, TGF-beta, and extracellular matrix down-regulation. Exp Cell Res. 2010;316:3207–26. doi: 10.1016/j.yexcr.2010.08.005. [DOI] [PubMed] [Google Scholar]
- 84.Coulson-Thomas YM, Gesteira TF, Norton AL, et al. The role of proteoglycans in the reactive stroma on tumor growth and progression. Histol Histopathol. 2015;30:33–41. doi: 10.14670/HH-30.33. [DOI] [PubMed] [Google Scholar]
- 85.Kang I, Barth JL, Sproul EP, et al. Expression of V3 versican by rat arterial sooth muscle cells promotes differentiated and anti-inflammatory phenotypes. J Biol Chem. 2015;290:21629–41. doi: 10.1074/jbc.M115.657486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lauer ME, Majors AK, Comhair S, et al. Hyaluronan and its heavy chain modification in asthma severity and experimental asthma exacerbation. J Biol Chem. 2015;290:23124–34. doi: 10.1074/jbc.M115.663823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lauer ME, Aytekin M, Comhair SA, et al. Modification of hyaluronan by heavy chains of inter-alpha-inhibitor in idiopathic pulmonary arterial hypertension. J Biol Chem. 2014;289:6791–8. doi: 10.1074/jbc.M113.512491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Forsberg E, Kjellen L. Heparan sulfate: lessons from knockout mice. J Clin Invest. 2001;108:175–80. doi: 10.1172/JCI13561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Coulson-Thomas VJ, Gesteira TF, Esko J, Kao W. Heparan sulfate regulates hair follicle and sebaceous gland morphogenesis and homeostasis. J Biol Chem. 2014;289:25211–26. doi: 10.1074/jbc.M114.572511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Mathews S, Bhonde R, Gupta PK, Totey S. Novel biomimetic tripolymer scaffolds consisting of chitosan, collagen type 1, and hyaluronic acid for bone marrow-derived human mesenchymal stem cells-based bone tissue engineering. J Biomed Mater Res B Appl Biomater. 2014;102:1825–34. doi: 10.1002/jbm.b.33152. [DOI] [PubMed] [Google Scholar]
- 91.Nemeth CL, Janebodin K, Yuan AE. Enhanced chondrogenic differentiation of dental pulp stem cells using nanopatterned PEG-GelMA-HA hydrogels. Tissue Eng Part A. 2014;20:2817–29. doi: 10.1089/ten.tea.2013.0614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lee RH, Yu JM, Foskett AM, et al. TSG-6 as a biomarker to predict efficacy of human mesenchymal stem/progenitor cells (hMSCs) in modulating sterile inflammation in vivo. Proc Natl Acad Sci U S A. 2014;111:16766–71. doi: 10.1073/pnas.1416121111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Oh JY, Kim MK, Shin MS, et al. The anti-inflammatory and anti-angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 2008;26:1047–55. doi: 10.1634/stemcells.2007-0737. [DOI] [PubMed] [Google Scholar]
- 94.Ye J, Yao K, Kim JC. Mesenchymal stem cell transplantation in a rabbit corneal alkali burn model: engraftment and involvement in wound healing. Eye. 2006;20:482–90. doi: 10.1038/sj.eye.6701913. [DOI] [PubMed] [Google Scholar]
- 95.Garzon I, Carriel V, Marin-Fernandez AB, et al. A combined approach for the assessment of cell viability and cell functionality of human fibrochondrocytes for use in tissue engineering. PloS One. 2012;7:e51961. doi: 10.1371/journal.pone.0051961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Arnalich-Montiel F, Pastor S, Blazquez-Martinez A, Fernandez-Delgado J, et al. Adipose-derived stem cells are a source for cell therapy of the corneal stroma. Stem Cells. 2008;26:570–9. doi: 10.1634/stemcells.2007-0653. [DOI] [PubMed] [Google Scholar]
- 97.Reinshagen H, Auw-Haedrich C, Sorg RV, et al. Corneal surface reconstruction using adult mesenchymal stem cells in experimental limbal stem cell deficiency in rabbits. Acta Ophthalmol. 2011;89:741–8. doi: 10.1111/j.1755-3768.2009.01812.x. [DOI] [PubMed] [Google Scholar]
- 98.Holan V, Trosan P, Cejka C, et al. A comparative study of the therapeutic potential of mesenchymal stem cells and limbal epithelial stem cells for ocular surface reconstruction. Stem Cells Transl Med. 2015;4:1052–63. doi: 10.5966/sctm.2015-0039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zhang L, Coulson-Thomas VJ, Kao W. Mesenchymal stem cells for treating ocular surface diseases. BMC Ophthalmol. 2015;15(Suppl 1):155. doi: 10.1186/s12886-015-0138-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Omoto M, Katikireddy KR, Rezazadeh A, et al. Mesenchymal stem cells home to inflamed ocular surface and suppress allosensitization in corneal transplantation. Invest Ophthalmol Vis Sci. 2014;55:6631–8. doi: 10.1167/iovs.14-15413. [DOI] [PubMed] [Google Scholar]
- 101.Lan Y, Kodati S, Lee HS, et al. Kinetics and function of mesenchymal stem cells in corneal injury. Invest Ophthalmol Vis Sci. 2012;53:3638–44. doi: 10.1167/iovs.11-9311. [DOI] [PubMed] [Google Scholar]
- 102.Almaliotis D, Koliakos G, Papakonstantinou E, et al. Mesenchymal stem cells improve healing of the cornea after alkali injury. Graefes Klin Exp Ophthalmol. 2015;253:1121–35. doi: 10.1007/s00417-015-3042-y. [DOI] [PubMed] [Google Scholar]
- 103.Oh JY, Ko JH, Kim MK, Wee WR. Effects of mesenchymal stem/stromal cells on cultures of corneal epithelial progenitor cells with ethanol injury. Invest Ophthalmol Vis Sci. 2014;55:7628–35. doi: 10.1167/iovs.14-15424. [DOI] [PubMed] [Google Scholar]
- 104.Holan V, Javorkova E. Mesenchymal stem cells, nanofiber scaffolds and ocular surface reconstruction. Stem Cell Rev. 2013;9:609–19. doi: 10.1007/s12015-013-9449-0. [DOI] [PubMed] [Google Scholar]
- 105.Cejkova J, Trosan P, Cejka C, et al. Suppression of alkali-induced oxidative injury in the cornea by mesenchymal stem cells growing on nanofiber scaffolds and transferred onto the damaged corneal surface. Exp Eye Res. 2013;116:312–23. doi: 10.1016/j.exer.2013.10.002. [DOI] [PubMed] [Google Scholar]
- 106.Treacy O, O'Flynn L, Ryan AE, et al. Mesenchymal stem cell therapy promotes corneal allograft survival in rats by local and systemic immunomodulation. Am J Transplant. 2014;14:2023–36. doi: 10.1111/ajt.12828. [DOI] [PubMed] [Google Scholar]
- 107.Pan CK, Heilweil G, Lanza R, Schwartz SD. Embryonic stem cells as a treatment for macular degeneration. Exp Opin Biol Ther. 2013;13:1125–33. doi: 10.1517/14712598.2013.793304. [DOI] [PubMed] [Google Scholar]
- 108.Schwartz SD, Hubschman JP, Heilwell G, et al. Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet. 2012;379:713–20. doi: 10.1016/S0140-6736(12)60028-2. [DOI] [PubMed] [Google Scholar]
- 109.Yanai A, Hafeli UO, Metcalfe AL, et al. Focused magnetic stem cell targeting to the retina using superparamagnetic iron oxide nanoparticles. Cell Transplant. 2012;21:1137–48. doi: 10.3727/096368911X627435. [DOI] [PubMed] [Google Scholar]
- 110.Mu Y, Zhao M, Su G. Stem cell-based therapies for age-related macular degeneration: current status and prospects. Int J Clin Exper Med. 2014;7:3843–52. [PMC free article] [PubMed] [Google Scholar]
- 111.Huang C, Zhang J, Ao M, Li Y, Zhang C, et al. Combination of retinal pigment epithelium cell-conditioned medium and photoreceptor outer segments stimulate mesenchymal stem cell differentiation toward a functional retinal pigment epithelium cell phenotype. J Cell Biochem. 2012;113:590–8. doi: 10.1002/jcb.23383. [DOI] [PubMed] [Google Scholar]
- 112.Dang Y, Zhang C, Zhu Y. Stem cell therapies for age-related macular degeneration: the past, present, and future. Clin Interven Aging. 2015;10:255–64. doi: 10.2147/CIA.S73705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Machalinska A, Kawa M, Pius-Sadowska E, et al. Long-term neuroprotective effects of NT-4-engineered mesenchymal stem cells injected intravitreally in a mouse model of acute retinal injury. Invest Ophthalmol Vis Sci. 2013;54:8292–305. doi: 10.1167/iovs.13-12221. [DOI] [PubMed] [Google Scholar]
- 114.Huang L, Xu W, Xu G. Transplantation of CX3CL1-expressing mesenchymal stem cells provides neuroprotective and immunomodulatory effects in a rat model of retinal degeneration. Ocul Immunol Inflam. 2013;21:276–85. doi: 10.3109/09273948.2013.791925. [DOI] [PubMed] [Google Scholar]
- 115.Leow SN, Luu CD, Hairul Nizam MH, et al. Safety and efficacy of human Wharton's jelly-derived mesenchymal stem cells therapy for retinal degeneration. PloS One. 2015;10:e0128973. doi: 10.1371/journal.pone.0128973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Shen HH, Chan EC, Lee JH, et al. Nanocarriers for treatment of ocular neovascularization in the back of the eye: new vehicles for ophthalmic drug delivery. Nanomedicine. 2015;10:2093–107. doi: 10.2217/nnm.15.47. [DOI] [PubMed] [Google Scholar]
- 117.Johnson TV, Bull ND, Martin KR. Transplantation prospects for the inner retina. Eye. 2009;23:1980–4. doi: 10.1038/eye.2008.376. [DOI] [PubMed] [Google Scholar]
- 118.Huo DM, Dong FT, Yu WH, Gao F. Differentiation of mesenchymal stem cell in the microenviroment of retinitis pigmentosa. Int J Ophthalmol. 2010;3:216–9. doi: 10.3980/j.issn.2222-3959.2010.03.08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Baker PS, Brown GC. Stem-cell therapy in retinal disease. Curr Opin Ophthalmol. 2009;20:175–81. doi: 10.1097/icu.0b013e328329b5f2. [DOI] [PubMed] [Google Scholar]
- 120.Ng TK, Fortino VR, Pelaez D, Cheung HS. Progress of mesenchymal stem cell therapy for neural and retinal diseases. World J Stem Cells. 2014;6:111–9. doi: 10.4252/wjsc.v6.i2.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kanno H. Regenerative therapy for neuronal diseases with transplantation of somatic stem cells. World J Stem Cells. 2013;5:163–71. doi: 10.4252/wjsc.v5.i4.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wang S, Lu B, Girman S, et al. Non-invasive stem cell therapy in a rat model for retinal degeneration and vascular pathology. PloS One. 2010;5:e9200. doi: 10.1371/journal.pone.0009200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363:1439–41. doi: 10.1016/S0140-6736(04)16104-7. [DOI] [PubMed] [Google Scholar]
- 124.Gnecchi M, He H, Melo LG, et al. Early beneficial effects of bone marrow-derived mesenchymal stem cells overexpressing Akt on cardiac metabolism after myocardial infarction. Stem Cells. 2009;27:971–9. doi: 10.1002/stem.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Estrada EJ, Valacchi F, Nicora E, et al. Combined treatment of intrapancreatic autologous bone marrow stem cells and hyperbaric oxygen in Type 2 diabetes mellitus. Cell Transplant. 2008;17:1295–304. doi: 10.3727/096368908787648119. [DOI] [PubMed] [Google Scholar]
- 126.Gonzalez-Rey E, Anderson P, Gonzalez MA, et al. Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis. Gut. 2009;58:929–39. doi: 10.1136/gut.2008.168534. [DOI] [PubMed] [Google Scholar]
- 127.Quittet MS, Touzani O, Sindji L, et al. Effects of mesenchymal stem cell therapy, in association with pharmacologically active microcarriers releasing VEGF, in an ischaemic stroke model in the rat. Acta Biomater. 2015;15:77–88. doi: 10.1016/j.actbio.2014.12.017. [DOI] [PubMed] [Google Scholar]
- 128.Qiao B, Shui W, Cai L, et al. Human mesenchymal stem cells as delivery of osteoprotegerin gene: homing and therapeutic effect for osteosarcoma. Drug Des Devel Ther. 2015;9:969–76. doi: 10.2147/DDDT.S77116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Knoop K, Schwenk N, Schmohl K, et al. Mesenchymal stem cell (MSC)-mediated, tumor stroma-targeted radioiodine therapy of metastatic colon cancer using the sodium iodide symporter as theranostic gene. J Nucl Med. 2015;56:600–6. doi: 10.2967/jnumed.114.146662. [DOI] [PubMed] [Google Scholar]
- 130.Smith CL, Chaichana KL, Lee YM, et al. Pre-exposure of human adipose mesenchymal stem cells to soluble factors enhances their homing to brain cancer. Stem Cells Transl Med. 2015;4:239–51. doi: 10.5966/sctm.2014-0149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Sadhukha T, O'Brien TD, Prabha S. Nano-engineered mesenchymal stem cells as targeted therapeutic carriers. J Control Release. 2014;196:243–51. doi: 10.1016/j.jconrel.2014.10.015. [DOI] [PubMed] [Google Scholar]
- 132.Zhao J, Vykoukal J, Abdelsalam M, et al. Stem cell-mediated delivery of SPIO-loaded gold nanoparticles for the theranosis of liver injury and hepatocellular carcinoma. Nanotechnology. 2014;25:405101. doi: 10.1088/0957-4484/25/40/405101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kim JH, Lee HJ, Song YS. Stem cell based gene therapy in prostate cancer. Biomed Res Int. 2014;2014:549136. doi: 10.1155/2014/549136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bartholomew A, Sturgeon C, Siatskas M, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol. 2002;30:42–8. doi: 10.1016/s0301-472x(01)00769-x. [DOI] [PubMed] [Google Scholar]
- 135.Di Nicola M, Carlo-Stella C, Magni M, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838–43. doi: 10.1182/blood.v99.10.3838. [DOI] [PubMed] [Google Scholar]
- 136.Krampera M, Glennie S, Dyson J, et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood. 2003;101:3722–9. doi: 10.1182/blood-2002-07-2104. [DOI] [PubMed] [Google Scholar]
- 137.Meisel R, Zibert A, Laryea M, et al. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004;103:4619–21. doi: 10.1182/blood-2003-11-3909. [DOI] [PubMed] [Google Scholar]
- 138.Tse WT, Pendleton JD, Beyer WM, et al. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation. 2003;75:389–97. doi: 10.1097/01.TP.0000045055.63901.A9. [DOI] [PubMed] [Google Scholar]
- 139.Li H, Guo Z, Jiang X, et al. Mesenchymal stem cells alter migratory property of T and dendritic cells to delay the development of murine lethal acute graft-versus-host disease. Stem Cells. 2008;26:2531–41. doi: 10.1634/stemcells.2008-0146. [DOI] [PubMed] [Google Scholar]
- 140.Garcia-Olmo D, Garcia-Arranz M, Herreros D, et al. A phase I clinical trial of the treatment of Crohn's fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum. 2005;48:1416–23. doi: 10.1007/s10350-005-0052-6. [DOI] [PubMed] [Google Scholar]
- 141.Casiraghi F, Azzollini N, Cassis P, et al. Pretransplant infusion of mesenchymal stem cells prolongs the survival of a semiallogeneic heart transplant through the generation of regulatory T cells. J Immunol. 2008;181:3933–46. doi: 10.4049/jimmunol.181.6.3933. [DOI] [PubMed] [Google Scholar]
- 142.Yang C, Robbins PD. Immunosuppressive exosomes: a new approach for treating arthritis. Int J Rheumatol. 2012;2012:573528. doi: 10.1155/2012/573528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kim SH, Bianco N, Menon R, et al. Exosomes derived from genetically modified DC expressing FasL are anti-inflammatory and immunosuppressive. Mol Ther. 2006;13:289–300. doi: 10.1016/j.ymthe.2005.09.015. [DOI] [PubMed] [Google Scholar]
- 144.Huber V, Filipazzi P, Iero M, et al. More insights into the immunosuppressive potential of tumor exosomes. J Transl Med. 2008;6:63. doi: 10.1186/1479-5876-6-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ekstrom EJ, Bergenfelz C, von Bulow V, et al. WNT5A induces release of exosomes containing pro-angiogenic and immunosuppressive factors from malignant melanoma cells. Mol Cancer. 2014;13:88. doi: 10.1186/1476-4598-13-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Cai Z, Zhang W, Yang F, et al. Immunosuppressive exosomes from TGF-beta1 gene-modified dendritic cells attenuate Th17-mediated inflammatory autoimmune disease by inducing regulatory T cells. Cell Res. 2012;22:607–10. doi: 10.1038/cr.2011.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–5. doi: 10.1038/nbt.1807. [DOI] [PubMed] [Google Scholar]
- 148.Bian S, Zhang L, Duan L, et al. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J Mol Med (Berl) 2014;92:387–97. doi: 10.1007/s00109-013-1110-5. [DOI] [PubMed] [Google Scholar]
- 149.van Poll D, Parekkadan B, Cho CH, et al. Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo. Hepatology. 2008;47:1634–43. doi: 10.1002/hep.22236. [DOI] [PubMed] [Google Scholar]
- 150.Yagi H, Soto-Gutierrez A, Kitagawa Y, et al. Bone marrow mesenchymal stromal cells attenuate organ injury induced by LPS and burn. Cell Transplant. 2010;19:823–30. doi: 10.3727/096368910X508942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Xie M, He X, Zhu J, et al. Experimental study of adipose-derived mesenchymal stem cells in the treatment of Crohn's disease. Zhonghua Wei Chang Wai Ke Za Zhi. 2015;18:58–64. [PubMed] [Google Scholar]
- 152.Dong LH, Jiang YY, Liu YJ, et al. The anti-fibrotic effects of mesenchymal stem cells on irradiated lungs via stimulating endogenous secretion of HGF and PGE2. Sci Rep. 2015;5:8713. doi: 10.1038/srep08713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Gu W, Song L, Li XM, et al. Mesenchymal stem cells alleviate airway inflammation and emphysema in COPD through down-regulation of cyclooxygenase-2 via p38 and ERK MAPK pathways. Sci Rep. 2015;5:8733. doi: 10.1038/srep08733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.El-Akabawy G, Rashed LA. Beneficial effects of bone marrow-derived mesenchymal stem cell transplantation in a non-immune model of demyelination. Ann Anat. 2015;198:11–20. doi: 10.1016/j.aanat.2014.12.002. [DOI] [PubMed] [Google Scholar]
- 155.Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008;451:937–42. doi: 10.1038/nature06800. [DOI] [PubMed] [Google Scholar]
- 156.McGinley LM, McMahon J, Stocca A, et al. Mesenchymal stem cell survival in the infarcted heart is enhanced by lentivirus vector-mediated heat shock protein 27 expression. Hum Gene Ther. 2013;24:840–51. doi: 10.1089/hum.2011.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Shi B, Long X, Zhao R, et al. Transplantation of mesenchymal stem cells carrying the human receptor activity-modifying protein 1 gene improves cardiac function and inhibits neointimal proliferation in the carotid angioplasty and myocardial infarction rabbit model. Exp Biol Med (Maywood) 2014;239:356–65. doi: 10.1177/1535370213517619. [DOI] [PubMed] [Google Scholar]
- 158.Wang CM, Guo Z, Xie YJ, et al. Co-treating mesenchymal stem cells with IL1beta and TNF-alpha increases VCAM-1 expression and improves post-ischemic myocardial function. Mol Med Rep. 2014;10:792–8. doi: 10.3892/mmr.2014.2236. [DOI] [PubMed] [Google Scholar]
- 159.Zeng B, Ren X, Lin G, et al. Paracrine action of HO-1-modified mesenchymal stem cells mediates cardiac protection and functional improvement. Cell Biol Int. 2008;32:1256–64. doi: 10.1016/j.cellbi.2008.07.010. [DOI] [PubMed] [Google Scholar]
- 160.Chullikana A, Majumdar AS, Gottipamula S, et al. Randomized, double-blind, phase I/II study of intravenous allogeneic mesenchymal stromal cells in acute myocardial infarction. Cytotherapy. 2015;17:250–61. doi: 10.1016/j.jcyt.2014.10.009. [DOI] [PubMed] [Google Scholar]
- 161.Gotherstrom C, Westgren M, Shaw SW, et al. Pre- and postnatal transplantation of fetal mesenchymal stem cells in osteogenesis imperfecta: a two-center experience. Stem Cells Transl Med. 2014;3:255–64. doi: 10.5966/sctm.2013-0090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Bhakta S, Hong P, Koc O. The surface adhesion molecule CXCR4 stimulates mesenchymal stem cell migration to stromal cell-derived factor-1 in vitro but does not decrease apoptosis under serum deprivation. Cardiovasc Revasc Med. 2006;7:19–24. doi: 10.1016/j.carrev.2005.10.008. [DOI] [PubMed] [Google Scholar]
- 163.Qin Y, Zhou Z, Zhang F, et al. Induction of regulatory B-cells by mesenchymal stem cells is affected by SDF-1alpha-CXCR7. Cell Physiol Biochem. 2015;37:117–30. doi: 10.1159/000430338. [DOI] [PubMed] [Google Scholar]
- 164.Gee K, Kryworuchko M, Kumar A. Recent advances in the regulation of CD44 expression and its role in inflammation and autoimmune diseases. Arch Immunol Ther Exp (Warsz) 2004;52:13–26. [PubMed] [Google Scholar]
- 165.Zhu H, Mitsuhashi N, Klein A, et al. The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix. Stem Cells. 2006;24:928–35. doi: 10.1634/stemcells.2005-0186. [DOI] [PubMed] [Google Scholar]
- 166.Chanmee T, Ontong P, Kimata K, Itano N. Key roles of hyaluronan and its CD44 receptor in the stemness and survival of cancer stem cells. Front Oncol. 2015;5:180. doi: 10.3389/fonc.2015.00180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Aldridge V, Garg A, Davies N, et al. Human mesenchymal stem cells are recruited to injured liver in a beta1-integrin and CD44 dependent manner. Hepatology. 2012;56:1063–73. doi: 10.1002/hep.25716. [DOI] [PubMed] [Google Scholar]
- 168.Poitz DM, Stolzel F, Arabanian L, et al. MiR-134-mediated beta1 integrin expression and function in mesenchymal stem cells. Biochim Biophys Acta. 2013;1833:3396–404. doi: 10.1016/j.bbamcr.2013.10.003. [DOI] [PubMed] [Google Scholar]
- 169.Veevers-Lowe J, Ball SG, Shuttleworth A, Kielty CM. Mesenchymal stem cell migration is regulated by fibronectin through alpha5beta1-integrin-mediated activation of PDGFR-beta and potentiation of growth factor signals. J Cell Sci. 2011;124:1288–300. doi: 10.1242/jcs.076935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Li YP, Paczesny S, Lauret E, et al. Human mesenchymal stem cells license adult CD34+ hemopoietic progenitor cells to differentiate into regulatory dendritic cells through activation of the Notch pathway. J Immunol. 2008;180:1598–608. doi: 10.4049/jimmunol.180.3.1598. [DOI] [PubMed] [Google Scholar]
- 171.Chang W, Kim R, Park SI, et al. Enhanced healing of rat calvarial bone defects with hypoxic conditioned medium from mesenchymal stem cells through increased endogenous stem cell migration via regulation of ICAM-1 targeted-microRNA-221. Mol Cells. 2015;38:643–50. doi: 10.14348/molcells.2015.0050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Levesque JP, Takamatsu Y, Nilsson SK, et al. Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases in the bone marrow following hematopoietic progenitor cell mobilization by granulocyte colony-stimulating factor. Blood. 2001;98:1289–97. doi: 10.1182/blood.v98.5.1289. [DOI] [PubMed] [Google Scholar]
- 173.Peister A, Mellad JA, Larson BL, et al. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood. 2004;103:1662–8. doi: 10.1182/blood-2003-09-3070. [DOI] [PubMed] [Google Scholar]
- 174.Kijima N, Hosen N, Kagawa N, et al. CD166/activated leukocyte cell adhesion molecule is expressed on glioblastoma progenitor cells and involved in the regulation of tumor cell invasion. Neuro-oncol. 2012;14:1254–64. doi: 10.1093/neuonc/nor202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Bruder SP, Ricalton NS, Boynton RE, et al. Mesenchymal stem cell surface antigen SB-10 corresponds to activated leukocyte cell adhesion molecule and is involved in osteogenic differentiation. J Bone Min Res. 1998;13:655–63. doi: 10.1359/jbmr.1998.13.4.655. [DOI] [PubMed] [Google Scholar]
- 176.Wiesmann A, Buhring HJ, Mentrup C, Wiesmann HP. Decreased CD90 expression in human mesenchymal stem cells by applying mechanical stimulation. Head Face Med. 2006;2:8. doi: 10.1186/1746-160X-2-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Campioni D, Rizzo R, Stignani M, et al. A decreased positivity for CD90 on human mesenchymal stromal cells (MSCs) is associated with a loss of immunosuppressive activity by MSCs. Cytometry B Clin Cytom. 2009;76:225–30. doi: 10.1002/cyto.b.20461. [DOI] [PubMed] [Google Scholar]
- 178.Hermiston ML, Xu Z, Weiss A. CD45: a critical regulator of signaling thresholds in immune cells. Annu Rev Immunol. 2003;21:107–37. doi: 10.1146/annurev.immunol.21.120601.140946. [DOI] [PubMed] [Google Scholar]
- 179.Drew E, Huettner CS, Tenen DG, McNagny KM. CD34 expression by mast cells: of mice and men. Blood. 2005;106:1885–7. doi: 10.1182/blood-2005-03-1291. [DOI] [PubMed] [Google Scholar]
- 180.Zanoni I, Ostuni R, Marek LR, et al. CD14 controls the LPS-induced endocytosis of Toll-like receptor 4. Cell. 2011;147:868–80. doi: 10.1016/j.cell.2011.09.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Pugin J, Heumann ID, Tomasz A, et al. CD14 is a pattern recognition receptor. Immunity. 1994;1:509–16. doi: 10.1016/1074-7613(94)90093-0. [DOI] [PubMed] [Google Scholar]
- 182.Ling GS, Bennett J, Woollard KJ, et al. Integrin CD11b positively regulates TLR4-induced signalling pathways in dendritic cells but not in macrophages. Nat Commun. 2014;5:3039. doi: 10.1038/ncomms4039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Homey B, Wang W, Soto H, et al. Cutting edge: the orphan chemokine receptor G protein-coupled receptor-2 (GPR-2, CCR10) binds the skin-associated chemokine CCL27 (CTACK/ALP/ILC). J Immunol. 2000;164:3465–70. doi: 10.4049/jimmunol.164.7.3465. [DOI] [PubMed] [Google Scholar]
- 184.Wang W, Soto H, Oldham ER, et al. Identification of a novel chemokine (CCL28), which binds CCR10 (GPR2). J Biol Chem. 2000;275:22313–23. doi: 10.1074/jbc.M001461200. [DOI] [PubMed] [Google Scholar]
- 185.Schulze MS, Wucherpfennig KW. The mechanism of HLA-DM induced peptide exchange in the MHC class II antigen presentation pathway. Curr Opin Immunol. 2012;24:105–11. doi: 10.1016/j.coi.2011.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Moretta L, Biassoni R, Bottino C, et al. Human NK cells and their receptors. Microbes Infect. 2002;4:1539–44. doi: 10.1016/s1286-4579(02)00037-0. [DOI] [PubMed] [Google Scholar]
- 187.Moretta A, Bottino C, Vitale M, et al. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu Rev Immunol. 2001;19:197–223. doi: 10.1146/annurev.immunol.19.1.197. [DOI] [PubMed] [Google Scholar]
- 188.Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009;9:265–73. doi: 10.1038/nrc2620. [DOI] [PubMed] [Google Scholar]
- 189.James D, Levine AJ, Besser D, Hemmati-Brivanlou A. TGFbeta/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development. 2005;132:1273–82. doi: 10.1242/dev.01706. [DOI] [PubMed] [Google Scholar]
- 190.Qu X, Liu X, Cheng K, et al. Mesenchymal stem cells inhibit Th17 cell differentiation by IL-10 secretion. Exp Hematol. 2012;40:761–70. doi: 10.1016/j.exphem.2012.05.006. [DOI] [PubMed] [Google Scholar]
- 191.Niu J, Yue W, Song Y, et al. Prevention of acute liver allograft rejection by IL-10-engineered mesenchymal stem cells. Clin Exp Immunol. 2014;176:473–84. doi: 10.1111/cei.12283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Liu GY, Xu Y, Li Y, et al. Secreted galectin-3 as a possible biomarker for the immunomodulatory potential of human umbilical cord mesenchymal stromal cells. Cytotherapy. 2013;15:1208–17. doi: 10.1016/j.jcyt.2013.05.011. [DOI] [PubMed] [Google Scholar]
- 193.Silverman AM, Nakata R, Shimada H, et al. A galectin-3-dependent pathway upregulates interleukin-6 in the microenvironment of human neuroblastoma. Cancer Res. 2012;72:2228–38. doi: 10.1158/0008-5472.CAN-11-2165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Mackinnon AC, Gibbons MA, Farnworth SL, et al. Regulation of transforming growth factor-beta1-driven lung fibrosis by galectin-3. Am J Resp Crit Care Med. 2012;185:537–46. doi: 10.1164/rccm.201106-0965OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Mathieu ME, Saucourt C, Mournetas V, et al. LIF-dependent signaling: new pieces in the Lego. Stem Cell Rev. 2012;8:1–15. doi: 10.1007/s12015-011-9261-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Maione F, Capano S, Regano D, et al. Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J Clin Invest. 2012;122:1832–48. doi: 10.1172/JCI58976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Maione F, Molla F, Meda C, et al. Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor growth and normalizes tumor vasculature in transgenic mouse models. J Clin Invest. 2009;119:3356–72. doi: 10.1172/JCI36308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Maher D, Blanc L, Raviglione M. WHO policies for tuberculosis control. Lancet. 2004;363:1911. doi: 10.1016/S0140-6736(04)16377-0. [DOI] [PubMed] [Google Scholar]




