151
|
Liu S, Yuan M, Hou K, Zhang L, Zheng X, Zhao B, Sui X, Xu W, Lu S, Guo Q. Immune characterization of mesenchymal stem cells in human umbilical cord Wharton's jelly and derived cartilage cells. Cell Immunol 2012; 278:35-44. [PMID: 23121974 DOI: 10.1016/j.cellimm.2012.06.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 06/21/2012] [Accepted: 06/27/2012] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells derived from human umbilical cord Wharton's jelly (hWJMSCs) became prospective seed cell candidate for tissue engineering and cell-based therapy because of its variety source, easy procurement, robust proliferation, and high purity compared with bone marrow- and adipose-derived MSCs. Such neonatal stem cells can be isolated from a variety of extraembryonic tissues and appear to be more primitive and have greater multi-potentiality than their adult counterparts. In this study, we investigated the immune characters of hWJMSCs and its derived cartilage cells (hWJMSC-Cs) by detecting the expression of major histocompatibility complex I/I(MHC-I/II), costimulatory molecules (CD40, CD80 and CD86) and immune inhibitors including human leukocyte antigen G (HLA-G), indoleamine-2,3-dioxygenase (IDO), and prostaglandin E2 (PGE2). We found that hWJMSCs did not express MHC-II and costimulatory molecules, but moderately expressed MHC-I, and positively expressed immune inhibitors as HLA-G, IDO, PGE2, demonstrating their very low immunogenicity and potential to induce immune tolerance microenvironment in hosts. The results of chondrogenic differentiated hWJMSCs(hWJMSC-Cs) are similar to those of undifferentiated cells, except for the slightly elevated MHC-II and costimulators expression. Additionally, we detected cytokine profile of hWJMSCs through cytokine antibody array and verified by western blot the positive expression of immune suppression-related molecules, HGF, VEGF, TGF, and IL-10. Furthermore, to investigate the in vivo immune response of the cells, hWJMSCs-scaffold constructs were implanted into rabbits and rats, and the result showed that hWJMSCs did not elicit immune rejection in the animals. Their intermediate state between adult and embryonic stem cells makes them an ideal candidate for reprogramming to the pluripotent status. Additional studies are necessary to clarify the potential of hWJMSCs to be used in cartilage and other tissue regeneration and cell-based therapies.
Collapse
Affiliation(s)
- Shuyun Liu
- Key Laboratory of the People's Liberation Army (PLA), Institute of Orthopedics, Chinese PLA General Hospital, No. 28 FuXing Road, Haidian District, Beijing 100853, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Lin BN, Whu SW, Chen CH, Hsu FY, Chen JC, Liu HW, Chen CH, Liou HM. Bone marrow mesenchymal stem cells, platelet-rich plasma and nanohydroxyapatite-type I collagen beads were integral parts of biomimetic bone substitutes for bone regeneration. J Tissue Eng Regen Med 2012; 7:841-54. [DOI: 10.1002/term.1472] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 10/25/2011] [Accepted: 01/05/2012] [Indexed: 01/19/2023]
Affiliation(s)
- Bo-Nian Lin
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital at Keelung, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Shu Wen Whu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital at Keelung, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Chih-Hwa Chen
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital at Keelung, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Fu-Yin Hsu
- Department of Life Sciences; National Taiwan Ocean University; Taipei Taiwan
| | - Jyh-Cheng Chen
- Department of Biomedical Imaging and Radiological Sciences; National Yang-Ming University; Taipei Taiwan
| | - Hsia-Wei Liu
- Department of Life Science; Fu Jen Catholic University; Taipei Taiwan
| | - Chien-Hao Chen
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital at Keelung, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Hau-Min Liou
- Department of Life Sciences; National Taiwan Ocean University; Taipei Taiwan
| |
Collapse
|
153
|
Wood KJ, Bushell A, Hester J. Regulatory immune cells in transplantation. Nat Rev Immunol 2012; 12:417-30. [DOI: 10.1038/nri3227] [Citation(s) in RCA: 340] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
154
|
Engela AU, Baan CC, Dor FJMF, Weimar W, Hoogduijn MJ. On the interactions between mesenchymal stem cells and regulatory T cells for immunomodulation in transplantation. Front Immunol 2012; 3:126. [PMID: 22629256 PMCID: PMC3355477 DOI: 10.3389/fimmu.2012.00126] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 05/03/2012] [Indexed: 12/13/2022] Open
Abstract
Experimental studies have established the use of mesenchymal stem cells (MSC) as a candidate immunosuppressive therapy. MSC exert their immunomodulatory function through the inhibition of CD4+ and CD8+ T cell proliferation. It is unknown whether MSC impair the immunosuppressive function of regulatory T cells (Treg). In vitro and in vivo studies suggest that MSC mediate their immunomodulatory effects through the induction of Treg. In this review we will focus on the interactions between MSC and Treg, and evaluate the consequences of these cellular interplays for prospective MSC immunotherapy in organ transplantation.
Collapse
Affiliation(s)
- Anja U Engela
- Transplantation Laboratory, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | | | | | | | | |
Collapse
|
155
|
Xu DM, Yu XF, Zhang D, Zhang MX, Zhou JF, Tan PH, Ding YC. Mesenchymal stem cells differentially mediate regulatory T cells and conventional effector T cells to protect fully allogeneic islet grafts in mice. Diabetologia 2012; 55:1091-102. [PMID: 22270222 DOI: 10.1007/s00125-011-2433-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 11/28/2011] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Limited information is available on the cellular interactions between regulatory T (T(reg)) cells and mesenchymal stem cells (MSCs). In particular, a direct effect of MSCs on the survival and proliferation of T(reg) cells has not been demonstrated. METHODS We investigated the effects of MSCs on effector T (T(eff)) cells and T(reg) cells, and the molecular mechanisms involved in the distinct regulation of these two cell populations by MSCs in vivo and in vitro. RESULTS We show that MSCs are capable of selectively suppressing T(eff) cells and fostering the generation of T(reg) cells. T(eff) cells, but not T(reg) cells, fail to respond to IL-2 and undergo profound apoptosis in the presence of MSCs. The differential regulations of these two T cell subsets by MSCs are associated with their distinct expressions of CD25, with MSCs specifically reducing the expression of CD25 on T(eff) and sparing T(reg) cells intact. In vivo, the administration of MSCs significantly delays the rejection of allogeneic islet grafts in adaptive transferred recipients by favouring the induction of T(reg) cells. In this model, MSCs inhibit the proliferation and development of alloreactive T(eff) but potently enhance the induction of T(reg) cells. CONCLUSIONS/INTERPRETATION We demonstrate that MSCs are capable of regulating T(eff) and T(reg) cells differentially in vitro. MSCs inhibit T(eff) cells by inducing apoptosis and impairing the proliferative response to IL-2 in T(eff) cells, but favour the survival and expansion of T(reg) cells. This result is further demonstrated in mice that have undergone allogeneic islet transplantation, in which MSCs suppress alloreactive T(eff) cells while favouring the induction of T(reg) cells, thus protecting the islet allografts from rejection.
Collapse
Affiliation(s)
- D M Xu
- Department of Haematology, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
156
|
Baiguera S, Jungebluth P, Mazzanti B, Macchiarini P. Mesenchymal stromal cells for tissue-engineered tissue and organ replacements. Transpl Int 2012; 25:369-82. [PMID: 22248229 DOI: 10.1111/j.1432-2277.2011.01426.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSCs), a rare heterogeneous subset of pluripotent stromal cells that can be easily isolated from different adult tissues, in vitro expanded and differentiated into multiple lineages, are immune privileged and, more important, display immunomodulatory capacities. Because of this, they are the preferred cell source in tissue-engineered replacements, not only in autogeneic conditions, where they do not evoke any immune response, but especially in the setting of allogeneic organ and tissue replacements. However, more preclinical and clinical studies are requested to completely understand MSC's immune biology and possible clinical applications. We herein review the immunogenicity and immunomodulatory properties of MSCs, their possible mechanisms and potential clinical use for tissue-engineered organ and tissue replacement.
Collapse
Affiliation(s)
- Silvia Baiguera
- BIOAIRlab, European Center of Thoracic Research (CERT), University Hospital Careggi, Florence, Italy
| | | | | | | |
Collapse
|
157
|
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-versus-host disease. Immunol Lett 2012; 143:161-9. [DOI: 10.1016/j.imlet.2012.01.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 12/30/2011] [Accepted: 01/31/2012] [Indexed: 12/13/2022]
|
158
|
Jui HY, Lin CH, Hsu WT, Liu YR, Hsu RB, Chiang BL, Tseng WYI, Chen MF, Wu KK, Lee CM. Autologous mesenchymal stem cells prevent transplant arteriosclerosis by enhancing local expression of interleukin-10, interferon-γ, and indoleamine 2,3-dioxygenase. Cell Transplant 2012; 21:971-84. [PMID: 22449499 DOI: 10.3727/096368911x627525] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Transplant arteriosclerosis (TA) remains the major limitation of long-term graft survival in heart transplantation despite the advances in immunosuppressants. Mesenchymal stem cells (MSCs) have been demonstrated to suppress allogeneic immune responses by numerous in vitro studies. However, the immunomodulatory effects of MSCs in vivo are controversial and the underlying molecular mechanisms are not conclusive. In this study, we investigated the therapeutic potential of autologous bone marrow-derived MSCs on TA in a porcine model of femoral artery transplantation. MSCs or saline were injected into the soft tissue surrounding the arterial grafts immediately postanastomosis. Four weeks after transplantation, neointimal formation increased significantly in untreated allografts compared with the MSC-treated grafts as assessed by intravascular ultrasound (maximum luminal area stenosis: 40 ± 12% vs. 18 ± 6%, p < 0.001). Grafts harvested at 4 weeks showed dense perivascular lymphocyte infiltration accompanied by significant intimal hyperplasia in the untreated but not in the MSC-treated allografts. Serial angiographic examination showed that all of the untreated allografts became occluded at the 8th week whereas the majority of the MSC-treated grafts remained patent at the 12th week posttransplantation (n = 12 each group, p < 0.001). Quantitative PCR analysis revealed that Foxp3 expression was comparable between the untreated and the MSC-treated groups. However, expression of interleukin-10 (IL-10), interferon-γ (IFN-γ), and indoleamine 2,3-dioxygenase (IDO) was increased significantly in the MSC-treated allografts compared with that in the allograft controls (p = 0.021 for IL-10, p = 0.003 for IFN-γ, and p = 0.008 for IDO). In conclusion, local delivery of autologous MSCs alleviates TA by inducing allograft tolerance via enhanced expression of IL-10, IFN-γ, and IDO but not Foxp3-positive cells in the vessel wall. These results suggest that MSCs induce immune tolerance by activating the type 1 regulatory T-like cells.
Collapse
Affiliation(s)
- Hsiang-Yiang Jui
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Dendritic cells and regulation of graft-versus-host disease and graft-versus-leukemia activity. Blood 2012; 119:5088-103. [PMID: 22403259 DOI: 10.1182/blood-2011-11-364091] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cell transplantation is the only curative treatment for many malignant hematologic diseases, with an often critical graft-versus-leukemia effect. Despite peritransplant prophylaxis, GVHD remains a significant cause of posthematopoietic stem cell transplantation morbidity and mortality. Traditional therapies have targeted T cells, yet immunostimulatory dendritic cells (DCs) are critical in the pathogenesis of GVHD. Furthermore, DCs also have tolerogenic properties. Monitoring of DC characteristics may be predictive of outcome, and therapies that target DCs are innovative and promising. DCs may be targeted in vivo or tolerogenic (tol) DCs may be generated in vitro and given in the peritransplant period. Other cellular therapies, notably regulatory T cells (T(reg)) and mesenchymal stem cells, mediate important effects through DCs and show promise for the prevention and treatment of GVHD in early human studies. Therapies are likely to be more effective if they have synergistic effects or target both DCs and T cells in vivo, such as tolDCs or T(reg). Given the effectiveness of tolDCs in experimental models of GVHD and their safety in early human studies for type 1 diabetes, it is crucial that tolDCs be investigated in the prevention and treatment of human GVHD while ensuring conservation of graft-versus-leukemia effects.
Collapse
|
160
|
|
161
|
Dissecting paracrine effectors for mesenchymal stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 129:137-52. [PMID: 22968371 DOI: 10.1007/10_2012_149] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
There has been increasing interest in the application of mesenchymal stem cells (MSCs) in regenerative medicine in recent years. In this context, the beneficial effects of MSCs have been ascribed mainly to a paracrine action rather than to direct replacement of the injured tissue. Indeed, MSCs produce a great variety of trophic and immunomodulatory factors. In this chapter, we provide an overview of growth factors and chemokines involved in stimulation of cell proliferation, inhibition of apoptosis, enhancement of angiogenesis, and suppression of inflammatory and immune response. In addition, we discuss the emerging role of the extracellular vesicles released from MSCs as possible paracrine mediators.
Collapse
|
162
|
Clinical Application of Mesenchymal Stem Cells in the Treatment and Prevention of Graft-versus-Host Disease. Adv Hematol 2011; 2011:427863. [PMID: 22190941 PMCID: PMC3235491 DOI: 10.1155/2011/427863] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 10/18/2011] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent a heterogeneous population of stromal cells with pluripotent mesenchymal differentiation potential. They have been found to have immunosuppressive properties and the ability to modulate angiogenesis and endogenous tissue repair by in vitro and animal studies. Clinical trials have examined the utility of these cells in autoimmune and inflammatory conditions. In particular, in allogeneic hematopoietic stem cell transplant (HSCT), multiple studies have been conducted to explore the use of MSC to treat acute and chronic graft-versus-host disease (GVHD) and for cotransplantation with HSCT to promote HSC engraftment and prevent GVHD. We review here the results of these studies and discuss some challenges of this treatment modality in this disease setting.
Collapse
|
163
|
English K, Mahon BP. Allogeneic mesenchymal stem cells: agents of immune modulation. J Cell Biochem 2011; 112:1963-8. [PMID: 21445861 DOI: 10.1002/jcb.23119] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adult mesenchymal stem cells possess a remarkably diverse array of immunosuppressive characteristics. The capacity to suppress the regular processes of allogeneic rejection, have allowed the use of tissue mismatched cells as therapeutic approaches in regenerative medicine and as agents of immune deviation. This review describes recent advances in understanding the mechanistic basis of mesenchymal stromal or stem cells (MSC) interaction with innate immunity. Particular emphasis is placed on the effect of Toll-like receptor signalling on MSC and a hypothesis that innate immune signals induce a 'licensing switch' in MSC is put forward. The mechanisms underlying MSC suppression of T cell responses and induction of regulatory populations are surveyed. Conflicting data regarding the influence of MSC on B cell function are outlined and discussed. Finally the limits to MSC mediated immune modulation are discussed with reference to the future clinical application of novel cell therapies.
Collapse
Affiliation(s)
- Karen English
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | |
Collapse
|
164
|
Saka Y, Furuhashi K, Katsuno T, Kim H, Ozaki T, Iwasaki K, Haneda M, Sato W, Tsuboi N, Ito Y, Matsuo S, Kobayashi T, Maruyama S. Adipose-derived stromal cells cultured in a low-serum medium, but not bone marrow-derived stromal cells, impede xenoantibody production. Xenotransplantation 2011; 18:196-208. [PMID: 21696449 DOI: 10.1111/j.1399-3089.2011.00640.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Although the immunomodulatory effects of mesenchymal stromal cells (MSC) on T cells have been elucidated, little is known about their effects on B cells. Recently, we have established a novel culture method for adipose-derived MSC (ASC) using low (2%) serum medium containing fibroblast growth factor-2. We showed that low serum-cultured ASC (LASC) was superior to high (20%) serum-cultured ASC (HASC) when used in regenerative therapy. The aim of this study was to compare the action of LASC, HASC, and bone marrow-derived MSC (BM-MSC), on xenoantibody production by B cells. METHODS Adipose-derived mesenchymal stromal cells and BM-MSC were obtained from humans or F344 rats and expanded in a low-serum or a high-serum culture medium. Proliferation of human peripheral mononuclear cells (PBMC) or rat splenocytes was induced by phytohemagglutinin (PHA) or anti-IgM-antibody. These cells were then co-cultured with LASC, HASC, or BM-MSC, and cell proliferation was studied. Porcine red blood cells (pRBC) were intraperitoneally injected into Lewis rats, and LASC, HASC, or BM-MSC obtained from F344 rats were injected intravenously or intraperitoneally. The levels of antibodies (IgM and IgG) against pRBC were examined using flow cytometry. RESULTS Human LASC suppressed PBMC proliferation more effectively than human HASC. Human LASC suppressed both T-cell and B-cell proliferation when incubated with PHA (a T-cell stimulus). However, human LASC did not suppress B-cell proliferation after incubation with anti-IgM-antibody (a T-cell-independent stimulus). Rat LASC suppressed PHA-stimulated splenocyte proliferation more effectively than rat HASC or rat BM-MSC. In vivo studies showed that intravenous injection of rat LASC significantly reduced the levels of IgG antibodies against pRBC, while intravenous administration of the other two types of MSC (rat HASC or rat BM-MSC) or intraperitoneal administration of rat LASC did not impede IgG production. A significant number of LASC were observed in the spleen when injected intravenously while only a few LASC were observed when given intraperitoneally. CONCLUSIONS Administration of LASC effectively impeded xenoantibody production by B cells through the inhibition of T-cell function, while HASC or BM-MSC showed less promising effects. These results suggest that intravenous injection of LASC may be useful in attenuating antibody-mediated rejection.
Collapse
Affiliation(s)
- Yosuke Saka
- Department of Nephrology, Nagoya University Graduate School of Medicine Department of Applied Immunology, Nagoya University School of Medicine, 65 Tsurumai-cho Showaku, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Anzalone R, Lo Iacono M, Loria T, Di Stefano A, Giannuzzi P, Farina F, La Rocca G. Wharton's jelly mesenchymal stem cells as candidates for beta cells regeneration: extending the differentiative and immunomodulatory benefits of adult mesenchymal stem cells for the treatment of type 1 diabetes. Stem Cell Rev Rep 2011; 7:342-63. [PMID: 20972649 DOI: 10.1007/s12015-010-9196-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSC) are uniquely capable of crossing germinative layers borders (i.e. are able to differentiate towards ectoderm-, mesoderm- and endoderm-derived cytotypes) and are viewed as promising cells for regenerative medicine approaches in several diseases. Type I diabetes therapy should potentially benefit from such differentiated cells: the search for alternatives to organ/islet transplantation strategies via stem cells differentiation is an ongoing task, significant goals having been achieved in most experimental settings (e.g. insulin production and euglycaemia restoration), though caution is still needed to ensure safe and durable effects in vivo. MSC are obtainable in high numbers via ex vivo culture and can be differentiated towards insulin-producing cells (IPC). Moreover, recent reports evidenced that MSC possess immunomodulatory activities (acting on both innate and acquired immunity effectors) which should result in a reduction of the immunogenicity of transplanted cells, thus limiting rejection. Moreover it has been proposed that MSC administration should be used to attenuate the autoimmune processes which lead to the destruction of beta cells. This review illustrates the recent advances made in differentiating human MSC to IPC. In particular, we compare the effectiveness of the differentiation protocols applied, the markers and functional assays used to characterize differentiated progeny, and the in vivo controls. We further speculate on how MSC derived from Wharton's jelly of human umbilical cord may represent a more promising regenerative medicine tool, as recently demonstrated for endoderm-derived organs (as liver) in human subjects, also considering their peculiar immunomodulatory features compared to other MSC populations.
Collapse
Affiliation(s)
- Rita Anzalone
- Sezione di Anatomia Umana, Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche, Università degli Studi di Palermo, Via del Vespro 129, Palermo, PA 90127, Italy
| | | | | | | | | | | | | |
Collapse
|
166
|
Eggenhofer E, Renner P, Soeder Y, Popp FC, Hoogduijn MJ, Geissler EK, Schlitt HJ, Dahlke MH. Features of synergism between mesenchymal stem cells and immunosuppressive drugs in a murine heart transplantation model. Transpl Immunol 2011; 25:141-7. [DOI: 10.1016/j.trim.2011.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/08/2011] [Accepted: 06/08/2011] [Indexed: 02/07/2023]
|
167
|
Popp FC, Fillenberg B, Eggenhofer E, Renner P, Dillmann J, Benseler V, Schnitzbauer AA, Hutchinson J, Deans R, Ladenheim D, Graveen CA, Zeman F, Koller M, Hoogduijn MJ, Geissler EK, Schlitt HJ, Dahlke MH. Safety and feasibility of third-party multipotent adult progenitor cells for immunomodulation therapy after liver transplantation--a phase I study (MISOT-I). J Transl Med 2011; 9:124. [PMID: 21798013 PMCID: PMC3166276 DOI: 10.1186/1479-5876-9-124] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 07/28/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Liver transplantation is the definitive treatment for many end-stage liver diseases. However, the life-long immunosuppression needed to prevent graft rejection causes clinically significant side effects. Cellular immunomodulatory therapies may allow the dose of immunosuppressive drugs to be reduced. In the current protocol, we propose to complement immunosuppressive pharmacotherapy with third-party multipotent adult progenitor cells (MAPCs), a culture-selected population of adult adherent stem cells derived from bone marrow that has been shown to display potent immunomodulatory and regenerative properties. In animal models, MAPCs reduce the need for pharmacological immunosuppression after experimental solid organ transplantation and regenerate damaged organs. METHODS Patients enrolled in this phase I, single-arm, single-center safety and feasibility study (n = 3-24) will receive 2 doses of third-party MAPCs after liver transplantation, on days 1 and 3, in addition to a calcineurin-inhibitor-free "bottom-up" immunosuppressive regimen with basiliximab, mycophenolic acid, and steroids. The study objective is to evaluate the safety and clinical feasibility of MAPC administration in this patient cohort. The primary endpoint of the study is safety, assessed by standardized dose-limiting toxicity events. One secondary endpoint is the time until first biopsy-proven acute rejection, in order to collect first evidence of efficacy. Dose escalation (150, 300, 450, and 600 million MAPCs) will be done according to a 3 + 3 classical escalation design (4 groups of 3-6 patients each). DISCUSSION If MAPCs are safe for patients undergoing liver transplantation in this study, a phase II/III trial will be conducted to assess their clinical efficacy.
Collapse
Affiliation(s)
- Felix C Popp
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Svobodova E, Krulova M, Zajicova A, Pokorna K, Prochazkova J, Trosan P, Holan V. The role of mouse mesenchymal stem cells in differentiation of naive T-cells into anti-inflammatory regulatory T-cell or proinflammatory helper T-cell 17 population. Stem Cells Dev 2011; 21:901-10. [PMID: 21663543 DOI: 10.1089/scd.2011.0157] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) modulate immune response and can produce significant levels of transforming growth factor-β (TGF-β) and interleukin-6 (IL-6). These 2 cytokines represent the key factors that reciprocally regulate the development and polarization of naive T-cells into regulatory T-cell (Treg) population or proinflammatory T helper 17 (Th17) cells. In the present study we demonstrate that MSCs and their products effectively regulate expression of transcription factors Foxp3 and RORγt and control the development of Tregs and Th17 cells in a population of alloantigen-activated mouse spleen cells or purified CD4(+)CD25(-) T-cells. The immunomodulatory effects of MSCs were more pronounced when these cells were stimulated to secrete TGF-β alone or TGF-β together with IL-6. Unstimulated MSCs produce TGF-β, but not IL-6, and the production of TGF-β can be further enhanced by the anti-inflammatory cytokines IL-10 or TGF-β. In the presence of proinflammatory cytokines, MSCs secrete significant levels of IL-6, in addition to a spontaneous production of TGF-β. MSCs producing TGF-β induced preferentially expression of Foxp3 and activation of Tregs, whereas MSC supernatants containing TGF-β together with IL-6 supported RORγt expression and development of Th17 cells. The effects of MSC supernatants were blocked by the inclusion of neutralization monoclonal antibody anti-TGF-β or anti-IL-6 into the culture system. The results showed that MSCs represent important players that reciprocally regulate the development and differentiation of uncommitted naive T-cells into anti-inflammatory Foxp3(+) Tregs or proinflammatory RORγt(+) Th17 cell population and thereby can modulate autoimmune, immunopathological, and transplantation reactions.
Collapse
Affiliation(s)
- Eliska Svobodova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
169
|
Chen PM, Yen ML, Liu KJ, Sytwu HK, Yen BL. Immunomodulatory properties of human adult and fetal multipotent mesenchymal stem cells. J Biomed Sci 2011; 18:49. [PMID: 21762539 PMCID: PMC3156728 DOI: 10.1186/1423-0127-18-49] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 07/18/2011] [Indexed: 02/06/2023] Open
Abstract
In recent years, a large number of studies have contributed to our understanding of the immunomodulatory mechanisms used by multipotent mesenchymal stem cells (MSCs). Initially isolated from the bone marrow (BM), MSCs have been found in many tissues but the strong immunomodulatory properties are best studied in BM MSCs. The immunomodulatory effects of BM MSCs are wide, extending to T lymphocytes and dendritic cells, and are therapeutically useful for treatment of immune-related diseases including graft-versus-host disease as well as possibly autoimmune diseases. However, BM MSCs are very rare cells and require an invasive procedure for procurement. Recently, MSCs have also been found in fetal-stage embryo-proper and extra-embryonic tissues, and these human fetal MSCs (F-MSCs) have a higher proliferative profile, and are capable of multilineage differentiation as well as exert strong immunomodulatory effects. As such, these F-MSCs can be viewed as alternative sources of MSCs. We review here the current understanding of the mechanisms behind the immunomodulatory properties of BM MSCs and F-MSCs. An increase in our understanding of MSC suppressor mechanisms will offer insights for prevalent clinical use of these versatile adult stem cells in the near future.
Collapse
Affiliation(s)
- Pei-Min Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | | | | | | | | |
Collapse
|
170
|
Abstract
In the past decade, more than 100 different composite tissue allotransplantation (CTA) procedures have been performed around the world including more than 50 hand and 8 facial transplants with encouraging graft survival and excellent functional outcomes. Broader clinical application of CTA, however, continues to be hampered by requirement for long-term, high-dose, multidrug maintenance immunosuppression to prevent graft rejection mediated particularly by composite tissue allograft's highly immunogenic skin component. Medication toxicity could result in severe adverse events including metabolic and infectious complications or malignancy. Notably, unlike in solid organs, clinical success is dictated not only by graft acceptance and survival but also by nerve regeneration, which determines ultimate functional outcomes. Novel strategies such as cellular and biologic therapies that integrate the concepts of immune regulation with those of nerve regeneration have shown promising results in small and large animal models. Clinical translation of these insights to reconstructive transplantation and CTA could further minimize the need of immunosuppression and optimize functional outcomes. This will enable wider application of such treatment options for patients in need of complex reconstructive surgery for congenital deformities or devastating injuries that are not amenable to standard methods of repair.
Collapse
Affiliation(s)
- Gerald Brandacher
- Divison of Plastic and Reconstructive Surgery, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | | |
Collapse
|
171
|
Griffin MD, Ritter T, Mahon BP. Immunological aspects of allogeneic mesenchymal stem cell therapies. Hum Gene Ther 2011; 21:1641-55. [PMID: 20718666 DOI: 10.1089/hum.2010.156] [Citation(s) in RCA: 229] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Allogeneic mesenchymal stem or stromal cells (MSCs) are proposed as cell therapies for degenerative, inflammatory, and autoimmune diseases. The feasibility of allogeneic MSC therapies rests heavily on the concept that these cells avoid or actively suppress the immunological responses that cause rejection of most allogeneic cells and tissues. In this article the validity of the immune privileged status of allogeneic MSCs is explored in the context of recent literature. Current data that provide the mechanistic basis for immune modulation by MSCs are reviewed with particular attention to how MSCs modify the triggering and effector functions of innate and adaptive immunity. The ability of MSCs to induce regulatory dendritic and T-cell populations is discussed with regard to cell therapy for autoimmune disease. Finally, we examine the evidence for and against the immune privileged status of allogeneic MSCs in vivo. Allogeneic MSCs emerge as cells that are responsive to local signals and exert wide-ranging, predominantly suppressive, effects on innate and adaptive immunity. Nonetheless, these cells also retain a degree of immunogenicity in some circumstances that may limit MSC longevity and attenuate their beneficial effects. Ultimately successful allogeneic cell therapies will rely on an improved understanding of the parameters of MSC-immune system interactions in vivo.
Collapse
Affiliation(s)
- Matthew D Griffin
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | | |
Collapse
|
172
|
Martinu T, Palmer SM, Ortiz LA. Lung-resident mesenchymal stromal cells. A new player in post-transplant bronchiolitis obliterans syndrome? Am J Respir Crit Care Med 2011; 183:968-70. [PMID: 21498820 DOI: 10.1164/rccm.201101-0006ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
173
|
Han KH, Ro H, Hong JH, Lee EM, Cho B, Yeom HJ, Kim MG, Oh KH, Ahn C, Yang J. Immunosuppressive mechanisms of embryonic stem cells and mesenchymal stem cells in alloimmune response. Transpl Immunol 2011; 25:7-15. [PMID: 21635949 DOI: 10.1016/j.trim.2011.05.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 12/29/2022]
Abstract
Although both embryonic stem cells (ESCs) and mesenchymal stem cells (MSCs) are known to have immunosuppressive effects, the mechanisms of immunosuppression are still controversial. Both types of stem cells suppressed not only the proliferation but also survival of CD4(+) T cells in vitro. They suppressed secretion of various cytokines (IL-2, IL-12, IFN-γ, TNF-α, IL-4, IL-5, IL-1β, and IL-10), whereas there was no change in the levels of TGF-β or IDO. Classic and modified transwell experiments demonstrated that immunosuppressive activities were mainly mediated by cell-to-cell contact. Granzyme B in the ESCs played a significant role in their immunosuppression, whereas PDL-1, Fas ligand, CD30 or perforin was not involved in the contact-dependent immunosuppression. However, none of the above molecules played a significant role in the immunosuppression by the MSCs. Interestingly, both stem cells increased the proportion of Foxp3(+) regulatory T cells. Our results showed that both ESCs and MSCs suppressed the survival as well as the proliferation of T cells by mainly contact-dependent mechanisms and increased the proportion of regulatory T cells. Granzyme B was involved in immunosuppression by the ESCs in a perforin-independent manner.
Collapse
Affiliation(s)
- Kyu Hyun Han
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Hass R, Kasper C, Böhm 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 2011; 9:12. [PMID: 21569606 PMCID: PMC3117820 DOI: 10.1186/1478-811x-9-12] [Citation(s) in RCA: 1220] [Impact Index Per Article: 87.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 05/14/2011] [Indexed: 12/11/2022] Open
Abstract
The mesenchymal stroma harbors an important population of cells that possess stem cell-like characteristics including self renewal and differentiation capacities and can be derived from a variety of different sources. These multipotent mesenchymal stem cells (MSC) can be found in nearly all tissues and are mostly located in perivascular niches. MSC have migratory abilities and can secrete protective factors and act as a primary matrix for tissue regeneration during inflammation, tissue injuries and certain cancers.These functions underlie the important physiological roles of MSC and underscore a significant potential for the clinical use of distinct populations from the various tissues. MSC derived from different adult (adipose tissue, peripheral blood, bone marrow) and neonatal tissues (particular parts of the placenta and umbilical cord) are therefore compared in this mini-review with respect to their cell biological properties, surface marker expression and proliferative capacities. In addition, several MSC functions including in vitro and in vivo differentiation capacities within a variety of lineages and immune-modulatory properties are highlighted. Differences in the extracellular milieu such as the presence of interacting neighbouring cell populations, exposure to proteases or a hypoxic microenvironment contribute to functional developments within MSC populations originating from different tissues, and intracellular conditions such as the expression levels of certain micro RNAs can additionally balance MSC function and fate.
Collapse
Affiliation(s)
- Ralf Hass
- Laboratory of Biochemistry and Tumor Biology, Gynecology Research Unit, Department of Obstetrics and Gynecology, Medical University, Hannover, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Cornelia Kasper
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstrasse 5, 30167 Hannover, Germany
| | - Stefanie Böhm
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstrasse 5, 30167 Hannover, Germany
| | - Roland Jacobs
- Department of Clinical Immunology and Rheumatology, Medical University, Hannover, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
175
|
Mesenchymal stromal cells: facilitators of successful transplantation? Cell Stem Cell 2011; 7:431-42. [PMID: 20887949 DOI: 10.1016/j.stem.2010.09.009] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 09/01/2010] [Accepted: 09/15/2010] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) possess immunomodulatory and reparative properties. Through specific interactions with immune cells that participate in both innate and adaptive responses, MSCs exposed to an inflammatory microenvironment can downregulate many immune effector functions. Clinical trials focusing on MSCs to treat graft-versus-host disease (GvHD) and autoimmune diseases are underway. Current analyses suggest that MSCs will improve cell and solid organ transplantation by ameliorating rejection and possibly eliminating the requirement for prolonged regimens of conventional immunosuppressive drugs. This review examines the in vitro and in vivo evidence for the clinical use of bone marrow derived MSCs.
Collapse
|
176
|
Shi M, Liu ZW, Wang FS, Peruzzi L, Daprà V, Loiacono E, Vatrano S, Rollino C, Sepe V, Rampino T, Dal Canton A. Immunomodulatory properties and therapeutic application of mesenchymal stem cells. Clin Exp Immunol 2011. [PMID: 21352202 DOI: 10.1111/j.1365-2249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multi-potent progenitor cells that are isolated from the bone marrow and several adult organs and tissues. These cells possess remarkable immunosuppressive properties and can inhibit the proliferation and function of the major immune cell populations, including T cells, B cells and natural killer (NK) cells; modulate the activities of dendritic cells (DCs); and induce regulatory T cells both in vivo and in vitro. These unique properties make MSCs ideal candidates for clinical application as immunosuppressants. The immunomodulatory effect of MSCs is mediated by a non-specific anti-proliferative action of these cells, which is dependent on cell-cell contact or secreted soluble factors such as indoleamine 2,3-dioxygenase (IDO), prostaglandin E(2) (PGE(2) ), nitric oxide (NO), histocompatibility leucocyte antigen-G (HLA-G), transforming growth factor (TGF)-β, interferon (IFN)-γ and interleukin (IL)-1β. Considerable progress has been obtained in preclinical studies on MSCs, including those on their ability to activate allogeneic cells. This review examines the current understanding of the immunomodulatory properties of MSCs and its therapeutic implication for immune-mediated diseases and transplant rejection.
Collapse
Affiliation(s)
- M Shi
- Beijing 302 Hospital, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Shi M, Liu ZW, Wang FS. Immunomodulatory properties and therapeutic application of mesenchymal stem cells. Clin Exp Immunol 2011; 164:1-8. [PMID: 21352202 DOI: 10.1111/j.1365-2249.2011.04327.x] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multi-potent progenitor cells that are isolated from the bone marrow and several adult organs and tissues. These cells possess remarkable immunosuppressive properties and can inhibit the proliferation and function of the major immune cell populations, including T cells, B cells and natural killer (NK) cells; modulate the activities of dendritic cells (DCs); and induce regulatory T cells both in vivo and in vitro. These unique properties make MSCs ideal candidates for clinical application as immunosuppressants. The immunomodulatory effect of MSCs is mediated by a non-specific anti-proliferative action of these cells, which is dependent on cell-cell contact or secreted soluble factors such as indoleamine 2,3-dioxygenase (IDO), prostaglandin E(2) (PGE(2) ), nitric oxide (NO), histocompatibility leucocyte antigen-G (HLA-G), transforming growth factor (TGF)-β, interferon (IFN)-γ and interleukin (IL)-1β. Considerable progress has been obtained in preclinical studies on MSCs, including those on their ability to activate allogeneic cells. This review examines the current understanding of the immunomodulatory properties of MSCs and its therapeutic implication for immune-mediated diseases and transplant rejection.
Collapse
Affiliation(s)
- M Shi
- Beijing 302 Hospital, Beijing, China
| | | | | |
Collapse
|
178
|
Regulatory T-cell generation and kidney allograft tolerance induced by mesenchymal stem cells associated with indoleamine 2,3-dioxygenase expression. Transplantation 2011; 90:1312-20. [PMID: 21042238 DOI: 10.1097/tp.0b013e3181fed001] [Citation(s) in RCA: 263] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The immunoregulatory properties of mesenchymal stem cells (MSCs) have been observed in vitro and in vivo. However, the underlying mechanisms of this immunomodulation remain undefined. Recent research demonstrated that MSCs express the tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO), known to suppress T-cell responses. This study was designed to address whether MSCs induce kidney allograft tolerance and whether IDO contributes to the immunoregulatory functions of MSCs in vivo. METHODS MSCs (1×10(6), intravenously) from wild-type (WT-MSCs) or IDO knockout (IDO(-/-)-MSCs) C57BL/6 mice were injected into BALB/c recipients 24 hr after receiving a life-supporting orthotopic C57BL/6 renal graft. RESULTS WT-MSC-treated recipients achieved allograft tolerance with normal histology and undetectable antidonor antibody levels. Tolerant recipients demonstrated increased circulating kynurenine levels and significantly high frequencies of tolerogenic dendritic cells. They also exhibited significantly impaired CD4+ T-cell responses consisting of decreased donor-specific proliferative ability and a Th2-dominant cytokine shift. In addition, high frequencies of CD4+CD25+Foxp3+ regulatory T cells (Tregs) were found in recipient spleens and donor grafts, with antibody-induced CD25+ cell depletion confirming the critical role of Tregs in the MSC-induced tolerance. Interestingly, renal allograft recipients treated with WT MSCs concomitant with the IDO inhibitor 1-methyl-tryptophan, or those treated with IDO(-/-)-MSCs alone, were unable to achieve allograft tolerance--revealing that functional IDO was necessary for the immunosuppression observed with WT-MSC treatment. CONCLUSIONS IDO secreted by MSCs was responsible, at least in part, for induction of kidney allograft tolerance through generation of Tregs. This study supports the clinical application of MSCs in transplantation.
Collapse
|
179
|
Grazia TJ, Plenter RJ, Lepper HM, Victorino F, Miyamoto SD, Crossno JT, Pietra BA, Gill RG, Zamora MR. Prolongation of cardiac allograft survival by a novel population of autologous CD117+ bone marrow-derived progenitor cells. Am J Transplant 2011; 11:34-44. [PMID: 21114653 PMCID: PMC3059253 DOI: 10.1111/j.1600-6143.2010.03335.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Autologous CD117(+) progenitor cells (PC) have been successfully utilized in myocardial infarction and ischemic injury, potentially through the replacement/repair of damaged vascular endothelium. To date, such cells have not been used to enhance solid organ transplant outcome. In this study, we determined whether autologous bone marrow-derived CD117(+) PC could benefit cardiac allograft survival, possibly by replacing donor vascular cells. Autologous, positively selected CD117(+) PC were administered posttransplantation and allografts were assessed for acute rejection. Although significant generation of recipient vascular cell chimerism was not observed, transferred PC disseminated both to the allograft and to peripheral lymphoid tissues and facilitated a significant, dose-dependent prolongation of allograft survival. While CD117(+) PC dramatically inhibited alloreactive T cell proliferation in vitro, this property did not differ from nonprotective CD117(-) bone marrow populations. In vivo, CD117(+) PC did not significantly inhibit T cell alloreactivity or increase peripheral regulatory T cell numbers. Thus, rather than inhibiting adaptive immunity to the allograft, CD117(+) PC may play a cytoprotective role in prolonging graft survival. Importantly, autologous CD117(+) PC appear to be distinct from bone marrow-derived mesenchymal stem cells (MSC) previously used to prolong allograft survival. As such, autologous CD117(+) PC represent a novel cellular therapy for promoting allograft survival.
Collapse
Affiliation(s)
- Todd J. Grazia
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Research 2, Box C272, 9th Floor, Rm 9118, 12700 East 19th Avenue, Aurora, CO 80045, Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045, Integrated Department of Immunology, National Jewish Medical and Research Center and University of Colorado Denver, 1400 Jackson St., K830, Denver, CO 80206
| | - Robert J. Plenter
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Research 2, Box C272, 9th Floor, Rm 9118, 12700 East 19th Avenue, Aurora, CO 80045, Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045
| | - Helen M. Lepper
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Research 2, Box C272, 9th Floor, Rm 9118, 12700 East 19th Avenue, Aurora, CO 80045, Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045
| | - Francisco Victorino
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Research 2, Box C272, 9th Floor, Rm 9118, 12700 East 19th Avenue, Aurora, CO 80045, Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045
| | - Shelley D. Miyamoto
- Department of Pediatrics, Division of Cardiology, The Children’s Hospital, University of Colorado Denver, Anschutz Medical Campus, Heart Institute, 13123 East 16th Avenue, Aurora, CO 80045
| | - Joseph T. Crossno
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Research 2, Box C272, 9th Floor, Rm 9118, 12700 East 19th Avenue, Aurora, CO 80045
| | - Biagio A. Pietra
- Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045, Department of Pediatrics, Division of Cardiology, The Children’s Hospital, University of Colorado Denver, Anschutz Medical Campus, Heart Institute, 13123 East 16th Avenue, Aurora, CO 80045
| | - Ronald G. Gill
- Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045, Integrated Department of Immunology, National Jewish Medical and Research Center and University of Colorado Denver, 1400 Jackson St., K830, Denver, CO 80206, Department of Surgery, Colorado Center for Transplantation Care, Research, and Education (CCTCARE) Institute, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, Aurora, CO 80045
| | - Martin R. Zamora
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Research 2, Box C272, 9th Floor, Rm 9118, 12700 East 19th Avenue, Aurora, CO 80045, Department of Medicine and Immunology, Barbara Davis Center for Childhood Diabetes, Transplantation Immunology, University of Colorado Denver, Anschutz Medical Campus, 1775 Aurora Court, Bldg. M20 Room 3202B, Box B-140, Aurora, CO 80045
| |
Collapse
|
180
|
Eggenhofer E, Steinmann JF, Renner P, Slowik P, Piso P, Geissler EK, Schlitt HJ, Dahlke MH, Popp FC. Mesenchymal stem cells together with mycophenolate mofetil inhibit antigen presenting cell and T cell infiltration into allogeneic heart grafts. Transpl Immunol 2010; 24:157-63. [PMID: 21194567 DOI: 10.1016/j.trim.2010.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 12/12/2010] [Accepted: 12/13/2010] [Indexed: 12/13/2022]
Abstract
Donor-derived mesenchymal stem cells (MSC) can induce long-term acceptance in a rat heart transplantation model when injected prior to transplantation in combination with mycophenolate mofetil (MMF). In contrast, MSC alone cause accelerated graft rejection. To better understand these conflicting data we studied the effects of MSC and MMF on lymphocyte populations in heart allografts and secondary lymphatic organs. Allogeneic MSC injected prior to transplantation are immunogenic in this model because activated CD4+ and CD8+ cells emerged earlier in secondary lymphatic organs of MSC- and MSC/MMF-treated animals, compared to animals not treated with MSC. Consequently T cells infiltrated the grafts of MSC-only treated animals promptly causing accelerated graft rejection. However, few T cells or antigen-presenting cells (APC) infiltrated the grafts of animals treated with MSC and MMF. Consistent with this finding, intercellular adhesion molecule 1 (ICAM-1) and E-selectin was down-regulated exclusively in MSC/MMF-treated grafts, indicating that MSC together with MMF interfere with endothelial activation. Additionally, the presence of interferon-gamma (IFN-γ) enhanced MSC capabilities to suppress T cell proliferation in vitro. Interestingly, MMF did not influence serum IFN-γ levels in vivo. Together, our data indicate that MSC pre-activate T cells, but co-treatment with MMF eliminates these T cells, decreases intragraft APC and T cell trafficking by inhibiting endothelial activation, and allows IFN-γ stimulation of suppressive MSC.
Collapse
Affiliation(s)
- E Eggenhofer
- Department of Surgery, University Medical Center Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Lan Z, Lian D, Liu W, Arp J, Charlton B, Ge W, Brand S, Healey D, DeBenedette M, Nicolette C, Garcia B, Wang H. Prevention of chronic renal allograft rejection by soluble CD83. Transplantation 2010; 90:1278-85. [PMID: 21079552 DOI: 10.1097/tp.0b013e318200005c] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Recombinant human soluble CD83 had previously exhibited significant immunosuppressive properties that involved interference with dendritic cell maturation in both mouse and humans, inhibition of autoimmunity in mice, and induction of antigen-specific mouse cardiac allograft tolerance when used in combination with other immunosuppressive drugs. Our current research focus turned to examining the effects of peritransplant soluble CD83 (sCD83) administration on prevention of chronic renal allograft rejection. METHODS Fisher344-to-Lewis orthotopic rat renal transplants were performed with sequential recipient killing on postoperative days (PODs) 2, 14, and 140 to examine both the acute and chronic effects of peritransplant sCD83 treatment in rat recipients. RESULTS Recipients treated with sCD83 exhibited a marked decrease in IgM and IgG deposition in the graft and antidonor antibody levels in the circulation, as early as POD14 and persisting until POD140. sCD83 treatment also reduced the infiltration of T cells and monocytes into the graft tissue and inhibited intragraft expression of MyD88 and inflammatory cytokine levels during the observation period. sCD83-treated grafts demonstrated normal histology beyond POD140, including dramatic reductions in tubular atrophy and interstitial fibrosis compared with untreated recipients. CONCLUSION We have demonstrated that peritransplant treatment with recombinant sCD83 attenuates both innate and adaptive immune responses and leads to prevention of chronic rejection in a rat renal transplant model. Because sCD83 is of human origin, the therapeutic approach used in our rodent transplant model holds significant promise for clinical transplantation.
Collapse
Affiliation(s)
- Zhu Lan
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Lan Z, Ge W, Arp J, Jiang J, Liu W, Gordon D, Healey D, DeBenedette M, Nicolette C, Garcia B, Wang H. Induction of kidney allograft tolerance by soluble CD83 associated with prevalence of tolerogenic dendritic cells and indoleamine 2,3-dioxygenase. Transplantation 2010; 90:1286-93. [PMID: 21076370 DOI: 10.1097/tp.0b013e3182007bbf] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tolerogenic dendritic cells (Tol-DCs) play a critical role in inducing and maintaining tolerance. Recognizing that both T-cell inactivation and activation are contingent on signals provided by DCs and that graft-specific activated T cells are major mediators of transplant rejection, we aimed to create an environment favoring Tol-DCs with a novel reagent, human soluble CD83 (hsCD83). METHODS Life-supporting orthotopic kidney transplantation was performed in a C57BL/6-to-BALB/c mouse model. The study group was treated with hsCD83 (100 μg/mouse/day, postoperative days -1 to +7, intravenously) and compared with untreated controls. RESULTS Treatment with hsCD83 achieved kidney allograft tolerance (>100 days), with negligible antidonor antibody detected. In contrast, kidney grafts in untreated recipients demonstrated severe rejection after 35 days, characterized by cellular infiltration, interstitial hemorrhage and edema, and glomerular and tubular necrosis, as well as high antidonor antibody titers. In addition, splenic DCs of tolerant recipients exhibited significantly decreased levels of surface major histocompatibility complex class II, CD40, CD80, and intracellular interleukin-12, as well as reduced allogeneic stimulatory capacity. Adoptive transfer of CD11c+ DCs from tolerant hsCD83-treated animals induced kidney allograft tolerance in syngeneic recipients. Blocking indoleamine 2,3-dioxygenase with 1-methyl-tryptophan (15 mg/mouse/day; gavage) prevented the immunosuppressive effect of hsCD83, abrogating hsCD83-induced Tol-DCs and graft tolerance, and leading to acute kidney graft rejection in 22 days. CONCLUSION hsCD83 alone was capable of inducing kidney allograft tolerance through a mechanism involving Tol-DC generation and, at least in part, indoleamine 2,3-dioxygenase activity. Because sCD83 is of human origin, the therapeutic approach used in our mouse transplant model holds significant promise for clinical transplantation.
Collapse
Affiliation(s)
- Zhu Lan
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Ge W, Arp J, Lian D, Liu W, Baroja ML, Jiang J, Ramcharran S, Eldeen FZ, Zinser E, Steinkasserer A, Chou P, Brand S, Nicolette C, Garcia B, Wang H. Immunosuppression involving soluble CD83 induces tolerogenic dendritic cells that prevent cardiac allograft rejection. Transplantation 2010; 90:1145-56. [PMID: 20861805 DOI: 10.1097/tp.0b013e3181f95718] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Dendritic cells (DCs) are crucial regulators of immunity and important in inducing and maintaining tolerance. Here, we investigated the potential of a novel DC-immunomodulating agent, soluble CD83 (sCD83), in inducing transplant tolerance. METHODS We used the C3H-to-C57BL/6 mouse cardiac transplantation model that exhibits a combination of severe cell-mediated rejection and moderate antibody-mediated rejection and investigated whether sCD83 could augment a combination therapy consisting of Rapamycin (Rapa) and anti-CD45RB monoclonal antibody (α-CD45) to prolong allograft survival. RESULTS Monotherapies consisting of Rapa and α-CD45 were incapable of preventing rejection. However, all treatments involving sCD83 were capable of (1) down-modulating expression of various DC surface molecules, such as major histocompatibility complex class II and costimulatory molecules, (2) reducing the allogeneic stimulatory capacity of the DCs, and (3) significantly inhibiting antidonor antibody responses. Most striking results were observed in the triple therapy-treated group, sCD83Rapaα-CD45, where cell-mediated rejection and antibody-mediated rejection were abrogated for over 100 days. Donor-specific tolerance was achieved in long-term surviving recipients, because donor skin transplants were readily accepted for an additional 100 days, whereas third-party skin grafts were rejected. Success of triple therapy treatment was accompanied by enhancement of tolerogenic-DCs that conferred antigen-specific protection on adoptive transfer to recipients of an allogeneic heart graft. CONCLUSIONS Our study revealed that sCD83 is capable of attenuating DC maturation and function, and inducing donor-specific allograft tolerance, in the absence of toxicity. Thus, sCD83 seems to be a safe and valuable counterpart to current DC-modulating agents.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/genetics
- Antigens, CD/pharmacology
- CD11c Antigen/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Drug Therapy, Combination
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Heart Transplantation/immunology
- Histocompatibility Antigens Class II/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunoglobulins/genetics
- Immunoglobulins/pharmacology
- Immunophenotyping
- Immunosuppressive Agents/pharmacology
- Leukocyte Common Antigens/immunology
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Protein Structure, Tertiary
- Recombinant Proteins/pharmacology
- Sirolimus/pharmacology
- Skin Transplantation
- Time Factors
- Transplantation Tolerance/drug effects
- Transplantation, Homologous
- CD83 Antigen
Collapse
Affiliation(s)
- Wei Ge
- Department of Surgery, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Perico N, Casiraghi F, Introna M, Gotti E, Todeschini M, Cavinato RA, Capelli C, Rambaldi A, Cassis P, Rizzo P, Cortinovis M, Marasà M, Golay J, Noris M, Remuzzi G. Autologous mesenchymal stromal cells and kidney transplantation: a pilot study of safety and clinical feasibility. Clin J Am Soc Nephrol 2010; 6:412-22. [PMID: 20930086 DOI: 10.2215/cjn.04950610] [Citation(s) in RCA: 227] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES Mesenchymal stromal cells (MSCs) abrogate alloimmune response in vitro, suggesting a novel cell-based approach in transplantation. Moving this concept toward clinical application in organ transplantation should be critically assessed. DESIGN, SETTING, PARTICIPANTS & MEASUREMENTS A safety and clinical feasibility study (ClinicalTrials.gov, NCT00752479) of autologous MSC infusion was conducted in two recipients of kidneys from living-related donors. Patients were given T cell-depleting induction therapy and maintenance immunosuppression with cyclosporine and mycophenolate mofetil. On day 7 posttransplant, MSCs were administered intravenously. Clinical and immunomonitoring of MSC-treated patients was performed up to day 360 postsurgery. RESULTS Serum creatinine levels increased 7 to 14 days after cell infusion in both MSC-treated patients. A graft biopsy in patient 2 excluded acute graft rejection, but showed a focal inflammatory infiltrate, mostly granulocytes. In patient 1 protocol biopsy at 1-year posttransplant showed a normal graft. Both MSC-treated patients are in good health with stable graft function. A progressive increase of the percentage of CD4+CD25highFoxP3+CD127- Treg and a marked inhibition of memory CD45RO+RA-CD8+ T cell expansion were observed posttransplant. Patient T cells showed a profound reduction of CD8+ T cell activity. CONCLUSIONS Findings from this study in the two patients show that MSC infusion in kidney transplant recipients is feasible, allows enlargement of Treg in the peripheral blood, and controls memory CD8+ T cell function. Future clinical trials with MSCs to look with the greatest care for unwanted side effects is advised.
Collapse
Affiliation(s)
- Norberto Perico
- Transplant Research Center Chiara Cucchi de Alessandri e Gilberto Crespi, Department of Immunology and Transplantation, Ospedali Riuniti di Bergamo, Mario Negri Institute for Pharmacological Research, Centro Anna Maria Astori, Parco Scientifico Tecnologico Kilometro Rosso, via Stezzano 87, 24126 Bergamo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev 2010; 62:1156-66. [PMID: 20828588 DOI: 10.1016/j.addr.2010.08.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Revised: 08/26/2010] [Accepted: 08/27/2010] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) possess a set of several fairly unique properties which make them ideally suited both for cellular therapies/regenerative medicine, and as vehicles for gene and drug delivery. These include: 1) relative ease of isolation; 2) the ability to differentiate into a wide variety of seemingly functional cell types of both mesenchymal and non-mesenchymal origin; 3) the ability to be extensively expanded in culture without a loss of differentiative capacity; 4) they are not only hypoimmunogenic, but they produce immunosuppression upon transplantation; 5) their pronounced anti-inflammatory properties; and 6) their ability to home to damaged tissues, tumors, and metastases following in vivo administration. In this review, we summarize the latest research in the use of mesenchymal stem cells in regenerative medicine, as immunomodulatory/anti-inflammatory agents, and as vehicles for transferring both therapeutic genes in genetic disease and genes designed to destroy malignant cells.
Collapse
|
186
|
Boyd AS, Fairchild PJ. Approaches for immunological tolerance induction to stem cell-derived cell replacement therapies. Expert Rev Clin Immunol 2010; 6:435-48. [PMID: 20441429 DOI: 10.1586/eci.10.20] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The shortage of donors for organ transplantation and also to treat degenerative diseases has led to the development of the new field of regenerative medicine. One aim of this field, in addition to in vivo induction of endogenous tissue regeneration, is to utilize stem cells as a supplementary source of cells to repair or replace tissues or organs that have ceased to function owing to ageing or autoimmunity. Embryonic stem cells hold promise in this respect because of their developmental capacity to generate all tissues within the body. More recently, the discovery of induced pluripotent stem cells, somatic cells reprogrammed to a primitive embryonic-like state by the introduction of pluripotency factors, may also act as an important cell source for cell replacement therapy. However, before cell replacement therapy can become a reality, one must consider how to overcome the potential transplant rejection of stem cell-derived products. There are several potential ways to circumvent the hurdles presented by the immune system in this setting, not least the induction of immunological tolerance in the host. In this review, we consider this and other approaches for engendering acceptance of stem cell-derived tissues.
Collapse
Affiliation(s)
- Ashleigh S Boyd
- Stem Cell Sciences Lab, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX13RE, UK.
| | | |
Collapse
|
187
|
Ge W, Jiang J, Liu W, Lian D, Saito A, Garcia B, Li XC, Wang H. Regulatory T cells are critical to tolerance induction in presensitized mouse transplant recipients through targeting memory T cells. Am J Transplant 2010; 10:1760-73. [PMID: 20636455 DOI: 10.1111/j.1600-6143.2010.03186.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Memory T cells are a significant barrier to induction of transplant tolerance. However, reliable means to target alloreactive memory T cells have remained elusive. In this study, presensitization of BALB/c mice with C57BL/6 skin grafts generated a large number of OX40(+)CD44(hi)effector/memory T cells and resulted in rapid rejection of donor heart allografts. Recognizing that anti-OX40L monoclonal antibody (mAb) (alpha-OX40L) monotherapy prolonged graft survival through inhibition and apoptosis of memory T cells in presensitized recipients, alpha-OX40L was added to the combined treatment protocol of LF15-0195 (LF) and anti-CD45RB (alpha-CD45RB) mAb-a protocol that induced heart allograft tolerance in non-presensitized recipients but failed to induce tolerance in presensitized recipients. Interestingly, this triple therapy restored donor-specific heart allograft tolerance in our presensitized model that was associated with induction of tolerogenic dendritic cells and CD4(+)CD25(+)Foxp3(+) T regulatory cells (Tregs). Of note, CD25(+) T cell depletion in triple therapy recipients prevented establishment of allograft tolerance. In addition, adoptive transfer of donor-primed effector/memory T cells into tolerant recipients markedly reduced levels of Tregs and broke tolerance. Our findings indicated that targeting memory T cells, by blocking OX40 costimulation in presensitized recipients was very important to expansion of Tregs, which proved critical to development of tolerance.
Collapse
Affiliation(s)
- W Ge
- Department of Surgery, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Hoogduijn MJ, Popp F, Verbeek R, Masoodi M, Nicolaou A, Baan C, Dahlke MH. The immunomodulatory properties of mesenchymal stem cells and their use for immunotherapy. Int Immunopharmacol 2010; 10:1496-500. [PMID: 20619384 DOI: 10.1016/j.intimp.2010.06.019] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 06/21/2010] [Accepted: 06/22/2010] [Indexed: 02/07/2023]
Abstract
There is growing interest in the use of mesenchymal stem cells (MSC) for immune therapy. Clinical trials that use MSC for treatment of therapy resistant graft versus host disease, Crohn's disease and organ transplantation have initiated. Nevertheless, the immunomodulatory effects of MSC are only partly understood. Clinical trials that are supported by basic research will lead to better understanding of the potential of MSC for immunomodulatory applications and to optimization of such therapies. In this manuscript we review some recent literature on the mechanisms of immunomodulation by MSC in vitro and animal models, present new data on the secretion of pro-inflammatory and anti-inflammatory cytokines, chemokines and prostaglandins by MSC under resting and inflammatory conditions and discuss the hopes and expectations of MSC-based immune therapy.
Collapse
Affiliation(s)
- Martin J Hoogduijn
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
189
|
Zhao S, Wehner R, Bornhäuser M, Wassmuth R, Bachmann M, Schmitz M. Immunomodulatory Properties of Mesenchymal Stromal Cells and Their Therapeutic Consequences for Immune-Mediated Disorders. Stem Cells Dev 2010; 19:607-14. [DOI: 10.1089/scd.2009.0345] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Senming Zhao
- Institute of Immunology, Technical University of Dresden, Dresden, Germany
| | - Rebekka Wehner
- Institute of Immunology, Technical University of Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Department of Medicine I, Medical Faculty, Technical University of Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ralf Wassmuth
- Department of Medicine I, Medical Faculty, Technical University of Dresden, Dresden, Germany
- DKMS Life Science Lab, Dresden, Germany
| | - Michael Bachmann
- Institute of Immunology, Technical University of Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Technical University of Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
190
|
Han KH, Kang HG, Gil HJ, Lee EM, Ahn C, Yang J. The immunosuppressive effect of embryonic stem cells and mesenchymal stem cells on both primary and secondary alloimmune responses. Transpl Immunol 2010; 23:141-6. [PMID: 20430098 DOI: 10.1016/j.trim.2010.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 02/21/2010] [Accepted: 04/21/2010] [Indexed: 01/12/2023]
Abstract
Recently, both embryonic stem cells and mesenchymal stem cells have been demonstrated to have immunosuppressive effects. The purpose of this study was to elucidate whether the embryonic stem cells and/or mesenchymal stem cells modulate both primary and secondary alloimmune responses. Both stem cells suppressed in vitro proliferation and cytokine production in primary alloimmune responses. They also suppressed in vitro proliferation and cytokine production of the allosensitized CD44+ memory T cells. However, they failed to prolong skin graft survival across both a major mismatch barrier (BALB/C, C57BL6/J) and a minor mismatch barrier (male to female). In conclusion, both embryonic stem cells and mesenchymal stem cells can suppress secondary alloimmune response in vitro as well as primary alloimmune responses; however, neither embryonic stem cells nor mesenchymal stem cells suppressed allograft rejection in stringent murine skin transplantation models.
Collapse
Affiliation(s)
- Kyu Hyun Han
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea
| | | | | | | | | | | |
Collapse
|
191
|
Patel S, King C, Lim P, Habiba U, Dave M, Porecha R, Rameshwar P. Personalizing Stem Cell Research and Therapy: The Arduous Road Ahead or Missed Opportunity? CURRENT PHARMACOGENOMICS AND PERSONALIZED MEDICINE 2010; 8:25-36. [PMID: 20563265 PMCID: PMC2886988 DOI: 10.2174/1875692111008010025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The euphoria of stem cell therapy has diminished, allowing scientists, clinicians and the general public to seriously re-examine how and what types of stem cells would effectively repair damaged tissue, prevent further tissue damage and/or replace lost cells. Importantly, there is a growing recognition that there are substantial person-to-person differences in the outcome of stem cell therapy. Even though the small molecule pharmaceuticals have long remained a primary focus of the personalized medicine research, individualized or targeted use of stem cells to suit a particular individual could help forecast potential failures of the therapy or identify, early on, the individuals who might benefit from stem cell interventions. This would however demand collaboration among several specialties such as pharmacology, immunology, genomics and transplantation medicine. Such transdisciplinary work could also inform how best to achieve efficient and predictable stem cell migration to sites of tissue damage, thereby facilitating tissue repair. This paper discusses the possibility of polarizing immune responses to rationalize and individualize therapy with stem cell interventions, since generalized "one-size-fits-all" therapy is difficult to achieve in the face of the diverse complexities posed by stem cell biology. We also present the challenges to stem cell delivery in the context of the host related factors. Although we focus on the mesenchymal stem cells in this paper, the overarching rationale can be extrapolated to other types of stem cells as well. Hence, the broader purpose of this paper is to initiate a dialogue within the personalized medicine community by expanding the scope of inquiry in the field from pharmaceuticals to stem cells and related cell-based health interventions.
Collapse
Affiliation(s)
- S.A. Patel
- Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - C.C. King
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - P.K. Lim
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - U. Habiba
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - M. Dave
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - R. Porecha
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - P. Rameshwar
- Department of Medicine, Division of Hematology and Oncology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| |
Collapse
|
192
|
Abstract
Since their discovery by Steinman and Cohn in 1973, dendritic cells (DCs) have become increasingly recognized for their crucial role as regulators of innate and adaptive immunity. DCs are exquisitely adept at acquiring, processing, and presenting antigens to T cells. They also adjust the context (and hence the outcome) of antigen presentation in response to a plethora of environmental inputs that signal the occurrence of pathogens or tissue damage. Such signals generally boost DC maturation, which promotes their migration from peripheral tissues into and within secondary lymphoid organs and their capacity to induce and regulate effector T cell responses. Conversely, more recent observations indicate that DCs are also crucial to ensure immunological peace. Indeed, DCs constantly present innocuous self- and nonself-antigens in a fashion that promotes tolerance, at least in part, through the control of regulatory T cells (Tregs). Tregs are specialized T cells that exert their immunosuppressive function through a variety of mechanisms affecting both DCs and effector cells. Here, we review recent advances in our understanding of the relationship between tolerogenic DCs and Tregs.
Collapse
|