J Rheum Dis.  2016 Oct;23(5):279-287. 10.4078/jrd.2016.23.5.279.

Immunomodulatory Function of Mesenchymal Stem Cells for Rheumatoid Arthritis

Affiliations
  • 1Division of Rheumatology, Department of Internal Medicine, Dankook University Medical College, Cheonan, Korea.
  • 2Division of Rheumatology, Department of Internal Medicine, Institute for Immunology and Immunologic Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea. yongbpark@yuhs.ac

Abstract

Developments in our comprehension of the autoimmune and inflammation mechanisms in rheumatoid arthritis (RA) have produced targeted therapies that block aberrant immune cells and cytokine networks, and improved treatment of RA patients considerably. Nevertheless, limitations of these treatments include incomplete treatment response, adverse effects requiring drug withdrawal, and refractory cases. Hence, many researchers have redirected efforts towards investigation of other biological aspects of RA, including the mechanisms driving joint tissue repair and balanced immune regulation. This investigation focuses on mesenchymal stem cell (MSC) research, with the ultimate goal of developing interventions for immune modulation and repair of damaged joints. MSCs are multipotent cells capable of differentiating into mesodermal lineage cells. These cells have also attracted interest for their anti-inflammatory and immunomodulatory capacities. They have many distinctive immunological properties, inhibiting the proliferation and production of cytokines by T, B, natural killer, and dendritic cells. Indeed, MSCs have the capacity to regulate immunity-induced peripheral tolerance, suggesting they can be used as therapeutic tools in RA. This review discusses properties of MSCs, in vitro studies, animal studies, and clinical trials involving MSCs. Our review discusses the current knowledge of the mechanisms of MSC-mediated immunosuppression and potential therapeutic uses of MSCs in RA.

Keyword

Inflammation; Mesenchymal stem cells; Rheumatoid arthritis

MeSH Terms

Animals
Arthritis, Rheumatoid*
Comprehension
Cytokines
Dendritic Cells
Humans
Immunosuppression
In Vitro Techniques
Inflammation
Joints
Mesenchymal Stromal Cells*
Mesoderm
Peripheral Tolerance
Therapeutic Uses
Cytokines
Therapeutic Uses

Figure

  • Figure 1. Suppressive effects of MSCs on immune cells. The effects of MSCs on cells of the immune system are anti-inflammatory. DCs: dendritic cells, MSCs: mesenchymal stem cells, NK cell: natural killer cell, TREG cell: regulatory T cell.


Reference

1. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature. 2003; 423:356–61.
Article
2. Gonzalez A, Maradit Kremers H, Crowson CS, Nicola PJ, Davis JM 3rd, Therneau TM, et al. The widening mortality gap between rheumatoid arthritis patients and the general population. Arthritis Rheum. 2007; 56:3583–7.
Article
3. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011; 365:2205–19.
Article
4. Espinoza F, Fabre S, Pers YM. Remission-induction therapies for early rheumatoid arthritis: evidence to date and clinical implications. Ther Adv Musculoskelet Dis. 2016; 8:107–18.
Article
5. Cohen G, Gossec L, Dougados M, Cantagrel A, Goupille P, Daures JP, et al. Radiological damage in patients with rheumatoid arthritis on sustained remission. Ann Rheum Dis. 2007; 66:358–63.
Article
6. Smolen JS, Aletaha D, Koeller M, Weisman MH, Emery P. New therapies for treatment of rheumatoid arthritis. Lancet. 2007; 370:1861–74.
Article
7. Chiang EY, Kolumam GA, Yu X, Francesco M, Ivelja S, Peng I, et al. Targeted depletion of lymphotoxin-alpha-expressing TH1 and TH17 cells inhibits autoimmune disease. Nat Med. 2009; 15:766–73.
8. Ehrenstein MR, Evans JG, Singh A, Moore S, Warnes G, Isenberg DA, et al. Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNFalpha therapy. J Exp Med. 2004; 200:277–85.
9. Tyndall A, van Laar JM. Stem cells in the treatment of inflammatory arthritis. Best Pract Res Clin Rheumatol. 2010; 24:565–74.
Article
10. Liang J, Li X, Zhang H, Wang D, Feng X, Wang H, et al. Allogeneic mesenchymal stem cells transplantation in patients with refractory RA. Clin Rheumatol. 2012; 31:157–61.
Article
11. Wang L, Wang L, Cong X, Liu G, Zhou J, Bai B, et al. Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: safety and efficacy. Stem Cells Dev. 2013; 22:3192–202.
Article
12. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999; 284:143–7.
Article
13. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8:315–7.
Article
14. Tyndall A, LeBlanc K. Stem cells and rheumatology: update on adult stem cell therapy in autoimmune diseases. Arthritis Rheum. 2006; 55:521–5.
Article
15. Sotiropoulou PA, Perez SA, Salagianni M, Baxevanis CN, Papamichail M. Characterization of the optimal culture conditions for clinical scale production of human mesenchymal stem cells. Stem Cells. 2006; 24:462–71.
Article
16. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringdén O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003; 31:890–6.
17. Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation. 2003; 75:389–97.
Article
18. Eliopoulos N, Stagg J, Lejeune L, Pommey S, Galipeau J. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood. 2005; 106:4057–65.
Article
19. Nauta AJ, Westerhuis G, Kruisselbrink AB, Lurvink EG, Willemze R, Fibbe WE. Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting. Blood. 2006; 108:2114–20.
Article
20. Prigozhina TB, Khitrin S, Elkin G, Eizik O, Morecki S, Slavin S. Mesenchymal stromal cells lose their immunosuppressive potential after allotransplantation. Exp Hematol. 2008; 36:1370–6.
Article
21. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008; 2:141–50.
Article
22. Dazzi F, Krampera M. Mesenchymal stem cells and autoimmune diseases. Best Pract Res Clin Haematol. 2011; 24:49–57.
Article
23. Meisel R, Zibert A, Laryea M, Göbel U, Däubener W, Dilloo D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004; 103:4619–21.
Article
24. Németh K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenu-ate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009; 15:42–9.
Article
25. Asari S, Itakura S, Ferreri K, Liu CP, Kuroda Y, Kandeel F, et al. Mesenchymal stem cells suppress B-cell terminal differentiation. Exp Hematol. 2009; 37:604–15.
Article
26. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005; 105:1815–22.
Article
27. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells. 2008; 26:212–22.
Article
28. Glennie S, Soeiro I, Dyson PJ, Lam EW, Dazzi F. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood. 2005; 105:2821–7.
Article
29. Rasmusson I, Ringdén O, Sundberg B, Le Blanc K. Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation. 2003; 76:1208–13.
Article
30. Prevosto C, Zancolli M, Canevali P, Zocchi MR, Poggi A. Generation of CD4+ or CD8+ regulatory T cells upon mesenchymal stem cell-lymphocyte interaction. Haematologica. 2007; 92:881–8.
Article
31. Tabera S, Pérez-Simón JA, Díez-Campelo M, Sánchez-Abarca LI, Blanco B, López A, et al. The effect of mesenchymal stem cells on the viability, proliferation and differentiation of B-lymphocytes. Haematologica. 2008; 93:1301–9.
Article
32. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008; 111:1327–33.
Article
33. Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol. 2006; 177:2080–7.
Article
34. Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells. 2006; 24:74–85.
Article
35. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, Giordano T, et al. Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood. 2005; 106:419–27.
Article
36. Stappenbeck TS, Miyoshi H. The role of stromal stem cells in tissue regeneration and wound repair. Science. 2009; 324:1666–9.
Article
37. Leibacher J, Henschler R. Biodistribution, migration and homing of systemically applied mesenchymal stem/stromal cells. Stem Cell Res Ther. 2016; 7:7.
Article
38. Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007; 56:1175–86.
Article
39. González MA, Gonzalez-Rey E, Rico L, Büscher D, Delgado M. Treatment of experimental arthritis by inducing immune tolerance with human adipose-derived mesenchymal stem cells. Arthritis Rheum. 2009; 60:1006–19.
Article
40. Choi JJ, Yoo SA, Park SJ, Kang YJ, Kim WU, Oh IH, et al. Mesenchymal stem cells overexpressing interleukin-10 at-tenuate collagen-induced arthritis in mice. Clin Exp Immunol. 2008; 153:269–76.
Article
41. Park MJ, Park HS, Cho ML, Oh HJ, Cho YG, Min SY, et al. Transforming growth factor β-transduced mesenchymal stem cells ameliorate experimental autoimmune arthritis through reciprocal regulation of Treg/Th17 cells and osteoclastogenesis. Arthritis Rheum. 2011; 63:1668–80.
Article
42. Liu Y, Mu R, Wang S, Long L, Liu X, Li R, et al. Therapeutic potential of human umbilical cord mesenchymal stem cells in the treatment of rheumatoid arthritis. Arthritis Res Ther. 2010; 12:R210.
Article
43. Bouffi C, Bony C, Courties G, Jorgensen C, Noël D. IL-6-de-pendent PGE2 secretion by mesenchymal stem cells inhibits local inflammation in experimental arthritis. PLoS One. 2010; 5:e14247.
Article
44. Djouad F, Fritz V, Apparailly F, Louis-Plence P, Bony C, Sany J, et al. Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor alpha in collagen-induced arthritis. Arthritis Rheum. 2005; 52:1595–603.
45. Chen B, Hu J, Liao L, Sun Z, Han Q, Song Z, et al. Flk-1+ mesenchymal stem cells aggravate collagen-induced arthritis by upregulating interleukin-6. Clin Exp Immunol. 2010; 159:292–302.
Article
46. Schurgers E, Kelchtermans H, Mitera T, Geboes L, Matthys P. Discrepancy between the in vitro and in vivo effects of murine mesenchymal stem cells on T-cell proliferation and collagen-induced arthritis. Arthritis Res Ther. 2010; 12:R31.
47. MacDonald GI, Augello A, De Bari C. Role of mesenchymal stem cells in reestablishing immunologic tolerance in autoimmune rheumatic diseases. Arthritis Rheum. 2011; 63:2547–57.
Article
48. Meyerrose TE, De Ugarte DA, Hofling AA, Herrbrich PE, Cordonnier TD, Shultz LD, et al. In vivo distribution of human adipose-derived mesenchymal stem cells in novel xen-otransplantation models. Stem Cells. 2007; 25:220–7.
Article
49. Park KH, Mun CH, Kang MI, Lee SW, Lee SK, Park YB. Treatment of collagen-induced arthritis using immune modulatory properties of human mesenchymal stem cells. Cell Transplant. 2016; 25:1057–72.
Article
50. Garimella MG, Kour S, Piprode V, Mittal M, Kumar A, Rani L, et al. Adipose-derived mesenchymal stem cells prevent systemic bone loss in collagen-induced arthritis. J Immunol. 2015; 195:5136–48.
Article
51. Ra JC, Kang SK, Shin IS, Park HG, Joo SA, Kim JG, et al. Stem cell treatment for patients with autoimmune disease by systemic infusion of culture-expanded autologous adipose tissue derived mesenchymal stem cells. J Transl Med. 2011; 9:181.
Article
52. Safety and efficacy study of human umbilical cord-mesenchymal stem cells for rheumatoid arthritis. Silver Spring (MD): U.S. Food and Drug Administration;2016 Jan. ClinicalTrials.gov Identifier: NCT02643823. Available from:. https://clinicaltrials.gov/ct2/show/NCT02643823?term=NCT02643823&rank=1.
53. Feasibility study of umbilical cord tissue drived mesenchymal stem cells (UC-MSC) in disease modifying antirheumatic drugs (DMARD) resistant rheumatoid arthritis. Panama: Panama: Ministry of Health;. 2013; Oct. ClinicalTrials.gov Identifier: NCT01985464. Available from:. https://clinicaltrials.gov/ct2/show/NCT01985464?term=NCT01985464&rank=1.
54. Phase Ib/IIa, Escalating dose, single blind, clinical trial to assess the safety of the i.v administration of allogeneic adi-pose-derived mesenchymal cells (eASCs) to refractory rheumatoid arthritis patients. Madrid: Spanish Agency for Medicines and Health Products;. 2011; Aug. ClinicalTrials. gov Identifier: NCT01663116. Available from:. https://clinicaltrials.gov/ct2/show/NCT01663116?term=NCT01663116&rank=1.
55. Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall JC, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012; 7:e47559.
Article
Full Text Links
  • JRD
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr