Immunosuppressive potential of human amnion epithelial cells in the treatment of experimental autoimmune encephalomyelitis

Courtney A McDonald, Natalie L Payne, Guizhi Sun, Leon Moussa, Christopher Siatskas, Rebecca Lim, Euan M Wallace, Graham Jenkin, Claude C A Bernard, Courtney A McDonald, Natalie L Payne, Guizhi Sun, Leon Moussa, Christopher Siatskas, Rebecca Lim, Euan M Wallace, Graham Jenkin, Claude C A Bernard

Abstract

Background: Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS). In recent years, it has been found that cells such as human amnion epithelial cells (hAECs) have the ability to modulate immune responses in vitro and in vivo and can differentiate into multiple cell lineages. Accordingly, we investigated the immunoregulatory effects of hAECs as a potential therapy in an MS-like disease, EAE (experimental autoimmune encephalomyelitis), in mice.

Methods: Using flow cytometry, the phenotypic profile of hAECs from different donors was assessed. The immunomodulatory properties of hAECs were examined in vitro using antigen-specific and one-way mixed lymphocyte proliferation assays. The therapeutic efficacy of hAECs was examined using a relapsing-remitting model of EAE in NOD/Lt mice. T cell responsiveness, cytokine secretion, T regulatory, and T helper cell phenotype were determined in the peripheral lymphoid organs and CNS of these animals.

Results: In vitro, hAECs suppressed both specific and non-specific T cell proliferation, decreased pro-inflammatory cytokine production, and inhibited the activation of stimulated T cells. Furthermore, T cells retained their naïve phenotype when co-cultured with hAECs. In vivo studies revealed that hAECs not only suppressed the development of EAE but also prevented disease relapse in these mice. T cell responses and production of the pro-inflammatory cytokine interleukin (IL)-17A were reduced in hAEC-treated mice, and this was coupled with a significant increase in the number of peripheral T regulatory cells and naïve CD4+ T cells. Furthermore, increased proportions of Th2 cells in the peripheral lymphoid organs and within the CNS were observed.

Conclusion: The therapeutic effect of hAECs is in part mediated by inducing an anti-inflammatory response within the CNS, demonstrating that hAECs hold promise for the treatment of autoimmune diseases like MS.

Figures

Fig. 1
Fig. 1
hAECs inhibit T cell responses and allogeneic and xenogeneic proliferation. Proliferative response of human PBMCs (a) or splenocytes from naïve C57BL/6 mice (b) stimulated with anti-CD3/CD28 antibodies in the presence or absence of hAECs at different hAECs to splenocyte ratios (n = 6 performed in triplicate, *P < 0.05, ***P < 0.001 compared to T cells). c Proliferative response of 2D2 splenocytes stimulated with 20 μg MOG35-55 in the presence or absence of hAECs at different ratios (n = 6 performed in triplicate, **P < 0.01, ***P < 0.001 compared to T cells). d Cytokine secretion profile in supernatant from co-cultures of hAECs and 2D2 splenocytes at 1:5 ratio (n = 5, **P < 0.01 ***P < 0.001 compared to T cells). e Proliferative response of C57BL/6 splenocytes co-cultured with hAECs (n = 4 performed in triplicate, ***P < 0.0001 compared to B6 + Bc). f Proliferative response of C57BL/6 splenocytes co-cultured with irradiated Balb/c splenocytes in the absence or presence of hAECs at hAEC to C57BL/6 T cell ratio of 1:5 (n = 4 preformed in triplicate, ***P < 0.0001 compared to B6 + Bc). g Cytokine secretion profile in supernatant from MLR cultures with hAECs. (n = 4, *P < 0.05,**P < 0.01, ***P < 0.001 compared to B6 + Bc)
Fig. 2
Fig. 2
hAECs inhibit upregulation of T cell activation markers and retain T cells in a naïve phenotype. Naïve CD4+ T cells were stimulated with anti-CD3/CD28 in the presence or absence of hAECs for 5 days and then analyzed by flow cytometry. a, b Representative profiles of CD25 expression on stimulated CD4+ T cells. c, d Representative profiles of CD44 and CD62L expression on stimulated CD4 + CD25+ T cells. CD62LhiCD44lo denotes naïve T cells, CD62LloCD44hi denotes memory cells. Data are representative of two independent experiments with n = 3 mice
Fig. 3
Fig. 3
Administration of hAECs on day 8, 10, and 12 suppresses the development of RR-EAE. a Clinical scores of NOD/Lt mice injected i.p. with one or five million hAECs or PBS on day 8, 10, and 12 (indicated by arrows) (n = 7, *P < 0.05 compared to PBS). Clinical scores (b) and cumulative disease score (c) of NOD/Lt mice injected i.p. with 0.1, 0.25, or 0.5 million hAECs or PBS on day 1 and 3 (indicated by arrows) (n = 6–7, **P < 0.01 compared to PBS). Representative spinal cord sections from PBS control mice stained with H&E (d), LFB (f), or Bielschowsky silver stain (h) or mice that received five million hAECs stained with H&E (e), LFB (g), or Bielschowsky silver stain (i) to assess inflammation, demyelination, and axonal damage, respectively. Magnification × 100. IBA-1+ immunoreactivity in the grey matter of spinal cord sections from PBS controls (j) or mice that received five million hAECs (k). Magnification × 400. (l) Total number of IBA-1+ cells/mm2 in different regions of the CNS in PBS controls and hAEC-treated mice (*P < 0.05 compared to PBS)
Fig. 4
Fig. 4
T cell responses in RR-EAE mice following hAEC administration. Mononuclear cells were isolated from the spleen of NOD/Lt mice at day 37. Proliferative response of splenocytes stimulated with rMOG (a) or anti-CD3/CD28 (g). bf Cytokine secretion profile in supernatant from rMOG stimulated splenocyte cultures. hl Cytokine secretion profile in supernatant from anti-CD3/CD28 stimulated splenocyte cultures (n = 6 performed in triplicate, *P < 0.05 compared to PBS)
Fig. 5
Fig. 5
Administration of hAECs increases the total number of Tregs and naïve CD4+ T cells in the periphery. Mononuclear cells were isolated from the lymph nodes and spleen at day 37, and the proportion and number of Tregs or naïve CD4+ T cells was analyzed by flow cytometry. a Total cell number in the lymph nodes. b Proportion of gated lymphocytes that are CD4 + CD25 + Foxp3+ Tregs in the lymph nodes. c Total Treg numbers in the lymph nodes. d Total cell number in the spleen. e Proportion of gated lymphocytes that are CD4 + CD25 + Foxp3+ Tregs in the spleen. f Total Treg numbers in the spleen. g Proportion of CD4+ that are naïve (CD62LhiCD44lo) T cells in the spleen. h Total number of CD4+ naïve (CD62LhiCD44lo) T cells in the spleen. (n = 6, *P < 0.05 compared to PBS)
Fig. 6
Fig. 6
Administration of hAECs increases Th2 cells in the CNS and shifts the inflammatory profile towards an anti-inflammatory environment. Mononuclear cells were isolated from NOD/Lt mice at day 37 and the phenotype of T helper cells was assessed by flow cytometry. Th1 (CD4 + IFN-γ+), Th17 (CD4 + IL-17+), and Th2 (CD4 + IL-4+) cell proportions and the Th1 to Th2 and Th17 to Th2 ratios were determined in the spleen (ae), lymph nodes (fj), and CNS (ko) (n = 6, *P < 0.05, **P < 0.01, ***P < 0.001)

References

    1. Ewing C, Bernard CCA. Insights into the aetiology and pathogenesis of multiple sclerosis. Immunol Cell Biol. 1998;76:47–54. doi: 10.1046/j.1440-1711.1998.00718.x.
    1. Lassmann H. Multiple sclerosis: is there neurodegeneration independent from inflammation? J Neurol Sci. 2007;259(1–2):3–6. doi: 10.1016/j.jns.2006.08.016.
    1. Payne N, Siatskas C, Bernard CC. The promise of stem cell and regenerative therapies for multiple sclerosis. J Autoimmun. 2008;31(3):288–94. doi: 10.1016/j.jaut.2008.04.002.
    1. Kieseier BC, Wiendl H, Leussink VI, Stuve O. Immunomodulatory treatment strategies in multiple sclerosis. J Neurol. 2008;255(Suppl 6):15–21. doi: 10.1007/s00415-008-6004-z.
    1. Li H, Niederkorn JY, Neelam S, Mayhew E, Word RA, McCulley JP, et al. Immunosuppressive factors secreted by human amniotic epithelial cells. Invest Ophthalmol Vis Sci. 2005;46(3):900–7. doi: 10.1167/iovs.04-0495.
    1. Whittle WL, Gibb W, Challis JR. The characterization of human amnion epithelial and mesenchymal cells: the cellular expression, activity and glucocorticoid regulation of prostaglandin output. Placenta. 2000;21(4):394–401. doi: 10.1053/plac.1999.0482.
    1. Sakuragawa N, Kakinuma K, Kikuchi A, Okano H, Uchida S, Kamo I, et al. Human amnion mesenchyme cells express phenotypes of neuroglial progenitor cells. J Neurosci Res. 2004;78(2):208–14. doi: 10.1002/jnr.20257.
    1. Miki T, Mitamura K, Ross MA, Stolz DB, Strom SC. Identification of stem cell marker-positive cells by immunofluorescence in term human amnion. J Reprod Immunol. 2007;75(2):91–6. doi: 10.1016/j.jri.2007.03.017.
    1. Ilancheran S, Michalska A, Peh G, Wallace EM, Pera M, Manuelpillai U. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biol Reprod. 2007;77(3):577–88. doi: 10.1095/biolreprod.106.055244.
    1. Toda A, Okabe M, Yoshida T, Nikaido T. The potential of amniotic membrane/amnion-derived cells for regeneration of various tissues. J Pharmacol Sci. 2007;105(3):215–28. doi: 10.1254/jphs.CR0070034.
    1. Diaz-Prado S, Muinos-Lopez E, Hermida-Gomez T, Rendal-Vazquez ME, Fuentes-Boquete I, de Toro FJ, et al. Multilineage differentiation potential of cells isolated from the human amniotic membrane. J Cell Biochem. 2010;111(4):846–57. doi: 10.1002/jcb.22769.
    1. Banas RA, Trumpower C, Bentlejewski C, Marshall V, Sing G, Zeevi A. Immunogenicity and immunomodulatory effects of amnion-derived multipotent progenitor cells. Hum Immunol. 2008;69(6):321–8. doi: 10.1016/j.humimm.2008.04.007.
    1. Murphy S, Rosli S, Acharya R, Mathias L, Lim R, Wallace E, et al. Amnion epithelial cell isolation and characterization for clinical use. Curr Protoc Stem Cell Biol. 2010;Chapter 1:Unit 1E 6.
    1. Miki T, Lehmann T, Cai H, Stolz DB, Strom SC. Stem cell characteristics of amniotic epithelial cells. Stem Cells. 2005;23(10):1549–59. doi: 10.1634/stemcells.2004-0357.
    1. Tan JL, Chan ST, Wallace EM, Lim R. Human amnion epithelial cells mediate lung repair by directly modulating macrophage recruitment and polarization. Cell Transplant. 2014;23(3):319–28. doi: 10.3727/096368912X661409.
    1. Wolbank S, Peterbauer A, Fahrner M, Hennerbichler S, van Griensven M, Stadler G, et al. Dose-dependent immunomodulatory effect of human stem cells from amniotic membrane: a comparison with human mesenchymal stem cells from adipose tissue. Tissue Eng. 2007;13(6):1173–83. doi: 10.1089/ten.2006.0313.
    1. McDonald C, Siatskas C, Bernard CCA. The emergence of amnion epithelial stem cells for the treatment of multiple sclerosis. Inflamm Regen. 2011;31(3):256–71. doi: 10.2492/inflammregen.31.256.
    1. Liu YH, Vaghjiani V, Tee JY, To K, Cui P, Oh DY, et al. Amniotic epithelial cells from the human placenta potently suppress a mouse model of multiple sclerosis. PLoS One. 2012;7(4):e35758. doi: 10.1371/journal.pone.0035758.
    1. Moodley Y, Illancheran S, Samuel C, Vaghijani V, Atienza D, Williams ED, et al. Human amnion epithelial cell transplantation abrogates lung fibrosis and augments repair. Am J Respir Crit Care Med. 2010;182(5):643–51. doi: 10.1164/rccm.201001-0014OC.
    1. Yawno T, Schuilwerve J, Moss TJ, Vosdoganes P, Westover AJ, Afandi E, et al. Human amnion epithelial cells reduce fetal brain injury in response to intrauterine inflammation. Dev Neurosci. 2013;35(2-3):272–282. doi: 10.1159/000346683.
    1. Kudriashov AF, Artarian AA, Putsillo MV. Use of amnion to repair a dural defect. Zh Vopr Neirokhir Im N N Burdenko. 1981;5:37–40.
    1. Mostaque AK, Abdur Rahman KB. Comparisons of the effects of biological membrane (amnion) and silver sulfadiazine in the management of burn wounds in children. J Burn Care Res. 2011;32(2):200–209. doi: 10.1097/BCR.0b013e31820aad94.
    1. Bujang-Safawi E, Halim AS, Khoo TL, Dorai AA. Dried irradiated human amniotic membrane as a biological dressing for facial burns–a 7-year case series. Burns. 2010;36(6):876–82. doi: 10.1016/j.burns.2009.07.001.
    1. Muraine M, Descargues G, Franck O, Villeroy F, Toubeau D, Menguy E, et al. Amniotic membrane graft in ocular surface disease. Prospective study with 31 cases. J Fr Ophtalmol. 2001;24(8):798–812.
    1. Zemba M, Andrei S, Cucu B, Bratulescu M, Stinghe A, Bobeico V, et al. Amniotic membrane transplantation in palliative treatment of bullous keratopathy. Oftalmologia. 2006;50(4):51–3.
    1. Motolese I, Mazzera L, Frezzotti P, Motolese PA, Motolese E. Use of amniotic membrane transplantation in isolated conjunctival Bowen disease: a case report. Eur J Ophthalmol. 2010;20(3):604–7.
    1. Altan-Yaycioglu R, Akova YA, Oto S. Amniotic membrane transplantation for treatment of symblepharon in a patient with recessive dystrophic epidermolysis bullosa. Cornea. 2006;25(8):971–3. doi: 10.1097/01.ico.0000225708.70135.d0.
    1. Payne NL, Sun G, Herszfeld D, Tat-Goh PA, Verma PJ, Parkington HC, et al. Comparative study on the therapeutic potential of neurally differentiated stem cells in a mouse model of multiple sclerosis. PLoS One. 2012;7(4):e35093. doi: 10.1371/journal.pone.0035093.
    1. Payne NL, Sun G, McDonald C, Layton D, Moussa L, Emerson-Webber A, et al. Distinct immunomodulatory and migratory mechanisms underpin the therapeutic potential of human mesenchymal stem cells in autoimmune demyelination. Cell Transplant. 2013;22(8):1409–25. doi: 10.3727/096368912X657620.
    1. Short MA, Campanale N, Litwak S, Bernard CC. Quantitative and phenotypic analysis of bone marrow-derived cells in the intact and inflamed central nervous system. Cell Adh Migr. 2011;5(5):373–81. doi: 10.4161/cam.5.5.17948.
    1. Parolini O, Alviano F, Bagnara GP, Bilic G, Buuhring HJ, Evangelista M, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells. 2008;26(2):300–11. doi: 10.1634/stemcells.2007-0594.
    1. Lim R, Chan ST, Tan JL, Mockler JC, Murphy SV, Wallace EM. Preterm human amnion epithelial cells have limited reparative potential. Placenta. 2013;34(6):486–92. doi: 10.1016/j.placenta.2013.03.010.
    1. 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(4):315–7. doi: 10.1080/14653240600855905.
    1. Pratama G, Vaghjiani V, Tee JY, Liu YH, Chan J, Tan C, et al. Changes in culture expanded human amniotic epithelial cells: implications for potential therapeutic applications. PLoS One. 2011;6(11):e26136. doi: 10.1371/journal.pone.0026136.
    1. Stadler G, Hennerbichler S, Lindenmair A, Peterbauer A, Hofer K, van Griensven M, et al. Phenotypic shift of human amniotic epithelial cells in culture is associated with reduced osteogenic differentiation in vitro. Cytotherapy. 2008;10(7):743–52. doi: 10.1080/14653240802345804.
    1. McQualter JL, Bernard CCA. Multiple sclerosis: a battle between destruction and repair. J Neurochem. 2007;100(2):295–306. doi: 10.1111/j.1471-4159.2006.04232.x.
    1. Hori J, Wang M, Kamiya K, Takahashi H, Sakuragawa N. Immunological characteristics of amniotic epithelium. Cornea. 2006;25(10 Suppl 1):S53–8. doi: 10.1097/01.ico.0000247214.31757.5c.
    1. Houlihan JM, Biro PA, Harper HM, Jenkinson HJ, Holmes CH. The human amnion is a site of MHC class Ib expression: evidence for the expression of HLA-E and HLA-G. J Immunol. 1995;154(11):5665–74.
    1. Kovats S, Main EK, Librach C, Stubblebine M, Fisher SJ, DeMars R. A class I antigen, HLA-G, expressed in human trophoblasts. Science. 1990;248(4952):220–3. doi: 10.1126/science.2326636.
    1. Lovett-Racke AE, Trotter JL, Lauber J, Perrin PJ, June CH, Racke MK. Decreased dependence of myelin basic protein-reactive T cells on CD28-mediated costimulation in multiple sclerosis patients. A marker of activated/memory T cells. J Clin Invest. 1998;101(4):725–30. doi: 10.1172/JCI1528.
    1. Guan H, Nagarkatti PS, Nagarkatti M. CD44 Reciprocally regulates the differentiation of encephalitogenic Th1/Th17 and Th2/regulatory T cells through epigenetic modulation involving DNA methylation of cytokine gene promoters, thereby controlling the development of experimental autoimmune encephalomyelitis. J Immunol. 2011;186(12):6955–64. doi: 10.4049/jimmunol.1004043.
    1. Brocke S, Piercy C, Steinman L, Weissman IL, Veromaa T. Antibodies to CD44 and integrin alpha4, but not L-selectin, prevent central nervous system inflammation and experimental encephalomyelitis by blocking secondary leukocyte recruitment. Proc Natl Acad Sci U S A. 1999;96(12):6896–901. doi: 10.1073/pnas.96.12.6896.
    1. Williams JL, Kithcart AP, Smith KM, Shawler T, Cox GM, Whitacre CC. Memory cells specific for myelin oligodendrocyte glycoprotein (MOG) govern the transfer of experimental autoimmune encephalomyelitis. J Neuroimmunol. 2011;234(1–2):84–92. doi: 10.1016/j.jneuroim.2011.02.008.
    1. Slavin AJ, Johns TG, Orain JM, Bernard CC. Regulation of myelin oligodendrocyte glycoprotein in different species throughout development. Dev Neurosci. 1997;19(1):69–78. doi: 10.1159/000111187.
    1. Khoury SJ, Hancock WW, Weiner HL. Oral tolerance to myelin basic protein and natural recovery from experimental autoimmune encephalomyelitis are associated with downregulation of inflammatory cytokines and differential upregulation of transforming growth factor beta, interleukin 4, and prostaglandin E expression in the brain. J Exp Med. 1992;176(5):1355–64. doi: 10.1084/jem.176.5.1355.
    1. Racke MK, Bonomo A, Scott DE, Cannella B, Levine A, Raine CS, et al. Cytokine-induced immune deviation as a therapy for inflammatory autoimmune disease. J Exp Med. 1994;180(5):1961–6. doi: 10.1084/jem.180.5.1961.
    1. Kohm AP, Carpentier PA, Anger HA, Miller SD. Cutting edge: CD4 + CD25+ regulatory T cells suppress antigen-specific autoreactive immune responses and central nervous system inflammation during active experimental autoimmune encephalomyelitis. J Immunol. 2002;169(9):4712–6. doi: 10.4049/jimmunol.169.9.4712.
    1. Tan JL, Chan ST, Lo CY, Deane JA, McDonald CA, Bernard CC, et al. Amnion cell mediated immune modulation following bleomycin challenge: controlling the regulatory T cell response. Stem Cell Res Ther. 2015;6(1):8.
    1. Pianta S, Bonassi-Signoroni P, Muradore I, Rodrigues MF, Rossi D, Silini A, et al. Amniotic membrane mesenchymal cells-derived factors skew T cell polarization toward Treg and downregulate Th1 and Th17 cells subsets. Stem Cell Rev. 2014; doi:10.1007/s12015-014-9558-4.
    1. Parolini O, Souza-Moreira L, O’Valle F, Magatti M, Hernandez-Cortes P, Gonzalez-Rey E, et al. Therapeutic effect of human amniotic membrane-derived cells on experimental arthritis and other inflammatory disorders. Arthritis Rheumatol. 2014;66(2):327–39. doi: 10.1002/art.38206.
    1. Furlan R, Poliani PL, Galbiati F, Bergami A, Grimaldi LM, Comi G, et al. Central nervous system delivery of interleukin 4 by a nonreplicative herpes simplex type 1 viral vector ameliorates autoimmune demyelination. Hum Gene Ther. 1998;9(17):2605–17. doi: 10.1089/hum.1998.9.17-2605.
    1. Butti E, Bergami A, Recchia A, Brambilla E, Del Carro U, Amadio S, et al. IL4 gene delivery to the CNS recruits regulatory T cells and induces clinical recovery in mouse models of multiple sclerosis. Gene Ther. 2008;15(7):504–15. doi: 10.1038/gt.2008.10.
    1. Lisak RP, Nedelkoska L, Studzinski D, Bealmear B, Xu W, Benjamins JA. Cytokines regulate neuronal gene expression: differential effects of Th1, Th2 and monocyte/macrophage cytokines. J Neuroimmunol. 2011;238(1–2):19–33. doi: 10.1016/j.jneuroim.2011.06.010.
    1. Razmkhah M, Jaberipour M, Erfani N, Habibagahi M, Talei AR, Ghaderi A. Adipose derived stem cells (ASCs) isolated from breast cancer tissue express IL-4, IL-10 and TGF-beta1 and upregulate expression of regulatory molecules on T cells: do they protect breast cancer cells from the immune response? Cell Immunol. 2011;266(2):116–22. doi: 10.1016/j.cellimm.2010.09.005.
    1. Komiya T, Sugiyama T, Takeda K, Watanabe N, Imai M, Kokubo M, et al. Suppressive effects of a novel CC chemokine receptor 4 antagonist on Th2 cell trafficking in ligand- and antigen-induced mouse models. Eur J Pharmacol. 2013;720(1–3):335–43. doi: 10.1016/j.ejphar.2013.10.006.
    1. Wang X, Kimbrel EA, Ijichi K, Paul D, Lazorchak AS, Chu J, et al. Human ESC-derived MSCs outperform bone marrow MSCs in the treatment of an EAE model of multiple sclerosis. Stem Cell Reports. 2014;3(1):115–30. doi: 10.1016/j.stemcr.2014.04.020.
    1. Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5(1):54–63. doi: 10.1016/j.stem.2009.05.003.
    1. Mathias LJ, Khong SM, Spyroglou L, Payne NL, Siatskas C, Thorburn AN, et al. Alveolar macrophages are critical for the inhibition of allergic asthma by mesenchymal stromal cells. J Immunol. 2013;191(12):5914–24. doi: 10.4049/jimmunol.1300667.
    1. Chen L, Coleman R, Leang R, Tran H, Kopf A, Walsh CM, et al. Human neural precursor cells promote neurologic recovery in a viral model of multiple sclerosis. Stem Cell Reports. 2014;2(6):825–37. doi: 10.1016/j.stemcr.2014.04.005.

Source: PubMed

3
Abonneren