Skip to main content

The Potential of Human Amnion Epithelial Cells as an Immunomodulatory and Neuroregenerative Treatment for Multiple Sclerosis

  • Chapter
  • First Online:
Perinatal Stem Cells

Abstract

Multiple sclerosis (MS) is a chronic autoimmune disease that involves inflammation, axonal damage, and demyelination of the brain and spinal cord. Whilst there are a number of treatments currently available for this often debilitating disease, they all target the immune system with variable efficacy and are often associated with potentially serious side effects. Most compellingly is that whilst these drugs can prevent relapses and minimize long-term disability, they are not curative, do not aid in the regeneration of damaged neural tissue or protect the CNS from neurodegeneration associated with progressive forms of MS. As such there is an urgent need for the development of new therapies that are effective for patients with relapsing–remitting as well as progressive MS and can not only prevent CNS injury but assist with repair. One such potential therapy is stem cell transplantation. A number of different stem cell types have been shown to be efficacious in murine models of MS, and thus clinical trials are being undertaken to investigate the efficacy in the clinic. A novel stem cell source that is gaining attention is human amnion epithelial cells (hAECs). These cells have been shown to suppress inflammation, migrate to inflamed sites within the CNS, engraft and differentiate toward neural lineages. Given their immunosuppressive and neuroregenerative potential, hAECs are attractive candidates for the treatment of MS and other neurodegenerative disorders that affect the CNS.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. McQualter JL, Bernard CCA. Multiple sclerosis: a battle between destruction and repair. J Neurochem. 2007;100:295–306.

    CAS  PubMed  Google Scholar 

  2. Hauser SL, Oksenberg JR. The neurobiology of multiple sclerosis: genes, inflammation, and neurodegeneration. Neuron. 2006; 52:61–76.

    CAS  PubMed  Google Scholar 

  3. Bernard CCA, et al. Myelin oligodendrocyte glycoprotein: a novel candidate autoantigen in multiple sclerosis. J Mol Med. 1997; 75:77–88.

    CAS  PubMed  Google Scholar 

  4. Ewing C, Bernard CCA. Insights into the aetiology and pathogenesis of multiple sclerosis. Immunol Cell Biol. 1998;76: 47–54.

    CAS  PubMed  Google Scholar 

  5. Keegan BM, Noseworthy JH. Multiple sclerosis. Annu Rev Med. 2002;53:285–302.

    CAS  PubMed  Google Scholar 

  6. Karussis D, et al. Neuroprotection in multiple sclerosis. Clin Neurol Neurosurg. 2006;108:250–4.

    PubMed  Google Scholar 

  7. Lassmann H, Brück W, Lucchinetti CF. The immunopathology of multiple sclerosis: an overview. Brain Pathol. 2007;17:210–8.

    PubMed  Google Scholar 

  8. Rovaris M, et al. Secondary progressive multiple sclerosis: current knowledge and future challenges. Lancet Neurol. 2006;5: 343–54.

    PubMed  Google Scholar 

  9. Dutta R, Trapp BD. Pathogenesis of axonal and neuronal damage in multiple sclerosis. Neurology. 2007;68:S22–31; discussion S43–54.

    PubMed  Google Scholar 

  10. Zuvich RL, McCauley JL, Pericak-Vance MA, Haines JL. Genetics and pathogenesis of multiple sclerosis. Semin Immunol. 2009; 21:328–33.

    CAS  PubMed Central  PubMed  Google Scholar 

  11. Nicholson MJ, Hahn M, Wucherpfennig KW. Unusual features of self-peptide/MHC binding by autoimmune T cell receptors. Immunity. 2005;23:351–60.

    CAS  PubMed Central  PubMed  Google Scholar 

  12. Manuel SL, Rahman R, Wigdahl B, Khan ZK, Jain P. Dendritic cells in autoimmune diseases and neuroinflammatory disorders. Front Biosci. 2007;12:4315–35.

    CAS  PubMed  Google Scholar 

  13. Payne N, Siatskas C, Barnard A, Bernard CC. The prospect of stem cells as multi-faceted purveyors of immune modulation, repair and regeneration in multiple sclerosis. Curr Stem Cell Res Ther. 2011;6(1):50–62.

    CAS  PubMed  Google Scholar 

  14. Gourraud PA, et al. Aggregation of multiple sclerosis genetic risk variants in multiple and single case families. Ann Neurol. 2011;69:65–74.

    PubMed Central  PubMed  Google Scholar 

  15. Ramagopalan SV, Dobson R, Meier UC, Giovannoni G. Multiple sclerosis: risk factors, prodromes, and potential causal pathways. Lancet Neurol. 2010;9:727–39.

    PubMed  Google Scholar 

  16. Marrosu MG, et al. Dissection of the HLA association with multiple sclerosis in the founder isolated population of Sardinia. Hum Mol Genet. 2001;10:2907–16.

    CAS  PubMed  Google Scholar 

  17. Sawcer S, et al. A high-density screen for linkage in multiple sclerosis. Am J Hum Genet. 2005;77:454–67.

    PubMed  Google Scholar 

  18. De Jager PL, et al. Meta-analysis of genome scans and replication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis susceptibility loci. Nat Genet. 2009;41:776–82.

    PubMed Central  PubMed  Google Scholar 

  19. Sawcer S, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476: 214–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Baranzini SE, et al. Genome, epigenome and RNA sequences of monozygotic twins discordant for multiple sclerosis. Nature. 2010;464:1351–6.

    CAS  PubMed Central  PubMed  Google Scholar 

  21. Kurtzke JF. Geographic distribution of multiple sclerosis: an update with special reference to Europe and the Mediterranean region. Acta Neurol Scand. 1980;62:65–80.

    CAS  PubMed  Google Scholar 

  22. Hammond SR, et al. The epidemiology of multiple sclerosis in three Australian cities: Perth, Newcastle and Hobart. Brain. 1988;111(Pt 1):1–25.

    PubMed  Google Scholar 

  23. Munger KL, Levin LI, Hollis BW, Howard NS, Ascherio A. Serum 25-hydroxyvitamin D levels and risk of multiple sclerosis. JAMA. 2006;296:2832–8.

    CAS  PubMed  Google Scholar 

  24. Agliardi C, et al. Vitamin D receptor (VDR) gene SNPs influence VDR expression and modulate protection from multiple sclerosis in HLA-DRB1*15-positive individuals. Brain Behav Immun. 2011;25(7):1460–7.

    CAS  PubMed  Google Scholar 

  25. Sundstrom P, et al. An altered immune response to Epstein-Barr virus in multiple sclerosis: a prospective study. Neurology. 2004; 62:2277–82.

    CAS  PubMed  Google Scholar 

  26. Mantzourani ED, Mavromoustakos TM, Platts JA, Matsoukas JM, Tselios TV. Structural requirements for binding of myelin basic protein (MBP) peptides to MHC II: effects on immune regulation. Curr Med Chem. 2005;12:1521–35.

    CAS  PubMed  Google Scholar 

  27. Purcell AW, McCluskey J, Rossjohn J. More than one reason to rethink the use of peptides in vaccine design. Nat Rev Drug Discov. 2007;6:404–14.

    CAS  PubMed  Google Scholar 

  28. Palmer E. Negative selection—clearing out the bad apples from the T-cell repertoire. Nat Rev Immunol. 2003;3:383–91.

    CAS  PubMed  Google Scholar 

  29. Walker LS, Abbas AK. The enemy within: keeping self-reactive T cells at bay in the periphery. Nat Rev Immunol. 2002;2:11–9.

    CAS  PubMed  Google Scholar 

  30. Weber MS, et al. B-cell activation influences T-cell polarization and outcome of anti-CD20 B-cell depletion in central nervous system autoimmunity. Ann Neurol. 2010;68:369–83.

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Lyons JA, Ramsbottom MJ, Cross AH. Critical role of antigen-specific antibody in experimental autoimmune encephalomyelitis induced by recombinant myelin oligodendrocyte glycoprotein. Eur J Immunol. 2002;32:1905–13.

    CAS  PubMed  Google Scholar 

  32. Bogie JF, Stinissen P, Hellings N, Hendriks JJ. Myelin-phagocytosing macrophages modulate autoreactive T cell proliferation. J Neuroinflammation. 2011;8:85.

    CAS  PubMed Central  PubMed  Google Scholar 

  33. Piani D, Frei K, Do KQ, Cuenod M, Fontana A. Murine brain macrophages induced NMDA receptor mediated neurotoxicity in vitro by secreting glutamate. Neurosci Lett. 1991;133:159–62.

    CAS  PubMed  Google Scholar 

  34. Boven LA, et al. Myelin-laden macrophages are anti-inflammatory, consistent with foam cells in multiple sclerosis. Brain. 2006; 129:517–26.

    PubMed  Google Scholar 

  35. Bailey SL, Schreiner B, McMahon EJ, Miller SD. CNS myeloid DCs presenting endogenous myelin peptides ‘preferentially’ polarize CD4+ T(H)-17 cells in relapsing EAE. Nat Immunol. 2007;8:172–80.

    CAS  PubMed  Google Scholar 

  36. Becher B, Blain M, Antel JP. CD40 engagement stimulates IL-12 p70 production by human microglial cells: basis for Th1 polarization in the CNS. J Neuroimmunol. 2000;102:44–50.

    CAS  PubMed  Google Scholar 

  37. Cua DJ, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421:744–8.

    CAS  PubMed  Google Scholar 

  38. Aloisi F. Immune function of microglia. Glia. 2001;36:165–79.

    CAS  PubMed  Google Scholar 

  39. Ford AL, Foulcher E, Lemckert FA, Sedgwick JD. Microglia induce CD4 T lymphocyte final effector function and death. J Exp Med. 1996;184:1737–45.

    CAS  PubMed  Google Scholar 

  40. Juedes AE, Ruddle NH. Resident and infiltrating central nervous system APCs regulate the emergence and resolution of experimental autoimmune encephalomyelitis. J Immunol. 2001;166: 5168–75.

    CAS  PubMed  Google Scholar 

  41. Zhang GX, et al. Induction of experimental autoimmune encephalomyelitis in IL-12 receptor-beta 2-deficient mice: IL-12 responsiveness is not required in the pathogenesis of inflammatory demyelination in the central nervous system. J Immunol. 2003; 170:2153–60.

    CAS  PubMed  Google Scholar 

  42. Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol. 2007;25:821–52.

    CAS  PubMed  Google Scholar 

  43. Goverman J. Autoimmune T, cell responses in the central nervous system. Nat Rev Immunol. 2009;9:393–407.

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Krakowski M, Owens T. Interferon-gamma confers resistance to experimental allergic encephalomyelitis. Eur J Immunol. 1996; 26:1641–6.

    CAS  PubMed  Google Scholar 

  45. Ferber IA, et al. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol. 1996;156:5–7.

    CAS  PubMed  Google Scholar 

  46. Willenborg DO, Fordham S, Bernard CC, Cowden WB, Ramshaw IA. IFN-gamma plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. J Immunol. 1996;157:3223–7.

    CAS  PubMed  Google Scholar 

  47. McKenzie BS, Kastelein RA, Cua DJ. Understanding the IL-23-IL-17 immune pathway. Trends Immunol. 2006;27:17–23.

    CAS  PubMed  Google Scholar 

  48. Chen Y, et al. Anti-IL-23 therapy inhibits multiple inflammatory pathways and ameliorates autoimmune encephalomyelitis. J Clin Invest. 2006;116:1317–26.

    CAS  PubMed Central  PubMed  Google Scholar 

  49. Bettelli E, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8.

    CAS  PubMed  Google Scholar 

  50. Li B, et al. FOXP3 ensembles in T-cell regulation. Immunol Rev. 2006;212:99–113.

    CAS  PubMed  Google Scholar 

  51. Komiyama Y, et al. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol. 2006;177:566–73.

    CAS  PubMed  Google Scholar 

  52. Hofstetter HH, et al. Therapeutic efficacy of IL-17 neutralization in murine experimental autoimmune encephalomyelitis. Cell Immunol. 2005;237:123–30.

    CAS  PubMed  Google Scholar 

  53. Lock C, et al. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med. 2002;8:500–8.

    CAS  PubMed  Google Scholar 

  54. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517.

    CAS  PubMed  Google Scholar 

  55. Chen W, et al. Conversion of peripheral CD4+ CD25- naive T cells to CD4+ CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–86.

    CAS  PubMed Central  PubMed  Google Scholar 

  56. Kishimoto T. Interleukin-6: from basic science to medicine— 40 years in immunology. Annu Rev Immunol. 2005;23:1–21.

    CAS  PubMed  Google Scholar 

  57. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+ CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6.

    CAS  PubMed  Google Scholar 

  58. Sakaguchi S. Naturally arising Foxp3-expressing CD25+ CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol. 2005;6:345–52.

    CAS  PubMed  Google Scholar 

  59. Campbell DJ, Ziegler SF. FOXP3 modifies the phenotypic and functional properties of regulatory T cells. Nat Rev Immunol. 2007;7:305–10.

    CAS  PubMed  Google Scholar 

  60. Bynoe MS, Bonorino P, Viret C. Control of experimental autoimmune encephalomyelitis by CD4+ suppressor T cells: peripheral versus in situ immunoregulation. J Neuroimmunol. 2007;191: 61–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  61. Bach JF. Regulatory T, cells under scrutiny. Nat Rev Immunol. 2003;3:189–98.

    PubMed  Google Scholar 

  62. Cottrez F, Groux H. Specialization in tolerance: innate CD(4+)CD(25+) versus acquired TR1 and TH3 regulatory T cells. Transplantation. 2004;77:S12–5.

    PubMed  Google Scholar 

  63. Groux H, et al. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature. 1997;389:737–42.

    CAS  PubMed  Google Scholar 

  64. Zamvil SS, Steinman L. Diverse targets for intervention during inflammatory and neurodegenerative phases of multiple sclerosis. Neuron. 2003;38:685–8.

    CAS  PubMed  Google Scholar 

  65. Fawcett JW, Asher RA. The glial scar and central nervous system repair. Brain Res Bull. 1999;49:377–91.

    CAS  PubMed  Google Scholar 

  66. Karnezis T, et al. The neurite outgrowth inhibitor Nogo A is involved in autoimmune-mediated demyelination. Nat Neurosci. 2004;7:736–44.

    CAS  PubMed  Google Scholar 

  67. Harel NY, Strittmatter SM. Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury? Nat Rev Neurosci. 2006;7:603–16.

    CAS  PubMed Central  PubMed  Google Scholar 

  68. Windrem MS, et al. Fetal and adult human oligodendrocyte progenitor cell isolates myelinate the congenitally dysmyelinated brain. Nat Med. 2004;10:93–7.

    CAS  PubMed  Google Scholar 

  69. Chang A, Tourtellotte WW, Rudick R, Trapp BD. Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N Engl J Med. 2002;346:165–73.

    PubMed  Google Scholar 

  70. Calza L, et al. Proliferation and phenotype regulation in the subventricular zone during experimental allergic encephalomyelitis: in vivo evidence of a role for nerve growth factor. Proc Natl Acad Sci U S A. 1998;95:3209–14.

    CAS  PubMed Central  PubMed  Google Scholar 

  71. Payne N, Siatskas C, Bernard CC. The promise of stem cell and regenerative therapies for multiple sclerosis. J Autoimmun. 2008; 31:288–94.

    CAS  PubMed  Google Scholar 

  72. Jones JL, Coles AJ. New treatment strategies in multiple sclerosis. Exp Neurol. 2010;225:34–9.

    PubMed  Google Scholar 

  73. Wiendl H, et al. Basic and escalating immunomodulatory treatments in multiple sclerosis: current therapeutic recommendations. J Neurol. 2008;255:1449–63.

    CAS  PubMed  Google Scholar 

  74. Miller DH, Leary SM. Primary-progressive multiple sclerosis. Lancet Neurol. 2007;6:903–12.

    PubMed  Google Scholar 

  75. Wolinsky JS, et al. Glatiramer acetate in primary progressive multiple sclerosis: results of a multinational, multicenter, double-blind, placebo-controlled trial. Ann Neurol. 2007;61:14–24.

    CAS  PubMed  Google Scholar 

  76. Bielekova B, et al. Effect of anti-CD25 antibody daclizumab in the inhibition of inflammation and stabilization of disease progression in multiple sclerosis. Arch Neurol. 2009;66:483–9.

    PubMed Central  PubMed  Google Scholar 

  77. Bielekova B, et al. Humanized anti-CD25 (daclizumab) inhibits disease activity in multiple sclerosis patients failing to respond to interferon beta. Proc Natl Acad Sci U S A. 2004;101:8705–8.

    CAS  PubMed Central  PubMed  Google Scholar 

  78. Wynn D, et al. Daclizumab in active relapsing multiple sclerosis (CHOICE study): a phase 2, randomised, double-blind, placebo-controlled, add-on trial with interferon beta. Lancet Neurol. 2010;9:381–90.

    CAS  PubMed  Google Scholar 

  79. Hauser SL, et al. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358:676–88.

    CAS  PubMed  Google Scholar 

  80. Carson KR, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood. 2009;113:4834–40.

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Coles AJ, et al. The window of therapeutic opportunity in multiple sclerosis: evidence from monoclonal antibody therapy. J Neurol. 2006;253:98–108.

    PubMed  Google Scholar 

  82. Coles AJ, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359:1786–801.

    PubMed  Google Scholar 

  83. Jones JL, et al. IL-21 drives secondary autoimmunity in patients with multiple sclerosis, following therapeutic lymphocyte depletion with alemtuzumab (Campath-1H). J Clin Invest. 2009;119: 2052–61.

    CAS  PubMed Central  PubMed  Google Scholar 

  84. Romine JS, Sipe JC, Koziol JA, Zyroff J, Beutler E. A double-blind, placebo-controlled, randomized trial of cladribine in relapsing-remitting multiple sclerosis. Proc Assoc Am Physicians. 1999;111:35–44.

    CAS  PubMed  Google Scholar 

  85. O’Connor P, et al. Oral fingolimod (FTY720) in multiple sclerosis: two-year results of a phase II extension study. Neurology. 2009;72:73–9.

    PubMed  Google Scholar 

  86. Kappos L, et al. Oral fingolimod (FTY720) for relapsing multiple sclerosis. N Engl J Med. 2006;355:1124–40.

    CAS  PubMed  Google Scholar 

  87. Kappos L, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362:387–401.

    CAS  PubMed  Google Scholar 

  88. Comi G, et al. Phase II study of oral fingolimod (FTY720) in multiple sclerosis: 3-year results. Mult Scler. 2010;16:197–207.

    CAS  PubMed  Google Scholar 

  89. Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell. 2008; 132:598–611.

    CAS  PubMed  Google Scholar 

  90. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22.

    CAS  PubMed  Google Scholar 

  91. Zappia E, et al. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood. 2005;106:1755–61.

    CAS  PubMed  Google Scholar 

  92. Uccelli A, Zappia E, Benvenuto F, Frassoni F, Mancardi G. Stem cells in inflammatory demyelinating disorders: a dual role for immunosuppression and neuroprotection. Expert Opin Biol Ther. 2006;6:17–22.

    CAS  PubMed  Google Scholar 

  93. Bochev I, et al. Mesenchymal stem cells from human bone marrow or adipose tissue differently modulate mitogen-stimulated B-cell immunoglobulin production in vitro. Cell Biol Int. 2008; 32:384–93.

    CAS  PubMed  Google Scholar 

  94. Kassis I, et al. Neuroprotection and immunomodulation with mesenchymal stem cells in chronic experimental autoimmune encephalomyelitis. Arch Neurol. 2008;65:753–61.

    PubMed  Google Scholar 

  95. 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;105:215–28.

    CAS  PubMed  Google Scholar 

  96. Freedman MS, et al. The therapeutic potential of mesenchymal stem cell transplantation as a treatment for multiple sclerosis: consensus report of the International MSCT Study Group. Mult Scler. 2010;16:503–10.

    PubMed  Google Scholar 

  97. Ilancheran S, Moodley Y, Manuelpillai U. Human fetal membranes: a source of stem cells for tissue regeneration and repair? Placenta. 2009;30:2–10.

    CAS  PubMed  Google Scholar 

  98. Murphy S et al. Amnion epithelial cell isolation and characterization for clinical use. Curr Protoc Stem Cell Biol. 2010;Chapter 1:Unit 1E 6.

    Google Scholar 

  99. Ilancheran S, et al. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biol Reprod. 2007;77:577–88.

    CAS  PubMed  Google Scholar 

  100. Diaz-Prado S, et al. Multilineage differentiation potential of cells isolated from the human amniotic membrane. J Cell Biochem. 2010;111:846–57.

    CAS  PubMed  Google Scholar 

  101. Gerdoni E, et al. Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol. 2007;61:219–27.

    CAS  PubMed  Google Scholar 

  102. Uccelli A, Prockop DJ. Why should mesenchymal stem cells (MSCs) cure autoimmune diseases? Curr Opin Immunol. 2010; 22:768–74.

    CAS  PubMed  Google Scholar 

  103. Li H, et al. Immunosuppressive factors secreted by human amniotic epithelial cells. Invest Ophthalmol Vis Sci. 2005;46: 900–7.

    PubMed  Google Scholar 

  104. Moon JH, et al. Successful vitrification of human amnion-derived mesenchymal stem cells. Hum Reprod. 2008;23:1760–70.

    PubMed  Google Scholar 

  105. Irony-Tur-Sinai M, et al. Immunomodulation of EAE by alpha-fetoprotein involves elevation of immune cell apoptosis markers and the transcription factor FoxP3. J Neurol Sci. 2009;279:80–7.

    CAS  PubMed  Google Scholar 

  106. 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;154:5665–74.

    CAS  PubMed  Google Scholar 

  107. Kovats S, et al. A class I antigen, HLA-G, expressed in human trophoblasts. Science. 1990;248:220–3.

    CAS  PubMed  Google Scholar 

  108. Banas RA, et al. Immunogenicity and immunomodulatory effects of amnion-derived multipotent progenitor cells. Hum Immunol. 2008;69:321–8.

    CAS  PubMed  Google Scholar 

  109. McDonald C, Siatskas C, Bernard CCA. The emergence of amnion epithelial stem cells for the treatment of Multiple Sclerosis. Inflamm Regen. 2011;31:256–71.

    CAS  Google Scholar 

  110. Wolbank S, 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;13:1173–83.

    CAS  PubMed  Google Scholar 

  111. Murphy S, et al. Human amnion epithelial cells prevent bleomycin-induced lung injury and preserve lung function. Cell Transplant. 2011;20(6):909–23.

    PubMed  Google Scholar 

  112. Murphy SV, et al. Human amnion epithelial cells do not abrogate pulmonary fibrosis in mice with impaired macrophage function. Cell Transplant. 2012;21:1477–92.

    PubMed  Google Scholar 

  113. Tan JL, Chan ST, Wallace EM, Lim R. Human amnion epithelial cells mediate lung repair by directly modulating macrophage recruitment and polarization. Cell Transplant. 2013;23(3): 319–28.

    PubMed  Google Scholar 

  114. Manuelpillai U, et al. Human amniotic epithelial cell transplantation induces markers of alternative macrophage activation and reduces established hepatic fibrosis. PLoS One. 2012;7:e38631.

    CAS  PubMed Central  PubMed  Google Scholar 

  115. Vogel DY, et al. Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status. J Neuroinflammation. 2013;10:35.

    CAS  PubMed Central  PubMed  Google Scholar 

  116. Liu YH, et al. Amniotic epithelial cells from the human placenta potently suppress a mouse model of multiple sclerosis. PLoS One. 2012;7:e35758.

    CAS  PubMed Central  PubMed  Google Scholar 

  117. Sankar V, Muthusamy R. Role of human amniotic epithelial cell transplantation in spinal cord injury repair research. Neuroscience. 2003;118:11–7.

    CAS  PubMed  Google Scholar 

  118. Dong W, Chen H, Yang X, Guo L, Hui G. Treatment of intracerebral haemorrhage in rats with intraventricular transplantation of human amniotic epithelial cells. Cell Biol Int. 2010;34:573–7.

    PubMed  Google Scholar 

  119. Vosdoganes P, et al. Human amnion epithelial cells as a treatment for inflammation-induced fetal lung injury in sheep. Am J Obstet Gynecol. 2011;205:156 e126–133.

    Google Scholar 

  120. Yawno T, et al. Human amnion epithelial cells reduce fetal brain injury in response to intrauterine inflammation. Dev Neurosci. 2013;35(2–3):272–82.

    CAS  PubMed  Google Scholar 

  121. Lopez-Diego RS, Weiner HL. Novel therapeutic strategies for multiple sclerosis—a multifaceted adversary. Nat Rev Drug Discov. 2008;7:909–25.

    CAS  PubMed  Google Scholar 

  122. Miki T, Lehmann T, Cai H, Stolz DB, Strom SC. Stem cell characteristics of amniotic epithelial cells. Stem Cells. 2005;23: 1549–59.

    CAS  PubMed  Google Scholar 

  123. Kong XY, et al. Transplantation of human amniotic cells exerts neuroprotection in MPTP-induced Parkinson disease mice. Brain Res. 2008;1205:108–15.

    CAS  PubMed  Google Scholar 

  124. Scaggiante B, et al. [Graft of cryopreserved human amniotic epithelial cells in a subject with type B Niemann-Pick disease]. Pediatr Med Chir. 1987;9:89–92.

    CAS  PubMed  Google Scholar 

  125. Liu T. Human amniotic epithelial cells ameliorate behavioral dysfunction and reduce infarct size in the rat middle cerebral artery occlusion model. Shock. 2008;29:603–11.

    PubMed  Google Scholar 

  126. Yang XX, Xue SR, Dong WL, Kong Y. Therapeutic effect of human amniotic epithelial cell transplantation into the lateral ventricle of hemiparkinsonian rats. Chin Med J (Engl). 2009;122:2449–54.

    CAS  Google Scholar 

  127. Okawa H, Okuda O, Arai H, Sakuragawa N, Sato K. Amniotic epithelial cells transform into neuron-like cells in the ischemic brain. Neuroreport. 2001;12:4003–7.

    CAS  PubMed  Google Scholar 

  128. Xue H, et al. Development of a chemically extracted acellular muscle scaffold seeded with amniotic epithelial cells to promote spinal cord repair. J Biomed Mater Res A. 2013;101:145–56.

    PubMed  Google Scholar 

  129. Kakishita K, Elwan MA, Nakao N, Itakura T, Sakuragawa N. Human amniotic epithelial cells produce dopamine and survive after implantation into the striatum of a rat model of Parkinson’s disease: a potential source of donor for transplantation therapy. Exp Neurol. 2000;165:27–34.

    CAS  PubMed  Google Scholar 

  130. Wu ZY, Hui GZ, Lu Y, Wu X, Guo LH. Transplantation of human amniotic epithelial cells improves hindlimb function in rats with spinal cord injury. Chin Med J (Engl). 2006;119:2101–7.

    Google Scholar 

  131. X-t M, et al. Co-transplantation of bFGF-expressing amniotic epithelial cells and neural stem cells promotes functional recovery in spinal cord-injured rats. Cell Biol Int. 2008;32:1546–58.

    Google Scholar 

  132. Uchida S, et al. Neurotrophic function of conditioned medium from human amniotic epithelial cells. J Neurosci Res. 2000;62: 585–90.

    CAS  PubMed  Google Scholar 

  133. Meng XT, Chen D, Dong ZY, Liu JM. Enhanced neural differentiation of neural stem cells and neurite growth by amniotic epithelial cell co-culture. Cell Biol Int. 2007;31:691–8.

    CAS  PubMed  Google Scholar 

  134. Sakuragawa N, Elwan MA, Uchida S, Fujii T, Kawashima K. Non-neuronal neurotransmitters and neurotrophic factors in amniotic epithelial cells: expression and function in humans and monkey. Jpn J Pharmacol. 2001;85:20–3.

    CAS  PubMed  Google Scholar 

  135. Venkatachalam S, et al. Novel neurotrophic factor secreted by amniotic epithelial cells. Biocell. 2009;33:81–9.

    CAS  PubMed  Google Scholar 

  136. Yang S, et al. Conditioned medium from human amniotic epithelial cells may induce the differentiation of human umbilical cord blood mesenchymal stem cells into dopaminergic neuron-like cells. J Neurosci Res. 2013;91:978–86.

    CAS  PubMed  Google Scholar 

  137. McClain CR, Sim FJ, Goldman SA. Pleiotrophin suppression of receptor protein tyrosine phosphatase-beta/zeta maintains the self-renewal competence of fetal human oligodendrocyte progenitor cells. J Neurosci. 2012;32:15066–75.

    CAS  PubMed Central  PubMed  Google Scholar 

  138. Taravini IR, et al. Pleiotrophin over-expression provides trophic support to dopaminergic neurons in parkinsonian rats. Mol Neurodegener. 2011;6:40.

    PubMed Central  PubMed  Google Scholar 

  139. Elwan MA, Sakuragawa N. Evidence for synthesis and release of catecholamines by human amniotic epithelial cells. Neuroreport. 1997;8:3435–8.

    CAS  PubMed  Google Scholar 

  140. Bankiewicz KS, Palmatier M, Plunkett RJ, Cummins A, Oldfield EH. Reversal of hemiparkinsonian syndrome in nonhuman primates by amnion implantation into caudate nucleus. J Neurosurg. 1994;81:869–76.

    CAS  PubMed  Google Scholar 

  141. Kakishita K, Nakao N, Sakuragawa N, Itakura T. Implantation of human amniotic epithelial cells prevents the degeneration of nigral dopamine neurons in rats with 6-hydroxydopamine lesions. Brain Res. 2003;980:48–56.

    CAS  PubMed  Google Scholar 

  142. Noh JS, et al. Neurotoxic and neuroprotective actions of catecholamines in cortical neurons. Exp Neurol. 1999;159:217–24.

    CAS  PubMed  Google Scholar 

  143. Takashima SS. Human amniotic epithelial cells possess hepatocyte-like characteristics and functions. Cell Struct Funct. 2004;29:73.

    CAS  PubMed  Google Scholar 

  144. 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.

    PubMed  Google Scholar 

  145. Zemba M, et al. Amniotic membrane transplantation in palliative treatment of bullous keratopathy. Oftalmologia. 2006;50:51–3.

    CAS  PubMed  Google Scholar 

  146. 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;20:604–7.

    PubMed  Google Scholar 

  147. 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;36:876–82.

    CAS  PubMed  Google Scholar 

  148. Wang M, et al. Immunogenicity and antigenicity of allogeneic amniotic epithelial transplants grafted to the cornea, conjunctiva, and anterior chamber. Invest Ophthalmol Vis Sci. 2006;47: 1522–32.

    PubMed  Google Scholar 

  149. Gabler B, Lohmann CP. Hypopyon after repeated transplantation of human amniotic membrane onto the corneal surface. Ophthalmology. 2000;107:1344–6.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Courtney A. McDonald B.Sc. (Hons.), Ph.D. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this chapter

Cite this chapter

McDonald, C.A., Short, M., Jenkin, G., Bernard, C.C.A. (2014). The Potential of Human Amnion Epithelial Cells as an Immunomodulatory and Neuroregenerative Treatment for Multiple Sclerosis. In: Atala, A., Murphy, S. (eds) Perinatal Stem Cells. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-1118-9_21

Download citation

Publish with us

Policies and ethics