J Clin Neurol.  2009 Mar;5(1):1-10.

Bone Marrow-Derived Mesenchymal Stem Cell Therapy as a Candidate Disease-Modifying Strategy in Parkinson's Disease and Multiple System Atrophy

Affiliations
  • 1Department of Neurology, Yonsei University College of Medicine, Seoul, Korea. phlee@yuhs.ac
  • 2Center for Neuroregeneration and Stem Cell Research, Ajou University College of Medicine, Suwon, Korea.

Abstract

Parkinson's disease (PD) and multiple system atrophy (MSA) are neurodegenerative diseases representative of alpha-synucleinopathies characterized pathologically by alpha-synuclein-abundant Lewy bodies and glial cytoplasmic inclusions, respectively. Embryonic stem cells, fetal mesencephalic neurons, and neural stem cells have been introduced as restorative strategies in PD animals and patients, but ethical and immunological problems as well as the serious side effects of tumorigenesis and disabling dyskinesia have limited clinical application of these stem cells. Meanwhile, cell therapy using mesenchymal stem cells (MSCs) is attractive clinically because these cells are free from ethical and immunological problems. MSCs are present in adult bone marrow and represent <0.01% of all nucleated bone marrow cells. MSCs are themselves capable of multipotency, differentiating under appropriate conditions into chondrocytes, skeletal myocytes, and neurons. According to recent studies, the neuroprotective effect of MSCs is mediated by their ability to produce various trophic factors that contribute to functional recovery, neuronal cell survival, and stimulation of endogenous regeneration and by immunoregulatory properties that not only inhibit nearly all cells participating in the immune response cell-cell-contact-dependent mechanism, but also release various soluble factors associated with immunosuppressive activity. However, the use of MSCs as neuroprotectives in PD and MSA has seldom been studied. Here we comprehensively review recent advances in the therapeutic roles of MSCs in PD and MSA, especially focusing on their neuroprotective properties and use in disease-modifying therapeutic strategies.

Keyword

mesenchymal stem cells; cell therapy; Parkinson's disease; multiple system atrophy; neuroprotection

MeSH Terms

Adult
Animals
Bone Marrow
Bone Marrow Cells
Cell Survival
Cell Transformation, Neoplastic
Chondrocytes
Dyskinesias
Embryonic Stem Cells
Humans
Inclusion Bodies
Lewy Bodies
Mesenchymal Stromal Cells
Multiple System Atrophy
Muscle Fibers, Skeletal
Neural Stem Cells
Neurodegenerative Diseases
Neurons
Neuroprotective Agents
Parkinson Disease
Regeneration
Stem Cells
Tissue Therapy
Neuroprotective Agents

Figure

  • Fig. 1 Effects of cell therapy with human mesenchymal stem cells (hMSCs) on animals treated with MG-132. Immunohistochemical analysis showed that hMSC treatment dramatically reduced the decline in the number of TH-ir cells in the SN of MG-132-treated rats (A). Stereological analysis revealed that the number of TH-ir cells was significantly higher in the hMSC-treatment group than in the group treated with MG-132 alone (n=5; p<0.05, B). Dopamine levels in the striatum (as assessed by gas chromatography-mass spectrometry) were significantly lower in MG-132-treated rats than in controls (p<0.01); however, hMSC treatment significantly increased the dopamine level in the striatum of MG-132-treated rats (n=5; p<0.05, C). MG-132 treatment resulted in the accumulation of polyubiquitinated proteins and a markedly increase in OX-6 immunoreactivity; however, hMSC treatment markedly decreased the accumulation of polyubiquitinated proteins and OX-6 immunoreactivity in MG-132-treated rats (D and E). The level of the cleaved form of caspase-3 was significantly lower in rats treated with hMSCs (F) than in MG-132-treated rats (n=3, G). Scale bar: 100 µm. *p<0.05, **p<0.01. SN: substantia nigra, TH-ir: tyrosine-hydroxylase-immunoreactive.

  • Fig. 2 Coculturing hMSCs with LPS-stimulated microglia in a Transwell culture chamber system decreased microglial activation and increased the expressions of IL-6, IL-10, and TGF-β. To identify soluble factors associated with modulation of microglial activation, we analyzed the expressions of IL-6, IL-10, and TGF-β in hMSCs cocultured with LPS-stimulated microglia and in hMSCs alone. The inclusion of hMSCs significantly decreased the number of process-bearing activated microglia at 6 and 24 h following hMSC treatment (A). When hMSCs were cocultured with LPS-stimulated microglia, IL-6 expression was significantly increased at 3 and 12 h, and the expressions of IL-10 and TGF-β at 12 h were significantly higher than those with hMSCs alone (B). Immunohistological evaluation of protective effect of hMSCs against LPS-induced damage to dopaminergic neurons in the SN. hMSC treatment considerably reduced the loss of TH-ir cells and microglial activation induced by LPS stimulation in the SN (C, Scale bar: 100 mm). Stereological analysis revealed that hMSC treatment significantly decreased the loss of TH-ir cells at 7 and 14 days following LPS stimulation (D, *p<0.05). The administration of hMSCs significantly down-regulated the LPS-induced increase in the expressions of TNF-α and iNOS mRNA at 3 days after LPS stimulation (E). hMSCs: human mesenchymal stem cells, LPS: lipopolysaccharide, IL: interleukin, TGF-β: transforming growth factor β, SN: substantia nigra, TNF-α: tumor necrosis factor-α.

  • Fig. 3 Changes from baseline scores (mean and SE values) on the Unified Multiple System Atrophy Rating Scale (UMSARS) for MSC-treated and control patients throughout the 12 months of follow-up (A). UMSARS I analysis between MSC-treated and control patients (B: black squares=MSC-treated patients; gray triangles=control patients). The improvement on the UMSARS was significantly greater in the MSC group than in the control group at all visits throughout the 12-month study period. Cerebral glucose metabolism in the MSC-treated patients was higher in the follow-up scan than in the initial scan in the cerebellum and white matter (C, red color), whereas in the control group it was significantly lower in the follow-up scan than in the initial scan in the cerebellum and brainstem (D, blue color). MSC: mesenchymal stem cell.


Reference

1. Moore DJ, West AB, Dawson VL, Dawson TM. Molecular pathophysiology of Parkinson's disease. Annu Rev Neurosci. 2005. 28:57–87.
Article
2. von Bohlen und Halbach O, Schober A, Krieglstein K. Genes, proteins, and neurotoxins involved in Parkinson's disease. Prog Neurobiol. 2004. 73:151–177.
Article
3. Adler CH. Nonmotor complications in Parkinson's disease. Mov Disord. 2005. 20:Suppl 11. S23–S29.
Article
4. Palmer MR, Granholm AC, van Horne CG, Giardina KE, Freund RK, Moorhead JW, et al. Intranigral transplantation of solid tissue ventral mesencephalon or striatal grafts induces behavioral recovery in 6-OHDA-lesioned rats. Brain Res. 2001. 890:86–99.
Article
5. Lindvall O. Stem cells for cell therapy in Parkinson's disease. Pharmacol Res. 2003. 47:279–287.
Article
6. Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W, Kao R, et al. Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N Engl J Med. 2001. 344:710–719.
Article
7. Olanow CW, Goetz CG, Kordower JH, Stoessl AJ, Sossi V, Brin MF, et al. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease. Ann Neurol. 2003. 54:403–414.
Article
8. Poewe W. Non-motor symptoms in Parkinson's disease. Eur J Neurol. 2008. 15 Suppl 1:14–20.
Article
9. Braak H, Del Tredici K. Invited Article: Nervous system pathology in sporadic Parkinson disease. Neurology. 2008. 70:1916–1925.
Article
10. Braak H, Ghebremedhin E, Rüb U, Bratzke H, Del Tredici K. Stages in the development of Parkinson's disease-related pathology. Cell Tissue Res. 2004. 318:121–134.
Article
11. Lee PH, Yeo SH, Kim HJ, Youm HY. Correlation between cardiac 123I-MIBG and odor identification in patients with Parkinson's disease and multiple system atrophy. Mov Disord. 2006. 21:1975–1977.
Article
12. Correia AS, Anisimov SV, Li JY, Brundin P. Stem cell-based therapy for Parkinson's disease. Ann Med. 2005. 37:487–498.
Article
13. Hagell P, Piccini P, Björklund A, Brundin P, Rehncrona S, Widner H, et al. Dyskinesias following neural transplantation in Parkinson's disease. Nat Neurosci. 2002. 5:627–628.
Article
14. Björklund A, Dunnett SB, Brundin P, Stoessl AJ, Freed CR, Breeze RE, et al. Neural transplantation for the treatment of Parkinson's disease. Lancet Neurol. 2003. 2:437–445.
Article
15. Winkler C, Kirik D, Björklund A. Cell transplantation in Parkinson's disease: how can we make it work? Trends Neurosci. 2005. 28:86–92.
Article
16. Isacson O, Bjorklund LM, Schumacher JM. Toward full restoration of synaptic and terminal function of the dopaminergic system in Parkinson's disease by stem cells. Ann Neurol. 2003. 53:Suppl 3. S135–S146. discussion S146-S148.
Article
17. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat Med. 2008. 14:504–506.
Article
18. Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. 2008. 14:501–503.
Article
19. Mendez I, Viñuela A, Astradsson A, Mukhida K, Hallett P, Robertson H, et al. Dopamine neurons implanted into people with Parkinson's disease survive without pathology for 14 years. Nat Med. 2008. 14:507–509.
Article
20. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999. 284:143–147.
Article
21. Woodbury D, Schwarz EJ, Prockop DJ, Black IB. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res. 2000. 61:364–370.
Article
22. Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med (Maywood). 2001. 226:507–520.
Article
23. Bonuccelli U, Del Dotto P. New pharmacologic horizons in the treatment of Parkinson disease. Neurology. 2006. 67:S30–S38.
Article
24. Li Y, Chen J, Chen XG, Wang L, Gautam SC, Xu YX, et al. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology. 2002. 59:514–523.
Article
25. Mahmood A, Lu D, Chopp M. Marrow stromal cell transplantation after traumatic brain injury promotes cellular proliferation within the brain. Neurosurgery. 2004. 55:1185–1193.
Article
26. Crigler L, Robey RC, Asawachaicharn A, Gaupp D, Phinney DG. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol. 2006. 198:54–64.
Article
27. Arnhold S, Klein H, Klinz FJ, Absenger Y, Schmidt A, Schinköthe T, et al. Human bone marrow stroma cells display certain neural characteristics and integrate in the subventricular compartment after injection into the liquor system. Eur J Cell Biol. 2006. 85:551–565.
Article
28. Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004. 36:568–584.
Article
29. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology. 1988. 38:1285–1291.
Article
30. Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA, Karluk D. Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure. Ann Neurol. 1999. 46:598–605.
Article
31. Ouchi Y, Yoshikawa E, Sekine Y, Futatsubashi M, Kanno T, Ogusu T, et al. Microglial activation and dopamine terminal loss in early Parkinson's disease. Ann Neurol. 2005. 57:168–175.
Article
32. Hunot S, Dugas N, Faucheux B, Hartmann A, Tardieu M, Debré P, et al. FcepsilonRII/CD23 is expressed in Parkinson's disease and induces, in vitro, production of nitric oxide and tumor necrosis factor-alpha in glial cells. J Neurosci. 1999. 19:3440–3447.
Article
33. Nagatsu T, Mogi M, Ichinose H, Togari A. Cytokines in Parkinson's disease. J Neural Transm Suppl. 2000. 143–151.
Article
34. Gao HM, Hong JS, Zhang W, Liu B. Distinct role for microglia in rotenone-induced degeneration of dopaminergic neurons. J Neurosci. 2002. 22:782–790.
Article
35. Cicchetti F, Brownell AL, Williams K, Chen YI, Livni E, Isacson O. Neuroinflammation of the nigrostriatal pathway during progressive 6-OHDA dopamine degeneration in rats monitored by immunohistochemistry and PET imaging. Eur J Neurosci. 2002. 15:991–998.
Article
36. Liberatore GT, Jackson-Lewis V, Vukosavic S, Mandir AS, Vila M, McAuliffe WG, et al. Inducible nitric oxide synthase stimulates dopaminergic neurodegeneration in the MPTP model of Parkinson disease. Nat Med. 1999. 5:1403–1409.
Article
37. Krampera M, Pasini A, Pizzolo G, Cosmi L, Romagnani S, Annunziato F. Regenerative and immunomodulatory potential of mesenchymal stem cells. Curr Opin Pharmacol. 2006. 6:435–441.
Article
38. Karussis D, Kassis I, Kurkalli BG, Slavin S. Immunomodulation and neuroprotection with mesenchymal bone marrow stem cells (MSCs): a proposed treatment for multiple sclerosis and other neuroimmunological/neurodegenerative diseases. J Neurol Sci. 2008. 265:131–135.
Article
39. Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal stromal cells. Blood. 2007. 110:3499–3506.
Article
40. Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E, et al. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood. 2005. 106:1755–1761.
Article
41. Gerdoni E, Gallo B, Casazza S, Musio S, Bonanni I, Pedemonte E, et al. Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol. 2007. 61:219–227.
Article
42. Guo J, Lin GS, Bao CY, Hu ZM, Hu MY. Anti-inflammation role for mesenchymal stem cells transplantation in myocardial infarction. Inflammation. 2007. 30:97–104.
Article
43. Aubin N, Curet O, Deffois A, Carter C. Aspirin and salicylate protect against MPTP-induced dopamine depletion in mice. J Neurochem. 1998. 71:1635–1642.
Article
44. Teismann P, Tieu K, Choi DK, Wu DC, Naini A, Hunot S, et al. Cyclooxygenase-2 is instrumental in Parkinson's disease neurodegeneration. Proc Natl Acad Sci U S A. 2003. 100:5473–5478.
Article
45. Chen H, Zhang SM, Hernán MA, Schwarzschild MA, Willett WC, Colditz GA, et al. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Arch Neurol. 2003. 60:1059–1064.
Article
46. Wahner AD, Bronstein JM, Bordelon YM, Ritz B. Nonsteroidal anti-inflammatory drugs may protect against Parkinson disease. Neurology. 2007. 69:1836–1842.
Article
47. Chen J, Li Y, Katakowski M, Chen X, Wang L, Lu D, et al. Intravenous bone marrow stromal cell therapy reduces apoptosis and promotes endogenous cell proliferation after stroke in female rat. J Neurosci Res. 2003. 73:778–786.
Article
48. Li Y, Chen J, Wang L, Zhang L, Lu M, Chopp M. Intracerebral transplantation of bone marrow stromal cells in a 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine mouse model of Parkinson's disease. Neurosci Lett. 2001. 316:67–70.
Article
49. Stumm RK, Rummel J, Junker V, Culmsee C, Pfeiffer M, Krieglstein J, et al. A dual role for the SDF-1/CXCR4 chemokine receptor system in adult brain: isoform-selective regulation of SDF-1 expression modulates CXCR4-dependent neuronal plasticity and cerebral leukocyte recruitment after focal ischemia. J Neurosci. 2002. 22:5865–5878.
Article
50. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007. 25:2739–2749.
Article
51. Banisadr G, Skrzydelski D, Kitabgi P, Rosténe W, Parsadaniantz SM. Highly regionalized distribution of stromal cell-derived factor-1/CXCL12 in adult rat brain: constitutive expression in cholinergic, dopaminergic and vasopressinergic neurons. Eur J Neurosci. 2003. 18:1593–1606.
Article
52. Emborg ME. Evaluation of animal models of Parkinson's disease for neuroprotective strategies. J Neurosci Methods. 2004. 139:121–143.
Article
53. McNaught KS, Perl DP, Brownell AL, Olanow CW. Systemic exposure to proteasome inhibitors causes a progressive model of Parkinson's disease. Ann Neurol. 2004. 56:149–162.
Article
54. Schapira AH, Cleeter MW, Muddle JR, Workman JM, Cooper JM, King RH. Proteasomal inhibition causes loss of nigral tyrosine hydroxylase neurons. Ann Neurol. 2006. 60:253–255.
Article
55. Zeng BY, Bukhatwa S, Hikima A, Rose S, Jenner P. Reproducible nigral cell loss after systemic proteasomal inhibitor administration to rats. Ann Neurol. 2006. 60:248–252.
Article
56. Bové J, Zhou C, Jackson-Lewis V, Taylor J, Chu Y, Rideout HJ, et al. Proteasome inhibition and Parkinson's disease modeling. Ann Neurol. 2006. 60:260–264.
Article
57. Kordower JH, Kanaan NM, Chu Y, Suresh Babu R, Stansell J 3rd, Terpstra BT, et al. Failure of proteasome inhibitor administration to provide a model of Parkinson's disease in rats and monkeys. Ann Neurol. 2006. 60:264–268.
Article
58. Manning-Boğ AB, Reaney SH, Chou VP, Johnston LC, McCormack AL, Johnston J, et al. Lack of nigrostriatal pathology in a rat model of proteasome inhibition. Ann Neurol. 2006. 60:256–260.
Article
59. Park HJ, Lee PH, Bang OY, Lee G, Ahn YH. Mesenchymal stem cells therapy exerts neuroprotection in a progressive animal model of Parkinson's disease. J Neurochem. 2008. 107:141–151.
Article
60. Tatton WG, Chalmers-Redman R, Brown D, Tatton N. Apoptosis in Parkinson's disease: signals for neuronal degradation. Ann Neurol. 2003. 53:Suppl 3. S61–S70. discussion S70-S72.
Article
61. Olanow CW, McNaught KS. Ubiquitin-proteasome system and Parkinson's disease. Mov Disord. 2006. 21:1806–1823.
Article
62. Kim YJ, Park HJ, Lee G, Bang OY, Ahn YH, Joe E, et al. Neuroprotective effects of human mesenchymal stem cells on dopaminergic neurons through anti-inflammatory action. Glia. 2009. 57:13–23.
Article
63. Mareschi K, Novara M, Rustichelli D, Ferrero I, Guido D, Carbone E, et al. Neural differentiation of human mesenchymal stem cells: Evidence for expression of neural markers and eag K+ channel types. Exp Hematol. 2006. 34:1563–1572.
Article
64. Blondheim NR, Levy YS, Ben-Zur T, Burshtein A, Cherlow T, Kan I, et al. Human mesenchymal stem cells express neural genes, suggesting a neural predisposition. Stem Cells Dev. 2006. 15:141–164.
Article
65. Barzilay R, Kan I, Ben-Zur T, Bulvik S, Melamed E, Offen D. Induction of human mesenchymal stem cells into dopamine-producing cells with different differentiation protocols. Stem Cells Dev. 2008. 17:547–554.
Article
66. Offen D, Barhum Y, Levy YS, Burshtein A, Panet H, Cherlow T, et al. Intrastriatal transplantation of mouse bone marrow-derived stem cells improves motor behavior in a mouse model of Parkinson's disease. J Neural Transm Suppl. 2007. 133–143.
Article
67. Ye M, Wang XJ, Zhang YH, Lu GQ, Liang L, Xu JY, et al. Therapeutic effects of differentiated bone marrow stromal cell transplantation on rat models of Parkinson's disease. Parkinsonism Relat Disord. 2007. 13:44–49.
Article
68. Wenning GK, Colosimo C, Geser F, Poewe W. Multiple system atrophy. Lancet Neurol. 2004. 3:93–103.
Article
69. Lee PH, Kim JW, Bang OY, Ahn YH, Joo IS, Huh K. Autologous mesenchymal stem cell therapy delays the progression of neurological deficits in patients with multiple system atrophy. Clin Pharmacol Ther. 2008. 83:723–730.
Article
70. Quinn N, Barker RA, Wenning GK. Are trials of intravascular infusions of autologous mesenchymal stem cells in patients with multiple system atrophy currently justified, and are they effective? Clin Pharmacol Ther. 2008. 83:663–665.
Article
71. Whone AL, Scolding NJ. Mesenchymal stem cells and neurodegenerative disease. Clin Pharmacol Ther. 2009. 85:19–20.
Article
Full Text Links
  • JCN
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