Skip to main content

Pathology and Cell-Based Therapy of Parkinson’s Disease

  • Chapter
  • First Online:
Pathology, Prevention and Therapeutics of Neurodegenerative Disease

Abstract

Parkinson’s disease (PD) comprises an age-related and the second most common disorders which is characterized by progressive motor symptoms such as bradykinesia, rigidity, akinesia, abnormal posture, and resting tremor. The pathology of PD involves the loss of dopaminergic (DA) neurons from the substantia nigra and the production α-synuclein-containing Lewy bodies and Lewy neurites. Current pharmacological treatments for PD target early symptoms by supplying dopamine precursors. These treatments regrettably have long-term side-effects, whereas stem cells-based therapies demonstrate safety and efficiency in preclinical studies through enhancement of dopamine uptake and motor symptoms. Therefore, stem cell transplantation shows great promise as a method to replace lost DA neurons; and for acquiring a better understanding of these neurodegenerative diseases mechanisms. In this review, we discuss recent preclinical studies of stem cell-based therapy for PD and important areas of future research.

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

Access this chapter

eBook
USD 16.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
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

Similar content being viewed by others

References

  1. Batista CE, Mariano ED, Marie SK, Teixeira MJ, Morgalla M, Tatagiba M, Li J, Lepski G. Stem cells in neurology—current perspectives. Arq Neuropsiquiatr. 2014;72:457–65.

    Article  Google Scholar 

  2. Trenkwalder C, Schwarz J, Gebhard J, Ruland D, Trenkwalder P, Hense HW, Oertel WH. Starnberg trial on epidemiology of Parkinsonism and hypertension in the elderly. Prevalence of Parkinson’s disease and related disorders assessed by a door-to-door survey of inhabitants older than 65 years. Arch Neurol. 1995;52:1017–22.

    Article  CAS  Google Scholar 

  3. von Campenhausen S, Bornschein B, Wick R, Botzel K, Sampaio C, Poewe W, Oertel W, Siebert U, Berger K, Dodel R. Prevalence and incidence of Parkinson’s disease in Europe. Eur Neuropsychopharmacol. 2005;15:473–90.

    Article  Google Scholar 

  4. Buttery PC, Barker RA. Treating Parkinson’s disease in the 21st century: can stem cell transplantation compete? J Comp Neurol. 2014;522:2802–16.

    Article  CAS  Google Scholar 

  5. Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55:181–4.

    Article  CAS  Google Scholar 

  6. Fahn S. Description of Parkinson’s disease as a clinical syndrome. Ann N Y Acad Sci. 2003;991:1–14.

    Article  CAS  Google Scholar 

  7. Chaudhuri KR, Healy DG, Schapira AH, National Institute for Clinical Excellence. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol. 2006;5:235–45.

    Article  Google Scholar 

  8. Savitt JM, Dawson VL, Dawson TM. Diagnosis and treatment of Parkinson disease: molecules to medicine. J Clin Invest. 2006;116:1744–54.

    Article  CAS  Google Scholar 

  9. Poewe W. Treatments for Parkinson disease—past achievements and current clinical needs. Neurology. 2009;72:S65–73.

    Article  CAS  Google Scholar 

  10. Prashanth LK, Fox S, Meissner WG. l-Dopa-induced dyskinesia-clinical presentation, genetics, and treatment. Int Rev Neurobiol. 2011;98:31–54.

    Article  CAS  Google Scholar 

  11. Huot P, Johnston TH, Koprich JB, Fox SH, Brotchie JM. The pharmacology of L-DOPA-induced dyskinesia in Parkinson’s disease. Pharmacol Rev. 2013;65:171–222.

    Article  CAS  Google Scholar 

  12. Jenner P. Dopamine agonists, receptor selectivity and dyskinesia induction in Parkinson’s disease. Curr Opin Neurol. 2003;16(Suppl 1):S3–7.

    Article  CAS  Google Scholar 

  13. Barker RA, Barrett J, Mason SL, Bjorklund A. Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson’s disease. Lancet Neurol. 2013;12:84–91.

    Article  CAS  Google Scholar 

  14. Bjorklund A, Dunnett SB. Dopamine neuron systems in the brain: an update. Trends Neurosci. 2007;30:194–202.

    Article  Google Scholar 

  15. Kefalopoulou Z, Politis M, Piccini P, Mencacci N, Bhatia K, Jahanshahi M, Widner H, Rehncrona S, Brundin P, Bjorklund A, Lindvall O, Limousin P, Quinn N, Foltynie T. Long-term clinical outcome of fetal cell transplantation for Parkinson disease: two case reports. JAMA Neurol. 2014;71:83–7.

    Article  Google Scholar 

  16. Kordower JH, Freeman TB, Snow BJ, Vingerhoets FJ, Mufson EJ, Sanberg PR, Hauser RA, Smith DA, Nauert GM, Perl DP, et al. Neuropathological evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson’s disease. N Engl J Med. 1995;332:1118–24.

    Article  CAS  Google Scholar 

  17. Petit GH, Olsson TT, Brundin P. The future of cell therapies and brain repair: Parkinson’s disease leads the way. Neuropathol Appl Neurobiol. 2014;40:60–70.

    Article  CAS  Google Scholar 

  18. Lindvall O, Sawle G, Widner H, Rothwell JC, Bjorklund A, Brooks D, Brundin P, Frackowiak R, Marsden CD, Odin P, et al. Evidence for long-term survival and function of dopaminergic grafts in progressive Parkinson’s disease. Ann Neurol. 1994;35:172–80.

    Article  CAS  Google Scholar 

  19. Piccini P, Brooks DJ, Bjorklund A, Gunn RN, Grasby PM, Rimoldi O, Brundin P, Hagell P, Rehncrona S, Widner H, Lindvall O. Dopamine release from nigral transplants visualized in vivo in a Parkinson’s patient. Nat Neurosci. 1999;2:1137–40.

    Article  CAS  Google Scholar 

  20. 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;14:504–6.

    Article  CAS  Google Scholar 

  21. Gonzalez C, Bonilla S, Flores AI, Cano E, Liste I. An update on human stem cell-based therapy in Parkinson’s disease. Curr Stem Cell Res Ther. 2016;11:561–8.

    Article  CAS  Google Scholar 

  22. Allan LE, Petit GH, Brundin P. Cell transplantation in Parkinson’s disease: problems and perspectives. Curr Opin Neurol. 2010;23:426–32.

    PubMed  Google Scholar 

  23. Disterhoft JF, Thompson LT, Moyer JR Jr, Mogul DJ. Calcium-dependent after hyperpolarization and learning in young and aging hippocampus. Life Sci. 1996;59:413–20.

    Article  CAS  Google Scholar 

  24. Jagmag SA, Tripathi N, Shukla SD, Maiti S, Khurana S. Evaluation of models of Parkinson’s disease. Front Neurosci. 2015;9:503.

    PubMed  Google Scholar 

  25. Beach TG, Adler CH, Sue LI, Vedders L, Lue L, White Iii CL, Akiyama H, Caviness JN, Shill HA, Sabbagh MN, Walker DG, Arizona Parkinson’s Disease Consortium. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol. 2010;119:689–702.

    Article  CAS  Google Scholar 

  26. McGeer PL, McGeer EG. Glial reactions in Parkinson’s disease. Mov Disord. 2008;23:474–83.

    Article  Google Scholar 

  27. More SV, Kumar H, Kim IS, Song SY, Choi DK. Cellular and molecular mediators of neuroinflammation in the pathogenesis of Parkinson’s disease. Mediat Inflamm. 2013;2013:952375.

    Article  Google Scholar 

  28. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.

    Article  Google Scholar 

  29. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40.

    Article  CAS  Google Scholar 

  30. Klein C, Westenberger A. Genetics of Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2:a008888.

    Article  Google Scholar 

  31. Volpicelli-Daley LA, Gamble KL, Schultheiss CE, Riddle DM, West AB, Lee VM. Formation of alpha-synuclein Lewy neurite-like aggregates in axons impedes the transport of distinct endosomes. Mol Biol Cell. 2014;25:4010–23.

    Article  Google Scholar 

  32. Halliday GM, Holton JL, Revesz T, Dickson DW. Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011;122:187–204.

    Article  CAS  Google Scholar 

  33. Irwin DJ, Lee VM, Trojanowski JQ. Parkinson’s disease dementia: convergence of alpha-synuclein, tau and amyloid-beta pathologies. Nat Rev Neurosci. 2013;14:626–36.

    Article  CAS  Google Scholar 

  34. McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, Cummings J, Duda JE, Lippa C, Perry EK, Aarsland D, Arai H, Ballard CG, Boeve B, Burn DJ, Costa D, Del Ser T, Dubois B, Galasko D, Gauthier S, Goetz CG, Gomez-Tortosa E, Halliday G, Hansen LA, Hardy J, Iwatsubo T, Kalaria RN, Kaufer D, Kenny RA, Korczyn A, Kosaka K, Lee VM, Lees A, Litvan I, Londos E, Lopez OL, Minoshima S, Mizuno Y, Molina JA, Mukaetova-Ladinska EB, Pasquier F, Perry RH, Schulz JB, Trojanowski JQ, Yamada M, Consortium on DLB. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65:1863–72.

    Article  CAS  Google Scholar 

  35. Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM, Oostra BA, Heutink P. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science. 2003;299:256–9.

    Article  CAS  Google Scholar 

  36. Dawson TM, Dawson VL. The role of parkin in familial and sporadic Parkinson’s disease. Mov Disord. 2010;25(Suppl 1):S32–9.

    Article  Google Scholar 

  37. Smith DK, He M, Zhang CL, Zheng JC. The therapeutic potential of cell identity reprogramming for the treatment of aging-related neurodegenerative disorders. Prog Neurobiol. 2017;157:212–29.

    Article  CAS  Google Scholar 

  38. Abou-Sleiman PM, Muqit MM, Wood NW. Expanding insights of mitochondrial dysfunction in Parkinson’s disease. Nat Rev Neurosci. 2006;7:207–19.

    Article  CAS  Google Scholar 

  39. Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol. 2008;183:795–803.

    Article  CAS  Google Scholar 

  40. Palacino JJ, Sagi D, Goldberg MS, Krauss S, Motz C, Wacker M, Klose J, Shen J. Mitochondrial dysfunction and oxidative damage in parkin-deficient mice. J Biol Chem. 2004;279:18614–22.

    Article  CAS  Google Scholar 

  41. Wood-Kaczmar A, Gandhi S, Yao Z, Abramov AY, Miljan EA, Keen G, Stanyer L, Hargreaves I, Klupsch K, Deas E, Downward J, Mansfield L, Jat P, Taylor J, Heales S, Duchen MR, Latchman D, Tabrizi SJ, Wood NW. PINK1 is necessary for long term survival and mitochondrial function in human dopaminergic neurons. PLoS One. 2008;3:e2455.

    Article  Google Scholar 

  42. Exner N, Treske B, Paquet D, Holmstrom K, Schiesling C, Gispert S, Carballo-Carbajal I, Berg D, Hoepken HH, Gasser T, Kruger R, Winklhofer KF, Vogel F, Reichert AS, Auburger G, Kahle PJ, Schmid B, Haass C. Loss-of-function of human PINK1 results in mitochondrial pathology and can be rescued by parkin. J Neurosci. 2007;27:12413–8.

    Article  CAS  Google Scholar 

  43. Abramov AY, Gegg M, Grunewald A, Wood NW, Klein C, Schapira AH. Bioenergetic consequences of PINK1 mutations in Parkinson disease. PLoS One. 2011;6:e25622.

    Article  CAS  Google Scholar 

  44. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010;8:e1000298.

    Article  Google Scholar 

  45. Rakovic A, Grunewald A, Seibler P, Ramirez A, Kock N, Orolicki S, Lohmann K, Klein C. Effect of endogenous mutant and wild-type PINK1 on Parkin in fibroblasts from Parkinson disease patients. Hum Mol Genet. 2010;19:3124–37.

    Article  CAS  Google Scholar 

  46. Seibler P, Graziotto J, Jeong H, Simunovic F, Klein C, Krainc D. Mitochondrial Parkin recruitment is impaired in neurons derived from mutant PINK1 induced pluripotent stem cells. J Neurosci. 2011;31:5970–6.

    Article  CAS  Google Scholar 

  47. Bosgraaf L, Van Haastert PJ. Roc, a Ras/GTPase domain in complex proteins. Biochim Biophys Acta. 2003;1643:5–10.

    Article  CAS  Google Scholar 

  48. MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A. The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron. 2006;52:587–93.

    Article  CAS  Google Scholar 

  49. Smith WW, Pei Z, Jiang H, Moore DJ, Liang Y, West AB, Dawson VL, Dawson TM, Ross CA. Leucine-rich repeat kinase 2 (LRRK2) interacts with parkin, and mutant LRRK2 induces neuronal degeneration. Proc Natl Acad Sci U S A. 2005;102:18676–81.

    Article  CAS  Google Scholar 

  50. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Muller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601–7.

    Article  CAS  Google Scholar 

  51. Nguyen HN, Byers B, Cord B, Shcheglovitov A, Byrne J, Gujar P, Kee K, Schule B, Dolmetsch RE, Langston W, Palmer TD, Pera RR. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell. 2011;8:267–80.

    Article  CAS  Google Scholar 

  52. Reinhardt P, Schmid B, Burbulla LF, Schondorf DC, Wagner L, Glatza M, Hoing S, Hargus G, Heck SA, Dhingra A, Wu G, Muller S, Brockmann K, Kluba T, Maisel M, Kruger R, Berg D, Tsytsyura Y, Thiel CS, Psathaki OE, Klingauf J, Kuhlmann T, Klewin M, Muller H, Gasser T, Scholer HR, Sterneckert J. Genetic correction of a LRRK2 mutation in human iPSCs links parkinsonian neurodegeneration to ERK-dependent changes in gene expression. Cell Stem Cell. 2013;12:354–67.

    Article  CAS  Google Scholar 

  53. Skibinski G, Nakamura K, Cookson MR, Finkbeiner S. Mutant LRRK2 toxicity in neurons depends on LRRK2 levels and synuclein but not kinase activity or inclusion bodies. J Neurosci. 2014;34:418–33.

    Article  CAS  Google Scholar 

  54. Martin I, Kim JW, Lee BD, Kang HC, Xu JC, Jia H, Stankowski J, Kim MS, Zhong J, Kumar M, Andrabi SA, Xiong Y, Dickson DW, Wszolek ZK, Pandey A, Dawson TM, Dawson VL. Ribosomal protein s15 phosphorylation mediates LRRK2 neurodegeneration in Parkinson’s disease. Cell. 2014;157:472–85.

    Article  CAS  Google Scholar 

  55. Su YC, Qi X. Inhibition of excessive mitochondrial fission reduced aberrant autophagy and neuronal damage caused by LRRK2 G2019S mutation. Hum Mol Genet. 2013;22:4545–61.

    Article  CAS  Google Scholar 

  56. Wang X, Yan MH, Fujioka H, Liu J, Wilson-Delfosse A, Chen SG, Perry G, Casadesus G, Zhu X. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet. 2012;21:1931–44.

    Article  CAS  Google Scholar 

  57. Melrose HL, Dachsel JC, Behrouz B, Lincoln SJ, Yue M, Hinkle KM, Kent CB, Korvatska E, Taylor JP, Witten L, Liang YQ, Beevers JE, Boules M, Dugger BN, Serna VA, Gaukhman A, Yu X, Castanedes-Casey M, Braithwaite AT, Ogholikhan S, Yu N, Bass D, Tyndall G, Schellenberg GD, Dickson DW, Janus C, Farrer MJ. Impaired dopaminergic neurotransmission and microtubule-associated protein tau alterations in human LRRK2 transgenic mice. Neurobiol Dis. 2010;40:503–17.

    Article  CAS  Google Scholar 

  58. Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, Braithwaite AT, Ogholikhan S, Aigner R, Winkler J, Farrer MJ, Gage FH. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis. 2011;41:706–16.

    Article  CAS  Google Scholar 

  59. Bjorklund A, Stenevi U. Reconstruction of the nigrostriatal dopamine pathway by intracerebral nigral transplants. Brain Res. 1979;177:555–60.

    Article  CAS  Google Scholar 

  60. Perlow MJ, Freed WJ, Hoffer BJ, Seiger A, Olson L, Wyatt RJ. Brain grafts reduce motor abnormalities produced by destruction of nigrostriatal dopamine system. Science. 1979;204:643–7.

    Article  CAS  Google Scholar 

  61. Grow DA, McCarrey JR, Navara CS. Advantages of nonhuman primates as preclinical models for evaluating stem cell-based therapies for Parkinson’s disease. Stem Cell Res. 2016;17:352–66.

    Article  CAS  Google Scholar 

  62. Vermilyea SC, Lu J, Olsen M, Guthrie S, Tao Y, Fekete EM, Riedel MK, Brunner K, Boettcher C, Bondarenko V, Brodsky E, Block WF, Alexander A, Zhang SC, Emborg ME. Real-time intraoperative MRI intracerebral delivery of induced pluripotent stem cell-derived neurons. Cell Transplant. 2017;26(4):613–24.

    Article  Google Scholar 

  63. Choi SS, Lee SR, Kim SU, Lee HJ. Alzheimer’s disease and stem cell therapy. Exp Neurobiol. 2014;23:45–52.

    Article  Google Scholar 

  64. Kim JH, Auerbach JM, Rodriguez-Gomez JA, Velasco I, Gavin D, Lumelsky N, Lee SH, Nguyen J, Sanchez-Pernaute R, Bankiewicz K, McKay R. Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson’s disease. Nature. 2002;418:50–6.

    Article  CAS  Google Scholar 

  65. Cho MS, Lee YE, Kim JY, Chung S, Cho YH, Kim DS, Kang SM, Lee H, Kim MH, Kim JH, Leem JW, Oh SK, Choi YM, Hwang DY, Chang JW, Kim DW. Highly efficient and large-scale generation of functional dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci U S A. 2008;105:3392–7.

    Article  CAS  Google Scholar 

  66. Chung S, Moon JI, Leung A, Aldrich D, Lukianov S, Kitayama Y, Park S, Li Y, Bolshakov VY, Lamonerie T, Kim KS. ES cell-derived renewable and functional midbrain dopaminergic progenitors. Proc Natl Acad Sci U S A. 2011;108:9703–8.

    Article  CAS  Google Scholar 

  67. Brederlau A, Correia AS, Anisimov SV, Elmi M, Paul G, Roybon L, Morizane A, Bergquist F, Riebe I, Nannmark U, Carta M, Hanse E, Takahashi J, Sasai Y, Funa K, Brundin P, Eriksson PS, Li JY. Transplantation of human embryonic stem cell-derived cells to a rat model of Parkinson’s disease: effect of in vitro differentiation on graft survival and teratoma formation. Stem Cells. 2006;24:1433–40.

    Article  CAS  Google Scholar 

  68. Tomaskovic-Crook E, Crook JM. Human embryonic stem cell therapies for neurodegenerative diseases. CNS Neurol Disord Drug Targets. 2011;10:440–8.

    Article  CAS  Google Scholar 

  69. Tabar VS. 133 the development of human embryonic stem cell-derived dopamine neurons for clinical use in Parkinson disease. Neurosurgery. 2016;63(Suppl 1):154–5.

    Article  Google Scholar 

  70. O’Keeffe FE, Scott SA, Tyers P, O’Keeffe GW, Dalley JW, Zufferey R, Caldwell MA. Induction of A9 dopaminergic neurons from neural stem cells improves motor function in an animal model of Parkinson’s disease. Brain. 2008;131:630–41.

    Article  Google Scholar 

  71. Redmond DE Jr, Bjugstad KB, Teng YD, Ourednik V, Ourednik J, Wakeman DR, Parsons XH, Gonzalez R, Blanchard BC, Kim SU, Gu Z, Lipton SA, Markakis EA, Roth RH, Elsworth JD, Sladek JR Jr, Sidman RL, Snyder EY. Behavioral improvement in a primate Parkinson’s model is associated with multiple homeostatic effects of human neural stem cells. Proc Natl Acad Sci U S A. 2007;104:12175–80.

    Article  CAS  Google Scholar 

  72. Shen Y, Huang J, Liu L, Xu X, Han C, Zhang G, Jiang H, Li J, Lin Z, Xiong N, Wang T. A compendium of preparation and application of stem cells in Parkinson’s disease: current status and future prospects. Front Aging Neurosci. 2016;8:117.

    PubMed  PubMed Central  Google Scholar 

  73. Chen D, Fu W, Zhuang W, Lv C, Li F, Wang X. Therapeutic effects of intranigral transplantation of mesenchymal stem cells in rat models of Parkinson’s disease. J Neurosci Res. 2017;95(3):907–17.

    Article  CAS  Google Scholar 

  74. Kim SU, Park IH, Kim TH, Kim KS, Choi HB, Hong SH, Bang JH, Lee MA, Joo IS, Lee CS, Kim YS. Brain transplantation of human neural stem cells transduced with tyrosine hydroxylase and GTP cyclohydrolase 1 provides functional improvement in animal models of Parkinson disease. Neuropathology. 2006b;26:129–40.

    Article  Google Scholar 

  75. Schwerk A, Altschuler J, Roch M, Gossen M, Winter C, Berg J, Kurtz A, Akyuz L, Steiner B. Adipose-derived human mesenchymal stem cells induce long-term neurogenic and anti-inflammatory effects and improve cognitive but not motor performance in a rat model of Parkinson’s disease. Regen Med. 2015;10:431–46.

    Article  CAS  Google Scholar 

  76. Zhou Y, Sun M, Li H, Yan M, He Z, Wang W, Wang W, Lu S. Recovery of behavioral symptoms in hemi-parkinsonian rhesus monkeys through combined gene and stem cell therapy. Cytotherapy. 2013;15:467–80.

    Article  CAS  Google Scholar 

  77. Rhee YH, Ko JY, Chang MY, Yi SH, Kim D, Kim CH, Shim JW, Jo AY, Kim BW, Lee H, Lee SH, Suh W, Park CH, Koh HC, Lee YS, Lanza R, Kim KS, Lee SH. Protein-based human iPS cells efficiently generate functional dopamine neurons and can treat a rat model of Parkinson disease. J Clin Invest. 2011;121:2326–35.

    Article  CAS  Google Scholar 

  78. Hallett PJ, Deleidi M, Astradsson A, Smith GA, Cooper O, Osborn TM, Sundberg M, Moore MA, Perez-Torres E, Brownell AL, Schumacher JM, Spealman RD, Isacson O. Successful function of autologous iPSC-derived dopamine neurons following transplantation in a non-human primate model of Parkinson’s disease. Cell Stem Cell. 2015;16:269–74.

    Article  CAS  Google Scholar 

  79. Nishimura K, Doi D, Samata B, Murayama S, Tahara T, Onoe H, Takahashi J. Estradiol facilitates functional integration of iPSC-derived dopaminergic neurons into striatal neuronal circuits via activation of integrin alpha5beta1. Stem Cell Reports. 2016;6:511–24.

    Article  CAS  Google Scholar 

  80. Jiang P, Huang P, Yen SH, Zubair AC, Dickson DW. Genetic modification of H2AX renders mesenchymal stromal cell-derived dopamine neurons more resistant to DNA damage and subsequent apoptosis. Cytotherapy. 2016;18:1483–92.

    Article  CAS  Google Scholar 

  81. Kim S, Honmou O, Kato K, Nonaka T, Houkin K, Hamada H, Kocsis JD. Neural differentiation potential of peripheral blood- and bone-marrow-derived precursor cells. Brain Res. 2006a;1123:27–33.

    Article  CAS  Google Scholar 

Download references

Acknowledgement

This work was carried out with the support of “Cooperative Research Program for Agriculture Science & Technology Development (Project No.PJ011792032017)” Rural Development Administration, Republic of Korea. This research was syupported by the Bio & Medical Technology Development Program of the NRF funded by the Korean gorverment, MSIP (NRF-2016M3A9B6900776).

Conflicts of Interest: The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Singapore Pte Ltd.

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Kim, S.Y., Choi, S.S., Lee, DS., Lee, S.H., Cha, S.H., Lee, H.J. (2019). Pathology and Cell-Based Therapy of Parkinson’s Disease. In: Singh, S., Joshi, N. (eds) Pathology, Prevention and Therapeutics of Neurodegenerative Disease. Springer, Singapore. https://doi.org/10.1007/978-981-13-0944-1_3

Download citation

  • DOI: https://doi.org/10.1007/978-981-13-0944-1_3

  • Published:

  • Publisher Name: Springer, Singapore

  • Print ISBN: 978-981-13-0943-4

  • Online ISBN: 978-981-13-0944-1

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics