Int J Stem Cells.  2015 May;8(1):106-114. 10.15283/ijsc.2015.8.1.106.

Direct Differentiation of Adult Ocular Progenitors into Striatal Dopaminergic Neurons

Affiliations
  • 1Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, United States. iahmad@unmc.edu
  • 2Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.
  • 3Department of Pharmacy Practice, Creighton University, Omaha, NE, United States.

Abstract

Parkinson's disease, characterized by motor dysfunction due to the loss of nigrostriatal dopaminergic neurons, is one of the most prevalent age-related neurodegenerative disorders. Given there is no current cure, the stem cell approach has emerged as a viable therapeutic option to replace the dopaminergic neurons that are progressively lost to the disease. The success of the approach is likely to depend upon accessible, renewable, immune compatible, and non-tumorigenic sources of neural progenitors from which stable dopaminergic neurons can be generated efficaciously. Here, we demonstrate that neural progenitors derived from limbus, a regenerative and accessible ocular tissue, represent a safe source of dopaminergic neurons. When the limbus-derived neural progenitors were subjected to a well-established protocol of directed differentiation under the influence of Shh and FGF8, they acquired the biochemical and functional phenotype of dopaminergic neurons that included the ability to synthesize dopamine. Their intrastriatal transplantation in the rat model of hemi-Parkinsonism was associated with a reduction in the amphetamine-induced rotation. No tumor formation was observed 6 weeks post-transplantation. Together, these observations posit limbus-derived neural progenitors as an accessible and safe source of dopaminergic neurons for a potential autologous ex-vivo stem cell approach to Parkinson's disease.

Keyword

Parkinson's disease; Dopaminergic neurons; Directed differentiation; Stem cells

MeSH Terms

Adult*
Dopamine
Dopaminergic Neurons*
Humans
Models, Animal
Neurodegenerative Diseases
Parkinson Disease
Phenotype
Stem Cells
Dopamine

Figure

  • Fig. 1 Generation and differentiation of limbus-derived neural progenitors. A schematic representation of the differentiation protocol is provided in A. Cell dissociates from limbus when cultured in the presence of FGF2 and Noggin generated neurospheres with BrdU+ and Nestin+ cells (B; C, dissociated neurospheres). BrdU-tagged neurospheres, when co-cultured with E18 hippocampal cells spread out and cells acquired neuronal morphology, co-expressing neuronal markers β-tubulin/DAPI (D). Whole cell voltage clamped recordings of differentiated cells with neuronal morphology revealed fast inward currents and sustained outward currents (E). The current-voltage (I~V) curve (F) exhibited a typical I-V relationship of fast inward currents due to the voltage gated Na+ channels and sustained outward currents due to the outwardly rectifying K+ channels.

  • Fig. 2 Differentiation of limbus-derived neural progenitors along the dopaminergic cell lineage. Limbus-derived neurospheres were subjected to three stages directed dopaminergic neuron differentiation protocol, using Shh and FGF8 (A). Cell dissociates from GFP-rat limbus when cultured in the presence of FGF2 and Noggin generated neurospheres with GFP+ neural progenitors. Neurospheres spread under the influence of ITSFn (Stage1) and cells therein acquired neuronal morphology under the influence of Shh and FGF8 (Stage II) and increased in number with ascorbate (Stage III).

  • Fig. 3 Molecular and biochemical properties of limbus-derived dopaminergic neurons. RT-PCR analysis of cells in neurospheres (lane 1) and cells at the end of Stage III of differentiation (lane 2) demonstrated the induction of transcripts corresponding to dopaminergic neuron regulator (Nurr1 and Ptx3) and phenotypic and functional markers (Calbindin, TH, DAT, and AADC, compared to the former (A). Immunocytochemical analysis of BrdU-tagged Stage III cells revealed BrdU+ cells, expressing dopaminergic neuron markers, TH (B, C, F) and En1 (D, E, F).

  • Fig. 4 Functional analysis of limbus-derived dopaminergic neurons. The schematic representation of the functional analysis of non-cell autonomously derived dopaminergic neurons is given in A. the HPLC-EC analysis revealed that limbus-derived Stage III cells release DA and DOPAC in the culture medium in response to KCL (B). Un-induced cells were negative for DA and DOPAC. Transplantation of limbus-derived GFP+ stage III cells in the striatum of 6OHDA-lesioned rats reduced the amphetamine-induced rotations to sham-lesioned controls one-week post-translation (C). Retrospective immunohistochemical analysis of sections through the striatum of transplanted rats revealed GFP+ limbus-derived stage III cells expressing TH in the host’s striatum (D~H).


Reference

References

1. Dyson SC, Barker RA. Cell-based therapies for Parkinson’s disease. Expert Rev Neurother. 2011; 11:831–844. PMID: 21651331.
Article
2. Kim JH, Auerbach JM, Rodríguez-Gómez JA, Velasco I, Gavin D, Lumelsky N, Lee SH, Nguyen J, Sánchez-Pernaute R, Bankiewicz K, McKay R. Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson’s disease. Nature. 2002; 418:50–56. DOI: 10.1038/nature00900. PMID: 12077607.
Article
3. Sundberg M, Isacson O. Advances in stem-cell--generated transplantation therapy for Parkinson’s disease. Expert Opin Biol Ther. 2014; 14:437–453. PMID: 24437368.
Article
4. Parameswaran S, Balasubramanian S, Rao MS, Ahmad I. Concise review: non-cell autonomous reprogramming: a nucleic acid-free approach to induction of pluripotency. Stem Cells. 2011; 29:1013–1020. DOI: 10.1002/stem.655. PMID: 21544901.
Article
5. Kitada M, Dezawa M. Parkinson’s disease and mesenchymal stem cells: potential for cell-based therapy. Parkinsons Dis. 2012; 2012:873706.
Article
6. Dezawa M, Kanno H, Hoshino M, Cho H, Matsumoto N, Itokazu Y, Tajima N, Yamada H, Sawada H, Ishikawa H, Mimura T, Kitada M, Suzuki Y, Ide C. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest. 2004; 113:1701–1710. PMID: 15199405. PMCID: 420509.
Article
7. Lavker RM, Tseng SC, Sun TT. Corneal epithelial stem cells at the limbus: looking at some old problems from a new angle. Exp Eye Res. 2004; 78:433–446. PMID: 15106923.
Article
8. Rama P, Matuska S, Paganoni G, Spinelli A, De Luca M, Pellegrini G. Limbal stem-cell therapy and long-term corneal regeneration. N Engl J Med. 2010; 363:147–155. PMID: 20573916.
Article
9. Zhao X, Das AV, Thoreson WB, James J, Wattnem TE, Rodriguez-Sierra J, Ahmad I. Adult corneal limbal epithelium: a model for studying neural potential of non-neural stem cells/progenitors. Dev Biol. 2002; 250:317–331. PMID: 12376106.
Article
10. Zhao X, Das AV, Bhattacharya S, Thoreson WB, Sierra JR, Mallya KB, Ahmad I. Derivation of neurons with functional properties from adult limbal epithelium: implications in autologous cell therapy for photoreceptor degeneration. Stem Cells. 2008; 26:939–949. DOI: 10.1634/stemcells.2007-0727. PMID: 18203675.
Article
11. Zetterström RH, Solomin L, Jansson L, Hoffer BJ, Olson L, Perlmann T. Dopamine neuron agenesis in Nurr1-deficient mice. Science. 1997; 276:248–250. DOI: 10.1126/science.276.5310.248. PMID: 9092472.
Article
12. Liberatore GT, Jackson-Lewis V, Vukosavic S, Mandir AS, Vila M, McAuliffe WG, Dawson VL, Dawson TM, Przedborski S. Inducible nitric oxide synthase stimulates dopaminergic neurodegeneration in the MPTP model of Parkinson disease. Nat Med. 1999; 5:1403–1409. DOI: 10.1038/70978. PMID: 10581083.
Article
13. Lee SH, Lumelsky N, Studer L, Auerbach JM, McKay RD. Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells. Nat Biotechnol. 2000; 18:675–679. DOI: 10.1038/76536. PMID: 10835609.
Article
14. Smidt MP, Asbreuk CH, Cox JJ, Chen H, Johnson RL, Burbach JP. A second independent pathway for development of mesencephalic dopaminergic neurons requires Lmx1b. Nat Neurosci. 2000; 3:337–341. DOI: 10.1038/73902. PMID: 10725922.
Article
15. Burbach JPH, Smits S, Smidt MP. Transcription factors in the development of midbrain dopamine neurons. Ann N Y Acad Sci. 2003; 991:61–68. DOI: 10.1111/j.1749-6632.2003.tb07463.x. PMID: 12846974.
Article
16. Cadet JL, Zhu SM. The intrastriatal 6-hydroxydopamine model of hemiparkinsonism: quantitative receptor autoradiographic evidence of correlation between circling behavior and presynaptic as well as postsynaptic nigrostriatal markers in the rat. Brain Res. 1992; 595:316–326. DOI: 10.1016/0006-8993(92)91066-N. PMID: 1467973.
Article
17. Parameswaran S, Balasubramanian S, Babai N, DelDebbio CB, Harms DW, Gurumurthy CB, Rao MS, Sharp JG, Ahmad I. Nucleic acid and non-nucleic acid-based reprogramming of adult limbal progenitors to pluripotency. PLoS One. 2012; 7:e46734. DOI: 10.1371/journal.pone.0046734. PMID: 23056428. PMCID: 3466310.
Article
18. Kim TE, Lee HS, Lee YB, Hong SH, Lee YS, Ichinose H, Kim SU, Lee MA. Sonic hedgehog and FGF8 collaborate to induce dopaminergic phenotypes in the Nurr1- overexpressing neural stem cell. Biochem Biophys Res Commun. 2003; 305:1040–1048. DOI: 10.1016/S0006-291X(03)00879-9. PMID: 12767935.
Article
19. Alves dos Santos MT, Smidt MP. En1 and Wnt signaling in midbrain dopaminergic neuronal development. Neural Dev. 2011; 6:23. DOI: 10.1186/1749-8104-6-23. PMID: 21569278. PMCID: 3104484.
Article
20. Nelson EL, Liang CL, Sinton CM, German DC. Midbrain dopaminergic neurons in the mouse: computer-assisted mapping. J Comp Neurol. 1996; 369:361–371. DOI: 10.1002/(SICI)1096-9861(19960603)369:3<361::AID-CNE3>3.0.CO;2-3. PMID: 8743418.
Article
21. González-Hernández T, Rodríguez M. Compartmental organization and chemical profile of dopaminergic and GABAergic neurons in the substantia nigra of the rat. J Comp Neurol. 2000; 421:107–135. PMID: 10813775.
Article
22. Capaccione KM, Pine SR. The Notch signaling pathway as a mediator of tumor survival. Carcinogenesis. 2013; 34:1420–1430. DOI: 10.1093/carcin/bgt127. PMID: 23585460. PMCID: 3697894.
Article
Full Text Links
  • IJSC
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