Development of iPSC technology in Parkinson’s Disease

Cell Gene Therapy Insights 2016; 2(3), 377-385.

10.18609/cgti.2016.048

Published: 4 October 2016
Expert Insight
Jun Takahashi

The central nervous system has very little potential for regeneration which is why cell replacement therapy offers great potential for functional recovery in neurodegenerative diseases. Following the discovery of human embryonic stem cells in 1998 and induced pluripotent stem cells in 2007, we are now able to manipulate the quantity and quality of donor cells utilized in stem cell-based therapies. For Parkinson’s disease in particular, which will be the focus of this article, precise protocols to establish induced pluripotent stem cells and to derive dopaminergic neurons have been developed to clinical grade, and preclinical data concerning the efficacy and safety of these cells now exists for rodent and monkey models. Based on these efforts, clinical trials for a number of neurodegenerative disease are expected to commence in the near term.

Due to the poor regenerative ability of the CNS, neurodegenerative diseases have become the focus of efforts to develop cell replacement strategies. In Parkinson’s disease (PD), midbrain dopaminergic (DA) neurons degenerate, and therefore cell transplantation represents a plausible approach to replace these lost neurons. Among neurodegenerative diseases, PD has seen a large number of clinical cases of cell transplantation since the late 1980’s [1]. Accordingly, the focus of this article is to describe the development of a relatively new cell source – induced pluripotent stem cells (iPSCs) – for cell therapies to treat PD, with other neurodegenerative diseases described briefly.

PD is caused by the progressive loss of nigrostriatal DA neurons, and the main symptoms are motor dysfunctions such as tremor, rigidity and akinesia. In 1987, early clinical research utilized fetal cells from the ventral mesencephalon which were transplanted into PD patients, with the results of clinical trials demonstrating that the grafted cells survived and functioned as DA neurons over 20 years in some patients [2]. However, ethical issues regarding the use of fetal tissues and limited amounts of accessible donor tissues prevented fetal cell transplantation from becoming a standard therapy to treat PD patients. Stem cells, in particular pluripotent stem cells such as iPSCs and embryonic stem cells (ESCs), therefore offered a promising alternative donor cell source.


Methods to establish iPSCs

ESCs were reported as the first human pluripotent stem cells in 1998 [3] and are derived from the inner cell mass of human blastocysts. Importantly, they are able to proliferate indefinitely and maintain their pluripotency, which makes them potentially ideal candidates for cell-based therapies. However, the inclusion of blastocysts in the ESC-generating process has raised a number of ethical questions thus making it difficult to use or even establish human ESCs. In addition, any cell therapies generated from ESCs will be allogeneic and thus life-long immunosuppression would be required, with immune rejection remaining a strong possibility in patients.

The first report of murine iPSCs was published in 2006 [4] and that of human iPSCs in 2007 [5,6]. Since then, iPSCs have been widely investigated as they hold the potential to produce numerous different cell types and tissues both in vitro and in vivo. They too are pluripotent and self-renewing and can be generated as autologous therapies, thus avoid both of the aforementioned issues with ESC use. Nevertheless, other issues have limited their clinical application thus far. In the original iPSC-generating method, four transcriptional factors (c-Myc, Oct4, Sox2, Klf4) were introduced into mouse embryonic or adult fibroblasts via retroviral vectors [4]. However, concerns arose regarding the use of the proto-oncogene c-Myc as this could increase the risk of tumorigenesis, so its expression in the method was replaced by Nanog and LIN28 [6], Glis1 [7], or L-Myc [8] or by inhibiting p53 [9,10]. Another problem concerned retroviral integration, which may cause genomic mutations [11]. This risk can be circumvented by integration-free methods using alternative vectors such as adenovirus [12], Sendai virus [13] or plasmids [14].

Additionally, the use of fibroblasts requires skin biopsies, which is an invasive procedure for the patient or donor volunteer. It was since determined that iPSCs can be established from a variety of somatic cells, and efficient iPSC induction from cord blood cells and peripheral blood cells was reported [15], the acquisition of which is less invasive. Finally, the original human iPSCs were established on feeder cells, namely mouse embryonic fibroblasts. However, for clinical applications xeno-derived cells should be avoided. Accordingly, a xeno-free matrix has been developed to achieve xeno-free, feeder-free culture of iPSCs [16], in addition to further improvements such as development of laminin fragment (LM511-E8), which supports not only iPSC culture [17,18], but also neural induction [19]. Overall, the methods to establish and expand human iPSCs have been steadily progressing towards clinical application (Table 1).


Protocols to induce neural cells

Leading on from the discovery and establishment of ESCs and iPSCs, protocols were developed to induce neural cells. Lineage specification from pluripotent stem cells is determined by several signals such as BMP, TGF/Activin/Nodal, and Wnt [20]. For efficient neural induction, the inhibition of both BMP and TGF/Activin/Nodal signaling is essential, and can be achieved by the inhibition of SMAD1/5/8 and SMAD2/3 (i.e., dual SMAD inhibition) [21]. By using this method, almost 100% of human ES/iPSCs can be differentiated into a neural lineage.

Furthermore, differentiation into specific neuronal subtypes from pluripotent stem cells has also been extensively studied. Based on established methods for in vitro differentiation of DA neurons, efficient protocols for dopaminergic differentiation of iPSCs were then developed. These protocols are based on the recapitulation of embryonic neural development and are performed by either neurosphere or monolayer culture. It is known that DA neurons of the nigro-striatal pathway are derived from the floor plate in the midbrain [22], thus to derive DA neurons from iPSCs requires a combination of dual SMAD inhibition, midbrain specification by Wnt signaling activation, and ventralization by Sonic hedgehog (Figure 1) [23–25].


Preclinical studies using animal models

Before moving towards clinical application of iPSC-derived neural cells, cell efficacy and safety must be examined carefully through animal experiments. These studies have indicated donor neurons derived from ESCs or iPSCs show similar properties. Therefore the following descriptions of these studies refers to either or both cell source interchangeably.

As discussed, for PD proof-of-concept of cell replacement therapy has already been established through early clinical trials using fetal ventral midbrain cells [1,2]. Whilst fetal cell transplantation for PD patients is certainly not perfect, there are many patients who have benefited from the therapy for nearly 20 years [32–34], and a new trial is ongoing to optimize the protocol [35]

With efficient protocols established to induce midbrain DA neurons, animal studies have looked at the impact of these induced DA neurons in animal models of PD. When grafted into the striatum of 6-OHDA-lesioned rats [24,26] or MPTP-treated monkeys [27], the neurons showed robust survival and function, which improved behavioral impairments in the animals. In addition, human ESC-derived DA neurons showed equal potency and efficacy to fetal midbrain DA neurons [28]. These results support the idea that ES/iPSCs can be used as a cell-based therapy for PD; however, it is important to note that residual undifferentiated stem cells or proliferating neural progenitor cells may cause tumor formation [27,29,30]. To reduce the tumorigenic potential of the ES/iPSC-based protocol, fluorescence activated cell sorting using antibodies for CORIN, a floor plate marker [19], or ALCAM, a CNS microvascular endothelium marker [31], have been developed. For example, by sorting CORIN+ cells, researchers can enrich DA progenitor cells as donor cells, which increases the number and density of DA neurons in the graft [19]. In addition, undifferentiated and proliferating cells can be removed, which prevents tumor formation by the grafted cells via this method. The differentiation efficiency differs from culture to culture, but by using this sorting procedure, we are always able to obtain high quality donor cells. These features are important, especially in clinical settings and this cell sorting technology can be utilized across a range of cell therapies for various diseases.


Autologous vs allogeneic transplantation

One of the advantages of iPSCs is that they enable autologous transplantation, which does not require immunosuppression, thus avoiding the adverse effects associated with long-term immunosuppressant drug use. iPSC-derived DA neurons autologously transplanted in non-human primates have already shown good survival without triggering an immune response by the host brain [32,33], indicating this treatment is ideal from the immunological point of view. Clinical-grade manufacturing of autologous iPSCs for a patient, however, is costly and laborious, and additionally there is concern regarding the vulnerability of iPSCs derived from patients who have disease-specific genetic backgrounds. These problems may be surmountable by harnessing new technologies, such as gene editing and automated cell culture systems, which would potentially allow the widespread use of autologous technologies.

Another strategy to reduce the immune response in cell transplantation is matching human leukocyte antigens (HLA), which in the case of organ transplantations such as kidney and bone marrow, matching HLA-A, -B, and -DR improves graft survival. Major histocompatibility complex (MHC)-matched transplantation of retinal pigment epithelial cells derived from MHC-homozygous iPSCs resulted in no immune response in monkeys [34]. Another advantage of iPSCs is that they can be established from individuals with homozygous HLA haplotypes, which simplifies HLA-matched transplantation, and could further allow clinical-grade manufacturing at affordable prices. It is estimated that a cell bank from HLA-homozygous volunteers of the 10 most frequent haplotypes would match 37.7% of the UK population, and 150 similar volunteers would match 93% [35]. According to another estimate, 50 lines would cover 90.7% [36] or 73% [37] of the Japanese population. Based on these estimates, several projects are underway to establish clinical-grade iPSC stocks from HLA-homozygous volunteers.


Getting treatments to the clinic

As mentioned, clinical trials in PD using fetal ventral midbrain cells have shown varying degrees of benefit [1,2]. Subsequent double-blind, placebo-controlled clinical trials failed to show significant differences between the transplanted and control group [38,39]. Moreover, these studies reported the appearance of graft-induced dyskinesia. These negative results were discouraging, but the studies also revealed that a subpopulation of the cases had significantly restored function and improved quality of life. In addition, several problems in the protocol, such as the absence of immunosuppression and a too short period of observation, were posited as potential reasons for the negative outcomes. Whilst fetal cell transplantation for PD patients is not ideal, recent studies have reported that there are many patients who have benefited from the therapy for nearly 20 years [40–42]. Currently, a new trial is ongoing to optimize the protocol and to study any benefits in a more rigorous and consistent way than previous trials [43]. The optimization of future clinical trials using ES/iPSC-based therapies for PD, including criteria for patient selection, observation time, and assessment of symptoms, will benefit from previous fetal-cell-based trials.


Other neurodegenerative diseases

While ES/iPSC-based therapies for PD is the focus of this Expert Insight, I would also like to give brief attention to neurodegenerative diseases that target other neuronal subtypes. Pluripotent stem cells, for example have been used to generate the cerebral neocortex in a 3D culture that allows a self-organization that mimics human neocorticogenesis [44]. Similar self-organization strategies can be applied to other types of neural cells such as pituitary gland [45], retina [46], and hippocampus [47]. Additionally, patient-derived iPSCs are a powerful tool for analyzing the pathology of specific diseases and drug discovery. In fact, iPSCs have already been established from patients of various neurological diseases including PD [48], AD [49], amyotrophic lateral sclerosis [50], and schizophrenia [51].

In neurodegenerative diseases such as AD, cell transplantation is more complicated than in PD because the degenerated neurons are widely spread. Mouse AD models have indicated that the transplantation of mesenchymal or neural stem cells could improve cognitive and memory function. More specifically, bone marrow-derived mesenchymal stem cells were shown to ameliorate Aβ-induced neurotoxicity and cognitive decline by inhibiting apoptotic cell death and oxidative stress in the hippocampus [52]. When hippocampal neural stem cells were injected into the bilateral hippocampi of aged triple transgenic mice that express pathogenic forms of amyloid precursor protein, presenilin, and tau, they enhanced hippocampal synaptic density in a manner mediated by brain-derived neurotrophic factor and ameliorated the complex behavioral deficits associated with widespread AD pathology [53]. However, in these cases, the grafted stem cells functioned via cytokine effects rather than by cell replacement. In fact, it remains unknown whether ES/iPSC-derived neural cells function in the brain of animal models of AD or other neurodegenerative diseases. Therefore, more studies about the pathology of the diseases and ES/iPSC technologies are required for the development of ES-iPSC-based therapies against neurodegenerative diseases.

With that said, clinical trials using fetal neural stem cells have been performed not only for PD but also for Huntington’s disease [54] and amyotrophic lateral sclerosis [55]. Unfortunately, like PD, the results varied widely between patients, making it premature to judge whether stem cell-based therapies can become the standard for neurodegenerative diseases. Two reasons are the complexity of the CNS and pathology of the diseases.


Conclusion

In the case of neuronal cell transplantation, survival of the grafted cells is not sufficient for an effective clinical outcome as the grafted cells must also extend neurites and form synapses with the host neurons for curative effects to be observed. In this context, not only the donor cells but also the host environment is important. Therefore, drugs or gene modifications that promote cell survival, neurite extension, and synapse formation would enhance the therapeutic effect of the grafted cells. Rehabilitation will also contribute to the construction of appropriate neuronal circuits by activity-dependent modifications. Thus, a key to successful regenerative medicine for neurodegenerative diseases is the combination of cell transplantation, medical treatment, and rehabilitation. Thanks to the development of ES/iPSCs, we now have technologies to manipulate the quantity and quality of donor cells to realize a new era of cell-based therapies.


Financial disclosure

The author has no relevant financial involvement with an organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock options or ownership, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.


Acknowledgement

The author thanks Dr. Peter Karagiannis for critical reading of the manuscript.

References

1. Barker RA, Drouin-Ouellet J, Parmar M. Cell-based therapies for Parkinson disease—past insights and future potential. Nat. Rev. Neurol. 2015; 11(9), 492–503. CrossRef

2. Barker RA, Barrett J, Mason SL, Björklund A. Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson’s disease. Lancet Neurol. 2013; 12(1), 84–91. CrossRef

3. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282(5391), 1145–7. CrossRef

4. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126, 663–76. CrossRef

5. Takahashi K, Tanabe K, Ohnuki M et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131, 861–72. CrossRef

6. Yu J, Vodyanik MA, Smuga-Otto K et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007; 318(5858), 1917–20. CrossRef

7. Maekawa M, Yamaguchi K, Nakamura T et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature 2011; 474(7350), 225–9. CrossRef

8. Nakagawa M, Takizawa N, Narita M, Ichisaka T, Yamanaka S. Promotion of direct reprogramming by transformation-deficient Myc. Proc. Natl. Acad. Sci. USA 2010; 107, 14152–7. CrossRef

9. Hong H, Takahashi K, Ichisaka T et al. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature 2009; 460, 1132–5. CrossRef

10. Gore A, Li Z, Fung HL et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 2011; 471, 63–7. CrossRef

11. Selvaraj V, Plane JM, Williams AJ, Deng W. Switching cell fate: the remarkable rise of induced pluripotent stem cells and lineage reprogramming technologies. Trends Biotechnol. 2010; 28(4), 214–23. CrossRef

12. Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem cells generated without viral integration. Science 2008; 322(5903), 945–9. CrossRef

13. Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad. Ser. B. Phys. Biol. Sci. 2009; 85, 348–62. CrossRef

14. Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science 2008; 322(5903), 949–53. CrossRef

15. Okita K, Yamakawa T, Matsumura Y et al. An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells 2013; 31(3), 458–66. CrossRef

16. Bergström R, Ström S, Holm F, Feki A, Hovatta O. Xeno-free culture of human pluripotent stem cells. Methods Mol. Biol. 2011; 767, 125–36. CrossRef

17. Miyazaki T, Futaki S, Suemori H et al. Laminin E8 fragments support efficient adhesion and expansion of dissociated human pluripotent stem cells. Nat. Commun. 2012; 3, 1236. CrossRef

18. Nakagawa M, Taniguchi Y, Senda S et al. A novel efficient feeder-free culture system for the derivation of human induced pluripotent stem cells. Sci. Rep. 2014; 4, 3594. CrossRef

19. Doi D, Samata B, Katsukawa M et al. Isolation of Human Induced Pluripotent Stem Cell-derived Dopaminergic Progenitors by Cell Sorting for Successful Transplantation. Stem Cell Rep. 2014; 2(3), 337–50. CrossRef

20. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 2008; 132(4), 661–80. CrossRef

21. Chambers SM, Fasano CA, Papapetrou EP et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol. 2009; 27, 275–80. CrossRef

22. Ono Y, Nakatani T, Sakamoto Y et al. Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells. Development 2007; 134, 3213–25. CrossRef

23. Kriks S, Shim JW, Piao J et al. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson’s disease. Nature 2011; 480, 547–51. CrossRef

24. Kirkeby A, Grealish S, Wolf D et al. Generation of Regionally Specified Neural Progenitors and Functional Neurons from Human Embryonic Stem Cells under Defined Conditions. Cell Rep. 2012; 1, 703–14. CrossRef

25. Xi J, Liu Y, Liu H, Chen H, Emborg ME, Zhang SC. Specification of Midbrain Dopamine Neurons from Primate Pluripotent Stem Cells. Stem Cells 2012; 30, 1655–63. CrossRef

26. Steinbeck JA, Choi SJ, Mrejeru A et al. Optogenetics enables functional analysis of human embryonic stem cell-derived grafts in a Parkinson’s disease model. Nat. Biotechnol. 2015; 33(2), 204–9. CrossRef

27. Doi D, Morizane A, Kikuchi T et al. Prolonged Maturation Culture Favors a Reduction in the Tumorigenicity and the Dopaminergic Function of Human ESC-Derived Neural Cells in a Primate Model of Parkinson’s Disease. Stem Cells 2012; 30, 935–45. CrossRef

28. Grealish S, Diguet E, Kirkeby A et al. Human ESC-derived dopamine neurons show similar preclinical efficacy and potency to fetal neurons when grafted in a rat model of Parkinson’s disease. Cell Stem Cell 2014; 15(5), 653–65. CrossRef

29. Brederlau A, Correia AS, Anisimov SV et al. 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. CrossRef

30. Elkabetz Y, Panagiotakos G, Al Shamy G, Socci ND, Tabar V, Studer L. Human ES cell-derived neural rosettes reveal a functionally distinct early neural stem cell stage. Genes Dev. 2008; 22, 152–65. CrossRef

31. Bye CR, Jonsson ME, Bjorklund A, Parish CL, Thompson LH. Transcriptome analysis reveals transmembrane targets on transplantable midbrain dopamine progenitors. Proc. Natl. Acad. Sci. USA 2015; 112, 1946–55. CrossRef

32. Morizane A, Doi D, Kikuchi T et al. Direct Comparison of Autologous and Allogeneic Transplantation of iPSC-Derived Neural Cells in the Brain of a Nonhuman Primate. Stem Cell Rep. 2013; 1, 283–92. CrossRef

33. Hallett P, Deleidi M, Astradsson A et al. 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. CrossRef

34. Sugita S, Iwasaki Y, Makabe K et al. Successful Transplantation of Retinal Pigment Epithelial Cells from MHC Homozygote iPSCs in MHC-Matched Models. Stem Cell Rep. 2016; pii S2213-6711(16), 30178–3. CrossRef

35. Evans JR, Mason SL, Barker RA. Current status of clinical trials of neural transplantation in Parkinson’s disease. Prog. Brain Res. 2012; 200, 169–98. CrossRef

36. Taylor CJ, Peacock S, Chaudhry AN, Bradley JA, Bolton EM. Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA types. Cell Stem Cell 2012; 11, 147–52. CrossRef

37. Nakatsuji N, Nakajima F, Tokunaga K. HLA-haplotype banking and iPS cells. Nat. Biotechnol. 2008; 26, 739–40. CrossRef

38. Okita K, Matsumura Y, Sato Y et al. A more efficient method to generate integration-free human iPS cells. Nat. Methods 2011; 8, 409–12. CrossRef

39. Freed CR, Greene PE, Breeze RE et al. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N. Engl. J. Med. 344; 710–719, 2001. CrossRef

40. Olanow CW, Goetz CG, Kordower JH et al. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson’s disease. Ann. Neurol. 54; 403–414, 2003. CrossRef

41. Kefalopoulou Z, Politis M, Piccini P et al. Long-term clinical outcome of fetal cell transplantation for Parkinson disease: two case reports. JAMA Neurol. 2014; 71, 83–7. CrossRef

42. Hallett PJ, Cooper O, Sadi D, Robertson H, Mendez I, Isacson O. Long-term health of dopaminergic neuron transplants in Parkinson’s disease patients. Cell Rep. 2014; 7(6), 1755-61. CrossRef

43. Li W, Englund E, Widner H et al. Extensive graft-derived dopaminergic innervation is maintained 24 years after transplantation in the degenerating parkinsonian brain. Proc. Natl. Acad. Sci. USA 2016; 113(23), 6544-9. CrossRef

44. Evans JR, Mason SL, Barker RA. Current status of clinical trials of neural transplantation in Parkinson’s disease. Prog. Brain Res. 2012; 200, 169-98. CrossRef

45. Kadoshima T, Sakaguchi H, Nakano T et al. Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell-derived neocortex. Proc. Natl. Acad. Sci. USA 2013; 110(50), 20284-9. CrossRef

46. Suga H, Kadoshima T, Minaguchi M et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature 2011; 480(7375), 57-62. CrossRef

47. Nakano T, Ando S, Takata N et al. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell 2012; 10(6), 771–85. CrossRef

48. Sakaguchi H, Kadoshima T, Soen M et al. Generation of functional hippocampal neurons from self-organizing human embryonic stem cell-derived dorsomedial telencephalic tissue. Nat. Commun. 2015; 6, 8896. CrossRef

49. Nguyen HN, Byers B, Cord B et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 201; 8(3), 267–80. CrossRef

50. Israel MA, Yuan SH, Bardy C et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012; 482(7384), 216–20. CrossRef

51. Egawa N, Kitaoka S, Tsukita K et al. Drug screening for ALS using patient-specific induced pluripotent stem cells. Sci. Transl. Med. 2012; 4(145), 145ra104. CrossRef

52. Brennand KJ, Simone A, Jou J et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature 2011; 473(7346), 221–5. CrossRef

53. Lee JK, Jin HK, Bae JS. Bone marrow-derived mesenchymal stem cells attenuate amyloid beta-induced memory impairment and apoptosis by inhibiting neuronal cell death. Curr. Alzheimer Res. 2010; 7, 540–8. CrossRef

54. Blurton-Jones M, Kitazawa M, Martinez-Coria H et al. Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc. Natl. Acad. Sci. USA 2009; 106(32), 13594–9. CrossRef

55. Bachoud-Levi AC, Gaura V, Brugie` res P et al. Effect of fetal neural transplants in patients with Huntington’s disease 6 years after surgery: a long-term follow-up study. Lancet Neurol. 2006; 5, 303–9. CrossRef

56. Glass JD, Boulis NM, Johe K et al. Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: results of a phase I trial in 12 patients. Stem Cells 2012; 30, 1144–51. CrossRef


Affiliations

Jun Takahashi MD, PhD
Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University,
53 Shogoin kawahara-cho,
Sakyo-ku, Kyoto 606-8507, Japan
Tel: #81-75-366-7052
Fax: #81-75-366-7071
jbtaka@cira.kyoto-u.ac.jp




Creative Commons License

This work is licensed under a Creative Commons Attribution- NonCommercial – NoDerivatives 4.0 International License.