Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease

Sonja Kriks, Jae-Won Shim, Jinghua Piao, Yosif M Ganat, Dustin R Wakeman, Zhong Xie, Luis Carrillo-Reid, Gordon Auyeung, Chris Antonacci, Amanda Buch, Lichuan Yang, M Flint Beal, D James Surmeier, Jeffrey H Kordower, Viviane Tabar, Lorenz Studer, Sonja Kriks, Jae-Won Shim, Jinghua Piao, Yosif M Ganat, Dustin R Wakeman, Zhong Xie, Luis Carrillo-Reid, Gordon Auyeung, Chris Antonacci, Amanda Buch, Lichuan Yang, M Flint Beal, D James Surmeier, Jeffrey H Kordower, Viviane Tabar, Lorenz Studer

Abstract

Human pluripotent stem cells (PSCs) are a promising source of cells for applications in regenerative medicine. Directed differentiation of PSCs into specialized cells such as spinal motoneurons or midbrain dopamine (DA) neurons has been achieved. However, the effective use of PSCs for cell therapy has lagged behind. Whereas mouse PSC-derived DA neurons have shown efficacy in models of Parkinson's disease, DA neurons from human PSCs generally show poor in vivo performance. There are also considerable safety concerns for PSCs related to their potential for teratoma formation or neural overgrowth. Here we present a novel floor-plate-based strategy for the derivation of human DA neurons that efficiently engraft in vivo, suggesting that past failures were due to incomplete specification rather than a specific vulnerability of the cells. Midbrain floor-plate precursors are derived from PSCs 11 days after exposure to small molecule activators of sonic hedgehog (SHH) and canonical WNT signalling. Engraftable midbrain DA neurons are obtained by day 25 and can be maintained in vitro for several months. Extensive molecular profiling, biochemical and electrophysiological data define developmental progression and confirm identity of PSC-derived midbrain DA neurons. In vivo survival and function is demonstrated in Parkinson's disease models using three host species. Long-term engraftment in 6-hydroxy-dopamine-lesioned mice and rats demonstrates robust survival of midbrain DA neurons derived from human embryonic stem (ES) cells, complete restoration of amphetamine-induced rotation behaviour and improvements in tests of forelimb use and akinesia. Finally, scalability is demonstrated by transplantation into parkinsonian monkeys. Excellent DA neuron survival, function and lack of neural overgrowth in the three animal models indicate promise for the development of cell-based therapies in Parkinson's disease.

Figures

Figure 1. Induction and neurogenic conversion of…
Figure 1. Induction and neurogenic conversion of hESC-derived midbrain FP precursors is dependent on CHIR99021 addition
a) Immunocytochemistry at day 11 for FOXA2 (red), NESTIN (green, upper panels), LMX1A (green, middle panels) and OTX2 (green, lower panels). b,c) Quantification of the data presented in (a); mean ± SEM, n=3 (independent experiments): *** p < 0.001; ** p < 0.01; p < 0.05 (compared to LSB, Dunnett test). d) Diagram of culture conditions. e,f) Selected lists of differentially expressed transcripts at day 11 comparing LSB/S/F8/CHIR versus LSB (e) or versus LSB/S/F8 (f). g,h) Temporal gene expression analysis for markers of midbrain DA precursor (g), forebrain and ventral non-DA precursor identity (h). Scale bars: 50 μm.
Figure 2
Figure 2
Immunocytochemical and molecular analysis of midbrain DA neuron fate in LSB/S/F8/CHIR treated versus LSB/S/F8 (hypothalamic) and forebrain LSB (dorsal forebrain) fates. a) Immunocytochemistry at day 25 for co-expression of FOXA2 (blue) with Tuj1(red)/LMX1A(green) (upper panels) and NURR1(red)/TH(green) (lower panels). b) Quantitative co-expression analysis for LSB/S/F8/CHIR; mean ± SEM, n=3 (independent experiments). c,d) Global gene expression analysis at day 25 (triplicates each). Selected lists of differentially expressed transcripts comparing day 13 versus day 25 in LSB/S/F8/CHIR (c) LSB/S/F8/CHIR versus LSB (d, left panel) and LSB/S/F8 (d, right panel). e) Gene expression analysis for key midbrain DA neuron markers. Significance compared to LSB: Dunnett test: *** p < 0.001; ** p < 0.01; * p < 0.05). Scale bars: 50 μm.
Figure 3. In vitro maturation and functional…
Figure 3. In vitro maturation and functional characterization of FP versus rosette-derived midbrain DA neurons
a) Immunocytochemistry at day 50 for TH (red), with LMX1A (green) and FOXA2 (blue; left panels) and NURR1 (green, right panels). b) Quantification of TH+, FOXA2+, LMX1+ and NURR1+ cells in rosette- versus FP-derived (LSB/S/F8/CHIR) cultures. c) Quantification of serotonin+ (5-HT), and GABA+ neuronal subtypes at day 50 in rosette-versus FP-derived cultures. d,e) HPLC analysis for DA and metabolites d) Representative HPLC chromatogram in a sample of FP-derived cultures. e) Levels of DA, DOPAC and HVA in FP-and rosette-derived cultures. f) Immunocytochemistry in FP-derived cultures (day 80) for TH (red) and synapsin (green). g-i) Electrophysiological analyses of FP cultures at day 80. Phase contrast image of a patched neuron (g) and corresponding recordings (h). i) Power analysis showing membrane potential oscillations characteristic of DA neuron identity (2~5Hz) Mean ± SEM; significance (panels b, c, e) comparing FP versus rosette-derived cultures: Student’s T-test: *** p < 0.001; ** p < 0.01; p < 0.05). Scale bars: 50 μm in (a), 20 μm in (f, upper panel), 5 μm in (f, lower panel) and 20 μm in (g)
Figure 4. In vivo survival and function…
Figure 4. In vivo survival and function of FP-derived human DA neurons in mouse, rat and monkey PD hosts
a-d) 6-OHDA lesioned adult mice (NOD-SCID IL2Rgc null strain): a) TH expression and graft morphology at 4.5 months after transplantation. b) Expression of human specific marker (hNCAM, blue), TH (green), and FOXA2 (red). c) Quantification of FOXA2+ and TH+ cells in FP-derived grafts (mean ± SEM, n=4 at 4.5 months post grafting). d) Amphetamine-induced rotation analysis in FP- (blue) versus rosette-derived (green) grafts. Scale bars: 500 μm in (a), and 100 and 40 μm in (b). e-p) 6-OHDA lesioned adult rats: Immunohistochemistry for TH (green) and human specific markers (red) hNA (e) and hNCAM (f). g) Stereological quantification of hNA+, TH+ and TH+ cells co-expressing FOXA2 (average graft volume = 2.6 ± 0.6 mm3). h-j) Co-expression of TH (green) with FOXA2, PITX3 and NURR1 (red). k-m) Behavioral analysis in FP- versus sham-grafted animals. k) Amphetamine-induced rotational asymmetry. l) stepping test: measuring forelimb akinesia in affected versus non-affected side. m) Cylinder test: measuring ipsi- versus contra-lateral paw preference. Grafted animals showed significant improvement in all three tests (p < 0.01 at 4.5–5 month; n=4–6 each). n-p) Immunohistochemistry for TH (green) and co-expression (red) with DAT (n), GIRK2 (o) and calbindin (p). Significance levels (panels d, k, l, m): ** p < 0.01; p < 0.05). Scale bars: 200 μm in (e), 50 μm in (f), 20 μm in (h-j) and 40 μm in (n-p). q-t) Adult MPTP lesioned rhesus monkeys. q) Representative graft at 1 month after transplantation expressing human specific cytoplasm marker SC-121 (green). r) TH expression in graft with surrounding TH+ fibers (arrows). s) Co-expression of SC-121 (red) and TH (green). t) Co-expression of FOXA2 (red) and TH+ (green). Scale bars: 2mm for (q), 500 μm for (r), 200 μm for (s), and 50 μm for (t).

References

    1. Li XJ, et al. Specification of motoneurons from human embryonic stem cells. Nat Biotechnol. 2005;23:215–221.
    1. Perrier AL, et al. From the Cover: Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci U S A. 2004;101:12543–8.
    1. Tabar V, et al. Therapeutic cloning in individual Parkinsonian mice. Nature Med. 2008;14:379–381.
    1. Wernig M, et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson’s disease. Proc Natl Acad Sci U S A. 2008;105:5856–5861.
    1. Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders--time for clinical translation? J Clin Invest. 2010;120:29–40.
    1. Elkabetz Y, et al. Human ES cell-derived neural rosettes reveal a functionally dinstinct early neural stem cell stage. Genes Dev. 2008;22:152–165.
    1. Roy NS, et al. Functional engraftment of human ES cell-derived dopaminergic neurons enriched by coculture with telomerase-immortalized midbrain astrocytes. Nature Med. 2006;12:1259–1268.
    1. Kittappa R, Chang WW, Awatramani RB, McKay RD. The foxa2 gene controls the birth spontaneous degeneration of dopamine neurons in old age. PLoS Biol. 2007;5:e325.
    1. Ferri AL, et al. Foxa1 and Foxa2 regulate multiple phases of midbrain dopaminergic neuron development in a dosage-dependent manner. Development. 2007;134:2761–2769.
    1. Roelink H, et al. Floor plate and motor neuron induction by vhh-1, a vertebrate homolog of hedgehog expressed by the notochord. Cell. 1994;76:761–775.
    1. Liem KF, Tremml G, Roelink H, Jessell TM. Dorsal differentiation of neural plate cells induced by BMP-mediated signals from epidermal ectoderm. Cell. 1995;82:969–979.
    1. Fasano CA, Chambers SM, Lee G, Tomishima MJ, Studer L. Efficient derivation of functional floor plate tissue from human embryonic stem cells. Cell Stem Cell. 2010;6:336–347.
    1. Chambers SM, et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275–280.
    1. Muroyama Y, Fujihara M, Ikeya M, Kondoh H, Takada S. Wnt signaling plays an essential role in neuronal specification of the dorsal spinal cord. Genes Dev. 2002;16:548–553.
    1. Joksimovic M, et al. Wnt antagonism of Shh facilitates midbrain floor plate neurogenesis. Nat Neurosci. 2009;12:125–131.
    1. Lyashenko N, et al. Differential requirement for the dual functions of beta-catenin in embryonic stem cell self-renewal and germ layer formation. Nat Cell Biol. 2011;13:753–761.
    1. VanDunk C, Hunter LA, Gray PA. Development, maturation, and necessity of transcription factors in the mouse suprachiasmatic nucleus. J Neurosci. 2011;31:6457–6467.
    1. Huang dW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57.
    1. Costa RH, Grayson DR, Darnell JE., Jr Multiple hepatocyte-enriched nuclear factors function in the regulation of transthyretin and alpha 1-antitrypsin genes. Mol Cell Biol. 1989;9:1415–1425.
    1. Soldner F, et al. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009;136:964–977.
    1. Guzman JN, Sanchez-Padilla J, Chan CS, Surmeier DJ. Robust pacemaking in substantia nigra dopaminergic neurons. J Neurosci. 2009;29:11011–11019.
    1. Nedergaard S, Flatman JA, Engberg I. Nifedipine- and omega-conotoxin-sensitive Ca2+ conductances in guinea-pig substantia nigra pars compacta neurones. J Physiol. 1993;466:727–747.
    1. Ferrari D, Sanchez-Pernaute R, Lee H, Studer L, Isacson O. Transplanted dopamine neurons derived from primate ES cells preferentially innervate DARPP-32 striatal progenitors within the graft. Eur J Neurosci. 2006;24:1885–1896.
    1. Olanow CW, Kordower JH, Freeman TB. Fetal nigral transplantation as a therapy for Parkinson’s disease. Trends Neurosci. 1996;19:102–109.
    1. Zetterström RH, et al. Dopamine neuron agenesis in Nurr1-deficient mice. Science. 1997;276:248–250.
    1. Quintana E, et al. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–598.
    1. Kim H, et al. miR-371-3 Expression Predicts Neural Differentiation Propensity in Human Pluripotent Stem Cells. Cell Stem Cell. 2011;8:695–706.
    1. Hargus G, et al. Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:15921–15926.
    1. Aubry L, et al. Striatal progenitors derived from human ES cells mature into DARPP32 neurons in vitro and in quinolinic acid-lesioned rats. Proc Natl Acad Sci U S A. 2008;105:16707–16712.

Source: PubMed

3
Abonnieren