IAP-Based Cell Sorting Results in Homogeneous Transplantable Dopaminergic Precursor Cells Derived from Human Pluripotent Stem Cells

Daniela Lehnen, Serena Barral, Tiago Cardoso, Shane Grealish, Andreas Heuer, Andrej Smiyakin, Agnete Kirkeby, Jutta Kollet, Harold Cremer, Malin Parmar, Andreas Bosio, Sebastian Knöbel, Daniela Lehnen, Serena Barral, Tiago Cardoso, Shane Grealish, Andreas Heuer, Andrej Smiyakin, Agnete Kirkeby, Jutta Kollet, Harold Cremer, Malin Parmar, Andreas Bosio, Sebastian Knöbel

Abstract

Human pluripotent stem cell (hPSC)-derived mesencephalic dopaminergic (mesDA) neurons can relieve motor deficits in animal models of Parkinson's disease (PD). Clinical translation of differentiation protocols requires standardization of production procedures, and surface-marker-based cell sorting is considered instrumental for reproducible generation of defined cell products. Here, we demonstrate that integrin-associated protein (IAP) is a cell surface marker suitable for enrichment of hPSC-derived mesDA progenitor cells. Immunomagnetically sorted IAP+ mesDA progenitors showed increased expression of ventral midbrain floor plate markers, lacked expression of pluripotency markers, and differentiated into mature dopaminergic (DA) neurons in vitro. Intrastriatal transplantation of IAP+ cells sorted at day 16 of differentiation in a rat model of PD resulted in functional recovery. Grafts from sorted IAP+ mesDA progenitors were more homogeneous in size and DA neuron density. Thus, we suggest IAP-based sorting for reproducible prospective enrichment of mesDA progenitor cells in clinical cell replacement strategies.

Keywords: CD47; IAP; PD; Parkinson's disease; cell replacement therapy; dopaminergic; floor plate; immunomagnetic sorting (MACS); mesDA progenitor cells; midbrain; regenerative medicine.

Copyright © 2017 Miltenyi Biotec GmbH. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Identification of IAP as a Cell Surface Marker Expressed in FOXA2+ mesDA Progenitor Cells (A) mesDA were differentiated according to the protocol of Kirkeby et al. (2012a). Cells were harvested for a flow-cytometry-based surface marker screening on d11 and d16. AA, ascorbic acid; FN, fibronectin; lam, laminin; MN, MACS Neuro medium; NB-21, NeuroBrew-21; PO, poly-L-ornithine. (B) hESCs (H9) and hiPSCs (hFF-iPS) were differentiated toward mesDA progenitor cells and screened for marker expression on d11 and d16 of differentiation. Surface markers expressed on >90% of the FOXA2+ mesDA progenitor cells are depicted in the Edwards-Venn diagram (Bardou et al., 2014); see also Table S5. Twelve surface markers were concomitantly expressed on d11 and d16 in both hESC and hiPSC-derived FOXA2+ cells. (C) Comparative analysis of the 12 surface markers expressed in hESCs and hiPSCs at d11 and d16 of differentiation. Shown is the ratio of the mean fluorescence intensity (MFI) for each marker for FOXA2+ and FOXA2− cells. IAP displayed the highest discrimination between FOXA2+ and FOXA2− cells on hiPSCs and hESCs at d11 and d16. (D) Schematic illustration of the gating strategy used for the cell surface marker screening. Single cells were distinguished by the FSC properties, and cells of interest were gated based on FSC/SSC characteristics. As shown for IAP, surface markers expressed by mesDA progenitors were identified based on the co-staining with FOXA2. See also Figure S1. (E) Immunofluorescence staining of mesDA progenitor cells on d11 showed co-expression of IAP (red) and FOXA2 (green); Cell nuclei were stained with DAPI (blue). Scale bar represents 100 μm.
Figure 2
Figure 2
Characterization of IAP Expression In Vitro and In Vivo (A and B) Flow cytometric analysis of IAP positive and negative cells on (A) d11 or (B) d16 of differentiation with respect to expression of known markers for pluripotent, neural (stem) cells. Bar graph shows the frequency of each marker expressed by the IAP+ and IAP− subsets for differentiated hESCs and hiPSCs as the mean of independent triplicates (mean ± SD). See also Figure S2. (C) Protein expression in the neural tube of the human fetal mesencephalon (7 weeks p.c.). FOXA2 (red) is expressed within the floor plate and basal plate, LMX1A (blue) delineates the floor plate and the roof plate in human VM. IAP (green) expression was observed in the floor plate as well as in the roof plate. Cell nuclei stained with DAPI are shown in white. Scale bar represents 500 μm in the overview and 50 μm in close up. (D) hiPSCs were differentiated toward floor plate, basal plate, alar plate, and roof plate fates, respectively. Color code indicates floor plate (black), basal plate (gray), alar plate (blue), and roof plate (white). Cells differentiated for 11 days in vitro were analyzed for IAP and FOXA2 expression using flow cytometry. Bar graphs illustrate the frequency (%) of FOXA2+ cells in the IAP+ fraction (n = 3; independent experiments; mean ± SD). Representative density plots are depicted for each differentiation condition.
Figure 3
Figure 3
Purification and Characterization of IAP+ mesDA Progenitor Cells (A) The frequency of IAP+ (d11) in the unsorted fraction accounted for 53% (left blot). After the immunomagnetic sorting, IAP+ cells were enriched to a purity of 98%. (B) Sorting efficiencies of IAP+ cells derived from d11 and d16 of differentiation measured by flow cytometry. Gray symbols indicate differentiated hESCs and black symbols represent differentiated hiPSCs (d11 n = 20; d16 n = 7; independent experiments; mean ± SD; ∗∗∗p < 0.005 according to Student's t test). (C) Immunofluorescence analysis of the unsorted and IAP+ cell fraction. LMX1A (green) and FOXA2 (red). LMX1A+/FOXA2+ cells are shown in the merged picture. In contrast to the unsorted cells, the IAP+ cells reveal homogeneous FOXA2/LMX1A co-expression 3 days after replating. Scale bar represents 100 μm. (D) Gene expression analysis for FOXA2, LMX1A, and CORIN on the unsorted, IAP−, and IAP+ cells by qRT-PCR. Values are shown as the relative expression of each gene compared with the unsorted fraction (n = 3; independent experiments; mean ± SD; ∗p < 0.05; ∗∗p < 0.005; according to Student's t test). (E) IAP+ versus IAP− cells sorted on d16 of differentiated hESCs and hiPSCs. The color represents log2 intensity values centered to the gene-wise median of all samples; saturation limits are indicated in the green-black-red color bar above the heatmap. The sample groups are depicted by orange/yellow color bars above the heatmap. Shown is a selection of significantly enriched pathways associated with the candidate genes (see color bars on the right-hand side of the heatmap). See also Figure S3. (F) Gene expression trends of hESC-derived mesDA progenitors d16 unsorted, IAP+, and IAP− samples are summarized in scatterplots. The unfiltered normalized log2 intensities of IAP− versus IAP+ cells are compared. The color code corresponds to the median values of the normalized log2 intensities of independent experiments of the unsorted fraction.
Figure 4
Figure 4
Gene Expression and Pathway Analysis of d16 Sorted IAP−/+ Fractions and d16 IAP+ Cells Terminally Differentiated until d50 (A) Immunofluorescence staining of differentiated hESCs, positively sorted for IAP on d16 and replated for terminal differentiation until d50. Mature dopaminergic neurons are detected by TH (green) and TUJ1 (red) co-staining (shown as merged picture). Cell nuclei were stained with DAPI (blue). Scale bar represents 100 μm. (B) Gene expression analysis (qRT-PCR) of differentiated hESCs at d50. Sorting was performed at d16 of differentiation of hESCs, and unsorted, IAP−, IAP+ cell fractions were replated for terminal differentiation. Shown is the mRNA expression of selected genes within the IAP+ and the IAP− fractions relative to the expression of the unsorted fraction (n = 3; independent experiments; mean ± SD; ∗p < 0.05; ∗∗p < 0.005; ∗∗∗p < 0.0005). See also Figure S4. (C) IAP+ cells sorted on d16 versus IAP+ cells sorted on d16 and subsequently differentiated until d50. The color represents log2 intensity values centered to the gene-wise median of all samples; saturation limits are indicated in the green-black-red color bar above the heatmap. The sample groups are depicted by blue color bars above the heatmaps. Shown is a selection of significantly enriched pathways associated with the candidate genes (see color bars on the right-hand side of the heatmaps). (D) The d16 sorted IAP+ sample set versus IAP+ cells sorted on d16 and subsequently differentiated until d50. The color code corresponds to the log2-fold change values of adult human dopaminergic neurons relative to human embryonic NSCs (GEO: GSE25931) (Marei et al., 2011).
Figure 5
Figure 5
Six Week Survival of Unsorted versus IAP+ hiPSC-Derived mesDA Progenitors Grafted to the Striatum Schematic experimental setup (A) and histological analysis of grafts (B–G) 6 weeks after intrastriatal transplantation. Cells were either transplanted on d11 (no grafts detected) or d16 immediately after IAP-based immunomagnetic enrichment (D and G) or sorted on d11, replated and transplanted at d16 (C and F), or transplanted on d16 without sorting (B and E). Immunohistochemistry for hNCAM (B–D) and TH (E–G) revealed that sorted cells gave rise to smaller grafts (I) with overall lower TH cell (H) count but higher TH density (J). Transplanted animals: unsorted, n = 4; sorted d11 tpl d16, n = 4; sorted d16, n = 5; mean ± SD; ∗p < 0.05 according to one-way ANOVA followed by Bonferroni post hoc test; using IBM SPPS Statistics. Scale bar represents 500 μm in (B–G) and 50 μm in close-up box (E–G). tpl, transplantation.
Figure 6
Figure 6
Functional Potency Assessment of Unsorted versus IAP+ hESC-Derived mesDA Progenitors Grafted to the Striatum Schematic experimental setup (A) and histological analysis of grafts (hNCAM: B, D; TH: C, E) 18 weeks after intrastriatal transplantation. Transplantation of immunomagnetically enriched d16 IAP+ mesDA progenitors yielded grafts with higher TH density than from unsorted control cells (F), see also Figure S5. Proportion of TH+ neurons coexpressing GIRK2 and Calbindin was comparable (G) the number of 5-HT+ cells was lower in grafts form IAP+ cells (H). IAP+ progenitors like the controls led to complete functional recovery in an amphetamine-induced rotation assay 18 weeks after grafting (I). Transplanted animals: unsorted, n = 3; sorted, n = 3; mean ± SD. Scale bar represents 500 μm in (B–E) and 50 μm in close-up box (C and E).

References

    1. Aguila J.C., Blak A., van Arensbergen J., Sousa A., Vazquez N., Aduriz A., Gayosso M., Lopez Mato M.P., Lopez de Maturana R., Hedlund E. Selection based on FOXA2 expression is not sufficient to enrich for dopamine neurons from human pluripotent stem cells. Stem Cells Transl. Med. 2014;3:1032–1042.
    1. Bardou P., Mariette J., Escudie F., Djemiel C., Klopp C. jvenn: an interactive Venn diagram viewer. BMC Bioinformatics. 2014;15:293.
    1. Barker R.A., Barrett J., Mason S.L., Bjorklund A. Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson's disease. Lancet Neurol. 2013;12:84–91.
    1. Barker R.A., Drouin-Ouellet J., Parmar M. Cell-based therapies for Parkinson disease-past insights and future potential. Nat. Rev. Neurol. 2015;11:492–503.
    1. Barral S., Ecklebe J., Tomiuk S., Tiveron M.C., Desoeuvre A., Eckardt D., Cremer H., Bosio A. Efficient neuronal in vitro and in vivo differentiation after immunomagnetic purification of mESC derived neuronal precursors. Stem Cell Res. 2013;10:133–146.
    1. Bosio A., Huppert V., Donath S., Hennemann P., Malchow M., Heinlein U.A. Isolation and enrichment of stem cells. Adv. Biochem. Eng. Biotechnol. 2009;114:23–72.
    1. Brederlau A., Correia A.S., Anisimov S.V., Elmi M., Paul G., Roybon L., Morizane A., Bergquist F., Riebe I., Nannmark U. 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;24:1433–1440.
    1. Bryson J.B., Machado C.B., Crossley M., Stevenson D., Bros-Facer V., Burrone J., Greensmith L., Lieberam I. Optical control of muscle function by transplantation of stem cell-derived motor neurons in mice. Science. 2014;344:94–97.
    1. Bye C.R., Jonsson M.E., Bjorklund A., Parish C.L., Thompson L.H. Transcriptome analysis reveals transmembrane targets on transplantable midbrain dopamine progenitors. Proc. Natl. Acad. Sci. USA. 2015;112:E1946–E1955.
    1. Carlsson T., Carta M., Winkler C., Bjorklund A., Kirik D. Serotonin neuron transplants exacerbate L-DOPA-induced dyskinesias in a rat model of Parkinson's disease. J. Neurosci. 2007;27:8011–8022.
    1. Despres D., Flohr T., Uppenkamp M., Baldus M., Hoffmann M., Huber C., Derigs H.G. CD34+ cell enrichment for autologous peripheral blood stem cell transplantation by use of the CliniMACs device. J. Hematother. Stem Cell Res. 2000;9:557–564.
    1. Doi D., Morizane A., Kikuchi T., Onoe H., Hayashi T., Kawasaki T., Motono M., Sasai Y., Saiki H., Gomi M. 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;30:935–945.
    1. Doi D., Samata B., Katsukawa M., Kikuchi T., Morizane A., Ono Y., Sekiguchi K., Nakagawa M., Parmar M., Takahashi J. Isolation of human induced pluripotent stem cell-derived dopaminergic progenitors by cell sorting for successful transplantation. Stem Cell Reports. 2014;2:337–350.
    1. Friling S., Andersson E., Thompson L.H., Jonsson M.E., Hebsgaard J.B., Nanou E., Alekseenko Z., Marklund U., Kjellander S., Volakakis N. Efficient production of mesencephalic dopamine neurons by Lmx1a expression in embryonic stem cells. Proc. Natl. Acad. Sci. USA. 2009;106:7613–7618.
    1. Ganat Y.M., Calder E.L., Kriks S., Nelander J., Tu E.Y., Jia F., Battista D., Harrison N., Parmar M., Tomishima M.J. Identification of embryonic stem cell-derived midbrain dopaminergic neurons for engraftment. J. Clin. Invest. 2012;122:2928–2939.
    1. Grealish S., Diguet E., Kirkeby A., Mattsson B., Heuer A., Bramoulle Y., Van Camp N., Perrier A.L., Hantraye P., Bjorklund A. 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;15:653–665.
    1. Hargus G., Cooper O., Deleidi M., Levy A., Lee K., Marlow E., Yow A., Soldner F., Hockemeyer D., Hallett P.J. Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc. Natl. Acad. Sci. USA. 2010;107:15921–15926.
    1. Huang A.M., Wang H.L., Tang Y.P., Lee E.H. Expression of integrin-associated protein gene associated with memory formation in rats. J. Neurosci. 1998;18:4305–4313.
    1. Jiang P., Lagenaur C.F., Narayanan V. Integrin-associated protein is a ligand for the P84 neural adhesion molecule. J. Biol. Chem. 1999;274:559–562.
    1. Joksimovic M., Yun B.A., Kittappa R., Anderegg A.M., Chang W.W., Taketo M.M., McKay R.D., Awatramani R.B. Wnt antagonism of Shh facilitates midbrain floor plate neurogenesis. Nat. Neurosci. 2009;12:125–131.
    1. Kefalopoulou Z., Politis M., Piccini P., Mencacci N., Bhatia K., Jahanshahi M., Widner H., Rehncrona S., Brundin P., Bjorklund A. Long-term clinical outcome of fetal cell transplantation for Parkinson disease: two case reports. JAMA Neurol. 2014;71:83–87.
    1. Kirkeby A., Grealish S., Wolf D.A., Nelander J., Wood J., Lundblad M., Lindvall O., Parmar M. Generation of regionally specified neural progenitors and functional neurons from human embryonic stem cells under defined conditions. Cell Rep. 2012;1:703–714.
    1. Kirkeby A., Nelander J., Parmar M. Generating regionalized neuronal cells from pluripotency, a step-by-step protocol. Front. Cell. Neurosci. 2012;6:64.
    1. Kirkeby A., Nolbrant S., Tiklova K., Heuer A., Kee N., Cardoso T., Ottosson D.R., Lelos M.J., Rifes P., Dunnett S.B. Predictive markers guide differentiation to improve graft outcome in clinical translation of hESC-based therapy for Parkinson's disease. Cell Stem Cell. 2017;20:135–148.
    1. Kriks S., Shim J.W., Piao J., Ganat Y.M., Wakeman D.R., Xie Z., Carrillo-Reid L., Auyeung G., Antonacci C., Buch A. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. Nature. 2011;480:547–551.
    1. La Manno G., Gyllborg D., Codeluppi S., Nishimura K., Salto C., Zeisel A., Borm L.E., Stott S.R., Toledo E.M., Villaescusa J.C. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell. 2016;167:566–580.e19.
    1. Lindvall O., Kokaia Z. Prospects of stem cell therapy for replacing dopamine neurons in Parkinson's disease. Trends Pharmacol. Sci. 2009;30:260–267.
    1. Marei H.E., Althani A., Afifi N., Michetti F., Pescatori M., Pallini R., Casalbore P., Cenciarelli C., Schwartz P., Ahmed A.E. Gene expression profiling of embryonic human neural stem cells and dopaminergic neurons from adult human substantia nigra. PLoS One. 2011;6:e28420.
    1. Metzakopian E., Lin W., Salmon-Divon M., Dvinge H., Andersson E., Ericson J., Perlmann T., Whitsett J.A., Bertone P., Ang S.L. Genome-wide characterization of Foxa2 targets reveals upregulation of floor plate genes and repression of ventrolateral genes in midbrain dopaminergic progenitors. Development. 2012;139:2625–2634.
    1. Mi Z.P., Jiang P., Weng W.L., Lindberg F.P., Narayanan V., Lagenaur C.F. Expression of a synapse-associated membrane protein, P84/SHPS-1, and its ligand, IAP/CD47, in mouse retina. J. Comp. Neurol. 2000;416:335–344.
    1. Ohnishi H., Kaneko Y., Okazawa H., Miyashita M., Sato R., Hayashi A., Tada K., Nagata S., Takahashi M., Matozaki T. Differential localization of Src homology 2 domain-containing protein tyrosine phosphatase substrate-1 and CD47 and its molecular mechanisms in cultured hippocampal neurons. J. Neurosci. 2005;25:2702–2711.
    1. Ono Y., Nakatani T., Sakamoto Y., Mizuhara E., Minaki Y., Kumai M., Hamaguchi A., Nishimura M., Inoue Y., Hayashi H. Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells. Development. 2007;134:3213–3225.
    1. Osafune K., Caron L., Borowiak M., Martinez R.J., Fitz-Gerald C.S., Sato Y., Cowan C.A., Chien K.R., Melton D.A. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat. Biotechnol. 2008;26:313–315.
    1. Politis M., Wu K., Loane C., Quinn N.P., Brooks D.J., Rehncrona S., Bjorklund A., Lindvall O., Piccini P. Serotonergic neurons mediate dyskinesia side effects in Parkinson's patients with neural transplants. Sci. Transl. Med. 2010;2:38ra46.
    1. Pruszak J., Sonntag K.C., Aung M.H., Sanchez-Pernaute R., Isacson O. Markers and methods for cell sorting of human embryonic stem cell-derived neural cell populations. Stem Cells. 2007;25:2257–2268.
    1. Rath A., Klein A., Papazoglou A., Pruszak J., Garcia J., Krause M., Maciaczyk J., Dunnett S.B., Nikkhah G. Survival and functional restoration of human fetal ventral mesencephalon following transplantation in a rat model of Parkinson's disease. Cell Transplant. 2013;22:1281–1293.
    1. Reinhold M.I., Lindberg F.P., Plas D., Reynolds S., Peters M.G., Brown E.J. In vivo expression of alternatively spliced forms of integrin-associated protein (CD47) J. Cell Sci. 1995;108(Pt 11):3419–3425.
    1. Samata B., Doi D., Nishimura K., Kikuchi T., Watanabe A., Sakamoto Y., Kakuta J., Ono Y., Takahashi J. Purification of functional human ES and iPSC-derived midbrain dopaminergic progenitors using LRTM1. Nat. Commun. 2016;7:13097.
    1. Schumm M., Lang P., Bethge W., Faul C., Feuchtinger T., Pfeiffer M., Vogel W., Huppert V., Handgretinger R. Depletion of T-cell receptor alpha/beta and CD19 positive cells from apheresis products with the CliniMACS device. Cytotherapy. 2013;15:1253–1258.
    1. Shimamura K., Hartigan D.J., Martinez S., Puelles L., Rubenstein J.L. Longitudinal organization of the anterior neural plate and neural tube. Development. 1995;121:3923–3933.
    1. Sonntag K.C., Pruszak J., Yoshizaki T., van Arensbergen J., Sanchez-Pernaute R., Isacson O. Enhanced yield of neuroepithelial precursors and midbrain-like dopaminergic neurons from human embryonic stem cells using the bone morphogenic protein antagonist noggin. Stem Cells. 2007;25:411–418.
    1. Steinbeck J.A., Choi S.J., Mrejeru A., Ganat Y., Deisseroth K., Sulzer D., Mosharov E.V., Studer L. Optogenetics enables functional analysis of human embryonic stem cell-derived grafts in a Parkinson's disease model. Nat. Biotechnol. 2015;33:204–209.
    1. Tabar V., Studer L. Pluripotent stem cells in regenerative medicine: challenges and recent progress. Nat. Rev. Genet. 2014;15:82–92.
    1. Verney C., Zecevic N., Puelles L. Structure of longitudinal brain zones that provide the origin for the substantia nigra and ventral tegmental area in human embryos, as revealed by cytoarchitecture and tyrosine hydroxylase, calretinin, calbindin, and GABA immunoreactions. J. Comp. Neurol. 2001;429:22–44.
    1. Villaescusa J.C., Arenas E. Transplantable midbrain dopamine neurons: a moving target. Exp. Neurol. 2010;222:173–178.
    1. Wood H.B., Episkopou V. Comparative expression of the mouse Sox1, Sox2 and Sox3 genes from pre-gastrulation to early somite stages. Mech. Dev. 1999;86:197–201.

Source: PubMed

3
Abonnieren