Induction of human neuronal cells by defined transcription factors

Zhiping P Pang, Nan Yang, Thomas Vierbuchen, Austin Ostermeier, Daniel R Fuentes, Troy Q Yang, Ami Citri, Vittorio Sebastiano, Samuele Marro, Thomas C Südhof, Marius Wernig, Zhiping P Pang, Nan Yang, Thomas Vierbuchen, Austin Ostermeier, Daniel R Fuentes, Troy Q Yang, Ami Citri, Vittorio Sebastiano, Samuele Marro, Thomas C Südhof, Marius Wernig

Abstract

Somatic cell nuclear transfer, cell fusion, or expression of lineage-specific factors have been shown to induce cell-fate changes in diverse somatic cell types. We recently observed that forced expression of a combination of three transcription factors, Brn2 (also known as Pou3f2), Ascl1 and Myt1l, can efficiently convert mouse fibroblasts into functional induced neuronal (iN) cells. Here we show that the same three factors can generate functional neurons from human pluripotent stem cells as early as 6 days after transgene activation. When combined with the basic helix-loop-helix transcription factor NeuroD1, these factors could also convert fetal and postnatal human fibroblasts into iN cells showing typical neuronal morphologies and expressing multiple neuronal markers, even after downregulation of the exogenous transcription factors. Importantly, the vast majority of human iN cells were able to generate action potentials and many matured to receive synaptic contacts when co-cultured with primary mouse cortical neurons. Our data demonstrate that non-neural human somatic cells, as well as pluripotent stem cells, can be converted directly into neurons by lineage-determining transcription factors. These methods may facilitate robust generation of patient-specific human neurons for in vitro disease modelling or future applications in regenerative medicine.

Figures

Figure 1. Rapid generation of functional neurons…
Figure 1. Rapid generation of functional neurons from human ES cells
a, Four days after induction, ES-iN cells exhibited bipolar neuronal morphologies. b–c, Eight days after induction, ES-iN cells expressed Tuj1 (b) and MAP2 (c). d, Spontaneous action potentials presumably caused by membrane potential fluctuations recorded from an ES-iN cell 6 days after induction. Arrow: pronounced AHP. e, Representative traces of action potentials in response to step current injections 15 days after induction. Membrane potential was maintained at ~ –63mV. f, Quantification of intrinsic membrane properties in control ES cells (0 day) before and after viral transduction. membrane input resistance (Rin), resting membrane potential (RMP), capacitance (Cm), after hyperpolarization potentials (AHP). Scale bars: 10µm (a,b,c). Numbers of cells recorded are labeled in the bars. Note the heterogeneity of the parameters (see also Suppl. Fig. 1). Data are presented with mean±SEM. * p<0.05.
Figure 2. NeuroD1 increases reprogramming efficiency in…
Figure 2. NeuroD1 increases reprogramming efficiency in primary human fetal fibroblasts
a, Quantification of Tuj1-positive BAM-iN cells with indicated factors, 3 weeks after dox. b–c, Three weeks after dox BAM+NeuroD1 iN cells exhibited neuronal morphologies (b) and expressed Tuj1 (c) . d–f, iN cells expressed NeuN (d,) PSA-NCAM (e), and MAP2 (f) 2 weeks after dox. g–h, An iN cell expressing MAP2 (g) and synapsin (h) 4 weeks after dox and co-cultured with primary astrocytes. i, Single cell gene expression profiling using Fluidigm dynamic arrays. Rows represent the evaluated genes and columns represent individual cells. Heatmap (blue to red) represents the threshold Ct values as indicated. Data in (a) are presented as mean±SD. Scale bars: 100 µm (b, c), 10 µm (d–h).
Figure 3. Membrane properties of fibroblast iN…
Figure 3. Membrane properties of fibroblast iN cells
a, Quantification of Tuj1-positive neuronal cells from HFFs (line HFF-A) 3 weeks after dox or HPFs (line HPF-B) 4 weeks after dox. N=3 independent experiments. b, Patch clamp recording was conducted on HFF-iN cells identified by EGFP fluorescence and DIC microcopy. c, Representative traces of membrane potentials in response to step current injections (lower panel) from an HFF-iN cell 19 days after dox. Membrane potential was maintained at ~ −63 mV. d, Representative traces of membrane currents recorded with a ramp protocol (lower panel). Fast activating and inactivating Na+ currents were prominent. Three traces are shown superimposed. e–g, HPF-iN cells express Tuj1 (red) and NeuN (green) (e), Neurofilament (green) (f) and MAP2 (green) (g). h, Representative traces of membrane potentials in response to step current injections in HPF-iN cells. Action potentials were generated in cultures without glia. i, Representative traces of membrane currents recorded following a ramp protocol (lower panel) in HPF-iN cells. The Na+ currents could be blocked by TTX. Data in (a) are presented as mean±SD. Scale bars: 10 µm (a, e–g).
Figure 4. Synaptic responses of HFF-iN cells
Figure 4. Synaptic responses of HFF-iN cells
a, An HFF-iN cell expressing EGFP co-cultured with mouse cortical neurons at day 35 after dox. b, Synapsin positive puncta co-localize with neurites extending from HFF-iN cells (arrow heads). c, Thirty-five days after dox, spontaneous PSCs were recorded in HFF-iN cells. d, The slow responses could be blocked by PTX. Bursting events of EPSCs were recorded in the presence of PTX. The insert shows the fast kinetics of the responses. e, In the presence of PTX and CNQX (50 µM), no spontaneous activities were observed. f, Evoked postsynaptic responses. Four traces were super imposed. Sti. = stimulation. g, In the presence of PTX, electric stimulation evoked fast-kinetic excitatory PSCs (EPSCs). h, No evoked synaptic responses were observed in the presence of PTX and CNQX. Scale bars: 100 µm (a); 10µm (b).

References

    1. Blau HM, et al. Plasticity of the differentiated state. Science. 1985;230(4727):758–766.
    1. Gurdon JB. From nuclear transfer to nuclear reprogramming: the reversal of cell differentiation. Annu Rev Cell Dev Biol. 2006;22:1–22.
    1. Heins N, et al. Glial cells generate neurons: the role of the transcription factor Pax6. Nat Neurosci. 2002;5(4):308–315.
    1. Ieda M, et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010;142(3):375–386.
    1. Shen CN, Slack JM, Tosh D. Molecular basis of transdifferentiation of pancreas to liver. Nat Cell Biol. 2000;2(12):879–887.
    1. Tada M, Takahama Y, Abe K, Nakatsuji N, Tada T. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr Biol. 2001;11(19):1553–1558.
    1. Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872.
    1. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian cells. Nature. 1997;385(6619):810–813.
    1. Xie H, Ye M, Feng R, Graf T. Stepwise reprogramming of B cells into macrophages. Cell. 2004;117(5):663–676.
    1. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455(7213):627–632.
    1. Graf T, Enver T. Forcing cells to change lineages. Nature. 2009;462(7273):587–594.
    1. Zhou Q, Melton DA. Extreme makeover: converting one cell into another. Cell Stem Cell. 2008;3(4):382–388.
    1. Vierbuchen T, et al. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010;463(7284):1035–1041.
    1. Hansen DV, Lui JH, Parker PR, Kriegstein AR. Neurogenic radial glia in the outer subventricular zone of human neocortex. Nature. 2010;464(7288):554–561.
    1. Kriegstein A, Noctor S, Martinez-Cerdeno V. Patterns of neural stem and progenitor cell division may underlie evolutionary cortical expansion. Nat Rev Neurosci. 2006;7(11):883–890.
    1. Zhang X, et al. Pax6 is a human neuroectoderm cell fate determinant. Cell Stem Cell. 2010;7(1):90–100.
    1. Bottenstein JE, Sato GH. Growth of a rat neuroblastoma cell line in serum-free supplemented medium. Proc Natl Acad Sci U S A. 1979;76(1):514–517.
    1. Guo G, et al. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675–685.
    1. Johnson MA, Weick JP, Pearce RA, Zhang SC. Functional neural development from human embryonic stem cells: accelerated synaptic activity via astrocyte coculture. J Neurosci. 2007;27(12):3069–3077.
    1. Wu H, et al. Integrative genomic and functional analyses reveal neuronal subtype differentiation bias in human embryonic stem cell lines. Proc Natl Acad Sci U S A. 2007;104(34):13821–13826.
    1. Koch P, Opitz T, Steinbeck JA, Ladewig J, Brustle O. A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. Proc Natl Acad Sci U S A. 2009;106(9):3225–3230.
    1. Marchetto MC, et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell. 2010;143(4):527–539.
    1. Hu BY, et al. Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc Natl Acad Sci U S A. 2010;107(9):4335–4340.
    1. Xu Y, et al. Revealing a core signaling regulatory mechanism for pluripotent stem cell survival and self-renewal by small molecules. Proc Natl Acad Sci U S A. 2010;107(18):8129–8134.
    1. Maximov A, Pang ZP, Tervo DG, Sudhof TC. Monitoring synaptic transmission in primary neuronal cultures using local extracellular stimulation. J Neurosci Methods. 2007;161(1):75–87.
    1. Wong CC, et al. Non-invasive imaging of human embryos before embryonic genome activation predicts development to the blastocyst stage. Nat Biotechnol. 2010;28(10):1115–1121.

Source: PubMed

3
Abonnere