W. Akhtar, J. De-jong, A. V. Pindyurin, L. Pagie, W. Meuleman et al., , 2013.

S. Averaimo, A. Assali, O. Ros, S. Couvet, Y. Zagar et al., A plasma membrane microdomain compartmentalizes ephrin-generated cAMP signals to prune developing retinal axon arbors, Nat. Commun, vol.7, p.12896, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01398119

P. Batard, M. Jordan, and F. Wurm, , 2001.

C. Beard, K. Hochedlinger, K. Plath, A. Wutz, J. et al., Efficient method to generate single-copy transgenic mice by site-specific integration in embryonic stem cells, Genesis, vol.44, pp.23-28, 2006.

J. B. Black, P. Perez-pinera, and C. A. Gersbach, Mammalian synthetic biology: engineering biological systems, Annu. Rev. Biomed. Eng, vol.19, pp.249-277, 2017.

J. A. Brzezinski, L. Prasov, and T. Glaser, Math5 defines the ganglion cell competence state in a subpopulation of retinal progenitor cells exiting the cell cycle, Dev. Biol, vol.365, pp.395-413, 2012.

A. J. Cammack, A. Moudgil, T. Lagunas, J. Chen, M. J. Vasek et al., Transposonmediated, cell type-specific transcription factor recording in the mouse brain, 2019.

R. E. Campbell, O. Tour, A. E. Palmer, P. A. Steinbach, G. S. Baird et al., A monomeric red fluorescent protein, Proc. Natl. Acad. Sci. USA, vol.99, pp.7877-7882, 2002.

C. Cepko, Retrovirus vectors and their applications in neurobiology, Neuron, vol.1, pp.345-353, 1988.

F. Chen and J. Loturco, A method for stable transgenesis of radial glia lineage in rat neocortex by piggyBac mediated transposition, J. Neurosci. Methods, vol.207, pp.172-180, 2012.

F. Chen, B. J. Maher, and J. J. Loturco, piggyBac transposon-mediated cellular transgenesis in mammalian forebrain by in utero electroporation, Cold Spring Harb. Protoc, pp.741-749, 2014.

M. B. Clark, P. P. Amaral, F. J. Schlesinger, M. E. Dinger, R. J. Taft et al., The reality of pervasive transcription, PLoS Biol, vol.9, pp.1000625-1001102, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02349846

S. Ding, X. Wu, G. Li, M. Han, Y. Zhuang et al., Efficient transposition of the piggyBac (PB) transposon in mammalian cells and mice, Cell, vol.122, pp.473-483, 2005.

J. G. Doench, Am I ready for CRISPR? A user's guide to genetic screens, Nat. Rev. Genet, vol.19, pp.67-80, 2018.

S. M. Dymecki and J. C. Kim, Molecular neuroanatomy's ''three Gs'': a primer, Neuron, vol.54, pp.17-34, 2007.

M. R. Ebrahimkhani and M. Ebisuya, Synthetic developmental biology: build and control multicellular systems, Curr. Opin. Chem. Biol, vol.52, pp.9-15, 2019.

D. M. Fekete, J. Perez-miguelsanz, E. F. Ryder, and C. L. Cepko, Clonal analysis in the chicken retina reveals tangential dispersion of clonally related cells, Dev. Biol, vol.166, pp.666-682, 1994.

L. Feng, Z. H. Xie, Q. Ding, X. Xie, R. T. Libby et al., MATH5 controls the acquisition of multiple retinal cell fates, Mol. Brain, vol.3, p.36, 2010.

M. Figueres-oñ-ate, J. García-marqué-s, and L. Ló-pez-mascaraque, , 2016.

, UbC-StarTrack, a clonal method to target the entire progeny of individual progenitors, Sci. Rep, vol.6, p.33896

M. J. Fraser, T. Ciszczon, T. Elick, and C. Bauser, Precise excision of TTAA-specific lepidopteran transposons piggyBac (IFP2) and tagalong (TFP3) from the baculovirus genome in cell lines from two species of Lepidoptera, Insect Mol. Biol, vol.5, pp.141-151, 1996.

Y. Gao, E. Hisey, T. W. Bradshaw, E. Erata, W. E. Brown et al., Plug-and-play protein modification using homology-independent universal genome engineering, Neuron, vol.103, pp.583-597, 2019.

F. García-moreno, N. A. Vasistha, J. Begbie, and Z. Molná-r, CLoNe is a new method to target single progenitors and study their progeny in mouse and chick, Development, vol.141, pp.1589-1598, 2014.

J. Goedhart, D. Von-stetten, M. Noirclerc-savoye, M. Lelimousin, L. Joosen et al., , 2012.

E. Haapaniemi, S. Botla, J. Persson, B. Schmierer, and J. Taipale, , 2018.

, CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response, Nat. Med, vol.24, pp.927-930

B. P. Hafler, N. Surzenko, K. T. Beier, C. Punzo, J. M. Trimarchi et al., Transcription factor Olig2 defines subpopulations of retinal progenitor cells biased toward specific cell fates, Proc. Natl. Acad. Sci. USA, vol.109, pp.7882-7887, 2012.

B. H?-ammerle, E. Ulin, J. Guimera, W. Becker, F. Guillemot et al., Transient expression of Mnb/Dyrk1a couples cell cycle exit and differentiation of neuronal precursors by inducing p27KIP1 expression and suppressing NOTCH signaling, Development, vol.138, pp.2543-2554, 2011.

J. He, G. Zhang, A. D. Almeida, M. Cayouette, B. D. Simons et al., How variable clones build an invariant retina, Neuron, vol.75, pp.786-798, 2012.

T. Hirsch, T. Rothoeft, N. Teig, J. W. Bauer, G. Pellegrini et al., , 2017.

, Regeneration of the entire human epidermis using transgenic stem cells, Nature, vol.551, pp.327-332

I. Holguera and C. Desplan, Neuronal specification in space and time, Science, vol.362, pp.176-180, 2018.

F. Inoue, M. Kircher, B. Martin, G. M. Cooper, D. M. Witten et al., A systematic comparison reveals substantial differences in chromosomal versus episomal encoding of enhancer activity, Genome Res, vol.27, pp.38-52, 2017.

Z. Ivics, M. A. Li, L. Má-té-s, J. D. Boeke, A. Nagy et al., Transposon-mediated genome manipulation in vertebrates, Nat. Methods, vol.6, pp.415-422, 2009.

G. S. Jefferis and J. Livet, Sparse and combinatorial neuron labelling, Curr. Opin. Neurobiol, vol.22, pp.101-110, 2012.

N. Jullien, F. Sampieri, A. Enjalbert, and J. P. Herman, Regulation of Cre recombinase by ligand-induced complementation of inactive fragments, Nucleic Acids Res, vol.31, p.131, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01775207

C. J. Jung, S. Mé-noret, L. Brusselle, L. Tesson, C. Usal et al., Comparative analysis of piggyBac, CRISPR/Cas9 and TALEN mediated BAC transgenesis in the zygote for the generation of humanized SIRPA rats, Sci. Rep, vol.6, p.31455, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02150699

K. Kawakami and T. Noda, Transposition of the Tol2 element, an Aclike element from the Japanese medaka fish Oryzias latipes, in mouse embryonic stem cells, Genetics, vol.166, pp.895-899, 2004.

K. Kawakami, D. A. Largaespada, and Z. Ivics, Transposons as tools for functional genomics in vertebrate models, Trends Genet, vol.33, pp.784-801, 2017.

S. E. Klompe, P. L. Vo, T. S. Halpin-healy, and S. H. Sternberg, , 2019.

, Transposon-encoded CRISPR-Cas systems direct RNA-guided DNA integration, Nature, vol.571, pp.219-225

O. Kobiler, Y. Lipman, K. Therkelsen, I. Daubechies, and L. W. Enquist, Herpesviruses carrying a Brainbow cassette reveal replication and expression of limited numbers of incoming genomes, Nat. Commun, vol.1, p.146, 2010.

T. Kondo, K. Imamura, M. Funayama, K. Tsukita, M. Miyake et al., iPSC-based compound screening and in vitro trials identify a synergistic anti-amyloid b combination for Alzheimer's disease, Cell Rep, vol.21, pp.2304-2312, 2017.

S. F. Landrette and T. Xu, Somatic genetics empowers the mouse for modeling and interrogating developmental and disease processes, PLoS Genet, vol.7, p.1002110, 2011.

S. M. Leber and J. R. Sanes, Migratory paths of neurons and glia in the embryonic chick spinal cord, J. Neurosci, vol.15, pp.1236-1248, 1995.

X. Li, X. Zhao, Y. Fang, X. Jiang, T. Duong et al., Generation of destabilized green fluorescent protein as a transcription reporter, J. Biol. Chem, vol.273, pp.34970-34975, 1998.

M. A. Li, D. J. Turner, Z. Ning, K. Yusa, Q. Liang et al., Mobilization of giant piggyBac transposons in the mouse genome, Nucleic Acids Res, vol.39, p.148, 2011.

G. Liang and Y. Zhang, Genetic and epigenetic variations in iPSCs: potential causes and implications for application, Cell Stem Cell, vol.13, pp.149-159, 2013.

K. Loulier, R. Barry, P. Mahou, Y. Le-franc, W. Supatto et al., Multiplex cell and lineage tracking with combinatorial labels, Neuron, vol.81, pp.505-520, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01086032

F. T. Merkle, S. Ghosh, N. Kamitaki, J. Mitchell, Y. Avior et al., Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations, Nature, vol.545, pp.229-233, 2017.

T. Mikuni, J. Nishiyama, Y. Sun, N. Kamasawa, Y. et al., Highthroughput, high-resolution mapping of protein localization in mammalian brain by in vivo genome editing, Cell, vol.165, pp.1803-1817, 2016.

R. Nehme, E. Zuccaro, S. D. Ghosh, C. Li, J. L. Sherwood et al., , 2018.

, Combining NGN2 programming with developmental patterning generates human excitatory neurons with NMDAR-mediated synaptic transmission, Cell Rep, vol.23, pp.2509-2523

J. Nishiyama, T. Mikuni, Y. , and R. , Virus-mediated genome editing via homology-directed repair in mitotic and postmitotic cells in mammalian brain, Neuron, vol.96, pp.755-768, 2017.

H. Niwa, K. Yamamura, and J. Miyazaki, Efficient selection for highexpression transfectants with a novel eukaryotic vector, Gene, vol.108, pp.193-199, 1991.

S. C. Noctor, A. C. Flint, T. A. Weissman, R. S. Dammerman, and A. R. Kriegstein, Neurons derived from radial glial cells establish radial units in neocortex, Nature, vol.409, pp.714-720, 2001.

T. Pierfelice, L. Alberi, and N. Gaiano, Notch in the vertebrate nervous system: an old dog with new tricks, Neuron, vol.69, pp.840-855, 2011.

S. Pontes-quero, L. Heredia, V. Casquero-garcía, M. Ferná-ndez-chacó-n, W. Luo et al., Dual ifgMosaic: a versatile method for multispectral and combinatorial mosaic gene-function analysis, Cell, vol.170, pp.800-814, 2017.

J. Price, D. Turner, and C. Cepko, Lineage analysis in the vertebrate nervous system by retrovirus-mediated gene transfer, Proc. Natl. Acad. Sci. USA, vol.84, pp.156-160, 1987.

I. Querques, A. Mades, C. Zuliani, C. Miskey, M. Alb et al., A highly soluble Sleeping Beauty transposase improves control of gene insertion, Nat. Biotechnol, vol.37, pp.1502-1512, 2019.

A. Rebsam, T. J. Petros, and C. A. Mason, Switching retinogeniculate axon laterality leads to normal targeting but abnormal eye-specific segregation that is activity dependent, J. Neurosci, vol.29, pp.14855-14863, 2009.

A. C. Rios, O. Serralbo, D. Salgado, M. , and C. , Neural crest regulates myogenesis through the transient activation of NOTCH, Nature, vol.473, pp.532-535, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01780066

S. B. Rompani and C. L. Cepko, Retinal progenitor cells can produce restricted subsets of horizontal cells, Proc. Natl. Acad. Sci. USA, vol.105, pp.192-197, 2008.

E. Schick, S. D. Mccaffery, E. E. Keblish, C. Thakurdin, and M. Emerson,

J. Schindelin, I. Arganda-carreras, E. Frise, V. Kaynig, M. Longair et al., Fiji: an open-source platform for biological-image analysis, Nat. Methods, vol.9, pp.676-682, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-02616466

O. Serralbo, C. A. Picard, M. , and C. , Long-term, inducible gene loss-of-function in the chicken embryo, Genesis, vol.51, pp.372-380, 2013.

B. Sharma, L. Ho, G. H. Ford, H. I. Chen, A. B. Goldstone et al., , 2017.

, Alternative progenitor cells compensate to rebuild the coronary vasculature in Elabela-and Apj-deficient hearts, Dev. Cell, vol.42, pp.655-666

D. M. Shcherbakova and V. V. Verkhusha, Near-infrared fluorescent proteins for multicolor in vivo imaging, Nat. Methods, vol.10, pp.751-754, 2013.

D. Skowronska-krawczyk, F. Chiodini, M. Ebeling, C. Alliod, A. Kundzewicz et al., Conserved regulatory sequences in Atoh7 mediate nonconserved regulatory responses in retina ontogenesis, Development, vol.136, pp.3767-3777, 2009.

B. Z. Stanger, A. J. Tanaka, and D. A. Melton, Organ size is limited by the number of embryonic progenitor cells in the pancreas but not the liver, Nature, vol.445, pp.886-891, 2007.

J. Strecker, A. Ladha, Z. Gardner, J. L. Schmid-burgk, K. S. Makarova et al., RNA-guided DNA insertion with CRISPRassociated transposases, Science, vol.365, pp.48-53, 2019.

S. C. Suzuki, A. Bleckert, P. R. Williams, M. Takechi, S. Kawamura et al., Cone photoreceptor types in zebrafish are generated by symmetric terminal divisions of dedicated precursors, Proc. Natl. Acad. Sci. USA, vol.110, pp.15109-15114, 2013.

K. Suzuki, Y. Tsunekawa, R. Hernandez-benitez, J. Wu, J. Zhu et al., In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration, Nature, vol.540, pp.144-149, 2016.

M. Tewary, N. Shakiba, and P. W. Zandstra, Stem cell bioengineering: building from stem cell biology, Nat. Rev. Genet, vol.19, pp.595-614, 2018.

S. W. Wang, B. S. Kim, K. Ding, H. Wang, D. Sun et al., Requirement for math5 in the development of retinal ganglion cells, Genes Dev, vol.15, pp.24-29, 2001.

Y. Wang, J. Wang, A. Devaraj, M. Singh, A. Jimenez-orgaz et al., Suicidal autointegration of sleeping beauty and piggyBac transposons in eukaryotic cells, PLoS Genet, vol.10, p.1004103, 2014.

T. A. Weissman and Y. A. Pan, Brainbow: new resources and emerging biological applications for multicolor genetic labeling and analysis, Genetics, vol.199, pp.293-306, 2015.

M. B. Woodworth, K. M. Girskis, and C. A. Walsh, Building a lineage from single cells: genetic techniques for cell lineage tracking, Nat. Rev. Genet, vol.18, pp.230-244, 2017.

N. Yang, S. Chanda, S. Marro, Y. Ng, J. A. Janas et al., Generation of pure GABAergic neurons by transcription factor programming, Nat. Methods, vol.14, pp.621-628, 2017.

J. Yu, K. Hu, K. Smuga-otto, S. Tian, R. Stewart et al., Human induced pluripotent stem cells free of vector and transgene sequences, Science, vol.324, pp.797-801, 2009.

Y. C. Yu, S. He, S. Chen, Y. Fu, K. N. Brown et al., Preferential electrical coupling regulates neocortical lineage-dependent microcircuit assembly, Nature, vol.486, pp.113-117, 2012.

K. Yusa, L. Zhou, M. A. Li, A. Bradley, and N. L. Craig, A hyperactive piggyBac transposase for mammalian applications, Proc. Natl. Acad. Sci. USA, vol.108, pp.1531-1536, 2011.

K. Ziegler, T. Bui, R. J. Frisque, A. Grandinetti, and V. R. Nerurkar, A rapid in vitro polyomavirus DNA replication assay, J. Virol. Methods, vol.122, pp.123-127, 2004.

H. Zong, J. S. Espinosa, H. H. Su, M. D. Muzumdar, and L. Luo, Mosaic analysis with double markers in mice, Cell, vol.121, pp.479-492, 2005.

M. , with incorporation of the TTAA footprint at a silent position (Leu104) and addition of a FLAG epitope (DYKDDDDK) at the protein Ct. To limit expression of the Cre Nt fragment prior to transposition, its coding sequence was positioned in frame (through the PB 5 0 TR) with a PEST degron (Li et al., 1998) followed by a translational stop. The membrane-restricted GFP was generated by adding a short Kras tethering sequence (Averaimo et al., 2016) at the Ct end of EGFP using annealed oligonucleotides. The TTAA-less LiOn* CAGNRFP vector was designed on the model of LiOn CAGNRFP with punctual substitutions intended not to affect vector replication and expression. The 3-color integration reporter was constructed by inserting in LiOn CAGNRFP two CMVdriven transcriptional units expressing mTurquoise2 and EYFP, 5 0 of CAG and 3 0 of the mRFP1 polyA, respectively. To assay Cre activity, we designed a floxed reporter ( Tol2 CAG::loxP-mCherry-loxP-EYFP, abbreviated as Tol2 CAG::RY) in which expression switches from mCherry to EYFP upon recombination, framed with Tol2 transposition endfeet to enable genomic integration. The LiOn-CAGNRFP-2A-NICD vector was assembled by introducing a P2A cleavage sequence between the RFP and NICD ORFs to enable their co-expression. As non-integrative control vectors, METHOD DETAILS DNA constructs A schematized map of the plasmids designed for this study can be found in Table S2, along with restriction sites available to exchange GOIs and promoters. All iOn and control piggyBac vectors were assembled in a pUC57-mini plasmid backbone (Genscript Inc) using a combination of DNA synthesis, p.2, 2002.

, For triple-color labeling experiments, we used 100 ng/well of each LiOn CAGNFP plasmid and 60 ng of PBase vector. To validate the LiOn CMVNCre transgene, 50 ng of the corresponding plasmid was co-transfected with 10 ng of PBase vector in a HEK293 cell line stably expressing the Tol2 CAG::RY reporter. This line was established by successive use of Tol2 transposition, drug selection with G418 (300 mg/ml, Sigma) and picking of RFP-positive clones. In some experiments, 50 ng of non-integrative plasmid expressing an FP marker distinct from the iOn vector (CMV::mTurquoise2, CMV::IRFP or CAG::GFP) were applied as transfection control. For FACS analysis, transfections were performed in 6-cm dishes with scaled up concentrations. HEK293 cell viability after iOn plasmids transfection was assessed by dye exclusion with Trypan blue solution (0.4%, Sigma). FP expression was either assayed by flow cytometry, epifluorescence or confocal microscopy, or an Arrayscan high-content system (Thermo Fisher Scientific) (see below). For fixed observations, cells grown on 13 mm coverslips coated with collagen (50 mg/ml, Sigma) were immersed in 4% paraformaldehyde (PFA), HEK293, HeLa and NIH 3T3 cell culture experiments iOn and piggyBac plasmids were transfected in human HEK293, HeLa or mouse NIH 3T3 cells using cationic lipids. Except when otherwise noted, 1 3 10 5 cells/well were plated in a 24-well dish and transfected at day 1 with 100 ng iOn vector with or without 20 ng of PBase-expressing plasmid (CAG::hyPBase) using 0.7 ml of Lipofectamine 2000 reagent (Invitrogen)

. For, 561 nm (RFP), 640 nm (IRFP). 10000 cells were analyzed for each condition; non-fluorescent controls were prepared from mock-transfected cells stained with DAPI. For clonal experiments, HEK293 cells were sorted as single cells two days after transfection. Selection windows were chosen to include most of the FP-positive population and exclude negative cells. For 3-color cell sorting, we first selected live dissociated cells and subsequently selected RFP+, IRFP+ cells within the GFP+ population. Cells were then plated as single cells in 96-well plates and grown for 7-10 days in 200 mL of 10% FBS/DMEM medium mixed 1:1 with filtrated HEK293-conditioned medium. FP expression was assayed by epifluorescence microscopy or Arrayscan High-Content imaging (see below). Some positive clones were expanded in larger dishes for sequencing. To this aim, genomic DNA was isolated from a confluent 3.5-or 10-cm dish with the Nucleospin Tissue Kit (Macherey-Nagel), HEK293 cells grown on 6-cm dishes were dissociated three days after transfection, stained with DAPI and analyzed on a MoFlo Astrios cell sorter (Beckman Coulter) using the following laser lines: 405 nm (DAPI), 488 nm (GFP)

, Episome extraction and characterization

. Ziegler, For the bacterial transformation assay, 5ng of episomal DNA was transformed into XL2-Blue Ultracompetent Cells (Stratagene); the TTAA footprint in activated LiOn CAGNRFP transgenes. Quantitative PCR analysis was performed on purified episomes using an Applied Biosystems realtime PCR machine (7500 Fast System) with PowerUp SYBR green master mix following the manufacturer's instructions. Results from PCR1 PCR2), Quantification of gene expression was based on the DeltaCt method in at least three independent biological experiments. Episomal DNA isolated from cells transfected with CAG::RFP was used as a reference, Extrachromosomal DNA was purified from HEK cells three days after transfection as in, 2009.

M. , Human iPS cell transfection and differentiation For iOn labeling of differentiating iPS cells, colonies were dissociated with Accutase (Life Technologies) and replated in 96-well plates coated with poly-L-ornithine (20 mg/ml, Sigma P4957) and laminin (3 mg/ml, Sigma 23017-015), 2015.

, 1 mass ratio) using Lipofectamine 2000 reagent. 48 hrs after transfection, GFP-positives cells (1.5%) were sorted using an Astrios MoFlo EQ cell sorter and plated at low density (10 3 cells/ 10-cm dish) on feeder cells. After eight days, GFP-positives clones were picked under a fluorescent stereomicroscope, Mouse ES cell transfection and clone selection KH2 ES cells were transfected with LiOn CAGNGFP-Kras and CAG::hyPBase plasmids, vol.4

. Loulier, A DNA mix containing 1-1.2 mg/ml of iOn vector, 0.5-1.2 mg/ml of non-integrative control plasmid and 0.2 mg/ml of CAG::hyPBase plasmid supplemented with fast green dye was injected with a glass capillary pipette into one lateral ventricle or one eye of E12.5 or E14.5 mice, or the optic cup or central spinal cord canal of E1.5 or E2 chick embryos, respectively. For multicolor labeling, the mix contained 1 mg/ml of each LiOn CAGNFP vector and 0.6 mg/ml of PBase vector. Embryos were left to develop until sacrifice. Tissues were fixed in 4% PFA. Postnatal mouse brains were sectioned at 200-mm thickness with a vibrating microtome (VT1000, Leica), while mouse retinas, chick E6 spinal cords and E6-E8 retinas were flat-mounted on glass slides, Mouse and chicken embryonic electroporation In utero and in ovo electroporation in mouse and chicken embryos were performed as previously described, 2009.

, Immunostaining For cell cultures: HEK293 cells plated on glass coverslips or iPS cells were fixed with 4% PFA, followed by washing in PBS and a 20-60 min blocking step at room temperature. Blocking solution for HEK293 and iPS cells respectively contained 10% normal goat serum (Sigma) or fetal bovine serum (Eurobio) and 0.5% or 0.2% Triton X-100 (Sigma)

, 1:100) in PBS-0.1% Triton-1% NDS. Following PBS washes, slides were incubated 1 hr with secondary antibody (Alexa 647 donkey anti-mouse or anti-rabbit, Invitrogen, 1:500) in the above buffer, washed and mounted with Vectashield medium. For whole chicken spinal cords: embryos were fixed overnight in 4% PFA. Spinal cords were dissected, washed in PBS and incubated with Alexa 647-conjugated phalloidin (Molecular Probes, 1:250) in PBS-0.1% Triton-1% NDS for 3 hr at room temperature. Following PBS washes, spinal cords mounted in Vectashield medium. Fluorescence imaging and image analysis Epifluorescence images were, C with primary antibody diluted in blocking solution (rabbit anti-FLAG, Sigma, 1:250 or mouse anti-Tuj1, Biolegend, 1:500). After washing in PBS and incubation with secondary antibody (Alexa 647 anti-goat IgG, 1:500, or Alexa 488 anti-goat IgG, 1:1000, vol.40, p.3