Self-organizing models of human trunk organogenesis recapitulate spinal cord and spine co-morphogenesis

Self-organizing models of human trunk organogenesis recapitulate spinal cord and spine co-morphogenesis

Data availability

All scRNA-seq and tomo-seq data are available at the Gene Expression Omnibus: GSE215983 (SuperSeries composed of two SubSeries (single-cell and tomo-seq files))—the tomo-seq data (GSE215982) and the scRNA-seq data (GSE215981). The human embryo scRNA-seq data used in Extended Data Fig. 10 can be found at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE171892 and https://www.ebi.ac.uk/biostudies/arrayexpress/studies/E-MTAB-9388. For single-cell analysis, we used GRCh38-2020-A (from 10x Genomics) based on hg38. The human genome CRCh38 is available at https://www.ncbi.nlm.nih.gov/datasets/genome/GCF_000001405.26/. Additional data supporting the findings of this study are available in the article and the Supplementary Information. Further information for resources or technical information are available upon reasonable request from the corresponding author. Requests for transcriptomic analysis should be sent to a.vanoudenaarden@hubrecht.eu.

Code availability

The tomo-seq analysis script is available at https://github.com/anna-alemany.

References

Sahu, S. & Sharan, S. K. Translating embryogenesis to generate organoids: novel approaches to personalized medicine. iScience23, 101485 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Shao, Y. & Fu, J. Engineering multiscale structural orders for high-fidelity embryoids and organoids. Cell Stem Cell29, 722–743 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Moris, N., Alev, C., Pera, M. & Martinez Arias, A. Biomedical and societal impacts of in vitro embryo models of mammalian development. Stem Cell Rep.16, 1021–1030 (2021).

Article 

Google Scholar 

Sozen, B., Conkar, D. & Veenvliet, J. V. Carnegie in 4D? Stem-cell-based models of human embryo development. Semin. Cell Dev. Biol.131, 44–57 (2022).

Wymeersch, F. J., Wilson, V. & Tsakiridis, A. Understanding axial progenitor biology in vivo and in vitro. Development148, dev180612 (2021).

Article 
CAS 
PubMed 

Google Scholar 

Pourquié, O., Al Tanoury, Z. & Chal, J. The long road to making muscle in vitro. In Current Topics in Developmental Biology Vol. 129 (ed Brivanlou, A. H.) 123–142 (Academic Press, 2018).

Henrique, D., Abranches, E., Verrier, L. & Storey, K. G. Neuromesodermal progenitors and the making of the spinal cord. Development142, 2864–2875 (2015).

Article 
CAS 
PubMed 

Google Scholar 

Nedelec, S. & Martinez-Arias, A. In vitro models of spinal motor circuit’s development in mammals: achievements and challenges. Curr. Opin. Neurobiol.66, 240–249 (2021).

Article 
CAS 
PubMed 

Google Scholar 

Deschamps, J. & Duboule, D. Embryonic timing, axial stem cells, chromatin dynamics, and the Hox clock. Genes Dev.31, 1406–1416 (2017).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Philippidou, P. & Dasen, J. S. Hox genes: choreographers in neural development, architects of circuit organization. Neuron80, 12–34 (2013).

Article 
CAS 
PubMed 

Google Scholar 

Abdel Fattah, A. R. et al. Actuation enhances patterning in human neural tube organoids. Nat. Commun.12, 3192 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Budjan, C. et al. Paraxial mesoderm organoids model development of human somites. eLlife11, e68925 (2022).

Article 
CAS 

Google Scholar 

Duval, N. et al. BMP4 patterns Smad activity and generates stereotyped cell fate organization in spinal organoids. Development146, dev175430 (2019).

Article 
CAS 
PubMed 

Google Scholar 

Faustino Martins, J.-M. et al. Self-organizing 3D human trunk neuromuscular organoids. Cell Stem Cell26, 172–186 (2020).

Article 
CAS 
PubMed 

Google Scholar 

Gouti, M. et al. In vitro generation of neuromesodermal progenitors reveals distinct roles for Wnt signalling in the specification of spinal cord and paraxial mesoderm identity. PLoS Biol.12, e1001937 (2014).

Article 
PubMed 
PubMed Central 

Google Scholar 

Zheng, Y. et al. Dorsal-ventral patterned neural cyst from human pluripotent stem cells in a neurogenic niche. Sci. Adv.5, eaax5933 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Lee, J.-H. et al. Production of human spinal-cord organoids recapitulating neural-tube morphogenesis. Nat. Biomed. Eng.6, 435–448 (2022).

Article 
CAS 
PubMed 

Google Scholar 

Libby, A. R. G. et al. Axial elongation of caudalized human organoids mimics aspects of neural tube development. Development148, dev198275 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Maury, Y. et al. Combinatorial analysis of developmental cues efficiently converts human pluripotent stem cells into multiple neuronal subtypes. Nat. Biotechnol.33, 89–96 (2015).

Article 
CAS 
PubMed 

Google Scholar 

Miao, Y. et al. Reconstruction and deconstruction of human somitogenesis in vitro. Nature614, 500–508 (2023).

Article 
CAS 
PubMed 

Google Scholar 

Mouilleau, V. et al. Dynamic extrinsic pacing of the HOX clock in human axial progenitors controls motor neuron subtype specification. Development148, dev194514 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Ogura, T., Sakaguchi, H., Miyamoto, S. & Takahashi, J. Three-dimensional induction of dorsal, intermediate and ventral spinal cord tissues from human pluripotent stem cells. Development145, dev162214 (2018).

Article 
PubMed 
PubMed Central 

Google Scholar 

Olmsted, Z. T. & Paluh, J. L. Co-development of central and peripheral neurons with trunk mesendoderm in human elongating multi-lineage organized gastruloids. Nat. Commun.12, 3020 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Sanaki-Matsumiya, M. et al. Periodic formation of epithelial somites from human pluripotent stem cells. Nat. Commun.13, 2325 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

van den Brink, S. C. et al. Single-cell and spatial transcriptomics reveal somitogenesis in gastruloids. Nature582, 405–409 (2020).

Article 
PubMed 

Google Scholar 

Veenvliet, J. V. et al. Mouse embryonic stem cells self-organize into trunk-like structures with neural tube and somites. Science370, eaba4937 (2020).

Article 
CAS 
PubMed 

Google Scholar 

Xu, P.-F. et al. Construction of a mammalian embryo model from stem cells organized by a morphogen signalling centre. Nat. Commun.12, 3277 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Xue, W. et al. Generation of dorsoventral human spinal cord organoids via functionalizing composite scaffold for drug testing. iScience26, 105898 (2022).

Yaman, Y. I. & Ramanathan, S. Controlling human organoid symmetry breaking reveals signaling gradients drive segmentation clock waves. Cell186, 513–527 (2023).

Article 
CAS 
PubMed 

Google Scholar 

Yamanaka, Y. et al. Reconstituting human somitogenesis in vitro. Nature614, 509–520 (2023).

Article 
CAS 
PubMed 

Google Scholar 

Diaz-Cuadros, M. et al. In vitro characterization of the human segmentation clock. Nature580, 113–118 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Park, J., Hsiung, H.-A., Khven, I., La Manno, G. & Lutolf, M. P. Self-organizing in vitro mouse neural tube organoids mimic embryonic development. Development149, dev201052 (2022).

Article 
CAS 
PubMed 

Google Scholar 

Beccari, L. et al. Multi-axial self-organization properties of mouse embryonic stem cells into gastruloids. Nature562, 272–276 (2018).

Article 
CAS 
PubMed 

Google Scholar 

Moris, N. et al. An in vitro model of early anteroposterior organization during human development. Nature582, 410–415 (2020).

Article 
CAS 
PubMed 

Google Scholar 

Rito, T., Libby, A. R. G., Demuth, M. & Briscoe, J. Notochord and axial progenitor generation by timely BMP and NODAL inhibition during vertebrate trunk formation. Preprint at bioRxiv https://doi.org/10.1101/2023.02.27.530267 (2023).

Van Den Brink, S. C. et al. Symmetry breaking, germ layer specification and axial organisation in aggregates of mouse embryonic stem cells. Development141, 4231–4242 (2014).

Article 
PubMed 
PubMed Central 

Google Scholar 

Olmsted, Z. T. & Paluh, J. L. Stem cell neurodevelopmental solutions for restorative treatments of the human trunk and spine. Front. Cell. Neurosci.15, 667590 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Chambers, S. M. et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol.27, 275–280 (2009).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Hemmati-Brivanlou, A., Kelly, O. G. & Melton, D. A. Follistatin, an antagonist of activin, is expressed in the Spemann organizer and displays direct neuralizing activity. Cell77, 283–295 (1994).

Article 
CAS 
PubMed 

Google Scholar 

Camus, A., Perea-Gomez, A., Moreau, A. & Collignon, J. Absence of Nodal signaling promotes precocious neural differentiation in the mouse embryo. Dev. Biol.295, 743–755 (2006).

Article 
CAS 
PubMed 

Google Scholar 

Sagner, A. & Briscoe, J. Establishing neuronal diversity in the spinal cord: a time and a place. Development146, dev182154 (2019).

Article 
CAS 
PubMed 

Google Scholar 

Tani, S., Chung, U.-I., Ohba, S. & Hojo, H. Understanding paraxial mesoderm development and sclerotome specification for skeletal repair. Exp. Mol. Med.52, 1166–1177 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Goto, H., Kimmey, S. C., Row, R. H., Matus, D. Q. & Martin, B. L. FGF and canonical Wnt signaling cooperate to induce paraxial mesoderm from tailbud neuromesodermal progenitors through regulation of a two-step epithelial to mesenchymal transition. Development144, 1412–1424 (2017).

CAS 
PubMed 
PubMed Central 

Google Scholar 

Araya, C., Carmona-Fontaine, C. & Clarke, J. D. W. Extracellular matrix couples the convergence movements of mesoderm and neural plate during the early stages of neurulation. Dev. Dyn.245, 580–589 (2016).

Article 
CAS 
PubMed 

Google Scholar 

Buckley, C. E. & St Johnston, D. Apical–basal polarity and the control of epithelial form and function. Nat. Rev. Mol. Cell Biol.23, 559–577 (2022).

Long, K. R. & Huttner, W. B. How the extracellular matrix shapes neural development. Open Biol.9, 180216 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Mongera, A., Michaut, A., Guillot, C., Xiong, F. & Pourquié, O. Mechanics of anteroposterior axis formation in vertebrates. Annu. Rev. Cell Dev. Biol.35, 259–283 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Boulet, A. M. & Capecchi, M. R. Signaling by FGF4 and FGF8 is required for axial elongation of the mouse embryo. Dev. Biol.371, 235–245 (2012).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Cunningham, T. J., Kumar, S., Yamaguchi, T. P. & Duester, G. Wnt8a and Wnt3a cooperate in the axial stem cell niche to promote mammalian body axis extension. Dev. Dyn.244, 797–807 (2015).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Ribes, V., Le Roux, I., Rhinn, M., Schuhbaur, B. & Dollé, P. Early mouse caudal development relies on crosstalk between retinoic acid, Shh and Fgf signalling pathways. Development136, 665–676 (2009).

Article 
CAS 
PubMed 

Google Scholar 

Anand, G. M. et al. Controlling organoid symmetry breaking uncovers an excitable system underlying human axial elongation. Cell186, 497–512 (2023).

Article 
CAS 
PubMed 

Google Scholar 

Padmanabhan, R. Retinoic acid-induced caudal regression syndrome in the mouse fetus. Reprod. Toxicol.12, 139–151 (1998).

Article 
CAS 
PubMed 

Google Scholar 

Tan, Y. & Cahan, P. SingleCellNet: a computational tool to classify single cell RNA-seq data across platforms and across species. Cell Syst.9, 207–213 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Tyser, R. C. V. et al. Single-cell transcriptomic characterization of a gastrulating human embryo. Nature600, 285–289 (2021).

Article 
CAS 
PubMed 

Google Scholar 

Rayon, T., Maizels, R. J., Barrington, C. & Briscoe, J. Single-cell transcriptome profiling of the human developing spinal cord reveals a conserved genetic programme with human-specific features. Development148, dev199711 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Mayeuf-Louchart, A. et al. Endothelial cell specification in the somite is compromised in Pax3-positive progenitors of Foxc1/2 conditional mutants, with loss of forelimb myogenesis. Development143, 872–879 (2016).

CAS 
PubMed 
PubMed Central 

Google Scholar 

Mayeuf-Louchart, A. et al. Notch regulation of myogenic versus endothelial fates of cells that migrate from the somite to the limb. Proc. Natl Acad. Sci. USA111, 8844–8849 (2014).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Prummel, K. D., Nieuwenhuize, S. & Mosimann, C. The lateral plate mesoderm. Development147, dev175059 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Junker, J. P. et al. Genome-wide RNA tomography in the zebrafish embryo. Cell159, 662–675 (2014).

Article 
CAS 
PubMed 

Google Scholar 

Rossi, G. et al. Capturing cardiogenesis in gastruloids. Cell Stem Cell28, 230–240 (2021).

Article 
CAS 
PubMed 

Google Scholar 

Rayon, T. et al. Species-specific pace of development is associated with differences in protein stability. Science369, eaba7667 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Marklund, U. et al. Detailed expression analysis of regulatory genes in the early developing human neural tube. Stem Cells Dev.23, 5–15 (2014).

Article 
CAS 
PubMed 

Google Scholar 

Matsuda, M. et al. Recapitulating the human segmentation clock with pluripotent stem cells. Nature580, 124–129 (2020).

Article 
CAS 
PubMed 

Google Scholar 

Blauwkamp, T. A., Nigam, S., Ardehali, R., Weissman, I. L. & Nusse, R. Endogenous Wnt signalling in human embryonic stem cells generates an equilibrium of distinct lineage-specified progenitors. Nat. Commun.3, 1070 (2012).

Article 
PubMed 

Google Scholar 

Kyprianou, C. et al. Basement membrane remodelling regulates mouse embryogenesis. Nature582, 253–258 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Rifes, P. et al. Redefining the role of ectoderm in somitogenesis: a player in the formation of the fibronectin matrix of presomitic mesoderm. Development134, 3155–3165 (2007).

Article 
CAS 
PubMed 

Google Scholar 

Radice, G. L. et al. Developmental defects in mouse embryos lacking N-cadherin. Dev. Biol.181, 64–78 (1997).

Article 
CAS 
PubMed 

Google Scholar 

Nikolopoulou, E., Galea, G. L., Rolo, A., Greene, N. D. E. & Copp, A. J. Neural tube closure: cellular, molecular and biomechanical mechanisms. Development144, 552–566 (2017).

Article 
CAS 
PubMed 

Google Scholar 

Ensini, M., Tsuchida, T. N., Belting, H. G. & Jessell, T. M. The control of rostrocaudal pattern in the developing spinal cord: specification of motor neuron subtype identity is initiated by signals from paraxial mesoderm. Development125, 969–982 (1998).

Article 
CAS 
PubMed 

Google Scholar 

Wolujewicz, P., Steele, J. W., Kaltschmidt, J. A., Finnell, R. H. & Ross, M. E. Unraveling the complex genetics of neural tube defects: From biological models to human genomics and back. Genesis59, e23459 (2021).

Article 
PubMed 
PubMed Central 

Google Scholar 

Andersen, J. et al. Generation of functional human 3D cortico-motor assembloids. Cell183, 1913–1929 (2020).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Barbeau, S., Tahraoui-Bories, J., Legay, C. & Martinat, C. Building neuromuscular junctions in vitro. Development147, dev193920 (2020).

Article 
CAS 
PubMed 

Google Scholar 

Cederquist, G. Y. et al. Specification of positional identity in forebrain organoids. Nat. Biotechnol.37, 436–444 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Renier, N. et al. Mapping of brain activity by automated volume analysis of immediate early genes. Cell165, 1789–1802 (2016).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Download references

Acknowledgements

We thank M. Russeau for help and assistance on pluripotent stem cell culture; P. Lacour, F. Luciani, T. Rashed and H. Malek for their help during the project; J. Dasen for providing guinea pig anti-HOXC9 antibody; C. Metin for sharing ARL13B antibody; and L. Goutebroze for providing the Phalloidin probes. We thank H. Wichterle and the Ribes laboratory, in particular V. Ribes and P. Gilardi, as well as F. Causeret for helpful discussions along the project and critical reading of the manuscript. We thank J.-A. Girault and I. Caillé for their comments on the manuscripts. Imaging experiments and iPSC cultures were carried out, respectively, at the Imaging Platform and the Cell and Tissue Engineering Facility of the Institut du Fer à Moulin. This project was supported by two grants from the ATIP/Avenir program (S.N. and J.F.), Laboratoire d’Excellence (LabEx) Biopsy (ANR-10-LABX-73, S.N.), ANR SYMASYM (ANR-18-CE16-0021-03, S.N. and X.M.) as well as the NWO Gravitation Project: BRAINSCAPES: A Roadmap from Neurogenetics to Neurobiology (NWO: 024.004.012, B.v.S. and A.v.O.) and a ZonMW PSIDER grant (GREAT, 40-46800-98-015, A.L. and A.v.O.).

Author information

Authors and Affiliations

Institut du Fer à Moulin, Paris, France

Simona Gribaudo, Rémi Robert, Julien Ferent & Stéphane Nedelec

Inserm, UMR-S 1270, Paris, France

Simona Gribaudo, Rémi Robert, Julien Ferent & Stéphane Nedelec

Sorbonne Université, Science and Engineering Faculty, Paris, France

Simona Gribaudo, Rémi Robert, Julien Ferent & Stéphane Nedelec

Oncode Institute, Utrecht, The Netherlands

Björn van Sambeek, Anna Lyubimova & Alexander van Oudenaarden

Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands

Björn van Sambeek, Anna Lyubimova & Alexander van Oudenaarden

Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France

Camil Mirdass

Institut de Biologie de l’ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France

Kamal Bouhali & Xavier Morin

Contributions

S.N. and S.G. conceived the project. S.N., S.G., X.M., A.v.O. and R.R. designed experiments. S.G., S.N., R.R. and C.M. performed most wet lab experiments. K.B., R.R., S.N. and X.M. performed time-lapse imaging. J.F. performed clearing and 3D imaging. A.v.O. and A.L. performed scRNA-seq and tomo-seq experiments. A.v.O. and B.v.S. performed scRNA-seq and tomo-seq analysis. Project administration: S.N., X.M. and A.v.O. Supervision: S.N., A.v.O. and X.M. Writing—original draft: S.G. and S.N. Writing—review and editing: all authors.

Corresponding author

Correspondence to
Stéphane Nedelec.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Alexandre Mayran and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Morphological evolution of hiPSC-derived aggregates over time.

a, Representative bright field images of day 7 structures derived from WTS2 or WTC11 cell lines and from independent experiments, n = 13 for WTS2 and n = 7 for WTC11. b, Aspect ratio (Length (L)/Width (W)) of WTS2-derived structures over time. At day 7, the mean aspect ratio is 1.7 ± 0.76. c, Distribution of (Lmax-Wmax) of WTS2-derived structures over time: bin center=50 µm. d, Box-plot of WTC11-derived structures width and length over time. At day 7, the mean length is 516 ± 285 µm. e, WTC11-derived structures aspect ratio over time. At day 7, the mean aspect ratio is 1.7 ± 0.75. f, Distribution of (Lmax-Wmax) of WTC11-derived structures over time: bin center=50 µm. For box plots, the central line indicates the median; box limits the 1st and 3rd quartiles and whiskers the range. Each dot corresponds to a single structure and different colors indicate independent experiments. For WTS2, n = 11 for day 3; n = 12 for day 4; n = 13 for day 7. For WTC11, n = 2 for day 3; n = 3 for day 4; n = 3 for day 7. Scale bar, 200 µm (a).

Extended Data Fig. 2 Cell fate specification and topographical organization.

a, Schematic of the caudal region of the mammalian embryo. Axial progenitors generate both spinal neural and somitic lineages. n.t: neural tube. pPSM, aPSM: posterior and anterior PSM. b-b’, Maximum-intensity projection of whole-mount immunostaining on day 4 for markers of axial (SOX2/TBXT/CDX2), neural (SOX2) and mesodermal (TBX6) progenitors. WTS2 (b) and WTC11 (b’) derived structures. c, Percentage of structures with regionalized SOX2/TBXT/CDX2 progenitors in the entire population (day 4, n = 7, 159 structures) or among the elongated structures (at day 6, n = 3, 74 structures; day 7, n = 4, 48 structures and day 8, n = 3, 35 structures). Error bars represents the standard deviation. d-g, Immunostaining of sections at day 7 for mitotic cells (P-H3), F-Actin (Phalloidin) and neural progenitors (SOX1/3) markers (d, n = 2), or for the ventral neural progenitor marker (NKX6.1) (e, n = 4), or for the somitic markers FOXC2, SOX9, PAX3 (f, n = 2) and MEOX1 (g, n = 5). h, Low magnification view of sections of structures at day 7, immunostained for SOX9, PAX3 and SOX1/3, n = 4 i,j, 3D maximum-intensity projections of whole mount immunostaining at day 7 for the neuronal (SOX1/3, PAX3 in (i) or SOX1/3, OLIG2 in (j)) and somitic (FOXC2, SOX9, PAX3) markers. Optical plane in j. Scale bars, 50 µm (b, d-f), 100 µm (g, i, j), 500 µm (h).

Extended Data Fig. 3 Analysis of structures derived from other hiPSC lines and factors impacting neuro/mesodermal differentiation.

a, Representative bright field images of day 7 structures generated from SOX2-GFP, WTC11 and KUTE4 cell lines (n = 3, n = 3, n = 2). b-d, Immunostaining of sections of day 7 structures for neural progenitors (SOX1/3) and somitic (PAX3) markers, n = 3 for SOX2-GFP and WTC11, n = 2 for KUTE4. e-h, Representative bright field images (e, f; n = 3 and n = 4) and sections immunostained for neural progenitors (SOX2) and somitic (FOXC2) markers (g, h; n = 3 and n = 4) of day 7 structures generated by aggregating different cell numbers at day 0 (120, 160 and 300 cells). i, Representative bright field images (left panel) and sections immunostained for SOX2 and FOXC2 markers (right panel) of day 7 structures generated by aggregating different cell numbers at day 0 (300 or 600 cells) in presence of CHIR or with the addition of LDN and SB (n = 3). j, Representative bright field images (left panel) and sections immunostained for SOX2 and FOXC2 markers (right panel) of day 7 structures in presence of CHIR or with the addition of bFGF (20 ng/ml) for the first 2 days of differentiation (n = 3). k, Immunostaining of plated day 3 aggregates for neural progenitor (SOX2) and PSM (TBX6) markers. Aggregates obtained with different initial cell numbers were cultured using different B27 supplement (percentage of aggregates with TBX6 staining at day 3: B27 control, 50 ± 13% (120 cells) versus 83 ± 8% (300 cells), n = 2; B27 batch #2450484, 14 ± 9% (120 cells) versus 32 ± 21% (300 cells), n = 2; B27 batch #2450486, 2% (120 cells) versus 8% (300 cells), n = 1). l, Representative bright field images of day 7 structures obtained with the different initial cell numbers and different B27 supplements (n as in k). Scale bars, 200 µm (a, e, f, g, h, i, j, l), 100 µm (b, c, d), 50 µm (k).

Extended Data Fig. 4 Morphometric analysis of day 7 matrigel embedded structures.

a, Percentage of elongation for structures derived from 3 different cells lines (WTS2, n = 9, 892 structures total; WTC11, n = 3, 161 structures total; and SOX2-GFP, n = 2, 76 structures total). b, Box-plot of aspect ratio (length/width) of elongated structures at day 7 (WTS2, n = 8, 100 structures total; WTC11, n = 4, 57 structures total; SOX2-GFP, n = 3, 34 structures total). In the box plots, the central line indicates the median; box limits the 1st and 3rd quartiles and whiskers the range. Each dot corresponds to a single aggregate and different colors indicate independent differentiations. c-e, Gallery of bright field images of day 7 structures derived from 3 different hiPSC lines. Numbers indicate series from independent experiments. Scale bars, 200 µm (c, d, e).

Extended Data Fig. 5 Live imaging of matrigel embedded structures.

a, Time-lapse imaging showing dynamic elongation of matrigel-embedded SOX2-GFP derived structures between day 4 and 5.5. Red arrows pinpoint SOX2 negative cells. b, Time-lapse of matrigel-embedded WTS2 derived structures between day 4 and 7. Post-imaging whole mount immunostaining at day 7 for axial progenitors (SOX2, CDX2) and somitic (FOXC2) markers for the corresponding structure. #3 and #4, examples of non-elongating aggregates. Scale bars, 100 µm (a, b).

Extended Data Fig. 6 WNT, FGF and RA pathways control neural tube formation.

a, Experimental design. b, Representative images of SOX2-GFP whole mount structures in the different culture conditions at day 7, n = 3. c-d, Quantification of the aspect ratio (length/width) in the different conditions for day 7 organoids derived from SOX2-GFP (c) (n = 3, 2 to 6 wells/n/condition) and WTS2 (d) (n = 1, 2 wells/n/condition) hiPSC lines. ERKi and FGFRi=FGF pathways inhibitors. IWP2=Wnt pathway inhibitor. In the box plots, the central line indicates the median; box limits the 1st and 3rd quartiles and whiskers the range. Each dot represents a single organoid. Statistical test in c: Kruskal-Wallis followed by Dunn’s multiple comparison test. *** P 
>>> Read full article>>>
Copyright for syndicated content belongs to the linked Source : Nature.com – https://www.nature.com/articles/s41587-023-01956-9

Exit mobile version