Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Differentiating visceral sensory ganglion organoids from induced pluripotent stem cells

Abstract

The ability to generate visceral sensory neurons (VSN) from induced pluripotent stem (iPS) cells may help to gain insights into how the gut–nerve–brain axis is involved in neurological disorders. We established a protocol to differentiate human iPS-cell-derived visceral sensory ganglion organoids (VSGOs). VSGOs exhibit canonical VSN markers, and single-cell RNA sequencing revealed heterogenous molecular signatures and developmental trajectories of VSGOs aligned with native VSN. We integrated VSGOs with human colon organoids on a microfluidic device and applied this axis-on-a-chip model to Alzheimer’s disease. Our results suggest that VSN could be a potential mediator for propagating gut-derived amyloid and tau to the brain in an APOE4- and LRP1-dependent manner. Furthermore, our approach was extended to include patient-derived iPS cells, which demonstrated a strong correlation with clinical data.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Differentiating OEPD and EpP from iPS cells.
Fig. 2: Canonical VSN marker expression in VSGOs differentiated from EpP.
Fig. 3: VSGO scRNA analysis.
Fig. 4: Establishment of 3D axis-on-a-chip connecting VSGO and HCO using a microfluidic chip.
Fig. 5: Diverse assays using VSGOs for comparing APOE3/4 lines.
Fig. 6: Aβ and tau propagation assay of patient-derived VSGOs.

Similar content being viewed by others

Data availability

The human reference genome (GRCh38-2024-A) for the scRNA analysis was provided by 10x Genomics and is available at https://www.10xgenomics.com/support/software/cell-ranger/downloads. The scRNA-seq and snRNA-seq transcriptome data generated in this study are available in the Gene Expression Omnibus (GEO) under accession numbers GSE264303 and GSE271257. The mouse vagal sensory neuron scRNA-seq data are available in the GEO under accession number GSE124312. Source data are provided with this paper.

Code availability

The codes used for data analysis are available on the GitHub repository at https://github.com/Hweeseon/scRNA_VSGO/ under an MIT license.

References

  1. Cryan, J. F., O’Riordan, K. J., Sandhu, K., Peterson, V. & Dinan, T. G. The gut microbiome in neurological disorders. Lancet Neurol. 19, 179–194 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Kim, M. S. et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut 69, 283–294 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Choi, H., Lee, D. & Mook-Jung, I. Gut microbiota as a hidden player in the pathogenesis of Alzheimer’s disease. J. Alzheimers Dis. 86, 1501–1526 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. Vargas-Caballero, M. et al. Vagus nerve stimulation as a potential therapy in early Alzheimer’s disease: a review. Front. Hum. Neurosci. 16, 866434 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kim, S. et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103, 627–641.e627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chen, C. et al. Gut inflammation triggers C/EBPβ/δ-secretase-dependent gut-to-brain propagation of Aβ and Tau fibrils in Alzheimer’s disease. EMBO J. 40, e106320 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sun, Y. et al. Intra-gastrointestinal amyloid-beta1-42 oligomers perturb enteric function and induce Alzheimer’s disease pathology. J. Physiol. 598, 4209–4223 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Crespo, M. et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nat. Med. 23, 878–884 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Paşca, A. M. et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 12, 671–678 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  10. De Santis, R. et al. Direct conversion of human pluripotent stem cells into cranial motor neurons using a piggyBac vector. Stem Cell Res. 29, 189–196 (2018).

    Article  PubMed  Google Scholar 

  11. Schwartzentruber, J. et al. Molecular and functional variation in iPSC-derived sensory neurons. Nat. Genet. 50, 54–61 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Kupari, J., Häring, M., Agirre, E., Castelo-Branco, G. & Ernfors, P. An atlas of vagal sensory neurons and their molecular specialization. Cell Rep. 27, 2508–2523.e2504 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Saint-Jeannet, J. P. & Moody, S. A. Establishing the pre-placodal region and breaking it into placodes with distinct identities. Dev. Biol. 389, 13–27 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ladher, R. K., O’Neill, P. & Begbie, J. From shared lineage to distinct functions: the development of the inner ear and epibranchial placodes. Development 137, 1777–1785 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Koehler, K. R. et al. Generation of inner ear organoids containing functional hair cells from human pluripotent stem cells. Nat. Biotechnol. 35, 583–589 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McCarroll, M. N. & Nechiporuk, A. V. Fgf3 and Fgf10a work in concert to promote maturation of the epibranchial placodes in zebrafish. PLoS ONE 8, e85087 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Vermeiren, S., Bellefroid, E. J. & Desiderio, S. Vertebrate sensory ganglia: common and divergent features of the transcriptional programs generating their functional specialization. Front. Cell Dev. Biol. 8, 587699 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Blentic, A., Chambers, D., Skinner, A., Begbie, J. & Graham, A. The formation of the cranial ganglia by placodally-derived sensory neuronal precursors. Mol. Cell Neurosci. 46, 452–459 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Wang, L. et al. Notch signalling regulates epibranchial placode patterning and segregation. Development 147, dev183665 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Katz, D. M., Erb, M., Lillis, R. & Neet, K. Trophic regulation of nodose ganglion cell development: evidence for an expanded role of nerve growth factor during embryogenesis in the rat. Exp. Neurol. 110, 1–10 (1990).

    Article  CAS  PubMed  Google Scholar 

  21. Erickson, J. T., Brosenitsch, T. A. & Katz, D. M. Brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor are required simultaneously for survival of dopaminergic primary sensory neurons in vivo. J. Neurosci. 21, 581–589 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Borgmann, D. et al. Gut-brain communication by distinct sensory neurons differently controls feeding and glucose metabolism. Cell Metab. 33, 1466–1482.e1467 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Adams, J. M. et al. Liraglutide modulates appetite and body weight through glucagon-like peptide 1 receptor-expressing glutamatergic neurons. Diabetes 67, 1538–1548 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Browning, K. N., Babic, T., Holmes, G. M., Swartz, E. & Travagli, R. A. A critical re-evaluation of the specificity of action of perivagal capsaicin. J. Physiol. 591, 1563–1580 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zhao, Q. et al. A multidimensional coding architecture of the vagal interoceptive system. Nature 603, 878–884 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lin, Y. T. et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 98, 1141–1154 e1147 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Avraham, O. et al. Profiling the molecular signature of satellite glial cells at the single cell level reveals high similarities between rodents and humans. Pain 163, 2348–2364 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Lowenstein, E. D., Misios, A., Buchert, S. & Ruffault, P. L. Molecular characterization of nodose ganglia development reveals a novel population of Phox2b+ glial progenitors in mice. J. Neurosci. 44, e1441232024 (2024).

    Article  CAS  PubMed  Google Scholar 

  29. Rosen, Y. et al. Toward universal cell embeddings: integrating single-cell RNA-seq datasets across species with SATURN. Nat. Methods 21, 1492–1500 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Thiery, A. P. et al. scRNA-sequencing in chick suggests a probabilistic model for cell fate allocation at the neural plate border. eLife 12, e82717 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schwarz, Q., Vieira, J. M., Howard, B., Eickholt, B. J. & Ruhrberg, C. Neuropilin 1 and 2 control cranial gangliogenesis and axon guidance through neural crest cells. Development 135, 1605–1613 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Smith, A. C., Fleenor, S. J. & Begbie, J. Changes in gene expression and cell shape characterise stages of epibranchial placode-derived neuron maturation in the chick. J. Anat. 227, 89–102 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tambalo, M., Anwar, M., Ahmed, M. & Streit, A. Enhancer activation by FGF signalling during otic induction. Dev. Biol. 457, 69–82 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kaelberer, M. M. et al. A gut-brain neural circuit for nutrient sensory transduction. Science 361, eaat5236 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Jameson, K. G., et al. Vagal interoception of microbial metabolites from the small intestinal lumen. Preprint at bioRxiv https://doi.org/10.1101/2023.12.18.572257 (2023).

  36. Yang, Y., Lin, Z., Lin, Q., Bei, W. & Guo, J. Pathological and therapeutic roles of bioactive peptide trefoil factor 3 in diverse diseases: recent progress and perspective. Cell Death Dis. 13, 62 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Barton, J. R. et al. Intestinal neuropod cell GUCY2C regulates visceral pain. J. Clin. Invest. 133, e165578 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dominguez Conde, C. et al. Cross-tissue immune cell analysis reveals tissue-specific features in humans. Science 376, eabl5197 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dimitrov, D. et al. LIANA+ provides an all-in-one framework for cell–cell communication inference. Nat. Cell Biol. 26, 1613–1622 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Koutsodendris, N. et al. Neuronal APOE4 removal protects against tau-mediated gliosis, neurodegeneration and myelin deficits. Nat. Aging 3, 275–296 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kim, H., Kim, S., Cho, B., Shin, J. & Kim, J. APOE epsilon4-dependent effects on the early amyloid pathology in induced neurons of patients with Alzheimer’s disease. Transl. Neurodegener. 11, 45 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lourenco, F. C. et al. Netrin-1 interacts with amyloid precursor protein and regulates amyloid-beta production. Cell Death Differ. 16, 655–663 (2009).

    Article  CAS  PubMed  Google Scholar 

  43. Kanekiyo, T. et al. Neuronal clearance of amyloid-beta by endocytic receptor LRP1. J. Neurosci. 33, 19276–19283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rauch, J. N. et al. LRP1 is a master regulator of tau uptake and spread. Nature 580, 381–385 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Takayama, Y. et al. Selective induction of human autonomic neurons enables precise control of cardiomyocyte beating. Sci. Rep. 10, 9464 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Park, J. C. et al. A logical network-based drug-screening platform for Alzheimer’s disease representing pathological features of human brain organoids. Nat. Commun. 12, 280 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Raimondi, M. T., Albani, D. & Giordano, C. An organ-on-a-chip engineered platform to study the microbiota-gut-brain axis in neurodegeneration. Trends Mol. Med. 25, 737–740 (2019).

    Article  PubMed  Google Scholar 

  48. Goff, L. A. et al. Spatiotemporal expression and transcriptional perturbations by long noncoding RNAs in the mouse brain. Proc. Natl Acad. Sci. USA 112, 6855–6862 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lionnet, A. et al. Characterisation of tau in the human and rodent enteric nervous system under physiological conditions and in tauopathy. Acta Neuropathol. Commun. 6, 65 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Parvizi, J., Van Hoesen, G. W. & Damasio, A. The selective vulnerability of brainstem nuclei to Alzheimer’s disease. Ann. Neurol. 49, 53–66 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Palmqvist, S. et al. Earliest accumulation of beta-amyloid occurs within the default-mode network and concurrently affects brain connectivity. Nat. Commun. 8, 1214 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Spuch, C., Ortolano, S. & Navarro, C. LRP-1 and LRP-2 receptors function in the membrane neuron. Trafficking mechanisms and proteolytic processing in Alzheimer’s disease. Front Physiol. 3, 269 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Suarez, A. N. et al. Gut vagal sensory signaling regulates hippocampus function through multi-order pathways. Nat. Commun. 9, 2181 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Balasubramaniam, M. et al. Rescue of ApoE4-related lysosomal autophagic failure in Alzheimer’s disease by targeted small molecules. Commun. Biol. 7, 60 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jack, C. R. Jr et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562 (2018).

    Article  PubMed  Google Scholar 

  56. Fleming, S. J. et al. Unsupervised removal of systematic background noise from droplet-based single-cell experiments using CellBender. Nat. Methods 20, 1323–1335 (2023).

    Article  CAS  PubMed  Google Scholar 

  57. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291 e289 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Gayoso, A. et al. A Python library for probabilistic analysis of single-cell omics data. Nat. Biotechnol. 40, 163–166 (2022).

    Article  CAS  PubMed  Google Scholar 

  60. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e727 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Lange, M. et al. CellRank for directed single-cell fate mapping. Nat. Methods 19, 159–170 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Lee (Department of Biomedical Sciences, College of Medicine, Seoul National University) for technical support in the differentiation of VSGOs. We also thank S. Kim (Department of Biomedical Sciences, College of Medicine, Seoul National University) for technical support in the maintenance of iPS cells. We thank H. Cho and S. Yu (Genomic Medicine Institute, Medical Research Center, Seoul National University) for technical support in scRNA-seq. NNC0090-1170 (Liraglutide) was provided by Novo Nordisk Compound Sharing. We thank Geninus Inc. for performing sequencing process of scRNA-seq at i-VSN. We thank Macrogen Inc. for performing sequencing process of scRNA-seq of OEPD, EpP and snRNA-seq of VSGOs. This research was supported by a grant of the MD-PhD/Medical Scientist Training Program through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant number: HI21C1212) to K.A. This work was supported by Samsung Research Funding and Incubation Center of Samsung Electronics under Project Number SRF-IT1901-51 to S. Chung. This work was supported by the National Research Foundation (NRF) grant funded by the Korea government (MSIT) (2022M3A9B6018217) to S. Chung. This work was supported by the NRF Grant funded by the Korean Government (MSIT) (No. 2018R1A5A2025964) to S.J.K. This research was supported by Basic Science Research Program through the NRF funded by the Ministry of Education (2020R1A6A1A03047972) to J.-I.K. Additionally, this research was supported by a grant of the Korea Dementia Research Project through the Korea Dementia Research Center (KDRC), funded by the Ministry of Health and Welfare and Ministry of Science and ICT, Republic of Korea (grant number: RS-2020-KH106747) to I.M.-J.

Author information

Authors and Affiliations

Authors

Contributions

K.A. and I.M.-J. conceptualized the research. K.A. designed protocol for differentiating VSGOs. K.A. performed RT−qPCR, ICC of OEPD, EpP, VMN, HCO and bundle chip experiments. K.A. and J.S. performed ICC of VSGOs. I.M.-J. and H.C. supervised the overall experimental process. S.B.H. and H.C. modified previous protocol for differentiating HCO from iPS cells. H.L. and S.J.K. performed and analyzed patch-clamp analysis. J.H., S. Choi and K.A. performed PRV analysis. J.W.H. and K.A. performed calcium channel imaging of VSGO. K.P., B.C., H.-S.P. and J.-I.K. constructed scRNA libraries. H.-S.P. analyzed scRNA-seq data and wrote the method of analyzing scRNA-seq data. J.A., S. Choi, and S. Chung designed bundle chips. S. Choi, M.K., H.-W.L. and H.S. cultured VSGOs and HCO at bundle chips. S. Choi wrote methods for bundle chips. S. Choi and J.A. performed calcium channel imaging of bundle chip. K.A., H.-S.P., S. Choi and I.M.-J. wrote the final version of the paper.

Corresponding authors

Correspondence to Seok Chung, Jong-Il Kim or Inhee Mook-Jung.

Ethics declarations

Competing interests

K.A. and I.M.-J. are inventors on a filed patent related to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Keqiang Ye and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Nina Vogt, in collaboration with the Nature Methods team.

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 Graphical abstract of entire experimental process.

VSGO, Visceral sensory ganglion organoid; iPSC, induced-pluripotent stem cell; EB, Embryonic body; OEPD, Otic-epibranchial progenitor domain; EpP, Epibranchial placode; VSN, visceral sensory neuron; RAP, Receptor-associated protein; HCO, human colon organoid; Aβ, amyloid beta; VMN, visceral motor neuron; NC, negative control; CN, cognitively normal; sAD, sporadic Alzheimer’s disease.

Extended Data Fig. 2 Comparing diverse methods for differentiating EpP from OEPD.

(a) Schematic of diverse protocols tested for differentiating EpP from OEPD. (b-d) RT-PCR results of PHOX2B (b), PAX2 (c), and PAX8 (d) for comparing diverse protocols (n = 4 for each formula, all n: biological replicates). two-sided ANOVA with Tukey’s post-hoc test *P < 0.05, **P < 0.01, ***P < 0.001. All data were presented as mean ± SEM.

Source data

Extended Data Fig. 3 Functional characterization of VSGOs.

(a) Representative live-cell images before and after treatment with capsaicin (10 µM) and liraglutide (10 µM) (Baseline indicates before treatment) (Representative images among 2 batches, Scale bars: 100μm). (b) Schematic of timeline for calcium imaging. (c) Venn diagram representing number of i-VSNs activated by capsaicin or liraglutide or both. (d) Schematic of timeline for patch-clamp recording. (e) Bright field image showing patch-clamp experiment on VSN (Representative image among 3 batches, Scale bars: 10μm). (f) The examples of voltage traces. (g) Increasing relationship of spike number vs current (total 26 cells analyzed among 3 batches, mean ± SEM). (h and i) i-VSN was activated responding to capsaicin (h) and GLP1R agonist DMB (i). (j) Venn diagram of i-VSN number ratio responding to capsaicin or DMB, or both.

Extended Data Fig. 4 scRNA-seq label transfer and trajectory analysis.

(a-f) UMAPs of representative markers for each cluster identified at VSGOs, VSN for SLC17A6, SC for MPZ, SGC for TRPM3, GP for FBLN5 and MKI67, EpPL for PAX2. (g) comparison of VSN proportions between VSGO and i-VSN, two-sided Mann-whitney’s U test (4 biological replicates /each group, mean ± SEM). (h) Expression matrixplot of genes among iVSN cultured days (x axis). (i) UMAP of mouse nodose ganglion and VSGOs. (j) List of gene membership among each Macrogenes. (k) Expression of VSN score 2 among each VSN subclusters. (l) Terminal states of trajectory analysis. (m) Directed partition-based graph abstraction (PAGA) cluster UMAP. (n, o) UMAP of pseudotime and latent time. (p) UMAP plots of random walks among each cluster. (q, r) Smoothed gene expression trends for VSN_1 (q) and VSN_2 (r). Along with each trends, driver genes for each cell states are presented.

Source data

Extended Data Fig. 5 3D Axis-on-a-chip for identifying cell-cell interactions.

(a) Entire immunostaining image of axis-on-a-chip indicating VSGO chamber by TUJ1 (red) and HCO chamber by DSG2 (yellow) (Representative image among 3 biological replicates). (b and c) Diffusion test was conducted in microfluidic chip using 10 µM of 40 kDa FITC-dextran solution. Fluorescence images showing diffusion of dextran within the microchannel when with no ECM hydrogel or with ECM hydrogel composed of Collagen I and matrigel. (d) Graph represents relative intensity comparison of dextran fluorescence of each condition. (e) PRV-infected colon organoid connected with neurite bundle of VSGO. PRV expressing GFP and neurite immunostained with TUJ1 (red) (Representative image among 3 biological replicates). ECM, Extracellular matrix; PRV, pseudorabies virus. Scale bars indicate 500 µm (a), 50 µm (b), 200 µm (c and d).

Source data

Extended Data Fig. 6 VSN subcluster scRNA-seq classification and Axis-on-a-chip scRNA-seq.

(a) A comprehensive comparison of various characteristics of VSN clusters revealed between our data and previously published results. (b) UMAPs for canonical markers of HCO clusters or previously described (c) Ligand-receptor interaction plot between HCO and VSN. (d) Expression of VSN score 2 among previous VSGO clusters and chip VSN 3 and 5.

Extended Data Fig. 7 ICC images of axis-on-a-chip used for Aβ and tau propagation assay.

VSGO and its neurite bundle immunostained with TUJ1 (red) and HCO with DSG2 (yellow). (a) Images of propagated FITC-oAβ (a) (Representative images among E3 n = 7, E4 n = 4, AD4 n = 3) and Atto-488 tauPFF (b) (Representative images among E3 n = 5, E4 n = 4, AD4 n = 3) via the bundle of each E3, E4, and AD4 line. All n are biological replicates. Scale bars indicate 500μm.

Extended Data Fig. 8 Investigating ApoE4 and LRP1 in Aβ and Tau Propagation through Transcriptomic Comparisons and RAP Inhibition.

(a) Quantifications of proportions of identified VSN subclusters among E3 and E4 VSN. (b) UMAP presenting E3 and E4 sample for VSN cluster. (c, d) UMAPs for APOE (c) and LRP1 expression (d) among VSN cluster. (e-g) Expressions of gene sets among GO term: Cholesterol Biosynthetic process (e), Regulation of Autophagy (f) and Chromatin Organization (g) among each sample. (h) Schematic explaining mechanism of RAP working on LRP1 of i-VSN neurite. Representative images of FITC-oAβ (i) and Atto-499 tauPFF (k) before and after treatment of RAP on i-VSN (TUJ1, yellow). Normalized propagation ratio of FITC-oAβ (j) (n = 9 for E4 RAP- and n = 11 for E4 RAP+ among 4 independent differentiations each) and Atto-488 tauPFF (l) (n = 10 for E4 RAP- and n = 12 for E4 RAP+ among 4 independent differentiations each) were shown as bar graphs. RAP, receptor-associated protein. Scale bars: 20 µm. (a) two-sided chi-square test (j and k) two-sided student t test. *P < 0.05, ***P < 0.001. All data were presented as mean ± SEM.

Source data

Extended Data Fig. 9 Comparative evaluation of Aβ and tau propagation in VSN versus VMN.

(a) Timeline of protocol used for generating VMN from iPSCs. (b) ICC characterization of VMN expressing TH (green) and CHAT (red). (c-f) Aβ or tau propagation in VSN neurite bundle compared to VMN. Rendering images of FITC-oligomeric Aβ (c) and Atto-488 tauPFF (e). Bar graphs showing normalized propagation ratio of FITC-oligomeric Aβ (d) and Atto-488 tauPFF (f). VMN, visceral motor neuron. n = 4 independent differentiations among three batches. Scale bars indicate 50 µm (c and e). (d, f) Two-sided Mann-Whitney’s U test. *P < 0.05. All data were presented as mean ± SEM.

Source data

Extended Data Fig. 10 Correlation analysis between axis-on-a-chip propagation analysis and clinical data of donors.

(a-e) Correlation analysis between donor’s clinical data and quantified amyloid or tau propagation amount at axis-on-a-chip. Donor data: a, MMSE-Z score; b, Semantic memory Z-score; c, Global amyloid PET SUVR; d, Brain MRI hippocampal adjusted volume (HVA); e, FDG PET SUVR at PCC (n=16). Each n are biological replicates. Used iPSC lines: 1-control, 4-sAD lines. (a-e) Kendall’s tau correlation analysis. ***P<0.001.

Supplementary information

Supplementary Information

Supplementary Figs. 1–13, Tables 1–9 and Protocol.

Reporting Summary

Supplementary Table 10

Lists of gene membership among each Macrogenes.

Supplementary Table 11

Additional data for Trajectory analysis.

Supplementary Video 1

Live-cell calcium imaging of VSGOs after treating with liraglutide.

Supplementary Video 2

Live-cell calcium imaging of VSGOs after treating with capsaicin.

Source data

Source Data Fig. 1

Image quantification, RT–PCR source data.

Source Data Fig. 2

Image quantification source data.

Source Data Fig. 3

scRNA-seq cell count data.

Source Data Fig. 4

Live-cell calcium imaging source data.

Source Data Fig. 5

Image quantification source data.

Source Data Fig. 6

Image quantification source data.

Source Data Extended Data Fig. 2

RT–PCR source data.

Source Data Extended Data Fig. 4

scRNA-seq cell count source data.

Source Data Extended Data Fig. 5

Diffusion test source data.

Source Data Extended Data Fig. 8

Image quantification source data.

Source Data Extended Data Fig. 9

Image quantification source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ahn, K., Park, HS., Choi, S. et al. Differentiating visceral sensory ganglion organoids from induced pluripotent stem cells. Nat Methods 21, 2135–2146 (2024). https://doi.org/10.1038/s41592-024-02455-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41592-024-02455-8

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing