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:

Geometric engineering of organoid culture for enhanced organogenesis in a dish

Abstract

Here, we introduce a facile, scalable engineering approach to enable long-term development and maturation of organoids. We have redesigned the configuration of conventional organoid culture to develop a platform that converts single injections of stem cell suspensions to radial arrays of organoids that can be maintained for extended periods without the need for passaging. Using this system, we demonstrate accelerated production of intestinal organoids with significantly enhanced structural and functional maturity, and their continuous development for over 4 weeks. Furthermore, we present a patient-derived organoid model of inflammatory bowel disease (IBD) and its interrogation using single-cell RNA sequencing to demonstrate its ability to reproduce key pathological features of IBD. Finally, we describe the extension of our approach to engineer vascularized, perfusable human enteroids, which can be used to model innate immune responses in IBD. This work provides an immediately deployable platform technology toward engineering more realistic organ-like structures in a dish.

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: Geometric engineering of conventional organoid culture using OCTOPUS.
Fig. 2: Effects of long-term culture on the maturation of intestinal organoids in OCTOPUS.
Fig. 3: Prolonged culture of human intestinal organoids in OCTOPUS.
Fig. 4: scRNA-seq of human enteroids in OCTOPUS.
Fig. 5: Organoid-based model of human IBD in OCTOPUS.
Fig. 6: Microengineering of vascularized human enteroids in OCTOPUS-EVO.

Similar content being viewed by others

Data availability

All the numeric data used in this study are included in the Source data files provided with this paper. The human enteroids scRNA-seq dataset analyzed during the current study is available at the NCBI Gene Expression Omnibus, under accession number GSE203380. The raw images are too large for public deposit and are available from the corresponding author on reasonable request.

References

  1. Clevers, H. Modeling development and disease with organoids. Cell 165, 1586–1597 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Lancaster, M. A. & Knoblich, J. A. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345, 1247125 (2014).

    Article  PubMed  Google Scholar 

  3. Dutta, D., Heo, I. & Clevers, H. Disease modeling in stem cell-derived 3D organoid systems. Trends Mol. Med. 23, 393–410 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Broutier, L. et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat. Protoc. 11, 1724–1743 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Shin, W. et al. Spatiotemporal gradient and instability of Wnt induce heterogeneous growth and differentiation of human intestinal organoids. iScience 21, 101372 (2020).

    Article  Google Scholar 

  6. Hu, H. et al. Long-term expansion of functional mouse and human hepatocytes as 3D organoids. Cell 175, 1591–1606 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Huch, M. & Koo, B. Modeling mouse and human development using organoid cultures. Development 142, 3113–3125 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Homan, K. A. et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat. Methods 16, 255–262 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lee, G. Y., Kenny, P. A., Lee, E. H. & Bissell, M. J. Three-dimensional culture models of normal and malignant breast epithelial cells. Nat. Methods 4, 359–365 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, Y. et al. Long-term culture captures injury-repair cycles of colonic stem cells. Cell 179, 1144–1159 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, Y. et al. Bioengineered systems and designer matrices that recapitulate the intestinal stem cell niche. Cell. Mol. Gastroenterol. Hepatol. 5, 440–453 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Walton, K. D. et al. Hedgehog-responsive mesenchymal clusters direct patterning and emergence of intestinal villi. Proc. Natl Acad. Sci. USA 109, 15817–15822 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Walker, E. M., Thompson, C. A., Kohlnhofer, B. M., Faber, M. L. & Battle, M. A. Characterization of the developing small intestine in the absence of either GATA4 or GATA6. BMC Res. Notes 7, 902 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Serra, D. et al. Self-organization and symmetry breaking in intestinal organoid development. Nature 569, 66–72 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sato, T. & Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science 340, 1190–1194 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Sumigray, K. D., Terwilliger, M. & Lechler, T. Morphogenesis and compartmentalization of the intestinal crypt. Dev. Cell 45, 183–197 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cattin, A. et al. Hepatocyte nuclear factor 4α, a key factor for homeostasis, cell architecture, and barrier function of the adult intestinal epithelium. Mol. Cell. Biol. 29, 6294–6308 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Spanier, B. Transcriptional and functional regulation of the intestinal peptide transporter PEPT1. J. Physiol. 592, 871–879 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Ferraris, R. P. & Diamond, J. Regulation of intestinal sugar transport. Physiol. Rev. 77, 257–302 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Chen, L., Tuo, B. & Dong, H. Regulation of intestinal glucose absorption by ion channels and transporters. Nutrients 8, 43 (2016).

    Article  PubMed Central  Google Scholar 

  21. Yang, X. et al. Molecular mechanisms of calcium signaling in the modulation of small intestinal ion transporters and bicarbonate secretion. Oncotarget 9, 3727–3740 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. MacDonald, P. E. et al. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes 51, S434–S442 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Kim, Y. S. & Ho, S. B. Intestinal goblet cells and mucins in health and disease: recent insights and progress. Curr. Gastroenterol. Rep. 12, 319–330 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Fuji, M. et al. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition. Cell Stem Cell 23, 787–792 (2018).

    Article  Google Scholar 

  25. Elmentaite, R. et al. Single-cell sequencing of developing human gut reveals transcriptional links to childhood Crohn’s disease. Dev. Cell 55, 771–783 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, Y. et al. Single-cell transcriptome analysis reveals differential nutrient absorption functions in human intestine. J. Exp. Med. 217, e20191130 (2020).

    Article  PubMed  Google Scholar 

  27. Ito, G. et al. Lineage-specific expression of bestrophin-2 and bestrophin-4 in human intestinal epithelial cells. PLoS ONE 8, e79693 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sprangers, J., Zaalberg, I. C. & Maurice, M. M. Organoid-based modeling of intestinal development, regeneration, and repair. Cell Death Differ. 28, 95–107 (2021).

    Article  PubMed  Google Scholar 

  29. Peterson, L. W. & Artis, D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 14, 141–153 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Modigliani, R. et al. Clinical, biological, and endoscopic picture of attacks of Crohn’s disease. Gastroenterology 98, 811–818 (1990).

    Article  CAS  PubMed  Google Scholar 

  31. Guo, C. & Shen, J. Cytoskeletal organization and cell polarity in the pathogenesis in Crohn’s disease. Clin. Rev. Allergy Immunol. 60, 164–174 (2021).

    Article  PubMed  Google Scholar 

  32. Schmitt, M. et al. Paneth cells respond to inflammation and contribute to tissue regeneration by acquiring stem-like features through SCF/c-Kit signaling. Cell Rep. 24, 2312–2328 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Spinelli, A., Correale, C., Szabo, H. & Montorsi, M. Intestinal fibrosis in Crohn’s disease: medical treatment or surgery? Curr. Drug Targets 11, 242–248 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Reimund, J. M. et al. Increased production of tumour necrosis factor-α, interleukin-1β, and interleukin-6 by morphologically normal intestinal biopsies from patients with Crohn’s disease. Gut 39, 684–689 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Grebenyuk, S. & Ranga, A. Engineering organoid vascularization. Front. Bioeng. Biotechnol. 7, 1–12 (2019).

    Article  Google Scholar 

  36. Deban, L. et al. Multiple pathogenic roles of microvasculature in inflammatory bowel disease: a Jack of all trades. Am. J. Pathol. 172, 1457–1466 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jones, G. et al. Dynamics of colon monocyte and macrophage activation during colitis. Front. Immunol. 27, 2764 (2018).

    Article  Google Scholar 

  38. Demers, C. J. et al. Development-on-a-chip: in vitro neural tube patterning with a microfluidic device. Development 143, 1884–1892 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kasendra, M. et al. Development of a primary human small intestine-on-a-chip using biopsy-derived organoids. Sci. Rep. 8, 2871 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Wu, F., Huang, Y., Dong, F. & Kwon, J. H. Ulcerative colitis-associated long noncoding RNA, BC012900, regulates intestinal epithelial cell apoptosis. Inflamm. Bowel Dis. 22, 782–795 (2016).

    Article  PubMed  Google Scholar 

  42. Gu, L. et al. Identification of a 5-lncRNA signature-based risk scoring system for survival prediction in colorectal cancer. Mol. Med. Rep. 18, 279–291 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Axelrad, J. E., Lichtiger, S. & Yajnik, V. Inflammatory bowel disease and cancer: the role of inflammation, immunosuppression, and cancer treatment. World J. Gastroenterol. 22, 4794–4801 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jaeger, N. et al. Single-cell analyses of Crohn’s disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions. Nat. Commun. 12, 1921 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  45. van de Watering, M. et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161, 933–945 (2015).

    Article  Google Scholar 

  46. Mansour, A. A. et al. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol. 36, 432–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Dye, B. R. et al. A bioengineered niche promotes in vivo engraftment and maturation of pluripotent stem cell derived human lung organoids. eLife 5, e19732 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Takahashi, Y., Takebe, T. & Taniguchi, H. Methods for generating vascularized islet-like organoids via self-condensation. Curr. Protoc. Stem Cell Biol. 45, e49 (2018).

    Article  PubMed  Google Scholar 

  49. Vargas-Valderrama, A., Messina, A., Mitjavila-Garcia, M. T. & Guenou, H. The endothelium, a key factor in organ development and hPSC-derived organoid vascularization. J. Biomed. Sci. 27, 67 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Park, S. E., Georgescu, A. & Huh, D. Organoids-on-a-chip. Science 364, 960–965 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. Wells, W. Yang and G. Al for their input. This work was supported by the National Institutes of Health (NIH) (grant nos. 1DP2HL127720-01, 1UG3DK122644) (D.D.H.); NIH grant no. R01DK124369 (K.E.H.); Children’s Hospital of Philadelphia Institutional Development Funds (K.E.H.); the National Science Foundation (grant no. CMMI:15-48571) (D.D.H.); the Ministry of Trade, Industry & Energy of the Republic of Korea (D.D.H.); the Kwanjeong Educational Foundation (S.E.P.); the Children’s Hospital of Philadelphia Gastrointestinal Epithelium Modeling Program (K.E.H.); and the University of Pennsylvania (D.D.H.). D.D.H. is a recipient of the NIH Director’s New Innovator Award and the Cancer Research Institute Technology Impact Award.

Author information

Authors and Affiliations

Authors

Contributions

S.E.P. designed the research, performed the experiments, analyzed the data and wrote the manuscript. S.K. performed experiments, analyzed data and created figures. J.P., A.G., J.C. and A.Y.Y. provided assistance in the experiments and the preparation of the manuscript. B.J.W. provided assistance in the analysis of the histological data. T.A.K. and K.E.H. provided materials and assistance in the experiments and analyzed the data. D.D.H. designed the research, analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Dan Dongeun Huh.

Ethics declarations

Competing interests

D.D.H. is a cofounder of Vivodyne Inc. and holds equity in Vivodyne Inc. and Emulate Inc. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Maxime Mahe and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Madhura Mukhopadhyay, 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 Co-culture of organoids in OCTOPUS.

Co-culture of mouse liver and intestinal organoids in OCTOPUS. The OCTOPUS insert shown in this example contains two juxtaposed spiral chambers with independent access ports. Scale bars, 300 μm (left image) and 100 μm (right images).

Extended Data Fig. 2 Diffusion of soluble factors in OCTOPUS.

a, b. Visualization of 70 kDa FITC-dextran diffusion into the inner and outer regions of the hydrogel scaffolds in Matrigel drop (a) and OCTOPUS (b). The organoids in the inner and outer regions were located 600 μm (OCTOPUS)/2400 μm (Matrigel drop) and 400 μm (both groups) from the hydrogel surface, respectively. Scale bars, 100 μm. c. Temporal profiles of mean fluorescence intensity (MFI) due to dextran diffusion. d, e. Visualization of temporal changes in oxygen concentration in the inner and outer regions of Matrigel scaffolds in conventional drop culture (d) and OCTOPUS (e). Quenching of blue fluorescence shown in the micrographs was caused by an increase in the level of oxygen. f. Temporal profiles of normalized fluorescence intensity due to oxygen diffusion into Matrigel (n=3 independent experiments).

Extended Data Fig. 3 Culture of mouse liver organoids in OCTOPUS.

a, b. Formation and extended culture of mouse liver organoids in OCTOPUS and conventional drop culture. Scale bars, 100 μm (n=5 biologically independent experiments). c. Confocal micro-graphs showing the expression of albumin (ALB) in mouse liver organoids. Scale bars, 10 μm. ELISA analysis of (d) albumin and (e) urea in conditioned media (n=3 biologically independent experiments). All data are presented as mean ± SEM and P values are from unpaired, two-sided t test.

Source data

Extended Data Fig. 4 Maturation of intestinal organoids in OCTOPUS.

a. Comparison of bud length. Confocal micrographs show the cross-section of buds extending from the main body of the organoids. In each image, the white dashed lines indicate the approximated positions of the base and tip of a bud. Organoids in OCTOPUS generate more elongated buds. Scale bars, 20 μm. b. Comparison of the number and size of organoid buds in vitro to those of mouse intestinal crypts in vivo. c, d. Quantification of Ki67, Lgr5, and EdU expression in mouse intestinal organoids (n = 5 and 3 biologically independent experiments for (c) and (d), respectively). e. Organoids developing in OCTOPUS exhibit increased expression of Hnf4α, a marker of mature intestinal epithelial cells, compared to the control group in Matrigel drop. Scale bars, 10 μm. f. Quantification of the fraction of Hnf4α+ cells and the level of Hnf4α expression. Immunofluorescence of Hnf4α was normalized with respect to the number of cells (n = 4). g. The graph shows the cellular composition of the intestinal epithelium in OCTOPUS. Quantification was performed by measuring the immunofluorescence of cell type-specific markers described in Fig. 2m. Undifferentiated cells were identified by positive DAPI staining without the expression of differentiation markers. The data indicate the abundance of enterocytes (79% of the differentiated cell population at day 14) (n = 3). All data are presented as mean ± SEM and P values are from unpaired, two-sided t test.

Source data

Extended Data Fig. 5 Effect of co-culture on organoid development in OCTOPUS.

a. Co-culture of intestinal organoids and fibroblasts to mimic the epithelial-stromal unit of the intestine. Scale bar, 300 μm. b, c. In comparison to monoculture, the co-culture organoids are larger and express higher levels of Hnf4α expression. Scale bars, 100 μm (b), and 10 μm (c). All data are presented as mean ± SEM and P values are from unpaired, two-sided t test (n = 5 biologically independent experiments).

Source data

Extended Data Fig. 6 Functional characterization of intestinal organoids in OCTOPUS.

a. Immunofluorescence analysis of PEPT1, a nutrient transporter responsible for intestinal uptake of peptides. In this analysis, organoids in Matrigel drops at the maximum duration of culture (7 days) were compared to those maintained in OCTOPUS for 14 days to examine the contribution of extended culture. Organoids in OCTOPUS exhibit robust immunostaining of PEPT1 predominantly in the villus domain of the organoids. PEPT1 expression in this region is restricted to the apical surface of the epithelium facing the organoid lumen (L), matching the localized distribution of PEPT1 on the brush border membrane of the native intestinal epithelium. This polarized expression of PEPT1 is also observed in the villus surface of the organoids in Matrigel drop culture but the level of expression is significantly lower than that in OCTOPUS. Scale bars, 20 μm. b. Comparison of GLUT immunofluorescence in the villus domain of organoids. GLUT is ex-pressed on both the apical and basal surfaces of the villus epithelium in OCTOPUS. This trans-porter is also present in drop culture but its immunofluorescence is much weaker. Scale bars, 20 μm (n = 3 biologically independent experiments). c. Imaging and quantification of intracellular calcium signaling in intestinal organoids treated with 100 μM ATP. In the plots of relative intensity, the time between organoid stimulation and maximum fluorescence intensity is shaded in pink. Data were normalized to fluorescence intensity at the initial time point (t = 0 min). Scale bars, 100 μm (n = 3). d. Fraction of responsive organoids. The fraction of organoids that respond to ATP and glucose stimulation is larger in OCTOPUS. e. ELISA analysis of an active form of GLP-1 and MUC2 secreted by intestinal organoids. Both analytes are produced in significantly higher concentrations in OCTOPUS, which also continue to increase over time during extended culture (n = 3 biologically independent experiments). All data are presented as mean ± SEM and P values are from unpaired, two-sided t test.

Extended Data Fig. 7 Passaging and expansion of intestinal organoids in OCTOPUS.

a. Experimental procedure for passaging organoids in OCTOPUS. Subculture and expansion of (b) mouse and (c) human intestinal organoids grown in OCTOPUS and Matrigel drop. For comparison, culture conditions (for example, seeding density, hydrogel volume, media composition) were kept the same between the two systems. After 7 days of culture, organoids at a given passage number (Passage N) were physically dissociated and then transferred to new OCTOPUS devices or Matrigel drops (Passage N+1) at densities of 200 crypts/100 μl and 100 crypts/60 μl for mouse and human intestinal organoids, respectively. White solid lines in the micrographs show the outline of culture chambers in the OCTOPUS device or a sessile drop of Matrigel. Scale bars, 1 mm.

Extended Data Fig. 8 IBD enteroids cultured in Matrigel drops.

a. When cultured in Matrigel drops, IBD enteroids show properly polarized epithelial cells (top right) that resemble those in the epithelium of normal enteroids. In comparison to IBD enteroids in OCTOPUS, they also retain the structural integrity of the epithelium as visualized by ZO-1 expression (bottom right). Scale bars, 5 μm. b. UMAP plots showing the expression of two representative LINC genes by IBD and normal enteroids cultured in Matrigel drops. c. UMAP plots showing the expression of 4 representative IBD-associated genes by IBD and normal enteroids cultured in Matrigel drops.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–23, Tables 1 and 2, and captions for videos.

Reporting Summary

Supplementary Video 1

How to use OCTOPUS. OCTOPUS can be easily and quickly set up in a standard multi-well plate. In the first step demonstrated in this video, the OCTOPUS inserts are placed in a 12-well plate. Next, a stem cell-containing ECM hydrogel precursor solution is injected manually into each device through an open access port at the center of the device using a 200-μl pipette tip—this step can be performed by an automated fluid handling system as shown in the lower inset of Fig. 1m. After gelation, organoid media can be pipetted directly into the OCTOPUS-containing wells to initiate organoid culture (not shown in the video).

Supplementary Video 2

Injection and distribution of hydrogel solution. This video shows a fluorescence view of hydrogel injection into OCTOPUS. The ECM solution is injected into the loading chamber without leakage, which is immediately followed by the flow and rapid distribution of the injected solution into the radial culture chambers. For the purpose of visualization, a total volume of 100 μl of Matrigel was mixed with 1-μm fluorescent (red) microbeads. The entire culture chambers were filled within 5 s (not shown in the video).

Supplementary Video 3

Diffusion of FITC-dextran into hydrogel scaffolds. The video shows direct comparison of FITC-dextran (70 kDa) diffusion into the inner regions of hydrogel scaffolds in OCTOPUS and Matrigel drop. In OCTOPUS, fluorescence becomes visible throughout the scaffold within 30 min, illustrating rapid and uniform diffusive transport.

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Extended Data Fig. 3

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 5

Numerical source data.

Source Data Extended Data Fig. 8

Numerical source data.

Rights and permissions

Springer Nature or its licensor 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

Park, S.E., Kang, S., Paek, J. et al. Geometric engineering of organoid culture for enhanced organogenesis in a dish. Nat Methods 19, 1449–1460 (2022). https://doi.org/10.1038/s41592-022-01643-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01643-8

This article is cited by

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