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:

Cloning and variation of ground state intestinal stem cells

Abstract

Stem cells of the gastrointestinal tract, pancreas, liver and other columnar epithelia collectively resist cloning in their elemental states. Here we demonstrate the cloning and propagation of highly clonogenic, ‘ground state’ stem cells of the human intestine and colon. We show that derived stem-cell pedigrees sustain limited copy number and sequence variation despite extensive serial passaging and display exquisitely precise, cell-autonomous commitment to epithelial differentiation consistent with their origins along the intestinal tract. This developmentally patterned and epigenetically maintained commitment of stem cells is likely to enforce the functional specificity of the adult intestinal tract. Using clonally derived colonic epithelia, we show that toxins A or B of the enteric pathogen Clostridium difficile recapitulate the salient features of pseudomembranous colitis. The stability of the epigenetic commitment programs of these stem cells, coupled with their unlimited replicative expansion and maintained clonogenicity, suggests certain advantages for their use in disease modelling and regenerative medicine.

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

Figure 1: Cloning stem cells from fetal intestine.
Figure 2: Stem cells from fetal small intestine.
Figure 3: Stem cells of fetal colon.
Figure 4: Differential gene expression in stem cells of stratified and columnar epithelia.
Figure 5: Genomic stability of ISC in culture.
Figure 6: C. difficile toxin B effects on in vitro-generated colonic epithelia.

Similar content being viewed by others

Accession codes

Primary accessions

European Nucleotide Archive

Gene Expression Omnibus

Data deposits

Data sets generated for this study have been submitted to the National Center for Biotechnology Information Gene Expression Omnibus (GEO) database and the European Nucleotide Archive under accession numbers GSE66749 and SRP056402.

References

  1. Tabar, V. & Studer, L. Pluripotent stem cells in regenerative medicine: challenges and recent progress. Nature Rev. Genet. 15, 82–92 (2014)

    Article  CAS  PubMed  Google Scholar 

  2. Okano, H. & Yamanaka, S. iPS cell technologies: significance and applications to CNS regeneration and disease. Mol. Brain 7, 22 (2014)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Müller, A. M. & Dzierzak, E. A. ES cells have only a limited lymphopoietic potential after adoptive transfer into mouse recipients. Development 118, 1343–1351 (1993)

    Article  PubMed  Google Scholar 

  4. Helgason, C. D., Sauvageau, G., Lawrence, H. J., Largman, C. & Humphries, R. K. Overexpression of HOXB4 enhances the hematopoietic potential of embryonic stem cells differentiated in vitro. Blood 87, 2740–2749 (1996)

    Article  CAS  PubMed  Google Scholar 

  5. Bonde, S., Dowden, A. M., Chan, K. M., Tabayoyong, W. B. & Zavazava, N. HOXB4 but not BMP4 confers self-renewal properties to ES-derived hematopoietic progenitor cells. Transplantation 86, 1803–1809 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Iuchi, S., Dabelsteen, S., Easley, K., Rheinwald, J. G. & Green, H. Immortalized keratinocyte lines derived from human embryonic stem cells. Proc. Natl Acad. Sci. USA 103, 1792–1797 (2006)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Amabile, G. et al. In vivo generation of transplantable human hematopoietic cells from induced pluripotent stem cells. Blood 121, 1255–1264 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Suzuki, N. et al. Generation of engraftable hematopoietic stem cells from induced pluripotent stem cells by way of teratoma formation. Mol. Ther. 21, 1424–1431 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rheinwatd, J. G. & Green, H. Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell 6, 331–343 (1975)

    Article  Google Scholar 

  10. Rama, P. et al. Limbal stem-cell therapy and long-term corneal regeneration. N. Engl. J. Med. 363, 147–155 (2010)

    Article  CAS  PubMed  Google Scholar 

  11. Senoo, M., Pinto, F., Crum, C. P. & McKeon, F. p63 is essential for the proliferative potential of stem cells of stratified epithelia. Cell 129, 523–536 (2007)

    Article  CAS  PubMed  Google Scholar 

  12. Kumar, P. A. et al. Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell 147, 525–538 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Matsuura, R. et al. Crucial transcription factors in endoderm and embryonic gut development are expressed in gut-like structures from mouse ES cells. Stem Cells 24, 624–630 (2006)

    Article  PubMed  CAS  Google Scholar 

  14. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009)

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Ootani, A. et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nature Med. 15, 701–706 (2009)

    Article  CAS  PubMed  Google Scholar 

  16. Sato, T. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418 (2011)

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Fordham, R. P. et al. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell 13, 734–744 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Middendorp, S. et al. Adult stem cells in the small intestine are intrinsically programmed with their location-specific function. Stem Cells 32, 1083–1091 (2014)

    Article  CAS  PubMed  Google Scholar 

  19. Yin, X. et al. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nature Methods 11, 106–112 (2014)

    Article  CAS  PubMed  Google Scholar 

  20. Kim, K. A. et al. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science 309, 1256–1259 (2005)

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Dreesen, O. & Brivanlou, A. H. Signaling pathways in cancer and embryonic stem cells. Stem Cell Rev. 3, 7–17 (2007)

    Article  CAS  PubMed  Google Scholar 

  22. Zhu, L. et al. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature 457, 603–607 (2009)

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  24. Powell, A. E. et al. The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 149, 146–158 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Botrugno, O. A. et al. Synergy between LRH-1 and beta-catenin induces G1 cyclin-mediated cell proliferation. Mol. Cell 15, 499–509 (2004)

    Article  CAS  PubMed  Google Scholar 

  26. Lessard, J. & Sauvageau, G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 423, 255–260 (2003)

    Article  ADS  CAS  PubMed  Google Scholar 

  27. Sangiorgi, E. & Capecchi, M. R. Bmi1 is expressed in vivo in intestinal stem cells. Nature Genet. 40, 915–920 (2008)

    Article  CAS  PubMed  Google Scholar 

  28. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Metcalfe, C., Kljavin, N. M., Ybarra, R. & de Sauvage, F. J. Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration. Cell Stem Cell 14, 149–159 (2014)

    Article  CAS  PubMed  Google Scholar 

  30. Battle, M. A. et al. GATA4 is essential for jejunal function in mice. Gastroenterology 135, 1676–1686 (2008)

    Article  CAS  PubMed  Google Scholar 

  31. Walker, E. M., Thompson, C. A. & Battle, M. A. GATA4 and GATA6 regulate intestinal epithelial cytodifferentiation during development. Dev. Biol. 392, 283–294 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dusing, M. R., Maier, E. A., Aronow, B. J. & Wiginton, D. A. Onecut-2 knockout mice fail to thrive during early postnatal period and have altered patterns of gene expression in small intestine. Physiol. Genomics 42, 115–125 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. International Stem Cell Initiative. Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage. Nature Biotechnol. 29, 1132–1144 (2011)

  34. Avery, S. et al. BCL-XL mediates the strong selective advantage of a 20q11.21 amplification commonly found in human embryonic stem cell cultures. Stem Cell Rep. 1, 379–386 (2013)

    Article  CAS  Google Scholar 

  35. Shultz, L. D. et al. Human cancer growth and therapy in immunodeficient mouse models. Cold Spring Harb Protoc. http://dx.doi.org/10.1101/pdb.top073585 (2014)

  36. Voth, D. E. & Ballard, J. D. Clostridium difficile toxins: mechanism of action and role in disease. Clin. Microbiol. Rev. 18, 247–263 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Carter, G. P., Rood, J. I. & Lyras, D. The role of toxin A and toxin B in the virulence of Clostridium difficile. Trends Microbiol. 20, 21–29 (2012)

    Article  CAS  PubMed  Google Scholar 

  38. Lyras, D. et al. Toxin B is essential for virulence of Clostridium difficile. Nature 458, 1176–1179 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Farrow, M. A. et al. Clostridium difficile toxin B-induced necrosis is mediated by the host epithelial cell NADPH oxidase complex. Proc. Natl Acad. Sci. USA 110, 18674–18679 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Huelsenbeck, J. et al. Upregulation of the immediate early gene product RhoB by exoenzyme C3 from Clostridium limosum and toxin B from Clostridium difficile. Biochemistry 46, 4923–4931 (2007)

    Article  CAS  PubMed  Google Scholar 

  41. Aktories, K., Schmidt, G. & Just, I. Rho GTPases as targets of bacterial protein toxins. Biol. Chem. 381, 421–426 (2000)

    Article  CAS  PubMed  Google Scholar 

  42. MacFie, T. S. et al. DUOX2 and DUOXA2 form the predominant enzyme system capable of producing the reactive oxygen species H2O2 in active ulcerative colitis and are modulated by 5-aminosalicylic acid. Inflamm. Bowel Dis. 20, 514–524 (2014)

    Article  PubMed  Google Scholar 

  43. Cheng, H. & Leblond, C. P. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. V. Unitarian theory of the origin of the four epithelial cell types. Am. J. Anat. 141, 537–561 (1974)

    Article  CAS  PubMed  Google Scholar 

  44. Yui, S. et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell. Nature Med. 18, 618–623 (2012)

    Article  CAS  PubMed  Google Scholar 

  45. Wang, F. et al. Isolation and characterization of intestinal stem cells based on surface marker combinations and colony-formation assay. Gastroenterology 145, 383–395 (2013)

    Article  CAS  PubMed  Google Scholar 

  46. Watson, C. L. et al. An in vivo model of human small intestine using pluripotent stem cells. Nature Med. 20, 1310–1314 (2014)

    Article  CAS  PubMed  Google Scholar 

  47. Sheaffer, K. L. & Kaestner, K. H. Transcriptional networks in liver and intestinal development. Cold Spring Harb. Perspect. Biol. 4, a008284 (2012)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Nicholson, J. K. et al. Host-gut microbiota metabolic interactions. Science 336, 1262–1267 (2012)

    Article  ADS  CAS  PubMed  Google Scholar 

  49. Brandl, K. & Beutler, B. Creating diseases to understand what prevents them: genetic analysis of inflammation in the gastrointestinal tract. Curr. Opin. Immunol. 24, 678–685 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lees, C. W., Barrett, J. C., Parkes, M. & Satsangi, J. New IBD genetics: common pathways with other diseases. Gut 60, 1739–1753 (2011)

    Article  CAS  PubMed  Google Scholar 

  51. Schmidt, D., Hübsch, U., Wurzer, H., Heppt, W. & Aufderheide, M. Development of an in vitro human nasal epithelial (HNE) cell model. Toxicol. Lett. 88, 75–79 (1996)

    Article  CAS  PubMed  Google Scholar 

  52. Chumbler, N. M. et al. Clostridium difficile toxin B causes epithelial cell necrosis through an autoprocessing-independent mechanism. PLoS Pathog. 8, e1003072 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang, K. et al. PennCNV: an integrated hidden Markov model designed for high-resolution copy number variation detection in whole-genome SNP genotyping data. Genome Res. 17, 1665–1674 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler Transform. Bioinformatics 25, 1754–1760 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009)

    Article  PubMed  PubMed Central  Google Scholar 

  58. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from Connecticut Innovations (W.X., F.M.), the Joint Council Office of the Agency for Science Technology Research Agency (A*STAR), Singapore (W.X., F.M.), the National Medical Research Council, Singapore (BNB101677A to H.K.Y., F.M., and W.X.; BnB11dec063 to N.N., F.M. and W.X.), the Department of Defense (W81XWH-10-1-0289 to C.P.C.) and the National Institute of Health (AI09575504 to D.B.L.). We thank M. LaLande, B. Lane and B. Seet for support, B. Tennent, B. Knowles and T. McLaughlin for comments on the manuscript, J. Hammer for artwork, L. Lapierre and J. Franklin for histology evaluation. We thank H. Green for advice and support.

Author information

Authors and Affiliations

Authors

Contributions

Experimental design and conception were done by W.X., F.M., D.B.L., K.Y.H. and C.P.C.; X.W. cloned and differentiated the intestinal stem cells with help from L.H.W., F.K., G.N., B.E.H. and Y.H.; Y.Y., X.W. prepared the genomic and gene expression analyses together with F.K., G.N., C.C.K. and L.W.; T.Z., D.B. and N.N. performed all computational and bioinformatics work. B.H. and C.P.C. obtained fetal tissues and F.A.S., J.S.H. and T.D. provided endoscopic biopsies, and R.B. analysed the xenografts. The C. difficile experiments were designed and executed by B.C., L.H.W., M.A.F. and D.B.L.; W.X. and F.M. wrote the manuscript with input from all other authors.

Corresponding authors

Correspondence to Frank McKeon or Wa Xian.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Loss of clonogenicity in differentiated ISC.

a, Schematic of ISC differentiation using either the γ-secretase inhibitor dibenzazepine (DBZ) or withdrawal of the Wnt regulator R-spondin 1 (Rspo1). ISCs were plated on day 0, DBZ added or Rspo1 removed at day 2, and colonies passaged en masse at day 7. At day 14, after 7 days of continuous growth, colonies were counted. b, Micrographs show immunofluorescence at day 7 colonies grown without Rspo1 or in the presence of DBZ for 5 days using antibodies to Ki67, chromogranin A (CHGA), keratin 20 (Krt20), E-cadherin (E-cad), and mucin 2 (Muc2). Scale bar, 50 μm; n = 4 technical replicates. c, Histogram shows colony formation in each condition normalized to control ISCs. n = 4 biological replicates; error bars, s.d. d, Staining of ALI-differentiated intestinal stem cells with monoclonal antibody HD6 directed to Paneth cells. Scale bar, 50 μm; n = 4 technical replicates.

Extended Data Figure 2 Intestinal stem cell expression profiles.

a, List of genes differentially expressed in ISC derived from duodenum, jejunum and ileum. These data correspond to heat map of Fig. 2b. b, Immunofluorescence labelling of ALI-differentiated ISCs from duodenum with antibodies against Tff2, mucin 5AC, villin, E-cadherin, and mucin 2. c, Immunofluorescence labelling of ALI-differentiated epithelia from jejunum stem cells with antibodies to E-cadherin, mucin 2, villin, and mucin 5AC. Scale bar, 50 μm; n = 10 technical replicates.

Extended Data Figure 3 Differential gene expression in epithelia derived from colonic stem cells.

Heat map of differentially expressed (>1.5-fold, P < 0.05) genes in ALI cultures derived from stem cell pedigrees of ascending, transverse, and descending colon.

Extended Data Figure 4 Differential gene expression across columnar and stratified epithelial stem cells.

a, Histograms of expression microarray signal intensity of selected genes across averaged intestine and colon ISCs, stratified epithelial stem cells, and stem cells of the fallopian tube (FT). Biological replicas n = 2–6 (FT = 2, stratified epithelia = 3, colon, intestine = 6); error bars, s.d. b, Dot plot showing expression microarray data of indicated genes for stem cell pedigrees (ISC; Duo, duodenum; Jej, jejunum; Ile, ileum; AC, ascending colon; TC, transverse colon; DC, descending colon) derived from various regions of the intestinal tract before and after air–liquid interface (ALI) differentiation. Biological replicas n = 2 (total 12 data sets) for stem cells, technical replicas n = 2 for ALI. c, Chart of aggregate P values by Student's t-test for gene expression changes between ground state stem cells and their ALI-differentiated counterparts.

Extended Data Figure 5 Genes affected by CNV and SNV events in intestinal stem cell pedigrees during passaging.

a, Summary of CNV (events (genes affected)) and non-synonymous SNV in pedigrees 1 and 2 at P5 to P20. b, Summary of genes altered by interstitial CNV amplifications (top) or deletions (bottom) in ISC pedigrees 3 to 7 at P5 and P25. c, Summary of genes sustaining non-synonymous SNV in five ISC pedigrees at P5 and P25.

Extended Data Figure 6 Whole-genome CNV profiles for intestinal stem cell pedigrees 3–7 at P5 and P25.

Regions marked by ovals represent aneuploidy.

Extended Data Figure 7 Impact of ISCGS passaging on ALI differentiation.

ALI differentiation of intestinal pedigree 2 initiated from cells at the indicated passage number. As indicated, histological sections of differentiated epithelia were stained with antibodies to either E-cadherin (ECAD, green) and mucin 2 (Muc2, red), or Ki67 (green) and chromogranin A (CHGA, red). Scale bar, 75 μm; n = 4 technical replicates.

Extended Data Figure 8 ISCGS tumorigenicity assays in immunodeficient mice.

a, Quantification of tumour formation assessments at 4–16 weeks following subcutaneous inoculation of two million cells of the indicated ISC pedigrees at passage 6 or passage 25 at 4–16 weeks. ‘Pool’ indicates total set of clones derived from P0 ileum culture before pedigree generation. ‘Cancer cells’ refers to propagating cells from case of high-grade serous ovarian cancer. b, Left, histological section through site of injection of 1 million cells from pedigree 3. Right, section of injection site stained with antibody (STEM121) to human epithelial cells (brown) revealing benign cysts. Scale bar, 15 μm.

Extended Data Figure 9 Dose- and time-dependency of TcdB pathology in ALI-generated colonic epithelia.

a, Immunofluorescence localization of adherens junction marker E-cadherin and tight junction marker claudin 3 in ALI-differentiated epithelia derived from transverse colon stem cells following exposure to 100 pM TcdB for the indicated durations. n = 4 technical replicates. Scale bar, 100 μm. b, Representative H&E images of ALI cultures at indicated times and concentration of TcdB exposure. Scale bar, 250 μm; n = 4 technical replicates. c, Gene set enrichment analysis of whole-genome expression data from colonic epithelia treated with 500pM TcdB for 24 h and control samples showing enriched KEGG pathway sets. NES, normalized enrichment score; NOM P value, nominal P value. d, 3D plot of upregulated genes at the indicated time points and dosages > twofold, P < 0.05). n = 2 technical replicates. e, Heat map of upregulated genes in 500 pM TcdB samples. The genes (237 genes) were chosen by cutoff values (> twofold, P < 0.05). Three time points (8, 16 and 24 h) are shown. f, 3D plot of downregulated genes at the indicated time points and dosages > twofold, P < 0.05). n = 2 technical replicates.

Extended Data Figure 10 Dose- and time-dependency of TcdA pathology in ALI-generated colonic epithelia.

a, Left, representative H&E images of ALI cultures at indicated times and concentration of TcdA exposure; right, immunofluorescence localization of adherens junction marker E-cadherin (ECAD; green) and mucin 2 (MUC2; red) in ALI-differentiated epithelia derived from transverse colon stem cells following incubation with 10 nM TcdA for the indicated durations. Scale bar, 100 μm; n = 4 technical replicates. b, 3D plot of histological scoring of representative H&E time points and concentrations performed by a gastrointestinal pathologist according to a standard 0–3 rating for colonic epithelial integrity. c, Distribution of tight junction marker claudin 3 (Cldn3) and adherens junction marker (Cdh17) following treatment of ALI colonic epithelium with TcdA for the indicated times and doses. Scale bar, 50 μm; n = 4 technical replicates. d, Histogram of permeability of ALI colonic epithelium (Papp) to small molecules (FD4, molecular mass 4,400 Da) following exposure to the indicated doses of TcdA for the indicated times.

Supplementary information

Supplementary Tables

This file contains Supplementary Tables 1-3 and a further table containing a summary of CNV and exome capture sequencing run analysis for intestinal stem cells. (PDF 334 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, X., Yamamoto, Y., Wilson, L. et al. Cloning and variation of ground state intestinal stem cells. Nature 522, 173–178 (2015). https://doi.org/10.1038/nature14484

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14484

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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