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:

Somatic inflammatory gene mutations in human ulcerative colitis epithelium

Abstract

With ageing, normal human tissues experience an expansion of somatic clones that carry cancer mutations1,2,3,4,5,6,7. However, whether such clonal expansion exists in the non-neoplastic intestine remains unknown. Here, using whole-exome sequencing data from 76 clonal human colon organoids, we identify a unique pattern of somatic mutagenesis in the inflamed epithelium of patients with ulcerative colitis. The affected epithelium accumulates somatic mutations in multiple genes that are related to IL-17 signalling—including NFKBIZ, ZC3H12A and PIGR, which are genes that are rarely affected in colon cancer. Targeted sequencing validates the pervasive spread of mutations that are related to IL-17 signalling. Unbiased CRISPR-based knockout screening in colon organoids reveals that the mutations confer resistance to the pro-apoptotic response that is induced by IL-17A. Some of these genetic mutations are known to exacerbate experimental colitis in mice8,9,10,11, and somatic mutagenesis in human colon epithelium may be causally linked to the inflammatory process. Our findings highlight a genetic landscape that adapts to a hostile microenvironment, and demonstrate its potential contribution to the pathogenesis of ulcerative colitis.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Somatic mutations in UC epithelium.
Fig. 2: Targeted sequencing of bulk UC organoids.
Fig. 3: Phenotypic characterization of UC-related mutations.
Fig. 4: iNOS mediates IL-17A-induced apoptosis.

Similar content being viewed by others

Data availability

Gene expression datasets are available from the Gene Expression Omnibus (GEO) under the accession code GSE127757. Whole-exome sequencing data have been deposited to the Japanese Genotype-phenotype Archive under the accession number JGAS00000000199. Source data for Figs. 1, 3 and 4 and Extended Data Figs. 4, 6, 8 and 9 are provided with the paper. All relevant data that are included with this study are available from the corresponding author on reasonable request.

References

  1. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019).

    Article  ADS  PubMed  CAS  Google Scholar 

  4. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362, 911–917 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  5. Martincorena, I. et al. Tumor evolution. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  6. Martincorena, I. & Campbell, P. J. Somatic mutation in cancer and normal cells. Science 349, 1483–1489 (2015).

    Article  ADS  PubMed  CAS  Google Scholar 

  7. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  8. Maxwell, J. R. et al. Differential roles for interleukin-23 and interleukin-17 in intestinal immunoregulation. Immunity 43, 739–750 (2015).

    Article  PubMed  CAS  Google Scholar 

  9. Murthy, A. K., Dubose, C. N., Banas, J. A., Coalson, J. J. & Arulanandam, B. P. Contribution of polymeric immunoglobulin receptor to regulation of intestinal inflammation in dextran sulfate sodium-induced colitis. J. Gastroenterol. Hepatol. 21, 1372–1380 (2006).

    PubMed  CAS  Google Scholar 

  10. Lee, J. S. et al. Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 43, 727–738 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Nagahama, Y. et al. Regnase-1 controls colon epithelial regeneration via regulation of mTOR and purine metabolism. Proc. Natl Acad. Sci. USA 115, 11036–11041 (2018).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  12. Brentnall, T. A. et al. Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology 107, 369–378 (1994).

    Article  PubMed  CAS  Google Scholar 

  13. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Yamamoto, M. et al. Regulation of Toll/IL-1-receptor-mediated gene expression by the inducible nuclear protein IκBζ. Nature 430, 218–222 (2004).

    Article  ADS  PubMed  CAS  Google Scholar 

  16. Karlsen, J. R., Borregaard, N. & Cowland, J. B. Induction of neutrophil gelatinase-associated lipocalin expression by co-stimulation with interleukin-17 and tumor necrosis factor-alpha is controlled by IκB-ζ but neither by C/EBP-β nor C/EBP-δ. J. Biol. Chem. 285, 14088–14100 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Kaetzel, C. S., Robinson, J. K., Chintalacharuvu, K. R., Vaerman, J. P. & Lamm, M. E. The polymeric immunoglobulin receptor (secretory component) mediates transport of immune complexes across epithelial cells: a local defense function for IgA. Proc. Natl Acad. Sci. USA 88, 8796–8800 (1991).

    Article  ADS  PubMed  CAS  PubMed Central  Google Scholar 

  18. Kumar, P. et al. Intestinal interleukin-17 receptor signaling mediates reciprocal control of the gut microbiota and autoimmune inflammation. Immunity 44, 659–671 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Cao, A. T., Yao, S., Gong, B., Elson, C. O. & Cong, Y. Th17 cells upregulate polymeric Ig receptor and intestinal IgA and contribute to intestinal homeostasis. J. Immunol. 189, 4666–4673 (2012).

    Article  PubMed  CAS  Google Scholar 

  20. Chang, S. H., Park, H. & Dong, C. Act1 adaptor protein is an immediate and essential signaling component of interleukin-17 receptor. J. Biol. Chem. 281, 35603–35607 (2006).

    Article  PubMed  CAS  Google Scholar 

  21. Robles, A. I. et al. Whole-exome sequencing analyses of inflammatory bowel disease-associated colorectal cancers. Gastroenterology 150, 931–943 (2016).

    Article  PubMed  CAS  Google Scholar 

  22. Fujita, M. et al. Genomic landscape of colitis-associated cancer indicates the impact of chronic inflammation and its stratification by mutations in the Wnt signaling. Oncotarget 9, 969–981 (2018).

    Article  PubMed  Google Scholar 

  23. Baker, A. M. et al. Evolutionary history of human colitis-associated colorectal cancer. Gut 68, 985–995 (2019).

    Article  PubMed  CAS  Google Scholar 

  24. Din, S. et al. Mutational analysis identifies therapeutic biomarkers in inflammatory bowel disease-associated colorectal cancers. Clin. Cancer Res. 24, 5133–5142 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Brandtzaeg, P., Baklien, K., Fausa, O. & Hoel, P. S. Immunohistochemical characterization of local immunoglobulin formation in ulcerative colitis. Gastroenterology 66, 1123–1136 (1974).

    Article  PubMed  CAS  Google Scholar 

  26. Das, K. M., Erber, W. F. & Rubinstein, A. Immunohistochemical changes in morphologically involved and uninvolved colonic mucosa of patients with idiopathic proctitis. J. Clin. Invest. 59, 379–385 (1977).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Uchima, H., Eishi, Y., Takemura, T. & Hirokawa, K. Immunohistochemical studies of ulcerative colitis. With special reference to localization of immunoglobulins, secretory component, and lysozyme in view of suffering periods. Acta Pathol. Jpn. 33, 1183–1196 (1983).

    PubMed  CAS  Google Scholar 

  28. Garg, A. V. et al. MCPIP1 endoribonuclease activity negatively regulates interleukin-17-mediated signaling and inflammation. Immunity 43, 475–487 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Iwasaki, H. et al. The IκB kinase complex regulates the stability of cytokine-encoding mRNA induced by TLR-IL-1R by controlling degradation of regnase-1. Nat. Immunol. 12, 1167–1175 (2011).

    Article  PubMed  CAS  Google Scholar 

  30. Howell, K. J. et al. DNA methylation and transcription patterns in intestinal epithelial cells from pediatric patients with inflammatory bowel diseases differentiate disease subtypes and associate with outcome. Gastroenterology 154, 585–598 (2018).

    Article  PubMed  CAS  Google Scholar 

  31. Kimura, H. et al. Increased expression of an inducible isoform of nitric oxide synthase and the formation of peroxynitrite in colonic mucosa of patients with active ulcerative colitis. Gut 42, 180–187 (1998).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Kolios, G., Rooney, N., Murphy, C. T., Robertson, D. A. & Westwick, J. Expression of inducible nitric oxide synthase activity in human colon epithelial cells: modulation by T lymphocyte derived cytokines. Gut 43, 56–63 (1998).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Parikh, K. et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 567, 49–55 (2019).

    Article  ADS  PubMed  CAS  Google Scholar 

  34. Hueber, W. et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 61, 1693–1700 (2012).

    Article  PubMed  CAS  Google Scholar 

  35. McGovern, D. P., Kugathasan, S. & Cho, J. H. Genetics of inflammatory bowel diseases. Gastroenterology 149, 1163–1176 (2015).

    Article  PubMed  CAS  Google Scholar 

  36. Magro, F. et al. Third European evidence-based consensus on diagnosis and management of ulcerative colitis. Part 1: definitions, diagnosis, extra-intestinal manifestations, pregnancy, cancer surveillance, surgery, and ileo-anal pouch disorders. J. Crohns Colitis 11, 649–670 (2017).

    Article  PubMed  Google Scholar 

  37. Fujii, M. et al. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition. Cell Stem Cell 23, 787–793 (2018).

    Article  PubMed  CAS  Google Scholar 

  38. Fujii, M. et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell 18, 827–838 (2016).

    Article  PubMed  CAS  Google Scholar 

  39. Mihara, E. et al. Active and water-soluble form of lipidated Wnt protein is maintained by a serum glycoprotein afamin/α-albumin. eLife 5, e11621 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Seino, T. et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell 22, 454–467 (2018).

    Article  PubMed  CAS  Google Scholar 

  41. Drost, J. et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 521, 43–47 (2015).

    Article  ADS  PubMed  CAS  Google Scholar 

  42. Smyth, G. K. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, 1544-6115 (2004).

    Article  ADS  MathSciNet  MATH  Google Scholar 

  43. Planell, N. et al. Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut 62, 967–976 (2013).

    Article  PubMed  CAS  Google Scholar 

  44. Vanhove, W. et al. Strong upregulation of AIM2 and IFI16 inflammasomes in the mucosa of patients with active inflammatory bowel disease. Inflamm. Bowel Dis. 21, 2673–2682 (2015).

    Article  PubMed  Google Scholar 

  45. Ramilowski, J. A. et al. A draft network of ligand-receptor-mediated multicellular signalling in human. Nat. Commun. 6, 7866 (2015).

    Article  ADS  PubMed  CAS  Google Scholar 

  46. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Rep. 17, 1193–1205 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Koo, B. K. et al. Controlled gene expression in primary Lgr5 organoid cultures. Nat Methods 9, 81–83 (2012).

    Article  CAS  Google Scholar 

  49. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  ADS  PubMed  CAS  Google Scholar 

  50. Fujii, M., Matano, M., Nanki, K. & Sato, T. Efficient genetic engineering of human intestinal organoids using electroporation. Nat. Protocols 10, 1474–1485 (2015).

    Article  PubMed  CAS  Google Scholar 

  51. Guo, Q. et al. ‘Cold shock’ increases the frequency of homology directed repair gene editing in induced pluripotent stem cells. Sci. Rep. 8, 2080 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  52. Hsiau, T. et al. Inference of CRISPR edits from Sanger trace data. Preprint at bioRxiv https://doi.org/10.1101/251082 (2019).

Download references

Acknowledgements

This work was supported by AMED (grant numbers JP19cm0106206 and JP19gm5010002), AMED-CREST (grant number JP19gm1210001) and the JSPS KAKENHI (grant numbers JP17H06176 and JP26115007). K.T., Y.O. and M.F. were supported by the Japan Society for the Promotion of Science Research Fellowships for Young Scientists. We also thank the Collaborative Research Resources at the School of Medicine, Keio University for providing technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

K.N., M.F. and T.S. designed the study. T.H., S.I. and T.K. provided specimens. Y.O. and S.T. performed histochemical analysis. K.N., M.F., M.M., S.S., K.K. and K.I. performed sample preparation. K.N., M.F., M.M., S.N., A.T., S.S., K.I., K.Y. and N.S. performed organoid experiments. M.F., M.S., S.D., Y.N. and K.T. performed sequencing and bioinformatics analysis. R.I. supervised statistical analyses. K.N., M.F. and T.S. wrote the manuscript.

Corresponding author

Correspondence to Toshiro Sato.

Ethics declarations

Competing interests

T.S. is an inventor on several patents related to organoid culture.

Additional information

Peer review information Nature thanks Judy Cho, Ramnik Xavier and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Study design.

a, Nomenclature of organoids. b, Patient demographics and the breakdown of the organoids used for each analysis.

Extended Data Fig. 2 Mutational signatures of HC, UC and CAN organoids.

a, Relative contribution of the nucleotide transition patterns in each sample group in healthy (n = 28), UCuninf (n = 15), UCinf without CAN (n = 15) and UCinf with CAN (n = 17) organoids. Data are shown as the mean (±s.d.) relative contribution of base substitution types in each sample type. b, Cosine similarity of the mutational signature of each organoid clone to the COSMIC signatures (1–30). UCinf organoids with an MLH1 mutation (UC08T_IS1, UC08T_IS2) are highlighted in red. One organoid line with a contribution of APOBEC signatures (signatures 2 and 13) is highlighted in green characters (UC10T_IS). c, Number of truncating mutations in the indicated genes in healthy, UCuninf (n = 43) and UCinf (n = 31) organoids. Genes with two or more organoid clones that have truncating mutations are shown. MLH1-mutated organoids (UC08T_IS1, 2) were excluded from this analysis. *P = 0.004; q < 0.1 (two-sided Fisher's exact test with Benjamini–Hochberg adjustment). d, dN/dS ratios of NFKBIZ and PIGR (genes with pglobal < 0.05 and qglobal < 0.1), calculated using the whole-exome sequencing data.

Extended Data Fig. 3 Copy-number analysis of UC organoids.

a, Heat map showing the average minor allele frequencies (MAFs) of SNPs that were heterozygous in the normal counterpart (bin size 25 Mb). Genomic regions with LOH are indicated with red boxes. Genomic positions of PIGR, NFKBIZ and TRAF3IP2 are shown at the top. Organoid lines that were subjected to CGH–SNP array analysis are highlighted in red. Organoid lines with MLH1 mutations (UC08) are not shown. b, DNA copy-number analysis using CGH–SNP microarray analysis. For each organoid line, the genome-wide chromosome state is demonstrated by the log2-transformed signal ratio (top) and the allelic difference (bottom) values. Genomic regions with CN-LOH are shown in red. Deletions in common fragile sites are shown in green. Genetic lesions associated with UC-related mutations are shown in black. c, Loss of PIGR protein in UC12T_IA organoids with CN-LOH at the PIGR locus. Intact PIGR protein expression in UC12T_IS organoids is also shown. β-actin was used as a loading control. Data are representative of n = 2 technical replicates with similar results. d, Focal deletion of fragile sites in UCinf organoids, detected by CGH analysis. Chromosome states are visualized using the log2-transformed signal ratio. Deletions in chromosomal fragile sites at positions 3p14 and 16p13.2 were uniquely detected in the UCinf epithelium.

Extended Data Fig. 4 Characterization of UC-related mutations.

a, Increased expression of NFKBIZ mRNA in purified UC epithelium (E-MTAB-5464). NFKBIZ expression was compared between healthy control individuals (n = 11) and patients with UC (n = 11) using the exact negative binomial test in edgeR (P = 5.0 × 10−6). Bars represent the median. b, Gene set enrichment analysis using public datasets (GSE38713, left; GSE59071, right) and in-house IL-17 target genes (Supplementary Table 3). NES; normalized enrichment score. Left, n = 15 (active colitis), n = 13 (healthy control); right, n = 82 (active colitis), n = 11 (healthy control). ch, IL-17A-induced expression of NFKBIZ (IκBζ) and PIGR in colon organoids. Organoids were stimulated with 100 ng ml−1 IL-17A for 3 h (c, f) or 24 h (d, e, g, h). The expression of NFKBIZ (IκBζ) and PIGR in control, NFKBIZ-mutant and TRAF3IP2-mutant organoids was analysed by quantitative PCR with reverse transcription (c, f) and immunoassay (d, e, g, h). Matched UCuninf organoids that were derived from the same patients were used as controls. mRNA expression levels are shown relative to the expression of ACTB. Expression levels of PIGR protein relative to β-actin are shown at the bottom of the pseudo-blot images. Results are shown from two (ce) and one (fh) biologically independent experiments, and two technical replicates (g, h). Data are shown as mean + s.e.m. (c, f). i, Genetic mutations detected by whole-exome sequencing in CAN organoids. Patterns of mutations are specified using the same symbols as Fig. 2a.

Source data

Extended Data Fig. 5 Targeted sequencing of UC-related genes.

a, Consistency of VAFs between fresh epithelium samples and organoids (n = 11 pairs). Red dots and lines show truncating and non-synonymous mutations in UC-mutated genes (NFKBIZ, PIGR, IL17RA, TRAF3IP2 and ZC3H12A) (n = 12 mutations). Black dots and lines show synonymous mutations in genes that are mutated in UC and any mutations in the other genes (n = 19 mutations). VAFs did not significantly differ between the sample pairs (P = 1, two-sided Wilcoxon signed-rank test). b, dN/dS ratios of the indicated genes in the targeted sequencing data. Genes with with q-values <0.1 (that is, statistically significant) are shown. c, Genetic mutations in UCuninf and control organoids, detected by targeted sequencing. Top, for each gene, the number of different variants is shown by density. Bottom, the VAF in each analysed sample. Each dot represents the fraction of each altered allele per total reads mapped to the gene. Genes are shown in different colours as specified on the left. d, PIGR (top), secretory IgA (middle) and cytokeratin20 (CK20) (bottom) immunostaining of UCinf epithelium (UC09T_IS2 and UC17T_IA) with focal loss of PIGR and secretory IgA expression. Magnified areas show boundaries between regions in which PIGR expression is intact and regions in which PIGR expression is lost. Images are representative images of four regions in which PIGR expression is lost, in eight patients with UC. Scale bars, 500 μm (low magnification); 100 μm (high magnification).

Extended Data Fig. 6 Targeted sequencing of UC-related genes (continued).

a, b, Reduced response to treatment with IL-17A in patient-derived cloned ZC3H12AD437Y UCinf organoids (UC23N_IS) in comparison to wild-type counterpart organoids (UC23N_UA). Results are representative of n = 2 technical replicates with similar results. c, Design of CRISPR–Cas9-mediated ZC3H12AD437Y knock-in. The donor vector contains two silent mutations at the protospacer adjacent motifs flanking the CRISPR targets, in addition to the D437Y mutation. Each allele of the knocked-in clone used for analysis is shown on the right. The clone is heterozygous for a knocked-in D437Y mutation and a 42-bp in-frame deletion. d, e, Reduced response to treatment with IL-17A in genetically engineered ZC3H12AD437Y-KI organoids compared to parental wild-type organoids. Results are representative of n = 2 technical replicates with similar results. f, Per-patient data of the targeted sequencing result. For each patient, mutations in organoids that were derived from the most-distal sample are shown. Top, for each gene, the number of different variants is shown by density. Bottom, dots represent the VAF of each variant. Genes are shown in different colours as specified on the left. Data are shown as mean + s.e.m. (a, d).

Source data

Extended Data Fig. 7 Targeted sequencing of cytokine and PAMP receptor genes and cancer driver genes.

ac, Mutations in cytokine and PAMP receptor genes (a) and sporadic colon cancer driver genes (b, c) identified in UCinf, UCuninf and control organoids by targeted sequencing. Number of different variants per gene are shown by density (top). Top, for each gene, the number of different variants is shown by density. Bottom, dots represent the VAF of each variant. Genes are shown in different colours as specified on the left. d, Sporadic colon cancer gene mutations identified in CAN organoids.

Extended Data Fig. 8 IL-17A-induced cytotoxic response in human colon organoids.

a, Fold-change values of individual sgRNAs in two experiments that used different lines of organoids. sgRNAs are coloured according to their targeting genes, as specified at the top. b, Response of wild-type organoids to treatment with IL-17A in differing growth factor conditions. The organoids showed a cytotoxic response to treatment with IL-17A only in the absence of Noggin. The WNRAIF condition is the medium condition including WNT3A, Noggin, R-spondin-1 (Rspo), A83-01, IGF-1 and FGF-2, but lacking EGF. Data are representative of n = 2 biologically independent experiments with similar results. c, sgRNA targets and confirmation of CRISPR–Cas9-mediated knockout of IL17RA, TRAF3IP2 and NFKBIZ. For knockout of NFKBIZ, three NFKBIZKO lines (#1–3) were generated from different parental organoids. Successful knockout was confirmed by Sanger sequencing in all lines. Loss of IκBζ protein was also confirmed by an immunoassay using an organoid stimulated with IL-17A for 24 h. Result is from a single experiment. d, IL17RAKO and TRAF3IP2KO organoids are resistant to treatment with IL-17A in a Noggin-deficient condition. Right, dots show the relative area of organoids in each well. Results are representative of n = 3 technical replicates. e, NFKBIZKO organoid line 1 tolerates treatment with IL-17A. The other two knockout lines showed a similar response. f, Patient-derived NFKBIZ-mutant organoids (UC02N_IS) are resistant to treatment with IL-17A. Wild-type counterpart organoids (UC02N_UA) were used as a control. Data are representative of n = 2 biologically independent experiments with similar results. g, Patient-derived ZC3H12AD437Y-mutant organoids (UC23N_IS) are tolerant of treatment with IL-17A compared to their wild-type counterparts (UC23N_UA). Data are representative of n = 2 technical replicates with similar results. h, Knockout of TP53 mitigates IL-17A-induced cytotoxicity. Data are representative of n = 2 biologically independent experiments with similar results. Mean + s.e.m (dh). P values determined by two-sided Welch’s t-test. Scale bars, 1 mm (b, dh).

Source data

Extended Data Fig. 9 iNOS-dependent apoptosis in human colon organoids treated with IL-17A.

a, Detection of cleaved caspase-3 by immunoassay. Treatment with IL-17A increases the expression of cleaved caspase-3 in wild-type, but not NFKBIZKO, organoids in the absence of Noggin. b, Analysis of NOS2 transcript expression by quantitative PCR. IL-17A induces expression of NOS2 transcript in wild-type, but not in NFKBIZKO, TRAF3IP2KO and IL17RAKO, organoids in the absence of Noggin.

Source data

Extended Data Table 1 Clinical information on the participants in the study

Supplementary information

Supplementary Figure

Supplementary Figure 1: Source images of capillary based immunodetection.

Reporting Summary

Supplementary Table

Supplementary Table 1: Somatic mutations identified by whole exome sequencing

Supplementary Table

Supplementary Table 2: Somatic mutations identified by targeted sequencing.

Supplementary Table

Supplementary Table 3: List of IL-17 target genes, and sequences of qPCR primers, sgRNA targets and double strand DNAs.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nanki, K., Fujii, M., Shimokawa, M. et al. Somatic inflammatory gene mutations in human ulcerative colitis epithelium. Nature 577, 254–259 (2020). https://doi.org/10.1038/s41586-019-1844-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1844-5

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