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:

Human autoinflammatory disease reveals ELF4 as a transcriptional regulator of inflammation

Abstract

Transcription factors specialized to limit the destructive potential of inflammatory immune cells remain ill-defined. We discovered loss-of-function variants in the X-linked ETS transcription factor gene ELF4 in multiple unrelated male patients with early onset mucosal autoinflammation and inflammatory bowel disease (IBD) characteristics, including fevers and ulcers that responded to interleukin-1 (IL-1), tumor necrosis factor or IL-12p40 blockade. Using cells from patients and newly generated mouse models, we uncovered ELF4-mutant macrophages having hyperinflammatory responses to a range of innate stimuli. In mouse macrophages, Elf4 both sustained the expression of anti-inflammatory genes, such as Il1rn, and limited the upregulation of inflammation amplifiers, including S100A8, Lcn2, Trem1 and neutrophil chemoattractants. Blockade of Trem1 reversed inflammation and intestine pathology after in vivo lipopolysaccharide challenge in mice carrying patient-derived variants in Elf4. Thus, ELF4 restrains inflammation and protects against mucosal disease, a discovery with broad translational relevance for human inflammatory disorders such as IBD.

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: Loss-of-function variants in ELF4 identified in males with X-linked autoinflammatory disease.
Fig. 2: Inflammation in vivo and clinical response to anakinra.
Fig. 3: ELF4 deficiency augments TH17 cell responses in situ, in vitro and in vivo.
Fig. 4: Innate inflammatory responses to PRR stimulation are augmented in vitro and in vivo.
Fig. 5: Elf4 in macrophages regulates anti- and proinflammatory genes, including Trem1.

Similar content being viewed by others

Data availability

RNA-seq data were deposited in the GEO database (accession no. GSE175569). WES data will not be made publicly available because they contain information that could compromise research participant privacy/consent. Source data are provided with this paper. Information on the WES raw data supporting the findings of the present study is available from the corresponding author, C.L.L., upon request. Mice harboring the Trp250Ser or KO allele for Elf4 are available from the corresponding author, C.L.L., upon request.

References

  1. Bousfiha, A. et al. Human inborn errors of immunity: 2019 update of the IUIS phenotypical classification. J. Clin. Immunol. 40, 66–81 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Tangye, S. G. et al. Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J. Clin. Immunol. 40, 24–64 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Adzhubei, I. A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Rentzsch, P., Witten, D., Cooper, G. M., Shendure, J. & Kircher, M. CADD: predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 47, D886–D894 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Vaser, R., Adusumalli, S., Leng, S. N., Sikic, M. & Ng, P. C. SIFT missense predictions for genomes. Nat. Protoc. 11, 1–9 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Poon, G. M. K. & Kim, H. M. Signatures of DNA target selectivity by ETS transcription factors. Transcription 8, 193–203 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sharrocks, A. D. The ETS-domain transcription factor family. Nat. Rev. Mol. Cell Biol. 2, 827–837 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Miyazaki, Y., Sun, X., Uchida, H., Zhang, J. & Nimer, S. MEF, a novel transcription factor with an Elf-1 like DNA binding domain but distinct transcriptional activating properties. Oncogene 13, 1721–1729 (1996).

    CAS  PubMed  Google Scholar 

  10. Lacorazza, H. D. et al. The ETS protein MEF plays a critical role in perforin gene expression and the development of natural killer and NK-T cells. Immunity 17, 437–449 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Yamada, T., Park, C. S., Mamonkin, M. & Lacorazza, H. D. Transcription factor ELF4 controls the proliferation and homing of CD8+ T cells via the Krüppel-like factors KLF4 and KLF2. Nat. Immunol. 10, 618–626 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. You, F. et al. ELF4 is critical for induction of type I interferon and the host antiviral response. Nat. Immunol. 14, 1237–1246 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Curina, A. et al. High constitutive activity of a broad panel of housekeeping and tissue-specific cis-regulatory elements depends on a subset of ETS proteins. Genes Dev. 31, 399–412 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lee, P.-H. et al. The transcription factor E74-like factor 4 suppresses differentiation of proliferating CD4+ T cells to the Th17 lineage. J. Immunol. 192, 178–188 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Bouchon, A., Facchetti, F., Weigand, M. A. & Colonna, M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 410, 1103–1107 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Uhlig, H. H. et al. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology 147, 990–1007.e3 (2014).

    Article  PubMed  Google Scholar 

  17. Sobreira, N., Schiettecatte, F., Valle, D. & Hamosh, A. GeneMatcher: a matching tool for connecting investigators with an interest in the same gene. Hum. Mutat. 36, 928–930 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kircher, M. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat. Genet. 46, 310–315 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Flannigan, K. L. et al. IL-17A-mediated neutrophil recruitment limits expansion of segmented filamentous bacteria. Mucosal Immunol. 10, 673–684 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Revu, S. et al. IL-23 and IL-1β drive human Th17 cell differentiation and metabolic reprogramming in absence of CD28 costimulation. Cell Rep 22, 2642–2653 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Stark, M. A. et al. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 22, 285–294 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Weaver, C. T., Elson, C. O., Fouser, L. A. & Kolls, J. K. The Th17 pathway and inflammatory diseases of the intestines, lungs, and skin. Annu. Rev. Pathol. Mechanisms Dis. 8, 477–512 (2013).

    Article  CAS  Google Scholar 

  24. Okayasu, I. et al. A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology 98, 694–702 (1990).

    Article  CAS  PubMed  Google Scholar 

  25. Wirtz, S., Neufert, C., Weigmann, B. & Neurath, M. F. Chemically induced mouse models of intestinal inflammation. Nat. Protoc. 2, 541–546 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Esplugues, E. et al. Control of TH17 cells occurs in the small intestine. Nature 475, 514–518 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mandal, P. et al. Caspase-8 collaborates with caspase-11 to drive tissue damage and execution of endotoxic shock. Immunity 49, 42–55.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Manthiram, K., Zhou, Q., Aksentijevich, I. & Kastner, D. L. The monogenic autoinflammatory diseases define new pathways in human innate immunity and inflammation. Nat. Immunol. 18, 832–842 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Stewart, D. M., Tian, L., Notarangelo, L. D. & Nelson, D. L. Update on X-linked hypogammaglobulinemia with isolated growth hormone deficiency. Curr. Opin. Allergy Clin. Immunol. 5, 510–512 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Beura, L. K. et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 532, 512–516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Takeda, A. J. et al. Human PI3Kγ deficiency and its microbiota-dependent mouse model reveal immunodeficiency and tissue immunopathology. Nat. Commun. 10, 4364 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Lu, Z. et al. MEF up-regulates human β-defensin 2 expression in epithelial cells. FEBS Lett. 561, 117–121 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Cao, L. et al. HIPK2 is necessary for type I interferon-mediated antiviral immunity. Sci. Signal. https://doi.org/10.1126/scisignal.aau4604 (2019).

  34. Seifert, L. L. et al. The ETS transcription factor ELF1 regulates a broadly antiviral program distinct from the type I interferon response. PLoS Pathog. 15, e1007634 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Guo, B., Chang, E. Y. & Cheng, G. The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice. J. Clin. Invest. 118, 1680–1690 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Guarda, G. et al. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity 34, 213–223 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Reboldi, A. et al. 25-Hydroxycholesterol suppresses interleukin-1-driven inflammation downstream of type I interferon. Science 345, 679–684 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Aksentijevich, I. et al. An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist. N. Engl. J. Med. 360, 2426–2437 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Reddy, S. et al. An autoinflammatory disease due to homozygous deletion of the IL1RN locus. N. Engl. J. Med. 360, 2438–2444 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Schenk, M., Bouchon, A., Seibold, F. & Mueller, C. TREM-1-expressing intestinal macrophages crucially amplify chronic inflammation in experimental colitis and inflammatory bowel diseases. J. Clin. Invest. 117, 3097–3106 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Garvie, C. W., Hagman, J. & Wolberger, C. Structural studies of Ets-1/Pax5 complex formation on DNA. Mol. Cell 8, 1267–1276 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. 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 

  43. 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  Google Scholar 

  44. Crowley, E. et al. Prevalence and clinical features of inflammatory bowel diseases associated with monogenic variants, identified by whole-exome sequencing in 1000 children at a single center. Gastroenterology 158, 2208–2220 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Pan, J., Thoeni, C., Muise, A., Yeger, H. & Cutz, E. Multilabel immunofluorescence and antigen reprobing on formalin-fixed paraffin-embedded sections: novel applications for precision pathology diagnosis. Mod. Pathol. 29, 557–569 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families for participating in the research and all clinical care staff for their contributions. We also thank P. Schwartzberg, P.-P. Axisa and J.-M. Carpier for critical feedback. We thank Prometheus for providing recombinant IL-2 used in T cell culture experiments and the Yale Cancer Center for support. We thank Yale New Haven Hospital and S. Bluell and J. Buell for their support of the Pediatric Genomics Discovery Program. C.L.L. is funded by the Mathers Foundation, National Institute of Allergy and Infectious Diseases/National Institutes of Health (grant no. R01AI150913), Immune Deficiency Foundation, Hood Foundation and Yale University. A.M.M. is funded by a Canada Research Chair (Tier 1) in Pediatric IBD, Canadian Institute of Health Research Foundation Grant, National Institute of Diabetes and Digestive and Kidney Diseases (grant no. RC2DK118640) and the Leona M. and Harry B. Helmsley Charitable Trust.

Author information

Authors and Affiliations

Authors

Contributions

P.M.T., M.L.B. and M.Z. performed experiments, analyzed the data and wrote the manuscript. T.J.M. and A.J.R. performed experiments and analyzed the data. W.J. performed the analysis of the genomics data from family A. N.W. identified and evaluated the ELF4 variant in patient B.1. J.P. performed staining of biopsy samples from patient B.1 and analyzed the data. R.M. provided pathology expertise for staining of biopsy samples from patient A.1. P.M. provided clinical care and insights for patient A.1. A.G. provided clinical care and insights for patient B.1. A.M.C.v.R. provided clinical care and insights for patient C.1. I.H.I.M.H. performed genetic analysis of family C. V.A.S.H.D. recruited and provided clinical care and insights for patient C.1. J.C. recruited and provided clinical care and insights for patient A.1. S.A.L. oversaw genetic analysis of family A. A.M.M. provided clinical care and oversaw genomics analysis and histology staining of biopsies from family B. C.L.L. supervised overall research and data analysis, performed experiments and wrote/edited the manuscript. All authors discussed and reviewed the manuscript.

Corresponding author

Correspondence to Carrie L. Lucas.

Ethics declarations

Competing interests

S.A.L. is part owner of Qiyas Higher Health and Victory Genomics, startup companies unrelated to this work. All other authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Extended DEX patient clinical and cellular findings and generation of Elf4 KO and Trp250Ser mice.

a, NK cell, (b) NKT cell, (c) CD4+ and CD8+ T cell, (d) monocyte, (e) B cell, (f) CD4+ memory and naïve, and (g) CD8+ memory and naïve flow cytometric immunophenotyping for the indicated markers on PBMCs from a healthy donor (Ctrl) and patient A.1. h, NK cytotoxicity assay using PBMCs from patient A.1 (red) compared to the normal range (grey shading). i, Human IFNα ELISA in supernatants of LPS-stimulated PBMCs from healthy donors (n = 3) and patient A.1 (n = 1). j, Western blot on THP1 lysates for ELF4. k, Histogram of missense variants in the gnomAD dabase in ELF4 gene. l, Western blot on 293T cells overexpressing variants of ELF4 (myc-tagged) reported in gnomAD. m, Schematic of mouse Elf4. n, Western blot for Elf4 in mouse thymus. o, Sanger sequencing genotyping of Trp250Ser mice. p, Relative allele usage of B.2 (X/Trp251Ser) PBMCs. q, Relative allele usage (X/Trp250Ser) of mouse CD4+ or CD8+ cells. r, Percentage of perforin+ CD8+ T cells (WT n = 6, Trp250Ser n = 3, Elf4 KO n = 3) 4 days with after anti-CD3 and anti-CD28. s, Perforin gene expression in blasting CD8+ T cells isolated from healthy controls and patient A.1 determined by qRT-PCR (Ctrl n = 3, A.1 n = 1). t, Histogram displaying perforin expression in NT and ELF4 CRISPR-edited human CD8 + T cells after 10 days of IL-2. u, Western blot showing CRISPR deletion of ELF4 from human CD8 + T cells by CRISPR. v, Perforin expression determined by flow cytometry at 24-hour time point following overexpression of myc-tagged Trp251Ser and WT ELF4 mRNA in patient A.1, B.1 (pink), and C.1 (red) CD8 + T cells. Data are presented as mean + /- S.E.M. with two-tailed unpaired t-test (r) or paired t-test (v) *p < 0.05, **p < 0.01, ***p < 0.001, ****p < .0001, no marking indicates not significant.

Source data

Extended Data Fig. 2 Extended serum analyses in patient A.1.

Concentrations of the indicated cytokine or chemokine in serum from independent blood draws of unrelated healthy controls (n = 4–6), patient A.1 (n = 3), mom (blue circles, n = 3), and dad (green circles, n = 1–3). Data from three independent experiments is presented as mean ± SEM. Statistical analysis was performed using two-tailed unpaired t-test. **p < 0.01, no marking indicates not significant.

Source data

Extended Data Fig. 3 Extended data on T cell differentiation and gene expression.

a, ELISA for IL-17A from human CD4+ cells (n = 1). b, ELISA for IL-17A from mouse CD4+ T cells (n = 1). (n = 1). c, Mouse IL-17A ELISA following naïve CD4+ Th17 in vitro differentiation under non-pathogenic conditions (TGFβ + IL-6) WT n = 3, Trp250Ser n = 3, Elf4 KO n = 3. d, e, Percentage of mouse or human naïve CD4 T cells in spleen (WT n = 3, Trp250Ser n = 3, Elf4 KO n = 3) or PBMC (Ctrl n = 3, A.1 n = 2), respectively. f, Western blot of cytoplasmic (Cyto) and nuclear (Nuc) fractions of effector T cells. g, Flow cytometry after treatment with anti-CD3 and anti-CD28 for 72 hours. h, List of gene sets and pathways associated with the differentially expressed genes in Elf4 KO naïve CD4+ T cells. i, Volcano plots of differentially expressed genes in Elf4 KO (1) or Trp250Ser (2) versus WT mouse naïve CD4+ T cells or Trp250Ser versus WT mouse in vitro differentiation Th17 cells after 48 hours under non-pathogenic (3) or pathogenic (4) conditions. j, Top ten upregulated and downregulated genes in Elf4 KO or Trp250Ser CD4+ T cells. Values shown as log2(FC). k, Naive CD4+ T cells differentiated in vitro to Th17 cells. l, Heat map showing Z-score summary of naive CD4+ T cell ATAC-seq peak results filtered for genes with p-value < 0.01 and FC > 2. m, Venn diagram displaying overlap between ATAC-seq peaks in Elf4 KO and WT naive CD4+ T cells. n, Heatmap displaying genes involved in chromatin regulation that were differentially expressed by RNAseq (WT vs Elf4 KO) and also display differences in accessibility by ATACseq. o, Reanalysis of DICE database 45. ELISA data are from a minimum of three experiments, each dot representing one ELISA well with two wells/technical replicates per sample. A minimum of n = 3 mice (biological replicates) was used for each genotype in mouse experiments. DEX patient samples represent blood from the same patient at different times. Data are presented as mean ± S.E.M. with two-tailed unpaired t-test *p < 0.05, **p < 0.01, ***p < 0.001, ****p < .0001, no marking indicates not significant.

Source data

Extended Data Fig. 4 Extended data on monocyte/macrophage cellular responses.

a, Indicated cytokine measured in culture supernatants from LPS-stimulated human PBMCs. Data are combined from two independent experiments (patient A.1 vs 5 controls, and patient B.1 vs 5 controls) and expressed as fold change of patient values normalized to the average of the controls (n = 13 healthy controls, n = 2 A.1 independent experiments, n = 1 B.1 experiment). b, PBMCs from patient A.1 and a healthy donor control were treated with LPS alone or LPS and a titration of IL-10 for 12 hours, and IL-6 was measured in culture supernatants (n = 1 patient and n = 1 healthy donor control). c, RT-PCR analysis of ELF4 gene expression in monocyte-derived macrophages from healthy donors after CRISPR targeting (NT: non-targeting gRNA, ELF4: ELF4 gRNA). d, IL-6 and CXCL1 measured in culture supernatants from 24hrs MDP/PolyIC/β-glucan-stimulated BMDMs isolated from Elf4 KO, Trp250Ser, or WT mice. e, Endotoxic shock was induced in groups of male WT and age-matched Elf4 KO and Trp250Ser mice by i.p. injection of 2 mg/kg ultra-pure (UP) LPS. Animals were scored for 0 h, 2 h, 4 h, 6 h and 16 h after LPS injection. f, Concentrations of the indicated cytokine or chemokine in mouse serum 4 hr after i.p. LPS challenge. Analytes in red are significantly different between genotypes. g, Endotoxic shock was induced in groups of female WT (n = 3) and age-matched heterozygous females (Elf4 KO n = 3 and Trp250Ser n = 3) by i.p. injection of 2 mg/kg ultra-pure (UP) LPS. h, Concentrations of the indicated cytokine or chemokine in mouse serum 4 hr after i.p. LPS challenge described in (g). Data are representative of three independent experiments and presented as mean ± SD. Statistical analysis was performed using two-tailed unpaired t-test. *p < 0.05, **p < 0.01, ***/###p < 0.001, ****p < 0.0001, no marking indicates not significant.

Source data

Extended Data Fig. 5 Extended data on macrophage gene expression and responses to Trem1 blockade.

a-c, Volcano plots (-log10(FDR) vs fold change) of differentially expressed genes in Elf4 KO versus WT mouse BMDMs as indicated. d-f, Heatmaps highlighting top 10 differentially expressed genes at each timepoint above. g, h, RT-PCR for Il10 and Il1rn in WT, Elf4 KO, or Elf4 Trp250Ser BMDMs at 16 hours after stimulation with LPS. For (g), (h), (k), and (l) n = 3 wt and n = 3 Trp250Ser mutant mice per group. (i) IL1RN reporter data as in Fig. 5C but with three individual 5′-GGAA sites mutated to 5′-AAAA to assess the contribution of each to ELF4-driven transcriptional activation of IL1RN reporter, n = 1 experimental replicate, representative of three independent experiments, ±SD. j, ChIP-sequencing traces for Elf4 bound near the indicated gene in mouse BMDM without (-) and with (+) 4 hr LPS stimulation. k, l, RT-PCR for S100a8 and Trem1 in WT, Elf4 KO, or Elf4 Trp250Ser BMDMs at 4 hours after stimulation with LPS. m, Functionally enriched gene ontology and KEGG pathways of upregulated differentially expressed genes in Elf4 KO compared to WT BMDMs 16 hrs after LPS stimulation. n-p, IL-6, IL-12p70, and IL-23 measured in culture supernatants at 24 hours after stimulation of indicated BMDMs with LPS or LPS and Trem1-Fc (n = 5/group). q, Endotoxic shock clinical score 16 hours after treatment (n = 8,7 WT/WT + Trem1-Fc; n = 3,4 Elf4 KO/Elf4 KO + Trem1 Fc; n = 5,4 Trp250Ser/Trp250Ser + Trem1 Fc). r, CXCL1 was measured in mouse serum at 4 hr after in vivo LPS challenge with the treatments indicated in (q). Data are representative of three independent experiments and presented as mean ± SD. Statistical analysis was performed using two-tailed unpaired t-test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, no marking indicates not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Tables 1–4.

Reporting Summary

Supplementary Table 5

Oligonucleotide sequences for gRNAs and primers.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tyler, P.M., Bucklin, M.L., Zhao, M. et al. Human autoinflammatory disease reveals ELF4 as a transcriptional regulator of inflammation. Nat Immunol 22, 1118–1126 (2021). https://doi.org/10.1038/s41590-021-00984-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00984-4

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