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.

  • Letter
  • Published:

The IL-15–AKT–XBP1s signaling pathway contributes to effector functions and survival in human NK cells

Abstract

Interleukin 15 (IL-15) is one of the most important cytokines that regulate the biology of natural killer (NK) cells1. Here we identified a signaling pathway—involving the serine-threonine kinase AKT and the transcription factor XBP1s, which regulates unfolded protein response genes2,3—that was activated in response to IL-15 in human NK cells. IL-15 induced the phosphorylation of AKT, which led to the deubiquitination, increased stability and nuclear accumulation of XBP1s protein. XBP1s bound to and recruited the transcription factor T-BET to the gene encoding granzyme B, leading to increased transcription. XBP1s positively regulated the cytolytic activity of NK cells against leukemia cells and was also required for IL-15-mediated NK cell survival through an anti-apoptotic mechanism. Thus, the newly identified IL-15–AKT–XBP1s signaling pathway contributes to enhanced effector functions and survival of human NK cells.

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: XBP1s is induced by IL-15 at the protein level and controls degranulation in NK cells.
Fig. 2: XBP1s regulates GZMB and interferon-γ expression in NK cells.
Fig. 3: XBP1s interacts with T-BET and binds to the proximal GZMB promoter.
Fig. 4: XBP1s contributes to IL-15-mediated NK cell survival.
Fig. 5: AKT mediates stability of XBP1s.

Similar content being viewed by others

Data availability

All summary or representative data generated and supporting the findings of this study are available within the paper. Raw data that support the findings of this study are available upon reasonable request.

References

  1. Becknell, B. & Caligiuri, M. A. Interleukin-2, interleukin-15, and their roles in human natural killer cells. Adv. Immunol. 86, 209–239 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Vidal, R. L. & Hetz, C. Unspliced XBP1 controls autophagy through FoxO1. Cell Res. 23, 463–464 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Calfon, M. et al. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 415, 92–96 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Uemura, A., Oku, M., Mori, K. & Yoshida, H. Unconventional splicing of XBP1 mRNA occurs in the cytoplasm during the mammalian unfolded protein response. J. Cell Sci. 122, 2877–2886 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Clauss, I. M., Chu, M., Zhao, J. L. & Glimcher, L. H. The basic domain/leucine zipper protein hXBP-1 preferentially binds to and transactivates CRE-like sequences containing an ACGT core. Nucleic Acids Res. 24, 1855–1864 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bettigole, S. E. et al. The transcription factor XBP1 is selectively required for eosinophil differentiation. Nat. Immunol. 16, 829–837 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kamimura, D. & Bevan, M. J. Endoplasmic reticulum stress regulator XBP-1 contributes to effector CD8+ T cell differentiation during acute infection. J. Immunol. 181, 5433–5441 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Shaffer, A. L. et al. XBP1, downstream of Blimp-1, expands the secretory apparatus and other organelles, and increases protein synthesis in plasma cell differentiation. Immunity 21, 81–93 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Cubillos-Ruiz, J. R. et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sadighi Akha, A. A. et al. Heightened induction of proapoptotic signals in response to endoplasmic reticulum stress in primary fibroblasts from a mouse model of longevity. J. Biol. Chem. 286, 30344–30351 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cross, B. C. et al. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proc. Natl Acad. Sci. USA 109, E869–E878 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mai, B. & Breeden, L. Xbp1, a stress-induced transcriptional repressor of the Saccharomyces cerevisiae Swi4/Mbp1 family. Mol. Cell. Biol. 17, 6491–6501 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Glimcher, L. H., Townsend, M. J., Sullivan, B. M. & Lord, G. M. Recent developments in the transcriptional regulation of cytolytic effector cells. Nat. Rev. Immunol. 4, 900–911 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Townsend, M. J. et al. T-bet regulates the terminal maturation and homeostasis of NK and Vα14i NKT cells. Immunity 20, 477–494 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Chen, X. et al. XBP1 promotes triple-negative breast cancer by controlling the HIF1α pathway. Nature 508, 103–107 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Acosta-Alvear, D. et al. XBP1 controls diverse cell type- and condition-specific transcriptional regulatory networks. Mol. Cell 27, 53–66 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Verdeil, G., Puthier, D., Nguyen, C., Schmitt-Verhulst, A. M. & Auphan-Anezin, N. STAT5-mediated signals sustain a TCR-initiated gene expression program toward differentiation of CD8 T cell effectors. J. Immunol. 176, 4834–4842 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Gotthardt, D. et al. STAT5 is a key regulator in NK cells and acts as a molecular switch from tumor surveillance to tumor promotion. Cancer Discov. 6, 414–429 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Jahrsdorfer, B. et al. Granzyme B produced by human plasmacytoid dendritic cells suppresses T-cell expansion. Blood 115, 1156–1165 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Romero-Ramirez, L. et al. XBP1 is essential for survival under hypoxic conditions and is required for tumor growth. Cancer Res. 64, 5943–5947 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Gupta, S. et al. HSP72 protects cells from ER stress-induced apoptosis via enhancement of IRE1α-XBP1 signaling through a physical interaction. PLoS Biol. 8, e1000410 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Carson, W. E. et al. A potential role for interleukin-15 in the regulation of human natural killer cell survival. J. Clin. Invest. 99, 937–943 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yano, S., Komine, M., Fujimoto, M., Okochi, H. & Tamaki, K. Interleukin 15 induces the signals of epidermal proliferation through ERK and PI 3-kinase in a human epidermal keratinocyte cell line, HaCaT. Biochem. Biophys. Res. Commun. 301, 841–847 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Wang, Y. et al. PI3K inhibitor LY294002, as opposed to wortmannin, enhances AKT phosphorylation in gemcitabine-resistant pancreatic cancer cells. Int. J. Oncol. 50, 606–612 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Kohn, A. D., Takeuchi, F. & Roth, R. A. Akt, a pleckstrin homology domain containing kinase, is activated primarily by phosphorylation. J. Biol. Chem. 271, 21920–21926 (1996).

    Article  CAS  PubMed  Google Scholar 

  26. McKeehan, W. & Hardesty, B. The mechanism of cycloheximide inhibition of protein synthesis in rabbit reticulocytes. Biochem. Biophys. Res. Commun. 36, 625–630 (1969).

    Article  CAS  PubMed  Google Scholar 

  27. Rock, K. L. et al. Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules. Cell 78, 761–771 (1994).

    Article  CAS  PubMed  Google Scholar 

  28. Zambricki, E. et al. Signaling T-cell survival and death by IL-2 and IL-15. Am. J. Transplant. 5, 2623–2631 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Xu, D. et al. Phosphorylation and activation of ubiquitin-specific protease-14 by Akt regulates the ubiquitin-proteasome system. eLife 4, e10510 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Lee, M. S. et al. PI3K/AKT activation induces PTEN ubiquitination and destabilization accelerating tumourigenesis. Nat. Commun. 6, 7769 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Guo, H., Samarakoon, A., Vanhaesebroeck, B. & Malarkannan, S. The p110δ of PI3K plays a critical role in NK cell terminal maturation and cytokine/chemokine generation. J. Exp. Med. 205, 2419–2435 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tassi, I. et al. p110γ and p110δ phosphoinositide 3-kinase signaling pathways synergize to control development and functions of murine NK cells. Immunity 27, 214–227 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Ali, K. et al. Inactivation of PI(3)K p110δ breaks regulatory T-cell-mediated immune tolerance to cancer. Nature 510, 407–411 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nandagopal, N., Ali, A. K., Komal, A. K. & Lee, S. H. The critical role of IL-15–PI3K–mTOR pathway in natural killer cell effector functions. Front. Immunol. 5, 187 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Zebedin, E. et al. Leukemic challenge unmasks a requirement for PI3Kδ in NK cell-mediated tumor surveillance. Blood 112, 4655–4664 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Liu, Y. et al. Preventing oxidative stress: a new role for XBP1. Cell Death Differ. 16, 847–857 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Jiang, S., Munker, R. & Andreeff, M. Bcl-2 is expressed in human natural killer cells and is regulated by interleukin-2. Nat. Immunol. 15, 312–317 (1996).

    CAS  Google Scholar 

  38. Viant, C. et al. Cell cycle progression dictates the requirement for BCL2 in natural killer cell survival. J. Exp. Med. 214, 491–510 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Minagawa, M. et al. Enforced expression of Bcl-2 restores the number of NK cells, but does not rescue the impaired development of NKT cells or intraepithelial lymphocytes, in IL-2/IL-15 receptor β-chain-deficient mice. J. Immunol. 169, 4153–4160 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Chu, J. et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia 28, 917–927 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Meinke, S. & Watzl, C. NK cell cytotoxicity mediated by 2B4 and NTB-A is dependent on SAP acting downstream of receptor phosphorylation. Front. Immunol. 4, 3 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Chu, J. et al. Genetic modification of T cells redirected toward CS1 enhances eradication of myeloma cells. Clin. Cancer Res. 20, 3989–4000 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Deng, Y. et al. Transcription factor Foxo1 is a negative regulator of natural killer cell maturation and function. Immunity 42, 457–470 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wang, Y. et al. CGK733-induced LC3 II formation is positively associated with the expression of cyclin-dependent kinase inhibitor p21Waf1/Cip1 through modulation of the AMPK and PERK/CHOP signaling pathways. Oncotarget 6, 39692–39701 (2015).

    PubMed  PubMed Central  Google Scholar 

  45. Wang, Y. et al. PERK/CHOP contributes to the CGK733-induced vesicular calcium sequestration which is accompanied by non-apoptotic cell death. Oncotarget 6, 25252–25265 (2015).

    PubMed  PubMed Central  Google Scholar 

  46. Yu, J. et al. Pro- and antiinflammatory cytokine signaling: reciprocal antagonism regulates interferon-γ production by human natural killer cells. Immunity 24, 575–590 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Institutes of Health (AI129582 and NS106170 to J.Y. and CA185301, CA210087 and CA068458 to M.A.C.), the Leukemia and Lymphoma Society (6503-17 and 1364-19 to J.Y.), the American Cancer Society (RSG-14-243-01-LIB to J.Y.) and the Gabrielle’s Angel Cancer Research Foundation (#87 to J.Y.).

Author information

Authors and Affiliations

Authors

Contributions

Y.W. performed experiments, designed research and wrote the manuscript; Y.Z., P.Y., W.D., Z.Z. and L.C. performed experiments; A.P.N. revised and proofread the manuscript; J.Z. performed statistical analyses; D.M.B., B.L.M-B., A.G.F. and M.A.C. designed research, reviewed the manuscript and/or acquired funding; J.Y. conceptualized the idea, designed research, wrote the manuscript, acquired funding and supervised the study.

Corresponding author

Correspondence to Jianhua Yu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Quantification of IL-15-induced XBP1s and its correlation with NK cell cytotoxicity.

a, Densitometric quantification of the level of XBP1s protein normalized to the level of β-Actin protein, assessed by immunoblotting shown in Fig. 1b. NK cells were treated with IL-2 (100 units/ml) or IL-15 (100 units/ml) for 24 h. Immunoblotting was performed with an anti-XBP1s antibody. Bar graphs display mean ± s.d. of n = 8 donors. ***P < 0.001 by linear mixed model. b, NK cells were treated with IL-15 (100 units/ml) for 24 h and co-cultured with MM.1S multiple myeloma cells for 4 h. CD107a surface expression and intracellular level of XBP1s were determined by flow cytometry using anti-CD107a and anti-XBP1s antibodies. n = 4 donors. ***P < 0.001 by two-tailed paired t test. MFI, mean fluorescence intensity.

Supplementary Figure 2 XBP1s positively regulates the expression of GZMB in primary NK cells.

a,b, NK cells were pre-treated with 1 µM of thapsigargin (Thap) for 1 h and then treated with or without IL-15 (100 units ml−1) for 4 h followed by qPCR (a) or for 24 h followed by immunoblotting to detect XBP1s or GZMB expression (b). Bar graphs display mean ± s.e.m. of n = 3 donors. *P < 0.05, **P < 0.01, ***P < 0.001 by linear mixed model (a). c, Densitometric quantification of the level of XBP1s or GZMB protein normalized to the level of β-Actin protein in (b). Bar graphs display mean ± s.d. of n = 3 donors. **P < 0.01 by linear mixed model. d, NK cells were transfected with XBP1 or a scramble siRNA by electroporation and then were cultured for 48 h, followed by immunoblotting with XBP1s, GZMB, and β-Actin antibodies. e, Densitometric quantification of the level of XBP1s or GZMB protein normalized to the level of β-Actin protein in (d). Bar graphs display mean ± s.d. of n = 3 donors. **P < 0.01, ***P < 0.001 by Student’s two-tailed unpaired t test. f,g,h, NK cells were pretreated with 50 µM of 4µ8C for 1 h and then treated with or without IL-15 (100 units ml−1) for 4 h followed by qPCR (f) or for 24 h followed by immunoblotting to detect XBP1s or GZMB expression (g,h). h, Densitometric quantification of the level of XBP1s or GZMB protein normalized to the level of β-Actin protein in (g). Bar graphs display mean ± s.e.m. (f) or mean ± s.d. (h) of n = 3 donors. **P < 0.01, ***P < 0.001 by linear mixed model. The experiment was repeated independently for three donors with similar results (b,d,g). N.S., no significance. Blot images (b,d,g) were cropped, and the full scans are shown in the supplementary figures.

Supplementary Figure 3 Interaction and co-localization of XBP1s with T-BET.

a, Densitometric quantification of the level of T-BET protein normalized to input (i.e., enrichment) in Fig. 3c. 293T cells were co-transfected with T-BET and FLAG-XBP1u, FLAG-XBP1s, or the control (pCDH) lentiviral vector by lentiviral infection, followed by a 48 h culture. Co-IP was performed with an anti-FLAG antibody. Bar graphs display mean ± s.d. of n = 3 independent experiments. **P < 0.01, ***P < 0.001 by linear mixed model. b, NK-92 cells were transduced with FLAG-XBP1s or the control (pCDH) lentiviral vector by lentiviral infection, followed by a 24 h culture. Immunofluorescent staining was performed with anti-FLAG and anti-T-BET antibodies. The nuclei were stained by DAPI. Yellow indicates overlap of red and green. c, NK-92 (upper panel) or primary NK cells (lower panel) were treated with IL-15 (100 units/ml) for 24 h, followed by immunofluorescent staining with an anti-T-BET antibody that was different from the antibody clone used in (b) and Fig. 3d for data validation. An anti-α-tubulin antibody was included to stain the cytoplasm. The nuclei were stained by DAPI. The experiment was repeated independently for one (b) or three (c) times with similar results. d, Primary human NK cells were treated with or without 50 μM of 4μ8c in the presence of IL-15 (100 units/ml) for 16 h. ChIP was performed with a T-BET antibody or control IgG. Precipitated DNA was then analyzed by PCR. Bar graphs display mean ± s.d. of n = 4 donors. **P < 0.01 by Student’s two-tailed unpaired t test.

Supplementary Figure 4 XBP1s positively regulates GZMB gene promoter activity in a STAT5-independent manner.

a,b, 293T cells were co-transfected with the following four plasmids: 1. STAT5A shRNA, STAT5B shRNA, or scramble shRNA; 2. XBP1s or empty vector pCDH (EV); 3. A pGL3 plasmid containing the GZMB promoter; 4. A pRL-TK plasmid (as a control for data normalization). The transfected cells were cultured for 48 h and then harvested and lysed to determine the expression level of XBP1s or STAT5 protein by immunoblotting (a) or GZMB promoter activity by luciferase reporter assays (b). Bar graphs display mean ± s.d. of n = 4 independent experiments. ***P < 0.001 by Student’s two-tailed unpaired t test. The moderate knockdown effect for STAT5A is likely because the antibody used (#25656, Cell Signaling Technology, Inc.) recognizes both STAT5A and STAT5B. N.S., no significance. The experiment in (a) was repeated independently for three times with similar results. Blot images (a) were cropped, and the full scans are shown in the supplementary figures.

Supplementary Figure 5 AKT signaling regulates the expressions of XBP1s and GZMB.

a, Densitometric quantification of the level of XBP1s protein normalized to the level of β-actin protein in Fig. 5c. NK cells were pre-treated with or without 10 µM of AKTi-1/2, a potent isozyme selective Akt1/2 kinase inhibitor, followed by stimulation with IL-15 (100 units ml−1) for 6 h. Cells were then harvested to make lysates for immunoblot analysis. Bar graphs display mean ± s.d. of n = 3 donors. *P < 0.05 by linear mixed model. b,c, AKT1 or scramble shRNA lentiviral construct (pLKO.1) was transduced by lentiviral infection in primary human NK cells for 24 h, followed by fluorescence-activated cell sorting (FACS) for transduced cells based on GFP expression. GZMB expression was analyzed at the protein level by flow cytometry (b) or at the mRNA level by qPCR (c). Bar graphs display mean ± s.d. in (b) or mean ± s.e.m. in (c) of n = 3 donors. **P < 0.01, ***P < 0.001 by Student’s two-tailed paired t test. d,e, myrAKTΔ4-129 lentiviral construct or empty vector (EV) was transduced by lentiviral infection in human primary NK cells for 24 h, followed by FACS-based cell sorting for GFP(+) cells. GZMB expression of the sorted cells was analyzed as described in (b, c). Bar graphs display mean ± s.d. in (d) or mean ± s.e.m. in (e) of n = 3 donors. P = 0.07, **P < 0.01, ***P < 0.001 by Student’s two-tailed paired t test. N.S., no significance.

Supplementary Figure 6 AKT signaling regulates XBP1s at the protein level.

a, Densitometric quantification of the level of XBP1s protein normalized to the level of β-actin in Fig. 5d. NK cells were pre-treated with IL-15 (100 units/ml) alone or IL-15 plus 10 µM of AKTi-1/2 for 1 h; then cells were cultured with or without 10 µg/ml of cycloheximide (CHX) for 30 min without washing cells. Cells were then harvested to make lysates for immunoblot analysis. Bar graphs display mean ± s.d. of n = 3 donors. P = 0.08, **P < 0.01 by linear mixed model. b, Densitometric quantification of the level of XBP1s protein normalized to the level of β-actin protein in Fig. 5e. 293T cells were co-transfected with 0.04, 0.2, or 1 µg of pECE-myrAKTΔ4-129 vector or pECE empty vector (EV) and pCDH-FLAG-tagged XBP1s (FLAG-XBP1s) vector or pCDH control vector. The transfected cells were cultured for 24 h, followed by detecting XBP1s protein expression by immunoblotting with an anti-XBP1s antibody. Bar graphs display mean ± s.d. of n = 3 independent experiments. P = 0.06, *P < 0.05, **P < 0.01 by linear mixed model. c, A plasmid containing myrAKTΔ4-129 with constitutively active AKT or a control plasmid was co-transfected with an XBP1s plasmid, a pGL3 plasmid containing the GZMB promoter, and a pRL-TK plasmid (as a control for data normalization) into 293T cells. The transfected cells were cultured for 48 h. The promoter activity of the GZMB gene was detected by luciferase reporter assays and the expression of XBP1s and myrAKTΔ4-129 at the protein levels were detected by immunoblot. Bar graphs display mean ± s.d. of n = 3 independent experiments. ***P < 0.001 by two-way ANOVA model. The blot experiment was repeated independently for three times with similar results. Blot images (c) were cropped, and the full scans are shown in the Supplementary Figures. d, Densitometric quantification of the level of XBP1s protein normalized to the level of β-actin protein in Fig. 5g. 293T cells were co-transfected with a pECE-myrAKTΔ4-129 vector or pECE empty vector (EV) and pCDH-FLAG-tagged XBP1s (FLAG-XBP1s) vector or pCDH control vector, followed by treatment with 10 µg/ml of CHX for 0, 1, 2 and 4 h in the presence or absence of 10 µM of MG132, a protein degradation inhibitor. Bar graphs display mean ± s.d. of n = 3 independent experiments. **P < 0.01, ***P < 0.001 by linear mixed model. N.S., no significance. Blot images (c) were cropped, and the full scans are shown in Supplementary Figures.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Dataset

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Zhang, Y., Yi, P. et al. The IL-15–AKT–XBP1s signaling pathway contributes to effector functions and survival in human NK cells. Nat Immunol 20, 10–17 (2019). https://doi.org/10.1038/s41590-018-0265-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0265-1

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