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:

Kainate receptor modulation by NETO2

This article has been updated

Abstract

Glutamate-gated kainate receptors are ubiquitous in the central nervous system of vertebrates, mediate synaptic transmission at the postsynapse and modulate transmitter release at the presynapse1,2,3,4,5,6,7. In the brain, the trafficking, gating kinetics and pharmacology of kainate receptors are tightly regulated by neuropilin and tolloid-like (NETO) proteins8,9,10,11. Here we report cryo-electron microscopy structures of homotetrameric GluK2 in complex with NETO2 at inhibited and desensitized states, illustrating variable stoichiometry of GluK2–NETO2 complexes, with one or two NETO2 subunits associating with GluK2. We find that NETO2 accesses only two broad faces of kainate receptors, intermolecularly crosslinking the lower lobe of ATDA/C, the upper lobe of LBDB/D and the lower lobe of LBDA/C, illustrating how NETO2 regulates receptor-gating kinetics. The transmembrane helix of NETO2 is positioned proximal to the selectivity filter and competes with the amphiphilic H1 helix after M4 for interaction with an intracellular cap domain formed by the M1–M2 linkers of the receptor, revealing how rectification is regulated by NETO2.

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: Architectures of the GluK2–NETO2 complexes.
Fig. 2: Extracellular interactions between GluK2 and NETO2.
Fig. 3: Ion conduction pore of GluK2 and the modulation mechanism of inward rectification.
Fig. 4: Architecture of the desensitized GluK2–NETO2 complex.

Similar content being viewed by others

Data availability

The 3D cryo-EM density maps of the antagonist DNQX-bound GluK2-1×NETO2, LBD–TMD of the GluK2-1×NETO2, GluK2-2×NETO2, GluK2-1×NETO2asymLBD complex and the agonist kainate-bound desensitized GluK2-1×NETO2des complex have been deposited in the Electron Microscopy Database under the accession codes EMD-31462, EMD-31464, EMD-31463, EMD-31459 and EMD-31460, respectively. The cryo-EM map of LBD–TMD of the GluK2-2×NETO2 complexes have been deposited as an additional map under entry EMD-31463. The coordinates for the structures have been deposited in the PDB under accession codes 7F59, 7F5B, 7F5A, 7F56 and 7F57, respectively. Source data are provided with this paper.

Change history

  • 27 September 2021

    The linking to some of the Supplementary Information files was originally incorrect and has now been amended.

References

  1. Herb, A. et al. The KA-2 subunit of excitatory amino acid receptors shows widespread expression in brain and forms ion channels with distantly related subunits. Neuron 8, 775–785 (1992).

    Article  CAS  PubMed  Google Scholar 

  2. Bettler, B. et al. Cloning of a novel glutamate receptor subunit, GluR5: expression in the nervous system during development. Neuron 5, 583–595 (1990).

    Article  CAS  PubMed  Google Scholar 

  3. Traynelis, S. F. et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol. Rev. 62, 405–496 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Castillo, P. E., Malenka, R. C. & Nicoll, R. A. Kainate receptors mediate a slow postsynaptic current in hippocampal CA3 neurons. Nature 388, 182–186 (1997).

    Article  ADS  CAS  PubMed  Google Scholar 

  5. Vignes, M. & Collingridge, G. L. The synaptic activation of kainate receptors. Nature 388, 179–182 (1997).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. MacDermott, A. B., Role, L. W. & Siegelbaum, S. A. Presynaptic ionotropic receptors and the control of transmitter release. Annu. Rev. Neurosci. 22, 443–485 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Chittajallu, R. et al. Regulation of glutamate release by presynaptic kainate receptors in the hippocampus. Nature 379, 78–81 (1996).

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Zhang, W. et al. A transmembrane accessory subunit that modulates kainate-type glutamate receptors. Neuron 61, 385–396 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Copits, B. A., Robbins, J. S., Frausto, S. & Swanson, G. T. Synaptic targeting and functional modulation of GluK1 kainate receptors by the auxiliary neuropilin and tolloid-like (NETO) proteins. J. Neurosci. 31, 7334–7340 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tang, M. et al. Neto1 is an auxiliary subunit of native synaptic kainate receptors. J. Neurosci. 31, 10009–10018 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tomita, S. & Castillo, P. E. Neto1 and Neto2: auxiliary subunits that determine key properties of native kainate receptors. J. Physiol. 590, 2217–2223 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Straub, C., Zhang, W. & Howe, J. R. Neto2 modulation of kainate receptors with different subunit compositions. J. Neurosci. 31, 8078–8082 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fisher, J. L. & Mott, D. D. The auxiliary subunits Neto1 and Neto2 reduce voltage-dependent inhibition of recombinant kainate receptors. J. Neurosci. 32, 12928–12933 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Meyerson, J. R. et al. Structural mechanism of glutamate receptor activation and desensitization. Nature 514, 328–334 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mayer, M. L. Crystal structures of the GluR5 and GluR6 ligand binding cores: molecular mechanisms underlying kainate receptor selectivity. Neuron 45, 539–552 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Kumari, J., Vinnakota, R. & Kumar, J. Structural and functional insights into GluK3–kainate receptor desensitization and recovery. Sci. Rep. 9, 1–16 (2019).

    Article  Google Scholar 

  17. Khanra, N., Brown, P. M., Perozzo, A. M., Bowie, D. & Meyerson, J. R. Architecture and structural dynamics of the heteromeric GluK2/K5 kainate receptor. Elife 10, e66097 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Meyerson, J. R. et al. Structural basis of kainate subtype glutamate receptor desensitization. Nature 537, 567–571 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Honore, T. et al. Quinoxalinediones: potent competitive non-NMDA glutamate receptor antagonists. Science 241, 701–703 (1988).

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Zhao, Y., Chen, S., Yoshioka, C., Baconguis, I. & Gouaux, E. Architecture of fully occupied GluA2 AMPA receptor–TARP complex elucidated by cryo-EM. Nature 536, 108–111 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Copits, B. A. & Swanson, G. T. Dancing partners at the synapse: auxiliary subunits that shape kainate receptor function. Nat. Rev. Neurosci. 13, 675–686 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kumar, J., Schuck, P. & Mayer, M. L. Structure and assembly mechanism for heteromeric kainate receptors. Neuron 71, 319–331 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pinheiro, P. S. et al. Selective block of postsynaptic kainate receptors reveals their function at hippocampal mossy fiber synapses. Cereb. Cortex 23, 323–331 (2013).

    Article  PubMed  Google Scholar 

  24. Li, Y.-J. et al. Neto proteins regulate gating of the kainate-type glutamate receptor GluK2 through two binding sites. J. Biol. Chem. 294, 17889–17902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Köhler, M., Burnashev, N., Sakmann, B. & Seeburg, P. H. Determinants of Ca2+ permeability in both TM1 and TM2 of high affinity kainate receptor channels: diversity by RNA editing. Neuron 10, 491–500 (1993).

    Article  PubMed  Google Scholar 

  26. Chen, S. et al. Activation and desensitization mechanism of AMPA receptor–TARP complex by cryo-EM. Cell 170, 1234–1246.e14 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, D., Watson, J. F., Matthews, P. M., Cais, O. & Greger, I. H. Gating and modulation of a hetero-octameric AMPA glutamate receptor. Nature 594, 454–458 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bowie, D. & Mayer, M. L. Inward rectification of both AMPA and kainate subtype glutamate receptors generated by polyamine-mediated ion channel block. Neuron 15, 453–462 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Kamboj, S. K., Swanson, G. T. & Cull-Candy, S. G. Intracellular spermine confers rectification on rat calcium‐permeable AMPA and kainate receptors. J. Physiol. 486, 297–303 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nayeem, N., Mayans, O. & Green, T. Correlating efficacy and desensitization with GluK2 ligand-binding domain movements. Open Biol. 3, 130051 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Goehring, A. et al. Screening and large-scale expression of membrane proteins in mammalian cells for structural studies. Nat. Protoc. 9, 2574–2585 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang, K. Gctf: real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bepler, T., Kelley, K., Noble, A. J. & Berger, B. Topaz-Denoise: general deep denoising models for cryoEM and cryoET. Nat. Commun. 11, 5208 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. Elife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Scheres, S. H. & Chen, S. Prevention of overfitting in cryo-EM structure determination. Nat. Methods 9, 853–854 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bordoli, L. et al. Protein structure homology modeling using SWISS-MODEL workspace. Nat. Protoc. 4, 1–13 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Briggs, D. C. et al. Metal ion-dependent heavy chain transfer activity of TSG-6 mediates assembly of the cumulus-oocyte matrix. J. Biol. Chem. 290, 28708–28723 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  PubMed  CAS  Google Scholar 

  41. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D Struct. Biol. 74, 531–544 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Appleton, B. A. et al. Structural studies of neuropilin/antibody complexes provide insights into semaphorin and VEGF binding. EMBO J. 26, 4902–4912 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. DeLano, W. L. Pymol: an open-source molecular graphics tool. CCP4 Newsletter on Protein Crystallography http://148.79.162.84/newsletters/newsletter40/11_pymol.html (2002).

Download references

Acknowledgements

We thank X. Huang, B. Zhu, X. Li, L. Chen and other staff members at the Center for Biological Imaging (CBI), Core Facilities for Protein Science at the Institute of Biophysics, Chinese Academy of Science (IBP, CAS) for the support in cryo-EM data collection; N. Sheng for providing the cDNAs of GluK2 and NETO2; and Y. Wu for his research assistant service. This work is funded by the Chinese Academy of Sciences Strategic Priority Research Program (grant XDB37030304 to Y.Z. and grant XDB37030301 to X.C.Z.), the National Key R & D Program of China (2019YFA0801603 to Y.S.S.), the National Natural Science Foundation of China (91849112 to Y.S.S. and 31971134 to X.C.Z.), the Natural Science Foundation of Jiangsu Province (BE2019707 to Y.S.S.) and the Fundamental Research Funds for the Central Universities (0903-14380029 to Y.S.S.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. conceived the project and supervised the research. L.H., J.S., W.A. and B.Y. carried out molecular cloning and the cell biology experiments. L.H. expressed and purified the protein complex sample. L.H., Y.D. and Y.W. prepared the sample for the cryo-EM study. Y. Gao. L.H., Y.W. and Y.D. carried out cryo-EM data collection. Y. Gao and Y.Z. processed the cryo-EM data and prepared the figures. B.L. and H.L. built and refined the atomic model. Y.Z. and X.C.Z. analysed the structure. Y.Z. and Y.S.S. designed the electrophysiological study. J.S. and Y. Ge performed the electrophysiological analysis. Y.Z. prepared the initial draft of the manuscript. X.C.Z., Y.S.S. and Y.Z. edited the manuscript with input from all authors in the final version.

Corresponding authors

Correspondence to Xuejun Cai Zhang, Yun Stone Shi or Yan Zhao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Ingo Greger and Geoffrey Swanson for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Functional study and purification of GluK2-Neto2 complex.

a, b, Outside-out recording of the WT GluK2 and GluK2F107L, in the absence or presence of Neto2. c, Statistical analysis of the desensitization time constant of the WT GluK2 and GluK2F107L, with or without Neto2 (GluK2, n = 12, GluK2 + Neto2, n = 14; GluK2F107L, n = 10; GluK2F107L + Neto2, n = 10). Each symbol represents a single cell recording, and n value represents biologically independent cells for statistical analysis. Significances were determined using two-sided unpaired t-test. ****, P < 0.0001. Similar results were reproduced from two independent experiments. Error bars stand for S.E.M. d, Fluorescence-detection size-exclusion chromatography (FSEC) analysis of the co-expressed GluK2-mCherry (red) and Neto2-GFP (green). The experiments were repeated independently with more than three times with similar results. e, Size-exclusion chromatography (SEC) profile of the purified GluK2-Neto2 complex. Fractions within the dashed lines were pooled for cryo-EM sample preparation. The experiments were repeated independently with more than three times with similar results. (f) Coomassie blue-stained SDS-PAGE gel of the pooled fractions. The gel was repeated three times from different batches of purification with similar results. The uncropped gel can be found in Supplementary Fig. 1.

Source data

Extended Data Fig. 2 Cryo-EM data analysis of GluK2-1×Neto2, GluK2-2×Neto2, and GluK2-1×Neto2asymLBD complex.

a, Flowchart of cryo-EM data processing. A total of 5,448 movie stacks were collected and motion-corrected, followed by CTF estimation and particle picking. A representative motion-corrected micrograph of this dataset is shown here (Scale bar = 40 nm). The experiments were repeated three times with similar results. Particles were cleaned and classified using several rounds of 2D and 3D classifications, which generated 3 classes, representing GluK2-1×Neto2, GluK2-2×Neto2, and GluK2-1×Neto2asymLBD, respectively. Particles were then submitted to further 3D classifications separately to improve resolutions. Focused classification and refinement of LBD-TMD were conducted on the particles of GluK2-1×Neto2 complex. Masks used in focused processing were overlaid on GluK2 map (green) as transparent grey surfaces alongside the arrows. b, e, h, k, Angular distribution of the particles contributing the final reconstruction for GluK2-1×Neto2asymLBD complex (b), GluK2-2×Neto2 complex (e), GluK2-1×Neto2 complex (h), and LBD-TMD (k). The length of each spike indicates of the number of particles in the designated orientation. c, f, i, l, Sharpened map of GluK2-1×Neto2asymLBD complex (c), GluK2-2×Neto2 complex (f), GluK2-1×Neto2 complex (i), and LBD-TMD (l), colored according to local resolution estimation. d, g, j, m, The half-map (red) and model-map (black) Fourier shell correlation (FSC) of GluK2-1×Neto2asymLBD complex (d), GluK2-2×Neto2 complex (g), GluK2-1×Neto2 complex (j), and LBD-TMD (m).

Source data

Extended Data Fig. 3 EM maps for transmembrane helices and the LBD-TMD.

a, Transmembrane helices M1−M4, and the M1-M2 loop. EM maps are shown as transparent grey surfaces. Some sidechains are shown as sticks. b, EM maps for LBD and TMD layers. CUB2 and LDLa of Neto2 are colored in orange. Receptor is colored in purple. N-glycans and a lipid tail are shown in sticks.

Extended Data Fig. 4 Structural comparison of ATD and LBD layers.

a, Superimposition of antagonist bound LBDs of 5KUH (grey) and GluK2-1×Neto2 (red). b, Comparison of the ATDA-LBDB and ATDC-LBDD segments between subunit A (grey) and C (blue) of the GluK2-1×Neto2, GluK2-2×Neto2 and GluK2-1×Neto2asymLBD complexes, using LBD as a reference. The COMs of the ATD R1/R2-lobe of subunits A and C are depicted as rectangles or triangles, respectively. The COMs of the LBD layer is marked as a circle. c, Superimposition of ATD-CUB1 interactions between GluK2-1×Neto2asymLBD (grey) and GluK2-1×Neto2 (red, orange and blue).

Extended Data Fig. 5 Sequence alignments and structural comparison of the KARs.

ad, Sequence alignments of the GluK members in Rat norvegicus, numbered according to full-length subunits. Secondary structures of GluK2 are marked above the sequence alignment. Dashes represent gaps. Conserved residues are shaded in grey. Residues which are involved in Neto2 interaction are indicated by triangle symbol. e, Structural comparison of the LBD of GluK2 with GluK1 (3FUZ, green), GluK3 (3U92, cyan), GluK4 (5IKB, magenta), and GluK5 (7KS0, yellow), respectively. D1- and D2- lobes and Loop 1 are indicated.

Extended Data Fig. 6 Representative desensitization and rectification traces.

a, Representative desensitization traces of GluK2 and mutants responded to 60 ms application of 10 mM glutamate were normalized and aligned to the peak. Superimposed responses of the receptor alone and the receptor-Neto2 complex were shown in black and green traces, respectively. b, Normalized current-voltage relationship of GluK2 mutants in the absence and presence of Neto2. n value represents independent cells for analysis. c, Representative desensitization traces and related statistical analysis (GluA2, n = 16; GluA2K2ICD, n = 12). Each symbol represents a single cell recording, and n value represents biologically independent cells for statistical analysis. Significances were determined using two-sided unpaired t-test. Not significant (ns), P = 0.0755. No adjustments were made for multiple comparisons. Error bars stand for S.E.M. The H1-helix is composed of residues 857FCSAMVEELRMSLK870 and removed in GluK2ΔH1 construct. The amino acid sequence of the ICD of GluK2 and GluA2 between M1-M2 helices are 587YEWYNPHPCNPDSDVVEN604 and 570YEWHTEEFEDGRETQSSESTNE591, respectively, which are involved in the ICD swapping constructs of GluA2K2ICD and GluK2A2ICD. d, I-V relationship for GluA2, GluK2 and related mutants. Desensitization curves (10 mM glutamate for 200 ms) were recorded at holding potential ranging from −100 to +100 mV in 20 mV increasement. Traces were normalized to the peak value at −100 mV.

Source data

Extended Data Fig. 7 Interactions stabilizing the pore helix M2.

a, EM density map of the LBD and TMD layers of the GluK2-1×Neto2 complex. Subunits A/C and B/D of GluK2 are colored in blue and red, respectively. The Neto2 protein is colored in orange. N-glycans are colored in yellow. b, The ion conduction pore and its profile of the GluK2-Neto2 complex. Pore loops are colored in green. A cation ion is shown as a grey sphere, overlaid with corresponding EM density colored in marine. Q621, T652, A656, and T660 are shown in sticks. Constriction sites are indicated in the pore profile. c, The TM helices of the GluK2 (red and blue) and the Neto2 (yellow) are shown as cartoon. The EM density of M2 helix are shown as transparent grey surface. Critical residues involved in interactions are shown as sticks. d, “Top-down” view of M2 helices and the pore loops. Q621 residues are shown in sticks.

Extended Data Fig. 8 Cryo-EM data analysis of GluK2-1×Neto2des complex.

a, Flowchart of cryo-EM data processing. A total of 2,957 movie stacks were collected and motion-corrected, followed by CTF estimation and particle picking. A representative motion-corrected micrograph of this dataset is shown here (Scale bar = 40 nm). The experiments were repeated three times with similar results. Three-dimensional classification generated 8 classes, 4 of which displayed classical structures of kainate receptors. Another round of 3D classification was then performed, followed by Ab-initio Reconstruction and Heterologous Refinement to furtherly improve the quality of map. b, Angular distribution of the particles contributing the final reconstruction of GluK2-1×Neto2des complex. The length of each spike indicates of the number of particles in the designated orientation. c, Sharpened map of GluK2-1×Neto2des complex, colored according to local resolution estimation. d, The half-map (red) and model-map (black) Fourier shell correlation.

Source data

Extended Data Fig. 9 Conformational change of GluK2-Neto2 complex upon desensitization.

a, Superimposition of agonist bound LBDs of 4BDM (grey) and GluK2-1×Neto2des (red). b, Superimposition of ATD-CUB1-LBD interactions between GluK2-1×Neto2asymLBD (grey) and GluK2-1×Neto2des (red, orange and blue). c, Organization of the D1 lobe of the GluK2-1×Neto2des (red and blue) and desensitized GluK2 alone (grey). COMs of the lobes are depicted as black dots. Distances and angles are indicated. d, The LBD rearrangement between GluK2-1×Neto2 and GluK2-1×Neto2des upon desensitization. e, Displacement of LBD at B-position of GluK2-1×Neto2des complex (red, blue and orange) compared with desensitized GluK2 (5KUF, grey).

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, L., Sun, J., Gao, Y. et al. Kainate receptor modulation by NETO2. Nature 599, 325–329 (2021). https://doi.org/10.1038/s41586-021-03936-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03936-y

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