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:

Oxytocin mediates early experience–dependent cross-modal plasticity in the sensory cortices

Abstract

Sensory experience is critical to development and plasticity of neural circuits. Here we report a new form of plasticity in neonatal mice, where early sensory experience cross-modally regulates development of all sensory cortices via oxytocin signaling. Unimodal sensory deprivation from birth through whisker deprivation or dark rearing reduced excitatory synaptic transmission in the correspondent sensory cortex and cross-modally in other sensory cortices. Sensory experience regulated synthesis and secretion of the neuropeptide oxytocin as well as its level in the cortex. Both in vivo oxytocin injection and increased sensory experience elevated excitatory synaptic transmission in multiple sensory cortices and significantly rescued the effects of sensory deprivation. Together, these results identify a new function for oxytocin in promoting cross-modal, experience-dependent cortical development. This link between sensory experience and oxytocin is particularly relevant to autism, where hypersensitivity or hyposensitivity to sensory inputs is prevalent and oxytocin is a hotly debated potential therapy.

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

Access options

Buy this article

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

Figure 1: Whisker deprivation from birth significantly reduced excitatory synaptic transmission in layer II/III pyramidal neurons of the S1 barrel field and cross-modally in other sensory cortices.
Figure 2: Whisker deprivation from birth significantly reduced visually evoked responses for neurons in V1.
Figure 3: Dark rearing from birth significantly reduced excitatory synaptic transmission in layer II/III pyramidal neurons of the primary visual cortex and cross-modally in other sensory cortices.
Figure 4: Sensory deprivation from birth reduced oxytocin level at P14.
Figure 5: Sensory deprivation from birth reduced oxytocin release.
Figure 6: Oxytocin application significantly increased excitatory synaptic transmission in S1 layer II/III pyramidal neurons via OXTR-mediated signaling.
Figure 7: In vivo oxytocin injection rescued the effect of sensory deprivation, whereas its antagonist reduced excitatory synaptic transmission.
Figure 8: Environmental enrichment rescued the effect of sensory deprivation.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

NCBI Reference Sequence

References

  1. Katz, L.C. & Shatz, C.J. Synaptic activity and the construction of cortical circuits. Science 274, 1133–1138 (1996).

    Article  CAS  Google Scholar 

  2. Crair, M.C. Neuronal activity during development: permissive or instructive? Curr. Opin. Neurobiol. 9, 88–93 (1999).

    Article  CAS  Google Scholar 

  3. Sur, M. & Rubenstein, J.L. Patterning and plasticity of the cerebral cortex. Science 310, 805–810 (2005).

    Article  CAS  Google Scholar 

  4. Wiesel, T.N. Postnatal development of the visual cortex and the influence of environment. Nature 299, 583–591 (1982).

    Article  CAS  Google Scholar 

  5. Fox, K. Anatomical pathways and molecular mechanisms for plasticity in the barrel cortex. Neuroscience 111, 799–814 (2002).

    Article  CAS  Google Scholar 

  6. Feldman, D.E. & Brecht, M. Map plasticity in somatosensory cortex. Science 310, 810–815 (2005).

    Article  CAS  Google Scholar 

  7. Fox, K. & Wong, R.O. A comparison of experience-dependent plasticity in the visual and somatosensory systems. Neuron 48, 465–477 (2005).

    Article  CAS  Google Scholar 

  8. Espinosa, J.S. & Stryker, M.P. Development and plasticity of the primary visual cortex. Neuron 75, 230–249 (2012).

    Article  CAS  Google Scholar 

  9. Nithianantharajah, J. & Hannan, A.J. Enriched environments, experience-dependent plasticity and disorders of the nervous system. Nat. Rev. Neurosci. 7, 697–709 (2006).

    Article  CAS  Google Scholar 

  10. Sale, A., Berardi, N. & Maffei, L. Enrich the environment to empower the brain. Trends Neurosci. 32, 233–239 (2009).

    Article  CAS  Google Scholar 

  11. van Praag, H., Kempermann, G. & Gage, F.H. Neural consequences of environmental enrichment. Nat. Rev. Neurosci. 1, 191–198 (2000).

    Article  CAS  Google Scholar 

  12. Feldman, D.E. Synaptic mechanisms for plasticity in neocortex. Annu. Rev. Neurosci. 32, 33–55 (2009).

    Article  CAS  Google Scholar 

  13. Bavelier, D. & Neville, H.J. Cross-modal plasticity: where and how? Nat. Rev. Neurosci. 3, 443–452 (2002).

    Article  CAS  Google Scholar 

  14. Bavelier, D., Dye, M.W. & Hauser, P.C. Do deaf individuals see better? Trends Cogn. Sci. 10, 512–518 (2006).

    Article  Google Scholar 

  15. Merabet, L.B. & Pascual-Leone, A. Neural reorganization following sensory loss: the opportunity of change. Nat. Rev. Neurosci. 11, 44–52 (2010).

    Article  CAS  Google Scholar 

  16. Frasnelli, J., Collignon, O., Voss, P. & Lepore, F. Crossmodal plasticity in sensory loss. Prog. Brain Res. 191, 233–249 (2011).

    Article  Google Scholar 

  17. Goel, A. et al. Cross-modal regulation of synaptic AMPA receptors in primary sensory cortices by visual experience. Nat. Neurosci. 9, 1001–1003 (2006).

    Article  CAS  Google Scholar 

  18. Jitsuki, S. et al. Serotonin mediates cross-modal reorganization of cortical circuits. Neuron 69, 780–792 (2011).

    Article  CAS  Google Scholar 

  19. He, K., Petrus, E., Gammon, N. & Lee, H.K. Distinct sensory requirements for unimodal and cross-modal homeostatic synaptic plasticity. J. Neurosci. 32, 8469–8474 (2012).

    Article  CAS  Google Scholar 

  20. Marco, E.J., Hinkley, L.B., Hill, S.S. & Nagarajan, S.S. Sensory processing in autism: a review of neurophysiologic findings. Pediatr. Res. 69, 48R–54R (2011).

    Article  Google Scholar 

  21. Suarez, M.A. Sensory processing in children with autism spectrum disorders and impact on functioning. Pediatr. Clin. North Am. 59, 203–214 (2012).

    Article  Google Scholar 

  22. Micheva, K.D. & Beaulieu, C. Quantitative aspects of synaptogenesis in the rat barrel field cortex with special reference to GABA circuitry. J. Comp. Neurol. 373, 340–354 (1996).

    Article  CAS  Google Scholar 

  23. Insel, T.R. The challenge of translation in social neuroscience: a review of oxytocin, vasopressin, and affiliative behavior. Neuron 65, 768–779 (2010).

    Article  CAS  Google Scholar 

  24. Lee, H.J., Macbeth, A.H., Pagani, J.H. & Young, W.S. III. Oxytocin: the great facilitator of life. Prog. Neurobiol. 88, 127–151 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Stoop, R. Neuromodulation by oxytocin and vasopressin. Neuron 76, 142–159 (2012).

    Article  CAS  Google Scholar 

  26. Green, J.J. & Hollander, E. Autism and oxytocin: new developments in translational approaches to therapeutics. Neurotherapeutics 7, 250–257 (2010).

    Article  CAS  Google Scholar 

  27. Miller, G. Neuroscience. The promise and perils of oxytocin. Science 339, 267–269 (2013).

    Article  CAS  Google Scholar 

  28. Yamasue, H. et al. Integrative approaches utilizing oxytocin to enhance prosocial behavior: from animal and human social behavior to autistic social dysfunction. J. Neurosci. 32, 14109–14117 (2012).

    Article  CAS  Google Scholar 

  29. Knobloch, H.S. et al. Evoked axonal oxytocin release in the central amygdala attenuates fear response. Neuron 73, 553–566 (2012).

    Article  CAS  Google Scholar 

  30. Landgraf, R. & Neumann, I.D. Vasopressin and oxytocin release within the brain: a dynamic concept of multiple and variable modes of neuropeptide communication. Front. Neuroendocrinol. 25, 150–176 (2004).

    Article  CAS  Google Scholar 

  31. Ludwig, M. & Leng, G. Dendritic peptide release and peptide-dependent behaviours. Nat. Rev. Neurosci. 7, 126–136 (2006).

    Article  CAS  Google Scholar 

  32. Leng, G. & Ludwig, M. Neurotransmitters and peptides: whispered secrets and public announcements. J. Physiol. 586, 5625–5632 (2008).

    Article  CAS  Google Scholar 

  33. Veening, J.G., de Jong, T. & Barendregt, H.P. Oxytocin-messages via the cerebrospinal fluid: behavioral effects; a review. Physiol. Behav. 101, 193–210 (2010).

    Article  CAS  Google Scholar 

  34. McEwen, B.B. Brain-fluid barriers: relevance for theoretical controversies regarding vasopressin and oxytocin memory research. Adv. Pharmacol. 50, 531–592, 655–708 (2004).

    Article  Google Scholar 

  35. Caldwell, H.K., Stephens, S.L. & Young, W.S. III. Oxytocin as a natural antipsychotic: a study using oxytocin knockout mice. Mol. Psychiatry 14, 190–196 (2009).

    Article  CAS  Google Scholar 

  36. Gimpl, G. & Fahrenholz, F. The oxytocin receptor system: structure, function, and regulation. Physiol. Rev. 81, 629–683 (2001).

    Article  CAS  Google Scholar 

  37. Tribollet, E., Dubois-Dauphin, M., Dreifuss, J.J., Barberis, C. & Jard, S. Oxytocin receptors in the central nervous system. Distribution, development, and species differences. Ann. NY Acad. Sci. 652, 29–38 (1992).

    Article  CAS  Google Scholar 

  38. Hammock, E. & Levitt, P. Oxytocin receptor ligand binding in embryonic tissue and postnatal brain development of the C57BL/6J mouse. Front. Behav. Neurosci. 7, 195 (2013).

    Article  Google Scholar 

  39. He, S., Ma, J., Liu, N. & Yu, X. Early enriched environment promotes neonatal GABAergic neurotransmission and accelerates synapse maturation. J. Neurosci. 30, 7910–7916 (2010).

    Article  CAS  Google Scholar 

  40. Huttenlocher, P.R. Neural Plasticity: The Effects of Environment on the Development of the Cerebral Cortex (Harvard University Press, 2002).

  41. Krug, K., Akerman, C.J. & Thompson, I.D. Responses of neurons in neonatal cortex and thalamus to patterned visual stimulation through the naturally closed lids. J. Neurophysiol. 85, 1436–1443 (2001).

    Article  CAS  Google Scholar 

  42. Hensch, T.K. Critical period regulation. Annu. Rev. Neurosci. 27, 549–579 (2004).

    Article  CAS  Google Scholar 

  43. Chevaleyre, V., Dayanithi, G., Moos, F.C. & Desarmenien, M.G. Developmental regulation of a local positive autocontrol of supraoptic neurons. J. Neurosci. 20, 5813–5819 (2000).

    Article  CAS  Google Scholar 

  44. Ludwig, M. et al. Intracellular calcium stores regulate activity-dependent neuropeptide release from dendrites. Nature 418, 85–89 (2002).

    Article  CAS  Google Scholar 

  45. Paxinos, G. & Franklin, K.B.J. The Mouse Brain in Stereotaxic Coordinates (Academic Press, San Diego, 2001).

  46. Jones, J.P. & Palmer, L.A. The two-dimensional spatial structure of simple receptive fields in cat striate cortex. J. Neurophysiol. 58, 1187–1211 (1987).

    Article  CAS  Google Scholar 

  47. Malone, B.J., Kumar, V.R. & Ringach, D.L. Dynamics of receptive field size in primary visual cortex. J. Neurophysiol. 97, 407–414 (2007).

    Article  Google Scholar 

  48. Yeh, C.I., Xing, D. & Shapley, R.M. “Black” responses dominate macaque primary visual cortex v1. J. Neurosci. 29, 11753–11760 (2009).

    Article  CAS  Google Scholar 

  49. Zhu, Y. & Yao, H. Modification of visual cortical receptive field induced by natural stimuli. Cereb. Cortex 23, 1923–1932 (2013).

    Article  Google Scholar 

  50. Liu, L. & Duff, K. A technique for serial collection of cerebrospinal fluid from the cisterna magna in mouse. J. Vis. Exp. 21, 960 (2008).

    Google Scholar 

  51. Hrabetova, S. & Nicholson, C. Biophysical Properties of Brain Extracellular Space Explored with Ion-Selective Microelectrodes, Integrative Optical Imaging and Related Techniques (CRC Press, 2007).

  52. Durand, S. et al. NMDA receptor regulation prevents regression of visual cortical function in the absence of Mecp2. Neuron 76, 1078–1090 (2012).

    Article  CAS  Google Scholar 

  53. Tabuchi, K. et al. A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science 318, 71–76 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Young (US National Institute of Mental Health) for the oxytocin knockout mice, V. Grinevich (Max Planck Institute, Heidelberg, Germany) for the AAV-OXT-Venus construct, Y. Lu, X. Zeng and S. He for technical assistance, and colleagues at ION and members of the Yu laboratory for suggestions and comments. This work was supported by grants from the Ministry of Science and Technology (2011CBA00400) to X.Y. and H.Y., the National Natural Science Foundation of China (31125015 and 31321091) to X.Y., the China Postdoctoral Science Foundation (2013M540393) and Postdoctor Research Program of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (2013KIP306) to S.-J.L.

Author information

Authors and Affiliations

Authors

Contributions

J.-J.Z., S.-J.L. and X.Y. designed the study and wrote the paper. J.-J.Z. performed and analyzed all in vitro electrophysiology experiments; S.-J.L. performed and analyzed biochemistry and immunohistochemistry experiments, with the help of W.-Y. M. and X.-D.Z.; X.-D.Z. performed stereotaxic injections with the help of J.-J.Z.; D.Z. performed in vivo electrophysiology experiments; D.Z. and H.Y. analyzed in vivo electrophysiology experiments. All authors edited the paper.

Corresponding author

Correspondence to Xiang Yu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 The effect of whisker deprivation on inhibitory synaptic transmission and early excitatory synaptic transmission in the sensory cortices.

(a,d) Representative mIPSC recordings (left) and average waveforms (right) for conditions as indicated from S1 (a) and V1 (d). (b,c) Whisker-deprivation (WD) did not significantly affect mIPSC frequencies [b, bar graphs: Ctrl, 1.04 ± 0.14 Hz, WD, 0.83 ± 0.09 Hz, P = 0.22, t(29) = 1.24; cumulative distributions, P = 0.29] or amplitudes [c, bar graphs: Ctrl, 26.33 ± 1.91 pA, WD, 28.37 ± 2.62 pA, P = 0.52, t(29) = 0.65; cumulative distributions: P = 0.58] in S1. (e,f) Whisker-deprivation did not significantly affect mIPSC frequencies [e, bar graphs: Ctrl, 1.47 ± 0.16 Hz, WD, 1.32 ± 0.13 Hz, P = 0.49, t(23) = 0.69; cumulative distributions: P = 0.45] or amplitudes [f, bar graphs: Ctrl, 26.96 ± 2.37 pA, WD, 30.88 ± 3.14 pA, P = 0.32, t(23) = 1.02; cumulative distributions: P = 0.12] in V1. (g,h) Whisker-deprivation reduced mEPSC frequencies at P7 in both S1 [g, Ctrl, 0.48 ± 0.06 Hz, WD, 0.31 ± 0.02 Hz, P = 0.018, t(21) = 2.58] and V1 [h, Ctrl, 0.38 ± 0.05 Hz, WD, 0.18 ± 0.02 Hz, P = 0.002, t(20) = 3.57]. (i,j) Dark-rearing (DR) reduced mEPSC frequencies at P7 in both S1 [i, Ctrl, 0.42 ± 0.10 Hz, DR, 0.11 ± 0.02 Hz, P = 0.003, t(35) = 3.18] and V1 [j, Ctrl, 0.42 ± 0.11 Hz, DR, 0.07 ± 0.01 Hz, P = 0.003, t(39) = 3.15]. Error bars denote s.e.m.; “n” as denoted inside bar graphs. *P < 0.05, **P < 0.01, using unpaired two-tailed Student's t-tests for bar graphs and Kolmogorov-Smirnov two-sample tests for cumulative distributions.

Supplementary Figure 2 Changes in the level of hypothalamic neuropeptides under dark-rearing and whisker-deprivation conditions.

(a) Microarray results showing changes in the mRNA level of multiple neuropeptides in the hypothalamus of dark-reared mice at P14, as compared to controls; n = 2 for each condition. (b,c) Real-time qPCR results showing changes in the mRNA level of neuropeptides from the hypothalamus in dark-reared (b, n = 4 for each condition) or whisker-deprived (c, n = 4 or 9 for each condition) mice at P14. Error bars denote s.e.m.; “n” represents the number of mice. *P < 0.05, **P < 0.01, ***P < 0.001, using paired t-test.

Supplementary Figure 3 Immunolabeling oxytocin neurons in the PVN and apoptosis assay under sensory-deprivation conditions.

(a) Display of a full set of sections containing oxytocin-positive neurons from the PVN; every 6th section (30 μm thick) was immunolabeled and counted. Scale bar: 200 μm. (b) Co-localization of oxytocin (red) and neurophysin I (green) in the mouse brain. Scale bar: 200 μm. (c-e) Representative immunofluorescence double-staining images of neurophysin I (green), TUNEL (red) and overlay (merge) in the PVN of Ctrl, whisker-deprived and dark-reared mice. Negative: negative control for TUNEL staining. Positive: positive control for TUNEL staining. Quantitation of TUNEL-positive neurons: 0/681 neurons for whisker-deprived mice, 0/746 for dark-reared mice and 0/1422 for combined controls. Scale bar: 200 μm. (f) AAV-OXT-Venus expression (green) was specific in oxytocin neurons (red, oxytocin-antibody staining). Scale bar: 200 μm. (g) Representative images of oxytocin immunoreactive neurons and fibers amplified using the Tyramide Signal Amplification systems. Left: coronal section of the whole brain, scale bar: 1 mm. Right: zoomed image of the PVN, showing fibers of oxytocin neurons projecting towards the 3rd ventricle, V: ventricle, scale bar: 40 μm.

Supplementary Figure 4 Retrograde tracing successfully labeled known direct projections to the primary somatosensory cortex in developing mice.

(a) Representative image of coronal brain slice from a P14 mouse injected with the retrograde tracer CTB (green). The site of injection is marked using an arrow. (b) Zoomed image of the contralateral cortex, showing neurons clearly labeled with CTB. (c) Zoomed image of the thalamus, showing neurons clearly labeled with CTB (green). Mice were injected with CTB at P9 and sacrificed at P14-15. Scale bar: 200 μm.

Supplementary Figure 5 Retrograde tracing successfully labeled known direct projections from PVN oxytocin neurons to the central amygdala of adult mice.

(a) Representative images (left, combined DIC and green fluorescence; right, green fluorescence alone) of coronal brain slices showing the site of CTB injection (green) in central amygdala (CeA). (b-d) Representative images of brain slices containing CeA-projecting oxytocin neurons. Location of sections is indicated by their bregma positions, and the positions of the zoomed areas are as indicated. Neurons co-labeling with oxytocin (red) and CTB (green) are indicated by arrows. CTB was injected into the central amygdala of adult female mice, which were sacrificed 7 days following injection. Scale bar:100 μm.

Supplementary Figure 6 Developmental expression profile of oxytocin and oxytocin receptor as well as acute effects of oxytocin on synaptic transmission.

(a,b) Developmental expression pattern of oxytocin peptide in the blood plasma [a, P <0.001, F(6, 39) = 13.50] and oxytocin mRNA in the hypothalamus [b, P < 0.001, F(6, 22) = 20.90]. (c) The effect of different concentrations of oxytocin on mEPSC frequencies in S1 at P14 mice [Ctrl, 0.78 ± 0.08 Hz, 0.1 nM OXT, 0.83 ± 0.12 Hz, 1 nM OXT, 1.30 ± 0.23 Hz, 10 nM OXT, 1.40 ± 0.12 Hz, 100 nM OXT, 1.56 ± 0.14 Hz, 1,000 nM OXT, 2.07 ± 0.30 Hz, P < 0.001, F(5, 62) = 6.78]. (d) Bath application of oxytocin significantly reduced mEPSC frequencies in S1 in adult, 2 month old mice [left, Ctrl, 3.95 ± 0.81 Hz, OXT, 1.90 ± 0.29 Hz, P = 0.019, t(16) = 2.60, unpaired two-tailed Student's t-tests], without affecting mEPSC amplitudes [right, Ctrl, 13.81 ± 2.06 pA, OXT, 12.44 ± 1.30 pA, P = 0.57, t(16) = 0.58, unpaired two-tailed Student's t-tests]. (e) Developmental expression pattern of oxytocin receptor mRNA in the PFC, S1 and V1 [P < 0.001, F(2, 9) = 31.62]. (f) Both Oxtr-RNAi-1 (pSuper-RNAi-1) and Oxtr-RNAi-2 (pSuper-RNAi-2) were effective in lowering the level of over-expressed OXTR (HA-mOXTR) in HEK 293T cells. GAPDH was used as the loading control. (g) Representative recordings of paired-pulse ratios for conditions as indicated. (h) Bath application of oxytocin did not affect paired-pulse ratios in S1 at P14 (P > 0.05, unpaired two-tailed Student's t-tests). Error bars denote s.e.m.; “n” as denoted inside bar graphs. *P < 0.05, **P < 0.01, ***P < 0.001, using one-way ANOVA unless otherwise specified.

Supplementary Figure 7 Environmental enrichment from birth significantly increased inhibitory synaptic transmission in layer II/III pyramidal neurons of S1 and V1.

(a) Illustration of standard and environmentally enriched (EE) housings. (b,e) Representative mIPSC recordings (left) and average waveforms (right) for conditions as indicated from S1 (b) and V1 (e). (c,d) Environmental enrichment significantly increased mIPSC frequencies in S1 at P14 [c, bar graphs: Ctrl, 1.59 ± 0.23 Hz, EE, 3.90 ± 0.58 Hz, P < 0.001, t(34) = 3.69; cumulative distributions: P < 0.001], without affecting mEPSC amplitudes [d, bar graphs: Ctrl, 30.79 ± 2.06 pA, EE, 28.87 ± 1.62 pA, P = 0.47, t(34) = 0.73; cumulative distributions: P = 0.42]. (f,g) Environmental enrichment significantly increased mIPSC frequencies in V1 at P14 [f, bar graphs: Ctrl, 1.37 ± 0.19 Hz, EE, 2.57 ± 0.28 Hz, P < 0.01, t(21) = 3.68; cumulative distributions: P < 0.001], without affecting mEPSC amplitudes [g, bar graphs: Ctrl, 24.70 ± 1.12 pA, EE, 25.15 ± 1.60 pA, P = 0.82, t(21) = 0.24; cumulative distributions: P = 0.84]. Error bars denote s.e.m.; “n” as denoted inside bar graphs. **P < 0.01, ***P < 0.001, using unpaired two-tailed Student's t-tests for bar graphs and Kolmogorov-Smirnov two-sample tests for cumulative distributions.

Supplementary Figure 8 Full-length images of immunoblots presented in the main figures.

Full-length images of cropped immunoblots presented in Fig. 1m (top), Fig. 3f (middle) and Fig. 6h (bottom) are shown.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1 (PDF 1329 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zheng, JJ., Li, SJ., Zhang, XD. et al. Oxytocin mediates early experience–dependent cross-modal plasticity in the sensory cortices. Nat Neurosci 17, 391–399 (2014). https://doi.org/10.1038/nn.3634

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3634

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