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:

Lymph node conduits transport virions for rapid T cell activation

Abstract

Despite intense interest in antiviral T cell priming, the routes by which virions move in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined the locations of virions and infected cells after administration to mice of vaccinia virus or Zika virus. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Through the use of confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus–infected cells in the LN paracortex, which led to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T 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: VACV and MVA rapidly infect T cell zone cells associated with conduits.
Fig. 2: Conduit-associated VACV-infected cells can be visualized in the LN within an hour of infection.
Fig. 3: VACV virions can be transported in LN conduits.
Fig. 4: VACV infects conduit-associated DCs.
Fig. 5: ZIKV virions are also transported by conduits.
Fig. 6: CD8+ T cells are rapidly activated by virus in the T cell zone.
Fig. 7: VACV infects paracortical DCs at lower viral doses.
Fig. 8: T cells activated at the highest viral doses traffic rapidly to the infected tissue.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. Gretz, J. E., Norbury, C. C., Anderson, A. O., Proudfoot, A. E. & Shaw, S. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J. Exp. Med. 192, 1425–1440 (2000).

    Article  CAS  Google Scholar 

  2. Itano, A. A. et al. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity 19, 47–57 (2003).

    Article  CAS  Google Scholar 

  3. Sixt, M. et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 22, 19–29 (2005).

    Article  CAS  Google Scholar 

  4. Pape, K. A., Catron, D. M., Itano, A. A. & Jenkins, M. K. The humoral immune response is initiated in lymph nodes by B cells that acquire soluble antigen directly in the follicles. Immunity 26, 491–502 (2007).

    Article  CAS  Google Scholar 

  5. Gerner, M. Y., Torabi-Parizi, P. & Germain, R. N. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 42, 172–185 (2015).

    Article  CAS  Google Scholar 

  6. Rantakari, P. et al. The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes. Nat. Immunol. 16, 386–396 (2015).

    Article  CAS  Google Scholar 

  7. Gerner, M. Y., Casey, K. A., Kastenmuller, W. & Germain, R. N. Dendritic cell and antigen dispersal landscapes regulate T cell immunity. J. Exp. Med. 214, 3105–3122 (2017).

    Article  CAS  Google Scholar 

  8. Thierry, G. R. et al. The conduit system exports locally secreted IgM from lymph nodes. J. Exp. Med. 215, 2972–2983 (2018).

    Article  CAS  Google Scholar 

  9. Norbury, C. C., Malide, D., Gibbs, J. S., Bennink, J. R. & Yewdell, J. W. Visualizing priming of virus-specific CD8+ T cells by infected dendritic cells in vivo. Nat. Immunol. 3, 265–271 (2002).

    Article  CAS  Google Scholar 

  10. Hickman, H. D. et al. Direct priming of antiviral CD8+ T cells in the peripheral interfollicular region of lymph nodes. Nat. Immunol. 9, 155–165 (2008).

    Article  CAS  Google Scholar 

  11. Kastenmuller, W., Torabi-Parizi, P., Subramanian, N., Lammermann, T. & Germain, R. N. A spatially-organized multicellular innate immune response in lymph nodes limits systemic pathogen spread. Cell 150, 1235–1248 (2012).

    Article  CAS  Google Scholar 

  12. Kastenmuller, W. et al. Peripheral prepositioning and local CXCL9 chemokine-mediated guidance orchestrate rapid memory CD8+ T cell responses in the lymph node. Immunity 38, 502–513 (2013).

    Article  CAS  Google Scholar 

  13. Gaya, M. et al. Inflammation-induced disruption of SCS macrophages impairs B cell responses to secondary infection. Science 347, 667–672 (2015).

    Article  CAS  Google Scholar 

  14. Sagoo, P. et al. In vivo imaging of inflammasome activation reveals a subcapsular macrophage burst response that mobilizes innate and adaptive immunity. Nat. Med. 22, 64–71 (2016).

    Article  CAS  Google Scholar 

  15. Brewitz, A. et al. CD8+ T cells orchestrate pDC-XCR1+ dendritic cell spatial and functional cooperativity to optimize priming. Immunity 46, 205–219 (2017).

    Article  CAS  Google Scholar 

  16. Junt, T. et al. Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells. Nature 450, 110–114 (2007).

    Article  CAS  Google Scholar 

  17. Iannacone, M. et al. Subcapsular sinus macrophages prevent CNS invasion on peripheral infection with a neurotropic virus. Nature 465, 1079–1083 (2010).

    Article  CAS  Google Scholar 

  18. Sung, J. H. et al. Chemokine guidance of central memory T cells is critical for antiviral recall responses in lymph nodes. Cell 150, 1249–1263 (2012).

    Article  CAS  Google Scholar 

  19. Hickman, H. D. et al. Chemokines control naive CD8+ T cell selection of optimal lymph node antigen presenting cells. J. Exp. Med. 208, 2511–2524 (2011).

    Article  CAS  Google Scholar 

  20. Woodruff, M. C. et al. Trans-nodal migration of resident dendritic cells into medullary interfollicular regions initiates immunity to influenza vaccine. J. Exp. Med. 211, 1611–1621 (2014).

    Article  CAS  Google Scholar 

  21. Palframan, R. T. et al. Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J. Exp. Med. 194, 1361–1373 (2001).

    Article  CAS  Google Scholar 

  22. Katakai, T. et al. A novel reticular stromal structure in lymph node cortex: an immuno-platform for interactions among dendritic cells, T cells and B cells. Int. Immunol. 16, 1133–1142 (2004).

    Article  CAS  Google Scholar 

  23. Bajenoff, M. & Germain, R. N. B-cell follicle development remodels the conduit system and allows soluble antigen delivery to follicular dendritic cells. Blood 114, 4989–4997 (2009).

    Article  CAS  Google Scholar 

  24. Moss, B. Poxviridae: the viruses and their replication. in Fields Virology (eds. Knipe, D. M. & Howley, P. M.) 2849–2884 (Lippincott-Raven, 2001).

  25. Moss, B. et al. Host range restricted, non-replicating vaccinia virus vectors as vaccine candidates. Adv. Exp. Med. Biol. 397, 7–13 (1996).

    Article  CAS  Google Scholar 

  26. Pierson, T. C. & Diamond, M. S. The emergence of Zika virus and its new clinical syndromes. Nature 560, 573–581 (2018).

    Article  CAS  Google Scholar 

  27. Baden, L. R. et al. First-in-human randomized, controlled trial of mosaic HIV-1 immunogens delivered via a modified vaccinia Ankara vector. J. Infect. Dis. 218, 633–644 (2018).

    Article  Google Scholar 

  28. La Rosa, C. et al. MVA vaccine encoding CMV antigens safely induces durable expansion of CMV-specific T cells in healthy adults. Blood 129, 114–125 (2017).

    Article  Google Scholar 

  29. Stan, R. V. et al. The diaphragms of fenestrated endothelia: gatekeepers of vascular permeability and blood composition. Dev. Cell 23, 1203–1218 (2012).

    Article  CAS  Google Scholar 

  30. Sirohi, D. et al. The 3.8 Å resolution cryo-EM structure of Zika virus. Science 352, 467–470 (2016).

    Article  CAS  Google Scholar 

  31. Hogquist, K. A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).

    Article  CAS  Google Scholar 

  32. Hickman, H. D. et al. CXCR3 chemokine receptor enables local CD8+ T cell migration for the destruction of virus-infected cells. Immunity 42, 524–537 (2015).

    Article  CAS  Google Scholar 

  33. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  CAS  Google Scholar 

  34. Belz, G. T. et al. Cutting edge: conventional CD8α+ dendritic cells are generally involved in priming CTL immunity to viruses. J. Immunol. 172, 1996–2000 (2004).

    Article  CAS  Google Scholar 

  35. Gerner, M. Y., Kastenmuller, W., Ifrim, I., Kabat, J. & Germain, R. N. Histo-cytometry: a method for highly multiplex quantitative tissue imaging analysis applied to dendritic cell subset microanatomy in lymph nodes. Immunity 37, 364–376 (2012).

    Article  CAS  Google Scholar 

  36. Link, A. et al. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat. Immunol. 8, 1255–1265 (2007).

    Article  CAS  Google Scholar 

  37. Denton, A. E., Roberts, E. W., Linterman, M. A. & Fearon, D. T. Fibroblastic reticular cells of the lymph node are required for retention of resting but not activated CD8+ T cells. Proc. Natl Acad. Sci. USA 111, 12139–12144 (2014).

    Article  CAS  Google Scholar 

  38. Cremasco, V. et al. B cell homeostasis and follicle confines are governed by fibroblastic reticular cells. Nat. Immunol. 15, 973–981 (2014).

    Article  CAS  Google Scholar 

  39. Bajenoff, M. et al. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity 25, 989–1001 (2006).

    Article  CAS  Google Scholar 

  40. Acton, S. E. et al. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature 514, 498–502 (2014).

    Article  CAS  Google Scholar 

  41. Astarita, J. L. et al. The CLEC-2–podoplanin axis controls the contractility of fibroblastic reticular cells and lymph node microarchitecture. Nat. Immunol. 16, 75–84 (2015).

    Article  CAS  Google Scholar 

  42. Yang, C. Y. et al. Trapping of naive lymphocytes triggers rapid growth and remodeling of the fibroblast network in reactive murine lymph nodes. Proc. Natl Acad. Sci. USA 111, E109–E118 (2014).

    Article  CAS  Google Scholar 

  43. Estes, J. D. Pathobiology of HIV/SIV-associated changes in secondary lymphoid tissues. Immunol. Rev. 254, 65–77 (2013).

    Article  Google Scholar 

  44. Mueller, S. N. et al. Viral targeting of fibroblastic reticular cells contributes to immunosuppression and persistence during chronic infection. Proc. Natl Acad. Sci. USA 104, 15430–15435 (2007).

    Article  CAS  Google Scholar 

  45. Ng, C. T., Nayak, B. P., Schmedt, C. & Oldstone, M. B. Immortalized clones of fibroblastic reticular cells activate virus-specific T cells during virus infection. Proc. Natl Acad. Sci. USA 109, 7823–7828 (2012).

    Article  CAS  Google Scholar 

  46. Scandella, E. et al. Restoration of lymphoid organ integrity through the interaction of lymphoid tissue–inducer cells with stroma of the T cell zone. Nat. Immunol. 9, 667–675 (2008).

    Article  CAS  Google Scholar 

  47. Davis, K. J. et al. Pathology of experimental Ebola virus infection in African green monkeys. Involvement of fibroblastic reticular cells. Arch. Pathol. Lab. Med. 121, 805–819 (1997).

    CAS  PubMed  Google Scholar 

  48. Steele, K. E., Anderson, A. O. & Mohamadzadeh, M. Fibroblastic reticular cell infection by hemorrhagic fever viruses. Immunotherapy 1, 187–197 (2009).

    Article  CAS  Google Scholar 

  49. Steele, K. E., Anderson, A. O. & Mohamadzadeh, M. Fibroblastic reticular cells and their role in viral hemorrhagic fevers. Expert Rev. Anti Infect. Ther. 7, 423–435 (2009).

    Article  CAS  Google Scholar 

  50. Twenhafel, N. A. et al. Pathology of experimental aerosol Zaire ebolavirus infection in rhesus macaques. Vet. Pathol. 50, 514–529 (2013).

    Article  CAS  Google Scholar 

  51. Srinivasan, R. S. et al. Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature. Genes Dev. 21, 2422–2432 (2007).

    Article  CAS  Google Scholar 

  52. Hickman, H. D. et al. Anatomically restricted synergistic antiviral activities of innate and adaptive immune cells in the skin. Cell Host Microbe 13, 155–168 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Intramural Research Program of NIAID, NIH (H.D.H.). R.V.S. was supported by NIH grants R21CA172983, R21CA175592, R01GM120592, and AHA 16GRNT27260362. We thank K. Dowd and T. Pierson (Viral Pathogenesis Section, Laboratory of Viral Diseases, NIAID) for assistance with ZIKV infection and J. Kabat (Biological Imaging Section, NIAID) for assistance with microscopy image analysis. G. Oliver (University of Chicago) provided Prox1-CreERT2tg/+ mice on the C57BL/6 genetic background.

Author information

Authors and Affiliations

Authors

Contributions

H.D.H. conceived the project. H.D.H., R.V.S., and J.W.Y. secured funding. H.D.H., G.V.R., A.S.W., J.P.S., D.T.M., L.S., and J.L.A. performed experiments and analyzed data. R.V.S. provided mice and technical expertise. H.D.H. wrote the manuscript. H.D.H., R.V.S., and J.W.Y. edited the manuscript.

Corresponding authors

Correspondence to Jonathan W. Yewdell or Heather D. Hickman.

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 Staining with an antibody to VACV-Ag confirms infection of cells expressing viral promoter–driven eGFP.

a) Maximum intensity projections (MIPs) of sections of popliteal LNs harvested 8 h after footpad infection with VACV-NP-S-eGFP. ERTR7 = red, rabbit polyclonal antibody against total VACV antigens = white, VACV-infected cells = green nuclear signal, B220 = blue. Scalebar = 200 ÎĽm. Boxes highlight different areas of the node in which infected cells are located. Higher magnification images are shown in the insets. Scalebars = 20 ÎĽm. For ease of viewing VACV-infected cells, the VACV-antigen-antibody signal (white) is omitted on the same inset image on the right hand side. b) As in a) but larger deconvolved images showing nuclear eGFP signal surround by white, cytoplasmic VACV Ag staining (VACV replicates in the cytoplasm). VACV eGFP signal (green) is omitted on the right. Scalebars = 20 ÎĽm. Images are representative of 6 nodes from 3 different experiments.

Supplementary Figure 2 VACV-infected cells can be visualized in all classified nodal locations by 8 h after infection.

Maximum intensity projections (MIPs) of sections of popliteal LNs harvested 8 h after footpad infection with VACV-NP-S-eGFP. ERTR7 = red, Lyve-1 = white, VACV-infected cells = green nuclear signal, B220 = blue. Scalebar = 200 ÎĽm. Boxes highlight different areas of the node in which infected cells are located. Higher magnification images are shown corresponding to boxed areas. Scalebars = 20 ÎĽm. Left panels show merged image; right panels show only B cells + conduits + VACV-infected cells. Images are representative of 6 nodes from 3 different experiments.

Supplementary Figure 3 GFP+ virus-infected cells can be visualized in the T cell zone by 4 h after infection.

Maximum intensity projections (MIPs) of sections of popliteal LNs harvested 4 h after footpad injection of MVA-(a-b) or VACV (c-d). ERTR7 = red, Lyve-1 = white, VACV- or MVA-infected cells = green nuclear signal, B220 = blue. Arrows point to some of the T cell zone virus-infected cells associated with conduits. Scalebars = 100 ÎĽm. Higher magnification images are shown to the right in (b and d). Scalebars = 15 ÎĽm. e) Deconvolved, high-magnification, blended-view images showing the association of T cell zone MVA-infected cells with conduits at 4 h p.i. Scalebar = 20 ÎĽm. Boxes indicate areas with increased magnification on the right. Scalebars = 5 ÎĽm. Images are representative of 6 nodes from 3 different experiments.

Supplementary Figure 4 Vaccinia virions are transported in nodal conduits.

a-b) Transmission electron micrographs of sections of popliteal LNs harvested 5 m after footpad infection of 108 PFU of 10 nM gold particle-conjugated VACV virions (indicated with an arrow). Panel a) shows a magnified image of panel b). Panel a) Scalebars = 200 nm. b) = 1 ÎĽm. Arrows indicate the same VACV virion in the two images. The conduits (labeled with a C) can be identified by organized collagen structures running either parallel (a-d) or anti-parallel (e-f) to the field of view and are lined by fibroblastic reticular cells (FRC). T = T cell. c-d) Transmission electron micrograph as in panel a-b, but in a different area of the T cell zone. Panel c) shows a magnified image of panel d). Panel c) Scalebars = 100 nm. d) = 1 ÎĽm. An arrow points to the same VACV virion in the two images. e-f) Transmission electron micrographs showing that cells near the conduits (labeled C) in the LN T cell zone of the node acquire VACV (indicated by arrows). In these images, the collagen in the conduit is running anti-parallel to the images and the FRCs that ensheath the conduit can be seen. Panel e) Scalebars = 500 nm. f) = 2 ÎĽm. Results are indicative of 14 grids from 2 separate experiments.

Supplementary Figure 5 Single-color stains for cell-surface markers on VACV-infected cells in the LN.

a-f) Maximum intensity projections (MIPs) of sections of popliteal LNs harvested 8 h after footpad injection of 108 PFU of VACV. Different cell surface markers are shown in white (indicated above each image). Staining in white for a) CD11b, b) CD169, c) F4/80, d) CD11c, e) CD205, f) SIGNR1. Scalebars = 100 ÎĽm. VACV-infected cells = green, B220 = blue. Some of the infected cells positive for each surface marker are indicated by tan circles. Higher magnification images of infected cells are shown below the whole LN section. Scalebars = 20 ÎĽm. Images are representative of 10 nodes from 4 different experiments.

Supplementary Figure 6 ZIKV analysis in popliteal LNs.

a) Maximum intensity projection (MIPs) of a section of a popliteal LN of an Ifnar -/- mouse infected with 104 FFU ZIKV in the footpad. LNs were harvested 24 h post-infection. Section was stained with secondary Ab only to show background staining for ZIKV-infected cells (white). Insets show higher magnification view without (left) or with (right) staining for CD11c (green). ERTR7 = red, B220 = blue. Scalebars = 200 μm (left), 20 μm (right higher mag.) Image is representative of 3 nodes from 2 different experiments. b-e) Electron micrographs of LNs infected and harvested as in (a). Conduits are labeled with a “C.” Arrows point to virions in the conduit. For clarity, the borders of conduits are lined with yellow dashed lines. b) Middle and right panels show higher magnification views of the same conduit in the left panels. ZIKV virions can be seen at three locations in this conduit. Left panel scalebar = 2 μm, middle = 500 nm, middle inset = 100 nm, right = 500 nm, right inset =100 nm. c) Right panel (scalebar= 500 nm) shows higher magnification view of left panel (scalebar = 100 nm). d-e) Two different conduits with ZIKV virions. Left scalebar = 200 nm, right scalebar = 50 nm. 24 grids from 6 nodes and 3 experiments were analyzed.

Supplementary Figure 7 CD69 expression is highest on T cells in the T cell zone at 8 h after infection.

a) Maximum intensity projections (MIPs) of sections of popliteal LNs harvested 8 h after footpad injection of 108 PFU of VACV-NP-S-eGFP. OT-I CD8+ T cells = red, VACV-infected cells = green, CD69 = white, B220 = blue. Left panel) all stains merged. Right panel) OT-I CD8+ T cell activation was assessed by colocalization of the red signal with the white signal (with the colocalized signal shown in purple). Arrows point to some of the activated CD8+ T cells. Scalebars = 100 ÎĽm. 10 LNs from 5 experiments were analyzed. b) MIP as in (a) with a VACV lacking SIINFEKL (VACV-NP-eGFP). Color-coded numbers indicating the CD69 mean fluorescent intensity (MFI) for each T cell (with SCS&IFA numbers in pink, medulla in green, T cell zone in yellow, B cell follicles in blue). Scalebar = 200 ÎĽm. c) Quantification of the CD69 MFIs of all OT-I CD8+ T cells in either the SCS&IFA (red), medulla (green), T cell zone (yellow), or B cell follicle (blue). Dots = Individual T cells from 1 LN section. n = 195 total T cells. Bar = mean. Statistics were calculated using a Mann-Whitney test. 6 LNs from 3 experiments were analyzed. d-e) as in (b-c) except with a VACV expressing SIINFEKL (VACV-NP-S-eGFP). Scalebar = 200 ÎĽm. e) Dots = Individual T cells from 1 LN section. n = 187 total T cells. Bar = mean. Statistics were calculated using a Mann-Whitney test. 6 LNs from 3 experiments were analyzed.

Supplementary Figure 8 OT-I CD8+ T cells are activated mainly in the LN SCS and IFA region at a low dose of virus and in the T cell zone at a high dose.

a) Maximum intensity projections (MIPs) of sections of popliteal LNs harvested 8 h after footpad injection of the indicated dose of VACV (top panels) or MVA (bottom panels). ERTR7 = white, infected cells = green nuclear signal. Left panels at each viral dose = merge; right panels = only infected cells. Scalebars = 100 ÎĽm. b) Number of infected cells at each LN location with varying viral dose. Dots represent the number of infected cells per LN section when popliteal LNs were harvested at 8 h post-footpad infection. n = 10 LNs/dose for all infections except: VACV at 108 PFU (n = 18); MVA and VACV at 107 PFU (n = 12). LNs came from at least 2 different experiments per group. c) The total number of infected cells per LN section correlates with the number of infected cells located in the T cell zone. Data from all viral doses in (b) were combined to create (c). d) MIPs of sections of popliteal LNs harvested 8 h after footpad injection at the indicated dose of VACV. OT-I cells = red, VACV-infected cells = green, B220 = blue, CD69 = white. Scalebars = 100 ÎĽm. The boxed area of the LN is magnified in the images below each section. Scalebars = 20 ÎĽm. In the far-right panels, OT-I CD8+ T cell activation was assessed by colocalization of the red signal with the white signal (with the colocalized signal shown in purple). Arrows point to activated CD8+ T cells. (Note that VACV-NP-eGFP does not contain SIINFEKL and should not activate OT-I CD8+ T cells, but does activate endogenous VACV-specific CD8+ T cells). Images are representative of 5 LNs per group from 2 different experiments.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reynoso, G.V., Weisberg, A.S., Shannon, J.P. et al. Lymph node conduits transport virions for rapid T cell activation. Nat Immunol 20, 602–612 (2019). https://doi.org/10.1038/s41590-019-0342-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0342-0

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