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:

Coupled analysis of transcriptome and BCR mutations reveals role of OXPHOS in affinity maturation

Abstract

Antigen-activated B cells diversify variable regions of B cell antigen receptors by somatic hypermutation in germinal centers (GCs). The positive selection of GC B cells that acquire high-affinity mutations enables antibody affinity maturation. In spite of considerable progress, the genomic states underlying this process remain to be elucidated. Single-cell RNA sequencing and topic modeling revealed increased expression of the oxidative phosphorylation (OXPHOS) module in GC B cells undergoing mitoses. Coupled analysis of somatic hypermutation in immunoglobulin heavy chain (Igh) variable gene regions showed that GC B cells acquiring higher-affinity mutations had further elevated expression of OXPHOS genes. Deletion of mitochondrial Cox10 in GC B cells resulted in reduced cell division and impaired positive selection. Correspondingly, augmentation of OXPHOS activity with oltipraz promoted affinity maturation. We propose that elevated OXPHOS activity promotes B cell clonal expansion and also positive selection by tuning cell division times.

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: Experimental design and scRNA-seq analysis of GC B cell response.
Fig. 2: Topic modeling uncovers prominent biological programs and reveals distinctive expression dynamics of OXPHOS and glycolysis in GC B cells.
Fig. 3: Coupled analysis of SHM and transcriptomes of NP-specific GC B cells implicates increased OXPHOS in positive selection.
Fig. 4: Coupled scRNA-seq analysis of OVA response reveals increased OXPHOS programming in positively selected GC B cell clones.
Fig. 5: Cox10 deficiency compromises cell division and positive selection of GC B cells.
Fig. 6: Oltipraz enhancement of OXPHOS activity during a GC response facilitates positive selection.

Similar content being viewed by others

Data availability

The scRNA-seq and bulk BCR-seq data have been deposited in the Gene Expression Omnibus database under accession number GSE154634. Source data are provided with this paper. All other data that support the findings of this study are available from the corresponding authors upon reasonable request.

Code availability

Code used in this study is available at Github: https://github.com/chendianyu/2021_NI_scGCB/.

References

  1. Victora, G. D. & Nussenzweig, M. C. Germinal centers. Annu. Rev. Immunol. 30, 429–457 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Mayer, C. T. et al. The microanatomic segregation of selection by apoptosis in the germinal center. Science 358, eaao2602 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Gitlin, A. D. et al. T cell help controls the speed of the cell cycle in germinal center B cells. Science 349, 643–646 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. De Silva, N. S. & Klein, U. Dynamics of B cells in germinal centres. Nat. Rev. Immunol. 15, 137–148 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Victora, G. D. et al. Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 143, 592–605 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Khalil, A. M., Cambier, J. C. & Shlomchik, M. J. B cell receptor signal transduction in the GC is short-circuited by high phosphatase activity. Science 336, 1178–1181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mueller, J., Matloubian, M. & Zikherman, J. Cutting edge: an in vivo reporter reveals active B cell receptor signaling in the germinal center. J. Immunol. 194, 2993–2997 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Nowosad, C. R., Spillane, K. M. & Tolar, P. Germinal center B cells recognize antigen through a specialized immune synapse architecture. Nat. Immunol. 17, 870–877 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Luo, W., Weisel, F. & Shlomchik, M. J. B cell receptor and CD40 signaling are rewired for synergistic induction of the c-Myc transcription factor in germinal center B cells. Immunity 48, 313–326.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mesin, L., Ersching, J. & Victora, G. D. Germinal center B cell dynamics. Immunity 45, 471–482 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shlomchik, M. J., Luo, W. & Weisel, F. Linking signaling and selection in the germinal center. Immunol. Rev. 288, 49–63 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kennedy, D. E. et al. Novel specialized cell state and spatial compartments within the germinal center. Nat. Immunol. 21, 660–670 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. King, H. W. et al. Single-cell analysis of human B cell maturation predicts how antibody class switching shapes selection dynamics. Sci. Immunol. 6, eabe6291 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Jellusova, J. et al. Gsk3 is a metabolic checkpoint regulator in B cells. Nat. Immunol. 18, 303–312 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cho, S. H. et al. Germinal centre hypoxia and regulation of antibody qualities by a hypoxia response system. Nature 537, 234–238 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Weisel, F. J. et al. Germinal center B cells selectively oxidize fatty acids for energy while conducting minimal glycolysis. Nat. Immunol. 21, 331–342 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wagner, A. et al. In silico modeling of metabolic state in single Th17 cells reveals novel regulators of inflammation and autoimmunity. J. Immunol. 204 (Suppl.), 150.22 (2020).

    Article  Google Scholar 

  18. Temate-Tiagueu, Y. et al. Inferring metabolic pathway activity levels from RNA-seq data. BMC Genomics 17, 542 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Hancock, T., Takigawa, I. & Mamitsuka, H. Mining metabolic pathways through gene expression. Bioinformatics 26, 2128–2135 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Furukawa, K., Akasako-Furukawa, A., Shirai, H., Nakamura, H. & Azuma, T. Junctional amino acids determine the maturation pathway of an antibody. Immunity 11, 329–338 (1999).

    Article  CAS  PubMed  Google Scholar 

  21. Jacob, J. & Kelsoe, G. In situ studies of the primary immune response to (4-hydroxy-3-nitrophenyl)acetyl. II. A common clonal origin for periarteriolar lymphoid sheath-associated foci and germinal centers. J. Exp. Med. 176, 679–687 (1992).

    Article  CAS  PubMed  Google Scholar 

  22. Allen, D., Simon, T., Sablitzky, F., Rajewsky, K. & Cumano, A. Antibody engineering for the analysis of affinity maturation of an anti-hapten response. EMBO J. 7, 1995–2001 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kowalczyk, M. S. et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 25, 1860–1872 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Blei, D. M., Ng, A. Y. & Jordan, M. I. Latent Dirichlet allocation. J. Mach. Learn. Res. 3, 993–1022 (2003).

    Google Scholar 

  25. Xu, H. et al. Transcriptional atlas of intestinal immune cells reveals that neuropeptide ɑ-CGRP modulates group 2 innate lymphoid cell responses. Immunity 51, 696–708.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bielecki, P. et al. Skin-resident innate lymphoid cells converge on a pathogenic effector state. Nature 592, 128–132 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ersching, J. et al. Germinal center selection and affinity maturation require dynamic regulation of mTORC1 kinase. Immunity 46, 1045–1058.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhu, X. et al. Analysis of the major patterns of B cell gene expression changes in response to short-term stimulation with 33 single ligands. J. Immunol. 173, 7141–7149 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Dominguez-Sola, D. et al. The proto-oncogene MYC is required for selection in the germinal center and cyclic reentry. Nat. Immunol. 13, 1083–1091 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Weiser, A. A. et al. Affinity maturation of B cells involves not only a few but a whole spectrum of relevant mutations. Int. Immunol. 23, 345–356 (2011).

    Article  CAS  PubMed  Google Scholar 

  31. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  32. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e27 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Turchaninova, M. A. et al. High-quality full-length immunoglobulin profiling with unique molecular barcoding. Nat. Protoc. 11, 1599–1616 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Diaz, F., Thomas, C. K., Garcia, S., Hernandez, D. & Moraes, C. T. Mice lacking COX10 in skeletal muscle recapitulate the phenotype of progressive mitochondrial myopathies associated with cytochrome c oxidase deficiency. Hum. Mol. Genet. 14, 2737–2748 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Xu, H. et al. Regulation of bifurcating B cell trajectories by mutual antagonism between transcription factors IRF4 and IRF8. Nat. Immunol. 16, 1274–1281 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Chamoto, K. et al. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc. Natl Acad. Sci. USA 114, E761–E770 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yu, Z. et al. Oltipraz upregulates the nuclear factor (erythroid-derived 2)-like 2 (NRF2) antioxidant system and prevents insulin resistance and obesity induced by a high-fat diet in C57BL/6J mice. Diabetologia 54, 922–934 (2011); erratum 54, 989 (2011).

  38. Nojima, T. et al. In-vitro derived germinal centre B cells differentially generate memory B or plasma cells in vivo. Nat. Commun. 2, 465 (2011).

    Article  PubMed  Google Scholar 

  39. Raybuck, A. L. et al. B cell–intrinsic mTORC1 promotes germinal center–defining transcription factor gene expression, somatic hypermutation, and memory B cell generation in humoral immunity. J. Immunol. 200, 2627–2639 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Jang, K. J. et al. Mitochondrial function provides instructive signals for activation-induced B-cell fates. Nat. Commun. 6, 6750 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Salazar-Roa, M. & Malumbres, M. Fueling the cell division cycle. Trends Cell Biol. 27, 69–81 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Wang, Z. et al. Cyclin B1/Cdk1 coordinates mitochondrial respiration for cell-cycle G2/M progression. Dev. Cell 29, 217–232 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Biton, M. et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell 175, 1307–1320.e22 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Huang for discussions and technical advice; W. Zhu for mouse genotyping; the Laboratory Animal Resources Center, the High-Performance Computing Center, the Flow Cytometry Core and the Genomics Core in Westlake University; and the Division of Laboratory Animal Resources at the University of Pittsburgh. Research was supported by the National Natural Science Foundation of China (grant nos. 31970842 and U20A20346; H.X.), the National Key R&D Program of China (grant nos. 2020YFA0804200 and 2019YFA0802900; H.X.) and the Education Foundation of Westlake University (H.X.). H.S. acknowledges support from the UPMC ITTC fund and NIH grant no. U01AI141990.

Author information

Authors and Affiliations

Authors

Contributions

H.X. and H.S. conceived and supervised this study. D.C. performed computational analysis with guidance from H.X. and N.S. Y.W. and G.K.M.V. performed the experiments with help from S.F., D.X. and C.W.N. D.H., H.X. and H.S. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Harinder Singh or Heping Xu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Laurence Morel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Experimental design, mitotic and spatial gene signature scores of GC B cells.

a, Experimental design. WT C57BL/6 mice were immunized with NP-KLH plus LPS and Alum. B220+FAShiGL7hi NP-specific GC B cells were sorted for scRNA-seq analysis on day 13 after immunization. b, A 2D embedding as in Fig. 1c, colored by batches. c, Correlation plot of DZ and LZ signature scores in GC B cells. Shown is the mitotic (top row) and spatial (bottom row) gene signature scores of individual cells in different clusters. d, Violin plots of the distribution of gray zone signature scores (Supplementary Table 2) of GC B cells in different clusters. Clusters enriched for cells with high mitotic activity are highlighted with grey background. Diamond indicates the mean; lines, first and third quartiles. Each symbol represents a cell (b,c).

Extended Data Fig. 2 Topic modeling, pseudotime and 2-NBDG uptake analysis of GC B cells.

a, Shown are 2D embedding as in Fig. 1c, colored by the topic’s weight in the cell. Biologically interested programs are highlighted. b, Violin plots of the distribution of CD40 signaling-responded gene signature score (Supplementary Table 2) of GC B cells. c,d, Pseudotime trajectory of GC B cells constructed by Monocle (c). Violin plots of the distribution of pseudotime scores of cells (d). e, Violin plots of the distribution of topic 1 weights of GC B cells. f, A representative flow plot showing the gating strategy for DZ versus LZ GC B cells (above) (Supplementary Gating Strategy). A representative flow histogram showing the intracellular ATP5A expression in DZ and LZ GC B cells (bottom). g, A representative flow histogram showing intracellular TOMM20 expression in DZ and LZ GC B cells. h, Violin plots of the distribution of fatty acid beta oxidation signature scores of GC B cells. i, A representative flow plot showing the gating strategy for analyzing its uptake in GC (red) and resting (black) B cells (top) (Supplementary Gating Strategy). The 2-NBDG labeling of GC and resting B cell pairs from individual mice (n = 8) are displayed, connected by lines (bottom). j, The 2-NBDG labeling of NP+ DZ and NP+ LZ GC B cell pairs from individual mice (n = 8) are displayed, connected by lines. Data are pooled from two independent experiments. Clusters enriched for cells with high mitotic activity are highlighted with grey background and diamond indicates the mean and lines indicate first and third quartiles (b,d,e,h). Data are representative of two (f,g) or the pool from two (i,j) independent experiments. Statistical significance was tested by one-way ANOVA followed by Tukey’s multiple comparisons test (b,d,e,h) or two-tailed paired t test (i,j).

Source data

Extended Data Fig. 3 Coupled analysis of Igh variable sequences with transcriptional states of NP-specific GC B cells.

a, Violin plots of the distribution of mutation frequencies of GC B cells in different clusters. b, Violin plots of the distribution of weights of different topics (Extended Data Fig. 2a) in scRNA-seq data of IgHV1-72*01-expressing GC B cells with or without HFMs. c-e, Violin plots of the distribution of OXPHOS signature scores in scRNA-seq data of IgHV1-72*01-expressing GC B cells in designated clusters (c), isotypes (d) or mutation numbers (e). Each dot represents a cell. Diamond indicates the mean. f, Violin plot of the distribution of signature score of glutamine metabolism (Supplementary Table 2) in scRNA-seq data of IgHV1-72*01-expressing GC B cells with or without indicated high affinity mutations. g, Differentially expressed genes in IgHV1-72*01-encoding GC B cells with or without indicated high affinity mutations. Top10 differentially expressed genes (dots) are labeled. Diamond indicates the mean and lines indicate first and third quartiles (a,b,f). Statistical significance was tested by two-tailed t test (b,f) or two-way ANOVA followed by Tukey’s multiple comparisons test (c-e).

Extended Data Fig. 4 Coupled analysis of Igh variable sequences and transcriptional states of GC B cells in response to OVA immunization.

a, Experimental design. WT C57BL/6 mice were immunized with OVA plus LPS and Alum. B220+FAShiGL7hi NP-specific GC B cells were sorted on day 13 after immunization for scRNA-seq analysis. b, Proportions of GC B cells (FashiGL-7hi) in splenic B cell (CD19+) compartments (Supplementary Gating Strategy) of mice on day 13 after immunization with adjuvants only (n = 3) or adjuvants plus OVA (n = 5). Each symbol represents an individual mouse. c, Bar plot showing the counts of top 40 assigned Igh variable genes (y-axis) in GC B cells analyzed on day 13 of the response to OVA immunization. d,e, Frequency of amino-acid substitutions across IgHV1-26*01 (d) and IgHV2-5*01 (e) sequences. The most frequent mutated positions are highlighted in red. f, Pie chart displays the proportions of IgHV2-5*01 sequences (166) with indicated amino acid substitutions at position 56. Each color represents designated amino acid, and the white sector indicates germline sequences (S). g, Violin plots of the distribution of OXPHOS signature scores (Methods, Supplementary Table 2) in scRNA-seq data of IgHV2-5*01-expressing GC B cells containing or lacking the dominant mutation. Each dot represents a cell, line indicates the mean. Data are pooled from three independent experiments shown as the mean±s.e.m. (b). Statistical significance was tested by two-tailed t test (b,g).

Source data

Extended Data Fig. 5 Seahorse, Flow cytometry and ELISA analysis of Cox10 deficient B cells.

a,b, Representative trace (a) and quantitative analyses of basal (b, left) or maximum OCR (b, left) of indicated cell lines with Cox10 gene edited via CRISPR-Cas9 in the Seahorse XFe96 flux analyzer (n = 9 cell cultures per experiment). c, Proportions of NP-specific GC B cells (CD38GL7hiNP+) in splenic B cell (B220+) compartments of Cox10+/+Aicda+/cre (n = 10 on dpi 7, n = 9 on dpi13, n = 6 on dpi21) and Cox10fl/flAicda+/cre (n = 5 on dpi 7, n = 6 on dpi13, n = 6 on dpi21) mice. d,e, ELISPOT analysis of splenocytes from Cox10+/+Aicda+/cre (n = 2 on dpi 7, n = 9 on dpi13, n = 6 on dpi21) and Cox10fl/flAicda+/cre (n = 3 on dpi 7, n = 6 on dpi13, n = 6 on dpi21) mice. Quantitative analysis at indicated dpi (d), representative images of NP-specific IgG1+ ASCs on dpi21 (e). f, Numbers of naive B cells (B220+CD38+GL7CD138) in Cox10+/+Aicda+/cre (n = 22) and Cox10fl/flAicda+/cre (n = 15) mice. g, Proportions of EdU+ cells in NP-specific GC B cell compartment (FashiGL7hiNP+) of Cox10+/+Aicda+/cre (n = 5) and Cox10fl/flAicda+/cre (n = 6) mice on dpi13, 2 hours post EdU administration. h, NP-specific IgG1 titers of Cox10+/+Aicda+/cre (n = 6) and Cox10fl/flAicda+/cre (n = 5) mice on dpi21. Shown are serial dilutions of serum samples for binding to NP5 (left) and NP30 (right). Data are representative of two (a,b) or five (e) or the pool from five (c,d,f) or two (g,h) independent experiments shown as the mean ± s.e.m. Each symbol represents an individual well (b) or mouse (f,g). Statistical significance was tested by two-tailed t test (b,f,g) or two-way ANOVA followed by Tukey’s multiple comparisons test (c,d).

Source data

Extended Data Fig. 6 Flow cytometry analysis of iCG B cells and TFH cells.

a, Experimental design. b, Representative flow plots showing the expression FAS and GL-7 of primary GC (left) and iGC (right) B cells cultured as in (a). Cells were gated on B220+ population (Supplementary Gating Strategy). c,d, Representative trace (c) and quantitative analyses of basal (d, left) or maximum OCR (d, right) of indicated cells in the Seahorse XFe96 flux analyzer (n = 6 cell cultures per experiment). e-i, Proportions of total and NP-specific GC B cells (e, f, n = 7 mice per group in e, n = 5 for DMSO and n = 6 for oltipraz groups in f), total and NP-specific plasma cells (g, h, n = 7 mice per group in g, n = 5 for DMSO and n = 6 for oltipraz groups in h), as well as NP-specific IgG1+ memory B cells (i, n = 7 mice per group on dpi13, n = 5 for DMSO and n = 6 on dpi21) of mice treated as in Fig. 6a at indicated time points after immunization. j, Proportions of TFH cells (CXCR5hiPD-1hi) in splenic activated CD4+ T cell (B220CD4+CD44+, Supplementary Gating Strategy) compartments of mice treated with oltipraz (n = 6) or DMSO (n = 5) as in Fig. 6a on dpi21. k, The expression of Il21 (left) and Il4 (right) in TFH cells of mice treated with oltipraz (n = 7) or DMSO (n = 7) as in Fig. 6a on dpi13. l, m, NP-specific IgG1 titers of mice treated as in Fig. 6a on dpi13 (l) and dpi21 (m). Shown are serial dilutions of serum samples for binding to NP2 (left) and NP27 (right). Data are representative of three independent experiments (b,c,d) or the pool from two (e-k) or four (l,m) independent experiments. Each symbol represents an individual well (d) mouse (e-k). Statistical significance was tested by two-tailed t test (d-k).

Source data

Supplementary information

Supplementary Information

Gating strategies for total and NP-specific GC B cells, DZ and LZ GC B cells, plasma cells, IgG1+ NP-specific memory B cells and TFH cells.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, D., Wang, Y., Manakkat Vijay, G.K. et al. Coupled analysis of transcriptome and BCR mutations reveals role of OXPHOS in affinity maturation. Nat Immunol 22, 904–913 (2021). https://doi.org/10.1038/s41590-021-00936-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00936-y

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