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:

Transmission of trained immunity and heterologous resistance to infections across generations

An Author Correction to this article was published on 18 January 2023

Matters Arising to this article was published on 20 January 2022

This article has been updated

Abstract

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.

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: Infection of male mice increases the resistance of offspring to infections.
Fig. 2: Infection of male mice increases progeny responsiveness.
Fig. 3: Training of male mice induces cellular changes in the bone marrow of offspring.
Fig. 4: Infection induces transcriptional changes in bone marrow progenitors of F1 control and F1 exposed offspring.
Fig. 5: Infection with C. albicans induces changes in the DNA methylation landscape of sperm.

Similar content being viewed by others

Data availability

Data from the RNA-seq, ATAC-seq and sperm DNA methylation (RRBS) experiments have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE130327. Raw sequencing data and sample annotations are available from the GEO under accession number GSE130327. The mouse reference genome (mm10, build GRCm38) was accessed from the UCSC genome browser (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/). Blacklisted genomic regions were downloaded from http://mitra.stanford.edu/kundaje/akundaje/release/blacklists/mm10-mouse/mm10.blacklist.bed.gz. Prebuilt gene sets for GSEA analysis were downloaded from the GO2MSIG (http://www.bioinformatics.org/go2msig/). The ATAC-seq peaks were annotated using annotatePeaks.pl (http://homer.ucsd.edu/homer/ngs/annotation.html), Reference SNPs and indels from the dbSNP were obtained from the NCBI (http://www.ncbi.nlm.nih.gov/SNP). Repeats were downloaded from Repbase (https://www.girinst.org/repbase/). Differentially methylated regions were annotated using the GENCODE gene model GRCm38.p6 release M20 (https://www.gencodegenes.org/mouse/release_M20.html). Data supporting the findings of this study are available within the article and supplementary information (Supplementary Tables 16). No restrictions on data availability apply. Correspondence and requests for materials should be addressed to M.G.N. (Mihai.Netea@radboudumc.nl). Source data are provided with this paper.

Code availability

The raw sequencing data within this study were processed using established software packages with indicated parameters and published datasets as stated in detail in the Methods and Reporting Summary. The code scripts used to analyze these data will be shared upon reasonable request.

Change history

References

  1. Beccaloni, G. W. & Smith, V. S. Celebrations for Darwin downplay Wallace’s role. Nature 451, 1050 (2008).

    Article  CAS  Google Scholar 

  2. Cubas, P., Vincent, C. & Coen, E. An epigenetic mutation responsible for natural variation in floral symmetry. Nature 401, 157–161 (1999).

    Article  CAS  Google Scholar 

  3. Luna, E., Bruce, T. J. A., Roberts, M. R., Flors, V. & Ton, J. Next-generation systemic acquired resistance. Plant Physiol. 158, 844–853 (2012).

    Article  CAS  Google Scholar 

  4. Belicard, T., Jareosettasin, P. & Sarkies, P. The piRNA pathway responds to environmental signals to establish intergenerational adaptation to stress. BMC Biol. 16, 103 (2018).

    Article  Google Scholar 

  5. Ferguson-Smith, A. C. & Patti, M.-E. You are what your dad ate. Cell Metab. 13, 115–117 (2011).

    Article  CAS  Google Scholar 

  6. Morgan, H. D., Sutherland, H. G., Martin, D. I. & Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet. 23, 314–318 (1999).

    Article  CAS  Google Scholar 

  7. Dias, B. G. & Ressler, K. J. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat. Neurosci. 17, 89–96 (2014).

    Article  CAS  Google Scholar 

  8. Rowe, A. H. & Rowe, M. P. Physiological resistance of grasshopper mice (Onychomys spp.) to Arizona bark scorpion (Centruroides exilicauda) venom. Toxicon 52, 597–605 (2008).

    Article  CAS  Google Scholar 

  9. Domínguez-Andrés, J. et al. Inflammatory Ly6Chigh monocytes protect against candidiasis through IL-15-driven NK cell/neutrophil activation. Immunity 46, 1059–1072.e4 (2017).

    Article  Google Scholar 

  10. Saeed, S. et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science 345, 1251086 (2014).

    Article  Google Scholar 

  11. Klengel, T., Dias, B. G. & Ressler, K. J. Models of intergenerational and transgenerational transmission of risk for psychopathology in mice. Neuropsychopharmacology 41, 219–231 (2016).

    Article  Google Scholar 

  12. Ciarlo, E. et al. Trained immunity confers broad-spectrum protection against bacterial infections. J. Infect. Dis. 222, 1869–1881 (2020).

    Article  CAS  Google Scholar 

  13. Mitroulis, I. et al. Modulation of myelopoiesis progenitors is an integral component of trained immunity. Cell 172, 147–161.e12 (2018).

    Article  CAS  Google Scholar 

  14. Mebratu, Y. & Tesfaigzi, Y. How ERK1/2 activation controls cell proliferation and cell death: is subcellular localization the answer? Cell Cycle 8, 1168–1175 (2009).

    Article  CAS  Google Scholar 

  15. Zhu, F. G., Reich, C. F. & Pisetsky, D. S. The role of the macrophage scavenger receptor in immune stimulation by bacterial DNA and synthetic oligonucleotides. Immunology 103, 226–234 (2001).

    Article  CAS  Google Scholar 

  16. Stein, J. V. et al. APRIL modulates B and T cell immunity. J. Clin. Invest. 109, 1587–1598 (2002).

    Article  CAS  Google Scholar 

  17. Volkova, O. et al. Development and characterization of domain-specific monoclonal antibodies produced against human SLAMF9. Monoclon. Antib. Immunodiagn. Immunother. 33, 209–214 (2014).

    Article  CAS  Google Scholar 

  18. Kaludercic, N. et al. Monoamine oxidase B prompts mitochondrial and cardiac dysfunction in pressure overloaded hearts. Antioxid. Redox Signal. 20, 267–280 (2014).

    Article  CAS  Google Scholar 

  19. Manicone, A. M. & McGuire, J. K. Matrix metalloproteinases as modulators of inflammation. Semin. Cell Dev. Biol. 19, 34–41 (2008).

    Article  CAS  Google Scholar 

  20. Nohra, R. et al. RGMA and IL21R show association with experimental inflammation and multiple sclerosis. Genes Immun. 11, 279–293 (2010).

    Article  CAS  Google Scholar 

  21. Hedl, M., Zheng, S. & Abraham, C. The IL18RAP region disease polymorphism decreases IL-18RAP/IL-18R1/IL-1R1 expression and signaling through innate receptor-initiated pathways. J. Immunol. 192, 5924–5932 (2014).

    Article  CAS  Google Scholar 

  22. Klaver, E. J. et al. Trichuris suis soluble products induce Rab7b expression and limit TLR4 responses in human dendritic cells. Genes Immun. 16, 378–387 (2015).

    Article  CAS  Google Scholar 

  23. Hrašovec, S., Hauptman, N., Glavač, D., Jelenc, F. & Ravnik-Glavač, M. TMEM25 is a candidate biomarker methylated and down-regulated in colorectal cancer. Dis. Markers 34, 93–104 (2013).

    Article  Google Scholar 

  24. Cramer, S. D., Ferree, P. M., Lin, K., Milliner, D. S. & Holmes, R. P. The gene encoding hydroxypyruvate reductase (GRHPR) is mutated in patients with primary hyperoxaluria type II. Hum. Mol. Genet. 8, 2063–2069 (1999).

    Article  CAS  Google Scholar 

  25. Hong, J. Y. et al. Antibody to FcεRIα suppresses immunoglobulin E binding to high-affinity receptor I in allergic inflammation. Yonsei Med. J. 57, 1412–1419 (2016).

    Article  CAS  Google Scholar 

  26. Emin, M. et al. Increased internalization of complement inhibitor CD59 may contribute to endothelial inflammation in obstructive sleep apnea. Sci. Transl. Med. 8, 320ra1 (2016).

    Article  Google Scholar 

  27. Dinarello, C. A. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood 117, 3720–3732 (2011).

    Article  CAS  Google Scholar 

  28. Yu, H.-B. et al. NFATc2 mediates epigenetic modification of dendritic cell cytokine and chemokine responses to dectin-1 stimulation. Nucleic Acids Res. 43, 836–847 (2015).

    Article  CAS  Google Scholar 

  29. Qin, Q., Lee, S. H., Liang, R. & Kalejta, R. F. Insertion of myeloid-active elements into the human cytomegalovirus major immediate early promoter is not sufficient to drive its activation upon infection of undifferentiated myeloid cells. Virology 448, 125–132 (2014).

    Article  CAS  Google Scholar 

  30. Lee, S. H. et al. Runx3 inhibits IL-4 production in T cells via physical interaction with NFAT. Biochem. Biophys. Res. Commun. 381, 214–217 (2009).

    Article  CAS  Google Scholar 

  31. Morris, V. A., Cummings, C. L., Korb, B., Boaglio, S. & Oehler, V. G. Deregulated KLF4 expression in myeloid leukemias alters cell proliferation and differentiation through microRNA and gene targets. Mol. Cell. Biol. 36, 559–573 (2016).

    Article  CAS  Google Scholar 

  32. Siklenka, K. et al. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science 350, aab2006 (2015).

    Article  Google Scholar 

  33. Sun, W. et al. Cold-induced epigenetic programming of the sperm enhances brown adipose tissue activity in the offspring. Nat. Med. 24, 1372–1383 (2018).

    Article  CAS  Google Scholar 

  34. Das, A. et al. High-resolution mapping and dynamics of the transcriptome, transcription factors, and transcription co-factor networks in classically and alternatively activated macrophages. Front. Immunol. 9, 22 (2018).

    Article  Google Scholar 

  35. Pundhir, S. et al. Enhancer and transcription factor dynamics during myeloid differentiation reveal an early differentiation block in Cebpa null progenitors. Cell Rep. 23, 2744–2757 (2018).

    Article  CAS  Google Scholar 

  36. Eggert, H., Kurtz, J. & Diddens-de Buhr, M. F. Different effects of paternal trans-generational immune priming on survival and immunity in step and genetic offspring. Proc. Biol. Sci. 281, 20142089 (2014).

    Google Scholar 

  37. Hernández López, J., Schuehly, W., Crailsheim, K. & Riessberger-Gallé, U. Trans-generational immune priming in honeybees. Proc. Biol. Sci. 281, 20140454 (2014).

    Google Scholar 

  38. Tidbury, H. J., Pedersen, A. B. & Boots, M. Within and transgenerational immune priming in an insect to a DNA virus. Proc. Biol. Sci. 278, 871–876 (2011).

    Google Scholar 

  39. Yue, F. et al. Maternal transfer of immunity in scallop Chlamys farreri and its trans-generational immune protection to offspring against bacterial challenge. Dev. Comp. Immunol. 41, 569–577 (2013).

    Article  CAS  Google Scholar 

  40. Ramanan, D. et al. An immunologic mode of multigenerational transmission governs a gut Treg setpoint. Cell 181, 1276–1290.e13 (2020).

    Article  CAS  Google Scholar 

  41. Weber-Stadlbauer, U. et al. Transgenerational transmission and modification of pathological traits induced by prenatal immune activation. Mol. Psychiatry 22, 102–112 (2017).

    Article  CAS  Google Scholar 

  42. Gerbault, P. et al. Evolution of lactase persistence: an example of human niche construction. Philos. Trans. R. Soc. Lond. B Biol. Sci. 366, 863–877 (2011).

    Article  CAS  Google Scholar 

  43. Maekawa, T. et al. ATF7 mediates TNF-α-induced telomere shortening. Nucleic Acids Res. 46, 4487–4504 (2018).

    Article  CAS  Google Scholar 

  44. Norouzitallab, P., Baruah, K., Biswas, P., Vanrompay, D. & Bossier, P. Probing the phenomenon of trained immunity in invertebrates during a transgenerational study, using brine shrimp Artemia as a model system. Sci. Rep. 6, 21166 (2016).

    Article  CAS  Google Scholar 

  45. Kazachenka, A. et al. Identification, characterization, and heritability of murine metastable epialleles: implications for non-genetic inheritance. Cell 175, 1259–1271.e13 (2018).

    Article  CAS  Google Scholar 

  46. Oey, H., Isbel, L., Hickey, P., Ebaid, B. & Whitelaw, E. Genetic and epigenetic variation among inbred mouse littermates: identification of inter-individual differentially methylated regions. Epigenetics Chromatin 8, 54 (2015).

    Article  Google Scholar 

  47. Nankabirwa, V. et al. Child survival and BCG vaccination: a community based prospective cohort study in Uganda. BMC Public Health 15, 175 (2015).

    Article  Google Scholar 

  48. Kovacs, E. J. et al. Aging and innate immunity in the mouse: impact of intrinsic and extrinsic factors. Trends Immunol. 30, 319–324 (2009).

    Article  CAS  Google Scholar 

  49. Roger, T. et al. Macrophage migration inhibitory factor deficiency is associated with impaired killing of gram-negative bacteria by macrophages and increased susceptibility to Klebsiella pneumoniae sepsis. J. Infect. Dis. 207, 331–339 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

M.G.N. was supported by a European Research Council advanced grant (no. 833247) and a Spinoza grant of the Netherlands Organization for Scientific Research. E.J.G.-B. is funded by the Hellenic Institute for the Study of Sepsis. T.R. was supported by the Swiss National Science Foundation (no. 310030_173123) and grants from Fondation Carigest/Promex Stiftung für die Forschung and Fondation de Recherche en Biochimie. J.D.-A. is supported by the Netherlands Organization for Scientific Research (VENI grant no. 09150161910024). G.R. is funded by the Horizon 2020 Marie Skłodowska-Curie Action-European Sepsis Academy-Innovative Training Network (no. 676129). A.S. holds an Emmy Noether fellowship of the Deutsche Forschungsgemeinschaft (DFG) (no. SCHL2116/1-1). M.G.N., A.S. and J.L.S. are funded by the DFG under Germany’s Excellence Strategy EXC2151 390873048. J.W. and K.L. are funded by the DFG within the SFB 1309 (Chemical Biology of Epigenetic Modifications). M.B. is a member of the excellence cluster ImmunoSensation2. We thank K. Nordström for bioinformatics support.

Author information

Authors and Affiliations

Authors

Contributions

N.K., J.D.-A., B.C., G.G., G.R., E.C., D.L.R., T.R., K.L., K.K., K.H., M.B. and H.T. designed and performed the experiments and analyzed the data. N.K. performed the cytokine measurements. N.K., B.C. and E.C. performed the flow cytometry. B.C. processed the RNA-seq and ATAC-seq experiments, pathway analysis and performed the colony assessment under the supervision of A.S. J.D.-A. performed the cytokine measurements and trained the immunity experiments under the supervision of M.G.N. G.R. designed and performed the experiments with mice and the infection assays under the supervision of E.J.G.-B. T.R. designed and E.C. performed the listeriosis model. K.L. and K.K. isolated the mouse sperm and performed the DNA methylation analysis with G.G. J.W., J.W.M.v.d.M., L.A.B.J., M.B. and J.L.S. provided guidance and advice. J.W. supervised the sperm and DNA methylation analysis. T.R., E.J.G.-B., A.S. and M.G.N. conceived the study and oversaw the research program. J.D.-A. wrote the first draft of the manuscript with all authors contributing to the writing and providing feedback.

Corresponding author

Correspondence to Jorge Domínguez-Andrés.

Ethics declarations

Competing interests

E.J.G.-B. has received honoraria (paid to the University of Athens) from AbbVie, Abbott, Biotest, Brahms, InflaRx, the Medicines Company, MSD and XBiotech. He has received independent educational grants from AbbVie, Abbott, Astellas Pharma, Axis Shield, bioMérieux, InflaRx, the Medicines Company and XBiotech. M.G.N. is a scientific founder of TTxD.

Additional information

Peer review information Nature Immunology thanks Søren Paludan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Zoltan Fehervari 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 Fungal burden in F1-control and F1-exposed mice.

(a) Fungal burden assessed in F1-control and F1-exposed mice in liver and kidney 3 and 7 days after infection with C. albicans, n = 5 per group. (b) Myeloperoxidase activity assessed in F1-control and F1-exposed mice in liver and kidney 3 and 7 days after infection with C. albicans, n = 5 per group. Statistical significance was calculated by two-tailed Mann Whitney U test. P values are depicted on the figures; ns, not significant.

Source data

Extended Data Fig. 2 No protective effects are seen in the F3-exposed offspring.

Bacterial burden assessed in F3-control and F3-exposed mice in liver, kidney and spleen 3 days after i.v. infection with E. coli. n = 5 per group; Mann Whitney U test. P values are depicted on the figures.

Source data

Extended Data Fig. 3 Cell counts in blood after infection of F1 mice.

Leukocyte subpopulations in blood collected just before (day 0), 2 and 3 days post-infection with L. monocytogenes (2 ×104 CFU i.v.) in F1-control and F1-exposed mice (n = 8). Statistical significance was calculated by two-tailed Mann Whitney U test. P values are depicted on the figures; ns, not significant.

Source data

Extended Data Fig. 4 Responses in the female progeny. Trained parents: females; progeny analyzed: F1 females.

(a) Survival of F1-control and F1-exposed mice after infection with L. monocytogenes. Data are presented as a Kaplan-Maier plot with a log rank test used to compare susceptibility between the two groups, n = 16 per group. (b) Percent of initial weight of mice 48 h after infection, n = 16 per group. (c, d) Bacteria in blood collected 48 (c) and 72 h after infection (d), n = 16 per group. P values are depicted on the figures, Mann-Whitney U test, unless otherwise stated. (e) Leukocyte subpopulations in blood collected just before (day 0), 2 and 3 days post-infection with L. monocytogenes in F1-control and F1-exposed mice, n = 16 per group. Statistical significance was calculated by two-tailed Mann Whitney U test. P values are depicted on the figures; ns, not significant.

Source data

Extended Data Fig. 5 Responses in the male progeny. Trained parents: females; progeny analyzed: F1 males.

(a) Survival of F1-control and F1-exposed mice after infection with L. monocytogenes. Data are presented as a Kaplan-Maier plot with a log rank test used to compare susceptibility between the two groups. n = 6 for F1-control, 8 for F1-exposed. (b) Percent of initial weight of mice 48 h after infection, n = 6 for F1-control, 8 for F1-exposed (c, d) Bacteria in blood collected 48 (c) and 72 h after infection (d), n = 6 for F1-control, 8 for F1-exposed. P values are depicted on the figures, Mann-Whitney U test, unless otherwise stated. (e) Leukocyte subpopulations in blood collected just before (day 0), 2 and 3 days post-infection with L. monocytogenes in F1-control and F1-exposed mice, n = 6 for F1-control, 8 for F1-exposed. Statistical significance was calculated by two-tailed Mann Whitney U test. P values are depicted on the figures; ns, not significant.

Source data

Extended Data Fig. 6 Tlr1-/-, Tlr2-/-, and Tlr6-/- mice are fully trainable by zymosan and fully resistant to infection with L. monocytogenes.

Survival of wild type, Tlr1-/-, Tlr2-/-, and Tlr6-/- mice female mice trained with zymosan before i.v. challenge with 1.1 ×105 CFU L. monocytogenes. Data are presented as a Kaplan-Maier plot with a log rank test used to compare susceptibility between the two groups. Number of mice per group and p value are depicted in the figure.

Source data

Extended Data Fig. 7 Phenotyping strategy.

(a) Manual gating strategy to phenotype BM myeloid lineages and progenitors using flow cytometry. GMP, cMoP and Ly6chigh monocytes (Mono) were sorted for RNA-seq or ATAC-seq. (b) Quantification of cell populations in the progeny of F1-control or F1-exposed mice. F1-control, n = 17, F1-exposed n = 13, Statistical significance was calculated by two-tailed unpaired t-test; ns, not significant.

Source data

Extended Data Fig. 8 Infection induces epigenetic changes in bone marrow progenitors of the F1-control and F1-exposed offspring.

(a) General distribution of all identified peaks by ATAC-seq relative to the distance to the closest gene transcription start site (TSS). (b) Annotation of all identified peaks according to the genomic location. UTR, untranslated region. (c) Hierarchical clustering and heatmap of all differentially accessible (DA) ATAC-seq peaks of sorted GMPs from F1-control and F1-exposed offspring. (1460 opening, 1402 closing regions). (d) Heatmap and hierarchical clustering of the subfraction of DA ATAC-seq peaks located within gene promoter regions (176 opening, 177 closing regions). F1-control/F1-exposed, n = 13 per group. See Supplementary Table 6 for the full list of differentially accessible regions.

Extended Data Fig. 9 DNA methylation landscape of sperm.

(a) Methylation density plot of global methylation levels. (b) Genomic annotation of DMRs. (c) Methylation levels at repetitive elements. (d) KEGG pathways enriched for hypomethylated and hypermethylated DMRs. n = 10 per group.

Supplementary information

Source data

Source Data Fig. 1

Statistical source data (Excel) for Fig. 1.

Source Data Fig. 2

Statistical source data (Excel) for Fig. 2.

Source Data Fig. 3

Statistical source data (Excel) for Fig. 3.

Source Data Extended Data Fig. 1

Statistical source data (Excel) for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data (Excel) for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data (Excel) for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data (Excel) for Extended Data Fig. 4

Source Data Extended Data Fig. 5

Statistical source data (Excel) for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data (Excel) for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data (Excel) for Extended Data Fig. 7.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Katzmarski, N., Domínguez-Andrés, J., Cirovic, B. et al. Transmission of trained immunity and heterologous resistance to infections across generations. Nat Immunol 22, 1382–1390 (2021). https://doi.org/10.1038/s41590-021-01052-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01052-7

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology