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.

Intergenerational transfer of antibiotic-perturbed microbiota enhances colitis in susceptible mice

Abstract

Antibiotic exposure in children has been associated with the risk of inflammatory bowel disease (IBD). Antibiotic use in children or in their pregnant mother can affect how the intestinal microbiome develops, so we asked whether the transfer of an antibiotic-perturbed microbiota from mothers to their children could affect their risk of developing IBD. Here we demonstrate that germ-free adult pregnant mice inoculated with a gut microbial community shaped by antibiotic exposure transmitted their perturbed microbiota to their offspring with high fidelity. Without any direct or continued exposure to antibiotics, this dysbiotic microbiota in the offspring remained distinct from controls for at least 21 weeks. By using both IL-10-deficient and wild-type mothers, we showed that both inoculum and genotype shape microbiota populations in the offspring. Because IL10−/− mice are genetically susceptible to colitis, we could assess the risk due to maternal transmission of an antibiotic-perturbed microbiota. We found that the IL10−/− offspring that had received the perturbed gut microbiota developed markedly increased colitis. Taken together, our findings indicate that antibiotic exposure shaping the maternal gut microbiota has effects that extend to the offspring, with both ecological and long-term disease consequences.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Microbiome transfer to pregnant germ-free mice colonized 23 dams and 112 pups.
Fig. 2: Intergenerational microbiota transfer efficiency and stability over time.
Fig. 3: STAT microbiota in IL10–/– mouse increases colonic inflammation.

References

  1. Jernberg, C., Löfmark, S., Edlund, C. & Jansson, J. K. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J. 1, 56–66 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Cox, L. M. et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158, 705–721 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Azad, M. B., Bridgman, S. L., Becker, A. B. & Kozyrskyj, A. L. Infant antibiotic exposure and the development of childhood overweight and central adiposity. Int. J. Obesity 38, 1290–1298 (2014).

    Article  CAS  Google Scholar 

  4. Arrieta, M.-C. et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 7, 307ra152 (2015).

    Article  PubMed  Google Scholar 

  5. Hviid, A., Svanstrom, H. & Frisch, M. Antibiotic use and inflammatory bowel diseases in childhood. Gut 60, 49–54 (2011).

    Article  PubMed  Google Scholar 

  6. Shaw, S. Y., Blanchard, J. F. & Bernstein, C. N. Association between the use of antibiotics and new diagnoses of Crohn’s disease and ulcerative colitis. Am. J. Gastroenterol. 106, 2133–2142 (2011).

    Article  PubMed  Google Scholar 

  7. Ng, S. C. et al. Geographical variability and environmental risk factors in inflammatory bowel disease. Gut 62, 630–649 (2013).

    Article  PubMed  Google Scholar 

  8. Kronman, M. P., Zaoutis, T. E., Haynes, K., Feng, R. & Coffin, S. E. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics 130, e794–e803 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Van Boeckel, T. P. et al. Global antibiotic consumption 2000 to 2010: an analysis of national pharmaceutical sales data. Lancet Infect. Dis. 14, 742–750 (2014).

    Article  PubMed  Google Scholar 

  10. Hicks, L. A. et al. US outpatient antibiotic prescribing variation according to geography, patient population, and provider specialty in 2011. Clin. Infect. Dis. 60, 1308–1316 (2015).

    PubMed  Google Scholar 

  11. Stokholm, J. et al. Prevalence and predictors of antibiotic administration during pregnancy and birth. PLoS ONE 8, e82932 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Andrade, S. E. et al. Prescription drug use in pregnancy. Am. J. Obstet. Gynecol. 191, 398–407 (2004).

    Article  PubMed  Google Scholar 

  13. Lacroix, I. et al. Prescription of drugs during pregnancy: a study using EFEMERIS, the new French database. Eur. J. Clin. Pharmacol. 65, 839–846 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Petersen, I., Gilbert, R., Evans, S., Ridolfi, A. & Nazareth, I. Oral antibiotic prescribing during pregnancy in primary care: UK population-based study. J. Antimicrob. Chemoth. 65, 2238–2246 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Metsälä, J. et al. Mother’s and offspring’s use of antibiotics and infant allergy to cow’s milk. Epidemiology 24, 303–309 (2013).

    Article  PubMed  Google Scholar 

  16. Moeller, A. H. et al. Cospeciation of gut microbiota. Science 353, 380–382 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Goodrich, J. K. et al. Human genetics shape the gut microbiome. Cell 159, 789–799 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sonnenburg, E. D. et al. Diet-induced extinctions in the gut microbiota compound over generations. Nature 529, 212–215 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kühn, R., Löhler, J., Rennick, D., Rajewsky, K. & Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75, 263–274 (1993).

    Article  PubMed  Google Scholar 

  20. Sellon, R. K. et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect. Immun. 66, 5224–5231 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Krause, P. et al. IL-10-producing intestinal macrophages prevent excessive antibacterial innate immunity by limiting IL-23 synthesis. Nat. Commun. 6, 7055 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sartor, R. B. & Wu, G. D. Roles for intestinal bacteria, viruses, and fungi in pathogenesis of inflammatory bowel diseases and therapeutic approaches. Gastroenterology 152, 327–339 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Jones-Hall, Y. L. & Grisham, M. B. Immunopathological characterization of selected mouse models of inflammatory bowel disease: comparison to human disease. Pathophysiology 21, 267–288 (2014).

    Article  PubMed  Google Scholar 

  24. Manichanh, C. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 55, 205–211 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Livanos, A. E. et al. Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat. Microbiol. 1, 16140 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Pedersen, H. K. et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535, 376–381 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Orešič, M. et al. Dysregulation of lipid and amino acid metabolism precedes islet autoimmunity in children who later progress to type 1 diabetes. J. Exp. Med. 205, 2975–2984 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Nagata, C. et al. Branched-chain amino acid intake and the risk of diabetes in a Japanese community: the Takayama study. Am. J. Epidemiol. 178, 1226–1232 (2013).

    Article  PubMed  Google Scholar 

  29. Lynch, C. J. & Adams, S. H. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat. Rev. Endocrinol. 10, 723–736 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Attene-Ramos, M. S. et al. DNA damage and toxicogenomic analyses of hydrogen sulfide in human intestinal epithelial FHs 74 Int cells. Environ. Mol. Mutagen. 51, 304–314 (2010).

    CAS  PubMed  Google Scholar 

  31. Van Passel, M. W. J. et al. The genome of Akkermansia muciniphila, a dedicated intestinal mucin degrader, and its use in exploring intestinal metagenomes. PLoS ONE 6, e16876 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Cohen, S. L., Moore, A. M. & Ward, W. E. Interleukin-10 knockout mouse: a model for studying bone metabolism during intestinal inflammation. Inflamm. Bowel Dis. 10, 557–563 (2004).

    Article  PubMed  Google Scholar 

  33. Mahana, D. et al. Antibiotic perturbation of the murine gut microbiome enhances the adiposity, insulin resistance, and liver disease associated with high-fat diet. Genome Med. 8, 48 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Spencer, D. M., Veldman, G. M., Banerjee, S., Willis, J. & Levine, A. D. Distinct inflammatory mechanisms mediate early versus late colitis in mice. Gastroenterology 122, 94–105 (2002).

    Article  PubMed  Google Scholar 

  35. Berg, D. J. et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4+ Th1-like responses. J. Clin. Invest. 98, 1010–1020 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lehmann, F. S., Burri, E. & Beglinger, C. The role and utility of faecal markers in inflammatory bowel disease. Ther. Adv. Gastroenter. 8, 23–36 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Chassaing, B. et al. Fecal lipocalin 2, a sensitive and broadly dynamic non-invasive biomarker for intestinal inflammation. PLoS ONE 7, e44328 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hansen, J. J., Holt, L. & Sartor, R. B. Gene expression patterns in experimental colitis in IL-10-deficient mice. Inflamm. Bowel Dis. 15, 890–899 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Miyoshi, J. et al. Peripartum antibiotics promote gut dysbiosis, loss of immune tolerance, and inflammatory bowel disease in genetically prone offspring. Cell Rep. 20, 491–504 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Metsälä, J. et al. Prenatal and post-natal exposure to antibiotics and risk of asthma in childhood. Clin. Exp. Allergy 45, 137–145 (2015).

    Article  PubMed  Google Scholar 

  42. Gensollen, T., Iyer, S. S., Kasper, D. L. & Blumberg, R. S. How colonization by microbiota in early life shapes the immune system. Science 352, 539–544 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gomez de Aguero, M. et al. The maternal microbiota drives early postnatal innate immune development. Science 351, 1296–1302 (2016).

    Article  PubMed  Google Scholar 

  44. Bokulich, N. A. et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 8, 343ra82 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Asnicar, F. et al. Studying vertical microbiome transmission from mothers to infants by strain-level metagenomic profiling. mSystems 2, e00164-16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Nayfach, S., Rodriguez-Mueller, B., Garud, N. & Pollard, K. S. An integrated metagenomics pipeline for strain profiling reveals novel patterns of bacterial transmission and biogeography. Genome Res. 26, 1612–1625 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Palm, N. W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Pascal, V. et al. A microbial signature for Crohn’s disease. Gut 66, 813–822 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Halfvarson, J. et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol. 2, 17004 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Gevers, D. et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 15, 382–392 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rogers, A. B. & Houghton, J. in Inflammation and Cancer: Methods in Molecular Biology Vol. 511 (ed. Kozlov, S. V.) 267–295 (Humana, New York, 2009).

  52. Cox, D. & Snell, E. J. Analysis of Binary Data (Chapman and Hall/CRC, London, 1989).

    Google Scholar 

  53. Caporaso, J. G. et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl Acad. Sci. USA 108, 4516–4522 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Aronesty, E. Comparison of sequencing utility programs. Open Bioinformatics J. 7, 1–8 (2013).

    Article  Google Scholar 

  56. Edgar, R. C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. McDonald, D. et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J. 6, 610–618 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Abubucker, S. et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol. 8, 1002358 (2012).

    Article  Google Scholar 

  60. Bittinger, K. qiimer: Work with QIIME Output Files in R. R package v.0.9.4 (CRAN, 2015); https://CRAN.R-project.org/package=qiimer

  61. McDonald, D. et al. The biological observation matrix (BIOM) format or: how I learned to stop worrying and love the ome-ome. GigaScience 1, 1–6 (2012)

  62. McMurdie, P. J. & Holmes, S. Phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Dixon, P. VEGAN, a package of R functions for community ecology. J. Veg. Sci. 14, 927–930 (2003).

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank M. Bower and the National Gnotobiotic Rodent Resource Center, University of North Carolina, Chapel Hill, for supplying mice, the NYUMC Genome Technology Center for help with sequencing (partially supported by a Cancer Center Support grant, P30CA016087, at the Laura and Isaac Perlmutter Cancer Center) and the NYUMC Histology Core for assistance preparing samples for histology. These studies were supported by NIH grants DK090989, OD010995 and DK034987 and the Crohn’s and Colitis Foundation of America, by the Ziff Family, Knapp Family and C&D funds, the Judith & Stewart Colton Center for Autoimmunity, and the Diane Belfer Program for Human Microbial Ecology.

Author information

Authors and Affiliations

Authors

Contributions

A.F.S., R.B.S. and M.J.B. designed experiments and interpreted data. A.F.S. performed experiments and participated in analysis of the data. Y.A. and V.E.R. contributed to interpretation of immunological data. Y.A. and M.H. performed protein expression assays. T.B. advised on microbiome analytical methods and performed data analyses. L.B. performed odds ratio calculations and other statistical tests. S.R., T.W. and D.K. contributed to microbiota stability analysis. A.B.R. performed histological analyses. L.M.C. and R.B.S. provided essential reagents and procedural advice. A.F.S. and M.J.B. were responsible for writing the manuscript, which was reviewed and edited by all authors.

Corresponding author

Correspondence to Martin J. Blaser.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

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

Electronic supplementary material

Supplementary Information

Supplementary Figures, Tables, Notes, Discussion and References.

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schulfer, A.F., Battaglia, T., Alvarez, Y. et al. Intergenerational transfer of antibiotic-perturbed microbiota enhances colitis in susceptible mice. Nat Microbiol 3, 234–242 (2018). https://doi.org/10.1038/s41564-017-0075-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-017-0075-5

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