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:

The maternal microbiome modulates fetal neurodevelopment in mice

Abstract

‘Dysbiosis’ of the maternal gut microbiome, in response to challenges such as infection1, altered diet2 and stress3 during pregnancy, has been increasingly associated with abnormalities in brain function and behaviour of the offspring4. However, it is unclear whether the maternal gut microbiome influences neurodevelopment during critical prenatal periods and in the absence of environmental challenges. Here we investigate how depletion and selective reconstitution of the maternal gut microbiome influences fetal neurodevelopment in mice. Embryos from antibiotic-treated and germ-free dams exhibited reduced brain expression of genes related to axonogenesis, deficient thalamocortical axons and impaired outgrowth of thalamic axons in response to cell-extrinsic factors. Gnotobiotic colonization of microbiome-depleted dams with a limited consortium of bacteria prevented abnormalities in fetal brain gene expression and thalamocortical axonogenesis. Metabolomic profiling revealed that the maternal microbiome regulates numerous small molecules in the maternal serum and the brains of fetal offspring. Select microbiota-dependent metabolites promoted axon outgrowth from fetal thalamic explants. Moreover, maternal supplementation with these metabolites abrogated deficiencies in fetal thalamocortical axons. Manipulation of the maternal microbiome and microbial metabolites during pregnancy yielded adult offspring with altered tactile sensitivity in two aversive somatosensory behavioural tasks, but no overt differences in many other sensorimotor behaviours. Together, our findings show that the maternal gut microbiome promotes fetal thalamocortical axonogenesis, probably through signalling by microbially modulated metabolites to neurons in the developing brain.

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: Depletion of the maternal microbiome impairs fetal thalamocortical axonogenesis.
Fig. 2: Colonization of the maternal microbiota prevents neurodevelopmental abnormalities induced by microbiome depletion.
Fig. 3: The maternal microbiota modulates maternal serum and fetal brain metabolites during pregnancy.
Fig. 4: The maternal microbiota modulates metabolites that promote fetal thalamocortical axonogenesis.

Similar content being viewed by others

Data availability

All data generated and analysed during this study are included in this published article and its Supplementary Information files. The 16S rRNA gene sequencing data that support the findings have also been deposited to the Qiita database (https://qiita.ucsd.edu/) with study IDs 13099, 13106 and 13107. Transcriptomic data that support the findings of this study have also been deposited to the Gene Expression Omnibus (GEO) repository with accession number GSE147183Source data are provided with this paper.

References

  1. Kim, S. et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 549, 528–532 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  2. Buffington, S. A. et al. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell 165, 1762–1775 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Jašarević, E. et al. The maternal vaginal microbiome partially mediates the effects of prenatal stress on offspring gut and hypothalamus. Nat. Neurosci. 21, 1061–1071 (2018).

    Article  PubMed  Google Scholar 

  4. Vuong, H. E., Yano, J. M., Fung, T. C. & Hsiao, E. Y. The microbiome and host behavior. Annu. Rev. Neurosci. 40, 21–49 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ferretti, P. et al. Mother-to-infant microbial transmission from different body sites shapes the developing infant gut microbiome. Cell Host Microbe 24, 133–145.e135 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nakashiba, T. et al. Netrin-G1: a novel glycosyl phosphatidylinositol-linked mammalian netrin that is functionally divergent from classical netrins. J. Neurosci. 20, 6540–6550 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rennie, S., Lotto, R. B. & Price, D. J. Growth-promoting interactions between the murine neocortex and thalamus in organotypic co-cultures. Neuroscience 61, 547–564 (1994).

    Article  CAS  PubMed  Google Scholar 

  8. Mitsogiannis, M. D., Little, G. E. & Mitchell, K. J. Semaphorin–plexin signaling influences early ventral telencephalic development and thalamocortical axon guidance. Neural Dev. 12, 6 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Braisted, J. E., Ringstedt, T. & O’Leary, D. D. Slits are chemorepellents endogenous to hypothalamus and steer thalamocortical axons into ventral telencephalon. Cereb. Cortex 19, i144–i151 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Song, H. et al. Conversion of neuronal growth cone responses from repulsion to attraction by cyclic nucleotides. Science 281, 1515–1518 (1998).

    Article  ADS  CAS  PubMed  Google Scholar 

  11. Stevens, A. & Jacobs, J. R. Integrins regulate responsiveness to slit repellent signals. J. Neurosci. 22, 4448–4455 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dontchev, V. D. & Letourneau, P. C. Nerve growth factor and semaphorin 3A signaling pathways interact in regulating sensory neuronal growth cone motility. J. Neurosci. 22, 6659–6669 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Enriquez-Barreto, L., Palazzetti, C., Brennaman, L. H., Maness, P. F. & Fairén, A. Neural cell adhesion molecule, NCAM, regulates thalamocortical axon pathfinding and the organization of the cortical somatosensory representation in mouse. Front. Mol. Neurosci. 5, 76 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chaplan, S. R., Bach, F. W., Pogrel, J. W., Chung, J. M. & Yaksh, T. L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 53, 55–63 (1994).

    Article  CAS  PubMed  Google Scholar 

  15. Bouet, V. et al. The adhesive removal test: a sensitive method to assess sensorimotor deficits in mice. Nat. Protocols 4, 1560–1564 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Eddy, N. B. & Leimbach, D. Synthetic analgesics. II. Dithienylbutenyl- and dithienylbutylamines. J. Pharmacol. Exp. Ther. 107, 385–393 (1953).

    CAS  PubMed  Google Scholar 

  17. Valsamis, B. & Schmid, S. Habituation and prepulse inhibition of acoustic startle in rodents. J. Vis. Exp. 55, e3446 (2011).

    Google Scholar 

  18. Fox, M. W. The visual cliff test for the study of visual depth perception in the mouse. Anim. Behav. 13, 232–233 (1965).

    Article  CAS  PubMed  Google Scholar 

  19. Wu, H. P., Ioffe, J. C., Iverson, M. M., Boon, J. M. & Dyck, R. H. Novel, whisker-dependent texture discrimination task for mice. Behav. Brain Res. 237, 238–242 (2013).

    Article  PubMed  Google Scholar 

  20. Deacon, R. M. Measuring motor coordination in mice. J. Vis. Exp. 75, e2609 (2013).

    Article  Google Scholar 

  21. Fujisaka, S. et al. Diet, genetics, and the gut microbiome drive dynamic changes in plasma metabolites. Cell Rep. 22, 3072–3086 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Saunders, N. R., Liddelow, S. A. & Dziegielewska, K. M. Barrier mechanisms in the developing brain. Front. Pharmacol. 3, 46 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Monk, C., Georgieff, M. K. & Osterholm, E. A. Research review: maternal prenatal distress and poor nutrition - mutually influencing risk factors affecting infant neurocognitive development. J. Child Psychol. Psychiatry 54, 115–130 (2013).

    Article  PubMed  Google Scholar 

  24. Olson, C. A. et al. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell 174, 497 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Matsumoto, M. et al. Cerebral low-molecular metabolites influenced by intestinal microbiota: a pilot study. Front. Syst. Neurosci. 7, 9 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Molnár, Z., Garel, S., López-Bendito, G., Maness, P. & Price, D. J. Mechanisms controlling the guidance of thalamocortical axons through the embryonic forebrain. Eur. J. Neurosci. 35, 1573–1585 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Vernocchi, P., Del Chierico, F. & Putignani, L. Gut microbiota profiling: metabolomics based approach to unravel compounds affecting human health. Front. Microbiol. 7, 1144 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Lu, J. et al. Microbiota influence the development of the brain and behaviors in C57BL/6J mice. PLoS One 13, e0201829 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Ong, I. M. et al. Gut microbiome populations are associated with structure-specific changes in white matter architecture. Transl. Psychiatry 8, 6 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Indrio, F. et al. Epigenetic matters: the link between early nutrition, microbiome, and long-term health development. Front Pediatr. 5, 178 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Keunen, K., van Elburg, R. M., van Bel, F. & Benders, M. J. Impact of nutrition on brain development and its neuroprotective implications following preterm birth. Pediatr. Res. 77, 148–155 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Shin Yim, Y. et al. Reversing behavioural abnormalities in mice exposed to maternal inflammation. Nature 549, 482–487 (2017).

    Article  ADS  PubMed  Google Scholar 

  33. Gehrig, J. L. et al. Effects of microbiota-directed foods in gnotobiotic animals and undernourished children. Science 365, eaau4732 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Hamad, A. F., Alessi-Severini, S., Mahmud, S. M., Brownell, M. & Kuo, I. F. Prenatal antibiotics exposure and the risk of autism spectrum disorders: a population-based cohort study. PLoS One 14, e0221921 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Atladóttir, H. O., Henriksen, T. B., Schendel, D. E. & Parner, E. T. Autism after infection, febrile episodes, and antibiotic use during pregnancy: an exploratory study. Pediatrics 130, e1447–e1454 (2012).

    Article  PubMed  Google Scholar 

  36. Reikvam, D. H. et al. Depletion of murine intestinal microbiota: effects on gut mucosa and epithelial gene expression. PLoS One 6, e17996 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. McVey Neufeld, K. A., Perez-Burgos, A., Mao, Y. K., Bienenstock, J. & Kunze, W. A. The gut microbiome restores intrinsic and extrinsic nerve function in germ-free mice accompanied by changes in calbindin. Neurogastroenterol. Motil. 27, 627–636 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Yano, J. M. et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161, 264–276 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pertea, M., Kim, D., Pertea, G. M., Leek, J. T. & Salzberg, S. L. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protocols 11, 1650–1667 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protocols 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  43. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Koh, A. et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell 175, 947–961 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Mishima, E. et al. Evaluation of the impact of gut microbiota on uremic solute accumulation by a CE-TOFMS-based metabolomics approach. Kidney Int. 92, 634–645 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Soga, T. et al. Differential metabolomics reveals ophthalmic acid as an oxidative stress biomarker indicating hepatic glutathione consumption. J. Biol. Chem. 281, 16768–16776 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Soga, T. et al. Quantitative metabolome analysis using capillary electrophoresis mass spectrometry. J. Proteome Res. 2, 488–494 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Kärkkäinen, O. et al. Whole grain intake associated molecule 5-aminovaleric acid betaine decreases β-oxidation of fatty acids in mouse cardiomyocytes. Sci. Rep. 8, 13036 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  50. Fothergill, J. C. & Guest, J. R. Catabolism of l-lysine by Pseudomonas aeruginosa. J. Gen. Microbiol. 99, 139–155 (1977).

    Article  CAS  PubMed  Google Scholar 

  51. Diehl, K. H. et al. A good practice guide to the administration of substances and removal of blood, including routes and volumes. J. Appl. Toxicol. 21, 15–23 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Erny, D. et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 18, 965–977 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Deuis, J. R., Dvorakova, L. S. & Vetter, I. Methods used to evaluate pain behaviors in rodents. Front. Mol. Neurosci. 10, 284 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Perry, W., Minassian, A., Lopez, B., Maron, L. & Lincoln, A. Sensorimotor gating deficits in adults with autism. Biol. Psychiatry 61, 482–486 (2007).

    Article  PubMed  Google Scholar 

  55. Barman, M. et al. Enteric salmonellosis disrupts the microbial ecology of the murine gastrointestinal tract. Infect. Immun. 76, 907–915 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. 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 (Suppl. 1), 4516–4522 (2011).

    Article  ADS  CAS  PubMed  Google Scholar 

  57. Amir, A. et al. Deblur rapidly resolves single-nucleotide community sequence patterns. mSystems 2, e00191-16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  60. Breiman, L. Random forests. Mach. Learn. 45, 5–32 (2001).

    Article  MATH  Google Scholar 

Download references

Acknowledgements

We thank members of the Hsiao lab for their critical review of the manuscript; T. Su for RNA sequencing advice; A. Oyler-Yaniv, J. Oyler-Yaniv and R. Wollman for assistance with initial light-sheet image acquisition; A. Rajbhandari and Irina Zhuravka of the UCLA Behavioral Testing Core for behavioural assay training; S. White for sharing ultrasonic vocalization equipment; and A. Collazo of the Caltech Beckman Institute Biological Imaging Facility for assistance with light-sheet image acquisition and analysis. Support for this research was provided by the Packard Fellowship in Science and Engineering and Klingenstein-Simons Award to E.Y.H.; UPLIFT: UCLA Postdocs’ Longitudinal Investment in Faculty Award (# K12 GM106996) and NICHD Pathway to Independence Award (#K99 HD101680) to H.E.V.; the Ruth L. Kirschstein National Research Service Awards (#F31 HD101270 to G.N.P. and #F30 DE025172 to D.W.W.), and the NSF Graduate Research Fellowship to E.J.L.C. E.Y.H. is a New York Stem Cell Foundation – Robertson Investigator. This research was supported in part by the New York Stem Cell Foundation.

Author information

Authors and Affiliations

Authors

Contributions

H.E.V. led and performed all experiments; G.N.P. assisted with sample collection and data analysis for imaging, 16S rRNA gene sequencing and metabolomic experiments; D.W.W. assisted with CLARITY, microcomputed tomography and imaging experiments; E.J.L.C., E.L.S., A.Q., M.K. and C.J.W. assisted with axon outgrowth, behavioural, immunofluorescence staining and/or imaging experiments; T.R. generated gnotobiotic mice; E.Y.H. supervised the study; and H.E.V., G.N.P. and E.Y.H. wrote the manuscript.

Corresponding author

Correspondence to Helen E. Vuong.

Ethics declarations

Competing interests

Findings regarding the manipulation of the maternal microbiome to influence fetal development and sensory behavior reported in the manuscript are the subject of provisional patent application US 62/844,503, owned by UCLA. The authors declare no competing interests.

Additional information

Peer review information Nature thanks Tianyi Mao and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Network analysis and qPCR validation of fetal brain RNA-seq data.

a, Gene ontology analysis of differentially expressed genes from E14.5 brains from ABX versus SPF dams (one-tailed Fisher exact, q = 0.0125, 0.017, 0.017, 0.0383, 0.0745, 0.0745, 0.0745, 0.1002, n = 3 dams). b, Quantitative RT–PCR for Ntng1 and LRRC4C expression in E14.5 brains from offspring of SPF, ABX or GF dams (two-way ANOVA+Tukey’s, n = 9, 15, 10 dams). c, Protein interaction network of genes downregulated in E14.5 brains from offspring of ABX vs. SPF dams (Benjamini-Hochberg, q < 0.05, n = 3 dams). d, Protein interaction networks of genes upregulated in E14.5 brains from offspring of ABX vs. SPF dams (Benjamini-Hochberg, q < 0.05, n = 3 dams). Mean ± SEM. *P < 0.05, n.s. = not statistically significant.

Source Data

Extended Data Fig. 2 Netrin-G1a thalamocortical axons in E14.5 brains of offspring from gnotobiotic dams.

a, Reference diagrams of coronal rostral to caudal E14.5 brain sections. be, Netrin-G1a in four independent E14.5 brains from offspring of SPF (b), ABX (c), GF (d), and Sp (e) dams. Scale = 500 μm. Yellow lines = matched control ROI. f, Netrin-G1a fluorescence per control ROI in E14.5 brain sections of offspring from SPF, ABX, GF and Sp dams. SPF, ABX, GF and Sp data are as presented in Fig. 1c and 2d. (Two-way ANOVA+Tukey’s, n = 5 dams). g, Total brain area E14.5 brain section of offspring from SPF, ABX, GF and Sp dams. (Two-way ANOVA+Tukey’s, n = 5 dams). h, Netrin-G1a per matched control ROI, normalized by total brain area in E14.5 brain sections of offspring from SPF, ABX, GF and Sp dams. (Two-way ANOVA+Tukey’s, n = 5 dams). i, Area of Netrin-G1a in E14.5 brain sections of SPF, ABX, GF and Sp dams. (Two-way ANOVA+Tukey’s, n = 5 dams). j, Area of Netrin-G1a+ staining normalized by total brain area in E14.5 brain sections of offspring from SPF, ABX, GF and Sp dams. (Two-way ANOVA+ Tukey’s, n = 5 dams). k, Netrin-G1a fluorescence in area of Netrin-G1a+ staining of E14.5 brains from SPF, ABX, GF, and Sp dams. (Two-way ANOVA+Tukey’s, n = 5 dams). l, DAPI per matched control ROI, normalized by total brain area in E14.5 brain sections from SPF, ABX, GF and Sp dams. (Two-way ANOVA+Tukey’s, n = 5, 7, 4, 5 dams). m, DAPI+ cell per matched control ROI, normalized by total brain area in E14.5 brain sections from SPF, ABX, GF and Sp dams. (Two-way ANOVA+ Tukey’s, n = 5, 7, 4, 5 dams). Mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, n.s. = not statistically significant.

Source Data

Extended Data Fig. 3 Whole brain volume and L1+ thalamocortical axons in E14.5 brains of offspring from gnotobiotic dams.

a, Micro-computed tomography of E14.5 fetal brain. Scale bar = 2 mm. b, Whole fetal brain volume of E14.5 offspring from SPF, ABX, GF and Sp dams. (One-way ANOVA+Tukey’s, n = 10, 5, 8, 5 dams). c, Volume of Netrin-G1a axons, normalized to E14.5 SPF, ABX, GF, and Sp whole brain volume. (SPF v ABX: two-tailed Mann–Whitney, SPF v GF, GF v Sp, and ABX v Sp: One-way ANOVA+Tukey’s, n = 5 offspring from different dams). d, Length of Netrin-G1a axons, normalized to the cubic root of E14.5 SPF, ABX, GF, and Sp whole brain volume. (SPF v ABX and SPF v GF: One-way ANOVA+Tukey’s, Sp v ABX and Sp v GF: two-tailed Mann–Whitney, n = 5 offspring from different dams). e, Distance from rostral tip of Netrin-G1a axon to cortex, normalized to the cubic root of E14.5 SPF, ABX, GF, and Sp whole brain volume. (SPF v ABX and Sp v ABX: One-way ANOVA+Tukey’s, SPF v GF and Sp v GF: two-tailed Mann–Whitney, n = 5 offspring from different dams). f, Circumference of Netrin-G1a axonal bundle at the internal capsule (IC), normalized to the cubic root of E14.5 SPF, ABX, GF, and Sp whole brain volume. (One-way ANOVA+Tukey‘s, n = 5 offspring from different dams). g, Netrin-G1a (magenta) and L1 (cyan) in E14.5 SPF, ABX, GF and Sp brain sections. Scale = 500 μm. Yellow lines = matched control ROI. h, L1 per matched control ROI, normalized by total brain area in E14.5 SPF, ABX, GF and Sp brain sections. (Two-way ANOVA+Tukey’s, n = 5 dams). i, L1 per matched control ROI in E14.5 brain sections of offspring from SPF, ABX, GF and Sp dams. SPF, ABX, GF, and Sp data are as in Fig. 1d and 2e. (Two-way ANOVA+Tukey’s, n = 5 dams). Mean ± SEM, *P < 0.05, **P < 0.01, ****P < 0.0001, n.s. = not statistically significant.

Source Data

Extended Data Fig. 4 Thalamic explant monocultures and co-cultures with striatal and hypothalamic explants.

a, Axon outgrowth from monoculture of thalamic explants (Th) from E14.5 offspring of SPF, ABX, GF and Sp dams. Scale = 250 μm. In Th monocultures: b, axon number, and c, axon length (One-way ANOVA+Tukey’s, n = 26, 20, 21, 10 explants). d, Axon outgrowth from (i) SPF Th+SPF hypothalamic explant (Hy), (ii) ABX Th+ABX Hy, (iii) SPF Th+ABX Hy, (iv) ABX Th+SPF Hy. Scale = 250 μm. Proximal to Hy: e, axon number /200 μm2 surface area, normalized to Th monoculture, and f, axon length, normalized to Th monoculture. (One-way ANOVA+Tukey’s, n = 14, 20, 9, 10 explants). g, Axon outgrowth from GF Th+GF St. Scale bar = 250 μm. Proximal to St: h, axon number/200 μm2 surface area, normalized to Th monoculture, and i, axon length, normalized to Th monoculture. (One-way ANOVA+Tukey’s, n = 16 GF explants). j, Axon outgrowth from GF Th+GF Hy. Scale bar = 250 μm. Proximal to Hy: k, axon number/200 μm2 surface area, normalized to Th monoculture, and l, axon length, normalized to Th monoculture. (One-way ANOVA+Tukey’s, n = 14 GF explants). m, Axon outgrowth from (i) SPF Th+SPF St, (ii) ABX Th+ABX St, (iii) Sp Th+Sp St, (iv) Sp Th+ABX St. Scale = 250 μm. Proximal to St: n, axon number/200 μm2 surface area, normalized to Th monoculture, and o, axon length, normalized to Th monoculture. (One-way ANOVA+Tukey’s, n = 14, 20, 14, 6 explants.) p, Axon outgrowth from (i) SPF Th+SPF Hy, (ii) ABX Th+ABX Hy, (iii) Sp Th+Sp Hy, (iv) Sp Th+ABX Hy. Arrows = sparse short axons. Scale = 250 μm. Proximal to Hy: q, axon number/200 μm perimeter, normalized to Th monoculture, and r, axon length, normalized to Th monoculture. (One-way ANOVA+Tukey’s, n = 14, 20, 14, 6 explants). SPF and ABX data in g-i and m-o are in Fig. 1k, 1l. Mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, n.s. = not statistically significant.

Source Data

Extended Data Fig. 5 Effects of maternal antibiotic treatment and gestational conventionalization on the faecal microbiota.

a, Bacterial load of the maternal faecal microbiota in response to antibiotic (ANVM = ampicillin, neomycin, vancomycin, metronidazole) treatment and conventionalization with SPF microbiota. Yellow line = Ct of germ-free control. (n = 5 cages). b, Principal coordinate analysis of 16S rRNA gene sequencing data (weighted Unifrac distances) for maternal faecal microbiota before antibiotic treatment (pre-ABX), on Day 2, 4, or 6 after ABX treatment, and on Day 0 (E14.5), 4 (E18.5), 6 (P2) and 8 (P4) after exposure to SPF bedding. (n = 5, 5, 5, 5, 3 cages). c, Class-level taxonomic diversity of maternal faecal microbiota pre-ABX and on Day 0 (E14.5), 4 (E18.5), 6 (P2) and 8 (P4) after exposure to SPF bedding. (n = 5, 5, 5, 5, 3 cages).

Source Data

Extended Data Fig. 6 Effects of the maternal microbiome on behaviours.

a, Maternal Sp colonization and SPF conventionalization. b, Force filament to induce 50% paw withdrawal (One-way ANOVA+Tukey’s, n = 38, 24, 45, 25 offspring). c, Latency to contact the adhesive, and d, latency to remove adhesive after first contact (One-way ANOVA+Tukey’s, n = 35, 19, 45, 38 offspring). e, f, Pairwise data for latency to contact and remove adhesive. (Two-way ANOVA+Sidak’s, n = 6, 6, 7, 7 dams). g, Force filament to induce 50% paw withdrawal by sex, per litter or individual (Two-way ANOVA+Tukey’s, n = 5, 7, 7, 7 dams). h, Latency to contact and i, remove adhesive after first contact (Two-way ANOVA+Tukey’s, n = 6, 6, 7, 11 dams). j, Latency to withdraw from hot plate (One-way ANOVA+Tukey’s; n = 37, 19, 45, 25 offspring). k, Habituation to acoustic tone (Two-way ANOVA+Tukey’s; n = 45, 25, 45, 52 offspring). l, Inhibitory effect of prepulse on startle (Two-way ANOVA+Tukey’s; n = 45, 25, 45, 52 offspring). m, Time on rotarod (One-way ANOVA+Tukey’s; n = 45, 20, 45, 50 offspring). n, Visual depth discrimination. y = 0.05, equal instances of exiting safe vs. cliff zone. (One-way ANOVA+Tukey’s; n = 38, 19, 45, 52 offspring). o, Percent time investigating novel texture (One-way ANOVA+Tukey’s; n = 20, 15, 29, 18 offspring). p, 4-MM treatment and SPF conventionalization. q, Force filament to induce 50% paw withdrawal (One-way ANOVA+Tukey’s, n = 10, 23, 15, 21 offspring). r, Latency to contact and s, remove adhesive after first contact (One-way ANOVA+Tukey’s, n = 25, 19, 15, 22 offspring). t, Pairwise data for latency to contact and remove adhesive. (Two-way ANOVA+Sidak’s, n = 5, 5 dams). u, Force filament to induce 50% paw withdrawal by sex, per litter or individual (Two-way ANOVA+Tukey’s, n = 5, 7, 5, 5 dams). v, Latency to contact and w, remove adhesive after first contact. (Two-way ANOVA+Tukey’s, n = 6, 6, 5, 5 dams). Data for SPF and ABX in uw are as in gi. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. n.s. = not statistically significant. Number of dams, sex of offspring per group and behavioural task are detailed in Supplementary Table 3.

Source Data

Extended Data Fig. 7 Maternal gut microbiota and fetal thalamocortical axons in dams colonized with a consortium of spore-forming bacteria or Bacteroides.

Faecal microbiota of E14.5 SPF and Sp dams (n = 4 dams): a, rarefaction curves of observed operational taxonomic units (OTUs). b, Principal coordinate analysis of weighted sequencing data, and c, order-level taxonomic diversity. d, Genes commonly differentially expressed in E14.5 brains from offspring of SPF and Sp vs. ABX dams (two-tailed Wald, n = 3 dams). Data for SPF and ABX are as in Fig. 1. Red indicates axonogenesis-related genes. e, PRR12 expression in E14.5 brains from offspring of SPF, ABX, and Sp dams (one-way ANOVA+Tukey’s; n = 11, 15, 8 offspring). f, g, Expression of axonogenesis-related genes in E14.5 brains from SPF, ABX, and Sp dams (Two-way ANOVA+Tukey’s; n = 11, 16, 8 offspring). Faecal microbiota of E14.5 SPF and BD dams (n = 4, 5 dams): h, rarefaction curves of the observed OTUs, i, principal coordinate analysis of weighted data, and j, order-level taxonomic diversity. k, Netrin-G1a (magenta) and L1 (cyan) in E14.5 brain sections of BD dams. Scale = 500 μm. Yellow lines = matched control ROI. l, L1 per matched control ROI, un-normalized (right) and normalized (left) by total brain area of E14.5 brain sections of SPF, ABX, and BD dams. Data for SPF and ABX are as in Figs. 1d and 2e, Extended Data Fig. 3g-i. (Two-way ANOVA+Tukey’s, n = 5 dams). m, Netrin-G1a per matched control ROI, un-normalized (middle) and normalized by total area of E14.5 brain sections (left). Netrin-G1a in area of Netrin-G1a+ staining in E14.5 brain sections (right). Data for SPF and ABX are as in Figs. 1c and 2d, Extended Data Fig. 2. (Two-way ANOVA+Tukey’s, n = 5 dams). Mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, n.s. = not statistically significant.

Source Data

Extended Data Fig. 8 Maternal serum and fetal brain metabolomic profiles from gnotobiotic dams.

a, Unsupervised hierarchical clustering of 753 maternal serum metabolites (n = 6 dams). b, Amino acid, lipid and xenobiotic metabolites dysregulated in E14.5 fetal brains of offspring from ABX vs. SPF dams (left) and ABX vs. Sp dams (right) (q<0.05; One-way ANOVA, n = 6 embryos from different dams). c, Amino acid, lipid and xenobiotic metabolites significantly dysregulated in E14.5 fetal brains of offspring from GF dams vs. SPF controls (left) and GF dams vs. Sp dams (right) (q<0.05; One-way ANOVA, n = offspring from 6 dams per group). d, Random Forest classification of top 30 metabolites in maternal serum that discriminate between SPF and Sp vs. ABX and GF dams. (n = 6 dams). e, Relative concentrations of N,N,N-trimethyl-5-aminovalerate (TMAV), trimethylamine N-oxide (TMAO), imidazole propionate (IP), hippurate (HIP), and 3-indoxyl-sulfate (3-IS) in maternal sera of SPF, ABX, GF, and Sp dams (One-way ANOVA with FDR contrasts; n = 6 dams). Mean ± SEM. *q<0.05, **q<0.01, ***q<0.001, ****q<0.0001, n.s. = not statistically significant.

Source Data

Extended Data Fig. 9 Effects of microbiome-dependent metabolites on thalamocortical axon outgrowth from GF explant co-cultures.

a, Axon number and b, axon length per 200 μm2 surface area proximal to striatal explant (St) and from (i) SPF thalamic explant (Th)+SPF St (ii) ABX Th+ABX St, and (iii) ABX Th+ABX St, supplemented with 1 nM, 100 nM, 10 μM of metabolites: trimethylamine N-oxide (TMAO), 5-aminovalerate (5-AV), imidazole propionate (IP), 3-indoxyl-sulfate (3-IS) or hippurate (HIP). (One-way ANOVA+Tukey’s, n = 14, 13, TMAO: 7, 6, 7, 5-AV: 3, 5, 7, IP: 5, 7, 7, 3-IS: 3, 7, 7, HIP: 6, 7, 8 explants). c, Axon number, and d, axon length per 200 μm2 surface area proximal to hypothalamic explant (Hy) from (i) SPF Th+SPF Hy), (ii) ABX Th+ABX Hy, and (iii) ABX Th+ABX Hy, supplemented with metabolites. (One-way ANOVA+Tukey’s, n = 14, 10, TMAO: 6, 6, 7, 5-AV: 3, 5, 7, IP: 3, 6, 7, 3-IS: 3, 7, 5, HIP: 5, 7, 8 explants). e, E14.5 GF Th proximal to GF St treated with metabolites. Scale = 250 μm. f, g, Axon number and h, i, axon length per 200 μm perimeter proximal to St (f, h) and Hy (g, i). (One-way ANOVA+Tukey’s, n = 14, 15, 14, 13, 12, 12, 16 explants). Mean ± SEM. *P < 0.05, **P < 0.01, ****P < 0.0001.

Source Data

Extended Data Fig. 10 Microbiome-dependent metabolites in E14.5 brains after maternal supplementation with 4-MM or SCFA and effects on netrin-G1a thalamocortical axons.

a, Netrin-G1a in four independent E14.5 brains from offspring of ABX+vehicle and ABX+4-MM dams. Scale = 500 μm, b, Netrin-G1a in area of Netrin-G1a+ staining in E14.5 brains from ABX+vehicle and ABX+4-MM dams. (Two-way ANOVA+Tukey’s, n = 8 dams). c, Area of Netrin-G1a+ staining in E14.5 brain sections. (Two-way ANOVA+Tukey’s, n = 8 dams). d, DAPI counts per matched ROI (yellow lines) normalized by total brain area. (Two-way ANOVA+Tukey’s, n = 4 dams). e, Levels of the metabolites (i) imidazole propionate (IP), (ii) trimethylamine oxide (TMAO), (iii) 3-indoxyl-sulfate (3-IS), (iv) hippurate (HIP) and (v) N, N, N-trimethyl-5-aminovalerate (TMAV) in E14.5 brain lysates from antibiotic-treated dams supplemented with 4-MM or vehicle (One-way ANOVA, n = 5 (ABX+vehicle), 3, 3, 5, 3, 5 (ABX+4-MM) dams). f, Levels of IP, TMAO, 3-IS, HIP, and TMAV in E14.5 brain lysates from BD dams. (One-way ANOVA+Tukey’s, n = 6, 6, 5 dams). g, SCFA administration. h, Netrin-G1a in four independent E14.5 brain sections from offspring of ABX and ABX+SCFA dams. Scale = 500 μm, i, Netrin-G1a and L1 in E14.5 brain sections from offspring of ABX and ABX+SCFA dams. j, Netrin-G1a per matched control ROI, normalized by total brain area. (Two-way ANOVA+Tukey’s, n = 4, 7 dams). k, Netrin-G1a in area of Netrin-G1a+ staining. (Two-way ANOVA+Tukey’s, n = 4, 7 dams). l, Area of Netrin-G1a+ staining. (Two-way ANOVA+Tukey’s, n = 4, 7 dams). m, L1 per matched control ROI, normalized by total brain area. (Two-way ANOVA+Tukey’s, n = 4, 7 dams). n, DAPI count per matched ROI, normalized by total brain area. (Two-way ANOVA+Tukey’s, n = 4, 7 dams). Mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, n.s. = not statistically significant.

Source Data

Supplementary information

Supplementary Information

Supplementary Discussion: Discussion of effects of the gut microbiota on axonogenesis and potential mechanisms of microbially modulated metabolites.

Reporting Summary

Supplementary Table

This file contains Supplementary Tables 1-8.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vuong, H.E., Pronovost, G.N., Williams, D.W. et al. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 586, 281–286 (2020). https://doi.org/10.1038/s41586-020-2745-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2745-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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