Abstract
Gut-resident microorganisms have time-limited effects in distant tissues during early life. However, the reasons behind this phenomenon are largely unknown. Here, using bacterial culture techniques, we show that a subset of live gut-resident bacteria translocate and disseminate to extraintestinal tissues (mesenteric lymph nodes and spleen) in preweaning (day of life 17), but not adult (day of life 35), mice. Translocation and dissemination in preweaning mice appeared physiologic as it did not induce an inflammatory response and required host goblet cells, the formation of goblet cell-associated antigen passages, sphingosine-1-phosphate receptor-dependent leukocyte trafficking and phagocytic cells. One translocating strain, Lactobacillus animalisWU, showed antimicrobial activity against the late-onset sepsis pathogen Escherichia coli ST69 in vitro, and its translocation was associated with protection from systemic sepsis in vivo. While limited in context, these findings challenge the idea that translocation of gut microbiota is pathological and show physiologic and beneficial translocation during early life.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others
Data availability
The stool sequencing data and L. animalisWU whole-genome sequencing data are archived in Sequence Read Archive (BioProject: PRJNA1067122 and PRJNA1066880). The RNA-seq data are archived in Gene Expression Omnibus (GSE278303).
References
Zheng, D., Liwinski, T. & Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 30, 492–506 (2020).
Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl Acad. Sci. USA 107, 12204–12209 (2010).
Verma, R. et al. Cell surface polysaccharides of Bifidobacterium bifidum induce the generation of Foxp3+ regulatory T cells. Sci. Immunol. 3, eaat6975 (2018).
Jeon, S. G. et al. Probiotic Bifidobacterium breve induces IL-10-producing Tr1 cells in the colon. PLoS Pathog. 8, e1002714 (2012).
Cervantes-Barragan, L. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357, 806–810 (2017).
Kim, Y. G. et al. Neonatal acquisition of Clostridia species protects against colonization by bacterial pathogens. Science 356, 315–319 (2017).
Becattini, S. et al. Commensal microbes provide first line defense against Listeria monocytogenes infection. J. Exp. Med. 214, 1973–1989 (2017).
Faber, F. et al. Host-mediated sugar oxidation promotes post-antibiotic pathogen expansion. Nature 534, 697–699 (2016).
Chen, R. Y. et al. A microbiota-directed food intervention for undernourished children. N. Engl. J. Med. 384, 1517–1528 (2021).
Rosado, M. M. et al. Spleen development is modulated by neonatal gut microbiota. Immunol. Lett. 199, 1–15 (2018).
Kolypetri, P. et al. Regulation of splenic monocyte homeostasis and function by gut microbial products. iScience 24, 102356 (2021).
Constantinides, M. G. et al. MAIT cells are imprinted by the microbiota in early life and promote tissue repair. Science 366, eaax6624 (2019).
Zegarra-Ruiz, D. F. et al. Thymic development of gut-microbiota-specific T cells. Nature 594, 413–417 (2021).
Morais, L. H., Schreiber, H. L. T. & Mazmanian, S. K. The gut microbiota–brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 19, 241–255 (2021).
Rodriguez, J. M. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv. Nutr. 5, 779–784 (2014).
Mazmanian, S. K., Liu, C. H., Tzianabos, A. O. & Kasper, D. L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122, 107–118 (2005).
Schretter, C. E. et al. A gut microbial factor modulates locomotor behaviour in Drosophila. Nature 563, 402–406 (2018).
Heilbronner, S., Krismer, B., Brotz-Oesterhelt, H. & Peschel, A. The microbiome-shaping roles of bacteriocins. Nat. Rev. Microbiol. 19, 726–739 (2021).
Furusawa, Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).
Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).
Campbell, C. et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 581, 475–479 (2020).
Cahenzli, J., Koller, Y., Wyss, M., Geuking, M. B. & McCoy, K. D. Intestinal microbial diversity during early-life colonization shapes long-term IgE levels. Cell Host Microbe 14, 559–570 (2013).
Karlsson, M. R., Kahu, H., Hanson, L. A., Telemo, E. & Dahlgren, U. I. Neonatal colonization of rats induces immunological tolerance to bacterial antigens. Eur. J. Immunol. 29, 109–118 (1999).
Olszak, T. et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489–493 (2012).
Knoop, K. A. et al. Microbial antigen encounter during a preweaning interval is critical for tolerance togut bacteria. Sci. Immunol. 2, eaao1314 (2017).
Doran, K. S., Banerjee, A., Disson, O. & Lecuit, M. Concepts and mechanisms: crossing host barriers. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.a010090 (2013).
Knoop, K. A., McDonald, K. G., McCrate, S., McDole, J. R. & Newberry, R. D. Microbial sensing by goblet cells controls immune surveillance of luminal antigens in the colon. Mucosal Immunol. 8, 198–210 (2015).
Balmer, M. L. et al. The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci. Transl. Med. 6, 237ra266 (2014).
Spadoni, I. et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 350, 830–834 (2015).
Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L. & Hooper, L. V. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host–microbial interface. Proc. Natl Acad. Sci. USA 105, 20858–20863 (2008).
Fulde, M. et al. Neonatal selection by Toll-like receptor 5 influences long-term gut microbiota composition. Nature 560, 489–493 (2018).
Pott, J. et al. Age-dependent TLR3 expression of the intestinal epithelium contributes to rotavirus susceptibility. PLoS Pathog. 8, e1002670 (2012).
Trahair, J. F. & Robinson, P. M. Enterocyte ultrastructure and uptake of immunoglobulins in the small intestine of the neonatal lamb. J. Anat. 166, 103–111 (1989).
Trahair, J. F., Wilson, J. M. & Neutra, M. R. Identification of a marker antigen for the endocytic stage of intestinal development in rat, sheep, and human. J. Pediatr. Gastroenterol. Nutr. 21, 277–287 (1995).
McDole, J. R. et al. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature 483, 345–349 (2012).
Knoop, K. A., McDonald, K. G., Kulkarni, D. H. & Newberry, R. D. Antibiotics promote inflammation through the translocation of native commensal colonic bacteria. Gut 65, 1100–1109 (2016).
Kulkarni, D. H. et al. Goblet cell associated antigen passages support the induction and maintenance of oral tolerance. Mucosal Immunol. 13, 271–282 (2020).
Knoop, K. A. et al. Maternal activation of the EGFR prevents translocation of gut-residing pathogenic Escherichia coli in a model of late-onset neonatal sepsis. Proc. Natl Acad. Sci. USA 117, 7941–7949 (2020).
Matloubian, M. et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427, 355–360 (2004).
Fung, T. C. et al. Lymphoid-tissue-resident commensal bacteria promote members of the IL-10 cytokine family to establish mutualism. Immunity 44, 634–646 (2016).
Udayan, S. et al. Identification of gut bacteria such as Lactobacillus johnsonii that disseminate to systemic tissues of wild type and MyD88−/− mice. Gut Microbes 14, 2007743 (2022).
St John, A. L. et al. S1P-dependent trafficking of intracellular Yersinia pestis through lymph nodes establishes buboes and systemic infection. Immunity 41, 440–450 (2014).
Louis, C. et al. Specific contributions of CSF-1 and GM-CSF to the dynamics of the mononuclear phagocyte system. J. Immunol. 195, 134–144 (2015).
Wang, L. et al. Selective depletion of CD11c+ CD11b+ dendritic cells partially abrogates tolerogenic effects of intravenous MOG in murine EAE. Eur. J. Immunol. 46, 2454–2466 (2016).
Durai, V. et al. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat. Immunol. 20, 1161–1173 (2019).
Niess, J. H. et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307, 254–258 (2005).
Van Rooijen, N. & Sanders, A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J. Immunol. Methods 174, 83–93 (1994).
Kozicky, L. K. & Sly, L. M. Depletion and reconstitution of macrophages in mice. Methods Mol. Biol. 1960, 101–112 (2019).
O’Boyle, C. J. et al. Microbiology of bacterial translocation in humans. Gut 42, 29–35 (1998).
Ribet, D. & Cossart, P. How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes Infect. 17, 173–183 (2015).
McPherson, A. C., Pandey, S. P., Bender, M. J. & Meisel, M. Systemic immunoregulatory consequences of gut commensal translocation. Trends Immunol. 42, 137–150 (2021).
Kleist, S. A. & Knoop, K. A. Understanding the elements of maternal protection from systemic bacterial infections during early life. Nutrients 12, 1045 (2020).
Greenfield, K. G., Badovinac, V. P., Griffith, T. S. & Knoop, K. A. Sepsis, cytokine storms, and immunopathology: the divide between neonates and adults. Immunohorizons 5, 512–522 (2021).
Round, J. L. & Mazmanian, S. K. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 9, 313–323 (2009).
Sefik, E. et al. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 349, 993–997 (2015).
Knoop, K. A. et al. Synchronization of mothers and offspring promotes tolerance and limits allergy. JCI Insight 5, e137943 (2020).
Blin, K. et al. antiSMASH 7.0: new and improved predictions for detection, regulation, chemical structures and visualisation. Nucleic Acids Res. 51, W46–W50 (2023).
Carl, M. A. et al. Sepsis from the gut: the enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. Clin. Infect. Dis. 58, 1211–1218 (2014).
Nocker, A., Cheung, C. Y. & Camper, A. K. Comparison of propidium monoazide with ethidium monoazide for differentiation of live vs. dead bacteria by selective removal of DNA from dead cells. J. Microbiol. Methods 67, 310–320 (2006).
Rawsthorne, H., Dock, C. N. & Jaykus, L. A. PCR-based method using propidium monoazide to distinguish viable from nonviable Bacillus subtilis spores. Appl. Environ. Microbiol. 75, 2936–2939 (2009).
Belkaid, Y. & Harrison, O. J. Homeostatic immunity and the microbiota. Immunity 46, 562–576 (2017).
Bain, C. C. et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat. Immunol. 15, 929–937 (2014).
Akagbosu, B. et al. Novel antigen-presenting cell imparts Treg-dependent tolerance to gut microbiota. Nature 610, 752–760 (2022).
Drumond, M. M. et al. Cell-free supernatant of probiotic bacteria exerted antibiofilm and antibacterial activities against Pseudomonas aeruginosa: a novel biotic therapy. Front. Pharmacol. 14, 1152588 (2023).
Corr, S. C. et al. Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118. Proc. Natl Acad. Sci. USA 104, 7617–7621 (2007).
Singer, J. R. et al. Preventing dysbiosis of the neonatal mouse intestinal microbiome protects against late-onset sepsis. Nat. Med. 25, 1772–1782 (2019).
Osteryoung, K. W. & Nunnari, J. The division of endosymbiotic organelles. Science 302, 1698–1704 (2003).
Albers, W. H., Tyler, C. W. & Boxerbaum, B. Asymptomatic bacteremia in the newborn infant. J. Pediatr. 69, 193–197 (1966).
Acknowledgements
This study was supported by National Institutes of Health grants R37AI112626, P30DK052574, R01DK097317, U01AI163073 and R01AI173220 awarded to R.D.N. B.A.R. was supported by K01DK125606 and P&F grant through DDRCC P30DK052574. V.J. was supported by the Crohn’s and Colitis Foundation grant number 902790. E.M.S. was supported by grants T32DK077653 and T32HD043010. cDC1 mice were a gift from K. Murphy, Washington University in Saint Louis School of Medicine.
Author information
Authors and Affiliations
Contributions
S.U. and R.D.N. conceptualized the project. S.U. designed the experiments. S.U., A.N.F., V.J., B.E.B., S.S., K.G.M., E.M.S., K.B.T., D.L.H., S.S., K.M.B., D.H.K., K.A.K., J.D.W., A.L.M., R.G. and E.L.J. conducted the studies. S.U. and B.A.R. conducted the bioinformatic analysis. L.D.W. performed the necropsy. S.U., B.A.R., J.T.P. and E.M.S. analysed the data. K.A.K., P.I.T., C.-S.H. and R.D.N. contributed reagents, materials and analysis tools. S.U. and R.D.N. wrote the paper with contributions from the other authors.
Corresponding author
Ethics declarations
Competing interests
R.D.N., K.G.M. and K.A.K. are inventors in patent US11,241,480 Methods for Modulation of Dietary and Microbial Exposure With Compounds Comprising An EGFR Ligand. All other authors declare no competing interests.
Peer review
Peer review information
Nature Microbiology thanks Gerard Eberl and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Bionomial distribution of translocation, characterization of colon bacterial taxa at family level in preweaning and adult mice, luminal vs mucosal residence of L. animalisWU, and changes in gut barrier with EGF.
(a) Pattern of number of CFUs recovered from the MLN and spleen of DOL17 mice (relates to supplementary table 2. b) Relative frequency of bacterial taxa at family level compared by 16S v4 sequencing of specific pathogen free laboratory mice at DOL17 (n = 3) and DOL35 (n = 4). Lactobacillaceae family (denoted by red box) was one of the most common translocating taxa, is present at both ages. c) Culture of luminal vs mucosal scrapings reveals that L. animalisWU can be present in both compartments. d) Schematic of EGF treatment to inhibit GAPs and FTY720 treatment to inhibit trafficking. e) Assessment of gut barrier function in L. animalisWU colonized mice in the presence and absence of EGF using the 4kD FITC dextran leak assay. Graphs represent the mean +/− SEM. P value calculated using two-tailed Student’s t-test.
Extended Data Fig. 2 S1PR modulation alters immune cell trafficking to spleen in preweaning mice.
Percentage of immune cells expressing S1PR (%S1PRGFP+CD45+) assessed in colon and spleen of S1PR-GFP reporter mice (B6.129P2-S1pr1tm1Hrose/J) treated at DOL17 with pan-S1PR inhibitor (FTY720) (n = 3) or not treated (control) (n = 3). Statistical analyses were performed by two-tailed Student’s t test in GraphPad Prism. Data represented as mean with individual values. P values are as denoted. Figure created with BioRender.com.
Extended Data Fig. 3 CSF1R blockade, loss of cDC1 cells, and CX3CR1 deletion does not impair L. animalisWU translocation in preweaning mice.
(a) Flow cytometry plots and (b-e) graphs demonstrate that CSFR1 blockade in preweaning mice reduces the CD45 + MHCII+ CD11c- colonic LP cellular population which can also express CX3CR1 and F4/80. (f) CFU/organ of L. animalisWU recovered from intestinal and extraintestinal tissues of L. animalisWU fed preweaning mice that were nontreated controls (n = 4) or treated with anti-CSF1R (n = 3). (g) CFU/organ of L. animalisWU recovered from intestinal and extraintestinal tissues of L. animalisWU fed wildtype, cDC1 deficient (Irf8 delta 32), or CX3CR1 deficient preweaning mice. Graphs represent mean +/− SEM. Statistical analyses were performed by two- tailed Student’s t test for B – F, one-way ANOVA for intestinal tissues in G, and one-sided cumulative binomial distribution probability test for extraintestinal tissues in F and G.
Extended Data Fig. 4 Induction of Th1, Th2 and Th17 cytokines by MLN and splenic CD4+ T cells from preweaning mice given L. animalisWU with and without EGF.
(a) Flow cytometry gating strategy for identifying Th1 cytokines (TNFα and IFNɣ) secreted by CD45+CD3+CD4+ T cells in MLNs and spleens. Frequency of TNFα+ and IFNɣ + CD4 + T cells in (b-c) MLNs and (d-e) spleens of nontreated (control), L. animalisWU fed and L. animalisWU + EGF fed mice. (f) Flow cytometry gating strategy for identifying Th2 cytokines (IL4 and IL13) secreted by CD45+CD3+CD4+ T cells in MLNs and spleens. Frequency of IL13+ and IL4 + CD4 + T cells in (g-h) MLNs and (i-j) spleens of nontreated (control), L. animalisWU fed and L. animalisWU + EGF fed mice. (k) Flow cytometry gating strategy for identifying Th17 cytokines (IL17 and IL22) secreted by CD45+CD3+CD4+ T cells in MLNs and spleens. Frequency of IL17+ and IL22 + CD4 + T cells in (l-m) MLNs and (n-o) spleens of nontreated (control), L. animalisWU fed and L. animalisWU + EGF fed mice. Statistical analyses were performed by one-way ANOVA with Dunnett’s post test. Graphs represent mean + /− SEM. P values are as denoted.
Extended Data Fig. 5 Peripheral blood neutrophils increase in preweaning mice infected with E. coli ST69.
Neutrophil (CD45+Ly6G/C+ cells) numbers in non-infected mice (n = 3) or mice infected with E. coli ST69 (n = 5) gavaged orally at DOL17. Statistical analyses were performed by two-tailed Student’s t test in GraphPad Prism. Graph represents mean +/− SEM. P values are as denoted.
Extended Data Fig. 6 L. animalisWU does not induce regulatory T cell subsets in preweaning mice.
(a) Graphs and b) representative flow plots of Foxp3+ and Foxp3+ regulatory T cell subsets (Foxp3+ percentages (%CD4+FOXP3+ and %CD4+FOXP3+RorɣT+) between mice not treated (Control) (n = 4) or fed with L. animalisWU (n = 4) from DOL10-20, in (A-B) MLN and Spleen. Statistical analyses were performed by one-way ANOVA in GraphPad Prism. Graphs represent mean +/− SEM. with a Dunnett’s post test.
Extended Data Fig. 7 Characterization of MLN and splenic bacterial taxa at family level in preweaning mice with and without L. animalisWU feeding.
Cellular populations from the a) MLN and b) spleen were isolated and treated with propidium monoazide and photoactivation, DNA was isolated and bacterial taxa were characterized by 16 s rRNA v4 sequencing. The number of different taxa identified were not dramatically altered by L. animalisWU feeding.
Extended Data Fig. 8 E. coli ST69 and L. animalisWU are sensitive to vancomycin, neomycin, ampicillin and metronidazole.
Colony forming units (CFU) of (A-D) E. coli ST69 and (E-H) L. animalisWU after plating bacteria treated with (A, E) Vancomycin (b, f) Neomycin (c, g) Ampicillin and (d, h) Metronidazole at the specified concentrations for 4 hours. Statistical analyses were performed by one way ANOVA with a Dunnett’s post test. Data represented as mean +/− SEM P values are as denoted.
Supplementary information
Supplementary Information
Supplementary Tables 1–4 and 8 and Supplementary Methods.
Supplementary Video 1
Video of the three-dimensional reconstruction of DOL17 colonic GC (UEA1+, green) containing bacteria (eubacteria FISH probe, red; nuclei, blue).
Supplementary Tables
Supplementary Tables 5–7.
Supplementary File 1
Sequence of regions in L. animalisWU.
Supplementary File 2
Sequence of regions in L. animalisWU.
Supplementary File 3
Sequence of regions in L. animalisWU.
Supplementary File 4
Sequence of regions in L. animalisWU.
Supplementary File 5
Sequence of regions in L. animalisWU.
Supplementary File 6
Sequence of regions in L. animalisWU.
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.
About this article
Cite this article
Udayan, S., Floyd, A.N., John, V. et al. Colonic goblet cell-associated antigen passages mediate physiologic and beneficial translocation of live gut bacteria in preweaning mice. Nat Microbiol 10, 927–938 (2025). https://doi.org/10.1038/s41564-025-01965-1
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41564-025-01965-1