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.

  • Review Article
  • Published:

Transmission of the gut microbiota: spreading of health

Key Points

  • The human intestinal microbiota is dominated by anaerobic health-associated bacteria that are established and maintained in individuals through host-to-host transmission. The transmission process incorporates excretion from a host in faecal matter, survival and persistence in the external environment, and concludes with ingestion and subsequent colonization of a new host.

  • Studies of the routes of transmission of intestinal pathogens provide a useful framework to better understand intestinal commensal transmission. Both share some common transmission features, such as the use of the faecal–oral transmission route and similar survival mechanisms to persist in the external environment.

  • Environmental survival mechanisms that are used by the intestinal microbiota once expelled by a host include sporulation, aerotolerance and entering a viable but non-culturable (VBNC) dormancy state. For anaerobic bacteria, these mechanisms protect against harmful oxygen, and in the case of sporulation and VBNC states, can provide varying resistance against other environmental conditions, such as desiccation and a lack of nutrients.

  • Reservoirs are a source or a sink for bacteria during transmission. Other people in the community are the principal reservoirs of intestinal bacteria, but food, water, animals and the built environment may also facilitate transmission.

  • Transmission of commensal bacteria may be disrupted by human sanitation practices, through the use of antibiotics or a long-term change in diet, which can eliminate species within an individual thereby preventing their onward transmission. Direct interventions to restore a depleted microbiota, such as faecal microbiota transplantation (FMT), are effective in some cases.

  • A greater general awareness of the transmission of the commensal microbiota is facilitated by technological advances in different disciplines, including microbiology, bioinformatics and genomics. Fostering the transmission of commensal bacteria between people through the maintenance of a healthy lifestyle and the discerning use of antibiotics and sanitation processes may promote human health.

Abstract

Transmission of commensal intestinal bacteria between humans could promote health by establishing, maintaining and replenishing microbial diversity in the microbiota of an individual. Unlike pathogens, the routes of transmission for commensal bacteria remain unappreciated and poorly understood, despite the likely commonalities between both. Consequently, broad infection control measures that are designed to prevent pathogen transmission and infection, such as oversanitation and the overuse of antibiotics, may inadvertently affect human health by altering normal commensal transmission. In this Review, we discuss the mechanisms and factors that influence host-to-host transmission of the intestinal microbiota and examine how a better understanding of these processes will identify new approaches to nurture and restore transmission routes that are used by beneficial bacteria.

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

Figure 1: Transmission of pathogenic and commensal intestinal bacteria.
Figure 2: Environmental survival mechanisms and sporulation transmission dynamics of the intestinal microbiota.
Figure 3: Inter-host transmission dynamics of spore-forming and non-spore-forming intestinal bacteria.
Figure 4: Transmission of commensal intestinal bacteria is influenced by donor health status.

Similar content being viewed by others

References

  1. Whitman, W. B., Coleman, D. C. & Wiebe, W. J. Prokaryotes: the unseen majority. Proc. Natl Acad. Sci. USA 95, 6578–6583 (1998).

    CAS  PubMed  Google Scholar 

  2. Sekirov, I., Russell, S. L., Antunes, L. C. M. & Finlay, B. B. Gut microbiota in health and disease. Physiol. Rev. 90, 859–904 (2010).

    CAS  PubMed  Google Scholar 

  3. Louis, P., Hold, G. L. & Flint, H. J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 12, 661–672 (2014).

    CAS  PubMed  Google Scholar 

  4. Lawley, T. D. & Walker, A. W. Intestinal colonization resistance. Immunology 138, 1–11 (2013).

    CAS  PubMed  Google Scholar 

  5. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214 (2012). This landmark study characterizes the microbiota across different human body sites.

  6. Rajilic´-Stojanovic´, M. & de Vos, W. M. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol. Rev. 38, 996–1047 (2014). This study provides a detailed description and phylogeny of more than 1,000 cultured species from the intestinal microbiota.

    PubMed  PubMed Central  Google Scholar 

  7. Eckburg, P. B. Diversity of the human intestinal microbial flora. Science 308, 1635–1638 (2005).

    PubMed  PubMed Central  Google Scholar 

  8. Rolfe, R. D., Hentges, D. J., Campbell, B. J. & Barrett, J. T. Factors related to the oxygen tolerance of anaerobic bacteria. Appl. Environ. Microbiol. 36, 306–313 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Andriantsoanirina, V., Allano, S., Butel, M. J. & Aires, J. Tolerance of Bifidobacterium human isolates to bile, acid and oxygen. Anaerobe 21, 39–42 (2013).

    CAS  PubMed  Google Scholar 

  10. Hug, L. A. et al. A new view of the tree of life. Nat. Microbiol. 1, 16048 (2016).

    CAS  PubMed  Google Scholar 

  11. Ley, R. E., Lozupone, C. A., Hamady, M., Knight, R. & Gordon, J. I. Worlds within worlds: evolution of the vertebrate gut microbiota. Nat. Rev. Microbiol. 6, 776–788 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Meehan, C. J. & Beiko, R. G. A. Phylogenomic view of ecological specialization in the Lachnospiraceae, a family of digestive tract-associated bacteria. Genome Biol. Evol. 6, 703–713 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Surana, N. K. & Kasper, D. L. The yin yang of bacterial polysaccharides: lessons learned from B. fragilis PSA. Immunol. Rev. 245, 13–26 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Wexler, H. M. Bacteroides: the good, the bad, and the nitty-gritty. Clin. Microbiol. Rev. 20, 593–621 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kamada, N., Chen, G. Y., Inohara, N. & Núñez, G. Control of pathogens and pathobionts by the gut microbiota. Nat. Immunol. 14, 685–690 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Chow, W.-H. et al. An inverse relation between cagA+ strains of Helicobacter pylori infection and risk of esophageal and gastric cardia adenocarcinoma. Cancer Res. 58, 588–590 (1998).

    CAS  PubMed  Google Scholar 

  17. Petersen, C. & Round, J. L. Defining dysbiosis and its influence on host immunity and disease. Cell. Microbiol. 16, 1024–1033 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Koenig, J. E. et al. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl Acad. Sci. USA 108, 4578–4585 (2010).

    PubMed  Google Scholar 

  19. Yatsunenko, T. et al. Human gut microbiome viewed across age and geography. Nature 486, 222–227 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Falkow, S. Who speaks for the microbes? Emerg. Infect. Dis. 4, 495–497 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Falkow, S. What is a pathogen? ASM News 63, 359–365 (1997).

    Google Scholar 

  22. Wexler, A. G. et al. Human symbionts inject and neutralize antibacterial toxins to persist in the gut. Proc. Natl Acad. Sci. USA 113, 3639–3644 (2016).

    CAS  PubMed  Google Scholar 

  23. Duncan, S. H., Louis, P. & Flint, H. J. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl. Environ. Microbiol. 70, 5810–5817 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Rakoff-Nahoum, S., Foster, K. R. & Comstock, L. E. The evolution of cooperation within the gut microbiota. Nature 533, 255–259 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Turroni, F. et al. Expression of sortase-dependent pili of Bifidobacterium bifidum PRL2010 in response to environmental gut conditions. FEMS Microbiol. Lett. 357, 23–33 (2014).

    CAS  PubMed  Google Scholar 

  26. Neville, B. A. et al. Pro-inflammatory flagellin proteins of prevalent motile commensal bacteria are variably abundant in the intestinal microbiome of elderly humans. PLoS ONE 8, e68919 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. He, M. et al. Emergence and global spread of epidemic healthcare-associated Clostridium difficile. Nat. Genet. 45, 109–113 (2013).

    CAS  PubMed  Google Scholar 

  28. Harris, S. R. et al. Evolution of MRSA during hospital transmission and intercontinental spread. Science 327, 469–474 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Chase-Topping, M., Gally, D., Low, C., Matthews, L. & Woolhouse, M. Super-shedding and the link between human infection and livestock carriage of Escherichia coli O157. Nat. Rev. Microbiol. 6, 904–912 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Flo, T. H. et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature 432, 917–921 (2004).

    CAS  PubMed  Google Scholar 

  31. Stecher, B. et al. Salmonella enterica serovar Typhimurium exploits inflammation to compete with the intestinal microbiota. PLoS Biol. 5, 2177–2189 (2007).

    CAS  PubMed  Google Scholar 

  32. Rivera-Chávez, F. et al. Depletion of butyrate-producing clostridia from the gut microbiota drives an aerobic luminal expansion of Salmonella. Cell Host Microbe 19, 443–454 (2016).

    PubMed  PubMed Central  Google Scholar 

  33. Turnbaugh, P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484 (2008).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Turpin, W. et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat. Genet. 48, 1413–1417 (2016).

    CAS  PubMed  Google Scholar 

  36. Pham, T. A. et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe 16, 504–516 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Bry, L., Falk, P. G., Midtvedt, T. & Gordon, J. I. A. Model of host–microbial interactions in an open mammalian ecosystem. Science 273, 1380–1383 (1996).

    CAS  PubMed  Google Scholar 

  38. Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., Edberg, S. & Medzhitov, R. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell 118, 229–241 (2004).

    CAS  PubMed  Google Scholar 

  39. Brinkman, B. M. et al. Gut microbiota affects sensitivity to acute DSS-induced colitis independently of host genotype. Inflamm. Bowel Dis. 19, 2560–2567 (2013).

    PubMed  Google Scholar 

  40. Cullen, T. W. et al. Gut microbiota. Antimicrobial peptide resistance mediates resilience of prominent gut commensals during inflammation. Science 347, 170–175 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Sela, D. A. et al. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc. Natl Acad. Sci. USA 105, 18964–18969 (2008).

    CAS  PubMed  Google Scholar 

  42. Charbonneau, M. R. et al. A microbial perspective of human developmental biology. Nature 535, 48–55 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Walker, A. W. et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 5, 220–230 (2010).

    PubMed  PubMed Central  Google Scholar 

  44. Flint, H. J., Scott, K. P., Louis, P. & Duncan, S. H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 9, 577–589 (2012).

    CAS  PubMed  Google Scholar 

  45. Setlow, P. I will survive: DNA protection in bacterial spores. Trends Microbiol. 15, 172–180 (2007).

    CAS  PubMed  Google Scholar 

  46. Errington, J. Regulation of endospore formation in Bacillus subtilis. Nat. Rev. Microbiol. 1, 117–126 (2003).

    CAS  PubMed  Google Scholar 

  47. Browne, H. P. et al. Culturing of 'unculturable' human microbiota reveals novel taxa and extensive sporulation. Nature 533, 543–546 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Atarashi, K. et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500, 232–236 (2013). This study shows that a selection of chloroform- resistant bacteria from human faeces can induce regulatory T cells in mice and have potential therapeutic properties through the alleviation of colitis in mice.

    CAS  PubMed  Google Scholar 

  49. Janoir, C. et al. Adaptive strategies and pathogenesis of Clostridium difficile from in vivo transcriptomics. Infect. Immun. 81, 3757–3769 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Mora-Uribe, P. et al. Characterization of the adherence of Clostridium difficile spores: the integrity of the outermost layer affects adherence properties of spores of the epidemic strain R20291 to components of the intestinal mucosa. Front. Cell. Infect. Microbiol. 6, 99 (2016).

    PubMed  PubMed Central  Google Scholar 

  51. Duncan, S. H. et al. Proposal of Roseburia faecis sp. nov., Roseburia hominis sp. nov. and Roseburia inulinivorans sp. nov., based on isolates from human faeces. Int. J. Syst. Evol. Microbiol. 56, 2437–2441 (2006).

    CAS  PubMed  Google Scholar 

  52. Flint, H. J., Duncan, S. H., Scott, K. P. & Louis, P. Interactions and competition within the microbial community of the human colon: links between diet and health. Environ. Microbiol. 9, 1101–1111 (2007).

    CAS  PubMed  Google Scholar 

  53. Tally, F. P., Stewart, P. R., Sutter, V. L. & Rosenblatt, J. E. Oxygen tolerance of fresh clinical anaerobic bacteria. J. Clin. Microbiol. 1, 161–164 (1975).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Miller, R. A. & Britigan, B. E. Role of oxidants in microbial pathophysiology. Clin. Microbiol. Rev. 10, 1–18 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Khan, M. T. et al. The gut anaerobe Faecalibacterium prausnitzii uses an extracellular electron shuttle to grow at oxic–anoxic interphases. ISME J. 6, 1578–1585 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Donaldson, G. P., Lee, S. M. & Mazmanian, S. K. Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 14, 20–32 (2016).

    CAS  PubMed  Google Scholar 

  57. Albenberg, L. et al. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology 147, 1055–1063.e8 (2014).

    PubMed  PubMed Central  Google Scholar 

  58. Khan, M. T., van Dijl, J. M. & Harmsen, H. J. M. Antioxidants keep the potentially probiotic but highly oxygen-sensitive human gut bacterium Faecalibacterium prausnitzii alive at ambient air. PLoS ONE 9, e96097 (2014).

    PubMed  PubMed Central  Google Scholar 

  59. Li, L., Mendis, N., Trigui, H., Oliver, J. D. & Faucher, S. P. The importance of the viable but non-culturable state in human bacterial pathogens. Front. Microbiol. 5, 258 (2014).

    PubMed  PubMed Central  Google Scholar 

  60. Signoretto, C., Lleo, M. M., Tafi, M. C. & Canepari, P. Cell wall chemical composition of Enterococcus faecalis in the viable but nonculturable state. Appl. Environ. Microbiol. 66, 1953–1959 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Rittershaus, E. S., Baek, S.-H. & Sassetti, C. M. The normalcy of dormancy: common themes in microbial quiescence. Cell Host Microbe 13, 643–651 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Senoh, M. et al. Conversion of viable but nonculturable Vibrio cholerae to the culturable state by co-culture with eukaryotic cells. Microbiol. Immunol. 54, 502–507 (2010).

    CAS  PubMed  Google Scholar 

  63. Amel, B. K., Amine, B. & Amina, B. Survival of Vibrio fluvialis in seawater under starvation conditions. Microbiol. Res. 163, 323–328 (2008).

    CAS  PubMed  Google Scholar 

  64. Xu, H. S. et al. Survival and viability of nonculturable Escherichia coli and Vibrio cholerae in the estuarine and marine environment. Microb. Ecol. 8, 313–323 (1982).

    CAS  PubMed  Google Scholar 

  65. Gupte, A. R., De Rezende, C. L. & Joseph, S. W. Induction and resuscitation of viable but nonculturable Salmonella enterica serovar Typhimurium DT104. Appl. Environ. Microbiol. 69, 6669–6675 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Dworkin, J. & Shah, I. M. Exit from dormancy in microbial organisms. Nat. Rev. Microbiol. 8, 890–896 (2010).

    CAS  PubMed  Google Scholar 

  67. Backhed, F. et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 17, 690–703 (2015).

    PubMed  Google Scholar 

  68. Dominguez-Bello, M. G. et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl Acad. Sci. USA 107, 11971–11975 (2010).

    PubMed  Google Scholar 

  69. Chu, D. M. et al. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 23, 314–326 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Blaser, M. J. & Falkow, S. What are the consequences of the disappearing human microbiota? Nat. Rev. Microbiol. 7, 887–894 (2009). This perspective proposes that changes in human ecology can compromise the composition of the human microbiota with associated negative effects on human health.

    CAS  PubMed  Google Scholar 

  71. Arrieta, M. C., Stiemsma, L. T., Amenyogbe, N., Brown, E. M. & Finlay, B. The intestinal microbiome in early life: health and disease. Front. Immunol. 5, 427 (2014).

    PubMed  PubMed Central  Google Scholar 

  72. Schloss, P. D., Iverson, K. D., Petrosino, J. F. & Schloss, S. J. The dynamics of a family's gut microbiota reveal variations on a theme. Microbiome 2, 25 (2014).

    PubMed  PubMed Central  Google Scholar 

  73. Song, S. J. et al. Cohabiting family members share microbiota with one another and with their dogs. eLife 2, e00458 (2013).

    PubMed  PubMed Central  Google Scholar 

  74. 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). In this study, a SNP-based metagenomic pipeline is developed to demonstrate mother-to-infant vertical transmission, with early-colonizing bacteria being predominately non-spore forming and late-colonizing bacteria being predominately spore-forming, which indicates temporal environmental survival by spore-forming bacteria.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Faith, J. J., Colombel, J.-F. & Gordon, J. I. Identifying strains that contribute to complex diseases through the study of microbial inheritance. Proc. Natl Acad. Sci. USA 112, 633–640 (2015).

    CAS  PubMed  Google Scholar 

  76. Tung, J. et al. Social networks predict gut microbiome composition in wild baboons. eLife 4, e05224 (2015).

    PubMed Central  Google Scholar 

  77. Moeller, A. H. et al. Social behavior shapes the chimpanzee pan-microbiome. Sci. Adv. 2, e1500997 (2016).

    PubMed  PubMed Central  Google Scholar 

  78. De Filippo, C. et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl Acad. Sci. USA 107, 14691–14696 (2010).

    PubMed  Google Scholar 

  79. Heikkila, M. P. & Saris, P. E. J. Inhibition of Staphylococcus aureus by the commensal bacteria of human milk. J. Appl. Microbiol. 95, 471–478 (2003).

    CAS  PubMed  Google Scholar 

  80. Fernandez, L. et al. The human milk microbiota: origin and potential roles in health and disease. Pharmacol. Res. 69, 1–10 (2013).

    CAS  PubMed  Google Scholar 

  81. Derrien, M. & van Hylckama Vlieg, J. E. Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol. 23, 354–366 (2015).

    CAS  PubMed  Google Scholar 

  82. Jost, T., Lacroix, C., Braegger, C. P., Rochat, F. & Chassard, C. Vertical mother–neonate transfer of maternal gut bacteria via breastfeeding. Environ. Microbiol. 16, 2891–2904 (2014).

    CAS  PubMed  Google Scholar 

  83. Vazquez-Torres, A. et al. Extraintestinal dissemination of Salmonella by CD18-expressing phagocytes. Nature 401, 804–808 (1999).

    CAS  PubMed  Google Scholar 

  84. Martin, R. et al. The commensal microflora of human milk: new perspectives for food bacteriotherapy and probiotics. Trends Food Sci. Technol. 15, 121–127 (2004).

    CAS  Google Scholar 

  85. Lang, J. M., Eisen, J. A. & Zivkovic, A. M. The microbes we eat: abundance and taxonomy of microbes consumed in a day's worth of meals for three diet types. PeerJ 2, e659 (2014).

    PubMed  PubMed Central  Google Scholar 

  86. David, L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563 (2014).

    CAS  PubMed  Google Scholar 

  87. Leff, J. W. & Fierer, N. Bacterial communities associated with the surfaces of fresh fruits and vegetables. PLoS ONE 8, e59310 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Brito, I. L. et al. Mobile genes in the human microbiome are structured from global to individual scales. Nature 535, 435–439 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Kalliomaki, M. et al. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet 357, 1076–1079 (2001).

    CAS  PubMed  Google Scholar 

  90. Allen, S. J., Martinez, E. G., Gregorio, G. V. & Dans, L. F. Probiotics for treating acute infectious diarrhoea. Cochrane Database Syst. Rev. 11, CD003048 (2010).

    Google Scholar 

  91. Hill, C. et al. Expert consensus document: the International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 11, 506–514 (2014).

    PubMed  Google Scholar 

  92. Malinen, E. et al. PCR-ELISAII: analysis of Bifidobacterium populations in human faecal samples from a consumption trial with Bifidobacterium lactis Bb-12 and a galacto-oligosaccharide preparation. Syst. Appl. Microbiol. 25, 249–258 (2002).

    CAS  PubMed  Google Scholar 

  93. Charbonneau, D., Gibb, R. D. & Quigley, E. M. Fecal excretion of Bifidobacterium infantis 35624 and changes in fecal microbiota after eight weeks of oral supplementation with encapsulated probiotic. Gut Microbes 4, 201–211 (2013).

    PubMed  PubMed Central  Google Scholar 

  94. McNulty, N. P. et al. The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Sci. Transl Med. 3, 106ra106 (2011).

    PubMed  PubMed Central  Google Scholar 

  95. Maldonado-Gómez, M. X. et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host Microbe 20, 515–526 (2016).

    PubMed  Google Scholar 

  96. Reid, G. et al. Responders and non-responders to probiotic interventions: how can we improve the odds? Gut Microbes 1, 200–204 (2010).

    PubMed  PubMed Central  Google Scholar 

  97. Sanders, M. E. et al. An update on the use and investigation of probiotics in health and disease. Gut 62, 787–796 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Natchu, U. C. & Bhatnagar, S. Diarrhoea in children: identifying the cause and burden. Lancet 382, 184–186 (2013).

    PubMed  Google Scholar 

  99. Tallon, P., Magajna, B., Lofranco, C. & Leung, K. T. Microbial indicators of faecal contamination in water: a current perspective. Water Air Soil Poll. 166, 139–166 (2005).

    CAS  Google Scholar 

  100. Koskey, A. M. et al. Blautia and Prevotella sequences distinguish human and animal fecal pollution in Brazil surface waters. Environ. Microbiol. Rep. 6, 696–704 (2014).

    PubMed  PubMed Central  Google Scholar 

  101. Handl, S., Dowd, S. E., Garcia-Mazcorro, J. F., Steiner, J. M. & Suchodolski, J. S. Massive parallel 16S rRNA gene pyrosequencing reveals highly diverse fecal bacterial and fungal communities in healthy dogs and cats. FEMS Microbiol. Ecol. 76, 301–310 (2011).

    CAS  PubMed  Google Scholar 

  102. Hand, D., Wallis, C., Colyer, A. & Penn, C. W. Pyrosequencing the canine faecal microbiota: breadth and depth of biodiversity. PLoS ONE 8, e53115 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Lamendella, R., Domingo, J. W., Ghosh, S., Martinson, J. & Oerther, D. B. Comparative fecal metagenomics unveils unique functional capacity of the swine gut. BMC Microbiol. 11, 103 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Ley, R. E. et al. Evolution of mammals and their gut microbes. Science 320, 1647–1651 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Moller-Stray, J. et al. Two outbreaks of diarrhoea in nurseries in Norway after farm visits, April to May 2009. Euro Surveill. 17, 20321 (2012).

    PubMed  Google Scholar 

  106. Toro, M. et al. Whole-genome sequencing analysis of Salmonella enterica serovar Enteritidis isolates in Chile provides insights into possible transmission between gulls, poultry, and humans. Appl. Environ. Microbiol. 82, 6223–6232 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Knetsch, C. W. et al. Whole genome sequencing reveals potential spread of Clostridium difficile between humans and farm animals in the Netherlands, 2002 to 2011. Euro Surveill. 19, 20954 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Iovine, N. M. & Blaser, M. J. Antibiotics in animal feed and spread of resistant Campylobacter from poultry to humans. Emerg. Infect. Dis. 10, 1158–1189 (2004).

    PubMed  PubMed Central  Google Scholar 

  109. Gupta, A. et al. Antimicrobial resistance among Campylobacter strains, United States, 1997–2001. Emerg. Infect. Dis. 10, 1102–1109 (2004).

    PubMed  PubMed Central  Google Scholar 

  110. Dunn, R. R., Fierer, N., Henley, J. B., Leff, J. W. & Menninger, H. L. Home life: factors structuring the bacterial diversity found within and between homes. PLoS ONE 8, e64133 (2013).

    PubMed  PubMed Central  Google Scholar 

  111. Lax, S. et al. Longitudinal analysis of microbial interaction between humans and the indoor environment. Science 345, 1048–1052 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Meadow, J. F. et al. Bacterial communities on classroom surfaces vary with human contact. Microbiome 2, 7 (2014).

    PubMed  PubMed Central  Google Scholar 

  113. Hsu, T. et al. Urban transit system microbial communities differ by surface type and interaction with humans and the environment. mSystems 1, e00018-16 (2016).

    PubMed  PubMed Central  Google Scholar 

  114. Kelley, S. T. & Gilbert, J. A. Studying the microbiology of the indoor environment. Genome Biol. 14, 202 (2013).

    PubMed  PubMed Central  Google Scholar 

  115. Kembel, S. W. et al. Architectural design influences the diversity and structure of the built environment microbiome. ISME J. 6, 1469–1479 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Barker, J. & Jones, M. V. The potential spread of infection caused by aerosol contamination of surfaces after flushing a domestic toilet. J. Appl. Microbiol. 99, 339–347 (2005).

    CAS  PubMed  Google Scholar 

  117. Perkins, S. D., Mayfield, J., Fraser, V. & Angenent, L. T. Potentially pathogenic bacteria in shower water and air of a stem cell transplant unit. Appl. Environ. Microbiol. 75, 5363–5372 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Flores, G. E. et al. Microbial biogeography of public restroom surfaces. PLoS ONE 6, e28132 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Gibbons, S. M. et al. Ecological succession and viability of human-associated microbiota on restroom surfaces. Appl. Environ. Microbiol. 81, 765–773 (2015).

    PubMed  PubMed Central  Google Scholar 

  120. Snelling, A. M., Saville, T., Stevens, D. & Beggs, C. B. Comparative evaluation of the hygienic efficacy of an ultra-rapid hand dryer versus conventional warm air hand dryers. J. Appl. Microbiol. 110, 19–26 (2010).

    PubMed  Google Scholar 

  121. Vaishampayan, P. et al. New perspectives on viable microbial communities in low-biomass cleanroom environments. ISME J. 7, 312–324 (2013).

    CAS  PubMed  Google Scholar 

  122. Tsai, F. C. & Macher, J. M. Concentrations of airborne culturable bacteria in 100 large US office buildings from the BASE study. Indoor Air 15, 71–81 (2005).

    PubMed  Google Scholar 

  123. David, L. A. et al. Host lifestyle affects human microbiota on daily timescales. Genome Biol. 15, R89 (2014).

    PubMed  PubMed Central  Google Scholar 

  124. Korpela, K. et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat. Commun. 7, 10410 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Claesson, M. J. et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 488, 178–184 (2012).

    CAS  PubMed  Google Scholar 

  126. Lozupone, C. A., Stombaugh, J. I., Gordon, J. I., Jansson, J. K. & Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 489, 220–230 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Faith, J. J. et al. The long-term stability of the human gut microbiota. Science 341, 1237439 (2013).

    PubMed  PubMed Central  Google Scholar 

  128. Schloissnig, S. et al. Genomic variation landscape of the human gut microbiome. Nature 493, 45–50 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Costello, E. K., Stagaman, K., Dethlefsen, L., Bohannan, B. J. & Relman, D. A. The application of ecological theory toward an understanding of the human microbiome. Science 336, 1255–1262 (2012). This review uses ecological metacommunity theory to provide a framework to understand how the intestinal microbiota is assembled and to predict how it responds to disturbances.

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Lee, S. M. et al. Bacterial colonization factors control specificity and stability of the gut microbiota. Nature 501, 426–429 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Lawley, T. D. et al. Targeted restoration of the intestinal microbiota with a simple, defined bacteriotherapy resolves relapsing Clostridium difficile disease in mice. PLoS Pathog. 8, e1002995 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Clemente, J. C. et al. The microbiome of uncontacted Amerindians. Sci. Adv. 1, e1500183 (2015).

    PubMed  PubMed Central  Google Scholar 

  133. Obregon-Tito, A. J. et al. Subsistence strategies in traditional societies distinguish gut microbiomes. Nat. Commun. 6, 6505 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Blaser, M. J. Antibiotic use and its consequences for the normal microbiome. Science 352, 544–545 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Martinez, I. et al. The gut microbiota of rural Papua New Guineans: composition, diversity patterns, and ecological processes. Cell Rep. 11, 527–538 (2015).

    CAS  PubMed  Google Scholar 

  137. Strachan, D. P. Hay fever, hygiene, and household size. BMJ 299, 1259–1260 (1989). Supporting the hygiene hypothesis, this study describes lower rates of hay fever and eczema in large families, which is associated with reduced hygiene among siblings in larger families.

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Beasley, R. Worldwide variation in prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and atopic eczema: ISAAC. Lancet 351, 1225–1232 (1998).

    Google Scholar 

  139. Stiemsma, L., Reynolds, L., Turvey, S. & Finlay, B. The hygiene hypothesis: current perspectives and future therapies. Immunotargets Ther. 4, 143–157 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  141. Blaser, M. J. Who are we? Indigenous microbes and the ecology of human diseases. EMBO Rep. 7, 956–960 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Shaw, S. Y., Blanchard, J. F. & Bernstein, C. N. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am. J. Gastroenterol. 105, 2687–2692 (2010).

    PubMed  Google Scholar 

  143. Dominguez-Bello, M. G. et al. Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat. Med. 22, 250–253 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. van Nood, E. et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 368, 407–415 (2013). This study demonstrates the efficacy of faecal microbiota transplants to resolve C. difficile infection in a controlled clinical trial in humans.

    CAS  PubMed  Google Scholar 

  145. Lagier, J.-C. et al. Culture of previously uncultured members of the human gut microbiota by culturomics. Nat. Microbiol. 1, 16203 (2016).

    CAS  PubMed  Google Scholar 

  146. Rettedal, E. A., Gumpert, H. & Sommer, M. O. A. Cultivation-based multiplex phenotyping of human gut microbiota allows targeted recovery of previously uncultured bacteria. Nat. Commun. 5, 4714 (2014).

    CAS  PubMed  Google Scholar 

  147. Lau, J. T. et al. Capturing the diversity of the human gut microbiota through culture-enriched molecular profiling. Genome Med. 8, 72 (2016).

    PubMed  PubMed Central  Google Scholar 

  148. Buffie, C. G. et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517, 205–208 (2014). This study shows that a single bacterial species, Clostridium scindens , can promote resistance to infection with C. difficile in a mouse model through the synthesis of secondary bile acids that are inhibitory to this species, thus demonstrating the potential of defined bacteria-based therapies for the treatment of intestinal-associated disease.

    PubMed  PubMed Central  Google Scholar 

  149. Czaplewski, L. et al. Alternatives to antibiotics — a pipeline portfolio review. Lancet Infect. Dis. 16, 239–251 (2016).

    CAS  PubMed  Google Scholar 

  150. Yee, A. L. & Gilbert, J. A. Microbiome. Is triclosan harming your microbiome? Science 353, 348–349 (2016).

    CAS  PubMed  Google Scholar 

  151. Vandegrift, R. et al. Cleanliness in context: reconciling hygiene with a modern microbial perspective. Preprint at bioRxiv http://dx.doi.org/10.1101/095745 (2016).

    Google Scholar 

  152. Falkow, S. Molecular Koch's postulates applied to bacterial pathogenicity — a personal recollection 15 years later. Nat. Rev. Microbiol. 2, 67–72 (2004).

    CAS  PubMed  Google Scholar 

  153. Parkhill, J. & Wren, B. W. Bacterial epidemiology and biology — lessons from genome sequencing. Genome Biol. 12, 230 (2011).

    PubMed  PubMed Central  Google Scholar 

  154. Brito, I. L. & Alm, E. J. Tracking strains in the microbiome: insights from metagenomics and models. Front. Microbiol. 7, 712 (2016).

    PubMed  PubMed Central  Google Scholar 

  155. Walker, A. W., Duncan, S. H., Louis, P. & Flint, H. J. Phylogeny, culturing, and metagenomics of the human gut microbiota. Trends Microbiol. 22, 267–274 (2014). This review highlights the importance of a multidisciplinary approach that combines metagenomic sequencing, bioinformatics and culturing to study the human intestinal microbiota.

    CAS  PubMed  Google Scholar 

  156. Li, S. S. et al. Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 352, 586–589 (2016).

    CAS  PubMed  Google Scholar 

  157. Subramanian, S. et al. Persistent gut microbiota immaturity in malnourished Bangladeshi children. Nature 510, 417–421 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Lax, S., Nagler, C. R. & Gilbert, J. A. Our interface with the built environment: immunity and the indoor microbiota. Trends Immunol. 36, 121–123 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Forster, S. C. et al. HPMCD: the database of human microbial communities from metagenomic datasets and microbial reference genomes. Nucleic Acids Res. 44, D604–D609 (2015).

    PubMed  PubMed Central  Google Scholar 

  160. Rupnik, M., Wilcox, M. H. & Gerding, D. N. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nat. Rev. Microbiol. 7, 526–536 (2009).

    CAS  PubMed  Google Scholar 

  161. Gough, E., Shaikh, H. & Manges, A. R. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin. Infect. Dis. 53, 994–1002 (2011).

    PubMed  Google Scholar 

  162. Vyas, D., Aekka, A. & Vyas, A. Fecal transplant policy and legislation. World J. Gastroenterol. 21, 6–11 (2015).

    PubMed  PubMed Central  Google Scholar 

  163. Petrof, E. O. et al. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: 'RePOOPulating' the gut. Microbiome 1, 3 (2013).

    PubMed  PubMed Central  Google Scholar 

  164. Allegretti, J. R. & Hamilton, M. J. Restoring the gut microbiome for the treatment of inflammatory bowel diseases. World J. Gastroenterol. 20, 3468–3474 (2014).

    PubMed  PubMed Central  Google Scholar 

  165. Olle, B. Medicines from microbiota. Nat. Biotechnol. 31, 309–315 (2013).

    CAS  PubMed  Google Scholar 

  166. Tvede, M. & Rask-Madsen, J. Bacteriotherapy for chronic relapsing Clostridium difficile diarrhoea in six patients. Lancet 1, 1156–1160 (1989).

    CAS  PubMed  Google Scholar 

  167. Jalanka, J. et al. Long-term effects on luminal and mucosal microbiota and commonly acquired taxa in faecal microbiota transplantation for recurrent Clostridium difficile infection. BMC Med. 14, 155 (2016).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank A. Zhu for critical reading of the manuscript during preparation. H.P.B. is funded through a Medical Research Council (MRC) grant (PF451), B.A.N. is funded through a Wellcome Trust grant (098051), S.C.F. is funded through an Australian National Health and Medical Research Council (NHMRC) grant (1091097). Work in the Lawley laboratory is supported through a MRC grant (PF451) and a Wellcome Trust grant (098051).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Trevor D. Lawley.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information Table 1

Aerotolerance of non-spore forming intestinal bacteria. (PDF 195 kb)

PowerPoint slides

Glossary

Peritoneum

A membrane that lines the abdominal cavity and provides a supportive role to internal body organs, including those of the gastrointestinal tract.

Pathobionts

Members of the commensal microbiota that may become pathogenic under certain circumstances.

Facultative anaerobic bacteria

Species that can grow and survive in aerobic and anaerobic conditions.

Infectious dose

The minimum number of bacteria required to cause an infection in a host.

Colonization resistance

The capacity of the resident microbiota to prevent the establishment of new species within the community, particularly the establishment of pathogens. Colonization resistance is a feature of a stable health-associated microbiota.

Homeostasis

A stable state. The overall maintenance of precise conditions in the microbiota that promote colonization resistance, even when subjected to external perturbations or stresses.

Colonizing dose

The minimum number of bacteria required to stably colonize a new host.

Super-shedding state

A host state, typically associated with infection by pathogens, that results in the release of numerous bacteria or spores into the external environment.

Antimicrobial peptides

A diverse range of proteins that are secreted by the host as a defence mechanism against pathogens or by microorganisms to target other microorganisms in close proximity.

Siderophores

Low-molecular-weight, iron-chelating agents that are secreted by bacteria and fungi to acquire iron from the surrounding environment.

Dysbiosis

A low-diversity microbiota with reduced colonization resistance that is typically associated with inflammation and outgrowth of facultative anaerobic Proteobacteria and pathogens.

Fucosylated

The attachment of a fucose molecule to a protein. Fucosylation of epithelial cells by the host can provide a protective role through the subsequent recruitment of commensal bacteria.

Toll-like receptors

A family of transmembrane protein receptors, characterized by the presence of a Toll and interleukin-1 receptor (TIR) domain, that recognize specific microorganism-associated molecular patterns and initiate an immune response.

Nucleotide-binding oligomerization domain-like receptors

(NOD-like receptors). A family of intracellular protein receptors that recognize microorganism-associated molecular patterns and initiate an immune response.

Desiccation

The process of liquid removal or drying out, which is usually deleterious to a bacterial cell.

Vegetative cell

The form of a bacterial cell that reproduces through binary fission.

Aerobic bacteria

Species that can only grow and survive in the presence of oxygen.

Obligate anaerobic bacteria

Species that can only grow and survive in the absence of oxygen.

Flavin–thiol electron shuttle

A process that involves the transfer of electrons to oxygen through riboflavin and thiol, which enables survival and growth in the presence of oxygen.

Social grooming

Cleaning and grooming carried out by animals, particularly primates, on other individuals in their community, which has hygienic and social roles.

Horizontal gene transfer

The transfer of genetic material between different strains or species that occurs independently of vertical transmission during replication.

Probiotics

Live microorganisms that when administered in adequate amounts confer a health benefit to the host.

Indicator microorganisms

Microorganisms that are used to assay hygiene levels of foods or water, in which the quantity of the microorganisms present is inversely related to the quality or hygiene level of the product being tested.

Undernutrition

A situation whereby an individual does not consume enough nutrients, which can have adverse effects on health.

α-diversity

The ecological diversity at a single site, as measured by the number of different species and their abundance.

β-diversity

A measure of the difference in ecological diversity between different sites.

Functional foods

Foods that contain additional elements to promote health, such as probiotics, prebiotics, vitamins or minerals.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Browne, H., Neville, B., Forster, S. et al. Transmission of the gut microbiota: spreading of health. Nat Rev Microbiol 15, 531–543 (2017). https://doi.org/10.1038/nrmicro.2017.50

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro.2017.50

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