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:

MICROBIOME

Culturing the human microbiota and culturomics

Abstract

The gut microbiota has an important role in the maintenance of human health and in disease pathogenesis. This importance was realized through the advent of omics technologies and their application to improve our knowledge of the gut microbial ecosystem. In particular, the use of metagenomics has revealed the diversity of the gut microbiota, but it has also highlighted that the majority of bacteria in the gut remain uncultured. Culturomics was developed to culture and identify unknown bacteria that inhabit the human gut as a part of the rebirth of culture techniques in microbiology. Consisting of multiple culture conditions combined with the rapid identification of bacteria, the culturomic approach has enabled the culture of hundreds of new microorganisms that are associated with humans, providing exciting new perspectives on host–bacteria relationships. In this Review, we discuss why and how culturomics was developed. We describe how culturomics has extended our understanding of bacterial diversity and then explore how culturomics can be applied to the study of the human microbiota and the potential implications for human health.

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: The strengths and weaknesses of metagenomic studies.
Fig. 2: The culturomic workflow.
Fig. 3: The history and future potential applications of culturomics in clinical microbiology.

Similar content being viewed by others

References

  1. Turnbaugh, P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484 (2009).

    Article  PubMed  CAS  Google Scholar 

  2. Lopez-Siles, M. et al. Mucosa-associated Faecalibacterium prausnitzii phylotype richness is reduced in patients with inflammatory bowel disease. Appl. Environ. Microbiol. 81, 7582–7592 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Torres-Fuentes, C., Schellekens, H., Dinan, T. G. & Cryan, J. F. The microbiota-gut-brain axis in obesity. Lancet Gastroenterol. Hepatol. 2, 747–756 (2017).

    Article  PubMed  Google Scholar 

  4. Metchnikoff, E. Les microbes intestinaux. Bull. l’Inst. Pasteur 6, 217–228 (1903). Metchnikoff is the first to describe probiotics.

    Google Scholar 

  5. Lagier, J. C. et al. Microbial culturomics: paradigm shift in the human gut microbiome study. Clin. Microbiol. Infect. 18, 1185–1193 (2012). This is the first reported microbial culturomic study.

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  7. Kaeberlein, T., Lewis, K. & Epstein, S. S. Isolating “uncultivable” microorganisms in pure culture in a simulated natural environment. Science 296, 1127–1129 (2002).

    Article  PubMed  CAS  Google Scholar 

  8. Nichols, D. et al. Use of ichip for high-throughput in situ cultivation of “uncultivable” microbial species. Appl. Environ. Microbiol. 76, 2445–2450 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Lagier, J. C., Khelaifia, S. & Tidjani-Alou, M. Culture of previously uncultured members of the human gut microbiota by culturomics. Nat. Microbiol. 1, 16203 (2016). This study drastically increases the number of known human-associated microbial species.

    Article  PubMed  CAS  Google Scholar 

  10. Venter, J. C. et al. Environmental genome shotgun sequencing of the Sargasso Sea. Science 304, 66–74 (2004).

    Article  PubMed  CAS  Google Scholar 

  11. Hold, G. L., Pryde, S. E., Russell, V. J., Furrie, E. & Flint, H. J. Assessment of microbial diversity in human colonic samples by 16S rDNA sequence analysis. FEMS Microbiol. Ecol. 39, 33–39 (2002).

    Article  PubMed  CAS  Google Scholar 

  12. Suau, A. et al. Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl. Environ. Microbiol. 65, 4799–4807 (1999).

    PubMed  PubMed Central  CAS  Google Scholar 

  13. Lagier, J. C., Million, M., Hugon, P., Armougom, F. & Raoult, D. Human gut microbiota: repertoire and variations. Front. Cell. Infect. Microbiol. 2, 136 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Jovel, J. et al. Characterization of the gut microbiome using 16S or shotgun metagenomics. Front. Microbiol. 7, 459 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Truong, D. T., Tett, A. & Pasolli, E. Microbial strain-level population structure and genetic diversity from metagenomes. Genome Res. 27, 626–638 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Dhariwal, A. et al. MicrobiomeAnalyst: a web-based tool for comprehensive statistical, visual and meta-analysis of microbiome data. Nucleic Acids Res. 45, W180–W188 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  18. Meyer, F. et al. The metagenomics RAST server — a public resource for the automatic phylogenetic and functional analysis of metagenomes. BMC Bioinformatics 9, 386 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Lynch, M. D. & Neufeld, J. D. Ecology and exploration of the rare biosphere. Nat. Rev. Microbiol. 13, 217–229 (2015).

    Article  PubMed  CAS  Google Scholar 

  20. Cleary, B. et al. Detection of low-abundance bacterial strains in metagenomic datasets by eigengenome partitioning. Genome Res. 33, 1053–1060 (2015).

    CAS  Google Scholar 

  21. Lanza, V. F. et al. In-depth resistome analysis by targeted metagenomics. Microbiome 6, 11 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Browne, H. P. et al. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature 533, 543–546 (2016). This study uses ethanol pretreatment to identify bacterial spores and increase the number of known human-associated bacterial species.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Goodman, A. L. et al. Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice. Proc. Natl Acad. Sci. USA 108, 6252–6257 (2011).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  24. Rinke, C. et al. Insights into the phylogeny and coding potential of microbial dark matter. Nature 499, 431–437 (2013). This study introduces the concept of microbial dark matter.

    Article  PubMed  CAS  Google Scholar 

  25. Cangelosi, G. A. & Meschke, J. S. Dead or alive: molecular assessment of microbial viability. Appl. Environ. Microbiol. 80, 5884–5891 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Angelakis, E. et al. Gut microbiome and dietary patterns in different Saudi populations and monkeys. Sci. Rep. 6, 32191 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Zapka, C. et al. Comparison of standard culture-based method to culture-independent method for evaluation of hygiene effects on the hand microbiome. mBio 8, e00093-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Bollmann, A., Lewis, K. & Epstein, S. S. Incubation of environmental samples in a diffusion chamber increases the diversity of recovered isolates. Appl. Environ. Microbiol. 73, 6386–6390 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Hugon, P. et al. A comprehensive repertoire of prokaryotic species identified in human beings. Lancet. Infect. Dis. 15, 1211–1219 (2015).

    Article  PubMed  Google Scholar 

  30. Staley, J. T. & Konopka, A. Measurement of in situ activities of nonphotosynthetic microorganisms in aquatic and terrestrial habitats. Annu. Rev. Microbiol. 39, 321–346 (1985).

    Article  PubMed  CAS  Google Scholar 

  31. Lagier, J. C. et al. The rebirth of culture in microbiology through the example of culturomics to study human gut microbiota. Clin. Microbiol. Rev. 28, 237–264 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Button, D. K., Schut, F., Quang, P., Martin, R. & Robertson, B. R. Viability and isolation of marine bacteria by dilution culture: theory, procedures, and initial results. Appl. Environ. Microbiol. 59, 881–891 (1993).

    PubMed  PubMed Central  CAS  Google Scholar 

  33. Rappe, M. S., Connon, S. A., Vergin, K. L. & Giovannoni, S. J. Cultivation of the ubiquitous SAR11 marine bacterioplankton clade. Nature 418, 630–633 (2002).

    Article  PubMed  CAS  Google Scholar 

  34. Dadon-Pilosof, A. et al. Surface properties of SAR11 bacteria facilitate grazing avoidance. Nat. Microbiol. 2, 1608–1615 (2017).

    Article  PubMed  CAS  Google Scholar 

  35. Stewart, E. J. Growing unculturable bacteria. J. Bacteriol. 194, 4151–4160 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Nichols, D. et al. Short peptide induces an “uncultivable” microorganism to grow in vitro. Appl. Environ. Microbiol. 74, 4889–4897 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Raoult, D. et al. Cultivation of the bacillus of Whipple’s disease. N. Engl. J. Med. 342, 620–625 (2000).

    Article  PubMed  CAS  Google Scholar 

  38. Raoult, D. et al. A flea-associated Rickettsia pathogenic for humans. Emerg. Infect. Dis. 7, 73–81 (2001).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Gouriet, F., Fenollar, F., Patrice, J. Y., Drancourt, M. & Raoult, D. Use of shell-vial cell culture assay for isolation of bacteria from clinical specimens: 13 years of experience. J. Clin. Microbiol. 43, 4993–5002 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Lagier, J. C. et al. Current and past strategies for bacterial culture in clinical microbiology. Clin. Microbiol. Rev. 28, 208–236 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Nottingham, P. M. & Hungate, R. E. Isolation of methanogenic bacteria from feces of man. J. Bacteriol. 96, 2178–2179 (1968).

    PubMed  PubMed Central  CAS  Google Scholar 

  42. Moore, W. E. & Holdeman, L. V. Human fecal flora: the normal flora of 20 Japanese-Hawaiians. Appl. Microbiol. 27, 961–979 (1974).

    PubMed  PubMed Central  CAS  Google Scholar 

  43. Finegold, S. M., Attebery, H. R. & Sutter, V. L. Effect of diet on human fecal flora: comparison of Japanese and American diets. Am. J. Clin. Nutr. 27, 1456–1469 (1974).

    Article  PubMed  CAS  Google Scholar 

  44. Aries, V., Crowther, J. S., Drasar, B. S., Hill, M. J. & Williams, R. E. Bacteria and the aetiology of cancer of the large bowel. Gut 10, 334–335 (1969).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Rajilic-Stojanovic, M., Smidt, H. & de Vos, W. M. Diversity of the human gastrointestinal tract microbiota revisited. Environ. Microbiol. 9, 2125–2136 (2007).

    Article  PubMed  Google Scholar 

  46. Moore, W. E. & Moore, L. H. Intestinal floras of populations that have a high risk of colon cancer. Appl. Environ. Microbiol. 61, 3202–3207 (1995).

    PubMed  PubMed Central  CAS  Google Scholar 

  47. 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).

    Article  PubMed  CAS  Google Scholar 

  48. La, S. B., Khelaifia, S., Lagier, J. C. & Raoult, D. Aerobic culture of anaerobic bacteria using antioxidants: a preliminary report. Eur. J. Clin. Microbiol. Infect. Dis. 33, 1781–1783 (2014).

    Article  CAS  Google Scholar 

  49. Dione, N., Khelaifia, S., La, S. B., Lagier, J. C. & Raoult, D. A quasi-universal medium to break the aerobic/anaerobic bacterial culture dichotomy in clinical microbiology. Clin. Microbiol. Infect. 22, 53–58 (2016).

    Article  PubMed  CAS  Google Scholar 

  50. Khelaifia, S. et al. Aerobic culture of methanogenic archaea without an external source of hydrogen. Eur. J. Clin. Microbiol. Infect. Dis. 35, 985–991 (2016).

    Article  PubMed  CAS  Google Scholar 

  51. Wolochow, H. Detection of Airborne Microorganisms through their Unique Compounds. Naval Biosciences Laboratory Technical Reports, Report No. 211170 (Armed Services Technical Information Agency, Arlington, VA, 1959).

  52. Anhalt, J. & Fenselau, C. Identification of bacteria using mass spectrometry. Anal. Chem. 47, 219–225 (1975).

    Article  CAS  Google Scholar 

  53. Fenselau, C. & Demirev, P. A. Characterization of intact microorganisms by MALDI mass spectrometry. Mass Spectrom. Rev. 20, 157–171 (2001).

    Article  PubMed  CAS  Google Scholar 

  54. Seng, P. et al. Ongoing revolution in bacteriology: routine identification of bacteria by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Clin. Infect. Dis. 49, 543–551 (2009).

    Article  PubMed  CAS  Google Scholar 

  55. Seng, P. et al. Identification of rare pathogenic bacteria in a clinical microbiology laboratory: impact of matrix-assisted laser desorption ionization-time of flight mass spectrometry. J. Clin. Microbiol. 51, 2182–2194 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Gross, A. et al. Technologies for single-cell isolation. Int. J. Mol. Sci. 16, 16897–16919 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Derrien, M., Vaughan, E. E., Plugge, C. M. & de Vos, W. M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 54, 1469–1476 (2004).

    Article  PubMed  CAS  Google Scholar 

  58. Duncan, S. H., Hold, G. L., Harmsen, H. J., Stewart, C. S. & Flint, H. J. Growth requirements and fermentation products of Fusobacterium prausnitzii, and a proposal to reclassify it as Faecalibacterium prausnitzii gen. nov., comb. nov. Int. J. Syst. Evol. Microbiol. 52, 2141–2146 (2002).

    PubMed  CAS  Google Scholar 

  59. Ma, L. et al. Gene-targeted microfluidic cultivation validated by isolation of a gut bacterium listed in Human Microbiome Project’s Most Wanted taxa. Proc. Natl Acad. Sci. USA 111, 9768–9773 (2014).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  60. Duncan, S. H. et al. Wheat bran promotes enrichment within the human colonic microbiota of butyrate-producing bacteria that release ferulic acid. Environ. Microbiol. 18, 2214–2225 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Walker, A. W., Duncan, S. H., McWilliam Leitch, E. C., Child, M. W. & Flint, H. J. pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl. Environ. Microbiol. 71, 3692–3700 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Chassard, C., Goumy, V., Leclerc, M., Del’homme, C. & Bernalier-Donadille, A. Characterization of the xylan-degrading microbial community from human faeces. FEMS Microbiol. Ecol. 61, 121–131 (2007).

    Article  PubMed  CAS  Google Scholar 

  63. Hugon, P. et al. A comprehensive repertoire of prokaryotic species identified in human beings. Lancet Infect. Dis. 15, 1211–1219 (2015).

    Article  PubMed  Google Scholar 

  64. Hill, M. J. & Drasar, B. S. The normal colonic bacterial flora. Gut 16, 318–323 (1975).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Shivaji, S. We are not alone: a case for the human microbiome in extra intestinal diseases. Gut Pathog. 9, 13 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Morotomi, M., Nagai, F. & Watanabe, Y. Description of Christensenella minuta gen. nov., sp. nov., isolated from human faeces, which forms a distinct branch in the order Clostridiales, and proposal of Christensenellaceae fam. nov. Int. J. Syst. Evol. Microbiol. 62, 144–149 (2012).

    Article  PubMed  CAS  Google Scholar 

  67. Dubourg, G. et al. First isolation of Akkermansia muciniphila in a blood-culture sample. Clin. Microbiol. Infect. 23, 682–683 (2017).

    Article  PubMed  CAS  Google Scholar 

  68. Fathi, P. & Wu, S. Isolation, detection, and characterization of enterotoxigenic Bacteroides fragilis in clinical samples. Open Microbiol. J. 10, 57–63 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Schloss, P. D. et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75, 7537–7541 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Huson, D. H., Mitra, S., Ruscheweyh, H. J., Weber, N. & Schuster, S. C. Integrative analysis of environmental sequences using MEGAN4. Genome Res. 21, 1552–1560 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Cassir, N. et al. Clostridium butyricum strains and dysbiosis linked to necrotizing enterocolitis in preterm neonates. Clin. Infect. Dis. 61, 1107–1115 (2015).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Million, M. et al. Increased gut redox and depletion of anaerobic and methanogenic prokaryotes in severe acute malnutrition. Sci. Rep. 6, 26051 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Tidjani, A. M. et al. Gut bacteria missing in severe acute malnutrition, can we identify potential probiotics by culturomics? Front. Microbiol. 8, 899 (2017).

    Article  Google Scholar 

  77. Wagner, V. E. et al. Effects of a gut pathobiont in a gnotobiotic mouse model of childhood undernutrition. Sci. Transl Med. 8, 366ra164 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Trehan, I. et al. Antibiotics as part of the management of severe acute malnutrition. N. Engl. J. Med. 368, 425–435 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Million, M., Diallo, A. & Raoult, D. Gut microbiota and malnutrition. Microb. Pathog. 106, 127–138 (2017).

    Article  PubMed  Google Scholar 

  80. Allegretti, J. R. et al. Recurrent Clostridium difficile infection associates with distinct bile acid and microbiome profiles. Aliment. Pharmacol. Ther. 43, 1142–1153 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Chang, J. Y. et al. Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea. J. Infect. Dis. 197, 435–438 (2008).

    Article  PubMed  Google Scholar 

  82. Schubert, A. M. et al. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls. mBio 5, e01021-14 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Theriot, C. M. & Young, V. B. Interactions between the gastrointestinal microbiome and Clostridium difficile. Annu. Rev. Microbiol. 69, 445–461 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Fuentes, S. et al. Reset of a critically disturbed microbial ecosystem: faecal transplant in recurrent Clostridium difficile infection. ISME J. 8, 1621–1633 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Seekatz, A. M. et al. Recovery of the gut microbiome following fecal microbiota transplantation. mBio 5, e00893-14 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Petrof, E. O. et al. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: ‘RePOOPulating’ the gut. Microbiome 1, 3 (2013). This paper presents a seminal study on the use of a cocktail of bacteria rather than a whole stool sample to perform FMT.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Tvede, M., Tinggaard, M. & Helms, M. Rectal bacteriotherapy for recurrent Clostridium difficile-associated diarrhoea: results from a case series of 55 patients in Denmark 2000–2012. Clin. Microbiol. Infect. 21, 48–53 (2015).

    Article  PubMed  CAS  Google Scholar 

  88. Khanna, S. et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J. Infect. Dis. 214, 173–181 (2016).

    Article  PubMed  Google Scholar 

  89. Gerding, D. N. et al. Administration of spores of nontoxigenic Clostridium difficile strain M3 for prevention of recurrent C. difficile infection: a randomized clinical trial. JAMA 313, 1719–1727 (2015).

    Article  PubMed  Google Scholar 

  90. Amrane, S., Bachar, D., Lagier, J. C. & Raoult, D. Clostridium scindens is present in the gut microbiota during Clostridium difficile infection. A metagenomic and culturomic analysis. J. Clin. Microbiol. 56, e01663–17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Buffie, C. G. et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517, 205–208 (2015).

    Article  PubMed  CAS  Google Scholar 

  92. Raoult, D. Is there a link between urinary microbiota and bladder cancer? Eur. J. Epidemiol. 32, 255 (2017).

    Article  PubMed  Google Scholar 

  93. Hilt, E. E. et al. Urine is not sterile: use of enhanced urine culture techniques to detect resident bacterial flora in the adult female bladder. J. Clin. Microbiol. 52, 871–876 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Whiteside, S. A., Razvi, H., Dave, S., Reid, G. & Burton, J. P. The microbiome of the urinary tract — a role beyond infection. Nat. Rev. Urol. 12, 81–90 (2015).

    Article  PubMed  Google Scholar 

  95. Fenollar, F. & Raoult, D. Does bacterial vaginosis result from fecal transplantation? J. Infect. Dis. 214, 1784 (2016).

    Article  PubMed  Google Scholar 

  96. Iida, N. et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 342, 967–970 (2013).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  97. Vetizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015). This study identifies relationships between the gut microbiota and the efficacy of anticancer immunotherapy.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Sivan, A. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350, 1084–1089 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Dubin, K. et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat. Commun. 7, 10391 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Tsilimigras, M. C., Fodor, A. & Jobin, C. Carcinogenesis and therapeutics: the microbiota perspective. Nat. Microbiol. 2, 17008 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  101. Daillere, R. et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity 45, 931–943 (2016).

    Article  PubMed  CAS  Google Scholar 

  102. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    Article  PubMed  CAS  Google Scholar 

  103. Zitvogel, L., Daillere, R., Roberti, M. P., Routy, B. & Kroemer, G. Anticancer effects of the microbiome and its products. Nat. Rev. Microbiol. 15, 465–478 (2017).

    Article  PubMed  CAS  Google Scholar 

  104. Dubourg, G., Abat, C. & Raoult, D. Why new antibiotics are not obviously useful now. Int. J. Antimicrob. Agents 49, 549–553 (2017).

    Article  PubMed  CAS  Google Scholar 

  105. Millette, M. et al. Purification and identification of the pediocin produced by Pediococcus acidilactici MM33, a new human intestinal strain. J. Appl. Microbiol. 104, 269–275 (2008).

    PubMed  CAS  Google Scholar 

  106. O’Shea, E. F. et al. Production of multiple bacteriocins from a single locus by gastrointestinal strains of Lactobacillus salivarius. J. Bacteriol. 193, 6973–6982 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. De, V. L. & Leroy, F. Bacteriocins from lactic acid bacteria: production, purification, and food applications. J. Mol. Microbiol. Biotechnol. 13, 194–199 (2007).

    Article  CAS  Google Scholar 

  108. Zheng, J., Ganzle, M. G., Lin, X. B., Ruan, L. & Sun, M. Diversity and dynamics of bacteriocins from human microbiome. Environ. Microbiol. 17, 2133–2143 (2015).

    Article  PubMed  CAS  Google Scholar 

  109. Zipperer, A. et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature 535, 511–516 (2016). This sequencing study demonstrates that the human microbiome contains many putative genes that encode antimicrobial agents.

    Article  PubMed  CAS  Google Scholar 

  110. Becattini, S. et al. Commensal microbes provide first line defense against Listeria monocytogenes infection. J. Exp. Med. 214, 1973–1989 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Szajewska, H. & Kolodziej, M. Systematic review with meta-analysis: Saccharomyces boulardii in the prevention of antibiotic-associated diarrhoea. Aliment. Pharmacol. Ther. 42, 793–801 (2015).

    Article  PubMed  CAS  Google Scholar 

  112. Weingarden, A. R. & Vaughn, B. P. Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease. Gut Microbes 8, 238–252 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Stackebrandt, E. & Ebers, J. Taxonomic parameters revisited: tarnished gold standards. Microbiol. Today 33, 152–155 (2006).

    Google Scholar 

  114. Sentausa, E. & Fournier, P. E. Advantages and limitations of genomics in prokaryotic taxonomy. Clin. Microbiol. Infect. 19, 790–795 (2013).

    Article  PubMed  CAS  Google Scholar 

  115. Fournier, P. E., Raoult, D. & Drancourt, M. New Species Announcement: a new format to prompt the description of new human microbial species. New Microbes New Infect. 15, 136–137 (2017).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This work has benefited from French State support, managed by the ‘Agence Nationale pour la Recherche’, including the ‘Programme d’Investissement d’Avenir’ under the reference Méditerranée Infection 10-IAHU-03. This work was also funded by the Prix Louis D. and by Région Provence Alpes Côte d’Azur and European funding FEDER PRIMI.

Author information

Authors and Affiliations

Authors

Contributions

J.-C.L., G.D., M.M., F.C., M.B., F.F., A.L. and D.R. researched the data for the article. J.-C.L., G.D., M.M., J.-M.R., P.-E.F. and D.R. substantially contributed to discussion of content. J.-C.L., G.D., M.M., F.C., M.B., F.F., A.L., P.-E.F. and D.R. wrote the article. J.-C.L., G.D., M.M., F.C., M.B., F.F., A.L., J.-M.R., P.-E.F. and D.R. reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Didier Raoult.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Supplementary information

Glossary

Microbiota

Consortia of microorganisms living in a defined environment.

Commensal

Microorganism that colonizes its host without causing disease.

Probiotics

Living microorganisms that can be beneficial for health.

Metagenomics

Shotgun sequencing of DNA isolated directly from a specific environment.

Bacteriotherapy

Administration of live microorganisms, alone or as complete ecosystems, to improve human health. FMT is an example of bacteriotherapy.

Microbial dark matter

Unassigned sequences from metagenomic studies.

MALDI-TOF mass spectrometry

Combination of a soft ionization method (MALDI) with mass spectrometry, enabling the identification of proteins through their mass and their charge number.

Diffusion chamber

A method that allows microorganisms to grow in their natural environment, the inoculum being sandwiched between semipermeable membranes of the chamber, allowing a free exchange of chemicals with the external milieu.

Obligate anaerobes

Organisms for which atmospheric oxygen concentration is toxic.

Shell-vial technique

A centrifuge-enhanced tissue culture assay.

Axenic media

Host-cell-free growth culture media.

Anaerobes

Organisms that do not require oxygen for growth.

Schaedler agar

Solid culture medium recommended for the isolation of anaerobic bacteria from clinical specimens.

Xylanolytic microbial communities

Communities containing prokaryotes with the ability to degrade xylan.

Taxonogenomics

Modern approach to describe new taxa isolated from culturomic studies combining genome sequencing and phenotypic information.

Operational taxonomic units.

Clusters of DNA sequences from unidentified organisms organized according to their DNA sequence similarity.

Microbiome

All genes and genomes of a microbiota.

Kwashiorkor

Clinical nutritional disease including irritability, diarrhoea with indigested food, swelling of the hands and feet (nutritional oedema), general puffiness of the face (moon face) and skin changes (depigmentation and thickened black and crumpled patches with peeling and rash).

Faecal microbiota transplantation

The transfer of faecal material from a healthy individual into an individual with a condition.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lagier, JC., Dubourg, G., Million, M. et al. Culturing the human microbiota and culturomics. Nat Rev Microbiol 16, 540–550 (2018). https://doi.org/10.1038/s41579-018-0041-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-018-0041-0

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