Metagenomic analyses reveal antibiotic-induced temporal and spatial changes in intestinal microbiota with associated alterations in immune cell homeostasis

D A Hill, C Hoffmann, M C Abt, Y Du, D Kobuley, T J Kirn, F D Bushman, D Artis, D A Hill, C Hoffmann, M C Abt, Y Du, D Kobuley, T J Kirn, F D Bushman, D Artis

Abstract

Despite widespread use of antibiotics, few studies have measured their effects on the burden or diversity of bacteria in the mammalian intestine. We developed an oral antibiotic treatment protocol and characterized its effects on murine intestinal bacterial communities and immune cell homeostasis. Antibiotic administration resulted in a 10-fold reduction in the amount of intestinal bacteria present and sequencing of 16S rDNA segments revealed significant temporal and spatial effects on luminal and mucosal-associated communities including reductions in luminal Firmicutes and mucosal-associated Lactobacillus species, and persistence of bacteria belonging to the Bacteroidetes and Proteobacteria phyla. Concurrently, antibiotic administration resulted in reduced RELM beta production, and reduced production of interferon-gamma and interleukin-17A by mucosal CD4(+) T lymphocytes. This comprehensive temporal and spatial metagenomic analyses will provide a resource and framework to test the influence of bacterial communities in murine models of human disease.

Figures

Figure 1
Figure 1
Antibiotic delivery in drinking water modulates intestinal communities but results in animal dehydration. A) 16S rDNA gene copies as quantified by real-time RT-PCR from stool pellets collected from naïve (H2O; N=5) or antibiotic-treated (ABX; N=5) animals (± s.e.m). B) Family level phylogenetic classification of 16S rDNA frequencies in stool pellets collected from naïve animals (H2O; N=5), antibiotic-treated animals (ABX; N=5), germ-free animals (GF; N=3), or autoclaved animal food (Chow; N=3). C) Weights of animals fed unsupplemented (H2O; N=4) or antibiotic supplemented (ABX; N=4) water (***, p

Figure 2

Antibiotic administration by gavage mimics…

Figure 2

Antibiotic administration by gavage mimics molecular, anatomic, histologic, and immunologic characteristics of reduced…

Figure 2
Antibiotic administration by gavage mimics molecular, anatomic, histologic, and immunologic characteristics of reduced microbial stimulation without animal dehydration. A) Weights of animals gavaged with unsupplemented (H2O; N=4) or antibiotic-supplemented (ABX; N=5) water (± s.e.m). B) 16S rDNA gene copies as quantified from stool pellets collected before (day 0) or over the course of antibiotic treatment (days 1, 5, 9) (N=5; ± s.e.m). C) Cecal images from control-treated (H2O), day 10 antibiotic-treated (ABX), or germ-free animals (GF) (bar, 1 cm). D) Photomicrographs of H&E stained cecal sections from control-treated (H2O), day 10 antibiotic-treated (ABX), or germ-free animals (GF) showing expansion of the lamina propria and enterocyte hyperplasia in ceca from antibiotic-treated and germ-free animals (bar, 50 μm). E) Quantification of cecal villus length and width from control-treated (H2O; N=4), day 10 antibiotic-treated (ABX; N=5), or germ-free animals (GF; N=3) (***, p

Figure 3

Global restructuring of intestinal bacterial…

Figure 3

Global restructuring of intestinal bacterial communities over time. A) Unweighted or B) weighted…

Figure 3
Global restructuring of intestinal bacterial communities over time. A) Unweighted or B) weighted UniFrac analysis of stool pellet, luminal content, and mucosal-associated samples from control-treated or antibiotic-treated animals. C) Unweighted or D) weighted UniFrac analysis of stool pellet samples from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals from day 1 to day 9 post treatment initiation. Colored symbols represent treatment day, white squares represent means, red line represents linear regression analysis (± s.e.m, significance determined by regression analysis). E) Genus level phylogenetic classification of 16S rDNA frequencies in stool pellets collected from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals from day 0 to day 9. F) Average frequency of bacterial groups before (Antibiotic Day 0) or during (Antibiotic Day 1, 9) antibiotic treatment. Mann-Whitney P-values of changes in group frequency with antibiotic treatment. Frequency reductions upon antibiotic treatment in red, increases in green, non-significant changes blank.

Figure 4

Antibiotics modify luminal and mucosal-associated…

Figure 4

Antibiotics modify luminal and mucosal-associated bacterial communities along the length of the colon.…

Figure 4
Antibiotics modify luminal and mucosal-associated bacterial communities along the length of the colon. A) Genus level phylogenetic classification of 16S rDNA frequencies in luminal samples collected from control-treated (H2O; N=4) or day 10 antibiotic-treated (ABX; N=5) animals from the cecum (Ce), proximal colon (Prox), or distal colon (Dist). B) Average frequency of bacterial groups in samples from control-treated (H2O; N=4) or antibiotic-treated (Antibiotic Day 10; N=5) animals. C) Genus level phylogenetic classification of 16S rDNA frequencies in mucosal-associated samples collected from control-treated (H2O; N=4) or day 10 antibiotic-treated (ABX; N=5) animals from the cecum (Ce), proximal colon (Prox), or distal colon (Dist). D) Average frequency of bacterial groups in mucosal-associated samples from control-treated (H2O; N=4) or antibiotic-treated (Antibiotic Day 10; N=5) animals. Mann-Whitney P-values of changes in group frequency with antibiotic treatment. Frequency reductions upon antibiotic treatment in red, increases in green, non-significant changes blank.

Figure 5

Antibiotic treatment alters mucosal CD4…

Figure 5

Antibiotic treatment alters mucosal CD4 + T lymphocyte homeostasis. A) Cytokine mRNA expression…

Figure 5
Antibiotic treatment alters mucosal CD4+ T lymphocyte homeostasis. A) Cytokine mRNA expression on day 10 as assessed by real-time RT-PCR of small intestine samples from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals (***, p<0.001; ± s.e.m). B) Expression of IFNγ or IL-17A by CD4+ T lymphocytes in the small intestinal lamina propria of control-treated (H2O) or day 10 antibiotic-treated (ABX) animals as analyzed by flow cytometry. C) Expression of IFNγ or IL-17A by CD4+ T lymphocytes in the mesenteric lymph nodes of control-treated (H2O) or day 10 antibiotic-treated (ABX) animals analyzed by flow cytometry. D) Statistical analysis of mesenteric lymph node CD4+ T lymphocyte cytokine expression from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals (*, p<0.05; ± s.e.m).
Similar articles
Cited by
References
    1. Eckburg PB, Lepp PW, Relman DA. Archaea and their potential role in human disease. Infect Immun. 2003;71:591–596. - PMC - PubMed
    1. Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: the unseen majority. Proc Natl Acad Sci U S A. 1998;95:6578–6583. - PMC - PubMed
    1. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–848. - PubMed
    1. Pace NR, Olsen GJ, Woese CR. Ribosomal RNA phylogeny and the primary lines of evolutionary descent. Cell. 1986;45:325–326. - PubMed
    1. Eckburg PB, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638. - PMC - PubMed
Show all 55 references
Publication types
MeSH terms
Substances
Related information
Grant support
Show all 19 grants
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 2
Figure 2
Antibiotic administration by gavage mimics molecular, anatomic, histologic, and immunologic characteristics of reduced microbial stimulation without animal dehydration. A) Weights of animals gavaged with unsupplemented (H2O; N=4) or antibiotic-supplemented (ABX; N=5) water (± s.e.m). B) 16S rDNA gene copies as quantified from stool pellets collected before (day 0) or over the course of antibiotic treatment (days 1, 5, 9) (N=5; ± s.e.m). C) Cecal images from control-treated (H2O), day 10 antibiotic-treated (ABX), or germ-free animals (GF) (bar, 1 cm). D) Photomicrographs of H&E stained cecal sections from control-treated (H2O), day 10 antibiotic-treated (ABX), or germ-free animals (GF) showing expansion of the lamina propria and enterocyte hyperplasia in ceca from antibiotic-treated and germ-free animals (bar, 50 μm). E) Quantification of cecal villus length and width from control-treated (H2O; N=4), day 10 antibiotic-treated (ABX; N=5), or germ-free animals (GF; N=3) (***, p

Figure 3

Global restructuring of intestinal bacterial…

Figure 3

Global restructuring of intestinal bacterial communities over time. A) Unweighted or B) weighted…

Figure 3
Global restructuring of intestinal bacterial communities over time. A) Unweighted or B) weighted UniFrac analysis of stool pellet, luminal content, and mucosal-associated samples from control-treated or antibiotic-treated animals. C) Unweighted or D) weighted UniFrac analysis of stool pellet samples from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals from day 1 to day 9 post treatment initiation. Colored symbols represent treatment day, white squares represent means, red line represents linear regression analysis (± s.e.m, significance determined by regression analysis). E) Genus level phylogenetic classification of 16S rDNA frequencies in stool pellets collected from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals from day 0 to day 9. F) Average frequency of bacterial groups before (Antibiotic Day 0) or during (Antibiotic Day 1, 9) antibiotic treatment. Mann-Whitney P-values of changes in group frequency with antibiotic treatment. Frequency reductions upon antibiotic treatment in red, increases in green, non-significant changes blank.

Figure 4

Antibiotics modify luminal and mucosal-associated…

Figure 4

Antibiotics modify luminal and mucosal-associated bacterial communities along the length of the colon.…

Figure 4
Antibiotics modify luminal and mucosal-associated bacterial communities along the length of the colon. A) Genus level phylogenetic classification of 16S rDNA frequencies in luminal samples collected from control-treated (H2O; N=4) or day 10 antibiotic-treated (ABX; N=5) animals from the cecum (Ce), proximal colon (Prox), or distal colon (Dist). B) Average frequency of bacterial groups in samples from control-treated (H2O; N=4) or antibiotic-treated (Antibiotic Day 10; N=5) animals. C) Genus level phylogenetic classification of 16S rDNA frequencies in mucosal-associated samples collected from control-treated (H2O; N=4) or day 10 antibiotic-treated (ABX; N=5) animals from the cecum (Ce), proximal colon (Prox), or distal colon (Dist). D) Average frequency of bacterial groups in mucosal-associated samples from control-treated (H2O; N=4) or antibiotic-treated (Antibiotic Day 10; N=5) animals. Mann-Whitney P-values of changes in group frequency with antibiotic treatment. Frequency reductions upon antibiotic treatment in red, increases in green, non-significant changes blank.

Figure 5

Antibiotic treatment alters mucosal CD4…

Figure 5

Antibiotic treatment alters mucosal CD4 + T lymphocyte homeostasis. A) Cytokine mRNA expression…

Figure 5
Antibiotic treatment alters mucosal CD4+ T lymphocyte homeostasis. A) Cytokine mRNA expression on day 10 as assessed by real-time RT-PCR of small intestine samples from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals (***, p<0.001; ± s.e.m). B) Expression of IFNγ or IL-17A by CD4+ T lymphocytes in the small intestinal lamina propria of control-treated (H2O) or day 10 antibiotic-treated (ABX) animals as analyzed by flow cytometry. C) Expression of IFNγ or IL-17A by CD4+ T lymphocytes in the mesenteric lymph nodes of control-treated (H2O) or day 10 antibiotic-treated (ABX) animals analyzed by flow cytometry. D) Statistical analysis of mesenteric lymph node CD4+ T lymphocyte cytokine expression from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals (*, p<0.05; ± s.e.m).
Figure 3
Figure 3
Global restructuring of intestinal bacterial communities over time. A) Unweighted or B) weighted UniFrac analysis of stool pellet, luminal content, and mucosal-associated samples from control-treated or antibiotic-treated animals. C) Unweighted or D) weighted UniFrac analysis of stool pellet samples from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals from day 1 to day 9 post treatment initiation. Colored symbols represent treatment day, white squares represent means, red line represents linear regression analysis (± s.e.m, significance determined by regression analysis). E) Genus level phylogenetic classification of 16S rDNA frequencies in stool pellets collected from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals from day 0 to day 9. F) Average frequency of bacterial groups before (Antibiotic Day 0) or during (Antibiotic Day 1, 9) antibiotic treatment. Mann-Whitney P-values of changes in group frequency with antibiotic treatment. Frequency reductions upon antibiotic treatment in red, increases in green, non-significant changes blank.
Figure 4
Figure 4
Antibiotics modify luminal and mucosal-associated bacterial communities along the length of the colon. A) Genus level phylogenetic classification of 16S rDNA frequencies in luminal samples collected from control-treated (H2O; N=4) or day 10 antibiotic-treated (ABX; N=5) animals from the cecum (Ce), proximal colon (Prox), or distal colon (Dist). B) Average frequency of bacterial groups in samples from control-treated (H2O; N=4) or antibiotic-treated (Antibiotic Day 10; N=5) animals. C) Genus level phylogenetic classification of 16S rDNA frequencies in mucosal-associated samples collected from control-treated (H2O; N=4) or day 10 antibiotic-treated (ABX; N=5) animals from the cecum (Ce), proximal colon (Prox), or distal colon (Dist). D) Average frequency of bacterial groups in mucosal-associated samples from control-treated (H2O; N=4) or antibiotic-treated (Antibiotic Day 10; N=5) animals. Mann-Whitney P-values of changes in group frequency with antibiotic treatment. Frequency reductions upon antibiotic treatment in red, increases in green, non-significant changes blank.
Figure 5
Figure 5
Antibiotic treatment alters mucosal CD4+ T lymphocyte homeostasis. A) Cytokine mRNA expression on day 10 as assessed by real-time RT-PCR of small intestine samples from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals (***, p<0.001; ± s.e.m). B) Expression of IFNγ or IL-17A by CD4+ T lymphocytes in the small intestinal lamina propria of control-treated (H2O) or day 10 antibiotic-treated (ABX) animals as analyzed by flow cytometry. C) Expression of IFNγ or IL-17A by CD4+ T lymphocytes in the mesenteric lymph nodes of control-treated (H2O) or day 10 antibiotic-treated (ABX) animals analyzed by flow cytometry. D) Statistical analysis of mesenteric lymph node CD4+ T lymphocyte cytokine expression from control-treated (H2O; N=4) or antibiotic-treated (ABX; N=5) animals (*, p<0.05; ± s.e.m).

References

    1. Eckburg PB, Lepp PW, Relman DA. Archaea and their potential role in human disease. Infect Immun. 2003;71:591–596.
    1. Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: the unseen majority. Proc Natl Acad Sci U S A. 1998;95:6578–6583.
    1. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–848.
    1. Pace NR, Olsen GJ, Woese CR. Ribosomal RNA phylogeny and the primary lines of evolutionary descent. Cell. 1986;45:325–326.
    1. Eckburg PB, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638.
    1. Palmer C, Bik EM, Digiulio DB, Relman DA, Brown PO. Development of the Human Infant Intestinal Microbiota. PLoS Biol. 2007;5:e177.
    1. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr. 2002;22:283–307.
    1. Backhed F, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101:15718–15723.
    1. Artis D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol. 2008;8:411–420.
    1. Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci U S A. 2002;99:15451–15455.
    1. Husebye E, Hellstrom PM, Midtvedt T. Intestinal microflora stimulates myoelectric activity of rat small intestine by promoting cyclic initiation and aboral propagation of migrating myoelectric complex. Dig Dis Sci. 1994;39:946–956.
    1. Cebra JJ. Influences of microbiota on intestinal immune system development. Am J Clin Nutr. 1999;69:1046S–1051S.
    1. Frank DN, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104:13780–13785.
    1. Bjorksten B, Sepp E, Julge K, Voor T, Mikelsaar M. Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol. 2001;108:516–520.
    1. Strober W, Fuss IJ, Blumberg RS. The immunology of mucosal models of inflammation. Annu Rev Immunol. 2002;20:495–549.
    1. Marra F, et al. Antibiotic use in children is associated with increased risk of asthma. Pediatrics. 2009;123:1003–1010.
    1. Hoban DJ. Antibiotics and collateral damage. Clin Cornerstone Suppl. 2003;3:S12–20.
    1. De La Cochetiere MF, et al. Effect of antibiotic therapy on human fecal microbiota and the relation to the development of Clostridium difficile. Microb Ecol. 2008;56:395–402.
    1. Hall JA, et al. Commensal DNA limits regulatory T cell conversion and is a natural adjuvant of intestinal immune responses. Immunity. 2008;29:637–649.
    1. Zaph C, et al. Commensal-dependent expression of IL-25 regulates the IL-23-IL-17 axis in the intestine. J Exp Med. 2008;205:2191–2198.
    1. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620–625.
    1. Stecher B, et al. Salmonella enterica serovar typhimurium exploits inflammation to compete with the intestinal microbiota. PLoS Biol. 2007;5:2177–2189.
    1. Sekirov I, et al. Antibiotic-induced perturbations of the intestinal microbiota alter host susceptibility to enteric infection. Infect Immun. 2008;76:4726–4736.
    1. Garner CD, et al. Perturbation of the small intestine microbial ecology by streptomycin alters pathology in a Salmonella enterica serovar typhimurium murine model of infection. Infect Immun. 2009;77:2691–2702.
    1. Croswell A, Amir E, Teggatz P, Barman M, Salzman NH. Prolonged impact of antibiotics on intestinal microbial ecology and susceptibility to enteric Salmonella infection. Infect Immun. 2009;77:2741–2753.
    1. Kang SS, et al. An antibiotic-responsive mouse model of fulminant ulcerative colitis. PLoS Med. 2008;5:e41.
    1. 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. 2004;118:229–241.
    1. Bashir ME, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. 2004;172:6978–6987.
    1. MILLER CP, BOHNHOFF M. Changes in the Mouse’s Enteric Microflora Associated with Enhanced Susceptibility to Salmonella Infection Following Streptomycin Treatment. J Infect Dis. 1963;113:59–66.
    1. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6:e280.
    1. Antonopoulos DA, et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun. 2009;77:2367–2375.
    1. Noverr MC, Noggle RM, Toews GB, Huffnagle GB. Role of antibiotics and fungal microbiota in driving pulmonary allergic responses. Infect Immun. 2004;72:4996–5003.
    1. Guseinov TS, Guseinova ST. Effect of dehydration on morphogenesis of the lymphatic network and immune structures in the small intestine. Bull Exp Biol Med. 2008;145:755–757.
    1. Rohde CL, Bartolini V, Jones N. The use of probiotics in the prevention and treatment of antibiotic-associated diarrhea with special interest in Clostridium difficile-associated diarrhea. Nutr Clin Pract. 2009;24:33–40.
    1. Thompson GR, Trexler PC. Gastrointestinal structure and function in germ-free or gnotobiotic animals. Gut. 1971;12:230–235.
    1. Wang ML, et al. Regulation of RELM/FIZZ isoform expression by Cdx2 in response to innate and adaptive immune stimulation in the intestine. Am J Physiol Gastrointest Liver Physiol. 2005;288:G1074–83.
    1. Lozupone CA, Hamady M, Kelley ST, Knight R. Quantitative and qualitative beta diversity measures lead to different insights into factors that structure microbial communities. Appl Environ Microbiol. 2007;73:1576–1585.
    1. Lozupone C, Hamady M, Knight R. UniFrac--an online tool for comparing microbial community diversity in a phylogenetic context. BMC Bioinformatics. 2006;7:371.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol. 2005;71:8228–8235.
    1. Zoetendal EG, et al. Mucosa-associated bacteria in the human gastrointestinal tract are uniformly distributed along the colon and differ from the community recovered from feces. Appl Environ Microbiol. 2002;68:3401–3407.
    1. Vael C, Nelen V, Verhulst SL, Goossens H, Desager KN. Early intestinal Bacteroides fragilis colonisation and development of asthma. BMC Pulm Med. 2008;8:19.
    1. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–1023.
    1. Turnbaugh PJ, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031.
    1. Grasselli E, et al. Evidence of horizontal gene transfer between human and animal commensal Escherichia coli strains identified by microarray. FEMS Immunol Med Microbiol. 2008;53:351–358.
    1. Karami N, et al. Transfer of an ampicillin resistance gene between two Escherichia coli strains in the bowel microbiota of an infant treated with antibiotics. J Antimicrob Chemother. 2007;60:1142–1145.
    1. Betsi GI, Papadavid E, Falagas ME. Probiotics for the treatment or prevention of atopic dermatitis: a review of the evidence from randomized controlled trials. Am J Clin Dermatol. 2008;9:93–103.
    1. Kozuch PL, Hanauer SB. Treatment of inflammatory bowel disease: a review of medical therapy. World J Gastroenterol. 2008;14:354–377.
    1. Ivanov II, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4:337–349.
    1. Atarashi K, et al. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455:808–812.
    1. Niess JH, Leithauser F, Adler G, Reimann J. Commensal gut flora drives the expansion of proinflammatory CD4 T cells in the colonic lamina propria under normal and inflammatory conditions. J Immunol. 2008;180:559–568.
    1. Artis D, et al. RELMbeta/FIZZ2 is a goblet cell-specific immune-effector molecule in the gastrointestinal tract. Proc Natl Acad Sci U S A. 2004;101:13596–13600.
    1. Ivanov II, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–1133.
    1. McKenna P, et al. The macaque gut microbiome in health, lentiviral infection, and chronic enterocolitis. PLoS Pathog. 2008;4:e20.
    1. Hugenholtz P. Exploring prokaryotic diversity in the genomic era. Genome Biol. 2002;3:REVIEWS0003.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73:5261–5267.

Source: PubMed

3
Suscribir