Antibiotic-Induced Alterations of the Gut Microbiota Alter Secondary Bile Acid Production and Allow for Clostridium difficile Spore Germination and Outgrowth in the Large Intestine

Casey M Theriot, Alison A Bowman, Vincent B Young, Casey M Theriot, Alison A Bowman, Vincent B Young

Abstract

It is hypothesized that the depletion of microbial members responsible for converting primary bile acids into secondary bile acids reduces resistance to Clostridium difficile colonization. To date, inhibition of C. difficile growth by secondary bile acids has only been shown in vitro. Using targeted bile acid metabolomics, we sought to define the physiologically relevant concentrations of primary and secondary bile acids present in the murine small and large intestinal tracts and how these impact C. difficile dynamics. We treated mice with a variety of antibiotics to create distinct microbial and metabolic (bile acid) environments and directly tested their ability to support or inhibit C. difficile spore germination and outgrowth ex vivo. Susceptibility to C. difficile in the large intestine was observed only after specific broad-spectrum antibiotic treatment (cefoperazone, clindamycin, and vancomycin) and was accompanied by a significant loss of secondary bile acids (deoxycholate, lithocholate, ursodeoxycholate, hyodeoxycholate, and ω-muricholate). These changes were correlated to the loss of specific microbiota community members, the Lachnospiraceae and Ruminococcaceae families. Additionally, physiological concentrations of secondary bile acids present during C. difficile resistance were able to inhibit spore germination and outgrowth in vitro. Interestingly, we observed that C. difficile spore germination and outgrowth were supported constantly in murine small intestinal content regardless of antibiotic perturbation, suggesting that targeting growth of C. difficile will prove most important for future therapeutics and that antibiotic-related changes are organ specific. Understanding how the gut microbiota regulates bile acids throughout the intestine will aid the development of future therapies for C. difficile infection and other metabolically relevant disorders such as obesity and diabetes. IMPORTANCE Antibiotics alter the gastrointestinal microbiota, allowing for Clostridium difficile infection, which is a significant public health problem. Changes in the structure of the gut microbiota alter the metabolome, specifically the production of secondary bile acids. Specific bile acids are able to initiate C. difficile spore germination and also inhibit C. difficile growth in vitro, although no study to date has defined physiologically relevant bile acids in the gastrointestinal tract. In this study, we define the bile acids C. difficile spores encounter in the small and large intestines before and after various antibiotic treatments. Antibiotics that alter the gut microbiota and deplete secondary bile acid production allow C. difficile colonization, representing a mechanism of colonization resistance. Multiple secondary bile acids in the large intestine were able to inhibit C. difficile spore germination and growth at physiological concentrations and represent new targets to combat C. difficile in the large intestine.

Keywords: Clostridium difficile; antibiotics; bile acids; colonization resistance; microbiota.

Figures

FIG 1
FIG 1
Antibiotic treatment scheme and experimental design. (A) C57BL/6 mice were treated with various antibiotics that would result in different microbial and metabolic environments. Red circles represent the time of necropsy for each group (n = 7 to 10 mice per group). (B) At the time of necropsy, ileal and cecal contents were collected. Ex vivo germination and outgrowth of C. difficile spores were measured in paired ileal and cecal contents from all treatment groups as well as by targeted bile acid analysis. In vitro spore germination and growth studies were done using relevant in vivo ileal and cecal bile acid concentrations. Microbiome analysis was also done to understand the relationship between gut bacteria and bile acids using correlation analysis. Abbreviations: noabx, no antibiotic; cef, cefoperazone; 1wk to 6wk, number of weeks off cefoperazone; 6wkC, 6 weeks off cefoperazone plus an intraperitoneal administration (IP) of clindamycin; vanco, vancomycin; metro, metronidazole; kana, kanamycin; clinda, clindamycin.
FIG 2
FIG 2
C. difficile ex vivo spore germination and outgrowth assays in murine ileal and cecal contents. Ex vivo germination and outgrowth of C. difficile spores were done in ileal (A) and cecal (B) contents collected from mice treated with various antibiotics. C. difficile VPI 10463 spores inoculated into the ileal contents of mice treated with or without antibiotics (noabx) allowed for spore germination and outgrowth after a 6-h period, whereas spores in non-antibiotic-treated cecal content did not. Only specific antibiotic treatments (cefoperazone [cef], 1 or 2 weeks off cefoperazone, 6 weeks off cefoperazone plus an intraperitoneal administration of clindamycin [6wkC], clindamycin [clinda], and vancomycin [vanco]) in the cecum supported spore germination and outgrowth. Black bars represent spores only, and gray bars represent spores and vegetative cells. Significance between groups was determined by Mann-Whitney nonparametric t test. Error bars represent the mean ± standard error of the mean (SEM) (*, P < 0.05; **, P < 0.01). n.s. not significant.
FIG 3
FIG 3
Targeted bile acid metabolomics of murine ileal and cecal contents. Bile acids were analyzed by LC-MS from paired ileal (A) and cecal (B) contents from ex vivo spore germination and outgrowth assays. A heat map shows the bile acid concentration present in micrograms per 100 mg of gut content, ranging from 1 to 120,000. The black boxes represent samples that did not reach significance in supporting spore germination and outgrowth of C. difficile spores in Fig. 2. (C) Bile acids present in cecal content that did not support spore germination and outgrowth in the black bars (resistance) or were able to support germination and outgrowth (susceptible) are compared. Significance between groups was determined by Mann-Whitney nonparametric t test. Error bars represent the mean ± SEM (****, P < 0.0001).
FIG 4
FIG 4
Secondary bile acids present during C. difficile resistance inhibit spore germination. In vitro spore germination inhibition assays were performed with C. difficile strain VPI 10463 to assess if the secondary bile acids ω-muricholate (ωMCA), hyodeoxycholate (HDCA), ursodeoxycholate (UDCA), lithocholate (LCA), and deoxycholate (DCA) were able to inhibit spore germination with known germinants TCA and DCA. Spores were incubated for 30 min in (A) BHI plus TCA (0.1%) or (B) BHI plus DCA (0.1%) supplemented with a range of secondary bile acids at relevant in vivo concentrations. Positive controls include BHI plus TCA (0.1%) or DCA (0.1%) alone with mock H2O and mock ethanol (EthOH) (red bars). A negative control was also used: BHI plus TCA supplemented with CDCA (0.04%) or BHI plus DCA supplemented with CDCA (0.04%), a known inhibitor of spore germination (blue bar). Negative controls include BHI alone without the addition of TCA or DCA. The data presented represent the mean ± standard deviation (SD) from triplicate experiments and were significant compared to the positive controls (A) TCA alone or (B) DCA alone (Student’s t test, *, P < 0.05; **, P < 0.01; ***, P < 0.001).
FIG 5
FIG 5
Secondary bile acids present during C. difficile resistance inhibit growth of C. difficile. (A) Growth rates (per hour) are shown. C. difficile was grown in BHI medium supplemented with various secondary bile acids with a range of in vivo concentrations (0.001% to 0.1%) present during resistance to C. difficile. The data presented represent the means ± SD from triplicate experiments and were significant compared to positive controls C. difficile and C. difficile with mock ethanol (EthOH) without bile acids (Students t test, **, P < 0.05; ***, P < 0.01). (B) Growth curve of representative secondary bile acids that significantly decreased the growth rate of C. difficile in panel A.
FIG 6
FIG 6
Correlation analysis of the gut microbiome and bile acids. Spearman’s correlation analysis was done with all 121 OTU (i.e., OTU that were greater than 1% of the total bacterial population) in the microbiome, color coded by phylum and grouped based on unsupervised clustering. All 26 bile acids detected were similarly clustered and are color coded based on structure. There were three distinct clusters of OTU (O1 to O3) and two distinct clusters of bile acids (B1 and B2). The heat map scale ranges from positively correlated, +0.7, to negatively correlated, −0.7.

References

    1. Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, Farley MM, Holzbauer SM, Meek JI, Phipps EC, Wilson LE, Winston LG, Cohen JA, Limbago BM, Fridkin SK, Gerding DN, McDonald LC. 2015. Burden of Clostridium difficile infection in the United States. N Engl J Med 372:825–834. doi:10.1056/NEJMoa1408913.
    1. Figueroa I, Johnson S, Sambol SP, Goldstein EJC, Citron DM, Gerding DN. 2012. Relapse versus reinfection: recurrent Clostridium difficile infection following treatment with fidaxomicin or vancomycin. Clin Infect Dis 55(Suppl 2):S104–S109. doi:10.1093/cid/cis357.
    1. Louie TJ, Miller MA, Mullane KM, Weiss K, Lentnek A, Golan Y, Gorbach S, Sears P, Shue Y. 2011. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med 364:422–431. doi:10.1056/NEJMoa0910812.
    1. Zar FA, Bakkanagari SR, Moorthi KM, Davis MB. 2007. A comparison of vancomycin and metronidazole for the treatment of Clostridium difficile-associated diarrhea, stratified by disease severity. Clin Infect Dis 45:302–307. doi:10.1086/519265.
    1. Cohen SH, Gerding DN, Kelly CP, Loo VG, McDonald LC, Pepin J, Wilcox M, Society for Healthcare Epidemiology of America, Infectious Diseases Society of America . 2010. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the Society for Healthcare Epidemiology of America (SHEA) and the Infectious Diseases Society of America (IDSA). Infect Control Hosp Epidemiol 31:431–455. doi:10.1086/651706.
    1. McFee RB. 2009. Clostridium difficile: emerging public health threat and other nosocomial or hospital acquired infections. Introduction. Dis Mon 55:419–421. doi:10.1016/j.disamonth.2009.03.013.
    1. Pepin J, Saheb N, Coulombe MA, Alary ME, Corriveau MP, Authier S, Leblanc M, Rivard G, Bettez M, Primeau V, Nguyen M, Jacob CE, Lanthier L. 2005. Emergence of fluoroquinolones as the predominant risk factor for Clostridium difficile-associated diarrhea: a cohort study during an epidemic in Quebec. Clin Infect Dis 41:1254–1260. doi:10.1086/496986.
    1. Reeves AE, Theriot CM, Bergin IL, Huffnagle GB, Schloss PD, Young VB. 2011. The interplay between microbiome dynamics and pathogen dynamics in a murine model of Clostridium difficile infection. Gut Microbes 2:145–158. doi:10.4161/gmic.2.3.16333.
    1. Buffie CG, Jarchum I, Equinda M, Lipuma L, Gobourne A, Viale A, Ubeda C, Xavier J, Pamer EG. 2012. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infect Immun 80:62–73. doi:10.1128/IAI.05496-11.
    1. Bassis CM, Theriot CM, Young VB. 2014. Alteration of the murine gastrointestinal microbiota by tigecycline leads to increased susceptibility to Clostridium difficile infection. Antimicrob Agents Chemother 58:2767–2774. doi:10.1128/AAC.02262-13.
    1. Antunes LC, Han J, Ferreira RB, Lolic P, Borchers CH, Finlay BB. 2011. The effect of antibiotic treatment on the intestinal metabolome. Antimicrob Agents Chemother 55:1494–1503. doi:10.1128/AAC.01664-10.
    1. Theriot CM, Koenigsknecht MJ, Carlson PE Jr, Hatton GE, Nelson AM, Li B, Huffnagle GB, Li JZ, Young VB. 2014. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat Commun 5:3114. doi:10.1038/ncomms4114.
    1. Vrieze A, Out C, Fuentes S, Jonker L, Reuling I, Kootte RS, van Nood E, Holleman F, Knaapen M, Romijn JA, Soeters MR, Blaak EE, Dallinga-Thie GM, Reijnders D, Ackermans MT, Serlie MJ, Knop FK, Holst JJ, van der Ley C, Kema IP, Zoetendal EG, de Vos WM, Hoekstra JB, Stroes ES, Groen AK, Nieuwdorp M. 2014. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol 60:824–831. doi:10.1016/j.jhep.2013.11.034.
    1. Sayin S, Wahlström A, Felin J, Jäntti S, Marschall H, Bamberg K, Angelin B, Hyötyläinen T, Orešič M, Bäckhed F. 2013. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab 17:225–235. doi:10.1016/j.cmet.2013.01.003.
    1. Yap IKS, Li JV, Saric J, Martin F, Davies H, Wang Y, Wilson ID, Nicholson JK, Utzinger J, Marchesi JR, Holmes E. 2008. Metabonomic and microbiological analysis of the dynamic effect of vancomycin-induced gut microbiota modification in the mouse. J Proteome Res 7:3718–3728. doi:10.1021/pr700864x.
    1. Ridlon JM, Kang D-J, Hylemon PB. 2006. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47:241–259. doi:10.1194/jlr.R500013-JLR200.
    1. Chiang JYL. 2009. Bile acids: regulation of synthesis. J Lipid Res 50:1955–1966. doi:10.1194/jlr.R900010-JLR200.
    1. Russell DW. 2003. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem 72:137–174. doi:10.1146/annurev.biochem.72.121801.161712.
    1. Claus SP, Ellero SL, Berger B, Krause L, Bruttin A, Molina J, Paris A, Want EJ, de Waziers I, Cloarec O, Richards SE, Wang Y, Dumas ME, Ross A, Rezzi S, Kochhar S, Van Bladeren P, Lindon JC, Holmes E, Nicholson JK. 2011. Colonization-induced host-gut microbial metabolic interaction. mBio 2:e00271–e00210. doi:10.1128/mBio.00271-10.
    1. Midtvedt T. 1974. Microbial bile acid transformation. Am J Clin Nutr 27:1341–1347.
    1. Kuipers F, Bloks VW, Groen AK. 2014. Beyond intestinal soap—bile acids in metabolic control. Nat Rev Endocrinol 10:488–498. doi:10.1038/nrendo.2014.60.
    1. Sorg JA, Sonenshein AL. 2008. Bile salts and glycine as cogerminants for Clostridium difficile spores. J Bacteriol 190:2505–2512. doi:10.1128/JB.01765-07.
    1. Wilson KH, Kennedy MJ, Fekety FR. 1982. Use of sodium taurocholate to enhance spore recovery on a medium selective for Clostridium difficile. J Clin Microbiol 15:443–446.
    1. Karlsson S, Lindberg A, Norin E, Burman LG, Akerlund T. 2000. Toxins, butyric acid, and other short-chain fatty acids are coordinately expressed and down-regulated by cysteine in Clostridium difficile. Infect Immun 68:5881–5888. doi:10.1128/IAI.68.10.5881-5888.2000.
    1. Sorg JA, Sonenshein AL. 2010. Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid. J Bacteriol 192:4983–4990. doi:10.1128/JB.00610-10.
    1. Francis MB, Allen CA, Sorg JA. 2013. Muricholic acids inhibit Clostridium difficile spore germination and growth. PLoS One 8:e73653. doi:10.1371/journal.pone.0073653.
    1. Sorg J. 2014. Microbial bile acid metabolic clusters: the bouncers at the bar. Cell Host Microbe 16:551–552. doi:10.1016/j.chom.2014.10.015.
    1. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, No D, Liu H, Kinnebrew M, Viale A, Littmann E, van den Brink MR, Jenq RR, Taur Y, Sander C, Cross JR, Toussaint NC, Xavier JB, Pamer EG. 2015. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517:205–208. doi:10.1038/nature13828.
    1. Weingarden AR, Chen C, Bobr A, Yao D, Lu Y, Nelson VM, Sadowsky MJ, Khoruts A. 2014. Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficile infection. Am J Physiol Gastrointest Liver Physiol 306:G310–G319. doi:10.1152/ajpgi.00282.2013.
    1. Wilson KH. 1983. Efficiency of various bile salt preparations for stimulation of Clostridium difficile spore germination. J Clin Microbiol 18:1017–1019.
    1. Koenigsknecht MJ, Theriot CM, Bergin IL, Schumacher CA, Schloss PD, Young VB. 2015. Dynamics and establishment of Clostridium difficile infection in the murine gastrointestinal tract. Infect Immun 83:934–941. doi:10.1128/IAI.02768-14.
    1. Sorg JA, Sonenshein AL. 2009. Chenodeoxycholate is an inhibitor of Clostridium difficile spore germination. J Bacteriol 191:1115–1117. doi:10.1128/JB.01260-08.
    1. Howerton A, Ramirez N, Abel-Santos E. 2011. Mapping interactions between germinants and Clostridium difficile spores. J Bacteriol 193:274–282. doi:10.1128/JB.00980-10.
    1. Hagio M, Matsumoto M, Fukushima M, Hara H, Ishizuka S. 2009. Improved analysis of bile acids in tissues and intestinal contents of rats using LC/ESI-MS. J Lipid Res 50:173–180. doi:10.1194/jlr.D800041-JLR200.
    1. Giel JL, Sorg JA, Sonenshein AL, Zhu J. 2010. Metabolism of bile salts in mice influences spore germination in Clostridium difficile. PLoS One 5:e8740. doi:10.1371/journal.pone.0008740.
    1. Wilson KH, Sheagren JN, Freter R. 1985. Population dynamics of ingested Clostridium difficile in the gastrointestinal tract of the Syrian hamster. J Infect Dis 151:355–361. doi:10.1093/infdis/151.2.355.
    1. Han J, Liu Y, Wang R, Yang J, Ling V, Borchers CH. 2015. Metabolic profiling of bile acids in human and mouse blood by LC-MS/MS in combination with phospholipid-depletion solid-phase extraction. Anal Chem 87:1127–1136. doi:10.1021/ac503816u.
    1. He G, Shankar RA, Chzhan M, Samouilov A, Kuppusamy P, Zweier JL. 1999. Noninvasive measurement of anatomic structure and intraluminal oxygenation in the gastrointestinal tract of living mice with spatial and spectral EPR imaging. Proc Natl Acad Sci U S A 96:4586–4591. doi:10.1073/pnas.96.8.4586.
    1. Crompton DW, Shrimpton DH, Silver IA. 1965. Measurements of the oxygen tension in the lumen of the small intestine of the domestic duck. J Exp Biol 43:473–478.
    1. Rogers WP. 1949. On the relative importance of aerobic metabolism in small nematode parasites of the alimentary tract: oxygen tensions in the normal environment of the parasites. Aust J Biol Sci 2:157–165.
    1. Owens R, Donskey C, Gaynes R, Loo V, Muto C. 2008. Antimicrobial-associated risk factors for Clostridium difficile infection. Clin Infect Dis 46(Suppl 1):S19–S31. doi:10.1086/521859.
    1. Freeman J, Wilcox MH. 1999. Antibiotics and Clostridium difficile. Microbes Infect 1:377–384. doi:10.1016/S1286-4579(99)80054-9.
    1. Zhang Y, Limaye PB, Renaud HJ, Klaassen CD. 2014. Effect of various antibiotics on modulation of intestinal microbiota and bile acid profile in mice. Toxicol Appl Pharmacol 277:138–145. doi:10.1016/j.taap.2014.03.009.
    1. Lewis BB, Buffie CG, Carter RA, Leiner I, Toussaint NC, Miller LC, Gobourne A, Ling L, Pamer EG. 2015. Loss of microbiota-mediated colonization resistance to Clostridium difficile infection with oral vancomycin compared with metronidazole. J Infect Dis 212:1656–1665. doi:10.1093/infdis/jiv256.
    1. Schubert AM, Sinani H, Schloss PD. 2015. Antibiotic-induced alterations of the murine gut microbiota and subsequent effects on colonization resistance against Clostridium difficile. mBio 6:e00974. doi:10.1128/mBio.00974-15.
    1. Wells JE, Hylemon PB. 2000. Identification and characterization of a bile acid 7alpha-dehydroxylation operon in Clostridium sp. strain TO-931, a highly active 7alpha-dehydroxylating strain isolated from human feces. Appl Environ Microbiol 66:1107–1113. doi:10.1128/AEM.66.3.1107-1113.2000.
    1. Mallonee DH, White WB, Hylemon PB. 1990. Cloning and sequencing of a bile acid-inducible operon from Eubacterium sp. strain vpi 12708. J Bacteriol 172:7011–7019.
    1. Ridlon JM, Kang D, Hylemon PB. 2010. Isolation and characterization of a bile acid inducible 7alpha-dehydroxylating operon in Clostridium hylemonae TN271. Anaerobe 16:137–146. doi:10.1016/j.anaerobe.2009.05.004.
    1. Lawley TD, Clare S, Walker AW, Stares MD, Connor TR, Raisen C, Goulding D, Rad R, Schreiber F, Brandt C, Deakin LJ, Pickard DJ, Duncan SH, Flint HJ, Clark TG, Parkhill J, Dougan G. 2012. Targeted restoration of the intestinal microbiota with a simple, defined bacteriotherapy resolves relapsing Clostridium difficile disease in mice. PLoS Pathog 8:e1002995. doi:10.1371/journal.ppat.1002995.
    1. Bernstein H, Bernstein C, Payne C, Dvorakova K, Garewal H. 2005. Bile acids as carcinogens in human gastrointestinal cancers. Mutat Res 589:47–65. doi:10.1016/j.mrrev.2004.08.001.
    1. McGarr SE, Ridlon JM, Hylemon PB. 2005. Diet, anaerobic bacterial metabolism, and colon cancer: a review of the literature. J Clin Gastroenterol 39:98–109.
    1. Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, Takei H, Muto A, Nittono H, Ridlon JM, White MB, Noble NA, Monteith P, Fuchs M, Thacker LR, Sikaroodi M, Bajaj JS. 2013. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 58:949–955. doi:10.1016/j.jhep.2013.01.003.
    1. Elkins CA, Moser SA, Savage DC. 2001. Genes encoding bile salt hydrolases and conjugated bile salt transporters in Lactobacillus johnsonii 100–100 and other Lactobacillus species. Microbiology 147:3403–3412. doi:10.1099/00221287-147-12-3403.
    1. Jones BV, Begley M, Hill C, Gahan CGM, Marchesi JR. 2008. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc Natl Acad Sci U S A 105:13580–13585. doi:10.1073/pnas.0804437105.
    1. Hamilton MJ, Weingarden AR, Unno T, Khoruts A, Sadowsky MJ. 2013. High-throughput DNA sequence analysis reveals stable engraftment of gut microbiota following transplantation of previously frozen fecal bacteria. Gut Microbes 4:125–135. doi:10.4161/gmic.23571.
    1. Carlson PE Jr, Walk ST, Bourgis AE, Liu MW, Kopliku F, Lo E, Young VB, Aronoff DM, Hanna PC. 2013. The relationship between phenotype, ribotype, and clinical disease in human Clostridium difficile isolates. Anaerobe 24:109–116. doi:10.1016/j.anaerobe.2013.04.003.
    1. Heeg D, Burns DA, Cartman ST, Minton NP. 2012. Spores of Clostridium difficile clinical isolates display a diverse germination response to bile salts. PLoS One 7:e32381. doi:10.1371/journal.pone.0032381.
    1. Kohli R, Myronovych A, Tan BK, Salazar-Gonzalez R, Miles L, Zhang W, Oehrle M, Sandoval DA, Ryan KK, Seeley RJ, Setchell KDR. 2015. Bile acid signaling: mechanism for bariatric surgery, cure for NASH? Dig Dis 33:440–446. doi:10.1159/000371699.
    1. National Institutes of Health 2002. Public Health Service policy on humane care and use of laboratory animals. Office of Laboratory Animal Welfare, National Institutes of Health, Bethesda, MD.
    1. Theriot CM, Koumpouras CC, Carlson PE, Bergin II, Aronoff DM, Young VB. 2011. Cefoperazone-treated mice as an experimental platform to assess differential virulence of Clostridium difficile strains. Gut Microbes 2:326–334. doi:10.4161/gmic.19142.
    1. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. 2013. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol 79:5112–5120. doi:10.1128/AEM.01043-13.
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ, Sahl JW, Stres B, Thallinger GG, Van Horn DJ, Weber CF. 2009. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol 75:7537–7541. doi:10.1128/AEM.01541-09.
    1. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, Peplies J, Glockner FO. 2007. Silva: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res 35:7188–7196. doi:10.1093/nar/gkm864.
    1. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glockner FO. 2013. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res 41:D590–D596. doi:10.1093/nar/gks1219.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. 2007. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol 73:5261–5267. doi:10.1128/AEM.00062-07.
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. 2011. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27:2194–2200. doi:10.1093/bioinformatics/btr381.

Source: PubMed

3
S'abonner