Recovery of the gut microbiome following fecal microbiota transplantation

Anna M Seekatz, Johannes Aas, Charles E Gessert, Timothy A Rubin, Daniel M Saman, Johan S Bakken, Vincent B Young, Anna M Seekatz, Johannes Aas, Charles E Gessert, Timothy A Rubin, Daniel M Saman, Johan S Bakken, Vincent B Young

Abstract

Clostridium difficile infection is one of the most common health care-associated infections, and up to 40% of patients suffer from recurrence of disease following standard antibiotic therapy. Recently, fecal microbiota transplantation (FMT) has been successfully used to treat recurrent C. difficile infection. It is hypothesized that FMT aids in recovery of a microbiota capable of colonization resistance to C. difficile. However, it is not fully understood how this occurs. Here we investigated changes in the fecal microbiota structure following FMT in patients with recurrent C. difficile infection, and imputed a hypothetical functional profile based on the 16S rRNA profile using a predictive metagenomic tool. Increased relative abundance of Bacteroidetes and decreased abundance of Proteobacteria were observed following FMT. The fecal microbiota of recipients following transplantation was more diverse and more similar to the donor profile than the microbiota prior to transplantation. Additionally, we observed differences in the imputed metagenomic profile. In particular, amino acid transport systems were overrepresented in samples collected prior to transplantation. These results suggest that functional changes accompany microbial structural changes following this therapy. Further identification of the specific community members and functions that promote colonization resistance may aid in the development of improved treatment methods for C. difficile infection.

Importance: Within the last decade, Clostridium difficile infection has surpassed other bacterial infections to become the leading cause of nosocomial infections. Antibiotic use, which disrupts the gut microbiota and its capability in providing colonization resistance against C. difficile, is a known risk factor in C. difficile infection. In particular, recurrent C. difficile remains difficult to treat with standard antibiotic therapy. Fecal microbiota transplantation (FMT) has provided a successful treatment method for some patients with recurrent C. difficile infection, but its mechanism and long-term effects remain unknown. Our results provide insight into the structural and potential metabolic changes that occur following FMT, which may aid in the development of new treatment methods for C. difficile infection.

Copyright © 2014 Seekatz et al.

Figures

FIG 1
FIG 1
Timeline of sample collection for each recipient-donor pair. Relative sample collection time, history of antibiotic use, and results for C. difficile clinical diagnostic tests (Illumigene assay) for the time of FMT for each recipient are indicated.
FIG 2
FIG 2
Relative abundances of most abundant OTUs in donor and pre- and post-FMT samples. Heat map of most abundant OTUs (>2% of all sequences), classified by phylum, family, and genus. Samples were clustered based on the Morisita-Horn distance matrix using the R package “vegan” and color coded by sample type (donor or pre- or post-FMT sample).
FIG 3
FIG 3
Estimated diversity in donor and pre- and post-FMT samples. The observed species richness (estimated number of OTUs) (A) or Shannon diversity index (B) for donor and pre- and post-FMT samples is shown. Each recipient-donor pair is color coded as indicated by the legend. Box plots specify the median (second), third, and fourth quartiles. The nonparametric Wilcoxon test was used to calculate significance among the different sample groups (*, P < 0.01; **, P < 0.001; ***, P < 0.0001).
FIG 4
FIG 4
Similarity between donor, pre-, and post-FMT samples. Shared species richness (number of shared OTUs) (A) or the Yue-Clayton theta similarity (θYC) (B) between pre-FMT and post-FMT (pre-post), donor and post-FMT (donor-post), and pre-FMT and donor (pre-donor) samples within recipient-donor pairs is shown. The nonparametric Wilcoxon test was used to calculate significance among the different sample groups (*, P < 0.01; **, P < 0.001; ***, P < 0.0001; ns, not significant).
FIG 5
FIG 5
Compositional comparison of donor, pre-, and post-FMT samples. Principal coordinates analysis (PCoA) of the theta similarity (θYC) for all samples, color coded by sample type (donor and pre- and post-FMT). The direction of 15 most abundant significantly correlated operational taxonomic units (OTUs) for axes 1 and 2 are labeled, as calculated using the Spearman correlation (P < 0.001).
FIG 6
FIG 6
Metagenomic functional predictions for donor, pre-, and post-FMT samples. Mean relative gene pathway abundance of significantly differentially abundant modules (ANOVA; P < 0.01) for donor and pre- and post-FMT samples. Gene pathway abundances were calculated using the HUMAnN pipeline and grouped by major functional categories.

References

    1. Ananthakrishnan AN. 2011. Clostridium difficile infection: epidemiology, risk factors and management. Nat. Rev. Gastroenterol. Hepatol. 8:17–26. 10.1038/nrgastro.2010.190
    1. Dubberke ER, Olsen MA. 2012. Burden of Clostridium difficile on the healthcare system. Clin. Infect. Dis. 55(Suppl 2):S88–S92. 10.1093/cid/cis335
    1. Cohen SH, Gerding DN, Johnson S, Kelly CP, Loo VG, McDonald LC, Pepin J, Wilcox MH, 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. 10.1086/651706
    1. Musher DM, Aslam S, Logan N, Nallacheru S, Bhaila I, Borchert F, Hamill RJ. 2005. Relatively poor outcome after treatment of Clostridium difficile colitis with metronidazole. Clin. Infect. Dis. 40:1586–1590. 10.1086/430311
    1. Kelly CP. 2012. Current strategies for management of initial Clostridium difficile infection. J. Hosp. Med. 7(Suppl 3):S5–S10. 10.1002/jhm.1909
    1. van der Waaij D. 1989. The ecology of the human intestine and its consequences for overgrowth by pathogens such as Clostridium difficile. Annu. Rev. Microbiol. 43:69–87. 10.1146/annurev.micro.43.1.69
    1. Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ. 2010. Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J. Clin. Gastroenterol. 44:354–360. 10.1097/MCG.0b013e3181c87e02
    1. Antharam VC, Li EC, Ishmael A, Sharma A, Mai V, Rand KH, Wang GP. 2013. Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J. Clin. Microbiol. 51:2884–2892. 10.1128/JCM.00845-13
    1. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, Schmidt TM, Young VB. 2008. Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea. J. Infect. Dis. 197:435–438. 10.1086/525047
    1. Theriot CM, Young VB. 2014. Microbial and metabolic interactions between the gastrointestinal tract and Clostridium difficile infection. Gut Microbes 5:86–95. 10.4161/gmic.27131
    1. van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, Visser CE, Kuijper EJ, Bartelsman JF, Tijssen JG, Speelman P, Dijkgraaf MG, Keller JJ. 2013. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 368:407–415. 10.1056/NEJMoa1205037
    1. Brandt LJ, Aroniadis OC. 2013. An overview of fecal microbiota transplantation: techniques, indications, and outcomes. Gastrointest. Endosc. 78:240–249. 10.1016/j.gie.2013.03.1329
    1. Yoon SS, Brandt LJ. 2010. Treatment of refractory/recurrent C. difficile-associated disease by donated stool transplanted via colonoscopy: a case series of 12 patients. J. Clin. Gastroenterol. 44:562–566. 10.1097/MCG.0b013e3181dac035
    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. 10.4161/gmic.23571
    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. 10.1371/journal.ppat.1002995
    1. Reeves AE, Koenigsknecht MJ, Bergin IL, Young VB. 2012. Suppression of Clostridium difficile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospiraceae. Infect. Immun. 80:3786–3794. 10.1128/IAI.00647-12
    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. 10.1152/ajpgi.00282.2013
    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. 10.1128/AEM.01541-09
    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. 10.1128/AEM.00062-07
    1. Langille MG, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, Huttenhower C. 2013. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat. Biotechnol. 31:814–821. 10.1038/nbt.2676
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, White O, Kelley ST, Methé B, Schloss PD, Gevers D, Mitreva M, Huttenhower C. 2012. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol. 8:e1002358. 10.1371/journal.pcbi.1002358
    1. Feehily C, Karatzas KA. 2013. Role of glutamate metabolism in bacterial responses towards acid and other stresses. J. Appl. Microbiol. 114:11–24. 10.1111/j.1365-2672.2012.05434.x
    1. Song Y, Garg S, Girotra M, Maddox C, von Rosenvinge EC, Dutta A, Dutta S, Fricke FW. 2013. Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection. PLoS One 8:e81330. 10.1371/journal.pone.0081330
    1. Angelberger S, Reinisch W, Makristathis A, Lichtenberger C, Dejaco C, Papay P, Novacek G, Trauner M, Loy A, Berry D. 2013. Temporal bacterial community dynamics vary among ulcerative colitis patients after fecal microbiota transplantation. Am. J. Gastroenterol. 108:1620–1630. 10.1038/ajg.2013.257
    1. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, LeLeiko N, Snapper SB, Bousvaros A, Korzenik J, Sands BE, Xavier RJ, Huttenhower C. 2012. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 13:R79. 10.1186/gb-2012-13-9-r79
    1. McHardy IH, Goudarzi M, Tong M, Ruegger PM, Schwager E, Weger JR, Graeber TG, Sonnenburg JL, Horvath S, Huttenhower C, McGovern DPB, Fornace AJ, Borneman J, Braun J. 2013. Integrative analysis of the microbiome and metabolome of the human intestinal mucosal surface reveals exquisite inter-relationships. Microbiome 1:17. 10.1186/2049-2618-1-17
    1. Karasawa T, Ikoma S, Yamakawa K, Nakamura S. 1995. A defined growth medium for Clostridium difficile. Microbiology 141:371–375. 10.1099/13500872-141-2-371
    1. Perez-Cobas AE, Artacho A, Knecht H, Ferrus ML, Friedrichs A, Ott SJ, Moya A, Latorre A, Gosalbes MJ. 2013. Differential effects of antibiotic therapy on the structure and function of human gut microbiota. PLoS One 8:e80201. 10.1371/journal.pone.0080201
    1. Theriot CM, Koenigsknecht MJ, Carlson PE, Hatton GE, Nelson AM, Li B, Huffnagle GB, Li JZ, Young VB. 2014. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increases susceptibility to Clostridium difficile infection. Nat. Commun 5:3114. 10.1038/ncomms4114
    1. Quera R, Espinoza R, Estay C, Rivera D. 2014. Bacteremia as an adverse event of fecal microbiota transplantation in a patient with Crohn’s disease and recurrent Clostridium difficile infection. J. Crohns Colitis 8:252–253. 10.1016/j.crohns.2013.10.002
    1. Schwartz M, Gluck M, Koon S. 2013. Norovirus gastroenteritis after fecal microbiota transplantation for treatment of Clostridium difficile infection despite asymptomatic donors and lack of sick contacts. Am. J. Gastroenterol. 108:1367. 10.1038/ajg.2013.164
    1. De Leon LM, Watson JB, Kelly CR. 2013. Transient flare of ulcerative colitis after fecal microbiota transplantation for recurrent Clostridium difficile infection. Clin. Gastroenterol. Hepatol. 11:1036–1038. 10.1016/j.cgh.2013.04.045
    1. Aas J, Gessert CE, Bakken JS. 2003. Recurrent Clostridium difficile colitis: case series involving 18 patients treated with donor stool administered via a nasogastric tube. Clin. Infect. Dis. 36:580–585. 10.1086/367657
    1. Hashway SA, Bergin IL, Bassis CM, Uchihashi M, Schmidt KC, Young VB, Aronoff DM, Patton DL, Bell JD. 2014. Impact of a hormone-releasing intrauterine system on the vaginal microbiome: a prospective baboon model. J. Med. Primatol. 43:89–99. 10.1111/jmp.12090
    1. Oksanen J, Blanchet FG, Kindt R, Legendre P, Minchin PR, O’Hara RB, Simpson GL, Solymos P, Stevens MHH, Wagner H. 2013. Vegan: Community Ecology R Package, v2.0-10.
    1. Warnes GR, Bolker B, Bonebakker L, Gentleman R, Liaw WHA, Lumley T, Maechler M, Magnusson A, Moeller S, Schwartz M, Venables B. 2013. Gplots: various R programming tools for plotting data, v2.12.1.
    1. Kuczynski J, Stombaugh J, Walters WA, Gonzalez A, Caporaso JG, Knight R. 2012. Using QIIME to analyze 16S rRNA gene sequences from microbial communities. Curr. Protoc. Microinform. 36:10.7.1–10.7.20. 10.1002/0471250953.bi1007s36 .
    1. Kanehisa M, Goto S. 2000. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 10.1093/nar/28.7.e27

Source: PubMed

3
Suscribir