Longitudinal microbiome analysis of single donor fecal microbiota transplantation in patients with recurrent Clostridium difficile infection and/or ulcerative colitis

Michael Mintz, Shanawaj Khair, Suman Grewal, Joseph F LaComb, Jiyhe Park, Breana Channer, Ramona Rajapakse, Juan Carlos Bucobo, Jonathan M Buscaglia, Farah Monzur, Anupama Chawla, Jie Yang, Charlie E Robertson, Daniel N Frank, Ellen Li, Michael Mintz, Shanawaj Khair, Suman Grewal, Joseph F LaComb, Jiyhe Park, Breana Channer, Ramona Rajapakse, Juan Carlos Bucobo, Jonathan M Buscaglia, Farah Monzur, Anupama Chawla, Jie Yang, Charlie E Robertson, Daniel N Frank, Ellen Li

Abstract

Background: Studies of colonoscopic fecal microbiota transplant (FMT) in patients with recurrent CDI, indicate that this is a very effective treatment for preventing further relapses. In order to provide this service at Stony Brook University Hospital, we initiated an open-label prospective study of single colonoscopic FMT among patients with ≥ 2 recurrences of CDI, with the intention of monitoring microbial composition in the recipient before and after FMT, as compared with their respective donor. We also initiated a concurrent open label prospective trial of single colonoscopic FMT of patients with ulcerative colitis (UC) not responsive to therapy, after obtaining an IND permit (IND 15642). To characterize how FMT alters the fecal microbiota in patients with recurrent Clostridia difficile infections (CDI) and/or UC, we report the results of a pilot microbiome analysis of 11 recipients with a history of 2 or more recurrences of C. difficile infections without inflammatory bowel disease (CDI-only), 3 UC recipients with recurrent C. difficile infections (CDI + UC), and 5 UC recipients without a history of C. difficile infections (UC-only).

Method: V3V4 Illumina 16S ribosomal RNA (rRNA) gene sequencing was performed on the pre-FMT, 1-week post-FMT, and 3-months post-FMT recipient fecal samples along with those collected from the healthy donors. Fitted linear mixed models were used to examine the effects of Group (CDI-only, CDI + UC, UC-only), timing of FMT (Donor, pre-FMT, 1-week post-FMT, 3-months post-FMT) and first order Group*FMT interactions on the diversity and composition of fecal microbiota. Pairwise comparisons were then carried out on the recipient vs. donor and between the pre-FMT, 1-week post-FMT, and 3-months post-FMT recipient samples within each group.

Results: Significant effects of FMT on overall microbiota composition (e.g., beta diversity) were observed for the CDI-only and CDI + UC groups. Marked decreases in the relative abundances of the strictly anaerobic Bacteroidetes phylum, and two Firmicutes sub-phyla associated with butyrate production (Ruminococcaceae and Lachnospiraceae) were observed between the CDI-only and CDI + UC recipient groups. There were corresponding increases in the microaerophilic Proteobacteria phylum and the Firmicutes/Bacilli group in the CDI-only and CDI + UC recipient groups. At a more granular level, significant effects of FMT were observed for 81 genus-level operational taxonomic units (OTUs) in at least one of the three recipient groups (p<0.00016 with Bonferroni correction). Pairwise comparisons of the estimated pre-FMT recipient/donor relative abundance ratios identified 6 Gammaproteobacteria OTUs, including the Escherichia-Shigella genus, and 2 Fusobacteria OTUs with significantly increased relative abundance in the pre-FMT samples of all three recipient groups (FDR < 0.05), however the magnitude of the fold change was much larger in the CDI-only and CDI + UC recipients than in the UC-only recipients. Depletion of butyrate producing OTUs, such as Faecalibacterium, in the CDI-only and CDI + UC recipients, were restored after FMT.

Conclusion: The results from this pilot study suggest that the microbial imbalances in the CDI + UC recipients more closely resemble those of the CDI-only recipients than the UC-only recipients.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT flow diagram.
Fig 1. CONSORT flow diagram.
Fig 2. Phyla/subphyla comparison of pre-FMT samples…
Fig 2. Phyla/subphyla comparison of pre-FMT samples of the CDI-only, UC +CDI, and UC-only recipient groups with the donor samples.
The average relative abundance of each of the phyla/subphyla groups is shown for the donor samples and the pre-FMT samples of the CDI-only, CDI + UC and UC-only recipient groups.
Fig 3. Principal coordinate analysis (PCoA) plots…
Fig 3. Principal coordinate analysis (PCoA) plots for Bray-Curtis distances (beta-diversity).
(A) CDI-only recipient group: Donors (), pre-FMT recipient (), 1-wk. post-FMT recipient (), 3-mos. post-FMT recipient (); (B) CDI + UC recipient group: Donors (♦), pre-FMT recipient (♦), 1-wk. post-FMT recipient (♦), 3-mos. post-FMT recipient (♦); (C) UC-only recipient group: Donors (●), pre-FMT recipient (●), 1-wk. post-FMT recipient (●), 3-mos. post-FMT recipient (●).

References

    1. Kelly CP, LaMont JT. Clostridium difficile—More difficult than ever. New Engl J Med. 2008;359(18):1932–40. doi:
    1. Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, et al. Burden of Clostridium difficile infection in the United States. The New England journal of medicine. 2015;372(9):825–34. doi: .
    1. Rao K, Higgins PDR. Epidemiology, Diagnosis, and Management of Clostridium difficile Infection in Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22(7):1744–54. doi:
    1. Nelson RL, Suda KJ, Evans CT. Antibiotic treatment for Clostridium difficile-associated diarrhoea in adults. Cochrane Db Syst Rev. 2017;(3). doi:
    1. Garey KW, Sethi S, Yadav Y, DuPont HL. Meta-analysis to assess risk factors for recurrent Clostridium difficile infection. The Journal of hospital infection. 2008;70(4):298–304. doi: .
    1. Johnson S. Recurrent Clostridium difficile infection: a review of risk factors, treatments, and outcomes. The Journal of infection. 2009;58(6):403–10.
    1. McFarland LV, Elmer GW, Surawicz CM. Breaking the cycle: treatment strategies for 163 cases of recurrent Clostridium difficile disease. The American journal of gastroenterology. 2002;97(7):1769–75. doi: .
    1. Olsen MA, Yan Y, Reske KA, Zilberberg MD, Dubberke ER. Recurrent Clostridium difficile infection is associated with increased mortality. Clinical microbiology and infection: the official publication of the European Society of Clinical Microbiology and Infectious Diseases. 2015;21(2):164–70.
    1. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, Schmidt TM, et al. Decreased diversity of the fecal Microbiome in recurrent Clostridium difficile-associated diarrhea. The Journal of infectious diseases. 2008;197(3):435–8. doi: .
    1. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(34):13780–5. doi: .
    1. Li E, Hamm CM, Gulati AS, Sartor RB, Chen H, Wu X, et al. Inflammatory bowel diseases phenotype, C. difficile and NOD2 genotype are associated with shifts in human ileum associated microbial composition. PloS one. 2012;7(6):e26284 doi: .
    1. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M, Zheng Z, et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010;139(6):1844–54 e1. doi: .
    1. Yoon SS, Brandt LJ. Treatment of refractory/recurrent C. difficile-associated disease by donated stool transplanted via colonoscopy: a case series of 12 patients. Journal of clinical gastroenterology. 2010;44(8):562–6. doi: .
    1. Bakken JS, Borody T, Brandt LJ, Brill JV, Demarco DC, Franzos MA, et al. Treating Clostridium difficile infection with fecal microbiota transplantation. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2011;9(12):1044–9. doi: .
    1. Mattila E, Uusitalo-Seppala R, Wuorela M, Lehtola L, Nurmi H, Ristikankare M, et al. Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection. Gastroenterology. 2012;142(3):490–6. doi: .
    1. Paine ER. Colonoscopic evaluation in ulcerative colitis. Gastroenterology report. 2014;2(3):161–8. doi: .
    1. Frank DN. BARCRAWL and BARTAB: software tools for the design and implementation of barcoded primers for highly multiplexed DNA sequencing. BMC bioinformatics. 2009;10:362 doi: .
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nature methods. 2012;9(4):357–9. doi: .
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011;27(16):2194–200. doi: .
    1. Schloss PD, Westcott SL. Assessing and improving methods used in operational taxonomic unit-based approaches for 16S rRNA gene sequence analysis. Applied and environmental microbiology. 2011;77(10):3219–26. doi: .
    1. Pruesse E, Peplies J, Glockner FO. SINA: accurate high-throughput multiple sequence alignment of ribosomal RNA genes. Bioinformatics. 2012;28(14):1823–9. doi: .
    1. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic acids research. 2013;41(Database issue):D590–6. doi: .
    1. Robertson CE, Harris JK, Wagner BD, Granger D, Browne K, Tatem B, et al. Explicet: graphical user interface software for metadata-driven management, analysis and visualization of microbiome data. Bioinformatics. 2013;29(23):3100–1. doi: .
    1. Anderson MJ, Crist TO, Chase JM, Vellend M, Inouye BD, Freestone AL, et al. Navigating the multiple meanings of beta diversity: a roadmap for the practicing ecologist. Ecology letters. 2011;14(1):19–28. doi: .
    1. Son JS, Zheng LJ, Rowehl LM, Tian X, Zhang Y, Zhu W, et al. Comparison of Fecal Microbiota in Children with Autism Spectrum Disorders and Neurotypical Siblings in the Simons Simplex Collection. PloS one. 2015;10(10):e0137725 doi: .
    1. Song Y, Garg S, Girotra M, Maddox C, von Rosenvinge EC, Dutta A, et al. Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection. PloS one. 2013;8(11):e81330 doi: .
    1. Weingarden AR, Chen C, Bobr A, Yao D, Lu Y, Nelson VM, et al. Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficile infection. American journal of physiology Gastrointestinal and liver physiology. 2014;306(4):G310–9. doi: .
    1. Hourigan SK, Chen LA, Grigoryan Z, Laroche G, Weidner M, Sears CL, et al. Microbiome changes associated with sustained eradication of Clostridium difficile after single faecal microbiota transplantation in children with and without inflammatory bowel disease. Alimentary pharmacology & therapeutics. 2015;42(6):741–52. doi: .
    1. Staley C, Kelly CR, Brandt LJ, Khoruts A, Sadowsky MJ. Complete Microbiota Engraftment Is Not Essential for Recovery from Recurrent Clostridium difficile Infection following Fecal Microbiota Transplantation. mBio. 2016;7(6):1–9. doi: .
    1. Khanna S, Vazquez-Baeza Y, Gonzalez A, Weiss S, Schmidt B, Muniz-Pedrogo DA, et al. Changes in microbial ecology after fecal microbiota transplantation for recurrent C. difficile infection affected by underlying inflammatory bowel disease. Microbiome. 2017;5(1):55 doi: .
    1. Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JH, Duflou A, et al. Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology. 2015;149(1):110–8 e4. doi: .
    1. Fuentes S, Rossen NG, van der Spek MJ, Hartman JH, Huuskonen L, Korpela K, et al. Microbial shifts and signatures of long-term remission in ulcerative colitis after faecal microbiota transplantation. The ISME journal. 2017;11(8):1877–89. doi: .
    1. Albenberg L, Esipova TV, Judge CP, Bittinger K, Chen J, Laughlin A, et al. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology. 2014;147(5):1055–63 e8. doi: .
    1. Hughes ER, Winter MG, Duerkop BA, Spiga L, Furtado de Carvalho T, Zhu W, et al. Microbial Respiration and Formate Oxidation as Metabolic Signatures of Inflammation-Associated Dysbiosis. Cell host & microbe. 2017;21(2):208–19. doi: .
    1. Petersen AM, Halkjaer SI, Gluud LL. Intestinal colonization with phylogenetic group B2 Escherichia coli related to inflammatory bowel disease: a systematic review and meta-analysis. Scandinavian journal of gastroenterology. 2015;50(10):1199–207. doi: .
    1. Garrett WS, Gallini CA, Yatsunenko T, Michaud M, DuBois A, Delaney ML, et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell host & microbe. 2010;8(3):292–300. doi: .
    1. Koroleva EP, Halperin S, Gubernatorova EO, Macho-Fernandez E, Spencer CM, Tumanov AV. Citrobacter rodentium-induced colitis: A robust model to study mucosal immune responses in the gut. Journal of immunological methods. 2015;421:61–72. doi: .
    1. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(43):16731–6. doi: .
    1. Lopez-Siles M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Faecalibacterium prausnitzii: from microbiology to diagnostics and prognostics. The ISME journal. 2017;11(4):841–52. doi: .

Source: PubMed

3
订阅