Effects of fecal microbiota transplantation in subjects with irritable bowel syndrome are mirrored by changes in gut microbiome

Rasmus Goll, Peter Holger Johnsen, Erik Hjerde, Joseph Diab, Per Christian Valle, Frank Hilpusch, Jorunn Pauline Cavanagh, Rasmus Goll, Peter Holger Johnsen, Erik Hjerde, Joseph Diab, Per Christian Valle, Frank Hilpusch, Jorunn Pauline Cavanagh

Abstract

Irritable bowel syndrome (IBS) is a common disorder of the lower gastrointestinal tract. The pathophysiology is far from settled, but a gut microbial dysbiosis is hypothesized to be a contributing factor. We earlier published a randomized double-blind placebo-controlled clinical trial on fecal microbiota transplantation (FMT) for IBS - the REFIT trial. The present data set describes the engraftment and includes participants from the study who received active FMT; 14 participants with effect of FMT (Effect) and 8 without (No effect). Samples were collected at baseline, after 6 and 12 months. Samples from the transplants (Donor) served as a comparator. In total 66 recipient samples and 17 donor samples were subjected to deep metagenomic sequencing, and taxonomic and functional analyses were performed. Alpha diversity measures showed a significantly increased diversity and evenness in the IBS groups compared to the donors. Taxonomic profiles showed higher relative abundance of phylum Firmicutes, and lower relative abundance of phylum Bacteroidetes, compared to donors at baseline. This profile was shifted toward the donor profile following FMT. Imputed growth rates showed that the resulting growth pattern was a conglomerate of donor and recipient activity. Thirty-four functional subclasses showed distinct differences between baseline samples and donors, most of which were shifted toward a donor-like profile after FMT. All of these changes were less pronounced in the No effect group. We conclude that FMT induces long-term changes in gut microbiota, and these changes mirror the clinical effect of the treatment. The study was registered in ClinicalTrials.gov (NCT02154867).

Keywords: Irritable bowel syndrome; diversity; engraftment; fecal microbiota transplantation; functional features; growth rate; metagenomic sequencing; taxonomic profile.

Figures

Figure 1.
Figure 1.
Panel A: Box plots with alpha diversity estimates where each sub-panel shows a different type of estimator. The colors group the samples into seven groups, and each point represents the richness estimate per sample. The data was filtered so that taxa occurring Effect group at baseline, after 12 months and the donor samples are visualized separate from the No effect group at baseline, after 12 months and the donor samples. The data were filtered so that taxa with a mean fractional abundance < 10−5 were excluded from the analysis.
Figure 1.
Figure 1.
Panel A: Box plots with alpha diversity estimates where each sub-panel shows a different type of estimator. The colors group the samples into seven groups, and each point represents the richness estimate per sample. The data was filtered so that taxa occurring Effect group at baseline, after 12 months and the donor samples are visualized separate from the No effect group at baseline, after 12 months and the donor samples. The data were filtered so that taxa with a mean fractional abundance < 10−5 were excluded from the analysis.
Figure 2.
Figure 2.
Panel A: Relative abundance of the most prevalent phyla for the seven groups. The data were filtered so that taxa not seen more than 5 times in at least 20% of the samples in the total dataset were removed. Following this, only taxa with a mean greater than 10−5 (fractional abundance > 0.00001) were kept before agglomeration at phylum level. Panel B: Differential abundance calculated with DESeq2 at phylum and family level, in the Effect group, 12 months vs. baseline. Positive log2FC indicate enriched taxa after FMT, negative log2FC indicate decreased taxa after FMT. Families indicated on the X-axis are colored according to phyla. FDR cut off for inclusion in the plot was < 10−4.
Figure 2.
Figure 2.
Panel A: Relative abundance of the most prevalent phyla for the seven groups. The data were filtered so that taxa not seen more than 5 times in at least 20% of the samples in the total dataset were removed. Following this, only taxa with a mean greater than 10−5 (fractional abundance > 0.00001) were kept before agglomeration at phylum level. Panel B: Differential abundance calculated with DESeq2 at phylum and family level, in the Effect group, 12 months vs. baseline. Positive log2FC indicate enriched taxa after FMT, negative log2FC indicate decreased taxa after FMT. Families indicated on the X-axis are colored according to phyla. FDR cut off for inclusion in the plot was < 10−4.
Figure 3.
Figure 3.
Multivariate regression analysis of the bacterial species. Each sample was labeled according to the corresponding study group. Panel A: This t1/t2-score plot of the orthogonal partial least squares projection to latent structures (OPLS) model (one predictive component and one orthogonal component) was built from the bacterial species composition in stool samples taken from the Donors, and from the Effect group at 3 time points: baseline, 6, and 12 months after FMT. The performance parameters R2Xcum, R2Ycum and Q2cum were 0.36, 0.61 and 0.32, respectively. Panel B: The top 50 bacterial species ranked by regression coefficients pertaining to the predictive components. To make the coefficients readily comparable, the independent variables for different taxa were scaled and centered prior to the analysis. The error bars indicate the confidence intervals of the coefficients. The coefficient is considered significant (above noise level), when the confidence interval does not include zero. Significant features are color-coded according to phylum.
Figure 3.
Figure 3.
Multivariate regression analysis of the bacterial species. Each sample was labeled according to the corresponding study group. Panel A: This t1/t2-score plot of the orthogonal partial least squares projection to latent structures (OPLS) model (one predictive component and one orthogonal component) was built from the bacterial species composition in stool samples taken from the Donors, and from the Effect group at 3 time points: baseline, 6, and 12 months after FMT. The performance parameters R2Xcum, R2Ycum and Q2cum were 0.36, 0.61 and 0.32, respectively. Panel B: The top 50 bacterial species ranked by regression coefficients pertaining to the predictive components. To make the coefficients readily comparable, the independent variables for different taxa were scaled and centered prior to the analysis. The error bars indicate the confidence intervals of the coefficients. The coefficient is considered significant (above noise level), when the confidence interval does not include zero. Significant features are color-coded according to phylum.
Figure 4.
Figure 4.
Growth rate score (GRiD) of the most frequent occurring species for samples in: (a) No effect group at baseline (green), after 12 months (purple) and the Donor samples (cyan); and (b) Effect group at baseline (pink), after 12 months (blue) and the Donor samples (cyan). GRiD score > 1 is an indication of bacteria in the growth phase, while GRiD score < 1 indicates bacteria in stationary or lag phase. The data were filtered so that species occurring < 15 times in < 30 samples were excluded from the analysis. FDR cut off for inclusion in the heat map was < 0.00005.
Figure 4.
Figure 4.
Growth rate score (GRiD) of the most frequent occurring species for samples in: (a) No effect group at baseline (green), after 12 months (purple) and the Donor samples (cyan); and (b) Effect group at baseline (pink), after 12 months (blue) and the Donor samples (cyan). GRiD score > 1 is an indication of bacteria in the growth phase, while GRiD score < 1 indicates bacteria in stationary or lag phase. The data were filtered so that species occurring < 15 times in < 30 samples were excluded from the analysis. FDR cut off for inclusion in the heat map was < 0.00005.
Figure 5.
Figure 5.
Differential abundance of functional subsystems with absolute abundance > 0.01% in the Effect group, 12 months vs. baseline. Positive log2FC indicate enriched subsystems after FMT, negative log2FC indicate decreased subsystems after FMT. Subsystem level 3 indicated on the X-axis are color-coded according to level 1 classification. FDR cut off for inclusion in the plot was < 0.05.
Figure 5.
Figure 5.
Differential abundance of functional subsystems with absolute abundance > 0.01% in the Effect group, 12 months vs. baseline. Positive log2FC indicate enriched subsystems after FMT, negative log2FC indicate decreased subsystems after FMT. Subsystem level 3 indicated on the X-axis are color-coded according to level 1 classification. FDR cut off for inclusion in the plot was < 0.05.
Figure 6.
Figure 6.
Multivariate regression analysis of the functional groups. Each sample was labeled according to the corresponding study group. Panel A: This t1/t2-score plot of the orthogonal partial least squares projection to latent structures (OPLS) model (one predictive component and one orthogonal component) was built from functional groups in stool samples taken from the Donors, and from the Effect group at 3 time points: baseline, 6, and 12 months after FMT. The performance parameters R2Xcum, R2Ycum and Q2cum were 0.55, 0.52 and 0.30, respectively. Panel B: The top 50 functional groups ranked by regression coefficients pertaining to the predictive components. To make the coefficients readily comparable, the independent variables for different functional groups were scaled and centered prior to the analysis. The error bars indicate the confidence intervals of the coefficients. The coefficient is considered significant (above noise level), when the confidence interval does not include zero. Significant features are color-coded according to level 1 classification.
Figure 6.
Figure 6.
Multivariate regression analysis of the functional groups. Each sample was labeled according to the corresponding study group. Panel A: This t1/t2-score plot of the orthogonal partial least squares projection to latent structures (OPLS) model (one predictive component and one orthogonal component) was built from functional groups in stool samples taken from the Donors, and from the Effect group at 3 time points: baseline, 6, and 12 months after FMT. The performance parameters R2Xcum, R2Ycum and Q2cum were 0.55, 0.52 and 0.30, respectively. Panel B: The top 50 functional groups ranked by regression coefficients pertaining to the predictive components. To make the coefficients readily comparable, the independent variables for different functional groups were scaled and centered prior to the analysis. The error bars indicate the confidence intervals of the coefficients. The coefficient is considered significant (above noise level), when the confidence interval does not include zero. Significant features are color-coded according to level 1 classification.

References

    1. Enck P, Aziz Q, Barbara G, Farmer AD, Fukudo S, Mayer EA, Niesler B, Quigley EMM, Rajilić-Stojanović M, Schemann M, et al. Irritable bowel syndrome. Nat Rev Dis Prim. 2016;2(1):16014. doi:10.1038/nrdp.2016.14.
    1. Marchesi JR, Adams DH, Fava F, Hermes GDA, Hirschfield GM, Hold G, Quraishi MN, Kinross J, Smidt H, Tuohy KM, et al. The gut microbiota and host health: a new clinical frontier. Gut. 2016;65(2):330–15. doi:10.1136/gutjnl-2015-309990.
    1. Ford AC, Lacy BE, Talley NJ.. Irritable bowel syndrome. N Engl J Med. 2017;376(26):2566–2578. doi:10.1056/NEJMra1607547.
    1. Crouzet L, Gaultier E, Del’Homme C, Cartier C, Delmas E, Dapoigny M, Fioramonti J, Bernalier-Donadille A. The hypersensitivity to colonic distension of IBS patients can be transferred to rats through their fecal microbiota. Neurogastroenterol Motil. 2013;25(4):e272–82. doi:10.1111/nmo.12103.
    1. Berg LK, Fagerli E, Martinussen M, Myhre A-O, Florholmen J, Goll R. Effect of fructose-reduced diet in patients with irritable bowel syndrome, and its correlation to a standard fructose breath test. Scand J Gastroenterol. 2013;48:936–943. doi:10.3109/00365521.2013.812139.
    1. Gibson PR, Shepherd SJ. Evidence-based dietary management of functional gastrointestinal symptoms: the FODMAP approach. J Gastroenterol Hepatol. 2010;25(2):252–258. doi:10.1111/j.1440-1746.2009.06149.x.
    1. Catassi G, Lionetti E, Gatti S, Catassi C. The low FODMAP diet: many question marks for a catchy acronym. Nutrients. 2017;9(3):292. doi:10.3390/nu9030292.
    1. Staudacher HM, Whelan K. The low FODMAP diet: recent advances in understanding its mechanisms and efficacy in IBS. Gut. 2017;66(8):1517–1527. doi:10.1136/gutjnl-2017-313750.
    1. Bennet SMP, Ohman L, Simren M. Gut microbiota as potential orchestrators of irritable bowel syndrome. Gut Liver. 2015;9:318–331. doi:10.5009/gnl14344.
    1. Ahmad OF, Akbar A. Microbiome, antibiotics and irritable bowel syndrome. Br Med Bull. 2016;120(1):91–99. doi:10.1093/bmb/ldw038.
    1. Vich VA, Imhann F, Collij V, Jankipersadsing SA, Gurry T, Mujagic Z, Kurilshikov A, Bonder MJ, Jiang X, Tigchelaar EF, et al. Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome. Sci Transl Med. 2018;10(472):eaap8914. doi:10.1126/scitranslmed.aap8914.
    1. Lee WJ, Lattimer LDN, Stephen S, Borum ML, Doman DB. Fecal microbiota transplantation: a review of emerging indications beyond relapsing clostridium difficile toxin colitis. Gastroenterol Hepatol (N Y). 2015;11:24–32.
    1. Simrén M, Barbara G, Flint HJ, Spiegel BMR, Spiller RC, Vanner S, Verdu EF, Whorwell PJ, Zoetendal EG. Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut. 2013;62(1):159–176. doi:10.1136/gutjnl-2012-302167.
    1. Kelly CR, Kahn S, Kashyap P, Laine L, Rubin D, Atreja A, Moore T, Wu G. Update on FMT 2015: indications, Methodologies, mechanisms and outlook. Gastroenterology. 2015;149(1):223–237. doi:10.1053/j.gastro.2015.05.008.
    1. Johnsen PH, Hilpüsch F, Cavanagh JP, Leikanger IS, Kolstad C, Valle PC, Goll R, et al. Faecal microbiota transplantation versus placebo for moderate-to-severe irritable bowel syndrome: a double-blind, randomised, placebo-controlled. Lancet Gastroenterol Hepatol. 2017;1253:17–24.
    1. Halkjær SI, Christensen AH, Lo BZS, Browne PD, Günther S, Hansen LH, Petersen AM. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut. 2018;67(12):2107–2115. doi:10.1136/gutjnl-2018-316434.
    1. Myneedu K, Deoker A, Schmulson MJ, Bashashati M. Fecal microbiota transplantation in irritable bowel syndrome: A systematic review and meta-analysis. United Eur Gastroenterol J. 2019;7(8):1033–1041. doi:10.1177/2050640619866990.
    1. Emiola A, Oh J. High throughput in situ metagenomic measurement of bacterial replication at ultra-low sequencing coverage. Nat Commun. 2018;9(1):4956. doi:10.1038/s41467-018-07240-8.
    1. Labus JS, Hollister EB, Jacobs J, Kirbach K, Oezguen N, Gupta A, Acosta J, Luna RA, Aagaard K, Versalovic J, et al. Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome. Microbiome. 2017;5(1):49. doi:10.1186/s40168-017-0260-z.
    1. Peter J, Fournier C, Keip B, Rittershaus N, Stephanou-Rieser N, Durdevic M, Dejaco C, Michalski M, Moser G, et al. Intestinal microbiome in irritable bowel syndrome before and after gut-directed hypnotherapy. Int J Mol Sci. 2018;19(11):3619. doi:10.3390/ijms19113619.
    1. Wang Z, Xu C-M, Liu Y-X, Wang X-Q, Zhang L, Li M, Zhu S-W, Xie Z-J, Wang P-H, Duan L-P, et al. Characteristic dysbiosis of gut microbiota of Chinese patients with diarrhea-predominant irritable bowel syndrome by an insight into the pan-microbiome. Chin Med J (Engl). 2019;132(8):889–904. doi:10.1097/CM9.0000000000000192.
    1. Lo Presti A, Zorzi F, Del Chierico F, Altomare A, Cocca S, Avola A, De Biasio F, Russo A, Cella E, Reddel S, Calabrese E, et al. Fecal and mucosal microbiota profiling in irritable bowel syndrome and inflammatory bowel disease. Front Microbiol. 2019;10:1655. doi:10.3389/fmicb.2019.01655.
    1. Pozuelo M, Panda S, Santiago A, Mendez S, Accarino A, Santos J, Guarner F, Azpiroz F, Manichanh C. Reduction of butyrate- and methane-producing microorganisms in patients with irritable bowel syndrome. Sci Rep. 2015;5(1):12693. doi:10.1038/srep12693.
    1. Hugerth LW, Andreasson A, Talley NJ, Forsberg AM, Kjellström L, Schmidt PT, Agreus L, Engstrand L, et al. No distinct microbiome signature of irritable bowel syndrome found in a Swedish random population. Gut. 2020;69(6):1076-1084. doi:10.1136/gutjnl-2019-318717.
    1. Pittayanon R, Lau JT, Yuan Y, Leontiadis GI, Tse F, Surette M, Moayyedi P. Gut microbiota in patients with irritable bowel syndrome—a systematic review. Gastroenterology. 2019;157(1):97–108. doi:10.1053/j.gastro.2019.03.049.
    1. Duan R, Zhu S, Wang B, Duan L. Alterations of gut microbiota in patients with irritable bowel syndrome based on 16S rRNA-targeted sequencing: a systematic review. Clin Transl Gastroenterol. 2019;10(2):e00012. doi:10.14309/ctg.0000000000000012.
    1. Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277. doi:10.3389/fimmu.2019.00277.
    1. Camilleri M. Intestinal secretory mechanisms in irritable bowel syndrome-diarrhea. Clin Gastroenterol Hepatol. 2015;13(6):1051–1057. doi:10.1016/j.cgh.2014.07.020.
    1. Cruz-Aguliar RM, Wantia N, Clavel T, Vehreschild MJGT, Buch T, Bajbouj M, Haller D, Busch D, Schmid R, Stein-Thoeringer C, et al. An open-labeled study on fecal microbiota transfer in irritable bowel syndrome patients reveals improvement in abdominal pain associated with the relative abundance of akkermansia muciniphila. Digestion. 2019;100:127–138. doi:10.1159/000494252.
    1. Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, Brown EM, Graham DB, Xavier RJ, Moon JJ, et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science. 2019;364(6446):1179–1184. doi:10.1126/science.aaw7479.
    1. Visconti A, Le Roy CI, Rosa F, Rossi N, Martin TC, Mohney RP, Li W, de Rinaldis E, Bell JT, Venter JC, Nelson KE, et al. Interplay between the human gut microbiome and host metabolism. Nat Commun. 2019;10(1):4505. doi:10.1038/s41467-019-12476-z.
    1. Hustoft TN, Hausken T, Ystad SO, Valeur J, Brokstad K, Hatlebakk JG, Lied GA, et al. Effects of varying dietary content of fermentable short-chain carbohydrates on symptoms, fecal microenvironment, and cytokine profiles in patients with irritable bowel syndrome. Neurogastroenterol Motil. 2017;29(4):e12969. doi:10.1111/nmo.12969.
    1. Venegas DP, De La Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Faber KN, Hermoso MA, et al. Short chain fatty acids (SCFAs)mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277. doi:10.3389/fimmu.2019.00277
    1. Fenn K, Strandwitz P, Stewart EJ, Dimise E, Rubin S, Gurubacharya S, Clardy J, Lewis K. Quinones are growth factors for the human gut microbiota. Microbiome. 2017;5(1):161. doi:10.1186/s40168-017-0380-5.
    1. Rossi M, Amaretti A, Raimondi S. Folate production by probiotic bacteria. Nutrients. 2011;3(1):118–134. doi:10.3390/nu3010118.
    1. Carbonero F, Benefiel AC, Alizadeh-Ghamsari AH, Gaskins HR. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front Physiol. 2012;3:448. doi:10.3389/fphys.2012.00448.
    1. Weiss G, Carver PL. Role of divalent metals in infectious disease susceptibility and outcome. Clin Microbiol Infect. 2018;24(1):16–23. doi:10.1016/j.cmi.2017.01.018.
    1. Porcheron G, Garénaux A, Proulx J, Sabri M, Dozois CM. Iron, copper, zinc, and manganese transport and regulation in pathogenic Enterobacteria: correlations between strains, site of infection and the relative importance of the different metal transport systems for virulence. Front Cell Infect Microbiol. 2013;3(DEC):90.. doi:10.3389/fcimb.2013.00090.
    1. Tedjo DI, Jonkers DMAE, Savelkoul PH, Masclee AA, Van BN, Pierik MJ, Penders J, et al. The effect of sampling and storage on the fecal microbiota composition in healthy and diseased subjects. PLoS One. 2015;10(5):e0126685. doi:10.1371/journal.pone.0126685.
    1. Wingett SW, Andrews S. FastQ screen: A tool for multi-genome mapping and quality control. F1000Research. 2018;7:1338. doi:10.12688/f1000research.15931.2.
    1. Chen S, Huang T, Zhou Y, Han Y, Xu M, Gu J. AfterQC: automatic filtering, trimming, error removing and quality control for fastq data. BMC Bioinform. 2017;18(S3):80. doi:10.1186/s12859-017-1469-3.
    1. BBMap download | . . Accessed 27June 2019.
    1. Menzel P, Ng KL, Krogh A. Fast and sensitive taxonomic classification for metagenomics with Kaiju. Nat Commun. 2016;7(1):11257. doi:10.1038/ncomms11257.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8:e61217. doi:10.1371/journal.pone.0061217.
    1. Silva GGZ, Green KT, Dutilh BE, Edwards RA. SUPER-FOCUS: a tool for agile functional analysis of shotgun metagenomic data. Bioinformatics. 2016;32(3):354–361. doi:10.1093/bioinformatics/btv584.
    1. Buchfink B, Xie C, Huson DH. Fast and sensitive protein alignment using DIAMOND. Nat Methods. 2015;12:59–60. doi:10.1038/nmeth.3176.
    1. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics. 2014;30(21):3123–3124. doi:10.1093/bioinformatics/btu494.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi:10.1186/s13059-014-0550-8.
    1. GRiD stool microbes database . . Accessed 26August 2019.
    1. Hong C, Manimaran S, Shen Y, Perez-Rogers JF, Byrd AL, Castro-Nallar E, Crandall KA, Johnson WE. PathoScope 2.0: a complete computational framework for strain identification in environmental or clinical sequencing samples. Microbiome. 2014;2(1):33. doi:10.1186/2049-2618-2-33.

Source: PubMed

3
Předplatit