Microbiota changes induced by microencapsulated sodium butyrate in patients with inflammatory bowel disease

Sonia Facchin, Nicola Vitulo, Matteo Calgaro, Andrea Buda, Chiara Romualdi, Daniel Pohl, Barbara Perini, Greta Lorenzon, Carla Marinelli, Renata D'Incà, Giacomo Carlo Sturniolo, Edoardo Vincenzo Savarino, Sonia Facchin, Nicola Vitulo, Matteo Calgaro, Andrea Buda, Chiara Romualdi, Daniel Pohl, Barbara Perini, Greta Lorenzon, Carla Marinelli, Renata D'Incà, Giacomo Carlo Sturniolo, Edoardo Vincenzo Savarino

Abstract

Background: Butyrate has shown anti-inflammatory and regenerative properties, providing symptomatic relief when orally supplemented in patients suffering from various colonic diseases. We investigated the effect of a colonic-delivery formulation of butyrate on the fecal microbiota of patients with inflammatory bowel diseases (IBDs).

Methods: In this double-blind, placebo-controlled, pilot study, 49 IBD patients (n = 19 Crohn's disease, CD and n = 30 ulcerative colitis, UC) were randomized to oral administration of microencapsulated-sodium-butyrate (BLM) or placebo for 2 months, in addition to conventional therapy. Eighteen healthy volunteers (HVs) were recruited to provide a healthy microbiota model of the local people. Fecal microbiota from stool samples was assessed by 16S sequencing. Clinical disease activity and quality of life (QoL) were evaluated before and after treatment.

Key results: At baseline, HVs showed a different microbiota composition compared with IBD patients. Sodium-butyrate altered the gut microbiota of IBD patients by increasing bacteria able to produce SCFA in UC patients (Lachnospiraceae spp.) and the butyrogenic colonic bacteria in CD patients (Butyricicoccus). In UC patients, QoL was positively affected by treatment.

Conclusions and inferences: Sodium-butyrate supplementation increases the growth of bacteria able to produce SCFA with potentially anti-inflammatory action. The clinical impact of this finding requires further investigation.

Keywords: 16S metabarcoding; gut inflammation; inflammatory bowel disease; prebiotics; short-chain fatty acid.

Conflict of interest statement

The authors report no conflict of interest.

© 2020 The Authors. Neurogastroenterology & Motility published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Graphic summary. Project study design: 18 healthy subjects and forty‐nine patients (21 on butyrose group and 28 on placebo group; 19 CD patients and 30 UC patients) were enrolled for this study. Pie charts show the microbial composition at phylum level in the different groups of samples
Figure 2
Figure 2
Box‐plot comparison of the alfa diversity calculated using Fisher metric between IBD and healthy group (A), timepoint T0 and T1 within the BLM group (B), and PBO group (C). Analysis performed with other distances confirms the same results (data not shown)
Figure 3
Figure 3
PERMANOVA tests if samples can be significantly separated accordingly to different variables (eg, treatment or type of disease). The figure shows the principal coordinate analysis considering the samples grouped according to (A) healthy and IBD status, (B) treatment (placebo and butyrose) at T0 = baseline and T1 = post‐treatment, (C) disease (UC and CD) undergoing BLM or PBO treatment at T0 and T1 timepoint, and (D) disease activity (1 indicates an active disease, while 0 a non‐active disease). The P‐value derived from the PERMANOVA test is reported for each comparison, and significant P‐value (<0.05) is indicated with a star
Figure 4
Figure 4
Barplot of the microbial composition at phylum level: average ASV abundance percentages of the samples stratified by treatment (butyrose, placebo, or control), disease (CD, UC, or control), and timepoint (T0 = baseline or T1 = post‐treatment)
Figure 5
Figure 5
sPLS‐DA analysis identified a subset of discriminant ASVs: for each ASV, a loading value that represents the discriminant power of that ASV in explaining differences between 2 examined conditions. The higher the absolute value, the bigger is the discriminative power. The loading value plots display the top 15 (panels A‐E) and top 6 (panel F) discriminant ASVs for each comparison. Percentages shown in the bars represent the mean relative abundances of each ASV in the considered conditions. The dark color and a light color of each bar represent the average relative abundance (in percentage) of an ASV in the considered comparison

References

    1. Sha S, Xu B, Wang X, et al. The biodiversity and composition of the dominant fecal microbiota in patients with inflammatory bowel disease. Diagn Microbiol Infect Dis. 2013;75(3):245‐251.
    1. Laserna‐Mendieta EJ, Clooney AG, Carretero‐Gomez JF, et al. Determinants of reduced genetic capacity for butyrate synthesis by the gut microbiome in Crohn’s disease and ulcerative colitis. J Crohn’s Colitis. 2018;12(2):204‐216.
    1. Stumpff F. A look at the smelly side of physiology: transport of short chain fatty acids. Pflugers Arch Eur J Physiol. 2018;470(4):571‐598.
    1. den Besten G, Lange K, Havinga R, et al. Gut‐derived short‐chain fatty acids are vividly assimilated into host carbohydrates and lipids. AJP Gastrointest Liver Physiol. 2013;305(12):G900‐G910.
    1. Nagalingam NA, Lynch SV. Role of the microbiota in inflammatory bowel diseases. Inflamm Bowel Dis. 2012;18(5):968‐980.
    1. Machiels K, Joossens M, Sabino J, et al. A decrease of the butyrate‐producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63(8):1275‐1283.
    1. Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer RJ. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther. 2008;27(2):104‐119.
    1. Załȩski A, Banaszkiewicz A, Walkowiak J. Butyric acid in irritable bowel syndrome. Prz Gastroenterol. 2013;8(6):350‐353.
    1. Louis P, Duncan SH, McCrae SI, Millar J, Jackson MS, Flint HJ. Restricted distribution of the butyrate kinase pathway among butyrate‐producing bacteria from the human colon. J Bacteriol. 2004;186(7):2099‐2106.
    1. Sossai P. Butyric acid: what is the future for this old substance? Swiss Med Wkly. 2012;142:w13596.
    1. Hamer HM, Jonkers DMAE, Vanhoutvin SALW, et al. Effect of butyrate enemas on inflammation and antioxidant status in the colonic mucosa of patients with ulcerative colitis in remission. Clin Nutr. 2010;29(6):738‐744.
    1. Assisi RF, GISDI Study Group . Combined butyric acid/mesalazine treatment in ulcerative colitis with mild‐moderate activity. Results of a multicentre pilot study. Minerva Gastroenterol Dietol. 2008;54(3):231‐238
    1. Sabatino AD, Morera R, Ciccocioppo R, et al. Oral butyrate for mildly to moderately active Crohn’s disease. Aliment Pharmacol Ther. 2005;22(9):789‐794.
    1. Vernia P, Annese V, Bresci G, et al. Topical butyrate improves efficacy of 5‐ASA in refractory distal ulcerative colitis: results of a multicentre trial. Eur J Clin Invest. 2003;33(3):244‐248.
    1. Vernia P, Monteleone G, Grandinetti G, et al. Combined oral sodium butyrate and mesalazine treatment compared to oral mesalazine alone in ulcerative colitis: Randomized, double‐blind, placebo‐ controlled pilot study. Dig Dis Sci. 2000;45(5):976‐981.
    1. Steinhart AH, Brzezinski A, Baker JP. Treatment of refractory ulcerative proctosigmoiditis with butyrate enemas. Am J Gastroenterol. 1994;89(2):179‐183.
    1. Vernia P, Marcheggiano A, Caprilli R, et al. Short‐chain fatty acid topical treatment in distal ulcerative colitis. Aliment Pharmacol Ther. 1995;9(3):309‐313.
    1. Kelly OB, O’Donnell S, Stempak JM, Steinhart AH, Silverberg MS. Therapeutic drug monitoring to guide infliximab dose adjustment is associated with better endoscopic outcomes than clinical decision making alone in active inflammatory bowel disease. Inflamm Bowel Dis. 2017;23(7):1202‐1209.
    1. Scheppach W, Sommer H, Kirchner T, et al. Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis. Gastroenterology. 1992;103(1):51‐56.
    1. Vanhoutvin SALW, Troost FJ, Kilkens TOC, et al. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil. 2009;21(9):952‐e76.
    1. Kannampalli P, Shaker R, Sengupta JN. Colonic butyrate‐algesic or analgesic? Neurogastroenterol Motil. 2011;23(11):975‐979.
    1. Patent “Process for the production of an n‐butyric acid compound in micro encapsulated form, for animal or human consumption”. . Accessed September 26, 2012.
    1. Lin J‐F, Chen J‐M, Zuo J‐H, et al. Meta‐analysis: fecal calprotectin for assessment of inflammatory bowel disease activity. Inflamm Bowel Dis. 2014;20(8):1407‐1415.
    1. Costa F, Mumolo MG, Ceccarelli L, et al. Calprotectin is a stronger predictive marker of relapse in ulcerative colitis than in Crohn’s disease. Gut. 2005;54(3):364‐368.
    1. Lewis JD, Chuai S, Nessel L, Lichtenstein GR, Aberra FN, Ellenberg JH. Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis. Inflamm Bowel Dis. 2008;14(12):1660‐1666.
    1. Vermeire S, Schreiber S, Sandborn WJ, Dubois C, Rutgeerts P. Correlation between the Crohn’s disease activity and Harvey‐Bradshaw indices in assessing Crohn’s disease severity. Clin Gastroenterol Hepatol. 2010;8(4):357‐363.
    1. Vuitton L, Marteau P, Sandborn WJ, et al. IOIBD technical review on endoscopic indices for Crohn’s disease clinical trials. Gut. 2016;65(9):1447‐1455.
    1. Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006;55(6):749‐753.
    1. Love J, Irvine E, Fedorak R. Quality of life in inflammatory bowel disease. J Clin Gastroenterol. 1992;14(1):15‐19.
    1. Brooks AW, Priya S, Blekhman R, Bordenstein SR. Gut microbiota diversity across ethnicities in the United States. PLoS Biol. 2018;16(12):e2006842.
    1. Takahashi S, Tomita J, Nishioka K, et al. Development of a prokaryotic universal primer for simultaneous analysis of Bacteria and Archaea using next‐generation sequencing. PLoS One. 2014. 10.1371/journal.pone.0105592
    1. Callahan BJ, Mcmurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2 paper supplementary information: high resolution sample inference from amplicon data. Nat Methods. 2016;13(7):581‐583.
    1. Yilmaz P, Parfrey LW, Yarza P, et al. The SILVA and “all‐species living tree project (LTP)” taxonomic frameworks. Nucleic Acids Res. 2014;42:D643‐D648.
    1. Wright ES. Using DECIPHER v2.0 to analyze big biological sequence data in R. R J. 2016;8(1):352.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8(4):e61217.
    1. Rohart F, Gautier B, Singh A, Lê Cao K‐A. mixOmics: An R package for ‘omics feature selection and multiple data integration. PLoS Comput Biol. 2017;13(11):e1005752.
    1. Rodriguez‐R LM, Konstantinidis KT. Estimating coverage in metagenomic data sets and why it matters. ISME J. 2014;8(11):2349.
    1. Sivaprakasam S, Bhutia YD, Yang S, Ganapathy V. Short‐chain fatty acid transporters: role in colonic homeostasis. Compr Physiol. 2018;8(1):299‐314.
    1. Zuo T, Ng SC. The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Front Microbiol. 2018;9:2247.
    1. Lane ER, Zisman TL, Suskind DL. The microbiota in inflammatory bowel disease: current and therapeutic insights. J Inflamm Res. 2017;10:63‐73.
    1. Lavelle A, Lennon G, Winter DC, O’Connell PR. Colonic biogeography in health and ulcerative colitis. Gut Microbes. 2016;7:435‐442.
    1. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti‐inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci. 2008;105(43):16731‐16736.
    1. Png CW, Lindén SK, Gilshenan KS, et al. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol. 2010;105(11):2420‐2428.
    1. Berger FK, Schwab N, Glanemann M, Bohle RM, Gärtner B, Groesdonk HV. Flavonifractor (Eubacterium) plautii bloodstream infection following acute cholecystitis. IDCases. 2018;14:e00461.
    1. Gomez‐Arango LF, Barrett HL, Wilkinson SA, et al. Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes. 2018;9(3):189‐201.
    1. Jones‐Hall YL, Kozik A, Nakatsu C. Ablation of tumor necrosis factor is associated with decreased inflammation and alterations of the microbiota in a mouse model of inflammatory bowel disease. PLoS One. 2015;10(3):e0119441.
    1. Spees AM, Lopez CA, Kingsbury DD, Winter SE, Bäumler AJ. Colonization resistance: battle of the bugs or ménage à trois with the host? PLoS Pathog. 2013;9(11):e1003730.
    1. Tedjo DI, Smolinska A, Savelkoul PH, et al. The fecal microbiota as a biomarker for disease activity in Crohn’s disease. Sci Rep. 2016;6:35216.
    1. Dinh DM, Volpe GE, Duffalo C, et al. Intestinal microbiota, microbial translocation, and systemic inflammation in chronic HIV infection. J Infect Dis. 2015;211(1):19‐27.
    1. Fernández‐Murga ML, Sanz Y. Safety assessment of Bacteroides uniformis CECT 7771 isolated from stools of healthy breast‐fed infants. PLoS One. 2016;11(1):e0145503.
    1. Canani RB, Di CM, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. 2011;17(12):1519‐1528.
    1. Banasiewicz T, Krokowicz Ł, Stojcev Z, et al. Microencapsulated sodium butyrate reduces the frequency of abdominal pain in patients with irritable bowel syndrome. Colorectal Dis. 2013;15(2):204‐209.
    1. Million M, Raoult D. Linking gut redox to human microbiome. Hum Microbiome J. 2018;10:27‐32.

Source: PubMed

3
Subscribe