Long-term diosmectite use does not alter the gut microbiota in adults with chronic diarrhea

Kévin Da Silva, Susie Guilly, Florence Thirion, Emmanuelle Le Chatelier, Nicolas Pons, Hugo Roume, Benoît Quinquis, Stanislav D Ehrlich, Nassima Bekkat, Hélène Mathiex-Fortunet, Harry Sokol, Joël Doré, Kévin Da Silva, Susie Guilly, Florence Thirion, Emmanuelle Le Chatelier, Nicolas Pons, Hugo Roume, Benoît Quinquis, Stanislav D Ehrlich, Nassima Bekkat, Hélène Mathiex-Fortunet, Harry Sokol, Joël Doré

Abstract

Background: Diosmectite, a natural colloidal clay, has been used worldwide for a number of approved indications, including the treatment of chronic functional diarrhea. Here, we used high-resolution whole metagenome shotgun sequencing to assess the impact of a 5 weeks administration of diosmectite (3 g/sachet, 3 sachets/day) on the fecal microbiota of 35 adults with functional chronic diarrhea.

Results: Gut microbiota was not impacted by diosmectite administration. In particular, richness remained stable and no microbial species displayed a significant evolution. Segregating patients either by diosmectite response (non responder, early responder, late responder) or by nationality (Great-Britain or Netherlands) yielded the same results.

Conclusion: We concluded that no microbiota-related physiological alterations are expected upon long-term treatment with diosmectite.

Trial registration: Clinicaltrials.gov NCT03045926.

Keywords: Chronic diarrhea; Diosmectite; Gut microbiota; Long-term diosmectite use; Loose stools; Microbiota composition; Shotgun metagenomics; Smecta®; Watery stools.

Conflict of interest statement

NB is employee of IPSEN. HMF is consultant of IPSEN. Other co-authors do not have any competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Bristol stool scale evolution according to visit. P-values associated with Wilcoxon signed-rank test are displayed. Boxes represent the median and interquartile ranges (IQRs) between the first and third quartiles; whiskers represent the lowest or highest values within 1.5 times IQR from the first or third quartiles
Fig. 2
Fig. 2
Microbiota evolution according to visit. A MGS richness evolution according to visit. B Boxplots of Bray–Curtis dissimilarity between MGS abundance at baseline (D-1) and MGS abundance at other time points (D-30, D-14, D8 or D35). P-values associated with Wilcoxon signed-rank test are displayed. Boxes represent the median and interquartile ranges (IQRs) between the first and third quartiles; whiskers represent the lowest or highest values within 1.5 times IQR from the first or third quartiles. C Principal Coordinates Analysis (PCoA) performed on Bray–Curtis dissimilarity matrix computed on MGS abundances. Patients are colored according to visit, and the analysis of similarity between different visits was computed through ANOSIM
Fig. 3
Fig. 3
Phyla distribution evolution. Mean phyla relative abundance along the different time points
Fig. 4
Fig. 4
Early-, late-, and non-responders. A Bristol stool scale evolution across the different time points according to responder subgroups. P-values associated with Wilcoxon signed-rank test are displayed. B Bristol stool scale at baseline (D-1) according to responder subgroups. P-values associated with Mann–Whitney test are displayed. C MGS richness at baseline (D-1) according to responder subgroups. P-values associated with Mann–Whitney test are displayed. Boxes represent the median and interquartile ranges (IQRs) between the first and third quartiles; whiskers represent the lowest or highest values within 1.5 times IQR from the first or third quartiles

References

    1. Moosavi M. Bentonite clay as a natural remedy: a brief review. Iran J Public Health. 2017;46:1176–1183.
    1. Guarino A, Lo Vecchio A, Pirozzi MR. Clinical role of diosmectite in the management of diarrhea. Expert Opin Drug Metab Toxicol. 2009;5:433–440. doi: 10.1517/17425250902865594.
    1. Dupont C, Foo JLK, Garnier P, Moore N, Mathiex-Fortunet H, Salazar-Lindo E. Oral diosmectite reduces stool output and diarrhea duration in children with acute watery diarrhea. Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc. 2009;7:456–62.
    1. Dupont C, Vernisse B. Anti-diarrheal effects of diosmectite in the treatment of acute diarrhea in children: a review. Paediatr Drugs. 2009;11:89–99. doi: 10.2165/00148581-200911020-00001.
    1. Khediri F, Mrad AI, Azzouz M, Doughi H, Najjar T, Mathiex-Fortunet H, et al. Efficacy of diosmectite (smecta) in the treatment of acute watery diarrhoea in adults: a multicentre, randomized, double-blind, placebo-controlled, parallel group study. Gastroenterol Res Pract. 2011;2011:783196. doi: 10.1155/2011/783196.
    1. Hu C, Song J, You Z, Luan Z, Li W. Zinc oxide-montmorillonite hybrid influences diarrhea, intestinal mucosal integrity, and digestive enzyme activity in weaned pigs. Biol Trace Elem Res United States. 2012;149:190–196. doi: 10.1007/s12011-012-9422-9.
    1. Chang F-Y, Lu C-L. Irritable bowel syndrome in the 21st century: perspectives from Asia or South-east Asia. J Gastroenterol Hepatol Australia. 2007;22:4–12. doi: 10.1111/j.1440-1746.2006.04672.x.
    1. Ducrotte P, Dapoigny M, Bonaz B, Siproudhis L. Symptomatic efficacy of beidellitic montmorillonite in irritable bowel syndrome: a randomized, controlled trial. Aliment Pharmacol Ther England. 2005;21:435–444. doi: 10.1111/j.1365-2036.2005.02330.x.
    1. Szajewska H, Dziechciarz P, Mrukowicz J. Meta-analysis: Smectite in the treatment of acute infectious diarrhoea in children. Aliment Pharmacol Ther England. 2006;23:217–227. doi: 10.1111/j.1365-2036.2006.02760.x.
    1. Droy MT, Drouet Y, Geraud G, Schatz B. Spinnability: a new approach to intestinal stress and its therapy. Gastroenterol Clin Biol France. 1985;9:119–121.
    1. Fioramonti J, Droy-Lefaix MT, Buéno L. Changes in gastro-intestinal motility induced by cholera toxin and experimental osmotic diarrhoea in dogs: effects of treatment with an argillaceous compound. Digestion. 1987;36:230–237. doi: 10.1159/000199423.
    1. Dupont C, Moreno JL, Barau E, Bargaoui K, Thiane E, Plique O. Effect of diosmectite on intestinal permeability changes in acute diarrhea: a double-blind placebo-controlled trial. J Pediatr Gastroenterol Nutr. 1992;14:413–419. doi: 10.1097/00005176-199205000-00007.
    1. Mahraoui L, Heyman M, Plique O, Droy-Lefaix MT, Desjeux JF. Apical effect of diosmectite on damage to the intestinal barrier induced by basal tumour necrosis factor-alpha. Gut. 1997;40:339–343. doi: 10.1136/gut.40.3.339.
    1. Eutamene H, Beaufrand C, Harkat C, Theodorou V. The role of mucoprotectants in the management of gastrointestinal disorders. Expert Rev Gastroenterol Hepatol England. 2018;12:83–90. doi: 10.1080/17474124.2018.1378573.
    1. Aguzzi C, Cerezo P, Viseras C, Caramella C. Use of clays as drug delivery systems: possibilities and limitations. Appl Clay Sci. 2007;36:22–36. doi: 10.1016/j.clay.2006.06.015.
    1. Droylefaix MT, Drouet Y, Schatz B. Sodium glycodeoxycholate and spinability of gastrointestinal mucus-protective effect of smectite. Gastroenterology. 1985;88:1369.
    1. González R, de Medina FS, Martínez-Augustin O, Nieto A, Gálvez J, Risco S, et al. Anti-inflammatory effect of diosmectite in hapten-induced colitis in the rat. Br J Pharmacol. 2004;141:951–960. doi: 10.1038/sj.bjp.0705710.
    1. Song M, Liu Y, Soares JA, Che TM, Osuna O, Maddox CW, et al. Dietary clays alleviate diarrhea of weaned pigs. J Anim Sci. 2012;90:345–360. doi: 10.2527/jas.2010-3662.
    1. Buccigrossi V, Russo C, Guarino A, de Freitas MB, Guarino A. Mechanisms of antidiarrhoeal effects by diosmectite in human intestinal cells. Gut Pathog. 2017;9:23. doi: 10.1186/s13099-017-0172-2.
    1. Pérez-Gaxiola G, Cuello-García CA, Florez ID, Pérez-Pico VM. Smectite for acute infectious diarrhoea in children. Cochrane database Syst Rev. 2018;4:CD011526.
    1. Yao-Zong Y, Shi-Rong L, Delvaux M. Comparative efficacy of dioctahedral smectite (Smecta) and a probiotic preparation in chronic functional diarrhoea. Dig liver Dis Off J Ital Soc Gastroenterol Ital Assoc Study Liver. 2004;36:824–8.
    1. Chang F-Y, Lu C-L, Chen C-Y, Luo J-C. Efficacy of dioctahedral smectite in treating patients of diarrhea-predominant irritable bowel syndrome. J Gastroenterol Hepatol Australia. 2007;22:2266–2272. doi: 10.1111/j.1440-1746.2007.04895.x.
    1. Dumitrascu DL, Stanculete M, Mitrea I, Dumitrascu DM, Farcas A. The effect of two antidiarrhoeal drugs on the psychosocial adjustment to illness in chronic functional diarrhoea. Rom J Intern Med Germany. 2004;42:191–197.
    1. Mearin F, Lacy BE, Chang L, Chey WD, Lembo AJ, Simren M, et al. Bowel Disorders. United States: Gastroenterology; 2016.
    1. Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–841. doi: 10.1038/nbt.2942.
    1. Nielsen HB, Almeida M, Juncker AS, Rasmussen S, Li J, Sunagawa S, et al. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat Biotechnol. 2014;32:822–828. doi: 10.1038/nbt.2939.
    1. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528:262–266. doi: 10.1038/nature15766.
    1. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555:623–628. doi: 10.1038/nature25979.
    1. Vieira-Silva S, Falony G, Belda E, Nielsen T, Aron-Wisnewsky J, Chakaroun R, et al. Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature England. 2020;581:310–315. doi: 10.1038/s41586-020-2269-x.
    1. Vandeputte D, Falony G, Vieira-Silva S, Tito RY, Joossens M, Raes J. Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut. 2016;65:57–62. doi: 10.1136/gutjnl-2015-309618.
    1. IHMS. Available from: 2020 Oct 5
    1. Costea PI, Zeller G, Sunagawa S, Pelletier E, Alberti A, Levenez F, et al. Towards standards for human fecal sample processing in metagenomic studies. Nat Biotechnol. 2017;35:1069–76. doi: 10.1038/nbt.3960.
    1. Pons N, Batto J-M, Kennedy S, Almeida M, Boumezbeur F, Moumen B, et al. METEOR -a plateform for quantitative metagenomic profiling of complex ecosystems. 2010. Available from:
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923.
    1. Criscuolo A, Brisse S. AlienTrimmer: a tool to quickly and accurately trim off multiple short contaminant sequences from high-throughput sequencing reads. Genomics. 2013;102:500–506. doi: 10.1016/j.ygeno.2013.07.011.
    1. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–546. doi: 10.1038/nature12506.
    1. Sayers EW, Agarwala R, Bolton EE, Brister JR, Canese K, Clark K, et al. Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2019;47:D23–D28. doi: 10.1093/nar/gky1069.
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol England. 1990;215:403–410. doi: 10.1016/S0022-2836(05)80360-2.
    1. R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria; 2017. Available from:
    1. Oksanen J, Blanchet FG, Friendly M, Kindt R, Legendre P, McGlinn D, et al. Vegan: community ecology package. 2019. Available from:
    1. Thioulouse J, Dray S, Dufour A--B, Siberchicot A, Jombart T, Pavoine S. Multivariate Analysis of Ecological Data with {ade4}. New York: Springer; 2018.
    1. Torchiano M. effsize: Efficient Effect Size Computation. 2018. Available from:
    1. Romano J, Kromrey JD, Coraggio J, Skowronek J. Appropriate statistics for ordinal level data: Should we really be using t-test and Cohen’sd for evaluating group differences on the NSSE and other surveys? Annu Meet Florida Assoc Institutional Res. 2006;177:1–3.
    1. de Gunzburg J, Ghozlane A, Ducher A, Le Chatelier E, Duval X, Ruppé E, et al. Protection of the human gut microbiome from antibiotics. J Infect Dis. 2018;217:628–636. doi: 10.1093/infdis/jix604.
    1. Luan Z, Sun G, Huang Y, Yang Y, Yang R, Li C, et al. Metagenomics study reveals changes in gut microbiota in centenarians: a cohort study of hainan centenarians. Front Microbiol. 2020;11:1474. doi: 10.3389/fmicb.2020.01474.
    1. Noguchi K, Gel YR, Brunner E, Konietschke F. {nparLD}: An {R} software package for the nonparametric analysis of longitudinal data in factorial experiments. J Stat Softw. 2012;50:1–23. Available from: .

Source: PubMed

3
Abonnere