A yeast fermentate improves gastrointestinal discomfort and constipation by modulation of the gut microbiome: results from a randomized double-blind placebo-controlled pilot trial

Iris Pinheiro, Larry Robinson, An Verhelst, Massimo Marzorati, Björn Winkens, Pieter Van den Abbeele, Sam Possemiers, Iris Pinheiro, Larry Robinson, An Verhelst, Massimo Marzorati, Björn Winkens, Pieter Van den Abbeele, Sam Possemiers

Abstract

Background: Constipation and symptoms of gastrointestinal discomfort such as bloating are common among otherwise healthy individuals, but with significant impact on quality of life. Despite the recognized contribution of the gut microbiome to this pathology, little is known about which group(s) of microorganism(s) are playing a role. A previous study performed in vitro suggests that EpiCor® fermentate has prebiotic-like properties, being able to favorably modulate the composition of the gut microbiome. Therefore, the aim of this study was to investigate the effects of EpiCor fermentate in a population with symptoms of gastrointestinal discomfort and reduced bowel movements and to evaluate its effect at the level of the gut microbiome.

Methods: This pilot study was performed according to a randomized, double-blind, placebo-controlled parallel design. Eighty subjects with symptoms of gastrointestinal discomfort and constipation were allocated to one of two trial arms (placebo or EpiCor fermentate). Randomization was done in a stratified manner according to symptom severity, resulting in two subgroups of patients: severe and moderate. Daily records of gastrointestinal symptoms were assessed on a 5-point scale, and also stool frequency and consistency were documented during a 2-week run-in and a 6-week intervention phases. Averages over two-week intervals were calculated. Constipation-associated quality of life and general perceived stress were assessed at baseline and after 3 and 6 weeks of intervention. Fecal samples were also collected at these same time points.

Results: EpiCor fermentate led to a significant improvement of symptoms such as bloating/distension (p = 0.033 and p = 0.024 after 2 and 4 weeks of intervention, respectively), feeling of fullness (p = 0.004 and p = 0.023 after 2 and 4 weeks of intervention, respectively) and general daily scores (p = 0.046 after 2 weeks of intervention) in the moderate subgroup. A significant improvement in stool consistency was observed for the total population (p = 0.023 after 2 weeks of intervention) as well as for the severe subgroup (p = 0.046 after 2 weeks of intervention), and a nearly significant increase in stool frequency was detected for the total cohort (p = 0.083 and p = 0.090 after 2 and 4 weeks of intervention, respectively). These effects were accompanied by an improvement in constipation-associated quality of life and general perceived stress, particularly in the moderate subgroup. Members of the families Bacteroidaceae and Prevotellaceae, two groups of bacteria that have been previously reported to be deficient in constipated patients, were found to increase with EpiCor fermentate in the severe subgroup. In the moderate subgroup, a significant increase in Akkermansia muciniphila was observed.

Conclusions: Despite the relatively low dose administered (500 mg/day), particularly when comparing to the high recommended doses for prebiotic fibers, EpiCor fermentate was able to modulate the composition of the gut microbiome, resulting in improvement of constipation-associated symptoms. Conversely, the reported increase in bowel movements may have altered the gut microbial community by increasing those groups of bacteria that are better adapted to a faster gastrointestinal transit time.

Trial registration: NCT03051399 at ClinicalTrials.gov. Retrospectively registered. Registration date: 13 February 2017.

Keywords: Bacteroides; Bacteroidetes; Constipation; EpiCor fermentate; GI transit time; Gastrointestinal discomfort; Gut microbiome; Human study; Prevotella; Saccharomyces cerevisiae.

Conflict of interest statement

Ethics approval and consent to participate

Ethical approval was obtained from the Independent Ethics Committee (IEC) / Institutional Review Board (IRB) from the Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium. All participants signed an informed consent prior to inclusion in the study.

Consent for publication

Not applicable.

Competing interests

This study was commissioned by Embria Health Sciences (USA) to ProDigest BVBA (Belgium), which subcontracted the D.R.U.G. Unit at the Ghent University hospital (Belgium) to perform the study. The project was coordinated by ProDigest but all parties involved took part on the discussion about study design, inclusion and exclusion criteria and outcome measurements. However, during study realization and data analysis, both ProDigest and D.R.U.G. Unit staff members involved in the study were blinded. Reporting was performed by ProDigest. Embria Health Sciences (USA) owns the proprietary product used in this study. Massimo Marzorati is one of the co-founders of ProDigest and Björn Winkens was subcontracted by ProDigest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Schematic diagram of the study flow (based on CONSORT 2010 guidelines). Legend: GI, gastrointestinal; PAC-QOL, Patient Assessment of Constipation Quality of Life; PSS, Perceived Stress Scale. a Gastrointestinal symptoms were assessed by asking the volunteers to grade daily in the evening the average severity over the previous 24 h on a 5-point scale from 0 (not at all) to 4 (extremely) for the following 5 GI characteristics: bloating/distension, passage of gas, GI rumbling, feeling of fullness and abdominal discomfort
Fig. 2
Fig. 2
Schematic diagram of primary and secondary objectives and instruments used for data/sample collection. Legend: GI, gastrointestinal; PAC-QOL, Patient Assessment of Constipation Quality of Life; PSS, Perceived Stress Scale. a Gastrointestinal symptoms were assessed by asking the volunteers to grade daily in the evening the average severity over the previous 24 h on a 5-point scale from 0 (not at all) to 4 (extremely) for the following 5 GI characteristics: bloating/distension, passage of gas, GI rumbling, feeling of fullness and abdominal discomfort. b Stool consistency was recorded according to the Bristol Stool Form Scale
Fig. 3
Fig. 3
Mean differences between EpiCor and placebo on gastrointestinal symptoms on the three cohorts. Legend: The enlisted gastrointestinal symptoms are BD (bloating/distension), GAS (passage of gas), RUM (GI rumbling), FF (feeling of fullness) and AD (abdominal discomfort). A daily total score (DTS) calculated as the sum of all items recorded each day is also shown. A linear mixed model analysis that takes into account the differences between groups at baseline was used (p-values <0.05 are depicted in bold text; p-values <0.1 are depicted in regular text)
Fig. 4
Fig. 4
Mean differences between EpiCor and placebo on stool frequency (a) and consistency (b) on the three cohorts. Legend: A linear mixed model analysis that takes into account the differences between groups at baseline was used (p-values <0.05 are depicted in bold text; p-values <0.1 are depicted in regular text)
Fig. 5
Fig. 5
Mean differences between EpiCor and placebo on constipation-associated quality of life (a) and perceived stress (b) on the three cohorts. Legend: The enlisted Patient Assessment of Constipation Quality of Life (PAC-QOL) items are PhD (physical discomfort), PsD (psychosocial discomfort), W&C (worries and concerns) and SAT (satisfaction). An instrument total score (ITS) calculated as the average of all 28 items recorded at each visit is also shown. PSS relates to the Perceived Stress Scale instrument. A linear mixed model analysis that takes into account the differences between groups at baseline was used (no p-values <0.05 were found; p-values <0.1 are depicted in regular text)
Fig. 6
Fig. 6
Bacteroidetes and Firmicutes relative abundances within the total cohort (a) and the two subgroups, severe (b) and moderate (c) that have been treated either with placebo or EpiCor. Each box represents median (50th percentile) and interquartile range (25th and 75th percentiles). The symbol (+) represents the mean. The outliers are indicated as dots (Tukey method). Significant (p < 0.05) and nearly significant (p < 0.1) p-values are also indicated within the boxplots (two-way repeated measures ANOVA with Dunnett’s multiple comparison’s test against V1). The inner plots (a*, b* and c*) correspond to the calculated Firmicutes/Bacteroidetes (F/B) ratio at each visit within the two treatment groups. Legend: V1, V2 and V3 correspond to visit 1 (baseline), visit 2 (3-weeks after treatment) and visit 3 (6-weeks after treatment), respectively
Fig. 7
Fig. 7
Principal component analysis (47.4%) of the relative fold-changes calculated for all taxonomic groups (family and genus levels). Each dot represents a treated group (either EpiCor or placebo) for all three cohorts (total cohort, severe and moderate). The first component (PC1) accounts for nearly 28% of the variance, and the second component (PC2) for nearly 20%. legend: E, Epicor; P, placebo; V, visit
Fig. 8
Fig. 8
Variables (taxa) correlation plot. Each vector represents a taxonomic group (family or genus levels). Shorter vectors only slightly contribute for differentiation between groups. Longer vectors have a bigger weight in groups’ differentiation. Those variables that mostly contribute the first component (PC1) are indicated in bold text
Fig. 9
Fig. 9
Hierarchical clustering heatmap of the Log2 relative fold-changes calculated for all taxonomic groups (family and genus levels). (a) Both taxa and treatment groups were subject to HCL analysis. (b) Shows a detail of the results for the groups’ HCL. The KMC analysis roughly revealed the presence of 7 major clusters (C1-C7) based on similarly of taxa relative fold-changes. Legend: C, cluster; E, Epicor; M, moderate subgroup, P, placebo; S, severe subgroup; TC, total cohort; V, visit

References

    1. Gray JR. What is chronic constipation? Definition and diagnosis. Can J Gastroenterol. 2011;25(Suppl B):7B–10B. doi: 10.1155/2011/548024.
    1. Suares NC, Ford AC. Prevalence of, and risk factors for, chronic idiopathic constipation in the community: systematic review and meta-analysis. Am J Gastroenterol. 2011;106:1582–1591. doi: 10.1038/ajg.2011.164.
    1. Bharucha AE. Constipation. Best Pract Res Clin Gastroenterol. 2007;21:709–731. doi: 10.1016/j.bpg.2007.07.001.
    1. Everhart JE, Ruhl CE. Burden of digestive diseases in the United States part I: overall and upper gastrointestinal diseases. Gastroenterology. 2009;136:376–386. doi: 10.1053/j.gastro.2008.12.015.
    1. Peery AF, Dellon ES, Lund J, Crockett SD, McGowan CE, Bulsiewicz WJ, et al. Burden of gastrointestinal disease in the United States: 2012 update. Gastroenterology. 2012;143:1173–1179. doi: 10.1053/j.gastro.2012.08.002.
    1. Nellesen D, Yee K, Chawla A, Lewis BE, Carson RT. A systematic review of the economic and humanistic burden of illness in irritable bowel syndrome and chronic constipation. J Manag Care Pharm. 2013;19:755–764.
    1. Wald A, Scarpignato C, Kamm MA, Mueller-Lissner S, Helfrich I, Schuijt C, et al. The burden of constipation on quality of life: results of a multinational survey. Aliment Pharmacol Ther. 2007;26:227–236. doi: 10.1111/j.1365-2036.2007.03376.x.
    1. Belsey J, Greenfield S, Candy D, Geraint M. Systematic review: impact of constipation on quality of life in adults and children. Aliment Pharmacol Ther. 2010;31:938–949.
    1. Curro D, Ianiro G, Pecere S, Bibbo S, Cammarota G. Probiotics, fibre and herbal medicinal products for functional and inflammatory bowel disorders. Br J Pharmacol. 2017;174:1426–1449. doi: 10.1111/bph.13632.
    1. Irvine EJ, Whitehead WE, Chey WD, Matsueda K, Shaw M, Talley NJ, et al. Design of treatment trials for functional gastrointestinal disorders. Gastroenterology. 2006;130:1538–1551. doi: 10.1053/j.gastro.2005.11.058.
    1. Spiller RC. Problems and challenges in the design of irritable bowel syndrome clinical trials: experience from published trials. Am J Med. 1999;107:91S–97S. doi: 10.1016/S0002-9343(99)00086-8.
    1. Veldhuyzen van Zanten SJ, Cleary C, Talley NJ, Peterson TC, Nyren O, Bradley LA, et al. Drug treatment of functional dyspepsia: a systematic analysis of trial methodology with recommendations for design of future trials. Am J Gastroenterol. 1996;91:660–673.
    1. Veldhuyzen van Zanten SJ, Talley NJ, Bytzer P, Klein KB, Whorwell PJ, Zinsmeister AR. Design of treatment trials for functional gastrointestinal disorders. Gut. 1999;45(Suppl 2):II69–II77.
    1. Buchwald-Werner S, Fujii H, Reule C, Schoen C. Perilla extract improves gastrointestinal discomfort in a randomized placebo controlled double blind human pilot study. BMC Complement Altern Med. 2014;14:173. doi: 10.1186/1472-6882-14-173.
    1. Bassotti G, Villanacci V, Cretoiu D, Cretoiu SM, Becheanu G. Cellular and molecular basis of chronic constipation: taking the functional/idiopathic label out. World J Gastroenterol; China. 2013;19:4099–105.
    1. Moyad MA, Robinson LE, Zawada ETJ, Kittelsrud JM, Chen D-G, Reeves SG, et al. Effects of a modified yeast supplement on cold/flu symptoms. Urol Nurs. 2008;28:50–55.
    1. Moyad MA, Robinson LE, Zawada ET, Kittelsrud J, Chen D-G, Reeves SG, et al. Immunogenic yeast-based fermentate for cold/flu-like symptoms in nonvaccinated individuals. J Altern Complement Med. 2010;16:213–218. doi: 10.1089/acm.2009.0310.
    1. Moyad MA, Robinson LE, Kittelsrud JM, Reeves SG, Weaver SE, Guzman AI, et al. Immunogenic yeast-based fermentation product reduces allergic rhinitis-induced nasal congestion: a randomized, double-blind, placebo-controlled trial. Adv Ther. 2009;26:795–804. doi: 10.1007/s12325-009-0057-y.
    1. Jensen GS, Hart AN, Schauss A. An antiinflammatory immunogen from yeast culture induces activation and alters chemokine receptor expression on human natural killer cells and B lymphocytes in vitro. Nutr Res. 2007;27:327–335. doi: 10.1016/j.nutres.2007.04.008.
    1. Jensen GS, Patterson KM, Yoon I. Yeast culture has anti-inflammatory effects and specifically activates NK cells. Comp Immunol Microbiol Infect Dis. 2008;31:487–500. doi: 10.1016/j.cimid.2007.08.005.
    1. Possemiers S, Pinheiro I, Verhelst A, Van den Abbeele P, Maignien L, Laukens D, et al. A dried yeast fermentate selectively modulates both the luminal and mucosal gut microbiota and protects against inflammation, as studied in an integrated in vitro approach. J Agric Food Chem. 2013;61:9380–9392. doi: 10.1021/jf402137r.
    1. Vinolo MAR, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011;3:858–876. doi: 10.3390/nu3100858.
    1. Havenaar R. Intestinal health functions of colonic microbial metabolites: a review. Benefic Microbes. 2011;2:103–114. doi: 10.3920/BM2011.0003.
    1. Kashyap PC, Marcobal A, Ursell LK, Larauche M, Duboc H, Earle KA, et al. Complex interactions among diet, gastrointestinal transit, and gut microbiota in humanized mice. Gastroenterology. 2013;144:967–977. doi: 10.1053/j.gastro.2013.01.047.
    1. Cherbut C. Motor effects of short-chain fatty acids and lactate in the gastrointestinal tract. Proc Nutr Soc. 2003;62:95–99. doi: 10.1079/PNS2002213.
    1. Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, et al. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology. 2010;138:1772–1782. doi: 10.1053/j.gastro.2010.01.053.
    1. Chassard C, Dapoigny M, Scott KP, Crouzet L, Del’homme C, Marquet P, et al. Functional dysbiosis within the gut microbiota of patients with constipated-irritable bowel syndrome. Aliment Pharmacol Ther. 2012;35:828–838. doi: 10.1111/j.1365-2036.2012.05007.x.
    1. de Meij TGJ, de Groot EFJ, Eck A, Budding AE, Kneepkens CMF, Benninga MA, et al. Characterization of microbiota in children with chronic functional constipation. PLoS One. 2016;11:e0164731. doi: 10.1371/journal.pone.0164731.
    1. de Moraes JG, Motta ME, Beltrao MF, Salviano TL, de Silva GA. Fecal microbiota and diet of children with chronic constipation. Int J Pediatr. 2016;2016:6787269. doi: 10.1155/2016/6787269.
    1. Khalif IL, Quigley EMM, Konovitch EA, Maximova ID. Alterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipation. Dig Liver Dis. 2005;37:838–849. doi: 10.1016/j.dld.2005.06.008.
    1. Zhu L, Liu W, Alkhouri R, Baker RD, Bard JE, Quigley EM, et al. Structural changes in the gut microbiome of constipated patients. Physiol Genomics. 2014;46:679–686. doi: 10.1152/physiolgenomics.00082.2014.
    1. EFSA Panel on Dietetic Products N and A (NDA) Guidance on the scientific requirements for health claims related to the immune system, the gastrointestinal tract and defence against pathogenic microorganisms. EFSA J. 2016;14:4369. doi: 10.2903/j.efsa.2016.4369.
    1. Trust, MAPI Research, Lyon F . Information booklet. 4rd 2015. PAC-QOL Patient-Assessment of Constipation Quality of Life.
    1. Jensen GS, Patterson KM, Barnes J, Schauss AG, Beaman R, Reeves SG, Robinson L. A double-blind placebo-controlled, randomized pilot study: consumption of a high-metabolite immunogen from yeast culture has beneficial effects on erythrocyte health and mucosal immune protection in healthy subjects. Open Nutr. 2008;2:68–75. doi: 10.2174/1874288200802010068.
    1. Suresh K. An overview of randomization techniques: an unbiased assessment of outcome in clinical research. J Hum Reprod Sci. 2011;4:8–11. doi: 10.4103/0974-1208.82352.
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol. 1997;32:920–924. doi: 10.3109/00365529709011203.
    1. Marquis P, De La Loge C, Dubois D, McDermott A, Chassany O. Development and validation of the patient assessment of constipation quality of life questionnaire. Scand J Gastroenterol. 2005;40:540–551. doi: 10.1080/00365520510012208.
    1. Drossman DA. The functional gastrointestinal disorders and the Rome III process. Gastroenterology. 2006;130:1377–1390. doi: 10.1053/j.gastro.2006.03.008.
    1. Cohen S, Kamarck T, Mermelstein R. A global measure of perceived stress. J Health Soc Behav. 1983;24:385–396. doi: 10.2307/2136404.
    1. Zoppi G, Cinquetti M, Luciano A, Benini A, Muner A, Bertazzoni ME. The intestinal ecosystem in chronic functional constipation. Acta Paediatr. 1998;87:836–841. doi: 10.1111/j.1651-2227.1998.tb01547.x.
    1. Vilchez-Vargas R, Junca H, Pieper DH. Metabolic networks, microbial ecology and “omics” technologies: towards understanding in situ biodegradation processes. Environ Microbiol. 2010;12:3089–3104. doi: 10.1111/j.1462-2920.2010.02340.x.
    1. Bohorquez LC, Delgado-Serrano L, Lopez G, Osorio-Forero C, Klepac-Ceraj V, Kolter R, et al. In-depth characterization via complementing culture-independent approaches of the microbial community in an acidic hot spring of the Colombian Andes. Microb Ecol. 2012;63:103–115. doi: 10.1007/s00248-011-9943-3.
    1. Camarinha-Silva A, Jauregui R, Chaves-Moreno D, Oxley APA, Schaumburg F, Becker K, et al. Comparing the anterior nare bacterial community of two discrete human populations using Illumina amplicon sequencing. Environ Microbiol. 2014;16:2939–2952. doi: 10.1111/1462-2920.12362.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–2461. doi: 10.1093/bioinformatics/btq461.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73:5261–5267. doi: 10.1128/AEM.00062-07.
    1. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, Peplies J, et al. SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res. 2007;35:7188–7196. doi: 10.1093/nar/gkm864.
    1. Lorenz MO. Methods of measuring concentration of wealth. J Am Stat Assoc. 1905;9
    1. Wittebolle L, Marzorati M, Clement L, Balloi A, Daffonchio D, Heylen K, et al. Initial community evenness favours functionality under selective stress. Nature. 2009;458:623–626. doi: 10.1038/nature07840.
    1. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, et al. TM4: a free, open-source system for microarray data management and analysis. BioTechniques. 2003;34:374–378.
    1. Garrigues V, Galvez C, Ortiz V, Ponce M, Nos P, Ponce J. Prevalence of constipation: agreement among several criteria and evaluation of the diagnostic accuracy of qualifying symptoms and self-reported definition in a population-based survey in Spain. Am J Epidemiol. 2004;159:520–526. doi: 10.1093/aje/kwh072.
    1. Tuteja AK, Talley NJ, Joos SK, Tolman KG, Hickam DH. Abdominal bloating in employed adults: prevalence, risk factors, and association with other bowel disorders. Am J Gastroenterol. 2008;103:1241–1248. doi: 10.1111/j.1572-0241.2007.01755.x.
    1. Quigley EMM. Microflora modulation of motility. J Neurogastroenterol Motil. 2011;17:140–147. doi: 10.5056/jnm.2011.17.2.140.
    1. Quigley EMM. The enteric microbiota in the pathogenesis and management of constipation. Best Pract Res Clin Gastroenterol. 2011;25:119–126. doi: 10.1016/j.bpg.2011.01.003.
    1. Leung L, Riutta T, Kotecha J, Rosser W. Chronic constipation: an evidence-based review. J Am Board Fam Med. 2011;24:436–451. doi: 10.3122/jabfm.2011.04.100272.
    1. Cherbut C, Aube AC, Blottiere HM, Galmiche JP. Effects of short-chain fatty acids on gastrointestinal motility. Scand J Gastroenterol. 1997;32(Suppl 2):58–61. doi: 10.1080/00365521.1997.11720720.
    1. Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol. 2015;11:577–591. doi: 10.1038/nrendo.2015.128.
    1. De Silva A, Bloom SR. Gut hormones and appetite control: a focus on PYY and GLP-1 as therapeutic targets in obesity. Gut Liver. 2012;6:10–20. doi: 10.5009/gnl.2012.6.1.10.
    1. Shekhar C, Monaghan PJ, Morris J, Issa B, Whorwell PJ, Keevil B, et al. Rome III functional constipation and irritable bowel syndrome with constipation are similar disorders within a spectrum of sensitization, regulated by serotonin. Gastroenterology. 2013;145:744–749. doi: 10.1053/j.gastro.2013.07.014.
    1. Kim S-E, Choi SC, Park KS, Park MI, Shin JE, Lee TH, et al. Change of fecal Flora and Effectiveness of the short-term VSL#3 Probiotic treatment in patients with functional constipation. J Neurogastroenterol Motil. 2015;21:111–120. doi: 10.5056/jnm14048.
    1. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen Y-Y, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105–108. doi: 10.1126/science.1208344.
    1. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107:14691–14696. doi: 10.1073/pnas.1005963107.
    1. Rajilic-Stojanovic M, Biagi E, Heilig HGHJ, Kajander K, Kekkonen RA, Tims S, et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011;141:1792–1801. doi: 10.1053/j.gastro.2011.07.043.
    1. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A. 2013;110:9066–9071. doi: 10.1073/pnas.1219451110.
    1. Bui TPN, de Vos WM, Plugge CM. Anaerostipes rhamnosivorans sp. nov., a human intestinal, butyrate-forming bacterium. Int J Syst Evol Microbiol. 2014;64:787–793. doi: 10.1099/ijs.0.055061-0.
    1. Borody TJ, Warren EF, Leis SM, Surace R, Ashman O, Siarakas S. Bacteriotherapy using fecal flora: toying with human motions. J Clin Gastroenterol. 2004;38:475–483. doi: 10.1097/01.mcg.0000128988.13808.dc.

Source: PubMed

3
Subscribe