Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects

Inés Martínez, Jaehyoung Kim, Patrick R Duffy, Vicki L Schlegel, Jens Walter, Inés Martínez, Jaehyoung Kim, Patrick R Duffy, Vicki L Schlegel, Jens Walter

Abstract

Background: To systematically develop dietary strategies based on resistant starch (RS) that modulate the human gut microbiome, detailed in vivo studies that evaluate the effects of different forms of RS on the community structure and population dynamics of the gut microbiota are necessary. The aim of the present study was to gain a community wide perspective of the effects of RS types 2 (RS2) and 4 (RS4) on the fecal microbiota in human individuals.

Methods and findings: Ten human subjects consumed crackers for three weeks each containing either RS2, RS4, or native starch in a double-blind, crossover design. Multiplex sequencing of 16S rRNA tags revealed that both types of RS induced several significant compositional alterations in the fecal microbial populations, with differential effects on community structure. RS4 but not RS2 induced phylum-level changes, significantly increasing Actinobacteria and Bacteroidetes while decreasing Firmicutes. At the species level, the changes evoked by RS4 were increases in Bifidobacterium adolescentis and Parabacteroides distasonis, while RS2 significantly raised the proportions of Ruminococcus bromii and Eubacterium rectale when compared to RS4. The population shifts caused by RS4 were numerically substantial for several taxa, leading for example, to a ten-fold increase in bifidobacteria in three of the subjects, enriching them to 18-30% of the fecal microbial community. The responses to RS and their magnitudes varied between individuals, and they were reversible and tightly associated with the consumption of RS.

Conclusion: Our results demonstrate that RS2 and RS4 show functional differences in their effect on human fecal microbiota composition, indicating that the chemical structure of RS determines its accessibility by groups of colonic bacteria. The findings imply that specific bacterial populations could be selectively targeted by well designed functional carbohydrates, but the inter-subject variations in the response to RS indicates that such strategies might benefit from more personalized approaches.

Conflict of interest statement

Competing Interests: The authors confirm that there has been no interference with an objective assessment of the manuscript, and that they adhere to all the PLoS ONE policies regarding the sharing of data and materials.

Figures

Figure 1. Experimental design used in this…
Figure 1. Experimental design used in this study.
Subjects (n = 10) participated in a 17-week double-blind crossover design, in which 3 dietary treatments were assessed: 100 g of crackers containing either native starch or 33 g of RS2 or RS4. An initial baseline period was proceeded by 3-week periods of each dietary treatment in succession interspersed by 2-week washout periods, and a final washout period. Weekly fecal samples were collected throughout the entire study.
Figure 2. Characterization of the fecal microbiota…
Figure 2. Characterization of the fecal microbiota in ten human subjects that consumed a random succession of crackers containing RS2, RS4, and native wheat starch (control) by multiplex pyrosequencing of 16S rRNA tags.
Phylogenetic trees that encompass the phyla (A) Firmicutes (with Clostridiales groups XIVa and IV labeled), (B) Actinobacteria and (C) Bacteroidetes are shown. The trees contain representative sequences of all OTUs detected to be impacted by RS in individual subjects together with sequences of related entries in the database (which included both type strains of known species and sequences from molecular studies of human fecal samples). Sequences were aligned in Muscle 3.6 and the trees were built using the neighbor-joining algorithm with 1,000 bootstrap replicates in MEGA 4.0. Open-black and closed-gray symbols were used to label sequences from individual subjects. OTUs that were not significantly affected in all ten subjects were labeled as ‘not significant’ (NS). The graphs next to the trees show the abundance of OTUs and bacterial groups that were significantly altered in the treatment groups (RS2, RS4, control). These graphs show mean proportions of the three individual samples taken during the treatment periods for each subject. Background refers to samples taken in periods were no crackers were consumed. Repeated measures ANOVA in combination with a Tukey's post-hoc test were performed to indentify differences between treatment groups, and the background was not included in the statistic analysis. *p<0.05; **p<0.01; ***p<0.001.
Figure 3. Bubble plots showing differences in…
Figure 3. Bubble plots showing differences in the proportions of bacterial taxa (as per cent of the total microbiota composition) detected between the RS4 (A) and RS2 (B) periods when compared to the control period.
The sizes of the bubbles are proportional to the magnitude of the difference. Black circles represent increases in proportions induced through RS treatment, and white circles show a decrease.
Figure 4. Temporal dynamics of the human…
Figure 4. Temporal dynamics of the human fecal microbiota in response to the consumption of crackers containing RS2, RS4, and native wheat starch (control) in five human subjects.
Graphs on the left show proportions of the three main phyla and four representative species (Bifidobacterium adolescentis, Parabacteroides distasonis, Ruminococcus bromii and Clostridium clostridioforme) as determined by pyrosequencing of 16S rRNA tags. Gel images on the right show molecular fingerprints generated by DGGE. Bands that represent Bifidobacterium adolescentis and Parabacteroides distasonis are labeled.

References

    1. Gordon HA, Pesti L. The gnotobiotic animal as a tool in the study of host microbial relationships. Bacteriol Rev. 1971;35:390–429.
    1. Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and personalized health care. Nat Rev Microbiol. 2005;3:431–438.
    1. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia. 2007;50:2374–2383.
    1. Flint HJ, Duncan SH, Scott KP, Louis P. Interactions and competition within the microbial community of the human colon: links between diet and health. Environ Microbiol. 2007;9:1101–1111.
    1. Tannock GW. The search for disease-associated compositional shifts in bowel bacterial communities of humans. Trends Microbiol. 2008;16:488–495.
    1. Louis P, Scott KP, Duncan SH, Flint HJ. Understanding the effects of diet on bacterial metabolism in the large intestine. J Appl Microbiol. 2007;102:1197–1208.
    1. Asp NG. Resistant starch. Proceedings from the second plenary meeting of EURESTA: European FLAIR-Concerted Action No 11 on the physiological implications of the consumption of resistant starch in man. Eur J Clin Nutr. 1992;46(Suppl2):S1.
    1. Behall KM, Scholfield DJ, Hallfrisch JG, Liljeberg-Elmstahl HG. Consumption of both resistant starch and beta-glucan improves postprandial plasma glucose and insulin in women. Diabetes Care. 2006;29:976–981.
    1. Nilsson AC, Ostman EM, Holst JJ, Bjorck IM. Including indigestible carbohydrates in the evening meal of healthy subjects improves glucose tolerance, lowers inflammatory markers, and increases satiety after a subsequent standardized breakfast. J Nutr. 2008;138:732–739.
    1. Nugent AP. Health properties of resistant starch. Nutr Bull. 2005;30:27–54.
    1. Bauer-Marinovic M, Florian S, Muller-Schmehl K, Glatt H, Jacobasch G. Dietary resistant starch type 3 prevents tumor induction by 1,2-dimethylhydrazine and alters proliferation, apoptosis and dedifferentiation in rat colon. Carcinogenesis. 2006;27:1849–1859.
    1. Le Leu RK, Brown IL, Hu Y, Esterman A, Young GP. Suppression of azoxymethane-induced colon cancer development in rats by dietary resistant starch. Cancer Biol Ther. 2007;6:1621–1626.
    1. Moreau NM, Martin LJ, Toquet CS, Laboisse CL, Nguyen PG, et al. Restoration of the integrity of rat caeco-colonic mucosa by resistant starch, but not by fructo-oligosaccharides, in dextran sulfate sodium-induced experimental colitis. Br J Nutr. 2003;90:75–85.
    1. Toden S, Bird AR, Topping DL, Conlon MA. Resistant starch prevents colonic DNA damage induced by high dietary cooked red meat or casein in rats. Cancer Biol Ther. 2006;5:267–272.
    1. Conlon MA, Bird AR. Interactive and individual effects of dietary non-digestible carbohydrates and oils on DNA damage, SCFA and bacteria in the large bowel of rats. Br J Nutr. 2009;101:1171–1177.
    1. Drzikova B, Dongowski G, Gebhardt E. Dietary fibre-rich oat-based products affect serum lipids, microbiota, formation of short-chain fatty acids and steroids in rats. Br J Nutr. 2005;94:1012–1025.
    1. Kleessen B, Stoof G, Proll J, Schmiedl D, Noack J, et al. Feeding resistant starch affects fecal and cecal microflora and short-chain fatty acids in rats. J Anim Sci. 1997;75:2453–2462.
    1. Wang X, Brown IL, Khaled D, Mahoney MC, Evans AJ, et al. Manipulation of colonic bacteria and volatile fatty acid production by dietary high amylose maize (amylomaize) starch granules. J Appl Microbiol. 2002;93:390–397.
    1. Lesmes U, Beards EJ, Gibson GR, Tuohy KM, Shimoni E. Effects of resistant starch type III polymorphs on human colon microbiota and short chain fatty acids in human gut models. J Agric Food Chem. 2008;56:5415–5421.
    1. Silvi S, Rumney CJ, Cresci A, Rowland IR. Resistant starch modifies gut microflora and microbial metabolism in human flora-associated rats inoculated with faeces from Italian and UK donors. J Appl Microbiol. 1999;86:521–530.
    1. Crittenden R, Playne MR. Modifying the human intestinal microbiota with prebiotics. In: Ouwehand AC, Vaughan EE, editors. Gastrointestinal microbiology. New York: Taylor and Francis; 2006.
    1. Abell GC, Cooke CM, Bennett CN, Conlon MA, McOrist AL. Phylotypes related to Ruminococcus bromii are abundant in the large bowel of humans and increase in response to a diet high in resistant starch. FEMS Microbiol Ecol. 2008;66:505–515.
    1. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J Epub ahead of print
    1. Robinson CJ, Bohannan BJ, Young VB. From structure to function: the ecology of host-associated microbial communities. Microbiol Mol Biol Rev. 74:453–476.
    1. Hamady M, Knight R. Microbial community profiling for human microbiome projects: Tools, techniques, and challenges. Genome Res. 2009;19:1141–1152.
    1. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6:e280.
    1. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A. 107:11971–11975.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484.
    1. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–1023.
    1. Claesson MJ, O'Sullivan O, Wang Q, Nikkila J, Marchesi JR, et al. Comparative analysis of pyrosequencing and a phylogenetic microarray for exploring microbial community structures in the human distal intestine. PLoS One. 2009;4:e6669.
    1. Martinez I, Wallace G, Zhang C, Legge R, Benson AK, et al. Diet-induced metabolic improvements in a hamster model of hypercholesterolemia are strongly linked to alterations of the gut microbiota. Appl Environ Microbiol. 2009;75:4175–4184.
    1. Tannock GW, Munro K, Bibiloni R, Simon MA, Hargreaves P, et al. Impact of consumption of oligosaccharide-containing biscuits on the fecal microbiota of humans. Appl Environ Microbiol. 2004;70:2129–2136.
    1. Tannock GW, Munro K, Harmsen HJ, Welling GW, Smart J, et al. Analysis of the fecal microflora of human subjects consuming a probiotic product containing Lactobacillus rhamnosus DR20. Appl Environ Microbiol. 2000;66:2578–2588.
    1. Kovatcheva-Datchary P, Egert M, Maathuis A, Rajilic-Stojanovic M, de Graaf AA, et al. Linking phylogenetic identities of bacteria to starch fermentation in an in vitro model of the large intestine by RNA-based stable isotope probing. Environ Microbiol. 2009;11:914–926.
    1. Leitch EC, Walker AW, Duncan SH, Holtrop G, Flint HJ. Selective colonization of insoluble substrates by human faecal bacteria. Environ Microbiol. 2007;9:667–679.
    1. Bird AR, Topping DL. Resistant starch as a prebiotic. In: Versalovic J, Wilson M, editors. Therapeutic microbiology: Prebiotics and related strategies. Washington, DC, USA: ASM Press; 2008. pp. 159–173.
    1. Cho KH, Cho D, Wang GR, Salyers AA. New regulatory gene that contributes to control of Bacteroides thetaiotaomicron starch utilization genes. J Bacteriol. 2001;183:7198–7205.
    1. D'Elia JN, Salyers AA. Contribution of a neopullulanase, a pullulanase, and an alpha-glucosidase to growth of Bacteroides thetaiotaomicron on starch. J Bacteriol. 1996;178:7173–7179.
    1. Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, et al. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol. 2007;73:1073–1078.
    1. Flint HJ, Bayer EA, Rincon MT, Lamed R, White BA. Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis. Nat Rev Microbiol. 2008;6:121–131.
    1. Ryan KJ. Normal Microbial Flora. In: Ryan KJ, Ray CG, editors. Medical Microbiology An intorduction to infectious diseases. 4th ed. New York: McGraw-Hill; 2004.
    1. Schell MA, Karmirantzou M, Snel B, Vilanova D, Berger B, et al. The genome sequence of Bifidobacterium longum reflects its adaptation to the human gastrointestinal tract. Proc Natl Acad Sci U S A. 2002;99:14422–14427.
    1. Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, et al. A genomic view of the human-Bacteroides thetaiotaomicron symbiosis. Science. 2003;299:2074–2076.
    1. Tap J, Mondot S, Levenez F, Pelletier E, Caron C, et al. Towards the human intestinal microbiota phylogenetic core. Environ Microbiol. 2009;11:2574–2584.
    1. Crittenden R, Laitila A, Forssell P, Matto J, Saarela M, et al. Adhesion of bifidobacteria to granular starch and its implications in probiotic technologies. Appl Environ Microbiol. 2001;67:3469–3475.
    1. Ramsay AG, Scott KP, Martin JC, Rincon MT, Flint HJ. Cell-associated alpha-amylases of butyrate-producing Firmicute bacteria from the human colon. Microbiology. 2006;152:3281–3290.
    1. McIntyre A, Gibson PR, Young GP. Butyrate production from dietary fibre and protection against large bowel cancer in a rat model. Gut. 1993;34:386–391.
    1. Scheppach W, Sommer H, Kirchner T, Paganelli GM, Bartram P, et al. Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis. Gastroenterology. 1992;103:51–56.
    1. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–848.
    1. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031.
    1. Cani PD, Delzenne NM. Interplay between obesity and associated metabolic disorders: new insights into the gut microbiota. Curr Opin Pharmacol. 2009;9:737–743.
    1. Woo KS, Seib PS. Cross-linked resitant starch: Preparation and properties. Cereal Chem. 2002;79:819–825.
    1. Cole JR, Wang Q, Cardenas E, Fish J, Chai B, et al. The Ribosomal Database Project: improved alignments and new tools for rRNA analysis. Nucleic Acids Res. 2009;37:D141–145.
    1. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 2004;32:1792–1797.
    1. Tamura K, Dudley J, Nei M, Kumar S. MEGA4: Molecular Evolutionary Genetics Analysis (MEGA) software version 4.0. Mol Biol Evol. 2007;24:1596–1599.
    1. Hall TA. BioEdit: a user-friendly biological sequence alignment editor analysis program for Windows 95/98/NT. Nucleic Acids Symposium Series. 1999;41:95–98.
    1. Walter J, Hertel C, Tannock GW, Lis CM, Munro K, et al. Detection of Lactobacillus, Pediococcus, Leuconostoc, and Weissella species in human feces by using group-specific PCR primers and denaturing gradient gel electrophoresis. Appl Environ Microbiol. 2001;67:2578–2585.
    1. Walter J, Tannock GW, Tilsala-Timisjarvi A, Rodtong S, Loach DM, et al. Detection and identification of gastrointestinal Lactobacillus species by using denaturing gradient gel electrophoresis and species-specific PCR primers. Appl Environ Microbiol. 2000;66:297–303.
    1. Rinttila T, Kassinen A, Malinen E, Krogius L, Palva A. Development of an extensive set of 16S rDNA-targeted primers for quantification of pathogenic and indigenous bacteria in faecal samples by real-time PCR. J Appl Microbiol. 2004;97:1166–1177.

Source: PubMed

3
Abonnieren