Resistant starch type-4 intake alters circulating bile acids in human subjects

Samitinjaya Dhakal, Moul Dey, Samitinjaya Dhakal, Moul Dey

Abstract

Background: Resistant starch (RS) type 4 (RS4) is a type of RS, a class of non-digestible prebiotic dietary fibers with a range of demonstrated metabolic health benefits to the host. On the other hand, bile acids (BA) have recently emerged as an important class of metabolic function mediators that involve host-microbiota interactions. RS consumption alters fecal and cecal BA in humans and rodents, respectively. The effect of RS intake on circulating BA concentrations remains unexplored in humans.

Methods and results: Using available plasma and stool samples from our previously reported double-blind, controlled, 2-arm crossover nutrition intervention trial (Clinicaltrials.gov: NCT01887964), a liquid-chromatography/mass-spectrometry-based targeted multiple reaction monitoring, and absolute quantifications, we assessed BA changes after 12 weeks of an average 12 g/day RS4-intake. Stool BA concentrations were lower post RS4 compared to the control, the two groups consuming similar macronutrients (n = 14/group). Partial least squares-discriminant analysis revealed distinct BA signatures in stool and plasma post interventions. The increased circulating BA concentrations were further investigated using linear mixed-effect modeling that controlled for potential confounders. A higher plasma abundance of several BA species post RS4 was observed (fold increase compared to control in parenthesis): taurocholic acid (1.92), taurodeoxycholic acid (1.60), glycochenodeoxycholic acid (1.58), glycodeoxycholic acid (1.79), and deoxycholic acid (1.77) (all, p < 0.05). Distinct microbiome ortholog-signatures were observed between RS4 and control groups (95% CI), derived using the Piphillin function-prediction algorithm and principal component analysis (PCA) of pre-existing 16S rRNA gene sequences. Association of Bifidobacterium adolescentis with secondary BA such as, deoxycholic acid (rho = 0.55, p = 0.05), glycodeoxycholic acid (rho = 0.65, p = 0.02), and taurodeoxycholic acid (rho = 0.56, p = 0.04) were observed in the RS4-group, but not in the control group (all, p > 0.05).

Conclusion: Our observations indicate a previously unknown in humans- RS4-associated systemic alteration of microbiota-derived secondary BA. Follow-up investigations of BA biosynthesis in the context of RS4 may provide molecular targets to understand and manipulate microbiome-host interactions.

Keywords: circulating bile acid; dietary fiber; metabolic syndrome; metabolomics; microbiota; resistant starch type 4.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Dhakal and Dey.

Figures

FIGURE 1
FIGURE 1
Participant flow in the study.
FIGURE 2
FIGURE 2
Estimated nutrient intakes at baseline and post diets. Bar graphs with mean ± SD showing similar macronutrient intakes at baseline and after interventions (all, p > 0.05). The difference in fiber intake was due to RS4 supplementation in the test group; n = 14/group; RS4, resistant starch type 4; CHO, Carbohydrates.
FIGURE 3
FIGURE 3
Effects of RS4 intervention on plasma and stool bile acids. Multivariate partial least squares-discriminant analysis (PLS-DA) score plots of control (brown) vs. RS4 (blue) in plasma (A) and stool (B) bile acids with their corresponding variable importance in projection (VIP) scores plots; Each dot represents the unique signature of 15 primary and secondary bile acid species for an individual; species with VIP > 1 are shown (C) bar graph showing fold change in bile acid group abundance; p calculated using linear mixed-effect model controlling for body weight, intervention sequence, and participant ID; n = 14/group; RS4, resistant starch type 4; LV, latent variable; CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; LCA, lithocholic acid; G, T, and U represent glyco-, tauro-, and urso-, respectively.
FIGURE 4
FIGURE 4
RS4 altered secondary bile acids. Box plots comparing concentrations of individual primary, secondary, and conjugated bile acid species: at baseline and after RS4 intake (A) after control vs. RS4 interventions (B); for (A,B)p-values were calculated using linear mixed-effect model controlling for body weight, intervention sequence, and participant ID; lower and upper hinges of the boxplot denote 25th and 75th percentile, line denotes median, and whiskers are drawn to minimum and maximum values but not further than 1.5x interquartile range. Outliers are displayed as black dots; (C) heatmap showing individual study participants on x-axis and bile acid species on y-axis to present differential abundance of circulating bile acids between individuals and groups; the colored cells denote abundance (red- high abundance, blue- low abundance); variables were autoscaled; For all panels: n = 14/group; RS4, resistant starch type 4; CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; LCA, lithocholic acid; G, T and U represent glyco-, tauro-, and urso-, respectively.
FIGURE 5
FIGURE 5
Impact of RS4 on gut microbiota functional capability. (A) Principal component analysis (PCA) of KEGG orthologs based on 16s rRNA gene sequences showing higher variability in control as illustrated by wider 95% CI (elliptical boundaries) vs. RS4 intervention that clustered the predicted orthologs toward zero on both axes within a narrow spatial area; the x and y axes explain 31 and 16.5% variability, respectively, in the control vs. RS4 the distinction between diets was less evident with baseline vs. RS4; each dot represents a unique signature of an individual participant’s gut microbiota for the 8,621 KEGG orthologs; dataset was log transformed. (B) Heatmap showing individual study participants on x-axis and KEGG orthologs on y-axis to present differential abundance of orthologs between individuals and groups; the colored cells denote abundance (red- high abundance, blue- low abundance); variables were autoscaled; and n = 14/group; RS4, resistant starch type 4; KEGG, Kyoto encyclopedia of genes and genomes.

References

    1. Fiorucci S, Mencarelli A, Palladino G, Cipriani S. Bile-acid-activated receptors: targeting tgr5 and farnesoid-X-receptor in lipid and glucose disorders. Trends Pharmacol Sci. (2009) 30:570–80. 10.1016/j.tips.2009.08.001
    1. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, et al. Tgr5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. (2009) 10:167–77. 10.1016/j.cmet.2009.08.001
    1. Chiang JY. Bile acids: regulation of synthesis. J Lip Res. (2009) 50:1955–66. 10.1194/jlr.R900010-JLR200
    1. Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. (2016) 24:41–50. 10.1016/j.cmet.2016.05.005
    1. Staley C, Weingarden AR, Khoruts A, Sadowsky MJ. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl Microbiol Biotechnol. (2017) 101:47–64. 10.1007/s00253-016-8006-6
    1. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol. (2014) 30:332–8. 10.1097/mog.0000000000000057
    1. Li T, Chiang JY. Bile acids as metabolic regulators. Curr Opin Gastroenterol. (2015) 31:159–65. 10.1097/mog.0000000000000156
    1. Upadhyaya B, McCormack L, Fardin-Kia AR, Juenemann R, Nichenametla S, Clapper J, et al. Impact of Dietary resistant starch type 4 on human gut microbiota and immunometabolic functions. Sci Rep. (2016) 6:28797. 10.1038/srep28797
    1. Martínez I, Kim J, Duffy PR, Schlegel VL, Walter J. Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects. PLoS One. (2010) 5:e15046. 10.1371/journal.pone.0015046
    1. Bindels LB, Walter J, Ramer-Tait AE. Resistant starches for the management of metabolic diseases. Curr Opin Clin Nutr Metab Care. (2015) 18:559–65. 10.1097/mco.0000000000000223
    1. Wanders AJ, van den Borne JJ, de Graaf C, Hulshof T, Jonathan MC, Kristensen M, et al. Effects of dietary fibre on subjective appetite, energy intake and body weight: a systematic review of randomized controlled trials. Obesity Rev Offi J Int Assoc Study Obesity. (2011) 12:724–39. 10.1111/j.1467-789X.2011.00895.x
    1. Clark MJ, Slavin JL. The effect of fiber on satiety and food intake: a systematic review. J Am College Nutr. (2013) 32:200–11. 10.1080/07315724.2013.791194
    1. Haub MD, Hubach KL, Al-Tamimi EK, Ornelas S, Seib PA. Different types of resistant starch elicit different glucose reponses in humans. J Nutr Metab. (2010) 2010:230501. 10.1155/2010/230501
    1. Englyst HN, Kingman SM, Cummings JH. Classification and measurement of nutritionally important starch fractions. Eur J Clin Nutr. (1992) 46:S33–50.
    1. Si X, Zhou Z, Strappe P, Blanchard CA. Comparison of Rs4-type resistant starch to Rs2-type resistant starch in suppressing oxidative stress in high-fat-diet-induced obese rats. Food Funct. (2017) 8:232–40. 10.1039/c6fo01225f
    1. Bindels LB, Segura Munoz RR, Gomes-Neto JC, Mutemberezi V, Martínez I, Salazar N, et al. Resistant starch can improve insulin sensitivity independently of the gut microbiota. Microbiome. (2017) 5:12. 10.1186/s40168-017-0230-5
    1. van Munster IP, Tangerman A, Nagengast FM. Effect of resistant starch on colonic fermentation, bile acid metabolism, and mucosal proliferation. Digest Dis Sci. (1994) 39:834–42. 10.1007/bf02087431
    1. Langkilde AM, Ekwall H, Björck I, Asp NG, Andersson H. Retrograded high-amylose corn starch reduces cholic acid excretion from the small bowel in ileostomy subjects. Eur J Clin Nutr. (1998) 52:790–5. 10.1038/sj.ejcn.1600648
    1. Grubben MJ, van den Braak CC, Essenberg M, Olthof M, Tangerman A, Katan MB, et al. Effect of resistant starch on potential biomarkers for colonic cancer risk in patients with colonic adenomas: a controlled trial. Digest Dis Sci. (2001) 46:750–6. 10.1023/a:1010787931002
    1. Hylla S, Gostner A, Dusel G, Anger H, Bartram HP, Christl SU, et al. Effects of resistant starch on the colon in healthy volunteers: possible implications for cancer prevention. Am J Clin Nutr. (1998) 67:136–42. 10.1093/ajcn/67.1.136
    1. Jacobasch G, Dongowski G, Schmiedl D, Müller-Schmehl K. Hydrothermal treatment of novelose 330 results in high yield of resistant starch type 3 with beneficial prebiotic properties and decreased secondary bile acid formation in rats. Br J Nutr. (2006) 95:1063–74. 10.1079/bjn20061713
    1. Trautwein EA, Forgbert K, Rieckhoff D, Erbersdobler HF. Impact of beta-cyclodextrin and resistant starch on bile acid metabolism and fecal steroid excretion in regard to their hypolipidemic action in hamsters. Biochim Biophys Acta. (1999) 1437:1–12. 10.1016/s0005-2760(98)00174-x
    1. Dongowski G, Jacobasch G, Schmiedl D. Structural stability and prebiotic properties of resistant starch type 3 increase bile acid turnover and lower secondary bile acid formation. J Agric Food Chem. (2005) 53:9257–67. 10.1021/jf0507792
    1. Han KH, Iijuka M, Shimada K, Sekikawa M, Kuramochi K, Ohba K, et al. Adzuki resistant starch lowered serum cholesterol and hepatic 3-hydroxy-3-methylglutaryl-coa mrna levels and increased hepatic ldl-receptor and cholesterol 7alpha-hydroxylase mrna levels in rats fed a cholesterol diet. Br J Nutr. (2005) 94:902–8. 10.1079/bjn20051598
    1. Liu X, Ogawa H, Kishida T, Ebihara K. Hypolipidaemic effect of maize starch with different amylose content in ovariectomized rats depends on intake amount of resistant starch. Br J Nutr. (2009) 101:328–39. 10.1017/s000711450801221x
    1. Verbeek MJ, De Deckere EA, Tijburg LB, Van Amelsvoort JM, Beynen AC. Influence of dietary retrograded starch on the metabolism of neutral steroids and bile acids in rats. Br J Nutr. (1995) 74:807–20.
    1. Shang W, Si X, Zhou Z, Li Y, Strappe P, Blanchard C. Characterization of fecal fat composition and gut derived fecal microbiota in high-fat diet fed rats following intervention with chito-oligosaccharide and resistant starch complexes. Food Funct. (2017) 8:4374–83. 10.1039/c7fo01244f
    1. Koay YC, Wali JA, Luk AWS, Macia L, Cogger VC, Pulpitel TJ, et al. Ingestion of resistant starch by mice markedly increases microbiome-derived metabolites. FASEB J Offi Publi Federat Am Soc Exp Biol. (2019) 33:8033–42. 10.1096/fj.201900177R
    1. Nichenametla SN, Weidauer LA, Wey HE, Beare TM, Specker BL, Dey M. Resistant starch type 4-enriched diet lowered blood cholesterols and improved body composition in a double blind controlled cross-over intervention. Mol Nutr Food Res. (2014) 58:1365–9. 10.1002/mnfr.201300829
    1. Woo KS, Seib PA. Cross-linked resistant starch: preparation and properties. Cereal Chem. (2002) 79:819–25. 10.1094/CCHEM.2002.79.6.819
    1. Emilien CH, Hsu WH, Hollis JH. Effect of resistant wheat starch on subjective appetite and food intake in healthy adults. Nutrition. (2017) 44:69–74. 10.1016/j.nut.2017.06.020
    1. Steele TJ, Maningat CC, Seib PA, Haub MD, Rosenkranz SK. Metabolic responses to native wheat starch (Midsol(Tm) 50) versus resistant wheat starch type 4 (Fibersym(®) Rw): standard versus marketplace testing protocols. Curr Dev Nutr. (2021) 5:nzab011. 10.1093/cdn/nzab011
    1. Mah E, Garcia-Campayo V, Liska D. Substitution of corn starch with resistant starch type 4 in a breakfast bar decreases postprandial glucose and insulin responses: a randomized, controlled, crossover study. Curr Dev Nutr. (2018) 2:nzy066. 10.1093/cdn/nzy066
    1. Stewart ML, Wilcox ML, Bell M, Buggia MA, Maki KC. Type-4 resistant starch in substitution for available carbohydrate reduces postprandial glycemic response and hunger in acute, randomized, double-blind, controlled study. Nutrients. (2018) 10:20129. 10.3390/nu10020129
    1. Barupal DK, Zhang Y, Shen T, Fan S, Roberts BS, Fitzgerald P, et al. A comprehensive plasma metabolomics dataset for a cohort of mouse knockouts within the international mouse phenotyping consortium. Metabolites. (2019) 9:50101. 10.3390/metabo9050101
    1. Wei R, Li G, Seymour AB. High-throughput and multiplexed Lc/Ms/Mrm method for targeted metabolomics. Analy Chem. (2010) 82:5527–33. 10.1021/ac100331b
    1. Cajka T, Fiehn O. Toward merging untargeted and targeted methods in mass spectrometry-based metabolomics and lipidomics. Analy Chem. (2016) 88:524–45. 10.1021/acs.analchem.5b04491
    1. La Frano MR, Hernandez-Carretero A, Weber N, Borkowski K, Pedersen TL, Osborn O, et al. Diet-induced obesity and weight loss alter bile acid concentrations and bile acid-sensitive gene expression in insulin target tissues of C57bl/6j mice. Nutr Res. (2017) 46:11–21. 10.1016/j.nutres.2017.07.006
    1. Dhakal S, McCormack L, Dey M. Association of the gut microbiota with weight-loss response within a retail weight-management program. Microorganisms. (2020) 8:1246. 10.3390/microorganisms8081246
    1. Dhakal S, Moazzami Z, Perry C, Dey M. Effects of lean pork on microbiota and microbial-metabolite trimethylamine-N-oxide: a randomized controlled non-inferiority feeding trial based on the dietary guidelines for americans. Mol Nutr Food Res. (2022) 2022:e2101136. 10.1002/mnfr.202101136
    1. Narayan NR, Weinmaier T, Laserna-Mendieta EJ, Claesson MJ, Shanahan F, Dabbagh K, et al. Piphillin predicts metagenomic composition and dynamics from dada2-corrected 16s rdna sequences. BMC Genomics. (2020) 21:56. 10.1186/s12864-019-6427-1
    1. Iwai S, Weinmaier T, Schmidt BL, Albertson DG, Poloso NJ, Dabbagh K, et al. Piphillin: improved prediction of metagenomic content by direct inference from human microbiomes. PLoS One. (2016) 11:e0166104. 10.1371/journal.pone.0166104
    1. Chong J, Liu P, Zhou G, Xia J. Using microbiomeanalyst for comprehensive statistical, functional, and meta-analysis of microbiome data. Nat Proto. (2020) 15:799–821. 10.1038/s41596-019-0264-1
    1. Pang Z, Chong J, Zhou G, de Lima Morais DA, Chang L, Barrette M, et al. Metaboanalyst 5.0: narrowing the gap between raw spectra and functional insights. Nucleic Acids Res. (2021) 49:W388–96. 10.1093/nar/gkab382
    1. Fava F, Gitau R, Griffin BA, Gibson GR, Tuohy KM, Lovegrove JA. The type and quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty acid excretion in a metabolic syndrome ‘at-risk’ population. Int J Obes. (2013) 37:216–23. 10.1038/ijo.2012.33
    1. Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr. (2011) 94:58–65. 10.3945/ajcn.110.010132
    1. Trumbo P, Schlicker S, Yates AA, Poos M. Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein and amino acids. J Am Dietetic Assoc. (2002) 102:1621–30. 10.1016/s0002-8223(02)90346-9
    1. Snetselaar LG, de Jesus JM, DeSilva DM, Stoody EE. Dietary guidelines for americans, 2020–2025. Nutr Today. (2021) 56:287–95.
    1. Ginos BNR, Navarro SL, Schwarz Y, Gu H, Wang D, Randolph TW, et al. Circulating bile acids in healthy adults respond differently to a dietary pattern characterized by whole grains, legumes and fruits and vegetables compared to a diet high in refined grains and added sugars: a randomized, controlled, crossover feeding study. Metabolism. (2018) 83:197–204. 10.1016/j.metabol.2018.02.006
    1. Chiang JY. Recent advances in understanding bile acid homeostasis. F1000Res. (2017) 6:2029. 10.12688/f1000research.12449.1
    1. Staels B, Fonseca VA. Bile acids and metabolic regulation: mechanisms and clinical responses to bile acid sequestration. Diabetes Care. (2009) 32:S237–45. 10.2337/dc09-S355
    1. Jiang Z, Sun TY, He Y, Gou W, Zuo LS, Fu Y, et al. Dietary fruit and vegetable intake, gut microbiota, and type 2 diabetes: results from two large human cohort studies. BMC Med. (2020) 18:371. 10.1186/s12916-020-01842-0
    1. Lu J, Wang S, Li M, Gao Z, Xu Y, Zhao X, et al. Association of serum bile acids profile and pathway dysregulation with the risk of developing diabetes among normoglycemic chinese adults: findings from the 4c study. Diabetes Care. (2021) 44:499–510. 10.2337/dc20-0884
    1. Paik D, Yao L, Zhang Y, Bae S, D’Agostino GD, Zhang M, et al. Human gut bacteria produce T (H)17-modulating bile acid metabolites. Nature. (2022) 603:907–12. 10.1038/s41586-022-04480-z
    1. Farr S, Stankovic B, Hoffman S, Masoudpoor H, Baker C, Taher J, et al. Bile acid treatment and Fxr agonism lower postprandial lipemia in mice. Am J Physiol Gastrointest Liver Physiol. (2020) 318:G682–93. 10.1152/ajpgi.00386.2018
    1. Sinha SR, Haileselassie Y, Nguyen LP, Tropini C, Wang M, Becker LS, et al. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe. (2020) 27:659.e–70.e. 10.1016/j.chom.2020.01.021
    1. Frommherz L, Bub A, Hummel E, Rist MJ, Roth A, Watzl B, et al. Age-related changes of plasma bile acid concentrations in healthy adults–results from the cross-sectional karmen study. PLoS One. (2016) 11:e0153959. 10.1371/journal.pone.0153959
    1. Ocvirk S, Wilson AS, Posma JM, Li JV, Koller KR, Day GM, et al. Prospective cohort analysis of gut microbial co-metabolism in alaska native and rural african people at high and low risk of colorectal cancer. Am J Clin Nutr. (2020) 111:406–19. 10.1093/ajcn/nqz301
    1. Winston JA, Rivera A, Cai J, Patterson AD, Theriot CM. Secondary bile acid ursodeoxycholic acid alters weight, the gut microbiota, and the bile acid pool in conventional mice. PLoS One. (2021) 16(2):e0246161. 10.1371/journal.pone.0246161
    1. Jung DH, Kim GY, Kim IY, Seo DH, Nam YD, Kang H, et al. Bifidobacterium adolescentis P2p3, a human gut bacterium having strong non-gelatinized resistant starch-degrading activity. J Microbiol Biotechnol. (2019) 29:1904–15. 10.4014/jmb.1909.09010
    1. Duranti S, Milani C, Lugli GA, Mancabelli L, Turroni F, Ferrario C, et al. Evaluation of genetic diversity among strains of the human gut commensal bifidobacterium adolescentis. Sci Rep. (2016) 6:23971. 10.1038/srep23971
    1. Tsai CC, Lin PP, Hsieh YM, Zhang ZY, Wu HC, Huang CC. Cholesterol-lowering potentials of lactic acid bacteria based on bile-salt hydrolase activity and effect of potent strains on cholesterol metabolism in vitro and in vivo. Sci World J. (2014) 2014:690752. 10.1155/2014/690752
    1. Choi SB, Lew LC, Yeo SK, Nair Parvathy S, Liong MT. Probiotics and the Bsh-related cholesterol lowering mechanism: a jekyll and hyde scenario. Crit Rev Biotechnol. (2015) 35:392–401. 10.3109/07388551.2014.889077

Source: PubMed

3
Subscribe