Ileal microbial shifts after Roux-en-Y gastric bypass orchestrate changes in glucose metabolism through modulation of bile acids and L-cell adaptation

Jerry T Dang, Valentin Mocanu, Heekuk Park, Michael Laffin, Caroline Tran, Naomi Hotte, Shahzeer Karmali, Daniel W Birch, Karen Madsen, Jerry T Dang, Valentin Mocanu, Heekuk Park, Michael Laffin, Caroline Tran, Naomi Hotte, Shahzeer Karmali, Daniel W Birch, Karen Madsen

Abstract

Roux-en-Y gastric bypass (RYGB)-induced glycemic improvement is associated with increases in glucagon-like-peptide-1 (GLP-1) secreted from ileal L-cells. We analyzed changes in ileal bile acids and ileal microbial composition in diet-induced-obesity rats after RYGB or sham surgery to elucidate the early and late effects on L-cells and glucose homeostasis. In early cohorts, there were no significant changes in L-cell density, GLP-1 or glucose tolerance. In late cohorts, RYGB demonstrated less weight regain, improved glucose tolerance, increased L-cell density, and increased villi height. No difference in the expression of GLP-1 genes was observed. There were lower concentrations of ileal bile acids in the late RYGB cohort. Microbial analysis demonstrated decreased alpha diversity in early RYGB cohorts which normalized in the late group. The early RYGB cohorts had higher abundances of Escherichia-Shigella but lower abundances of Lactobacillus, Adlercreutzia, and Proteus while the late cohorts demonstrated higher abundances of Escherichia-Shigella and lower abundances of Lactobacillus. Shifts in Lactobacillus and Escherichia-Shigella correlated with decreases in multiple conjugated bile acids. In conclusion, RYGB caused a late and substantial increase in L-cell quantity with associated changes in bile acids which correlated to shifts in Escherichia-Shigella and Lactobacillus. This proliferation of L-cells contributed to improved glucose homeostasis.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Figure 1
Figure 1
Pre- and post-operative absolute weight on high fat diet; RYGB, Roux-en-Y gastric bypass.
Figure 2
Figure 2
Ileal morphological changes and L-cell density amongst groups: early Roux-en-Y gastric bypass (RYGB) (n = 7), early sham (n = 8), late RYGB (n = 8), late sham (n = 9). Error bars on figures represent standard error of the means and asterisks represent statistical significance with * as p < 0.05, ** as p < 0.01, *** as p < 0.001, **** as p < 0.0001.
Figure 3
Figure 3
Intraperitoneal glucose tolerance testing in gastric bypass (n = 8) versus sham (n = 9) in the late cohorts; RYGB, Roux-en-Y gastric bypass.
Figure 4
Figure 4
Differences in microbial abundance between Roux-en-Y gastric bypass and sham at early and late timepoints. (A) Taxonomic differences in microbial relative abundance between groups. (B) Between group differences in α diversity using the Chao1 and Shannon indices. (C) Between-group differences in β diversity using the Bray–Curtis dissimilarity index.
Figure 5
Figure 5
Heatmap of ileal bile acid concentrations after logarithmic transformation of data.
Figure 6
Figure 6
Heatmap of Spearman correlations of differential microbes and bile acids between late RYGB compared to late sham.

References

    1. Thaler JP, Cummings DE. Minireview: Hormonal and metabolic mechanisms of diabetes remission after gastrointestinal surgery. Endocrinology. 2009;150:2518–2525.
    1. Mingrone G, Castagneto-Gissey L. Mechanisms of early improvement/resolution of type 2 diabetes after bariatric surgery. Diabetes Metab. 2009;35:518–523.
    1. Arora T, et al. Microbial regulation of the L cell transcriptome. Sci. Rep. 2018;8:1–9.
    1. Sweeney TE, Morton JM. Metabolic surgery: Action via hormonal milieu changes, changes in bile acids or gut microbiota? A summary of the literature. Best Pract. Res. Clin. Gastroenterol. 2014;28:727–740.
    1. Thomas C, et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009;10:167–177.
    1. Schaap FG, Trauner M, Jansen PLM. Bile acid receptors as targets for drug development. Nat. Rev. Gastroenterol. Hepatol. 2013;11:55–67.
    1. Graessler J, et al. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: Correlation with inflammatory and metabolic parameters. Pharmacogenomics J. 2013;13:514–522.
    1. Li JV, et al. Experimental bariatric surgery in rats generates a cytotoxic chemical environment in the gut contents. Front. Microbiol. 2011;2:1–9.
    1. Osto M, et al. Roux-en-Y gastric bypass surgery in rats alters gut microbiota profile along the intestine. Physiol. Behav. 2013;119:92–96.
    1. Zhang H, et al. Human gut microbiota in obesity and after gastric bypass. Proc. Natl. Acad. Sci. U. S. A. 2009;106:2365–2370.
    1. Le Roux CW, et al. Gut hypertrophy after gastric bypass is associated with increased glucagon-like peptide 2 and intestinal crypt cell proliferation. Ann. Surg. 2010;252:50–56.
    1. Dudrick SJ, Daly JM, Castro G, Akhtar M. Gastrointestinal adaptation following small bowel bypass for obesity. Ann. Surg. 1977;185:642.
    1. Hansen CF, et al. The effect of ileal interposition surgery on enteroendocrine cell numbers in the UC Davis type 2 diabetes mellitus rat. Regul. Pept. 2014;189:31–39.
    1. Mumphrey MB, Patterson LM, Zheng H. Roux-en-Y gastric bypass surgery increases number but not density of CCK-, GLP-1-, 5-HT-, and neurotensin-expressing enteroendocrine cells in rats. Neurogastroenterol. Motil. 2013;25:70–79. doi: 10.1111/nmo.12034.
    1. Rhee NA, et al. Effect of Roux-en-Y gastric bypass on the distribution and hormone expression of small-intestinal enteroendocrine cells in obese patients with type 2 diabetes. Diabetologia. 2015;58:2254–2258.
    1. Larraufie P, et al. Important role of the GLP-1 axis for glucose homeostasis after bariatric surgery. Cell Rep. 2019;26:1399–1408.e6.
    1. Awad WA, Ghareeb K, Böhm J. Effect of addition of a probiotic micro-organism to broiler diet on intestinal mucosal architecture and electrophysiological parameters. J. Anim. Physiol. Anim. Nutr. (Berl) 2010;94:486–494.
    1. Simonen M, et al. Conjugated bile acids associate with altered rates of glucose and lipid oxidation after Roux-en-Y gastric bypass. Obes. Surg. 2012;22:1473–1480.
    1. Patti ME, et al. Serum bile acids are higher in humans with prior gastric bypass: Potential contribution to improved glucose and lipid metabolism. Obesity. 2009;17:1671–1677.
    1. Kohli R, et al. Weight loss induced by Roux-en-Y gastric bypass but not laparoscopic adjustable gastric banding increases circulating bile acids. J. Clin. Endocrinol. Metab. 2013;98:6–10.
    1. Bhutta HY, et al. Effect of Roux-en-Y gastric bypass surgery on bile acid metabolism in normal and obese diabetic rats. PLoS ONE. 2015;10:1–17.
    1. Kohli R, et al. Intestinal adaptation after ileal interposition surgery increases bile acid recycling and protects against obesity-related comorbidities. Am. J. Physiol. Gastrointest. Liver Physiol. 2010;299:652–660.
    1. Smith CD, et al. Gastric acid secretion and vitamin B12 absorption after vertical Roux-en-Y gastric bypass for morbid obesity. Ann. Surg. 1993;218:91–96.
    1. Engevik MA, Hickerson A, Shull GE, Worrell RT. Acidic conditions in the NHE2 −/− mouse intestine result in an altered mucosa-associated bacterial population with changes in mucus oligosaccharides. Cell. Physiol. Biochem. 2013;32:111–128.
    1. Furet J-P, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss links with metabolic and low-grade inflammation markers. Diabetes. 2010;59:3049–3057.
    1. Kong LC, et al. Gut microbiota after gastric bypass in human obesity: Increased richness and associations of bacterial genera with adipose tissue genes. Am. J. Clin. Nutr. 2013;98:16–24.
    1. Liong MT, Shah NP. Bile salt deconjugation ability, bile salt hydrolase activity and cholesterol co-precipitation ability of lactobacilli strains. Int. Dairy J. 2005;15:391–398.
    1. Urdaneta V, Casadesús J. Interactions between bacteria and bile salts in the gastrointestinal and hepatobiliary tracts. Front. Med. 2017;4:1–13.
    1. Yadav H, Jain S, Sinha PR. Oral administration of dahi containing probiotic Lactobacillus acidophilus and Lactobacillus casei delayed the progression of streptozotocin-induced diabetes in rats. J. Dairy Res. 2008;75:189–195.
    1. Wickens KL, et al. Early pregnancy probiotic supplementation with Lactobacillus rhamnosus HN001 may reduce the prevalence of gestational diabetes mellitus: A randomised controlled trial. Br. J. Nutr. 2017;117:804–813.
    1. Foley MH, et al. Lactobacillus bile salt hydrolase substrate specificity governs bacterial fitness and host colonization. Proc. Natl. Acad. Sci. U. S. A. 2021;118:e2017709118.
    1. Lund ML, et al. L-cell differentiation is induced by bile acids through GpBAR1 and paracrine GLP-1 and serotonin signaling. Diabetes. 2020;69:614–623.
    1. Trabelsi M-S, et al. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat. Commun. 2015;6:7629.
    1. Potthoff MJ, et al. Colesevelam suppresses hepatic glycogenolysis by TGR5-mediated induction of GLP-1 action in DIO mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2013;304:371–380.
    1. Harach T, et al. TGR5 potentiates GLP-1 secretion in response to anionic exchange resins. Sci. Rep. 2012;2:2–8.
    1. Kaur A, et al. Loss of Cyp8b1 improves glucose homeostasis by increasing GLP-1. Diabetes. 2015;64:1168–1179.
    1. Li F, et al. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat. Commun. 2013;4:1–10.
    1. Yoon HS, et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 2021;6:563–573.
    1. Dao MC, et al. Akkermansia muciniphila abundance is lower in severe obesity, but its increased level after bariatric surgery is not associated with metabolic health improvement. Am. J. Physiol. Endocrinol. Metab. 2019;317:E446–E459.
    1. Yan M, Song M-M, Bai R-X, Cheng S, Yan W-M. Effect of Roux-en-Y gastric bypass surgery on intestinal Akkermansia muciniphila. World J. Gastrointest. Surg. 2016;8:301.
    1. Zhou K. Strategies to promote abundance of Akkermansia muciniphila, an emerging probiotics in the gut, evidence from dietary intervention studies. J. Funct. Foods. 2017;33:194–201.
    1. Roopchand DE, et al. Dietary polyphenols promote growth of the gut bacterium Akkermansia muciniphila and attenuate high-fat diet-induced metabolic syndrome. Diabetes. 2015;64:2847–2858.
    1. Chambers AP, et al. Weight-independent changes in blood glucose homeostasis after gastric bypass or vertical sleeve gastrectomy in rats. Gastroenterology. 2011;141:950–958.
    1. Dang JT, et al. A protocol for Roux-en-Y gastric bypass in rats using linear staplers. J. Vis. Exp. 2021;174:1–12. doi: 10.3791/62575.
    1. NYU Langone Medical Center. Tissue preparation and cryopreservation with sucrose for frozen tissue sections. .
    1. Abcam . Immunocytochemistry and Immunofluorescence Protocol. Abcam; 1998. pp. 1–6.
    1. Callahan BJ, et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods. 2016;13:581–583.
    1. Callahan BJ, McMurdie PJ, Holmes SP. Exact sequence variants should replace operational taxonomic units in marker-gene data analysis. ISME J. 2017;11:2639–2643.
    1. McMurdie PJ, Holmes S. Phyloseq: An R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE. 2013;8:e61217.
    1. Ni Y, et al. M2IA: A web server for microbiome and metabolome integrative analysis. Bioinformatics. 2020;36:3493–3498.

Source: PubMed

3
Prenumerera