Ileo-colonic delivery of conjugated bile acids improves glucose homeostasis via colonic GLP-1-producing enteroendocrine cells in human obesity and diabetes

Gerardo Calderon, Alison McRae, Juraj Rievaj, Judith Davis, Inuk Zandvakili, Sara Linker-Nord, Duane Burton, Geoffrey Roberts, Frank Reimann, Bronislava Gedulin, Adrian Vella, Nicholas F LaRusso, Michael Camilleri, Fiona M Gribble, Andres Acosta, Gerardo Calderon, Alison McRae, Juraj Rievaj, Judith Davis, Inuk Zandvakili, Sara Linker-Nord, Duane Burton, Geoffrey Roberts, Frank Reimann, Bronislava Gedulin, Adrian Vella, Nicholas F LaRusso, Michael Camilleri, Fiona M Gribble, Andres Acosta

Abstract

Background: The bile acid (BA) pathway plays a role in regulation of food intake and glucose metabolism, based mainly on findings in animal models. Our aim was to determine whether the BA pathway is altered and correctable in human obesity and diabetes.

Methods: We conducted 3 investigations: 1) BA receptor pathways were studied in NCI-H716 enteroendocrine cell (EEC) line, whole human colonic mucosal tissue and in human colonic EEC isolated by Fluorescence-activated Cell Sorting (ex vivo) from endoscopically-obtained biopsies colon mucosa; 2) We characterized the BA pathway in 307 participants by measuring during fasting and postprandial levels of FGF19, 7αC4 and serum BA; 3) In a placebo-controlled, double-blind, randomised, 28-day trial, we studied the effect of ileo-colonic delivery of conjugated BAs (IC-CBAS) on glucose metabolism, incretins, and lipids, in participants with obesity and diabetes.

Findings: Human colonic GLP-1-producing EECs express TGR5, and upon treatment with bile acids in vitro, human EEC differentially expressed GLP-1 at the protein and mRNA level. In Ussing Chamber, GLP-1 release was stimulated by Taurocholic acid in either the apical or basolateral compartment. FGF19 was decreased in obesity and diabetes compared to controls. When compared to placebo, IC-CBAS significantly decreased postprandial glucose, fructosamine, fasting insulin, fasting LDL, and postprandial FGF19 and increased postprandial GLP-1 and C-peptide. Increase in faecal BA was associated with weight loss and with decreased fructosamine.

Interpretations: In humans, BA signalling machinery is expressed in colonic EECs, deficient in obesity and diabetes, and when stimulated with IC-CBAS, improved glucose homeostasis. ClinicalTrials.gov number, NCT02871882, NCT02033876.

Funding: Research support and drug was provided by Satiogen Pharmaceuticals (San Diego, CA). AA, MC, and NFL report grants (AA- C-Sig P30DK84567, K23 DK114460; MC- NIH R01 DK67071; NFL- R01 DK057993) from the NIH. JR was supported by an Early Career Grant from Society for Endocrinology.

Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Co-Expression of GLP-1 with BA pathway signalling molecules in human colon and in vitro NCI-H716 L-cell model. A) Double immunofluorescence staining of whole human colonic tissue zoomed to GLP-1 (green) positive EECs co-expressing TGR5 (pink), with channel and merged displays of staining. Red bars for all images indicate 10 µm. Differentiated NCI-H716 cells, incubated with 1 mM CBAS in control media or control media alone for 2 h increments over the course of 6 h. Media was replaced in-between time point. Resulting culture supernatants were assayed for protein levels of b) GLP-1 c) FGF19. Differentiated H716 cell mRNA expression of d) GCG and e) FGF19 after 24 h treatments with 1 mM CBAS, 1 mM CDCA, compared to control media expression. Data are presented as mean±SEM. Significance testing used the student t-test for protein secretion studies, and ANOVA for mRNA expression; *p<0·05, **p<0·01, ***p<0·001.
Fig. 2
Fig. 2
Co-Expression of TGR5 in FACS-isolated human colonic EEC a) diagram and b) corresponding FACS-plots demonstrating a sequential flow-cytometric gating strategy to isolate GLP-1+ EEC from single cells suspensions of human colonic biopsies. c) Triple immunofluorescence staining of (from top) GLP-1+ FACS-isolated human enteroendocrine cells, GLP-1- FACS-isolated human colonic cells, and no primary antibody controls. Both sorted GLP-1 positive and negative cells contained populations of TGR5 (pink) compared to negative control. D) Quantification of TGR-5 positive cells in GLP-1+ and GLP-1- FACS-isolated human colonic cells. Red bar indicates 10 µm. Data are presented as mean±SEM. Significance testing used the student t-test; *p<0·05, **p<0·01, ***p<0·001.
Fig. 3
Fig. 3
Basolateral application of taurodeoxycholic acid triggers stronger GLP-1 response compared to apical application in human and mouse colon. GLP-1 release was measured in the basolateral compartment following addition of taurodeoxycholic Acid (TDCA, 100 μM) to the apical (AP) or basolateral (BAS) compartments of colonic tissue sections of a) human and b) mouse mounted in Ussing chambers. Water was added for control experiments (CTR). Human: Adj p<0·05 AP vs CTR, p<0·0001 BAS vs CTR, p<0·001 AP vs BAS; Mouse: p<0·01 BAS vs CTR, p<0·01 AP vs BAS. c) Double immunofluorescence staining of whole human colonic tissue with GLP-1 (green) positive EEC and colonocytes producing ASBT (pink) with individual channel and merged displays of staining. Red bars for all images indicate 20 µm.
Fig. 4
Fig. 4
Fasting serum Fibroblast Growth Factor (FGF) 19, 7aC4 and bile acids levels in patients with healthy weight, overweight and obesity based on BMI and type 2 diabetes. a) Fasting FGF19, c) serum fasting bile acids, and e) fasting 7aC4 in participants with normal weight, overweigh and obesity; b) Fasting FGF19, d) serum fasting bile acids, and f) fasting 7aC4 in participants with obesity with or without diabetes, Data are represented as median [IQR]. ANOVA*p<0·05.
Fig. 5
Fig. 5
Effect of Ileocolonic delivery of conjugated bile acids (IC-CBAS) in glucose homeostasis and glucose regulation in patients with obesity and type 2 diabetes. (a,b) Glucose concentrations at baseline and after treatment with (a) placebo, and (b) with IC—CBAS (grey curve= at baseline; black curve= after treatment). Arrows at 0 and 240 min indicate meal ingestion. (c) delta fasting glucose, (d) delta AAB00-120 min and (e) delta AUC0-120 min of postprandial glucose, and (f) delta fasting fructosamine after treatment period. <GLP-1 concentrations at baseline and after treatment with g) placebo and h) IC—CBAS (i) delta GLP-1 AUC0-360 min, (j) delta AAB0-360 min c-peptide, (k) delta fasting insulin, and (l) delta AAB0-120 min insulin after treatment period. Data are presented as mean±SEM. Wilcoxon test treatment vs. placebo: #p<0·1. *p<0·05, **p<0·01.
Fig. 6
Fig. 6
Effect of Ileocolonic delivery of conjugated bile acids (IC-CBAS) on bile acid pathways in patients with obesity and type 2 diabetes. (a) Total faecal bile acids (circles, baseline; and squares, treatment); (b) delta total fecal bile acids and (c) delta primary faecal bile acids; (d) delta total faecal bile acids by group (circles, placebo; and squares, IC-CBAS); (e) delta conjugated faecal bile acids, (f) delta postprandial serum bile acids, (g) delta C4, (h) delta fasting FGF19, (i) delta fasting total cholesterol, (j) delta fasting cLDL, (k) correlation between weight and ∆ faecal bile acids; (l) correlation between fructosamine with ∆ faecal bile acids after 1-month treatment with IC-CBAS, and (m). correlation between postprandial GLP-1 at 45 min with ∆ faecal bile acids after 1-month treatment with IC-CBAS Data are presented as mean±SEM. Wilcoxon, *p<0·05, ** p<0·01. ANCOVA, ^p<0·05, (Figs. a–j).

References

    1. Camilleri M., Gores G.J. Therapeutic targeting of bile acids. Am J Physiol Gastrointest Liver Physiol. 2015;309(4):G209–G215.
    1. Chiang J.Y. Bile acid metabolism and signaling. Compr Physiol. 2013;3(3):1191–1212.
    1. Simmonds W.J., Hofmann A.F., Theodor E. Absorption of cholesterol from a micellar solution: intestinal perfusion studies in man. J Clin Invest. 1967;46(5):874–890.
    1. Mekhjian H.S., Phillips S.F., Hofmann A.F. Colonic absorption of unconjugated bile acids: perfusion studies in man. Dig Dis Sci. 1979;24(7):545–550.
    1. Parks D.J., Blanchard S.G., Bledsoe R.K., Chandra G., Consler T.G., Kliewer S.A. Bile acids: natural ligands for an orphan nuclear receptor. Science. 1999;284(5418):1365–1368.
    1. Symonds E.L., Peiris M., Page A.J., Chia B., Dogra H., Masding A. Mechanisms of activation of mouse and human enteroendocrine cells by nutrients. Gut. 2015;64(4):618–626.
    1. Thomas C., Gioiello A., Noriega L., Strehle A., Oury J., Rizzo G. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009;10(3):167–177.
    1. Tomlinson E., Fu L., John L., Hultgren B., Huang X., Renz M. Transgenic mice expressing human fibroblast growth factor-19 display increased metabolic rate and decreased adiposity. Endocrinology. 2002;143(5):1741–1747.
    1. Lan T., Morgan D.A., Rahmouni K., Sonoda J., Fu X., Burgess S.C. FGF19, FGF21, and an FGFR1/beta-Klotho-Activating antibody act on the nervous system to regulate body weight and glycemia. Cell Metab. 2017;26(5):709–718. e3.
    1. Watanabe M., Horai Y., Houten S.M., Morimoto K., Sugizaki T., Arita E. Lowering bile acid pool size with a synthetic farnesoid x receptor (FXR) agonist induces obesity and diabetes through reduced energy expenditure. J Biol Chem. 2011;286(30):26913–26920.
    1. Li F., Jiang C.T., Krausz K.W., Li Y.F., Albert I., Hao H.P. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun. 2013;4:2384.
    1. Fang S., Suh J.M., Reilly S.M., Yu E., Osborn O., Lackey D. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med. 2015;21(2):159–165.
    1. Ahmad N., Pfalzer A., Kaplan L. Roux-en-Y gastric bypass normalizes the blunted postprandial bile acid excursion associated with obesity. Int J Obes. 2005:1553–1559. 2013.
    1. Dutia R., Embrey M., O'Brien S., Haeusler R.A., Agenor K.K., Homel P. Temporal changes in bile acid levels and 12alpha-hydroxylation after Roux-En-Y gastric bypass surgery in type 2 diabetes. Int J Obes. 2016;40(3):554.
    1. Kohli R., Setchell K., Kirby M., Myronovych A., Ryan K., Ibrahim S. A surgical model in male obese rats uncovers protective effects of bile acids post-bariatric surgery. Endocrinology. 2013
    1. Kohli R., Kirby M., Setchell K.J.Intestinal adaptation after ileal interposition surgery increases bile acid recycling and protects against obesity-related comorbidities2010.
    1. Kohli R., Bradley D., Setchell K., Eagon J., Abumrad N., Klein S. 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(4):12.
    1. Pournaras D.J., Glicksman C., Vincent R.P., Kuganolipava S., Alaghband-Zadeh J., Mahon D. The role of bile after roux-en-y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology. 2012;153(8):3613–3619.
    1. Wu T., Bound M., Standfield S., Jones K., Horowitz M., Rayner C. Effects of taurocholic acid on glycemic, glucagon-like peptide-1, and insulin responses to small intestinal glucose infusion in healthy humans. J Clin Endocrinol Metab. 2013;98(4):22.
    1. Kars M., Yang L., Gregor M., Mohammed B., Pietka T., Finck B. Tauroursodeoxycholic acid may improve liver and muscle but not adipose tissue insulin sensitivity in obese men and women. Diabetes. 2010;59(8):1899–1905.
    1. Ryan K.K., Tremaroli V., Clemmensen C., Kovatcheva-Datchary P., Myronovych A., Karns R. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509(7499):183–188.
    1. Bozadjieva N., Heppner K.M., Seeley R.J. Targeting fxr and FGF19 to treat metabolic diseases-lessons learned from bariatric surgery. Diabetes. 2018;67(9):1720–1728.
    1. Zhang J.H., Nolan J.D., Kennie S.L., Johnston I.M., Dew T., Dixon P.H. Potent stimulation of fibroblast growth factor 19 expression in the human ileum by bile acids. Am J Physiol Gastrointest Liver Physiol. 2013;304(10):G940–G948.
    1. de Bruine A.P., Dinjens W.N., van der Linden E.P., Pijls M.M., Moerkerk P.T., Bosman F.T. Extracellular matrix components induce endocrine differentiation in vitro in NCI-H716 cells. Am J Pathol. 1993;142(3):773–782.
    1. Habib A.M., Richards P., Cairns L.S., Rogers G.J., Bannon C.A., Parker H.E. Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry. Endocrinology. 2012;153(7):3054–3065.
    1. Brighton C.A., Rievaj J., Kuhre R.E., Glass L.L., Schoonjans K., Holst J.J. Bile acids trigger GLP-1 release predominantly by accessing basolaterally located g protein-coupled bile acid receptors. Endocrinology. 2015;156(11):3961–3970.
    1. Acosta A., Camilleri M., Shin A., Vazquez-Roque M.I., Iturrino J., Burton D. Quantitative gastrointestinal and psychological traits associated with obesity and response to weight-loss therapy. Gastroenterology. 2015;148(3):537–5460000.
    1. Vijayvargiya P., Busciglio I., Burton D., Donato L., Lueke A., Camilleri M. Bile acid deficiency in a subgroup of patients with irritable bowel syndrome with constipation based on biomarkers in serum and fecal samples. Clin Gastroenterol Hepatol. 2018;16(4):522–527.
    1. Donato L.J., Lueke A., Kenyon S.M., Meeusen J.W., Camilleri M. Description of analytical method and clinical utility of measuring serum 7-alpha-hydroxy-4-cholesten-3-one (7aC4) by mass spectrometry. Clin Biochem. 2018;52:106–111.
    1. Smushkin G., Sathananthan M., Piccinini F., Dalla Man C., Law J., Cobelli C. The effect of a bile acid sequestrant on glucose metabolism in subjects with type 2 diabetes. Diabetes. 2013;62(4):1094–1101.
    1. Adrian T.E., Gariballa S., Parekh K.A., Thomas S.A., Saadi H., Al Kaabi J. Rectal taurocholate increases L cell and insulin secretion, and decreases blood glucose and food intake in obese type 2 diabetic volunteers. Diabetologia. 2012;55(9):2343–2347.
    1. Trabelsi M.-S.S., Daoudi M., Prawitt J., Ducastel S., Touche V., Sayin S.I. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine l cells. Nat Commun. 2015;6:7629.
    1. Christiansen C.B., Trammell S.A.J., Wewer Albrechtsen N.J., Schoonjans K., Albrechtsen R., Gillum M.P. Bile acids drive colonic secretion of glucagon-like-peptide 1 and peptide-YY in rodents. Am J Physiol Gastrointest Liver Physiol. 2019;316(5):G574–GG84.
    1. Habib A.M., Richards P., Rogers G.J., Reimann F., Gribble F.M. Co-localisation and secretion of glucagon-like peptide 1 and peptide YY from primary cultured human L cells. Diabetologia. 2013;56(6):1413–1416.
    1. Haeusler R.A., Astiarraga B., Camastra S., Accili D., Ferrannini E. Human insulin resistance is associated with increased plasma levels of 12alpha-hydroxylated bile acids. Diabetes. 2013;62(12):4184–4191.
    1. Sachdev S., Wang Q., Billington C., Connett J., Ahmed L., Inabnet W. FGF 19 and bile acids increase following roux-en-y gastric bypass but not after medical management in patients with type 2 diabetes. Obes Surg. 2016;26(5):957–965.
    1. Proano M., Camilleri M., Phillips S.F., Brown M.L., Thomforde G.M. Transit of solids through the human colon: regional quantification in the unprepared bowel. Am J Physiol. 1990;258(6 Pt 1):G856–G862.
    1. Cremonini F., Mullan B., Camilleri M., Burton D., Rank M. Performance characteristics of scintigraphic transit measurements for studies of experimental therapies. Aliment Pharmacol Ther. 2002;16(10):1781–1790.
    1. Odunsi-Shiyanbade S.T., Camilleri M., McKinzie S., Burton D., Carlson P., Busciglio I.A. Effects of chenodeoxycholate and a bile acid sequestrant, colesevelam, on intestinal transit and bowel function. Clin Gastroenterol Hepatol. 2010;8(2):159–165.
    1. Malmstrom H., Walldius G., Grill V., Jungner I., Gudbjornsdottir S., Hammar N. Fructosamine is a useful indicator of hyperglycaemia and glucose control in clinical and epidemiological studies–cross-sectional and longitudinal experience from the Amoris cohort. PLoS ONE. 2014;9(10)
    1. Ma K., Saha P.K., Chan L., Moore D.D. Farnesoid x receptor is essential for normal glucose homeostasis. J Clin Invest. 2006;116(4):1102–1109.
    1. Bonde Y., Eggertsen G., Rudling M. Mice abundant in muricholic bile acids show resistance to dietary induced steatosis, weight gain, and to impaired glucose metabolism. PLoS ONE. 2016;11(1)
    1. Rudling M. Understanding mouse bile acid formation: is it time to unwind why mice and rats make unique bile acids? J Lipid Res. 2016;57(12):2097–2098.
    1. de Bruine A.P., Dinjens W.N., Pijls M.M., vd Linden E.P., Rousch M.J., Moerkerk P.T. NCI-H716 cells as a model for endocrine differentiation in colorectal cancer. Virchows Arch B Cell Pathol Incl Mol Pathol. 1992;62(5):311–320.
    1. Kuhre R.E., Wewer Albrechtsen N.J., Deacon C.F., Balk-Moller E., Rehfeld J.F., Reimann F. Peptide production and secretion in GLUTag, NCI-H716, and STC-1 cells: a comparison to native L-cells. J Mol Endocrinol. 2016;56(3):201–211.
    1. Valentin N., Camilleri M., Altayar O., Vijayvargiya P., Acosta A., Nelson A.D. Biomarkers for bile acid diarrhoea in functional bowel disorder with diarrhoea: a systematic review and meta-analysis. Gut. 2016;65(12):1951–1959.
    1. Keely S.J., Walters J.R. The farnesoid X receptor: good for bad. Cell Mol Gastroenterol Hepatol. 2016;2(6):725–732.
    1. Kliewer S.A., Mangelsdorf D.J. Bile acids as hormones: the FXR-FGF15/19 pathway. Dig Dis. 2015;33(3):327–331.
    1. Wojcik M., Janus D., Dolezal-Oltarzewska K., Kalicka-Kasperczyk A., Poplawska K., Drozdz D. A decrease in fasting FGF19 levels is associated with the development of non-alcoholic fatty liver disease in obese adolescents. J Pediatr Endocrinol Metab. 2012;25(11–12):1089–1093.
    1. Gomez-Ambrosi J., Gallego-Escuredo J.M., Catalan V., Rodriguez A., Domingo P., Moncada R. FGF19 and FGF21 serum concentrations in human obesity and type 2 diabetes behave differently after diet- or surgically-induced weight loss. Clin Nutr. 2017;36(3):861–868.
    1. Gallego-Escuredo J.M., Gomez-Ambrosi J., Catalan V., Domingo P., Giralt M., Fruhbeck G. Opposite alterations in FGF21 and FGF19 levels and disturbed expression of the receptor machinery for endocrine FGFs in obese patients. Int J Obes (Lond) 2015;39(1):121–129.
    1. Mannucci E., Ognibene A., Cremasco F., Bardini G., Mencucci A., Pierazzuoli E. Glucagon-like peptide (GLP)-1 and leptin concentrations in obese patients with type 2 diabetes mellitus. Diabet Med. 2000;17(10):713–719.
    1. Vilsboll T., Krarup T., Deacon C.F., Madsbad S., Holst J.J. Reduced postprandial concentrations of intact biologically active glucagon-like peptide 1 in type 2 diabetic patients. Diabetes. 2001;50(3):609–613.
    1. Batterham R.L., Cohen M.A., Ellis S.M., Le Roux C.W., Withers D.J., Frost G.S. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med. 2003;349(10):941–948.
    1. Gribble F.M., Meek C.L., Reimann F. Targeted intestinal delivery of incretin secretagogues-towards new diabetes and obesity therapies. Peptides. 2018;100:68–74.
    1. Camilleri M., Iturrino J., Bharucha A., Burton D., Shin A., Jeong I.D. Performance characteristics of scintigraphic measurement of gastric emptying of solids in healthy participants. Neurogastroenterol Motil. 2012;24(12):1076. the official journal of the European Gastrointestinal Motility Society.
    1. Pedersen O., Nielsen O., Bak J., Richelsen B., Beck-Nielsen H., Sorensen N. The effects of metformin on adipocyte insulin action and metabolic control in obese subjects with type 2 diabetes. Diabet Med. 1989;6(3):249–256.
    1. Evans J.L., Heymann C.J., Goldfine I.D., Gavin L.A. Pharmacokinetics, tolerability, and fructosamine-lowering effect of a novel, controlled-release formulation of alpha-lipoic acid. Endocr Pract. 2002;8(1):29–35.
    1. Cefalu W.T., Schneider D.J., Carlson H.E., Migdal P., Gan Lim L., Izon M.P. Effect of combination glipizide GITS/metformin on fibrinolytic and metabolic parameters in poorly controlled type 2 diabetic subjects. Diabetes Care. 2002;25(12):2123–2128.
    1. Bronden A., Alber A., Rohde U., Gasbjerg L.S., Rehfeld J.F., Holst J.J. The bile acid-sequestering resin sevelamer eliminates the acute GLP-1 stimulatory effect of endogenously released bile acids in patients with type 2 diabetes. Diabetes Obesity Metabolism. 2018;20(2):362–369.
    1. Potthoff M.J., Potts A., He T., Duarte J.A., Taussig R., Mangelsdorf D.J. Colesevelam suppresses hepatic glycogenolysis by TGR5-mediated induction of GLP-1 action in DIO mice. Am J Physiol Gastrointest Liver Physiol. 2013;304(4):G371–G380.
    1. Shang Q., Saumoy M., Holst J.J., Salen G., Xu G. Colesevelam improves insulin resistance in a diet-induced obesity (F-DIO) rat model by increasing the release of GLP-1. Am J Physiol Gastrointest Liver Physiol. 2010;298(3):G419–G424.
    1. Sun L., Xie C., Wang G., Wu Y., Wu Q., Wang X. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat Med. 2018;24(12):1919–1929.

Source: PubMed

3
구독하다