Berberine protects against diet-induced obesity through regulating metabolic endotoxemia and gut hormone levels

Jian Hui Xu, Xing Zhen Liu, Wei Pan, Da Jin Zou, Jian Hui Xu, Xing Zhen Liu, Wei Pan, Da Jin Zou

Abstract

Systemic inflammation, which can be induced by metabolic endotoxemia, and corresponding high‑fat diet‑mediated metabolic disorders are associated with gut microbiota. In the present study reverse transcription-polymerase chain reaction, immunofluorescence, pyrosequencing, ELISA and Oil Red O staining were performed to assess whether berberine can protect against diet-induced obesity, through modulating the gut microbiota and consequently improving metabolic endotoxemia and gastrointestinal hormone levels. Alterations in the gut microbiota induced by berberine resulted in a significant reduction in bacterial lipopolysaccharide levels in portal plasma. Levels of inflammatory and oxidative stress markers, as well as the mRNA expression levels of macrophage infiltration markers in visceral adipose tissue, were also reduced by berberine. Inhibition of the inflammatory response was associated with a reduction in intestinal permeability and an increase in the expression of tight junction proteins. In addition, berberine was reported to restore aberrant levels of gut hormones in the portal plasma, such as glucagon‑like peptide‑1 and ‑2, peptide YY, glucose‑dependent insulinotropic polypeptide and pancreatic polypeptide. The present findings indicated that berberine, through modulating gut microbiota, restored the gut barrier, reduced metabolic endotoxemia and systemic inflammation, and improved gut peptide levels in high‑fat diet‑fed rats. The present study suggests that berberine may be an effective therapeutic strategy for the treatment of obesity and insulin resistance.

Figures

Figure 1.
Figure 1.
Berberine reduced weight gain and improved glucose homeostasis in HFD-fed rats. (A) Body weight. (B) Daily food intake per rat. (C) Fasting plasma glucose concentration. (D) Fasting plasma insulin concentration. (E) HOMA-IR index, calculated using the following equation: FBG (mmol/l) × FINS (mU/l)/22.5. (F) Oral glucose tolerance test. Data are expressed as the mean ± standard deviation. *P#P<0.05, ###P<0.001 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; HOMA-IR, homeostasis model assessment of insulin resistance; FBG, fasting blood glucose; FINS, fasting blood insulin; OGTT, oral glucose tolerance test.
Figure 2.
Figure 2.
Berberine attenuated HFD-induced hepatic injury and hepatic steatosis. (A) Hepatic triglyceride content. (B) Hepatic total cholesterol content. (C) ALT plasma concentration. (D) AST plasma concentration. (E) Fresh-frozen liver sections were stained with Oil Red O (magnification, ×100). Data are expressed as the mean ± standard deviation. *P#P<0.05, ##P<0.01 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; ALT, alanine aminotransferase; AST, aspartate aminotransferase.
Figure 3.
Figure 3.
Berberine reduced endotoxemia and visceral adipose tissue inflammation in HFD-fed rats. (A) Portal plasma LPS concentrations. (B) TNF-α mRNA expression levels. (C) IL-1β mRNA expression levels. (D) PAI-1 mRNA expression levels. (E) NADPHox mRNA expression levels. (F) STAMP-2 mRNA expression levels. (G) MCP-1 mRNA expression levels. (H) F4/80 mRNA expression levels. Data are expressed as the mean ± standard deviation. **P##P<0.01, ###P<0.001 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; LPS, lipopolysaccharide; EU, endotoxin unit; TNF, tumor necrosis factor; IL, interleukin; PAI-1, plasminogen activator inhibitor-1; NADPHox, nicotinamide-adenine dinucleotide phosphate oxidase; STAMP-2, six transmembrane protein of prostate-2; MCP-1, monocyte chemotactic protein-1; F4/80, EGF-like module-containing mucin-like hormone receptor-like 1.
Figure 4.
Figure 4.
Metabolic endotoxemia was positively correlated with inflammation, oxidative stress and macrophage infiltration markers. Correlation between: Portal plasma LPS level and (A) TNF-α, (B) IL-1β, (C) PAI-1, (D) NADPHox, (E) STAMP-2, (F) MCP-1 and (G) F4/80 mRNA levels in visceral adipose tissue of Ctl, HF and HB rats. The inset corresponds to Pearson's correlation and corresponding P-value. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; LPS, lipopolysaccharide; TNF, tumor necrosis factor; IL, interleukin; PAI-1, plasminogen activator inhibitor-1; NADPHox, nicotinamide-adenine dinucleotide phosphate oxidase; STAMP-2, six transmembrane protein of prostate-2; MCP-1, monocyte chemotactic protein-1; F4/80, EGF-like module-containing mucin-like hormone receptor-like 1; EU, endotoxin unit.
Figure 5.
Figure 5.
Berberine reduced intestinal permeability in HFD-fed rats. (A) Plasma FITC-dextran concentration. (B) Total plasma FITC-dextran concentration. (C) Correlation between portal plasma LPS levels and plasma FITC-dextran concentration. The inset corresponds to Pearson's correlation and the corresponding P-value. Data are expressed as the mean ± standard deviation. **P##P<0.01 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; FITC, fluorescein isothiocyanate; LPS, lipopolysaccharide; EU, endotoxin unit.
Figure 6.
Figure 6.
Berberine restored the expression of tight junction proteins in HFD-fed rats. mRNA expression levels of colonic epithelial tight junction proteins (A) claudin-1, (B) claudin-2, (C) occludin and (D) ZO-1. Data are expressed as the mean ± standard deviation. *P#P<0.05, ###P<0.001 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; ZO-1, zonula occludens-1.
Figure 7.
Figure 7.
Berberine restored the expression and distribution of claudin-1 and claudin-2 in HFD-fed rats. (A) Immunofluorescent staining for claudin-1 (magnification, ×100). (B) Immunohistochemistry score for claudin-1. (C) Immunofluorescent staining for claudin-2 (magnification, ×100). (D) Immunohistochemistry score for claudin-2. Data are expressed as the mean ± standard deviation. *P##P<0.01 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet.
Figure 8.
Figure 8.
Berberine modulated intestinal hormone levels in portal plasma of HFD-fed rats. (A) Portal plasma GLP-1 levels. (B) Portal plasma GLP-2 levels. (C) Portal plasma GIP, PP and PYY levels. (D) Colonic proglucagon mRNA expression levels. (E) Colonic L-cell numbers. Data are expressed as the mean ± standard deviation. *P#P<0.05, ###P<0.001 compared with the HF group. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; GLP, glucagon-like peptide; GIP, gastric inhibitory polypeptide; PP, pancreatic polypeptide; PYY, peptide YY.
Figure 9.
Figure 9.
Berberine altered the gut microbiota composition in HFD-fed rats. (A) Rarefaction curves. (B) Weighted UniFrac PCoA. Each point represents the microbiota of each rat in the Ctl, HF and HB groups. (C) PCA. P-values were calculated by MCPP. (D) RDA between the Ctl and HF groups. Berberine altered the gut microbiota composition in HFD-fed rats. (E) RDA between the Ctl and HB groups. (F) RDA between the HB and HF groups. P-values were calculated by MCPP. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; PCoA, principal coordinates analysis; PCA, principal components analysis; RDA, redundancy analysis; MCPP, Monte Carlo permutation tests.
Figure 9.
Figure 9.
Berberine altered the gut microbiota composition in HFD-fed rats. (A) Rarefaction curves. (B) Weighted UniFrac PCoA. Each point represents the microbiota of each rat in the Ctl, HF and HB groups. (C) PCA. P-values were calculated by MCPP. (D) RDA between the Ctl and HF groups. Berberine altered the gut microbiota composition in HFD-fed rats. (E) RDA between the Ctl and HB groups. (F) RDA between the HB and HF groups. P-values were calculated by MCPP. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; PCoA, principal coordinates analysis; PCA, principal components analysis; RDA, redundancy analysis; MCPP, Monte Carlo permutation tests.
Figure 9.
Figure 9.
Berberine altered the gut microbiota composition in HFD-fed rats. (A) Rarefaction curves. (B) Weighted UniFrac PCoA. Each point represents the microbiota of each rat in the Ctl, HF and HB groups. (C) PCA. P-values were calculated by MCPP. (D) RDA between the Ctl and HF groups. Berberine altered the gut microbiota composition in HFD-fed rats. (E) RDA between the Ctl and HB groups. (F) RDA between the HB and HF groups. P-values were calculated by MCPP. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; PCoA, principal coordinates analysis; PCA, principal components analysis; RDA, redundancy analysis; MCPP, Monte Carlo permutation tests.
Figure 9.
Figure 9.
Berberine altered the gut microbiota composition in HFD-fed rats. (A) Rarefaction curves. (B) Weighted UniFrac PCoA. Each point represents the microbiota of each rat in the Ctl, HF and HB groups. (C) PCA. P-values were calculated by MCPP. (D) RDA between the Ctl and HF groups. Berberine altered the gut microbiota composition in HFD-fed rats. (E) RDA between the Ctl and HB groups. (F) RDA between the HB and HF groups. P-values were calculated by MCPP. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; PCoA, principal coordinates analysis; PCA, principal components analysis; RDA, redundancy analysis; MCPP, Monte Carlo permutation tests.
Figure 9.
Figure 9.
Berberine altered the gut microbiota composition in HFD-fed rats. (A) Rarefaction curves. (B) Weighted UniFrac PCoA. Each point represents the microbiota of each rat in the Ctl, HF and HB groups. (C) PCA. P-values were calculated by MCPP. (D) RDA between the Ctl and HF groups. Berberine altered the gut microbiota composition in HFD-fed rats. (E) RDA between the Ctl and HB groups. (F) RDA between the HB and HF groups. P-values were calculated by MCPP. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; PCoA, principal coordinates analysis; PCA, principal components analysis; RDA, redundancy analysis; MCPP, Monte Carlo permutation tests.
Figure 9.
Figure 9.
Berberine altered the gut microbiota composition in HFD-fed rats. (A) Rarefaction curves. (B) Weighted UniFrac PCoA. Each point represents the microbiota of each rat in the Ctl, HF and HB groups. (C) PCA. P-values were calculated by MCPP. (D) RDA between the Ctl and HF groups. Berberine altered the gut microbiota composition in HFD-fed rats. (E) RDA between the Ctl and HB groups. (F) RDA between the HB and HF groups. P-values were calculated by MCPP. Ctl, normal diet; HF, HFD; HB, HFD supplemented with berberine for 6 weeks; HFD, high-fat diet; PCoA, principal coordinates analysis; PCA, principal components analysis; RDA, redundancy analysis; MCPP, Monte Carlo permutation tests.

References

    1. Zhang Y, Li X, Zou D, Liu W, Yang J, Zhu N, Huo L, Wang M, Hong J, Wu P, et al. Treatment of type 2 diabetes and dyslipidemia with the natural plant alkaloid berberine. J Clin Endocrinol Metab. 2008;93:2559–2565. doi: 10.1210/jc.2007-2404.
    1. Xie W, Gu D, Li J, Cui K, Zhang Y. Effects and action mechanisms of berberine and Rhizoma coptidis on gut microbes and obesity in high-fat diet-fed C57BL/6J mice. PLoS One. 2011;6:e24520. doi: 10.1371/journal.pone.0024520.
    1. Gomes AP, Duarte FV, Nunes P, Hubbard BP, Teodoro JS, Varela AT, Jones JG, Sinclair DA, Palmeira CM, Rolo AP. Berberine protects against high fat diet-induced dysfunction in muscle mitochondria by inducing SIRT1-dependent mitochondrial biogenesis. Biochim Biophys Acta. 2012;1822:185–195. doi: 10.1016/j.bbadis.2011.10.008.
    1. Lee YS, Kim WS, Kim KH, Yoon MJ, Cho HJ, Shen Y, Ye JM, Lee CH, Oh WK, Kim CT, et al. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Diabetes. 2006;55:2256–2264. doi: 10.2337/db06-0006.
    1. Liu L, Yu YL, Yang JS, Li Y, Liu YW, Liang Y, Liu XD, Xie L, Wang GJ. Berberine suppresses intestinal disaccharidases with beneficial metabolic effects in diabetic states, evidences from in vivo and in vitro study. Naunyn Schmiedebergs Arch Pharmacol. 2010;381:371–381. doi: 10.1007/s00210-010-0502-0.
    1. Pan GY, Huang ZJ, Wang GJ, Fawcett JP, Liu XD, Zhao XC, Sun JG, Xie YY. The antihyperglycaemic activity of berberine arises from a decrease of glucose absorption. Planta Med. 2003;69:632–636. doi: 10.1055/s-2003-41121.
    1. Kong W, Wei J, Abidi P, Lin M, Inaba S, Li C, Wang Y, Wang Z, Si S, Pan H, et al. Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat Med. 2004;10:1344–1351. doi: 10.1038/nm1135.
    1. Wang Y, Yi X, Ghanam K, Zhang S, Zhao T, Zhu X. Berberine decreases cholesterol levels in rats through multiple mechanisms, including inhibition of cholesterol absorption. Metabolism. 2014;63:1167–1177. doi: 10.1016/j.metabol.2014.05.013.
    1. Lu SS, Yu YL, Zhu HJ, Liu XD, Liu L, Liu YW, Wang P, Xie L, Wang GJ. Berberine promotes glucagon-like peptide-1 (7–36) amide secretion in streptozotocin-induced diabetic rats. J Endocrinol. 2009;200:159–165. doi: 10.1677/JOE-08-0419.
    1. Yu Y, Liu L, Wang X, Liu X, Liu X, Xie L, Wang G. Modulation of glucagon-like peptide-1 release by berberine: In vivo and in vitro studies. Biochem Pharmacol. 2010;79:1000–1006. doi: 10.1016/j.bcp.2009.11.017.
    1. Liu YT, Hao HP, Xie HG, Lai L, Wang Q, Liu CX, Wang GJ. Extensive intestinal first-pass elimination and predominant hepatic distribution of berberine explain its low plasma levels in rats. Drug Metab Dispos. 2010;38:1779–1784. doi: 10.1124/dmd.110.033936.
    1. Chen W, Miao YQ, Fan DJ, Yang SS, Lin X, Meng LK, Tang X. Bioavailability study of berberine and the enhancing effects of TPGS on intestinal absorption in rats. AAPS PharmSciTech. 2011;12:705–711. doi: 10.1208/s12249-011-9632-z.
    1. Aronsson L, Huang Y, Parini P, Korach-André M, Håkansson J, Gustafsson JÅ, Pettersson S, Arulampalam V, Rafter J. Decreased fat storage by Lactobacillus paracasei is associated with increased levels of angiopoietin-like 4 protein (ANGPTL4) PLoS One. 2010;5:e13087. doi: 10.1371/journal.pone.0013087.
    1. Zhang X, Zhao Y, Zhang M, Pang X, Xu J, Kang C, Li M, Zhang C, Zhang Z, Zhang Y, et al. Structural changes of gut microbiota during berberine-mediated prevention of obesity and insulin resistance in high-fat diet-fed rats. PLoS One. 2012;7:e42529. doi: 10.1371/journal.pone.0042529.
    1. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58:1091–1103. doi: 10.1136/gut.2008.165886.
    1. Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, De Backer F, Neyrinck AM, Delzenne NM. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr. 2009;90:1236–1243. doi: 10.3945/ajcn.2009.28095.
    1. Cani PD, Neyrinck AM, Maton N, Delzenne NM. Oligofructose promotes satiety in rats fed a high-fat diet: Involvement of glucagon-like peptide-1. Obes Res. 2005;13:1000–1007. doi: 10.1038/oby.2005.117.
    1. Cani PD, Hoste S, Guiot Y, Delzenne NM. Dietary non-digestible carbohydrates promote L-cell differentiation in the proximal colon of rats. Br J Nutr. 2007;98:32–37. doi: 10.1017/S0007114507691648.
    1. Tazoe H, Otomo Y, Karaki S, Kato I, Fukami Y, Terasaki M, Kuwahara A. Expression of short-chain fatty acid receptor GPR41 in the human colon. Biomed Res. 2009;30:149–156. doi: 10.2220/biomedres.30.149.
    1. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, Cameron J, Grosse J, Reimann F, Gribble FM. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes. 2012;61:364–371. doi: 10.2337/db11-1019.
    1. des Rieux A, Pourcelle V, Cani PD, Marchand-Brynaert J, Préat V. Targeted nanoparticles with novel non-peptidic ligands for oral delivery. Adv Drug Deliv Rev. 2013;65:833–844. doi: 10.1016/j.addr.2013.01.002.
    1. Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, Ghatei MA, Bloom SR, Frost G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes (Lond) 2015;39:424–429. doi: 10.1038/ijo.2014.153.
    1. Feng Y, Li Y, Chen C, Lin X, Yang Y, Cai H, Lv Z, Cao M, Li K, Xu J, et al. Inhibiting roles of berberine in gut movement of rodents are related to activation of the endogenous opioid system. Phytother Res. 2013;27:1564–1571.
    1. Shan CY, Yang JH, Kong Y, Wang XY, Zheng MY, Xu YG, Wang Y, Ren HZ, Chang BC, Chen LM. Alteration of the intestinal barrier and GLP2 secretion in Berberine-treated type 2 diabetic rats. J Endocrinol. 2013;218:255–262. doi: 10.1530/JOE-13-0184.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Kim KA, Gu W, Lee IA, Joh EH, Kim DH. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One. 2012;7:e47713. doi: 10.1371/journal.pone.0047713.
    1. Glass CK, Olefsky JM. Inflammation and lipid signaling in the etiology of insulin resistance. Cell Metab. 2012;15:635–645. doi: 10.1016/j.cmet.2012.04.001.
    1. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114:1752–1761. doi: 10.1172/JCI21625.
    1. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. 2006;116:1793–1801. doi: 10.1172/JCI29069C1.
    1. Shoelson SE, Herrero L, Naaz A. Obesity, inflammation and insulin resistance. Gastroenterology. 2007;132:2169–2180. doi: 10.1053/j.gastro.2007.03.059.
    1. Jeong HW, Hsu KC, Lee JW, Ham M, Huh JY, Shin HJ, Kim WS, Kim JB. Berberine suppresses proinflammatory responses through AMPK activation in macrophages. Am J Physiol Endocrinol Metab. 2009;296:E955–E964. doi: 10.1152/ajpendo.90599.2008.
    1. Everard A, Cani PD. Diabetes, obesity and gut microbiota. Best Pract Res Clin Gastroenterol. 2013;27:73–83. doi: 10.1016/j.bpg.2013.03.007.
    1. Amasheh M, Fromm A, Krug SM, Amasheh S, Andres S, Zeitz M, Fromm M, Schulzke JD. TNFalpha-induced and berberine-antagonized tight junction barrier impairment via tyrosine kinase, Akt and NFkappaB signaling. J Cell Sci. 2010;123:4145–4155. doi: 10.1242/jcs.070896.
    1. Ma JY, Feng R, Tan XS, Ma C, Shou JW, Fu J, Huang M, He CY, Chen SN, Zhao ZX, et al. Excretion of berberine and its metabolites in oral administration in rats. J Pharm Sci. 2013;102:4181–4192. doi: 10.1002/jps.23718.
    1. Yu LC, Wang JT, Wei SC, Ni YH. Host-microbial interactions and regulation of intestinal epithelial barrier function: From physiology to pathology. World J Gastrointest Pathophysiol. 2012;3:27–43. doi: 10.4291/wjgp.v3.i1.27.
    1. Greiner T, Bäckhed F. Effects of the gut microbiota on obesity and glucose homeostasis. Trends Endocrinol Metab. 2011;22:117–123. doi: 10.1016/j.tem.2011.01.002.
    1. Ichikawa H, Sakata T. Stimulation of epithelial cell proliferation of isolated distal colon of rats by continuous colonic infusion of ammonia or short-chain fatty acids is nonadditive. J Nutr. 1998;128:843–847.
    1. Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr. 2009;139:1619–1625. doi: 10.3945/jn.109.104638.
    1. Lam Y, Ha CW, Campbell CR, Mitchell AJ, Dinudom A, Oscarsson J, Cook DI, Hunt NH, Caterson ID, Holmes AJ, et al. Increased gut permeability and microbiota change associate with mesenteric fat inflammation and metabolic dysfunction in diet-induced obese mice. PLoS One. 2012;7:e34233. doi: 10.1371/journal.pone.0034233.
    1. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA. 2013;110:9066–9071. doi: 10.1073/pnas.1219451110.
    1. Anhê FF, Roy D, Pilon G, Dudonné S, Matamoros S, Varin TV, Garofalo C, Moine Q, Desjardins Y, Levy E, et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut. 2015;64:872–883. doi: 10.1136/gutjnl-2014-307142.
    1. Cani PD, Everard A, Duparc T. Gut microbiota, enteroendocrine functions and metabolism. Curr Opin Pharmacol. 2013;13:935–940. doi: 10.1016/j.coph.2013.09.008.
    1. Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GG, Neyrinck AM, Possemiers S, Van Holle A, François P, de Vos WM, et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes. 2011;60:2775–2786. doi: 10.2337/db11-0227.
    1. Hansen KB, Rosenkilde MM, Knop FK, Wellner N, Diep TA, Rehfeld JF, Andersen UB, Holst JJ, Hansen HS. 2-Oleoyl glycerol is a GPR119 agonist and signals GLP-1 release in humans. J Clin Endocrinol Metab. 2011;96:E1409–E1417. doi: 10.1210/jc.2011-0647.

Source: PubMed

3
Sottoscrivi