Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability

P D Cani, S Possemiers, T Van de Wiele, Y Guiot, A Everard, O Rottier, L Geurts, D Naslain, A Neyrinck, D M Lambert, G G Muccioli, N M Delzenne, P D Cani, S Possemiers, T Van de Wiele, Y Guiot, A Everard, O Rottier, L Geurts, D Naslain, A Neyrinck, D M Lambert, G G Muccioli, N M Delzenne

Abstract

Background and aims: Obese and diabetic mice display enhanced intestinal permeability and metabolic endotoxaemia that participate in the occurrence of metabolic disorders. Our recent data support the idea that a selective increase of Bifidobacterium spp. reduces the impact of high-fat diet-induced metabolic endotoxaemia and inflammatory disorders. Here, we hypothesised that prebiotic modulation of gut microbiota lowers intestinal permeability, by a mechanism involving glucagon-like peptide-2 (GLP-2) thereby improving inflammation and metabolic disorders during obesity and diabetes.

Methods: Study 1: ob/ob mice (Ob-CT) were treated with either prebiotic (Ob-Pre) or non-prebiotic carbohydrates as control (Ob-Cell). Study 2: Ob-CT and Ob-Pre mice were treated with GLP-2 antagonist or saline. Study 3: Ob-CT mice were treated with a GLP-2 agonist or saline. We assessed changes in the gut microbiota, intestinal permeability, gut peptides, intestinal epithelial tight-junction proteins ZO-1 and occludin (qPCR and immunohistochemistry), hepatic and systemic inflammation.

Results: Prebiotic-treated mice exhibited a lower plasma lipopolysaccharide (LPS) and cytokines, and a decreased hepatic expression of inflammatory and oxidative stress markers. This decreased inflammatory tone was associated with a lower intestinal permeability and improved tight-junction integrity compared to controls. Prebiotic increased the endogenous intestinotrophic proglucagon-derived peptide (GLP-2) production whereas the GLP-2 antagonist abolished most of the prebiotic effects. Finally, pharmacological GLP-2 treatment decreased gut permeability, systemic and hepatic inflammatory phenotype associated with obesity to a similar extent as that observed following prebiotic-induced changes in gut microbiota.

Conclusion: We found that a selective gut microbiota change controls and increases endogenous GLP-2 production, and consequently improves gut barrier functions by a GLP-2-dependent mechanism, contributing to the improvement of gut barrier functions during obesity and diabetes.

Conflict of interest statement

Competing interests: None.

Figures

Figure 1
Figure 1
Prebiotic-associated changes in the gut microbiota. DGGE fingerprint patterns of the caecal microbial community of selected ob/ob mice fed a normal-diet (Ob-CT, white symbols), non-prebiotic control diet (Ob-Cell, grey symbols) or prebiotic diet (Ob-Pre, black symbols) for 5 weeks. The DGGE profiles were constructed using primers for (A) Total bacteria (B) Bifidobacterium spp (C) Lactobacillus spp. Cluster analysis is based on the Pearson product–moment correlation coefficient and UPGMA linkage. (D) Three-dimensional multidimensional scaling analysis conducted on the DGGE fingerprinting composite data set (total bacteria, Bifidobacterium spp. and Lactobacillus spp). CT, Cell and Pre respectively refer to the selected ob/ob mice fed a normal-diet, non-prebiotic control diet or prebiotic diet for 5 weeks. DGGE, denaturing gradient gel electrophoresis; UPGMA, unweighted pair group method with arithmetic mean clustering algorithm.
Figure 2
Figure 2
Prebiotic treatment reduces intestinal permeability. (A) Intestinal permeability assay: Plasma DX-4000–FITC (μg/ml) oral challenge measured in ob/ob mice fed a normal diet (Ob-CT), non-prebiotic control diet (Ob-Cell), prebiotic diet (Ob-Pre) for 5 weeks. The inset corresponds to the area under curve (AUC) in the same groups. (B) Plasma endotoxin (LPS) concentrations (EU/ml); the inset corresponds to correlation between plasma LPS levels and plasma DX-4000–FITC (Pearson’s r correlation and corresponding p value). (C,F) Jejunum epithelial tight-junction protein markers (ZO-1 and occludin mRNA concentrations) relative expression to Ob-CT. Data are mean with the SEM. Data with different superscript letters are significantly different (p<0.05), according to the post hoc ANOVA statistical analysis. (D,G) Correlations between intestinal permeability markers: plasma DX-4000–FITC and ZO-1 and occludin mRNA concentrations (p<0.05); the inset corresponds to Pearson’s r correlation and corresponding p value. (E,H) Immunohistochemistry score of the jejunum epithelial tight-junction proteins (ZO-1 and occludin) in wild-type (CT), Ob-CT, Ob-Cell or Ob-Pre mice. DX-4000, dextran of molecular weight 4000 Da; EU, endotoxin unit; FITC, fluroescein isothiocyanate; LPS, lipopolysaccharide.
Figure 3
Figure 3
Prebiotic treatment changes tight-junction proteins distribution. Representative immunofluorescence staining for (A) ZO-1 and (B) occludin in ob/ob mice fed a normal diet (Ob-CT), non-prebiotic control diet (Ob-Cell), prebiotic diet (Ob-Pre) for 5 weeks. WT, wild-type.
Figure 4
Figure 4
Prebiotic treatment reduces the occurrence of systemic inflammation. (A) IL1α, (B) IL1b, (C) TNFα, (D) MCP-1, (E) MIP-1a, (F) INFγ, (G) IL6, (H) IL10, (I) IL18 and (J) IL15 plasma levels (pg/ml) in ob/ob mice fed a normal diet (Ob-CT), non-prebiotic control diet (Ob-Cell) or prebiotic diet (Ob-Pre) for 5 weeks. Data are mean with the SEM. Data with different superscript letters are significantly different (p<0.05), according to the post hoc ANOVA statistical analysis. IFN, interferon; IL, interleukin; MCP-1, monocyte chemoattratant protein-1; MIP-1a, macrophage inflammatory protein-1a; TNF, tumour necrosis factor.
Figure 5
Figure 5
Changes in the gut microbiota control hepatic inflammation, oxidative stress and macrophage infiltration markers. (A,I) Inflammation: PAI-1, TNFα mRNA concentrations; (B,J) Macrophage infiltration markers: CD68, TLR4 mRNA concentrations; (E,F) Oxidative stress markers: NADPHox, iNOS mRNA concentrations in ob/ob mice fed a normal diet (Ob-CT), non-prebiotic control diet (Ob-Cell) or prebiotic diet (Ob-Pre) for 5 weeks. Data are mean with the SEM. Data with different superscript letters are significantly different (p<0.05), according to the post hoc ANOVA statistical analysis. Correlations between liver NADPHox mRNA and (C) liver CD68 mRNA, (D) TLR4 mRNA or (K) plasma MCP-1 levels. Correlations between plasma MCP-1 and (H) liver CD68 mRNA or (L) TLR4 mRNA. (G) Correlation between liver CD68 mRNA and TLR4 mRNA concentrations (p<0.05) the inset corresponds to Pearson’s r correlation and corresponding p value. iNOS, inducible nitric oxide synthase; MCP-1, monocyte chemoattractant protein-1; NADPHox, NADPH oxidase; PAI-1, plasminogen activator inhibitor; TLR, toll-like receptor; TNF, tumour necrosis factor.
Figure 6
Figure 6
Prebiotic administration increases intestinal proglucagon mRNA and correlates with intestinal permeability markers. (A) Jejunum proglucagon mRNA concentrations; (B) proximal colon proglucagon mRNA concentrations in ob/ob mice fed a normal diet (Ob-CT), non-prebiotic control diet (Ob-Cell), prebiotic diet (Ob-Pre) for 5 weeks. Data are mean with the SEM. Data with different superscript letters are significantly different (p<0.05), according to the post hoc ANOVA statistical analysis. Correlations between plasma GLP-2 levels and (C) plasma LPS plasma; or (D) DX-4000–FITC plasma levels. Correlations between proglucagon mRNA and (E) jejunum ZO-1 or (F) occludin mRNA concentrations; the inset corresponds to Pearson’s r correlation and corresponding p value. DX 4000, dextran of molecular weight 4000 Da; FITC, fluorescein isothiocyanate; GLP-2, glucogon-like protein-2; LPS, lipopolysaccharide.
Figure 7
Figure 7
Chronic GLP-2 antagonist treatment blunts prebiotic-induced reduction in endotoxaemia and hepatic inflammatory tone. (A) Plasma LPS concentrations (EU/ml); (B,D) Macrophage infiltration markers: TLR4, CD68mRNA concentrations; (C,E) oxidative stress markers: NADPHox, iNOS mRNA concentrations; (F,G) hepatic inflammation: PAI-1, TNFα mRNA concentrations; (H) jejunum epithelial tight-junction protein markers (ZO-1 and occludin mRNA) concentrations; (I) jejunum proglucagon mRNA concentrations in ob/ob mice fed a normal diet and injected twice daily with a saline (Ob-CT), prebiotic diet and injected twice daily with a saline (Ob-Pre), prebiotic diet and injected twice daily with GLP-2 antagonist (ObPre-Ant), normal diet and injected twice daily with GLP-2 antagonist (Ob-Ant) for 4 weeks. Data are mean with the SEM. *Significantly different (p<0.05) from Ob-CT and Ob-Pre-Ant according to the Kruskal–Wallis test analysis. Data with different superscript letters are significantly different (p<0.05), according to the post hoc ANOVA statistical analysis. EU, endotoxin unit; GLP-2, glucagon-like protein-2; iNOS, inducible nitric oxide synthase; NADPHox, NADPH oxidase; PAI-1, plasminogen activator inhibitor-1; PG, proglucagon; TJ, tight junction; TLR, toll-like receptor; TNF, tumour necrosis factor.
Figure 8
Figure 8
Chronic GLP-2 treatment lowers endotoxaemia, improves gut permeability markers, and reduces systemic and hepatic inflammation, oxidative stress and macrophage infiltration markers. (A) Plasma LPS concentrations (EU/ml); Cytokines and chemokines plasma levels (pg/ml): (B) IL1α; (C) IL10; (D) MIP-1a; (E) MCP-1; (F,J) hepatic inflammation: TNFα, PAI-1 mRNA concentrations; (G,I) macrophage infiltration markers: TLR4, CD68mRNA concentrations; (H,K) oxidative stress markers: iNOS, NADPHox mRNA; (L) immunohistochemistry score and (M) representative immunofluorescence staining of the jejunum epithelial tight-junction proteins (ZO-1 and occludin) measured in ob/ob mice injected twice daily with a saline vehicle (Ob-CT) or GLP-2 (Ob-GLP-2) for 12 days. Data are mean with the SEM. *Significantly different (p<0.05) from Ob-CT mice according to the two-tailed Student t test. EU, endotoxin unit; GLP-2, glucogon-like protein-2; iNOS, inducible nitric oxide synthase; IL, interleukin; LPS, lipopolysaccharide; MCP-1, monocyte chemoattractant protein-1; MIP-1a, macrophage inflammatory protein-1a; NADPHox, NADPH oxidase; TLR, toll-like receptor; TNF, tumour necrosis factor.

References

    1. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome – a new worldwide definition. Lancet 2005;366:1059–62
    1. Matarese G, Mantzoros C, La CA. Leptin and adipocytokines: bridging the gap between immunity and atherosclerosis. Curr Pharm Des 2007;13:3676–80
    1. Backhed F, Manchester JK, Semenkovich CF, et al. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A 2007;104:979–84
    1. Ley RE, Backhed F, Turnbaugh P, et al. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A 2005;102:11070–5
    1. Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 2004;101:15718–23
    1. Cani PD, Bibiloni R, Knauf C, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008;57:1470–81
    1. Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007;56:1761–72
    1. Cani PD, Neyrinck AM, Fava F, 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;50:2374–83
    1. Cai D, Yuan M, Frantz DF, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med 2005;11:183–90
    1. Shi H, Kokoeva MV, Inouye K, et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 2006;116:3015–25
    1. Poggi M, Bastelica D, Gual P, et al. C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia 2007;50:1267–76
    1. Song MJ, Kim KH, Yoon JM, et al. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun 2006;346:739–45
    1. Suganami T, Mieda T, Itoh M, et al. Attenuation of obesity-induced adipose tissue inflammation in C3H/HeJ mice carrying a Toll-like receptor 4 mutation. Biochem Biophys Res Commun 2007;354:45–9
    1. Tsukumo DM, Carvalho-Filho MA, Carvalheira JB, et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 2007;56:1986–98
    1. Adachi Y, Moore LE, Bradford BU, et al. Antibiotics prevent liver injury in rats following long-term exposure to ethanol. Gastroenterology 1995;108:218–24
    1. Lichtman SN, Keku J, Schwab JH, et al. Hepatic injury associated with small bowel bacterial overgrowth in rats is prevented by metronidazole and tetracycline. Gastroenterology 1991;100:513–9
    1. Lichtman SN, Sartor RB, Keku J, et al. Hepatic inflammation in rats with experimental small intestinal bacterial overgrowth. Gastroenterology 1990;98:414–23
    1. Nanji AA, Khettry U, Sadrzadeh SM, et al. Severity of liver injury in experimental alcoholic liver disease. Correlation with plasma endotoxin, prostaglandin E2, leukotriene B4, and thromboxane B2. Am J Pathol 1993;142:367–73
    1. Nishida J, Ekataksin W, McDonnell D, et al. Ethanol exacerbates hepatic microvascular dysfunction, endotoxemia, and lethality in septic mice. Shock 1994;1:413–8
    1. Enomoto N, Ikejima K, Yamashina S, et al. Kupffer cell sensitization by alcohol involves increased permeability to gut-derived endotoxin. Alcohol Clin Exp Res 2001;256 Suppl:51S–4S
    1. Enomoto N, Ikejima K, Bradford B, et al. Alcohol causes both tolerance and sensitization of rat Kupffer cells via mechanisms dependent on endotoxin. Gastroenterology 1998;115:443–51
    1. Rivera CA, Bradford BU, Seabra V, et al. Role of endotoxin in the hypermetabolic state after acute ethanol exposure. Am J Physiol 1998;2756 Pt 1:G1252–8
    1. Rivera CA, Tcharmtchi MH, Mendoza L, et al. Endotoxemia and hepatic injury in a rodent model of hindlimb unloading. J Appl Physiol 2003;95:1656–63
    1. Mazzon E, Cuzzocrea S. Role of TNF-alpha in ileum tight junction alteration in mouse model of restraint stress. Am J Physiol Gastrointest Liver Physiol 2008;294:G1268–80
    1. Paulos CM, Wrzesinski C, Kaiser A, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8 T cells via TLR4 signaling. J Clin Invest 2007;117:2197–204
    1. Brun P, Castagliuolo I, Leo VD, et al. Increased intestinal permeability in obese mice: new evidence in the pathogenesis of nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2007;292:G518–25
    1. Turnbaugh PJ, Backhed F, Fulton L, et al. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008;3:213–23
    1. Wang Z, Xiao G, Yao Y, et al. The role of bifidobacteria in gut barrier function after thermal injury in rats. J Trauma 2006;61:650–7
    1. Griffiths EA, Duffy LC, Schanbacher FL, et al. In vivo effects of bifidobacteria and lactoferrin on gut endotoxin concentration and mucosal immunity in Balb/c mice. Dig Dis Sci 2004;49:579–89
    1. Wang ZT, Yao YM, Xiao GX, et al. Risk factors of development of gut-derived bacterial translocation in thermally injured rats. World J Gastroenterol 2004;10:1619–24
    1. Commane DM, Shortt CT, Silvi S, et al. Effects of fermentation products of pro- and prebiotics on trans-epithelial electrical resistance in an in vitro model of the colon. Nutr Cancer 2005;51:102–9
    1. Ruan X, Shi H, Xia G, et al. Encapsulated Bifidobacteria reduced bacterial translocation in rats following hemorrhagic shock and resuscitation. Nutrition 2007;23:754–61
    1. Cani PD, Knauf C, Iglesias MA, et al. Improvement of glucose tolerance and hepatic insulin sensitivity by oligofructose requires a functional glucagon-like peptide 1 receptor. Diabetes 2006;55:1484–90
    1. Shin ED, Estall JL, Izzo A, et al. Mucosal adaptation to enteral nutrients is dependent on the physiologic actions of glucagon-like peptide-2 in mice. Gastroenterology 2005;128:1340–53
    1. Thulesen J, Knudsen LB, Hartmann B, et al. The truncated metabolite GLP-2 (3-33) interacts with the GLP-2 receptor as a partial agonist. Regul Pept 2002;103:9–15
    1. Nelson DW, Murali SG, Liu X, et al. Insulin-like growth factor I and glucagon-like peptide-2 responses to fasting followed by controlled or ad libitum refeeding in rats. Am J Physiol Regul Integr Comp Physiol 2008;294:R1175–84
    1. Thulesen J, Hartmann B, Hare KJ, et al. Glucagon-like peptide 2 (GLP-2) accelerates the growth of colonic neoplasms in mice. Gut 2004;53:1145–50
    1. Kitchen PA, FitzGerald AJ, Goodlad RA, et al. Glucagon-like peptide-2 increases sucrase-isomaltase but not caudal-related homeobox protein-2 gene expression. Am J Physiol Gastrointest Liver Physiol 2000;278:G425–8
    1. Dube PE, Rowland KJ, Brubaker PL. Glucagon-like peptide-2 activates beta-catenin signaling in the mouse intestinal crypt: role of insulin-like growth factor-I. Endocrinology 2008;149:291–301
    1. Van de Wiele T, Boon N, Possemiers S, et al. Prebiotic effects of chicory inulin in the simulator of the human intestinal microbial ecosystem. FEMS Microbiol Ecol 2004;51:143–53
    1. Boon N, De Windt W, Verstraete W, et al. Evaluation of nested PCR–DGGE (denaturing gradient gel electrophoresis) with group-specific 16S rRNA primers for the analysis of bacterial communities from different wastewater treatment plants. Fems Microbiol Ecol 2002;39:101–12
    1. Possemiers S, Bolca S, Grootaert C, et al. The prenylflavonoid isoxanthohumol from hops (Humulus lupulus L.) is activated into the potent phytoestrogen 8-prenylnaringenin in vitro and in the human intestine. J Nutr 2006;136:1862–7
    1. Wang Q, Fang CH, Hasselgren PO. Intestinal permeability is reduced and IL-10 levels are increased in septic IL-6 knockout mice. Am J Physiol Regul Integr Comp Physiol 2001;281:R1013–23
    1. Cani PD, Hoste S, Guiot Y, et al. Dietary non-digestible carbohydrates promote L-cell differentiation in the proximal colon of rats. Br J Nutr 2007;98:32–7
    1. Delzenne NM, Cani PD, Neyrinck AM. Modulation of glucagon-like peptide 1 and energy metabolism by inulin and oligofructose: experimental data. J Nutr 2007;137:2547S–51S
    1. Cani PD, Neyrinck AM, Maton N, et al. Oligofructose promotes satiety in rats fed a high-fat diet: involvement of glucagon-like peptide-1. Obes Res 2005;13:1000–7
    1. Cani PD, Daubioul CA, Reusens B, et al. Involvement of endogenous glucagon-like peptide-1(7-36) amide on glycaemia-lowering effect of oligofructose in streptozotocin-treated rats. J Endocrinol 2005;185:457–65
    1. Cani PD, Dewever C, Delzenne NM. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. Br J Nutr 2004;92:521–6
    1. McClean PL, Irwin N, Cassidy RS, et al. GIP receptor antagonism reverses obesity, insulin resistance, and associated metabolic disturbances induced in mice by prolonged consumption of high-fat diet. Am J Physiol Endocrinol Metab 2007;293:E1746–55
    1. Gault VA, McClean PL, Cassidy RS, et al. Chemical gastric inhibitory polypeptide receptor antagonism protects against obesity, insulin resistance, glucose intolerance and associated disturbances in mice fed high-fat and cafeteria diets. Diabetologia 2007;50:1752–62
    1. Althage MC, Ford EL, Wang S, et al. Targeted ablation of GIP-producing cells in transgenic mice reduces obesity and insulin resistance induced by a high fat diet. J Biol Chem 2008;283:18365–76
    1. Reimer RA, McBurney MI. Dietary fiber modulates intestinal proglucagon messenger ribonucleic acid and postprandial secretion of glucagon-like peptide-1 and insulin in rats. Endocrinology 1996;137:3948–56
    1. Delzenne NM, Kok N, Deloyer P, et al. Dietary fructans modulate polyamine concentration in the cecum of rats. J Nutr 2000;130:2456–60
    1. Kok NN, Morgan LM, Williams CM, et al. Insulin, glucagon-like peptide 1, glucose-dependent insulinotropic polypeptide and insulin-like growth factor I as putative mediators of the hypolipidemic effect of oligofructose in rats. J Nutr 1998;128:1099–103
    1. Urias-Silvas JE, Cani PD, Delmee E, et al. Physiological effects of dietary fructans extracted from Agave tequilana Gto. and Dasylirion spp. Br J Nutr 2008;99:254–61
    1. Delmee E, Cani PD, Gual G, et al. Relation between colonic proglucagon expression and metabolic response to oligofructose in high fat diet-fed mice. Life Sci 2006;79:1007–13
    1. Dube PE, Brubaker PL. Frontiers in glucagon-like peptide-2: multiple actions, multiple mediators. Am J Physiol Endocrinol Metab 2007;293:E460–5
    1. Drucker DJ, Erlich P, Asa SL, et al. Induction of intestinal epithelial proliferation by glucagon-like peptide 2. Proc Natl Acad Sci U S A 1996;93:7911–6
    1. Jasleen J, Ashley SW, Shimoda N, et al. Glucagon-like peptide 2 stimulates intestinal epithelial proliferation in vitro. Dig Dis Sci 2002;47:1135–40
    1. Cameron HL, Perdue MH. Stress impairs murine intestinal barrier function: improvement by glucagon-like peptide-2. J Pharmacol Exp Ther 2005;314:214–20
    1. Cameron HL, Yang PC, Perdue MH. Glucagon-like peptide-2-enhanced barrier function reduces pathophysiology in a model of food allergy. Am J Physiol Gastrointest Liver Physiol 2003;284:G905–12
    1. Benjamin MA, McKay DM, Yang PC, et al. Glucagon-like peptide-2 enhances intestinal epithelial barrier function of both transcellular and paracellular pathways in the mouse. Gut 2000;47:112–9
    1. Cani PD, Delzenne NM. Gut microflora as a target for energy and metabolic homeostasis. Curr Opin Clin Nutr Metab Care 2007;10:729–34
    1. Bigorgne AE, Bouchet-Delbos L, Naveau S, et al. Obesity-induced lymphocyte hyperresponsiveness to chemokines: a new mechanism of fatty liver inflammation in obese mice. Gastroenterology 2008;134:1459–69
    1. Membrez M, Blancher F, Jaquet M, et al. Gut microbiota modulation with norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J 2008;22:2416–26
    1. Yang R, Han X, Uchiyama T, et al. IL-6 is essential for development of gut barrier dysfunction after hemorrhagic shock and resuscitation in mice. Am J Physiol Gastrointest Liver Physiol 2003;285:G621–9
    1. Adams RB, Planchon SM, Roche JK. IFN-gamma modulation of epithelial barrier function. Time course, reversibility, and site of cytokine binding. J Immunol 1993;150:2356–63
    1. Coyne CB, Vanhook MK, Gambling TM, et al. Regulation of airway tight junctions by proinflammatory cytokines. Mol Biol Cell 2002;13:3218–34
    1. Nusrat A, Turner JR, Madara JL. Molecular physiology and pathophysiology of tight junctions. IV. Regulation of tight junctions by extracellular stimuli: nutrients, cytokines, and immune cells. Am J Physiol Gastrointest Liver Physiol 2000;279:G851–7
    1. Sakaguchi S, Furusawa S. Oxidative stress and septic shock: metabolic aspects of oxygen-derived free radicals generated in the liver during endotoxemia. FEMS Immunol Med Microbiol 2006;47:167–77
    1. Caplan MS, Miller-Catchpole R, Kaup S, et al. Bifidobacterial supplementation reduces the incidence of necrotizing enterocolitis in a neonatal rat model. Gastroenterology 1999;117:577–83
    1. Ruseler-van Embden JG, van Lieshout LM, Gosselink MJ, et al. Inability of Lactobacillus casei strain GG, L. acidophilus, and Bifidobacterium bifidum to degrade intestinal mucus glycoproteins. Scand J Gastroenterol 1995;30:675–80
    1. Kleessen B, Hartmann L, Blaut M. Fructans in the diet cause alterations of intestinal mucosal architecture, released mucins and mucosa-associated bifidobacteria in gnotobiotic rats. Br J Nutr 2003;89:597–606
    1. Bartholome AL, Albin DM, Baker DH, et al. Supplementation of total parenteral nutrition with butyrate acutely increases structural aspects of intestinal adaptation after an 80% jejunoileal resection in neonatal piglets. JPEN J Parenter Enteral Nutr 2004;28:210–22
    1. Tappenden KA, Drozdowski LA, Thomson AB, et al. Short-chain fatty acid-supplemented total parenteral nutrition alters intestinal structure, glucose transporter 2 (GLUT2) mRNA and protein, and proglucagon mRNA abundance in normal rats. Am J Clin Nutr 1998;68:118–25
    1. Johnson-Henry KC, Donato KA, Shen-Tu G, et al. Lactobacillus rhamnosus strain GG prevents enterohemorrhagic Escherichia coli O157:H7-induced changes in epithelial barrier function. Infect Immun 2008;76:1340–8
    1. Parassol N, Freitas M, Thoreux K, et al. Lactobacillus casei DN-114 001 inhibits the increase in paracellular permeability of enteropathogenic Escherichia coli-infected T84 cells. Res Microbiol 2005;156:256–62
    1. Martin GR, Wallace LE, Hartmann B, et al. Nutrient-stimulated GLP-2 release and crypt cell proliferation in experimental short bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2005;288:G431–8
    1. Thulesen J, Hartmann B, Kissow H, et al. Intestinal growth adaptation and glucagon-like peptide 2 in rats with ileal–jejunal transposition or small bowel resection. Dig Dis Sci 2001;46:379–88
    1. Jeppesen PB, Hartmann B, Thulesen J, et al. Glucagon-like peptide 2 improves nutrient absorption and nutritional status in short-bowel patients with no colon. Gastroenterology 2001;120:806–15
    1. Chiba M, Sanada Y, Kawano S, et al. Glicentin inhibits internalization of enteric bacteria by cultured INT-407 enterocytes. Pediatr Surg Int 2007;23:551–4
    1. Zhang W, Zhu W, Zhang J, et al. Protective effects of glucagon-like peptide 2 on intestinal ischemia-reperfusion rats. Microsurgery 2008;28:285–90
    1. Tsai CH, Hill M, Asa SL, et al. Intestinal growth-promoting properties of glucagon-like peptide-2 in mice. Am J Physiol 1997;2731 Pt 1:E77–84
    1. Dube PE, Forse CL, Bahrami J, et al. The essential role of insulin-like growth factor-1 in the intestinal tropic effects of glucagon-like peptide-2 in mice. Gastroenterology 2006;131:589–605

Source: PubMed

3
Se inscrever