Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent

Laura Wrzosek, Sylvie Miquel, Marie-Louise Noordine, Stephan Bouet, Marie Joncquel Chevalier-Curt, Véronique Robert, Catherine Philippe, Chantal Bridonneau, Claire Cherbuy, Catherine Robbe-Masselot, Philippe Langella, Muriel Thomas, Laura Wrzosek, Sylvie Miquel, Marie-Louise Noordine, Stephan Bouet, Marie Joncquel Chevalier-Curt, Véronique Robert, Catherine Philippe, Chantal Bridonneau, Claire Cherbuy, Catherine Robbe-Masselot, Philippe Langella, Muriel Thomas

Abstract

Background: The intestinal mucus layer plays a key role in the maintenance of host-microbiota homeostasis. To document the crosstalk between the host and microbiota, we used gnotobiotic models to study the influence of two major commensal bacteria, Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii, on this intestinal mucus layer. B. thetaiotaomicron is known to use polysaccharides from mucus, but its effect on goblet cells has not been addressed so far. F. prausnitzii is of particular physiological importance because it can be considered as a sensor and a marker of human health. We determined whether B. thetaiotaomicron affected goblet cell differentiation, mucin synthesis and glycosylation in the colonic epithelium. We then investigated how F. prausnitzii influenced the colonic epithelial responses to B. thetaiotaomicron.

Results: B. thetaiotaomicron, an acetate producer, increased goblet cell differentiation, expression of mucus-related genes and the ratio of sialylated to sulfated mucins in mono-associated rats. B. thetaiotaomicron, therefore, stimulates the secretory lineage, favoring mucus production. When B. thetaiotaomicron was associated with F. prausnitzii, an acetate consumer and a butyrate producer, the effects on goblet cells and mucin glycosylation were diminished. F. prausnitzii, by attenuating the effects of B. thetaiotaomicron on mucus, may help the epithelium to maintain appropriate proportions of different cell types of the secretory lineage. Using a mucus-producing cell line, we showed that acetate up-regulated KLF4, a transcription factor involved in goblet cell differentiation.

Conclusions: B. thetaiotaomicron and F. prausnitzii, which are metabolically complementary, modulate, in vivo, the intestinal mucus barrier by modifying goblet cells and mucin glycosylation. Our study reveals the importance of the balance between two main commensal bacteria in maintaining colonic epithelial homeostasis via their respective effects on mucus.

Figures

Figure 1
Figure 1
Time course analysis of GIT in B. thetaiotaomicron mono-associated rats. Germfree (GF) rats were inoculated with a culture of B. thetaiotaomicron (107 CFU) and euthanized 2 days (Bt-2d) or 30 days (Bt-30d) after inoculation. (A) Establishment of B. thetaiotaomicron in the gastrointestinal tract (GIT) of Bt-30d rats (n = 13) was monitored weekly by enumeration of the bacterial counts in the feces. (B) Scanning electron microscopy images of B. thetaiotaomicron in the cecum of Bt-2d and Bt-30d rats; scale bars, 1 μm. (C) Measurement of cecal pH in GF (n = 12), Bt-2d (n = 13) and Bt-30d rats (n = 19). (D) Cecal concentration of short-chain fatty acids (SCFA) in GF (n = 16), Bt-2d (n = 13) and Bt-30d rats (n = 19); only results for acetate, propionate and butyrate are shown, other SCFA were not detected; results are expressed in mM. Means with different letters are significantly different (P-value <0.05).
Figure 2
Figure 2
Characterization of the colonic epithelial response in B. thetaiotaomicron mono-associated rats. (A) Colonic crypt depth and total number of cells per crypt were determined on sections from GF (n = 3), Bt-2d (n = 5) and Bt-30d rats (n = 7) stained with Haematoxylin-Eosin-Safran (HES). (B, C) Representative pictures and graphs showing counts of goblet cells staining with (B) alcian blue (indicated as AB) and (C) periodic acid Schiff (indicated as PAS) in GF (n = 3), Bt-2d (n = 6) and Bt-30d (n = 7) samples. Scale bars, 50 μm. (D) Representative Western blots and densitometric analyses of proteins involved in the differentiation pathway of the secretory lineage, KLF4 and Chromogranin A (ChgA), in GF (n = 7), Bt-2d (n = 6) and Bt-30d (n = 7) samples. Means with different letters are significantly different (P-value <0.05).
Figure 3
Figure 3
Effects of bacterial metabolites on KLF4 protein in HT29-MTX cells. Representative Western blot and densitometric analyses of KLF4 and P21 proteins in HT29-MTX cells incubated with (A) 0, 10, 20 mM acetate, (B) 0, 5, 10 mM propionate, and (C) 0, 1, 5 mM butyrate. Each graph reports means of three independent experiments with three internal repeats per experiment. Means with different letters are significantly different (P-value <0.05).
Figure 4
Figure 4
Behavior of B. thetaiotaomicron and F. prausnitzii in combination in vitro and in vivo. (A) Acetate, butyrate and propionate concentrations from culture media, pure cultures of B. thetaiotaomicron (Bt), F. prausnitzii (Fp) and cocultures. (B) Establishment of B. thetaiotaomicron and F. prausnitzii in the gastrointestinal tract (GIT) of Bt + Fp-30d rats (n = 16) was monitored weekly by enumeration in the feces; arrows represent the various inoculations with F. prausnitzii before successful colonization. (C) Cecal concentration of short chain fatty acids (SCFA) in the cecum of germfree (GF) (n = 12), Bt-30d rats (n = 19), and Bt + Fp-30d rats (n = 16). (D) Measurement of oxidoreduction potential (mV) in the cecal contents of GF (n = 11), Bt-2d (n = 13), Bt-30d (n = 19) and Bt + Fp-30d rats (n = 16). Means with different letters are significantly different (P-value <0.05).
Figure 5
Figure 5
Characterization of the colonic epithelial response in B. thetaiotaomicron and F. prausnitzii di-associated rats. (A) Measurement of colonic crypt depth, total number of cells per colonic crypt and counts of (B) alcian blue- (indicated as AB) and periodic acid Schiff- (indicated as PAS) positive cells per crypt in colonic sections of Bt-30d (n = 7) and Bt + Fp-30d rats (n = 6). (C) Representative Western blot and densitometric analyses of proteins involved in the differentiation pathway of the secretory lineage, KLF4 and Chromogranin A (ChgA) in Bt-30d (n = 5) and Bt + Fp-30d rats (n = 4); protein fold induction in Bt-30d rats was used as a reference and arbitrarily defined as 1. (D) Immunostaining for MUC2 in germfree (GF) (n = 3), Bt-2d (n = 6), Bt-30d (n = 7) and Bt + Fp-30d rats (n = 6); scale bars, 50 μm. The asterisk indicates a statistical difference compared to Bt-30d rats (P-value <0.05); n.s., not significant.
Figure 6
Figure 6
Effects of bacterial colonization on colonic mucin glycosylation. (A) Proportion of neutral and acidic oligosaccharides (carrying sulfate, N-acetyl neuraminic acid or N-glycolyl acid residues) in germfree (GF) rats (n = 3), Bt-2d (n = 2), Bt-30d (n = 3) and Bt + Fp 30d (n = 3) rats. (B) Ratio of sialylated to sulfated O-glycan chain in each group of rats.

References

    1. Neish AS. Microbes in gastrointestinal health and disease. Gastroenterology. 2009;136:65–80. doi: 10.1053/j.gastro.2008.10.080.
    1. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638. doi: 10.1126/science.1110591.
    1. Ley RE, Hamady M, Lozupone C, Turnbaugh PJ, Ramey RR, Bircher JS, Schlegel ML, Tucker TA, Schrenzel MD, Knight R, Gordon JI. Evolution of mammals and their gut microbes. Science. 2008;320:1647–1651. doi: 10.1126/science.1155725. Erratum in: Science 2008, 322:1188.
    1. Tap J, Mondot S, Levenez F, Pelletier E, Caron C, Furet JP, Ugarte E, Munoz-Tamayo R, Paslier DL, Nalin R, Dore J, Leclerc M. Towards the human intestinal microbiota phylogenetic core. Environ Microbiol. 2009;11:2574–2584. doi: 10.1111/j.1462-2920.2009.01982.x.
    1. Duncan SH, Hold GL, Harmsen HJ, Stewart CS, Flint HJ. Growth requirements and fermentation products of Fusobacterium prausnitzii, and a proposal to reclassify it as Faecalibacterium prausnitzii gen. nov., comb. nov. Int J Syst Evol Microbiol. 2002;52:2141–2146. doi: 10.1099/ijs.0.02241-0.
    1. Mahowald MA, Rey FE, Seedorf H, Turnbaugh PJ, Fulton RS, Wollam A, Shah N, Wang C, Magrini V, Wilson RK, Cantarel BL, Coutinho PM, Henrissat B, Crock LW, Russell A, Verberkmoes NC, Hettich RL, Gordon JI. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proc Natl Acad Sci U S A. 2009;106:5859–5864. doi: 10.1073/pnas.0901529106.
    1. Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X, Wei H, Chen Y, Lu H, Zuo J, Su M, Qiu Y, Jia W, Xiao C, Smith LM, Yang S, Holmes E, Tang H, Zhao G, Nicholson JK, Li L, Zhao L. Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci U S A. 2008;105:2117–2122. doi: 10.1073/pnas.0712038105.
    1. Johansson ME, Phillipson M, Petersson J, Velcich A, Holm L, Hansson GC. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A. 2008;105:15064–15069. doi: 10.1073/pnas.0803124105.
    1. Kim YS, Ho SB. Intestinal goblet cells and mucins in health and disease: recent insights and progress. Curr Gastroenterol Rep. 2010;12:319–330. doi: 10.1007/s11894-010-0131-2.
    1. Comelli EM, Simmering R, Faure M, Donnicola D, Mansourian R, Rochat F, Corthesy-Theulaz I, Cherbut C. Multifaceted transcriptional regulation of the murine intestinal mucus layer by endogenous microbiota. Genomics. 2008;91:70–77. doi: 10.1016/j.ygeno.2007.09.006.
    1. Kandori H, Hirayama K, Takeda M, Doi K. Histochemical, lectin-histochemical and morphometrical characteristics of intestinal goblet cells of germfree and conventional mice. Exp Anim. 1996;45:155–160. doi: 10.1538/expanim.45.155.
    1. Tomas J, Wrzosek L, Bouznad N, Bouet S, Mayeur C, Noordine ML, Honvo-Houeto E, Langella P, Thomas M, Cherbuy C. Primocolonization is associated with colonic epithelial maturation during conventionalization. FASEB J. 2013;27:645–655. doi: 10.1096/fj.12-216861.
    1. Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012;3:289–306. doi: 10.4161/gmic.19897.
    1. Koropatkin NM, Cameron EA, Martens EC. How glycan metabolism shapes the human gut microbiota. Nat Rev Microbiol. 2012;10:323–335.
    1. Kurokawa K, Itoh T, Kuwahara T, Oshima K, Toh H, Toyoda A, Takami H, Morita H, Sharma VK, Srivastava TP, Taylor TD, Noguchi H, Mori H, Ogura Y, Ehrlich DS, Itoh K, Takagi T, Sakaki Y, Hayashi T, Hattori M. Comparative metagenomics revealed commonly enriched gene sets in human gut microbiomes. DNA Res. 2007;14:169–181. doi: 10.1093/dnares/dsm018.
    1. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi: 10.1038/nature08821.
    1. Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, Chiang HC, Hooper LV, Gordon JI. A genomic view of the human-Bacteroides thetaiotaomicron symbiosis. Science. 2003;299:2074–2076. doi: 10.1126/science.1080029.
    1. Martens EC, Chiang HC, Gordon JI. Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont. Cell Host Microbe. 2008;4:447–457. doi: 10.1016/j.chom.2008.09.007.
    1. Bjursell MK, Martens EC, Gordon JI. Functional genomic and metabolic studies of the adaptations of a prominent adult human gut symbiont, Bacteroides thetaiotaomicron, to the suckling period. J Biol Chem. 2006;281:36269–36279. doi: 10.1074/jbc.M606509200.
    1. Sonnenburg JL, Xu J, Leip DD, Chen CH, Westover BP, Weatherford J, Buhler JD, Gordon JI. Glycan foraging in vivo by an intestine-adapted bacterial symbiont. Science. 2005;307:1955–1959. doi: 10.1126/science.1109051.
    1. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G, Grangette C, Vasquez N, Pochart P, Trugnan G, Thomas G, Blottière HM, Doré J, Marteau P, Seksik P, Langella P. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105:16731–16736. doi: 10.1073/pnas.0804812105.
    1. Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E, Frangeul L, Nalin R, Jarrin C, Chardon P, Marteau P, Roca J, Dore J. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006;55:205–211. doi: 10.1136/gut.2005.073817.
    1. Seksik P, Sokol H, Lepage P, Vasquez N, Manichanh C, Mangin I, Pochart P, Dore J, Marteau P. Review article: the role of bacteria in onset and perpetuation of inflammatory bowel disease. Aliment Pharmacol Ther. 2006;24:11–18. doi: 10.1111/j.1365-2036.2006.03053.x.
    1. Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, Cosnes J, Corthier G, Marteau P, Dore J. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis. 2009;15:1183–1189. doi: 10.1002/ibd.20903.
    1. Swidsinski A, Ladhoff A, Pernthaler A, Swidsinski S, Loening-Baucke V, Ortner M, Weber J, Hoffmann U, Schreiber S, Dietel M, Lochs H. Mucosal flora in inflammatory bowel disease. Gastroenterology. 2002;122:44–54. doi: 10.1053/gast.2002.30294.
    1. Rajilic-Stojanovic M, Biagi E, Heilig HG, Kajander K, Kekkonen RA, Tims S, de Vos WM. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011;141:1792–1801. doi: 10.1053/j.gastro.2011.07.043.
    1. Balamurugan R, Rajendiran E, George S, Samuel GV, Ramakrishna BS. Real-time polymerase chain reaction quantification of specific butyrate-producing bacteria, Desulfovibrio and Enterococcus faecalis in the feces of patients with colorectal cancer. J Gastroenterol Hepatol. 2008;23:1298–1303. doi: 10.1111/j.1440-1746.2008.05490.x.
    1. Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Tran Van Nhieu J, Furet JP. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One. 2011;6:e16393. doi: 10.1371/journal.pone.0016393.
    1. Graessler J, Qin Y, Zhong H, Zhang J, Licinio J, Wong ML, Xu A, Chavakis T, Bornstein AB, Ehrhart-Bornstein M, Lamounier-Zepter V, Lohmann T, Wolf T, Bornstein SR. 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. 2012. [Epub ahead of print.]
    1. Katz JP, Perreault N, Goldstein BG, Lee CS, Labosky PA, Yang VW, Kaestner KH. The zinc-finger transcription factor Klf4 is required for terminal differentiation of goblet cells in the colon. Development. 2002;129:2619–2628.
    1. Lesuffleur T, Porchet N, Aubert JP, Swallow D, Gum JR, Kim YS, Real FX, Zweibaum A. Differential expression of the human mucin genes MUC1 to MUC5 in relation to growth and differentiation of different mucus-secreting HT-29 cell subpopulations. J Cell Sci. 1993;106:771–783.
    1. Archer SY, Meng S, Shei A, Hodin RA. p21(WAF1) is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc Natl Acad Sci U S A. 1998;95:6791–6796. doi: 10.1073/pnas.95.12.6791.
    1. Burger-van Paassen N, Vincent A, Puiman PJ, van der Sluis M, Bouma J, Boehm G, van Goudoever JB, van Seuningen I, Renes IB. The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection. Biochem J. 2009;420:211–219. doi: 10.1042/BJ20082222.
    1. Macfarlane GT, Macfarlane S. Fermentation in the human large intestine: its physiologic consequences and the potential contribution of prebiotics. J Clin Gastroenterol. 2011;45:S120–S127.
    1. Coyne MJ, Chatzidaki-Livanis M, Paoletti LC, Comstock LE. Role of glycan synthesis in colonization of the mammalian gut by the bacterial symbiont Bacteroides fragilis. Proc Natl Acad Sci U S A. 2008;105:13099–13104. doi: 10.1073/pnas.0804220105.
    1. Coyne MJ, Reinap B, Lee MM, Comstock LE. Human symbionts use a host-like pathway for surface fucosylation. Science. 2005;307:1778–1781. doi: 10.1126/science.1106469.
    1. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122:107–118. doi: 10.1016/j.cell.2005.05.007.
    1. Evrard B, Balestrino D, Dosgilbert A, Bouya-Gachancard JL, Charbonnel N, Forestier C, Tridon A. Roles of capsule and lipopolysaccharide O antigen in interactions of human monocyte-derived dendritic cells and Klebsiella pneumoniae. Infect Immun. 2010;78:210–219. doi: 10.1128/IAI.00864-09.
    1. Kachlany SC, Levery SB, Kim JS, Reuhs BL, Lion LW, Ghiorse WC. Structure and carbohydrate analysis of the exopolysaccharide capsule of Pseudomonas putida G7. Environ Microbiol. 2001;3:774–784. doi: 10.1046/j.1462-2920.2001.00248.x.
    1. Turpin W, Humblot C, Noordine ML, Wrzosek L, Tomas J, Mayeur C, Cherbuy C, Guyot JP, Thomas M. Behavior of lactobacilli isolated from fermented slurry (ben-saalga) in gnotobiotic rats. PLoS One. 2013;8:e57711. doi: 10.1371/journal.pone.0057711.
    1. Cherbuy C, Honvo-Houeto E, Bruneau A, Bridonneau C, Mayeur C, Duee PH, Langella P, Thomas M. Microbiota matures colonic epithelium through a coordinated induction of cell cycle-related proteins in gnotobiotic rat. Am J Physiol Gastrointest Liver Physiol. 2010;299:G348–G357. doi: 10.1152/ajpgi.00384.2009.
    1. Rul F, Ben-Yahia L, Chegdani F, Wrzosek L, Thomas S, Noordine ML, Gitton C, Cherbuy C, Langella P, Thomas M. Impact of the metabolic activity of Streptococcus thermophilus on the colon epithelium of gnotobiotic rats. J Biol Chem. 2011;286:10288–10296. doi: 10.1074/jbc.M110.168666.
    1. Thomas M, Wrzosek L, Ben-Yahia L, Noordine ML, Gitton C, Chevret D, Langella P, Mayeur C, Cherbuy C, Rul F. Carbohydrate metabolism is essential for the colonization of Streptococcus thermophilus in the digestive tract of gnotobiotic rats. PLoS One. 2011;6:e28789. doi: 10.1371/journal.pone.0028789.
    1. Freitas M, Axelsson LG, Cayuela C, Midtvedt T, Trugnan G. Indigenous microbes and their soluble factors differentially modulate intestinal glycosylation steps in vivo. Use of a “lectin assay” to survey in vivo glycosylation changes. Histochem Cell Biol. 2005;124:423–433. doi: 10.1007/s00418-005-0004-1.
    1. Bry L, Falk PG, Midtvedt T, Gordon JI. A model of host-microbial interactions in an open mammalian ecosystem. Science. 1996;273:1380–1383. doi: 10.1126/science.273.5280.1380.
    1. Benjdia A, Martens EC, Gordon JI, Berteau O. Sulfatases and a radical S-adenosyl-L-methionine (AdoMet) enzyme are key for mucosal foraging and fitness of the prominent human gut symbiont Bacteroides thetaiotaomicron. J Biol Chem. 2011;286:25973–25982. doi: 10.1074/jbc.M111.228841.
    1. Joly F, Mayeur C, Bruneau A, Noordine ML, Meylheuc T, Langella P, Messing B, Duee PH, Cherbuy C, Thomas M. Drastic changes in fecal and mucosa-associated microbiota in adult patients with short bowel syndrome. Biochimie. 2010;92:753–761. doi: 10.1016/j.biochi.2010.02.015.
    1. Ben-Yahia L, Mayeur C, Rul F, Thomas M. Growth advantage of Streptococcus thermophilus over Lactobacillus bulgaricus in vitro and in the gastrointestinal tract of gnotobiotic rats. Benef Microbes. 2012;3:211–219. doi: 10.3920/BM2012.0012.
    1. Gaudier E, Jarry A, Blottiere HM, de Coppet P, Buisine MP, Aubert JP, Laboisse C, Cherbut C, Hoebler C. Butyrate specifically modulates MUC gene expression in intestinal epithelial goblet cells deprived of glucose. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1168–G1174. doi: 10.1152/ajpgi.00219.2004.
    1. Gaudier E, Rival M, Buisine MP, Robineau I, Hoebler C. Butyrate enemas upregulate Muc genes expression but decrease adherent mucus thickness in mice colon. Physiol Res. 2009;58:111–119.
    1. Hatayama H, Iwashita J, Kuwajima A, Abe T. The short chain fatty acid, butyrate, stimulates MUC2 mucin production in the human colon cancer cell line, LS174T. Biochem Biophys Res Commun. 2007;356:599–603. doi: 10.1016/j.bbrc.2007.03.025.
    1. Chen ZY, Rex S, Tseng CC. Kruppel-like factor 4 is transactivated by butyrate in colon cancer cells. J Nutr. 2004;134:792–798.
    1. Augenlicht L, Shi L, Mariadason J, Laboisse C, Velcich A. Repression of MUC2 gene expression by butyrate, a physiological regulator of intestinal cell maturation. Oncogene. 2003;22:4983–4992. doi: 10.1038/sj.onc.1206521.
    1. Van der Sluis M, De Koning BA, De Bruijn AC, Velcich A, Meijerink JP, Van Goudoever JB, Buller HA, Dekker J, Van Seuningen I, Renes IB, Einerhand AW. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology. 2006;131:117–129. doi: 10.1053/j.gastro.2006.04.020.
    1. Gersemann M, Becker S, Kubler I, Koslowski M, Wang G, Herrlinger KR, Griger J, Fritz P, Fellermann K, Schwab M, Wehkamp J, Stange EF. Differences in goblet cell differentiation between Crohn’s disease and ulcerative colitis. Differentiation. 2009;77:84–94. doi: 10.1016/j.diff.2008.09.008.
    1. Johansson ME, Gustafsson JK, Sjoberg KE, Petersson J, Holm L, Sjovall H, Hansson GC. Bacteria penetrate the inner mucus layer before inflammation in the dextran sulfate colitis model. PLoS One. 2010;5:e12238. doi: 10.1371/journal.pone.0012238.
    1. Tobisawa Y, Imai Y, Fukuda M, Kawashima H. Sulfation of colonic mucins by N-acetylglucosamine 6-O-sulfotransferase-2 and its protective function in experimental colitis in mice. J Biol Chem. 2010;285:6750–6760. doi: 10.1074/jbc.M109.067082.
    1. Raouf AH, Tsai HH, Parker N, Hoffman J, Walker RJ, Rhodes JM. Sulphation of colonic and rectal mucin in inflammatory bowel disease: reduced sulphation of rectal mucus in ulcerative colitis. Clin Sci (Lond) 1992;83:623–626.
    1. Gaudier E, Forestier L, Gouyer V, Huet G, Julien R, Hoebler C. Butyrate regulation of glycosylation-related gene expression: evidence for galectin-1 upregulation in human intestinal epithelial goblet cells. Biochem Biophys Res Commun. 2004;325:1044–1051. doi: 10.1016/j.bbrc.2004.10.141.
    1. Dallo SF, Kannan TR, Blaylock MW, Baseman JB. Elongation factor Tu and E1 beta subunit of pyruvate dehydrogenase complex act as fibronectin binding proteins in Mycoplasma pneumoniae. Mol Microbiol. 2002;46:1041–1051. doi: 10.1046/j.1365-2958.2002.03207.x.
    1. Lan A, Bruneau A, Bensaada M, Philippe C, Bellaud P, Rabot S, Jan G. Increased induction of apoptosis by Propionibacterium freudenreichii TL133 in colonic mucosal crypts of human microbiota-associated rats treated with 1,2-dimethylhydrazine. Br J Nutr. 2008;100:1251–1259. doi: 10.1017/S0007114508978284.
    1. Martin F, Cachon R, Pernin K, De Coninck J, Gervais P, Guichard E, Cayot N. Effect of oxidoreduction potential on aroma biosynthesis by lactic acid bacteria in nonfat yogurt. J Dairy Sci. 2011;94:614–622. doi: 10.3168/jds.2010-3372.
    1. Cherbuy C, Darcy-Vrillon B, Morel MT, Pegorier JP, Duee PH. Effect of germfree state on the capacities of isolated rat colonocytes to metabolize n-butyrate, glucose, and glutamine. Gastroenterology. 1995;109:1890–1899. doi: 10.1016/0016-5085(95)90756-4.
    1. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265–275.
    1. Chomczynski P, Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem. 1987;162:156–159.
    1. Rossez Y, Maes E, Lefebvre Darroman T, Gosset P, Ecobichon C, Joncquel Chevalier Curt M, Boneca IG, Michalski JC, Robbe-Masselot C. Almost all human gastric mucin O-glycans harbor blood group A, B or H antigens and are potential binding sites for Helicobacter pylori. Glycobiology. 2012;22:1193–1206. doi: 10.1093/glycob/cws072.

Source: PubMed

3
Subscribe