Primary human colonic mucosal barrier crosstalk with super oxygen-sensitive Faecalibacterium prausnitzii in continuous culture

The gut microbiome plays an important role in human health and disease. Gnotobiotic animal and in vitro cell-based models provide some informative insights into mechanistic crosstalk. However, there is no existing system for a chronic co-culture of a human colonic mucosal barrier with super oxygen-sensitive commensal microbes, hindering the study of human-microbe interactions in a controlled manner. Here, we investigated the effects of an abundant super oxygen-sensitive commensal anaerobe, Faecalibacterium prausnitzii, on a primary human mucosal barrier using a Gut-MIcrobiome (GuMI) physiome platform that we designed and fabricated. Chronic continuous co-culture of F. prausnitzii for two days with colon epithelia, enabled by continuous flow of completely anoxic apical media and aerobic basal media, resulted in a strictly anaerobic apical environment fostering growth of and butyrate production by F. prausnitzii, while maintaining a stable colon epithelial barrier. We identified elevated differentiation and hypoxia-responsive genes and pathways in the platform compared with conventional aerobic static culture of the colon epithelia, attributable to a combination of anaerobic environment and continuous medium replenishment. Furthermore, we demonstrated anti-inflammatory effects of F. prausnitzii through HDAC and the TLR-NFKB axis. Finally, we identified that butyrate largely contributes to the anti-inflammatory effects by downregulating TLR3 and TLR4. Our results are consistent with some clinical observations regarding F. prausnitzii, thus motivating further studies employing this platform with more complex engineered colon tissues for understanding the interaction between the human colonic mucosal barrier and microbiota, pathogens, or engineered bacteria.

[1]  Christopher A. Voigt,et al.  Genetic circuit design automation for the gut resident species Bacteroides thetaiotaomicron , 2020, Nature Biotechnology.

[2]  Nobuo Sasaki,et al.  Development of a scalable co-culture system for gut anaerobes and human colon epithelium. , 2020, Gastroenterology.

[3]  M. Henn,et al.  Fecal microbiota transplantation for the improvement of metabolism in obesity: The FMT-TRIM double-blind placebo-controlled pilot trial , 2020, PLoS medicine.

[4]  Mitchell H. Murdock,et al.  The microbiota regulate neuronal function and fear extinction learning , 2019, Nature.

[5]  F. Vaziri,et al.  Induction effects of Faecalibacterium prausnitzii and its extracellular vesicles on toll-like receptor signaling pathway gene expression and cytokine level in human intestinal epithelial cells. , 2019, Cytokine.

[6]  Linda G. Griffith,et al.  Gut-Liver physiomimetics reveal paradoxical modulation of IBD-related inflammation by short-chain fatty acids , 2019, bioRxiv.

[7]  Lili Li,et al.  Dietary butyrate suppresses inflammation through modulating gut microbiota in high-fat diet-fed mice. , 2019, FEMS microbiology letters.

[8]  Raehyun Kim,et al.  Primary Cell-Derived Intestinal Models: Recapitulating Physiology. , 2019, Trends in biotechnology.

[9]  Craig P. McEntee,et al.  Divergent Roles for the IL-1 Family in Gastrointestinal Homeostasis and Inflammation , 2019, Front. Immunol..

[10]  M. Ajami,et al.  Sodium Butyrate as a Histone Deacetylase Inhibitor Affects Toll-Like Receptor 4 Expression in Colorectal Cancer Cell Lines , 2019, Immunological investigations.

[11]  S. Sunagawa,et al.  Gut microbial beta-glucuronidase and glycerol/diol dehydratase activity contribute to dietary heterocyclic amine biotransformation , 2019, BMC Microbiology.

[12]  Qiuhong Wang,et al.  Tetrandrine inhibits colon carcinoma HT-29 cells growth via the Bcl-2/Caspase 3/PARP pathway and G1/S phase , 2019, Bioscience reports.

[13]  J. Setubal,et al.  Butyrate Protects Mice from Clostridium difficile-Induced Colitis through an HIF-1-Dependent Mechanism. , 2019, Cell reports.

[14]  K. Faber,et al.  Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases , 2019, Front. Immunol..

[15]  J. Petrosino,et al.  A novel human enteroid-anaerobe co-culture system to study microbial-host interaction under physiological hypoxia , 2019, bioRxiv.

[16]  Dong-Woo Lee,et al.  A Robust Longitudinal Co-culture of Obligate Anaerobic Gut Microbiome With Human Intestinal Epithelium in an Anoxic-Oxic Interface-on-a-Chip , 2019, Front. Bioeng. Biotechnol..

[17]  Jie Huang,et al.  Sodium Butyrate Inhibits the Inflammation of Lipopolysaccharide-Induced Acute Lung Injury in Mice by Regulating the Toll-Like Receptor 4/Nuclear Factor κB Signaling Pathway. , 2019, Journal of agricultural and food chemistry.

[18]  W. S. Denney,et al.  An engineered E. coli Nissle improves hyperammonemia and survival in mice and shows dose-dependent exposure in healthy humans , 2019, Science Translational Medicine.

[19]  Diogo M. Camacho,et al.  Complex human gut microbiome cultured in anaerobic human intestine chips , 2018, bioRxiv.

[20]  Xiaotian Chen,et al.  Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. , 2018, Inflammatory bowel diseases.

[21]  S. Xia,et al.  Butyrate upregulates the TLR4 expression and the phosphorylation of MAPKs and NK-κB in colon cancer cell in vitro , 2018, Oncology letters.

[22]  Nathan Cermak,et al.  Rapid, inexpensive measurement of synthetic bacterial community composition by Sanger sequencing , 2018, bioRxiv.

[23]  Kelsey N. Retting,et al.  Bioprinted 3D Primary Human Intestinal Tissues Model Aspects of Native Physiology and ADME/Tox Functions , 2018, iScience.

[24]  P. Bork,et al.  Nutritional preferences of human gut bacteria reveal their metabolic idiosyncrasies , 2018, Nature Microbiology.

[25]  Murat Cirit,et al.  Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies , 2018, Scientific Reports.

[26]  N. Roy,et al.  Live Faecalibacterium prausnitzii induces greater TLR2 and TLR2/6 activation than the dead bacterium in an apical anaerobic co‐culture system , 2018, Cellular microbiology.

[27]  B. Patel,et al.  Sodium Butyrate Controls Cardiac Hypertrophy in Experimental Models of Rats , 2017, Cardiovascular Toxicology.

[28]  J. Clemente,et al.  The role of the gut microbiome in systemic inflammatory disease , 2018, British Medical Journal.

[29]  Y. Bhutia,et al.  Short-Chain Fatty Acid Transporters: Role in Colonic Homeostasis. , 2017, Comprehensive Physiology.

[30]  N. Roy,et al.  Live Faecalibacterium prausnitzii Does Not Enhance Epithelial Barrier Integrity in an Apical Anaerobic Co-Culture Model of the Large Intestine , 2017, Nutrients.

[31]  J. Kaur,et al.  Impact of novel N-aryl piperamide NO donors on NF-κB translocation in neuroinflammation: rational drug-designing synthesis and biological evaluation , 2017, Innate immunity.

[32]  J. Caldwell,et al.  Development and Characterization of a Human and Mouse Intestinal Epithelial Cell Monolayer Platform , 2017, Stem cell reports.

[33]  J. S. Kim,et al.  Sodium butyrate inhibits the NF‐kappa B signaling pathway and histone deacetylation, and attenuates experimental colitis in an IL‐10 independent manner , 2017, International immunopharmacology.

[34]  N. Allbritton,et al.  In Vitro Generation of Mouse Colon Crypts. , 2017, ACS biomaterials science & engineering.

[35]  M. Moyer,et al.  Epithelial TNF Receptor Signaling Promotes Mucosal Repair in Inflammatory Bowel Disease , 2017, The Journal of Immunology.

[36]  C. Lebrilla,et al.  Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion , 2017, Science.

[37]  Shao-Cong Sun,et al.  NF-κB signaling in inflammation , 2017, Signal Transduction and Targeted Therapy.

[38]  Murat Cirit,et al.  Integrated gut/liver microphysiological systems elucidates inflammatory inter‐tissue crosstalk , 2017, Biotechnology and bioengineering.

[39]  Douglas A Lauffenburger,et al.  Local remodeling of synthetic extracellular matrix microenvironments by co-cultured endometrial epithelial and stromal cells enables long-term dynamic physiological function. , 2017, Integrative biology : quantitative biosciences from nano to macro.

[40]  C. Schlieve,et al.  Fibroblast Growth Factors in the Gastrointestinal Tract: Twists and Turns , 2017, Developmental dynamics : an official publication of the American Association of Anatomists.

[41]  M. Donowitz,et al.  A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions , 2017, Scientific Reports.

[42]  J. Ragoussis,et al.  Update on hypoxia-inducible factors and hydroxylases in oxygen regulatory pathways: from physiology to therapeutics , 2017, Hypoxia.

[43]  G. Lal,et al.  Role of chemokine receptors and intestinal epithelial cells in the mucosal inflammation and tolerance , 2017, Journal of leukocyte biology.

[44]  J. A. Garrote,et al.  New IL-15 receptor-α splicing variants identified in intestinal epithelial Caco-2 cells , 2017, Innate immunity.

[45]  Michael A. Gurney,et al.  Pathophysiology of Intestinal Na+/H+ Exchange , 2016, Cellular and molecular gastroenterology and hepatology.

[46]  John T. Sauls,et al.  Effect of flow and peristaltic mixing on bacterial growth in a gut-like channel , 2016, Proceedings of the National Academy of Sciences.

[47]  C. Kuo,et al.  Wnt pathway regulation of intestinal stem cells , 2016, The Journal of physiology.

[48]  Anping Li,et al.  The CXCL8-CXCR1/2 pathways in cancer , 2016, Cytokine & growth factor reviews.

[49]  S. Iino,et al.  Epidermal growth factor is a critical regulator of the cytokine IL‐33 in intestinal epithelial cells , 2016, British journal of pharmacology.

[50]  Paul Wilmes,et al.  A microfluidics-based in vitro model of the gastrointestinal human–microbe interface , 2016, Nature Communications.

[51]  C. Lebrilla,et al.  Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella. , 2016, Cell host & microbe.

[52]  J. Collins,et al.  Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut-on-a-chip , 2015, Proceedings of the National Academy of Sciences.

[53]  E. Elinav,et al.  Epithelial IL-18 Equilibrium Controls Barrier Function in Colitis , 2015, Cell.

[54]  H. Akiyama,et al.  SOX9 maintains reserve stem cells and preserves radioresistance in mouse small intestine. , 2015, Gastroenterology.

[55]  S. Colgan,et al.  Physiologic hypoxia and oxygen homeostasis in the healthy intestine. A Review in the Theme: Cellular Responses to Hypoxia. , 2015, American journal of physiology. Cell physiology.

[56]  H. Sokol,et al.  Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease , 2015, Gut.

[57]  T. Weir,et al.  Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. , 2015, Cell host & microbe.

[58]  S. Kersten,et al.  The role of the gut microbiota in metabolic health , 2015, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[59]  M. Leclerc,et al.  Identification of Metabolic Signatures Linked to Anti-Inflammatory Effects of Faecalibacterium prausnitzii , 2015, mBio.

[60]  T. Keku,et al.  The gastrointestinal microbiota and colorectal cancer. , 2015, American journal of physiology. Gastrointestinal and liver physiology.

[61]  P. Moughan,et al.  Live Faecalibacterium prausnitzii in an apical anaerobic model of the intestinal epithelial barrier , 2015, Cellular microbiology.

[62]  K. Schulze The imaging and modelling of the physical processes involved in digestion and absorption , 2015, Acta physiologica.

[63]  Mandy B. Esch,et al.  TEER Measurement Techniques for In Vitro Barrier Model Systems , 2015, Journal of laboratory automation.

[64]  C. Borchers,et al.  An isotope-labeled chemical derivatization method for the quantitation of short-chain fatty acids in human feces by liquid chromatography-tandem mass spectrometry. , 2015, Analytica chimica acta.

[65]  Jeroen Raes,et al.  How informative is the mouse for human gut microbiota research? , 2015, Disease Models & Mechanisms.

[66]  Bin Wu,et al.  PDGF-BB and bFGF ameliorate radiation-induced intestinal progenitor/stem cell apoptosis via Akt/p53 signaling in mice. , 2014, American journal of physiology. Gastrointestinal and liver physiology.

[67]  H. Young,et al.  Does the microbiota play a role in the pathogenesis of autoimmune diseases? , 2014, Gut.

[68]  Taro Kawai,et al.  Toll-Like Receptor Signaling Pathways , 2014, Front. Immunol..

[69]  Yan Wang,et al.  The role of microbiome in central nervous system disorders , 2014, Brain, Behavior, and Immunity.

[70]  Jun Shen,et al.  Association between Faecalibacterium prausnitzii Reduction and Inflammatory Bowel Disease: A Meta-Analysis and Systematic Review of the Literature , 2014, Gastroenterology research and practice.

[71]  I. Pinchuk,et al.  G-CSF and G-CSFR are highly expressed in human gastric and colon cancers and promote carcinoma cell proliferation and migration , 2014, British Journal of Cancer.

[72]  J. Petrosino,et al.  Microbiota Modulate Behavioral and Physiological Abnormalities Associated with Neurodevelopmental Disorders , 2013, Cell.

[73]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

[74]  J. Imlay,et al.  An anaerobic bacterium, Bacteroides thetaiotaomicron, uses a consortium of enzymes to scavenge hydrogen peroxide , 2013, Molecular microbiology.

[75]  Jan Verhaegen,et al.  A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis , 2013, Gut.

[76]  J. Skotheim,et al.  Control of cell cycle transcription during G1 and S phases , 2013, Nature Reviews Molecular Cell Biology.

[77]  C. Manichanh,et al.  Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis , 2013, Alimentary pharmacology & therapeutics.

[78]  H. Sokol,et al.  Faecalibacterium prausnitzii and human intestinal health. , 2013, Current opinion in microbiology.

[79]  C. Philippe,et al.  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 , 2013, BMC Biology.

[80]  A. Andoh,et al.  Decreased abundance of Faecalibacterium prausnitzii in the gut microbiota of Crohn's disease , 2013, Journal of gastroenterology and hepatology.

[81]  M. Kagnoff,et al.  GM-CSF Produced by Nonhematopoietic Cells Is Required for Early Epithelial Cell Proliferation and Repair of Injured Colonic Mucosa , 2013, The Journal of Immunology.

[82]  Wei Sun,et al.  The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. , 2012, Molecular cell.

[83]  H. Flint,et al.  Role of the gut microbiota in nutrition and health , 2018, British Medical Journal.

[84]  D. Ingber,et al.  Human gut-on-a-chip inhabited by microbial flora that experiences intestinal peristalsis-like motions and flow. , 2012, Lab on a chip.

[85]  Zhiping Weng,et al.  Gene set enrichment analysis: performance evaluation and usage guidelines , 2012, Briefings Bioinform..

[86]  J. Clemente,et al.  The Impact of the Gut Microbiota on Human Health: An Integrative View , 2012, Cell.

[87]  M. Swartz A decade later. , 2011, Journal of pediatric health care : official publication of National Association of Pediatric Nurse Associates & Practitioners.

[88]  Gunnar C. Hansson,et al.  The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host–microbial interactions , 2010, Proceedings of the National Academy of Sciences.

[89]  Elke Cario,et al.  Toll-like receptors in inflammatory bowel diseases: A decade later , 2010, Inflammatory bowel diseases.

[90]  Cees van Kooten,et al.  The CD40-CD40L pathway contributes to the proinflammatory function of intestinal epithelial cells in inflammatory bowel disease. , 2010, The American journal of pathology.

[91]  Maria T. Abreu,et al.  Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function , 2010, Nature Reviews Immunology.

[92]  Andrew J. Wilson,et al.  Apoptotic sensitivity of colon cancer cells to histone deacetylase inhibitors is mediated by an Sp1/Sp3-activated transcriptional program involving immediate-early gene induction. , 2010, Cancer research.

[93]  Ian R. Holzman,et al.  Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. , 2009, The Journal of nutrition.

[94]  A. Zorn,et al.  Interactions between SOX factors and Wnt/β‐catenin signaling in development and disease , 2009, Developmental dynamics : an official publication of the American Association of Anatomists.

[95]  David H. Kim,et al.  Colorectal anatomy in adults at computed tomography colonography: normal distribution and the effect of age, sex, and body mass index , 2009, Endoscopy.

[96]  D. Threadgill,et al.  ERBBs in the gastrointestinal tract: recent progress and new perspectives. , 2009, Experimental cell research.

[97]  J. Doré,et al.  Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients , 2008, Proceedings of the National Academy of Sciences.

[98]  W. Tong,et al.  Transactivation of EGF receptor and ErbB2 protects intestinal epithelial cells from TNF-induced apoptosis , 2008, Proceedings of the National Academy of Sciences.

[99]  D. Lauffenburger,et al.  An inducible autocrine cascade regulates rat hepatocyte proliferation and apoptosis responses to tumor necrosis factor‐α , 2008, Hepatology.

[100]  Charles Giardina,et al.  HDAC3 overexpression and colon cancer cell proliferation and differentiation , 2008, Molecular carcinogenesis.

[101]  I. Wierstra,et al.  FOXM1, a typical proliferation-associated transcription factor , 2007, Biological chemistry.

[102]  C. Taylor,et al.  Hypoxia and gastrointestinal disease , 2007, Journal of Molecular Medicine.

[103]  Shizuo Akira,et al.  Signaling to NF-?B by Toll-like receptors , 2007 .

[104]  D. Jonkers,et al.  Review article: the role of butyrate on colonic function , 2007, Alimentary pharmacology & therapeutics.

[105]  Linda G. Griffith,et al.  Design, modeling and fabrication of a constant flow pneumatic micropump , 2007 .

[106]  J. Kiefer,et al.  Mixtures of SCFA, composed according to physiologically available concentrations in the gut lumen, modulate histone acetylation in human HT29 colon cancer cells , 2006, British Journal of Nutrition.

[107]  Jiannis Ragoussis,et al.  Concordant Regulation of Gene Expression by Hypoxia and 2-Oxoglutarate-dependent Dioxygenase Inhibition , 2006, Journal of Biological Chemistry.

[108]  Christopher J Schofield,et al.  Signalling hypoxia by HIF hydroxylases. , 2005, Biochemical and biophysical research communications.

[109]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[110]  T. Schaefer,et al.  Secretion of cytokines and chemokines by polarized human epithelial cells from the female reproductive tract. , 2005, Human reproduction.

[111]  Kimiko Yamamoto,et al.  Fluid shear stress induces differentiation of Flk-1-positive embryonic stem cells into vascular endothelial cells in vitro. , 2005, American journal of physiology. Heart and circulatory physiology.

[112]  Hans Clevers,et al.  FoxM1 is required for execution of the mitotic programme and chromosome stability , 2005, Nature Cell Biology.

[113]  G. Semenza,et al.  Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. , 2005, Blood.

[114]  Christopher J. Schofield,et al.  Oxygen sensing by HIF hydroxylases , 2004, Nature Reviews Molecular Cell Biology.

[115]  S. Kyrylenko,et al.  Differential regulation of the Sir2 histone deacetylase gene family by inhibitors of class I and II histone deacetylases , 2003, Cellular and Molecular Life Sciences CMLS.

[116]  M. Daly,et al.  PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes , 2003, Nature Genetics.

[117]  J. Davie Inhibition of histone deacetylase activity by butyrate. , 2003, The Journal of nutrition.

[118]  A. Keshavarzian,et al.  Intestinal barrier: An interface between health and disease , 2003, Journal of gastroenterology and hepatology.

[119]  S. Fukuyama,et al.  Autocrine IL-15 Mediates Intestinal Epithelial Cell Death Via the Activation of Neighboring Intraepithelial NK Cells1 , 2002, The Journal of Immunology.

[120]  H. Harmsen,et al.  Growth requirements and fermentation products of Fusobacterium prausnitzii, and a proposal to reclassify it as Faecalibacterium prausnitzii gen. nov., comb. nov. , 2002, International journal of systematic and evolutionary microbiology.

[121]  N. Koyama,et al.  Enhanced production of IL‐18 in butyrate‐treated intestinal epithelium by stimulation of the proximal promoter region , 2002, European journal of immunology.

[122]  D A Lauffenburger,et al.  Spatial range of autocrine signaling: modeling and computational analysis. , 2001, Biophysical journal.

[123]  V. Della Pietra,et al.  Genes modulated by histone acetylation as new effectors of butyrate activity , 2001, FEBS letters.

[124]  D. Podolsky,et al.  Differential Alteration in Intestinal Epithelial Cell Expression of Toll-Like Receptor 3 (TLR3) and TLR4 in Inflammatory Bowel Disease , 2000, Infection and Immunity.

[125]  Penny A. Johnson,et al.  Distribution of the interleukin‐8 receptors, CXCR1 and CXCR2, in inflamed gut tissue , 2000, The Journal of pathology.

[126]  L. Yin,et al.  The luminal short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell line. , 2000, Gastroenterology.

[127]  Elizabeth M. Topp,et al.  Transport processes in pharmaceutical systems , 1999 .

[128]  K. Audus,et al.  Characterization of the A549 cell line as a type II pulmonary epithelial cell model for drug metabolism. , 1998, Experimental cell research.

[129]  S. Gullans,et al.  Differential expression of human histone deacetylase mRNAs in response to immune cell apoptosis induction by trichostatin A and butyrate. , 1998, Biochemical and biophysical research communications.

[130]  M. Gassmann,et al.  Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1 alpha. , 1998, Genes & development.

[131]  R. Macdermott,et al.  Intestinal epithelial cells both express and respond to interleukin 15. , 1996, Gastroenterology.

[132]  G. Amidon,et al.  HT29-MTX/Caco-2 cocultures as an in vitro model for the intestinal epithelium: in vitro-in vivo correlation with permeability data from rats and humans. , 1996, Journal of pharmaceutical sciences.

[133]  G. Spindler,et al.  An Integrative View , 1992 .

[134]  L. Mallucci,et al.  Identification of an autocrine negative growth factor: Mouse β-galactoside-binding protein is a cytostatic factor and cell growth regulator , 1991, Cell.

[135]  G. Macfarlane,et al.  Short chain fatty acids in human large intestine, portal, hepatic and venous blood. , 1987, Gut.

[136]  S. Truelove,et al.  Luminal ions and short chain fatty acids as markers of functional activity of the mucosa in ulcerative colitis. , 1982, Journal of clinical pathology.

[137]  H. Flint,et al.  Formation of propionate and butyrate by the human colonic microbiota. , 2017, Environmental microbiology.

[138]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[139]  D. Hourcade,et al.  The Quantification of Glycosaminoglycans: A Comparison of HPLC, Carbazole, and Alcian Blue Methods. , 2008, Open glycoscience.

[140]  U. Kalina,et al.  Responsiveness of intestinal epithelial cell lines to lipopolysaccharide is correlated with Toll-like receptor 4 but not Toll-like receptor 2 or CD14 expression , 2002, International Journal of Colorectal Disease.

[141]  Paul Flecknell,et al.  Replacement, reduction and refinement. , 2002, ALTEX.