Register      Login
Animal Production Science Animal Production Science Society
Food, fibre and pharmaceuticals from animals
REVIEW

Signalling from the gut lumen

John B. Furness A B C and Jeremy J. Cottrell A
+ Author Affiliations
- Author Affiliations

A Department of Agriculture and Food, The University of Melbourne, Parkville, Vic. 3010, Australia.

B Florey Institute of Neuroscience and Mental Health, Parkville, Vic. 3010, Australia.

C Corresponding author. Email: j.furness@unimelb.edu.au

Animal Production Science 57(11) 2175-2187 https://doi.org/10.1071/AN17276
Submitted: 2 May 2017  Accepted: 3 July 2017   Published: 19 July 2017

Abstract

The lining of the gastrointestinal tract needs to be easily accessible to nutrients and, at the same time, defend against pathogens and chemical challenges. This lining is the largest and most vulnerable surface that faces the outside world. To manage the dual problems of effective nutrient conversion and defence, the gut lining has a sophisticated system for detection of individual chemical entities, pathogenic organisms and their products, and physico-chemical properties of its contents. Detection is through specific receptors that signal to the gut endocrine system, the nervous system, the immune system and local tissue defence systems. These effectors, in turn, modify digestive functions and contribute to tissue defence. Receptors for nutrients include taste receptors for sweet, bitter and savoury, free fatty acid receptors, peptide and phytochemical receptors, that are primarily located on enteroendocrine cells. Hormones released by enteroendocrine cells act locally, through the circulation and via the nervous system, to optimise digestion and mucosal health. Pathogen detection is both through antigen presentation to T-cells and through pattern-recognition receptors (PRRs). Activation of PRRs triggers local tissue defence, for example, by causing release of antimicrobials from Paneth cells. Toxic chemicals, including plant toxins, are sensed and then avoided, expelled or metabolised. It continues to be a major challenge to develop a comprehensive understanding of the integrated responses of the gastrointestinal tract to its luminal contents.

Additional keywords: microbiota, mucosal biology, nutrient receptors.


References

Abbott CR, Monterio M, Small C, Sajedi A, Smith KL, Parkinson JRC, Ghatei MA, Bloom SR (2005) The inhibitory effects of peripheral administration of peptide YY3-36 and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal–brainstem–hypothalamic pathway. Brain Research 1044, 127–131.
The inhibitory effects of peripheral administration of peptide YY3-36 and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal–brainstem–hypothalamic pathway.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXjvVOiu7s%3D&md5=b06e874da4157cd799fd13323ebfcf07CAS |

Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition and innate immunity. Cell 124, 783–801.
Pathogen recognition and innate immunity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xit1Kltb8%3D&md5=5ceb15056e3216c5a292f8bf0b03d92aCAS |

Andrews PLR, Davis CJ, Bingham S, Davidson HI, Hawthorn J, Maskell L (1990) The abdominal visceral innervation and the emetic reflex: pathways, pharmacology, and plasticity. Canadian Journal of Physiology and Pharmacology 68, 325–345.
The abdominal visceral innervation and the emetic reflex: pathways, pharmacology, and plasticity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3cXhslahs7Y%3D&md5=ef4955df201e9ae00835dd0d1eb18a69CAS |

Artis D (2008) Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nature Reviews. Immunology 8, 411–420.
Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXmt1OjtrY%3D&md5=163cfa982802939748ee49aa9eed8eafCAS |

Avau B, Depoortere I (2016) The bitter truth about bitter taste receptors: beyond sensing bitter in the oral cavity. Acta Physiologica 216, 407–420.
The bitter truth about bitter taste receptors: beyond sensing bitter in the oral cavity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXhvVens73L&md5=390837e9c7d344eff1935f722dfebe8cCAS |

Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI (2004) The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences, USA 101, 15718–15723.
The gut microbiota as an environmental factor that regulates fat storage.Crossref | GoogleScholarGoogle Scholar |

Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI (2005) Host–bacterial mutualism in the human intestine. Science 307, 1915–1920.
Host–bacterial mutualism in the human intestine.Crossref | GoogleScholarGoogle Scholar |

Berthoud HR, Kressel M, Raybould HE, Neuhuber WL (1995) Vagal sensors in the rat duodenal mucosa: distribution and structure as revealed by in vivo DiI tracing. Anatomy and Embryology 191, 203–212.
Vagal sensors in the rat duodenal mucosa: distribution and structure as revealed by in vivo DiI tracing.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2M3ovFKhsw%3D%3D&md5=437b57757ffd8837b6a07f48df0f707aCAS |

Berthoud HR, Patterson LM, Neumann F, Neuhuber WL (1997) Distribution and structure of vagal afferent intraganglionic laminar endings IGLEs in the rat gastrointestinal tract. Anatomy and Embryology 195, 183–191.
Distribution and structure of vagal afferent intraganglionic laminar endings IGLEs in the rat gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2s7pslKqsA%3D%3D&md5=7d8d2142eda682b91224aff17532f27bCAS |

Bevins CL, Salzman NH (2011) Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. National Review 9, 356–368.

Bingham JR, Ingelfinger FJ, Smithwick RH (1950) The effect of sympathectomy on abdominal pain in man. Gastroenterology 15, 18–31.

Birlouez-Aragon I, Saavedra G, Tessier FJ, Galinier A, Ait-Ameur L, Lacoste F, Niamba C-N, Alt N, Somoza V, Lecerf J-M (2010) A diet based on high-heat-treated foods promotes risk factors for diabetes mellitus and cardiovascular diseases. The American Journal of Clinical Nutrition 91, 1220–1226.
A diet based on high-heat-treated foods promotes risk factors for diabetes mellitus and cardiovascular diseases.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXlsVWrtLw%3D&md5=ba62147542009c5e28366232fbb05c83CAS |

Blanton LV, Charbonneau MR, Salih T, Barratt MJ, Venkatesh S, Ilkaveya O, Subramanian S, Manary MJ, Trehan I, Jorgensen JM, Fan Y-M, Henrissat B, Leyn SA, Rodionov DA, Osterman AL, Maleta KM, Newgard CB, Ashorn P, Dewey KG, Gordon JI (2016) Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science 351, aad3311
Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children.Crossref | GoogleScholarGoogle Scholar |

Bogunovic M, Dave SH, Tilstra JS, Chang DTW, Harpaz N, Xiong H, Mayer LF, Plevy SE (2007) Enteroendocrine cells express functional toll-like receptors. American Journal of Physiology. Gastrointestinal and Liver Physiology 292, G1770–G1783.
Enteroendocrine cells express functional toll-like receptors.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXmvF2lsLs%3D&md5=77f2591532e6ce3017fc8d56461f6d85CAS |

Braun T, Voland P, Kunz L, Prinz C, Gratzl M (2007) Enterochromaffin cells of the human gut: sensors for spices and odorants. Gastroenterology 132, 1890–1901.
Enterochromaffin cells of the human gut: sensors for spices and odorants.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXmsVymsLo%3D&md5=85d8ad48e33ce351a60305bb88bbd39fCAS |

Bremholm L, Hornum M, Henriksen BM, Larsen S, Holst JJ (2009) Glucagon-like peptide-2 increases mesenteric blood flow in humans. Scandinavian Journal of Gastroenterology 44, 314–319.
Glucagon-like peptide-2 increases mesenteric blood flow in humans.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXit1equr8%3D&md5=3c415dbd1e240c3b580cac249579afc8CAS |

Brookes SJH, Spencer NJ, Costa M, Zagorodnyuk VP (2013) Extrinsic primary afferent signalling in the gut. Nature Reviews. Gastroenterology & Hepatology 10, 286–296.
Extrinsic primary afferent signalling in the gut.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXntFylt7c%3D&md5=3db297f36d131db7adf83782961fd727CAS |

Bucinskaite V, Tolessa T, Pedersen J, Rydqvist B, Zerihun L, Holst JJ, Hellstrom PM (2009) Receptor-mediated activation of gastric vagal afferents by glucagon-like peptide-1 in the rat. Neurogastroenterology and Motility 21, 978–e78.
Receptor-mediated activation of gastric vagal afferents by glucagon-like peptide-1 in the rat.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtFCgsbnO&md5=653df92259e4fb3f791a3f1b699bbdc2CAS |

Bülbring E, Crema A (1958) Observations concerning the action of 5-hydroxytryptamine on the peristaltic refllex. British Journal of Pharmacology 13, 444–457.

Burrough ER, Arruda BL, Patience JF, Plummer PJ (2015) Alterations in the colonic microbiota of pigs associated with feeding distillers dried grains with solubles. PLoS One 10, e0141337
Alterations in the colonic microbiota of pigs associated with feeding distillers dried grains with solubles.Crossref | GoogleScholarGoogle Scholar |

Castelucci P, Robbins HL, Furness JB (2003) P2X2 purine receptor immunoreactivity of intraganglionic laminar endings in the mouse gastrointestinal tract. Cell and Tissue Research 312, 167–174.

Cerf-Bensussan N, Gaboriau-Routhiau V (2010) The immune system and the gut microbiota: friends or foes? Nature Reviews. Immunology 10, 735–744.
The immune system and the gut microbiota: friends or foes?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXht1SlurzP&md5=a266b9d5824eb696da43710a6f2ebe83CAS |

Chaudhri OB, Field BCT, Bloom SR (2008) Gastrointestinal satiety signals. International Journal of Obesity 32, S28–S31.
Gastrointestinal satiety signals.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXksl2ruw%3D%3D&md5=275ad4d2b95c030d175b55eb05b896adCAS |

Chen P, Zhao J, Gregersen H (2012) Up-regulated expression of advanced glycation end-products and their receptor in the small intestine and colon of diabetic rats. Digestive Diseases and Sciences 57, 48–57.
Up-regulated expression of advanced glycation end-products and their receptor in the small intestine and colon of diabetic rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XmtFOluw%3D%3D&md5=2e93b203a0a9492fb5c8b8528537f3a8CAS |

Cho H-J, Kosari S, Hunne B, Callaghan B, Rivera LR, Bravo DM, Furness JB (2015) Differences in hormone localisation patterns of K and L type enteroendocrine cells in the mouse and pig small intestine and colon. Cell and Tissue Research 359, 693–698.
Differences in hormone localisation patterns of K and L type enteroendocrine cells in the mouse and pig small intestine and colon.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhvFShsb%2FJ&md5=394234a9c1ac1bb6983459644423d698CAS |

Cordier-Bussat M, Bernard C, Levenez F, Klages N, Laser-Ritz B, Philippe J, Chayvialle JA, Cuber JC (1998) Peptones stimulate both the secretion of the incretin hormone glucagon-like peptide 1 and the transcription of the proglucagon gene. Diabetes 47, 1038–1045.
Peptones stimulate both the secretion of the incretin hormone glucagon-like peptide 1 and the transcription of the proglucagon gene.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXkt1ajtbo%3D&md5=2fbe8161651da7489de631ff5fe90d3aCAS |

Cox HM (2016) Neuroendocrine peptide mechanisms controlling intestinal epithelial function. Current Opinion in Pharmacology 31, 50–56.
Neuroendocrine peptide mechanisms controlling intestinal epithelial function.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC28XhsVWksb7M&md5=703e7941e99aefe91b999612b3ce7ad0CAS |

Daly K, Al-Rammahi M, Moran A, Marcello M, Niomiya Y, Shirazi-Beechey SP (2013) Sensing of amino acids by the gut-expressed taste receptor T1R1–T1R3 stimulates CCK secretion. American Journal of Physiology. Gastrointestinal and Liver Physiology 304, G271–G282.
Sensing of amino acids by the gut-expressed taste receptor T1R1–T1R3 stimulates CCK secretion.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXjsFKrtLk%3D&md5=652d6fe6a2db23edea9f7fa0a38822e2CAS |

Dann SM, Eckmann L (2007) Innate immune defenses in the intestinal tract. Current Opinion in Gastroenterology 23, 115–120.
Innate immune defenses in the intestinal tract.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXht1Kkt7c%3D&md5=0085387893cafa88c2188ce10b007e8fCAS |

Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, Kangawa K, Nakazato M (2002) The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology 123, 1120–1128.
The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xotl2gu7s%3D&md5=af4201a4acaaa7a4b6c6e34744d0be59CAS |

Diwakarla S, Fothergill LJ, Fakhry J, Callaghan B, Furness JB (2017) Heterogeneity of enterochromaffin cells within the gastrointestinal tract. Neurogastroenterology and Motility 29, e13101
Heterogeneity of enterochromaffin cells within the gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar |

Edfalk S, Steneberg P, Edlund H (2008) Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes 57, 2280–2287.
Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtFSrtrvP&md5=2cbb3aca863d60e720d324327170da10CAS |

Egerod KL, Engelstoft MS, Grunddal KV, Nøhr MK, Secher A, Sakata I, Pedersen J, Windeløv JA, Füchtbauer E-M, Olsen J, Sundler F, Christensen JP, Wierup N, Olsen JV, Holst JJ, Zigman JM, Poulsen SS, Schwartz TW (2012) A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin. Endocrinology 153, 5782–5795.
A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhvVahtbvI&md5=8e10b7887086e229e46b2673bbcb78fcCAS |

Ellis M, Chambers JD, Gwynne RM, Bornstein JC (2013) Serotonin and cholecystokinin mediate nutrient-induced segmentation in guinea pig small intestine. American Journal of Physiology. Gastrointestinal and Liver Physiology 304, G749–G761.
Serotonin and cholecystokinin mediate nutrient-induced segmentation in guinea pig small intestine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXntlGqurg%3D&md5=bc2e0a231e948d3b1327989b5f60fd8aCAS |

Engelstoft MS, Egerod KL, Holst B, Schwartz TW (2008) A gut feeling for obesity: 7TM sensors on enteroendocrine cells. Cell Metabolism 8, 447–449.
A gut feeling for obesity: 7TM sensors on enteroendocrine cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhsV2qtbjJ&md5=3a6f11f596c406f9a199ccf5343442d0CAS |

Fagarasan S, Kawamoto S, Kanagawa O, Suzuki K (2010) Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annual Review of Immunology 28, 243–273.
Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXlsVSms7w%3D&md5=437913f1f522833f38a550afafab4a5fCAS |

Feng B, La J-h, Schwartz ES, Tanaka T, McMurray TP, Gebhart GF (2012) Long-term sensitization of mechanosensitive and -insensitive afferents in mice with persistent colorectal hypersensitivity. The American Journal of Physiology 302, G676–G683.

Ferraris RP, Lee PP, Diamond JM (1989) Origin of regional and species differences in intestinal glucose uptake. The American Journal of Physiology 257, G689–G697.

Field M (2003) Intestinal ion transport and the pathophysiology of diarrhea. The Journal of Clinical Investigation 111, 931–943.
Intestinal ion transport and the pathophysiology of diarrhea.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXislOgsLc%3D&md5=825eb56e988de57e2f4d2a3873f78083CAS |

Fischbach MA, Segre JA (2016) Signaling in host-associated microbial communities. Cell 164, 1288–1300.
Signaling in host-associated microbial communities.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC28XktFKjs74%3D&md5=bdd5fb48f7e06b69aa4fcff03a4ac8c4CAS |

Fothergill LJ, Callaghan B, Hunne B, Bravo DM, Furness JB (2017) Costorage of enteroendocrine hormones evaluated at the cell and subcellular levels in male mice. Endocrinology 158, 2113–2123.
Costorage of enteroendocrine hormones evaluated at the cell and subcellular levels in male mice.Crossref | GoogleScholarGoogle Scholar |

Furness JB (2006) ‘The enteric nervous system.’ (Blackwell: Oxford)

Furness JB, Bravo DM (2015) Humans as cucinivores: comparisons with other species. Journal of Comparative Physiology. B, Biochemical, Systemic, and Environmental Physiology 185, 825–834.
Humans as cucinivores: comparisons with other species.Crossref | GoogleScholarGoogle Scholar |

Furness JB, Papka RE, Della NG, Costa M, Eskay RL (1982) Substance P-like immunoreactivity in nerves associated with the vascular system in guinea-pigs. Neuroscience 7, 447–459.
Substance P-like immunoreactivity in nerves associated with the vascular system in guinea-pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL38XhsFKhsrY%3D&md5=de46b7a7d35cc7fd67037ff160944baeCAS |

Furness JB, Jones C, Nurgali K, Clerc N (2004) Intrinsic primary afferent neurons and nerve circuits within the intestine. Progress in Neurobiology 72, 143–164.
Intrinsic primary afferent neurons and nerve circuits within the intestine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXivVemt74%3D&md5=e07bc9f9c6a69c7c183a0ea0c731ed44CAS |

Furness JB, Callaghan B, Rivera LR, Cho HJ (2014) The enteric nervous system and gastrointestinal innervation: integrated local and central control. Advances in Experimental Medicine and Biology 817, 39–71.
The enteric nervous system and gastrointestinal innervation: integrated local and central control.Crossref | GoogleScholarGoogle Scholar |

Ganusov VV, De Boer RJ (2007) Do most lymphocytes in humans really reside in the gut? Trends in Immunology 28, 514–518.
Do most lymphocytes in humans really reside in the gut?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtlajtrbL&md5=6dbba489b28e6fccf6651040a807e188CAS |

Garrett WS, Gordon JI, Glimcher LH (2010) Homeostasis and inflammation in the intestine. Cell 140, 859–870.
Homeostasis and inflammation in the intestine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXlsVSgu7o%3D&md5=2aa30de2268da2b2dd658282bb72b04eCAS |

Gershon MD, Kirchgessner AL (1991) Identification, characterization and projections of intrinsic primary afferent neurones of the submucosal plexus: activity- induced expression of c-fos immunoreactivity. Journal of the Autonomic Nervous System 33, 185–187.
Identification, characterization and projections of intrinsic primary afferent neurones of the submucosal plexus: activity- induced expression of c-fos immunoreactivity.Crossref | GoogleScholarGoogle Scholar |

Gerspach AC, Steinert RE, Schönenberger L, Graber-Maier A, Beglinger C (2011) The role of the gut sweet taste receptor in regulating GLP-1, PYY, and CCK release in humans. The American Journal of Physiology 301, E317–E325.

Ghia J-E, Li N, Wang H, Collins M, Deng Y, El-Sharkawy RT, Côté F, Mallet J, Khan WI (2009) Serotonin has a key role in pathogenesis of experimental colitis. Gastroenterology 137, 1649–1660.
Serotonin has a key role in pathogenesis of experimental colitis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhsVyhsb%2FI&md5=e5b20edccf6efaa49d635abeb1547169CAS |

Gribble FM, Reimann F (2015) Enteroendocrine cells: chemosensors in the intestinal epithelium. Annual Review of Physiology 78, 3.1–3.23.
Enteroendocrine cells: chemosensors in the intestinal epithelium.Crossref | GoogleScholarGoogle Scholar |

Gwynne RM, Clarke AJ, Furness JB, Bornstein JC (2014) Both exogenous 5-HT and endogenous 5-HT, released by fluoxetine, enhance distension evoked propulsion in guinea-pig ileum in vitro. Frontiers in Neuroscience 8, 1–9.
Both exogenous 5-HT and endogenous 5-HT, released by fluoxetine, enhance distension evoked propulsion in guinea-pig ileum in vitro.Crossref | GoogleScholarGoogle Scholar |

Habib AM, Richards P, Cairns LS, Rogers GJ, Bannon CAM, Parker HE, Morley TCE, Yeo GSH, Reimann F, Gribble FM (2012) Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry. Endocrinology 153, 3054–3065.
Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XpsFWgtbw%3D&md5=168fa328575d421328347db67827149dCAS |

Hagenbuch B, Gui C (2008) Xenobiotic transporters of the human organic anion transporting polypeptides (OATP) family. Xenobiotica 38, 778–801.
Xenobiotic transporters of the human organic anion transporting polypeptides (OATP) family.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXpt1Snurk%3D&md5=f3c6f74a393d326943a1a21482f9be1fCAS |

Haid D, Widmayer P, Breer H (2011) Nutrient sensing receptors in gastic endocrine cells. Journal of Molecular Histology 42, 355–364.
Nutrient sensing receptors in gastic endocrine cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXovFCks70%3D&md5=5fc738268e51259e9f46b06c3f906faaCAS |

Haid D, Jordan-Biegger C, Widmayer P, Breer H (2012) Receptors responsive to protein breakdown products in G-cells and D-cells of mouse, swine and human. Frontiers in Physiology 3, 1–15.
Receptors responsive to protein breakdown products in G-cells and D-cells of mouse, swine and human.Crossref | GoogleScholarGoogle Scholar |

Hao S, Sternini C, Raybould HE (2008) Role of CCK1 and Y2 receptors in activation of hindbrain neurons induced by intragastric administration of bitter taste receptor ligands. The American Journal of Physiology 294, R33–R38.

Hass N, Schwarzenbacher K, Breer H (2010) T1R3 is expressed in brush cells and ghrelin-producing cells of murine stomach. Cell and Tissue Research 339, 493–504.
T1R3 is expressed in brush cells and ghrelin-producing cells of murine stomach.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXis1yjs7g%3D&md5=b02d9f6f5b42d551d8b8f2af581ee945CAS |

Hayes MR, Covasa M (2005) CCK and 5-HT act synergistically to suppress food intake through simultaneous activation of CCK-1 and 5-HT3 receptors. Peptides 26, 2322–2330.
CCK and 5-HT act synergistically to suppress food intake through simultaneous activation of CCK-1 and 5-HT3 receptors.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXhtFylurvP&md5=416fff2bcb149de92a413a991df46889CAS |

Helander HF, Fändriks L (2012) The enteroendocrine ‘letter cells’: time for a new nomenclature? Scandinavian Journal of Gastroenterology 47, 3–12.
The enteroendocrine ‘letter cells’: time for a new nomenclature?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhs1CjurbK&md5=ff3900165e75fb36db60d43d31dfa218CAS |

Heredia DJ, Gershon MD, Koh SD, Corrigan RD, Okamoto T, Smith TK (2013) Important role of mucosal serotonin in colonic propulsion and peristaltic reflexes: in vitro analyses in mice lacking tryptophan hydroxylase 1. The Journal of Physiology 591, 5939–5957.
Important role of mucosal serotonin in colonic propulsion and peristaltic reflexes: in vitro analyses in mice lacking tryptophan hydroxylase 1.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhvVGhsLnF&md5=f8588926b5ae1062c39e4242e6cb313aCAS |

Holzer P (2007) Role of visceral afferent neurons in mucosal inflammation and defense. Current Opinion in Pharmacology 7, 563–569.
Role of visceral afferent neurons in mucosal inflammation and defense.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtlyksrrF&md5=5a689c8157696a35ac2d8f84b120d57dCAS |

Hosoi T, Okuma Y, Matsuda T, Nomura Y (2005) Novel pathway for LPS-induced afferent vagus nerve activation: possible role of nodose ganglion. Autonomic Neuroscience 120, 104–107.
Novel pathway for LPS-induced afferent vagus nerve activation: possible role of nodose ganglion.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXlvFKltLg%3D&md5=27c085fe62422a1b8605c90249ed0afaCAS |

Huang AL, Chen X, Hoon MA, Chandrashekar J, Guo W, Tränkner D, Ryba NJP, Zuker CS (2006) The cells and logic for mammalian sour taste detection. Nature 442, 934–938.
The cells and logic for mammalian sour taste detection.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XosVOgu7c%3D&md5=6579527df41df85e35f9eb8dc3092cd2CAS |

Hughes PA, Brierley SM, Martin CM (2009) TRPV1-expressing sensory fibres and IBS: links with immune function. Gut 58, 465–466.
TRPV1-expressing sensory fibres and IBS: links with immune function.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1M7jsFygtw%3D%3D&md5=3c878b348ae1f47614ff72cb0e2988acCAS |

Husted AS, Trauelsen M, Rudenko O, Hjorth SA, Schwartz TW (2017) GPCR-mediated signaling of metabolites. Cell Metabolism 25, 777–796.
GPCR-mediated signaling of metabolites.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2sXls1yltro%3D&md5=130d6a916e50eb8591aa26527d91e594CAS |

Irwin N, Flatt PR (2015) New perspectives on exploitation of incretin peptides for the treatment of diabetes and related disorders. World Journal of Diabetes 6, 1285–1295.
New perspectives on exploitation of incretin peptides for the treatment of diabetes and related disorders.Crossref | GoogleScholarGoogle Scholar |

Iwatsuki K, Uneyama H (2012) Sense of taste in the gastrointestinal tract. Journal of Pharmacological Sciences 118, 123–128.
Sense of taste in the gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xjt12iu7o%3D&md5=ab715b1061e4d7e0e62ee19e06cb70aaCAS |

Iwatsuki K, Ichikawa R, Uematsu A, Kitamura A, Uneyama H, Torii K (2012) Detecting sweet and umami tastes in the gastrointestinal tract. Acta Physiologica (Oxford, England) 204, 169–177.
Detecting sweet and umami tastes in the gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xht1eitbs%3D&md5=1303d8a0872de3751a0aba30ed2915e6CAS |

Janssen S, Depoortere I (2013) Nutrient sensing in the gut: new roads to therapeutics? Trends in Endocrinology and Metabolism 24, 92–100.
Nutrient sensing in the gut: new roads to therapeutics?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhvV2ku7jJ&md5=4ff790218c76ef6245e4cb043c9ec294CAS |

Janssen S, Laermans J, Verhulst P-J, Thijs T, Tack J, Depoortere I (2011) Bitter taste receptors and α-gustducin regulate the secretion of ghrelin with functional effects on food intake and gastric emptying. Proceedings of the National Academy of Sciences, USA 108, 2094–2099.
Bitter taste receptors and α-gustducin regulate the secretion of ghrelin with functional effects on food intake and gastric emptying.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhslyjt7c%3D&md5=4d689bf953ef86466d2bbbec67864af0CAS |

Jeyabal PVS, Kumar R, Gangula PRR, Micci M-A, Pasricha PJ (2008) Inhibitors of advanced glycation end-products prevent loss of enteric neuronal nitric oxide synthase in diabetic rats. Neurogastroenterology and Motility 20, 253–261.
Inhibitors of advanced glycation end-products prevent loss of enteric neuronal nitric oxide synthase in diabetic rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXjs1emsL0%3D&md5=a9bb624181271ebedf46ba77e8bdfcacCAS |

Keating DJ, Spencer NJ (2010) Release of 5-hydroxytryptamine from the mucosa is not required for the generation or propagation of colonic migrating motor complexes. Gastroenterology 138, 659–670.e2.
Release of 5-hydroxytryptamine from the mucosa is not required for the generation or propagation of colonic migrating motor complexes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXivVCns70%3D&md5=78772861fe6184303e44a0a4ace083e9CAS |

Kentish S, Li H, Philp LK, O’Donnell TA, Isaacs NJ, Young RL, Wittert GA, Blackshaw LA, Page AJ (2012) Diet-induced adaptation of vagal afferent function. The Journal of Physiology 590, 209–221.
Diet-induced adaptation of vagal afferent function.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhsVOhu70%3D&md5=26de0fd1a6d078d6d6e710e64eb88e35CAS |

Klaasen HLBM, Van der Heijden PJ, Stok W, Poelma FGJ, Koopman JP, Van der Brink ME, Bakker MH, Eling WMC, Beynen AC (1993) Apathogenic, intestinal, segmented, filamentous bacteria stimulate the mucosal immune system of mice. Infection and Immunity 61, 303–306.

Kyd JM, Cripps AW (2008) Functional differences between M cells and enterocytes in sampling luminal antigens. Vaccine 26, 6221–6224.
Functional differences between M cells and enterocytes in sampling luminal antigens.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtlCru77O&md5=b06edc8c6bad237e14c0cd0b5c8c2e64CAS |

Lee S-J, Lee J, Li KK, Holland D, Maughan H, Guttman DS, Yusta B, Drucker DJ (2012) Disruption of the murine Glp2r impairs Paneth cell function and increases susceptibility to small bowel enteritis. Endocrinology 153, 1141–1151.
Disruption of the murine Glp2r impairs Paneth cell function and increases susceptibility to small bowel enteritis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xjs1antb4%3D&md5=015a76ded7caab7745feae87d05228d3CAS |

Leung C, Herath CB, Jia Z, Andrikopoulos S, Brown BE, Davies MJ, Rivera LR, Furness JB, Forbes JM, Angus PW (2016a) Dietary advanced glycation end-products aggravate non-alcoholic fatty liver disease. World Journal of Gastroenterology 22, 8026–8040.
Dietary advanced glycation end-products aggravate non-alcoholic fatty liver disease.Crossref | GoogleScholarGoogle Scholar |

Leung C, Rivera L, Furness JB, Angus PW (2016b) The role of the gut microbiota in NAFLD. Nature Reviews. Gastroenterology & Hepatology 13, 412–425.
The role of the gut microbiota in NAFLD.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC28XptFyisLk%3D&md5=e443ffebceee19b422b93738b697100bCAS |

Li Y, Hao Y, Zhu J, Owyang C (2000) Serotonin released from intestinal enterochromaffin cells mediates luminal non-cholecystokinin-stimulated pancreatic secretion in rats. Gastroenterology 118, 1197–1207.
Serotonin released from intestinal enterochromaffin cells mediates luminal non-cholecystokinin-stimulated pancreatic secretion in rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXktlaiu7k%3D&md5=c02bd0872f719f1c25b36ea6523cb869CAS |

Li N, Ghia J-E, Wang H, McClemens J, Cote F, Suehiro Y, Mallet J, Khan WI (2011) Serotonin activates dendritic cell function in the context of gut inflammation. American Journal of Pathology 178, 662–671.
Serotonin activates dendritic cell function in the context of gut inflammation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXjtVOltrs%3D&md5=394766209afebc9fc388b61f1eb940b3CAS |

Lin HC, Zhao X-T, Wang L, Wong H (1996) Fat-induced ileal brake in the dog depends on peptide YY. Gastroenterology 110, 1491–1495.
Fat-induced ileal brake in the dog depends on peptide YY.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28XjtV2htLY%3D&md5=e8cc1369b94a0b9d4313d1d9a24c7e6bCAS |

Lundgren O, Peregrin AT, Persson K, Kordasti S, Uhnoo I, Svensson L (2000) Role of the enteric nervous system in the fluid and electrolyte secretion of rotavirus diarrhea. Science 287, 491–495.
Role of the enteric nervous system in the fluid and electrolyte secretion of rotavirus diarrhea.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXnsVWltQ%3D%3D&md5=1c8c0e7683e9f354b557809653fef3fdCAS |

Lynn PA, Olsson C, Zagorodnyuk V, Costa M, Brookes SJH (2003) Rectal intraganglionic laminar endings are transduction sites of extrinsic mechanoreceptors in the guinea pig rectum. Gastroenterology 125, 786–794.
Rectal intraganglionic laminar endings are transduction sites of extrinsic mechanoreceptors in the guinea pig rectum.Crossref | GoogleScholarGoogle Scholar |

MacDonald TT, Monteleone G (2005) Immunity, inflammation, and allergy in the gut. Science 307, 1920–1925.
Immunity, inflammation, and allergy in the gut.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXisVyiu7w%3D&md5=f869ba69067467f561254d881f0d2a10CAS |

Martin AM, Young RL, Leong L, Rogers GB, Spencer NJ, Jessup CF, Keating DJ (2017) The diverse metabolic roles of peripheral serotonin. Endocrinology 158, 1049–1063.
The diverse metabolic roles of peripheral serotonin.Crossref | GoogleScholarGoogle Scholar |

Mawe GM (1998) Nerves and hormones interact to control gallbladder function. News in Physiological Sciences 13, 84–90.

Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL (2005) An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122, 107–118.
An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXmsFeiurw%3D&md5=836901deb21a9057d42f6b70a4f108e9CAS |

Moran AW, Al-Rammahi MA, Arora DK, Batchelor DJ, Coulter EA, Daly K, Ionescu C, Bravo D, Shirazi-Beechey SP (2010) Expression of Na+/glucose co-transporter 1 (SGLT1) is enhanced by supplementation of the diet of weaning piglets with artificial sweeteners. British Journal of Nutrition 104, 637–646.
Expression of Na+/glucose co-transporter 1 (SGLT1) is enhanced by supplementation of the diet of weaning piglets with artificial sweeteners.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhtFCktbrE&md5=194f263c90f18a3544ff7da1ef28345fCAS |

Moran AW, Al-Rammahi M, Zhang C, Bravo D, Calsamiglia S, Shirazi-Beechey SP (2014) Sweet taste receptor expression in ruminant intestine and its activation by artificial sweeteners to regulate glucose absorption. Journal of Dairy Science 97, 4955–4972.
Sweet taste receptor expression in ruminant intestine and its activation by artificial sweeteners to regulate glucose absorption.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXpslCgtLY%3D&md5=a647f576f35bad45c62d4e7008aee4bbCAS |

Mowat AM (2003) Anatomical basis of tolerance and immunity to intestinal antigens. Nature Reviews. Immunology 3, 331–341.
Anatomical basis of tolerance and immunity to intestinal antigens.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXisFWku7s%3D&md5=fa25309ffae4befab529a8ee7479d251CAS |

Ness TJ, Gebhart GF (1990) Visceral pain: a review of experimental studies. Pain 41, 167–234.
Visceral pain: a review of experimental studies.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3czhtVGnug%3D%3D&md5=8528e02621ef3bd5660d16a0468f326aCAS |

Neutra MR, Mantis NJ, Kraehenbuhl JP (2001) Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nature Reviews. Immunology 2, 1004–1009.
Collaboration of epithelial cells with organized mucosal lymphoid tissues.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXotF2gsbg%3D&md5=f3f52244c1953b7755f44fc51d109765CAS |

Ogawa N, Ito M, Yamaguchi H, Shiuchi T, Okamoto S, Wakitani K, Minokoshi Y, Nakazato M (2012) Intestinal fatty acid infusion modulates food preference as well as calorie intake via the vagal nerve and mid-brain-hypothalamic neural pathways in rats. Metabolism: Clinical and Experimental 61, 1312–1320.
Intestinal fatty acid infusion modulates food preference as well as calorie intake via the vagal nerve and mid-brain-hypothalamic neural pathways in rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XkvFSqtL8%3D&md5=7f91e87c7c095a374c92344c0443dfc0CAS |

Ouellette AJ (2010) Paneth cells and innate mucosal immunity. Current Opinion in Gastroenterology 26, 547–553.
Paneth cells and innate mucosal immunity.Crossref | GoogleScholarGoogle Scholar |

Perez-Burgos A, Wang B, Mao Y-K, Mistry B, McVey Neufeld K-A, Bienenstock J, Kunze W (2013) Psychoactive bacteria Lactobacillus rhamnosus (JB-1) elicits rapid frequency facilitation in vagal afferents. The American Journal of Physiology 304, G211–G220.

Picó C, Oliver P, Sánchez J, Palou A (2003) Gastric leptin: a putative role in the short-term regulation of food intake. British Journal of Nutrition 90, 735–741.
Gastric leptin: a putative role in the short-term regulation of food intake.Crossref | GoogleScholarGoogle Scholar |

Powley TL, Phillips RJ (2011) Vagal intramuscular array afferents form complexes with interstitial cells of cajal in gastrointestinal smooth muscle: analogues of muscle spindle organs? Neuroscience 186, 188–200.
Vagal intramuscular array afferents form complexes with interstitial cells of cajal in gastrointestinal smooth muscle: analogues of muscle spindle organs?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXntV2ltL8%3D&md5=f13a79e74a90af80333ce788ef60a4f3CAS |

Psichas A, Reimann F, Gribble FM (2015) Gut chemosensing mechanisms. The Journal of Clinical Investigation 125, 908–917.
Gut chemosensing mechanisms.Crossref | GoogleScholarGoogle Scholar |

Ray BS, Neill CL (1947) Abdominal visceral sensation in man. Annals of Surgery 126, 709–724.
Abdominal visceral sensation in man.Crossref | GoogleScholarGoogle Scholar |

Raybould HE (2007) Mechanisms of CCK signaling from gut to brain. Current Opinion in Pharmacology 7, 570–574.
Mechanisms of CCK signaling from gut to brain.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtlyksrvN&md5=56f45616c92df1624111d7514873a7ccCAS |

Rehfeld JF (2004) A centenary of gastrointestinal endocrinology. Hormone and Metabolic Research. Hormon- und Stoffwechselforschung. Hormones et Metabolisme 36, 735–741.
A centenary of gastrointestinal endocrinology.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXnvFWltw%3D%3D&md5=cc877b5cddbc522b11a37f8c57b860a1CAS |

Reigstad CS, Salmonson CE, Rainey JF, Szurszewski JH, Linden DR, Sonnenburg JL, Farrugia G, Kashyap PC (2015) Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. The FASEB Journal 29, 1395–1403.
Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXmsFahsr4%3D&md5=5d19c80ff8bb241121b47f27c7780223CAS |

Rivera LR, Leung C, Pustovit RV, Hunne B, Andrikopoulos S, Herath C, Testro A, Angus PW, Furness JB (2014) Damage to enteric neurons occurs in mice that develop fatty liver disease but not diabetes in response to a high-fat diet. Neurogastroenterology and Motility 26, 1188–1199.
Damage to enteric neurons occurs in mice that develop fatty liver disease but not diabetes in response to a high-fat diet.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtFyntrjJ&md5=24949673c6a35acd7ee20628d18619c0CAS |

Rowland KJ, Brubaker PL (2011) The ‘cryptic’ mechanism of action of glucagon-like peptide-2. The American Journal of Physiology 301, G1–G8.

Rubio-Aliaga I, Daniel H (2008) Peptide transporters and their roles in physiological processes and drug disposition. Xenobiotica 38, 1022–1042.
Peptide transporters and their roles in physiological processes and drug disposition.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXpt1SntLc%3D&md5=9468fe9b2d941e372f31277af92720a4CAS |

Sanger GJ, Andrews PLR (2006) Treatment of nausea and vomiting: gaps in our knowledge. Autonomic Neuroscience 129, 3–16.
Treatment of nausea and vomiting: gaps in our knowledge.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtVGju7nF&md5=8a771fa26f4161830c64891b73649da8CAS |

Schmidt JB, Gregersen NT, Pedersen SD, Arentoft JL, Ritz C, Schwartz TW, Holst JJ, Astrup A, Sjödin A (2014) Effects of PYY3–36 and GLP-1 on energy intake, energy expenditure and appetite in overweight men. American Journal of Physiology. Endocrinology and Metabolism 306, E1248–E1256.
Effects of PYY3–36 and GLP-1 on energy intake, energy expenditure and appetite in overweight men.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtFaltbnF&md5=5aa6310a6ea1532c1bc9d54d551be36dCAS |

Shirazi-Beechey SP, Moran AW, Batchelor DJ, Daly K, Al-Rammahi M (2011) Influences of food constituents on gut health glucose sensing and signalling; regulation of intestinal glucose transport. The Proceedings of the Nutrition Society 70, 185–193.
Influences of food constituents on gut health glucose sensing and signalling; regulation of intestinal glucose transport.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXlslWjtL4%3D&md5=6c2c6e20fac2419bfe2f66273be47310CAS |

Sigalet DL, Wallace LE, Holst JJ, Martin GR, Kaji T, Tanaka H, Sharkey KA (2007) Enteric neural pathways mediate the anti-inflammatory actions of glucagon-like peptide 2. The American Journal of Physiology 293, G211–G221.

Smith TK, Gershon MD (2015) CrossTalk proposal: 5-HT is necessary for peristalsis. The Journal of Physiology 593, 3225–3227.
CrossTalk proposal: 5-HT is necessary for peristalsis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXht1Grt73P&md5=1272aecbc085da9a2d5ba70509f7ef7fCAS |

Smith DE, Clemencon B, Hediger MA (2013) Proton-coupled oligopeptide transporter family SLC15: physiological, pharmacological and pathological implications. Molecular Aspects of Medicine 34, 323–336.
Proton-coupled oligopeptide transporter family SLC15: physiological, pharmacological and pathological implications.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXksVyrurs%3D&md5=b3eebb6e558ccfe3c7237e2ae75b4c45CAS |

Spencer NJ, Sia TC, Brookes SJ, Costa M, Keating DJ (2015) CrossTalk opposing view: 5-HT is not necessary for peristalsis. The Journal of Physiology 593, 3229–3231.
CrossTalk opposing view: 5-HT is not necessary for peristalsis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXht1Grt73I&md5=daeccc090605686e7199e6f98d218f9dCAS |

Strader AD, Woods SC (2005) Gastrointestinal hormones and food intake. Gastroenterology 128, 175–191.
Gastrointestinal hormones and food intake.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXhtFaht7c%3D&md5=7991f66f83629b2b455f15b7b0e94f60CAS |

Szurszewski JH, Miller SM (1994) Physiology of prevertebral ganglia. In ‘Physiology of the gastrointestinal tract’. 3rd edn. (Ed. LR Johnson) pp. 795–877. (Raven Press: New York)

Thelen K, Dressman JB (2009) Cytochrome P450-mediated metabolism in the human gut wall. The Journal of Pharmacy and Pharmacology 61, 541–558.
Cytochrome P450-mediated metabolism in the human gut wall.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXmtVentA%3D%3D&md5=64827d017f89691356ec57d4cb74ac80CAS |

Treiner E, Duban L, Bahram S, Radosavljevic M, Wanner V, Tilloy F, Affaticati P, Gilfillan S, Lantz O (2003) Selection of evolutionarily conserved mucosal-associated invariant T cell by MR1. Nature 422, 164–169.
Selection of evolutionarily conserved mucosal-associated invariant T cell by MR1.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXhvFKgsr8%3D&md5=604982a419dba41d8ff1f4aba17a0bdfCAS |

Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV (2008) Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host–microbial interface. Proceedings of the National Academy of Sciences, USA 105, 20858–20863.
Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host–microbial interface.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXksFSntQ%3D%3D&md5=6b175621bf3048feca3565ddf0d1a3feCAS |

Voigt J-P, Fink H, Marsden CA (1995) Evidence for the involvement of the 5-HT1A receptor in CCK induced satiety in rats. Naunyn-Schmiedeberg’s Archives of Pharmacology 351, 217–220.
Evidence for the involvement of the 5-HT1A receptor in CCK induced satiety in rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXkvFWmsb0%3D&md5=4783cd2b373010fd94052bfee1989a3cCAS |

Wang F-B, Powley TL (2007) Vagal innervation of intestines: afferent pathways mapped with new en bloc horseradish peroxidase adaptation. Cell and Tissue Research 329, 221–230.
Vagal innervation of intestines: afferent pathways mapped with new en bloc horseradish peroxidase adaptation.Crossref | GoogleScholarGoogle Scholar |

Wang Y, Chandra R, Samasa LA, Gooch B, Fee BE, Cook JM, Vigna SR, Grant AO, Liddle RA (2011) Amino acids stimulate cholecystokinin release through the Ca2+-sensing receptor. American Journal of Physiology. Gastrointestinal and Liver Physiology 300, G528–G537.
Amino acids stimulate cholecystokinin release through the Ca2+-sensing receptor.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXlt1Ortrg%3D&md5=56a5be63e057d8a55178d6d71c3329d5CAS |

Wu SV, Rozengurt N, Yang M, Young SH, Sinnett Smith J, Rozengurt E (2002) Expression of bitter taste receptors of the T2R family in the gastrointestinal tract and enteroendocrine STC-1 cells. Proceedings of the National Academy of Sciences, USA 99, 2392–2397.
Expression of bitter taste receptors of the T2R family in the gastrointestinal tract and enteroendocrine STC-1 cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XitVSru7o%3D&md5=fd49d502af713412e7dd3a140e3d2a5bCAS |

Xiong Y, Swaminath G, Cao Q, Yang L, Guo Q, Salomonis H, Lu J, Houze JB, Dransfield PJ, Wang Y, Liu J, Wong S, Schwandner R, Steger F, Baribault H, Liu L, Coberly S, Miao L, Zhang J, Lin DCH, Schwarz M (2013) Activation of FFA1 mediates GLP-1 secretion in mice. Evidence for allosterism at FFA1. Molecular and Cellular Endocrinology 369, 119–129.
Activation of FFA1 mediates GLP-1 secretion in mice. Evidence for allosterism at FFA1.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXjtl2it70%3D&md5=0cc076e890c582fe7d48b7cacc5046bbCAS |

Yeoman CJ, White BA (2014) Gastrointestinal tract microbiota and probiotics in production animals. Annual Review of Animal Biosciences 2, 469–486.
Gastrointestinal tract microbiota and probiotics in production animals.Crossref | GoogleScholarGoogle Scholar |

Young RL (2011) Sensing via intestinal sweet taste pathways. Frontiers in Neuroscience 5, 1–13.
Sensing via intestinal sweet taste pathways.Crossref | GoogleScholarGoogle Scholar |

Yusta B, Holland D, Waschek JA, Drucker DJ (2012) Intestinotrophic glucagon-like peptide-2 (GLP-2) activates intestinal gene expression and growth factor-dependent pathways independent of the vasoactive intestinal peptide gene in mice. Endocrinology 153, 2623–2632.
Intestinotrophic glucagon-like peptide-2 (GLP-2) activates intestinal gene expression and growth factor-dependent pathways independent of the vasoactive intestinal peptide gene in mice.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xns1eitL4%3D&md5=384945ed776c5046e6b36017acca8b14CAS |

Zagorodnyuk VP, Chen BN, Brookes SJH (2001) Intraganglionic laminar endings are mechano-transduction sites of vagal tension receptors in the guinea-pig stomach. The Journal of Physiology 534, 255–268.
Intraganglionic laminar endings are mechano-transduction sites of vagal tension receptors in the guinea-pig stomach.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXltlejtLs%3D&md5=fba64c332ddefcdb7b83f4600ec8f37eCAS |

Zong H, Madden A, Ward M, Mooney MH, Elliott CT, Stitt AW (2010) Homodimerization is essential for the receptor for advanced glycation end products (RAGE)-mediated signal transduction. The Journal of Biological Chemistry 285, 23137–23146.
Homodimerization is essential for the receptor for advanced glycation end products (RAGE)-mediated signal transduction.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXovFelsrw%3D&md5=4d7f12430c947e5233eedc776ff19463CAS |