6,377
Views
3
CrossRef citations to date
0
Altmetric
Research Paper

Microbial stimulation of oxytocin release from the intestinal epithelium via secretin signaling

, , , & ORCID Icon
Article: 2256043 | Received 26 May 2023, Accepted 01 Sep 2023, Published online: 12 Sep 2023

References

  • Ojetti V, Saviano A, Brigida M, Petruzziello C, Caronna M, Gayani G, Franceschi F. Randomized control trial on the efficacy of Limosilactobacillus reuteri ATCC PTA 4659 in reducing inflammatory markers in acute uncomplicated diverticulitis. Eur J Gastroenterol Hepatol. 2022;34(5):496–21. doi:10.1097/MEG.0000000000002342.
  • Thomas CM, Saulnier DMA, Spinler JK, Hemarajata P, Gao C, Jones SE, Grimm A, Balderas MA, Burstein MD, Morra C, et al. FolC2-mediated folate metabolism contributes to suppression of inflammation by probiotic Lactobacillus reuteri. MicrobiologyOpen. 2016;5(5):802–818. doi:10.1002/mbo3.371.
  • Thomas CM, Hong T, van Pijkeren JP, Hemarajata P, Trinh DV, Hu W, Britton RA, Kalkum M, Versalovic J. Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling. PloS One. 2012;7(2):e31951. doi:10.1371/journal.pone.0031951.
  • Gao C, Major A, Rendon D, Lugo M, Jackson V, Shi Z, Mori-Akiyama Y, Versalovic J. Histamine H2 receptor-mediated suppression of intestinal inflammation by probiotic Lactobacillus reuteri. mBio. 2015;6(6):e01358–15. doi:10.1128/mBio.01358-15.
  • Hemarajata P, Gao C, Pflughoeft KJ, Thomas CM, Saulnier DM, Spinler JK, Versalovic J. Lactobacillus reuteri-specific immunoregulatory gene rsiR modulates histamine production and immunomodulation by Lactobacillus reuteri. J Bacteriol. 2013;195(24):5567–5576. doi:10.1128/JB.00261-13.
  • McCabe LR, Irwin R, Schaefer L, Britton RA. Probiotic use decreases intestinal inflammation and increases bone density in healthy male but not female mice. J Cell Physiol. 2013;228(8):1793–1798. doi:10.1002/jcp.24340.
  • Britton RA, Irwin R, Quach D, Schaefer L, Zhang J, Lee T, Parameswaran N, McCabe LR. Probiotic L. reuteri treatment prevents bone loss in a menopausal ovariectomized mouse model. J Cell Physiol. 2014;229(11):1822–1830. doi:10.1002/jcp.24636.
  • Zhang J, Motyl KJ, Irwin R, MacDougald OA, Britton RA, McCabe LR. Loss of bone and Wnt10b expression in male type 1 diabetic mice is blocked by the probiotic Lactobacillus reuteri. Endocrinology. 2015;156(9):3169–3182. doi:10.1210/EN.2015-1308.
  • Schepper JD, Collins FL, Rios-Arce ND, Raehtz S, Schaefer L, Gardinier JD, Britton RA, Parameswaran N, McCabe LR. Probiotic Lactobacillus reuteri prevents postantibiotic bone loss by reducing intestinal dysbiosis and preventing barrier disruption. J Bone Miner Res. 2019;34(4):681–698. doi:10.1002/jbmr.3635.
  • Schepper JD, Collins F, Rios-Arce ND, Kang HJ, Schaefer L, Gardinier JD, Raghuvanshi R, Quinn RA, Britton R, Parameswaran N, et al. Involvement of the gut microbiota and barrier function in glucocorticoid-induced osteoporosis. J Bone Miner Res. 2020;35(4):801–820. doi:10.1002/jbmr.3947.
  • Rios-Arce ND, Schepper JD, Dagenais A, Schaefer L, Daly-Seiler CS, Gardinier JD, Britton RA, McCabe LR, Parameswaran N. Post-antibiotic gut dysbiosis-induced trabecular bone loss is dependent on lymphocytes. Bone. 2020;134:115269. doi:10.1016/j.bone.2020.115269.
  • Nilsson AG, Sundh D, Bäckhed F, Lorentzon M. Lactobacillus reuteri reduces bone loss in older women with low bone mineral density: a randomized, placebo-controlled, double-blind, clinical trial. J Intern Med. 2018;284(3):307–317. doi:10.1111/joim.12805.
  • Poutahidis T, Kearney SM, Levkovich T, Qi P, Varian BJ, Lakritz JR, Ibrahim YM, Chatzigiagkos A, Alm EJ, Erdman SE. Microbial symbionts accelerate wound healing via the neuropeptide hormone oxytocin. PloS One. 2013;8(10):e78898. doi:10.1371/journal.pone.0078898.
  • Varian BJ, Poutahidis T, DiBenedictis BT, Levkovich T, Ibrahim Y, Didyk E, Shikhman L, Cheung HK, Hardas A, Ricciardi CE, et al. Microbial lysate upregulates host oxytocin. Brain Behav Immun. 2017;61:36–49. doi:10.1016/j.bbi.2016.11.002.
  • Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, Petrosino JF, Costa-Mattioli M. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell. 2016;165(7):1762–1775. doi:10.1016/j.cell.2016.06.001.
  • Sgritta M, Dooling SW, Buffington SA, Momin EN, Francis MB, Britton RA, Costa-Mattioli M. Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder. Neuron. 2019;101(2):246–259.e6. doi:10.1016/j.neuron.2018.11.018.
  • Buffington SA, Dooling SW, Sgritta M, Noecker C, Murillo OD, Felice DF, Turnbaugh PJ, Costa-Mattioli M. Dissecting the contribution of host genetics and the microbiome in complex behaviors. Cell. 2021;184(7):1740–1756.e16. doi:10.1016/j.cell.2021.02.009.
  • Stoop R. Neuromodulation by oxytocin and vasopressin. Neuron. 2012;76(1):142–159. doi:10.1016/j.neuron.2012.09.025.
  • Welch MG, Tamir H, Gross KJ, Chen J, Anwar M, Gershon MD. Expression and developmental regulation of oxytocin (OT) and oxytocin receptors (OTR) in the enteric nervous system (ENS) and intestinal epithelium. J Comp Neurol. 2009;512(2):256–270. doi:10.1002/cne.21872.
  • Walum H, Young LJ. The neural mechanisms and circuitry of the pair bond. Nat Rev Neurosci. 2018;19(11):643–654. doi:10.1038/s41583-018-0072-6.
  • Tamma R, Colaianni G, Zhu L, DiBenedetto A, Greco G, Montemurro G, Patano N, Strippoli M, Vergari R, Mancini L, et al. Oxytocin is an anabolic bone hormone. Proc Natl Acad Sci USA. 2009;106(17):7149–7154. doi:10.1073/pnas.0901890106.
  • Detillion CE, Craft TKS, Glasper ER, Prendergast BJ, DeVries AC. Social facilitation of wound healing. Psychoneuroendocrinology. 2004;29(8):1004–1011. doi:10.1016/j.psyneuen.2003.10.003.
  • Welch MG, Margolis KG, Li Z, Gershon MD. Oxytocin regulates gastrointestinal motility, inflammation, macromolecular permeability, and mucosal maintenance in mice. Am J Physiol Gastrointest Liver Physiol. 2014;307(8):G848–62. doi:10.1152/ajpgi.00176.2014.
  • İ̇şeri SÖ, Gedik İE, Erzik C, Uslu B, Arbak S, Gedik N, Yeğen BÇ. Oxytocin ameliorates skin damage and oxidant gastric injury in rats with thermal trauma. Burns. 2008;34(3):361–369. doi:10.1016/j.burns.2007.03.022.
  • Walter J, Britton RA, Roos S. Host-microbial symbiosis in the vertebrate gastrointestinal tract and the Lactobacillus reuteri paradigm. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4645–4652. doi:10.1073/pnas.1000099107.
  • Lin YP, Thibodeaux CH, Peña JA, Ferry GD, Versalovic J. Probiotic Lactobacillus reuteri suppress proinflammatory cytokines via c-jun. Inflamm Bowel Dis. 2008;14(8):1068–1083. doi:10.1002/ibd.20448.
  • Quach D, Parameswaran N, McCabe L, Britton RA. Characterizing how probiotic Lactobacillus reuteri 6475 and lactobacillic acid mediate suppression of osteoclast differentiation. Bone Reports. 2019;11:100227. doi:10.1016/j.bonr.2019.100227.
  • Erdman S, Poutahidis T. Probiotic ‘glow of health’: it’s more than skin deep. Benef Microbes. 2014;5(2):109–119. doi:10.3920/BM2013.0042.
  • Karimi S, Jonsson H, Lundh T, Roos S. Lactobacillus reuteri strains protect epithelial barrier integrity of IPEC-J2 monolayers from the detrimental effect of enterotoxigenic Escherichia coli. Physiol Rep. 2018;6(2):e13514. doi:10.14814/phy2.13514.
  • Reuter G. The Lactobacillus and Bifidobacterium microflora of the human intestine: composition and succession. Curr Issues Intest Microbiol. 2001;2:43–53.
  • Li F, Li X, Cheng CC, Bujdoš D, Tollenaar S, Simpson DJ, Tasseva G, Perez-Muñoz ME, Frese S, Gänzle MG, et al. A phylogenomic analysis of Limosilactobacillus reuteri reveals ancient and stable evolutionary relationships with rodents and birds and zoonotic transmission to humans. BMC Biol. 2023;21(1):53. doi:10.1186/s12915-023-01541-1.
  • Elmentaite R, Kumasaka N, Roberts K, Fleming A, Dann E, King HW, Kleshchevnikov V, Dabrowska M, Pritchard S, Bolt L, et al. Cells of the human intestinal tract mapped across space and time. Nature. 2021;597(7875):250–255. doi:10.1038/s41586-021-03852-1.
  • Fawkner-Corbett D, Antanaviciute A, Parikh K, Jagielowicz M, Gerós AS, Gupta T, Ashley N, Khamis D, Fowler D, Morrissey E, et al. Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell. 2021;184(3):810–826.e23. doi:10.1016/j.cell.2020.12.016.
  • Fujii M, Matano M, Toshimitsu K, Takano A, Mikami Y, Nishikori S, Sugimoto S, Sato T. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition. Cell Stem Cell. 2018;23(6):787–793.e6. doi:10.1016/j.stem.2018.11.016.
  • Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, Burgin G, Delorey TM, Howitt MR, Katz Y, et al. A single-cell survey of the small intestinal epithelium. Nature. 2017;551(7680):333–339. doi:10.1038/nature24489.
  • Han X, Zhou Z, Fei L, Sun H, Wang R, Chen Y, Chen H, Wang J, Tang H, Ge W, et al. Construction of a human cell landscape at single-cell level. Nature. 2020;581(7808):303–309. doi:10.1038/s41586-020-2157-4.
  • Li H, Wang X, Wang Y, Zhang M, Hong F, Wang H, Cui A, Zhao J, Ji W, Chen Y-G. Cross-species single-cell transcriptomic analysis reveals divergence of cell composition and functions in mammalian ileum epithelium. Cell Regeneration. 2022;11(1):19. doi:10.1186/s13619-022-00118-7.
  • Beumer J, Puschhof J, Bauzá-Martinez J, Martínez-Silgado A, Elmentaite R, James KR, Ross A, Hendriks D, Artegiani B, Busslinger GA, et al. High-resolution mRNA and secretome atlas of human enteroendocrine cells. Cell. 2020;181(6):1291–1306.e19. doi:10.1016/j.cell.2020.04.036.
  • Beumer J, Artegiani B, Post Y, Reimann F, Gribble F, Nguyen TN, Zeng H, Van den Born M, Van Es JH, Clevers H. Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signaling gradient. Nat Cell Biol. 2018;20(8):909–916. doi:10.1038/s41556-018-0143-y.
  • Grün D, Lyubimova A, Kester L, Wiebrands K, Basak O, Sasaki N, Clevers H, van Oudenaarden A. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature. 2015;525(7568):251–255. doi:10.1038/nature14966.
  • Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–265. doi:10.1038/nature07935.
  • Chang-Graham AL, Danhof HA, Engevik MA, Tomaro-Duchesneau C, Karandikar UC, Estes MK, Versalovic J, Britton RA, Hyser JM. Human intestinal enteroids with inducible neurogenin-3 expression as a novel model of gut hormone secretion. Cell Mol Gastroenterol Hepatol. 2019;8(2):209–229. doi:10.1016/j.jcmgh.2019.04.010.
  • Gribble FM, Reimann F. Enteroendocrine cells: chemosensors in the intestinal epithelium. Annu Rev Physiol. 2016;78(1):277–299. doi:10.1146/annurev-physiol-021115-105439.
  • Takayanagi Y, Yoshida M, Takashima A, Takanami K, Yoshida S, Nishimori K, Nishijima I, Sakamoto H, Yamagata T, Onaka T. Activation of supraoptic oxytocin neurons by secretin facilitates social recognition. Biol Psychiatry. 2017;81(3):243–251. doi:10.1016/j.biopsych.2015.11.021.
  • Nishijima I, Yamagata T, Spencer CM, Weeber EJ, Alekseyenko O, Sweatt JD, Momoi MY, Ito M, Armstrong DL, Nelson DL, et al. Secretin receptor-deficient mice exhibit impaired synaptic plasticity and social behavior. Hum Mol Genet. 2006;15(21):3241–3250. doi:10.1093/hmg/ddl402.
  • Afroze S, Meng F, Jensen K, McDaniel K, Rahal K, Onori P, Gaudio E, Alpini G, Glaser SS. The physiological roles of secretin and its receptor. Ann Transl Med. 2013;1(3):29. doi:10.3978/j.issn.2305-5839.2012.12.01.
  • Peñagarikano O, Lázaro MT, Lu X-H, Gordon A, Dong H, Lam HA, Peles E, Maidment NT, Murphy NP, Yang XW, et al. Exogenous and evoked oxytocin restores social behavior in the Cntnap2 mouse model of autism. Sci Transl Med. 2015;7(271):271ra8. doi:10.1126/scitranslmed.3010257.
  • Human Protein Atlas Group. Human Protein Atlas [Internet]. 2022; http://www.proteinatlas.org
  • Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al. Tissue-based map of the human proteome. Sci. 2015;347(6220):1260419. doi:10.1126/science.1260419.
  • Welch MG, Anwar M, Chang CY, Gross KJ, Ruggiero DA, Tamir H, Gershon MD. Combined administration of secretin and oxytocin inhibits chronic colitis and associated activation of forebrain neurons. Neurogastroent Motil. 2010;22(6):654–e202. doi:10.1111/j.1365-2982.2010.01477.x.
  • Krishnaswami S, McPheeters ML, Veenstra-VanderWeele J. A systematic review of secretin for children with autism spectrum disorders. Pediatrics. 2011;127(5):e1322–5. doi:10.1542/peds.2011-0428.
  • Roth KA, Gordon JI. Spatial differentiation of the intestinal epithelium: analysis of enteroendocrine cells containing immunoreactive serotonin, secretin, and substance P in normal and transgenic mice. Proc Natl Acad Sci USA. 1990;87(16):6408–6412. doi:10.1073/pnas.87.16.6408.
  • Jurek B, Neumann ID. The oxytocin receptor: from intracellular signaling to behavior. Physiol Rev. 2018;98(3):1805–1908. doi:10.1152/physrev.00031.2017.
  • Kerem L, Lawson EA. The effects of oxytocin on appetite regulation, food intake and metabolism in humans. IJMS. 2021;22(14):7737. doi:10.3390/ijms22147737.
  • Clodi M, Vila G, Geyeregger R, Riedl M, Stulnig TM, Struck J, Luger TA, Luger A. Oxytocin alleviates the neuroendocrine and cytokine response to bacterial endotoxin in healthy men. Am J Physiol-Endoc M. 2008;295(3):E686–91. doi:10.1152/ajpendo.90263.2008.
  • Kingsbury MA, Bilbo SD. The inflammatory event of birth: how oxytocin signaling may guide the development of the brain and gastrointestinal system. Front Neuroendocrinol. 2019;55:100794. doi:10.1016/j.yfrne.2019.100794.
  • Geenen V, Legros J-J, Franchimont P, Baudrihaye M, Defresne M-P, Boniver J. The neuroendocrine thymus: coexistence of oxytocin and neurophysin in the human thymus. Sci. 1986;232(4749):508–511. doi:10.1126/science.3961493.
  • Jankowski M, Wang D, Hajjar F, Mukaddam-Daher S, McCann SM, Gutkowska J. Oxytocin and its receptors are synthesized in the rat vasculature. Proc Natl Acad Sci USA. 2000;97(11):6207–6211. doi:10.1073/pnas.110137497.
  • Chibbar R, Miller FD, Mitchell BF. Synthesis of oxytocin in amnion, chorion, and decidua may influence the timing of human parturition. J Clin Invest. 1993;91(1):185–192. doi:10.1172/JCI116169.
  • Thackare H, Nicholson HD, Whittington K. Oxytocin—its role in male reproduction and new potential therapeutic uses. Hum Reprod Update. 2006;12(4):437–448. doi:10.1093/humupd/dmk002.
  • Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, Luo G, Jones SA, Goodwin B, Richardson JA, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab. 2005;2(4):217–225. doi:10.1016/j.cmet.2005.09.001.
  • Jones S. Mini-review: endocrine actions of fibroblast growth factor 19. Mol Pharmaceutics. 2008;5(1):42–48. doi:10.1021/mp700105z.
  • Romanov RA, Zeisel A, Bakker J, Girach F, Hellysaz A, Tomer R, Alpár A, Mulder J, Clotman F, Keimpema E, et al. Molecular interrogation of hypothalamic organization reveals distinct dopamine neuronal subtypes. Nat Neurosci. 2017;20(2):176–188. doi:10.1038/nn.4462.
  • Ding C, Leow MK‐S, Magkos F. Oxytocin in metabolic homeostasis: implications for obesity and diabetes management. Obes Rev. 2019;20(1):22–40. doi:10.1111/obr.12757.
  • Wewer Albrechtsen NJ, Kuhre RE, Toräng S, Holst JJ. The intestinal distribution pattern of appetite- and glucose regulatory peptides in mice, rats and pigs. BMC Res Notes. 2016;9(1):60. doi:10.1186/s13104-016-1872-2.
  • Polak JM, Coulling I, Bloom S, Pearse AGE. Immunofluorescent localization of secretin and enteroglucagon in human intestinal mucosa. Scand J Gastroenterol. 1971;6(8):739–744. doi:10.3109/00365527109179946.
  • Bealer SL, Crowley WR. Stimulation of central and systemic oxytocin release by histamine in the paraventricular hypothalamic nucleus: evidence for an interaction with norepinephrine. Endocrinology. 1999;140(3):1158–1164. doi:10.1210/endo.140.3.6601.
  • Kjaer A, Knigge U, Warberg J. Involvement of oxytocin in histamine- and stress-induced ACTH and prolactin secretion. Neuroendocrinology. 1995;61(6):704–713. doi:10.1159/000126898.
  • Ciosek J, Guzek JW. Neurohypophysial function and pteridines: effect of (6R)-5,6,7, 8-tetrahydro-alpha-biopterin on bioassayed hypothalamo-neurohypophysial vasopressin and oxytocin in the rat. Folia Med Cracov. 1992;33:25–35.
  • Ciosek J, Guzek JW, Orłowska-Majdak M. Neurohypophysial vasopressin and oxytocin as influenced by (6 R)-5,6,7,8-tetrahydro- α -biopterin in euhydrated and dehydrated rats. Biol Chem Hoppe Seyler. 1992;373(2):1079–1084. doi:10.1515/bchm3.1992.373.2.1079.
  • Verbalis JG, McCann MJ, McHale CM, Stricker EM. Oxytocin secretion in response to cholecystokinin and food: differentiation of nausea from satiety. Sci. 1986;232(4756):1417–1419. doi:10.1126/science.3715453.
  • Ueta Y, Kannan H, Higuchi T, Negoro H, Yamaguchi K, Yamashita H. Activation of gastric afferents increases noradrenaline release in the paraventricular nucleus and plasma oxytocin level. J Auton Nerv Syst. 2000;78(2–3):69–76. doi:10.1016/S0165-1838(99)00049-1.
  • Bondy CA, Jensen RT, Brady LS, Gainer H. Cholecystokinin evokes secretion of oxytocin and vasopressin from rat neural lobe independent of external calcium. 1989;86(13):5198–5201. doi:10.1073/pnas.86.13.5198.
  • Hashimoto H, Onaka T, Kawasaki M, Chen L, Mera T, Soya A, Saito T, Fujihara H, Sei H, Morita Y, et al. Effects of cholecystokinin (CCK)-8 on hypothalamic oxytocin-secreting neurons in rats lacking CCK-A receptor. Auton Neurosci. 2005;121(1–2):16–25. doi:10.1016/j.autneu.2005.05.002.
  • Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, Hao Y, Stoeckius M, Smibert P, Satija R. Comprehensive integration of single-cell data. Cell. 2019;177(7):1888–1902.e21. doi:10.1016/j.cell.2019.05.031.
  • Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, Preibisch S, Rueden C, Saalfeld S, Schmid B, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9(7):676–682. doi:10.1038/nmeth.2019.
  • Saxena K, Blutt SE, Ettayebi K, Zeng X-L, Broughman JR, Crawford SE, Karandikar UC, Sastri NP, Conner ME, Opekun AR, et al. Human intestinal enteroids: a new model to study human rotavirus infection, host restriction, and pathophysiology. J Virol. 2015;90(1):43–56. doi:10.1128/JVI.01930-15.
  • Sato T, Stange DE, Ferrante M, Vries RGJ, van Es JH, van den Brink S, van Houdt WJ, Pronk A, van Gorp J, Siersema PD, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–1772. doi:10.1053/j.gastro.2011.07.050.
  • Dekkers JF, Alieva M, Wellens LM, Ariese HCR, Jamieson PR, Vonk AM, Amatngalim GD, Hu H, Oost KC, Snippert HJG, et al. High-resolution 3D imaging of fixed and cleared organoids. Nat Protoc. 2019;14(6):1756–1771. doi:10.1038/s41596-019-0160-8.
  • Engevik MA, Morra CN, Röth D, Engevik K, Spinler JK, Devaraj S, Crawford SE, Estes MK, Kalkum M, Versalovic J. Microbial metabolic capacity for intestinal folate production and modulation of host folate receptors. Front Microbiol. 2019;10:2305. doi:10.3389/fmicb.2019.02305.
  • R Core Team. R: a language and environment for Statistical computing [internet]. Vienna, Austria: R Foundation for Statistical Computing; 2021. https://www.R-project.org/
  • Lenth RV. Emmeans: estimated marginal means, aka least-squares means. R package version 180 2022; https://CRAN.R-project.org/package=emmeans.
  • Bates D, Mächler M, Bolker B, Walker S. Fitting linear mixed-effects models using lme4. J Stat Softw. 2015;67(1):1–48. doi:10.18637/jss.v067.i01.