Gut Bacteria and Neurotransmitters
Abstract
:1. Introduction
2. Bacteria Dominates the Human Gut Microbiome
3. Wiring of the Gut Wall to the CNS
4. The Smaller Brain in Our Gut
5. Neurotransmitters and Neuropeptides
5.1. Glutamate (Glu)
5.2. Gamma-Aminobutyric Acid (GABA)
5.3. Glycine
5.4. Dopamine (DA)
5.5. Norepinephrine (NE) or Noradrenaline (NAd)
5.6. Serotonin (5-HT)
5.7. Histamine
6. Role of Short Chain Fatty Acids in Neurotransmission
7. Conclusions
Funding
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Dicks, L.M.T.; Hurn, D.; Hermanus, D. Gut Bacteria and Neuropsychiatric Disorders. Microorganisms 2021, 9, 2583. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Lou, J.; Shan, W.; Ding, J.; Jin, Z.; Hu, Y.; Du, Q.; Liao, Q.; Xie, R.; Xu, J. Pathophysiologic Role of Neurotransmitters in Digestive Diseases. Front. Physiol. 2021, 12, 567650. [Google Scholar] [CrossRef] [PubMed]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef] [PubMed]
- Van De Wouw, M.; Boehme, M.; Lyte, J.M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain-gut axis alterations. J. Physiol. 2018, 596, 4923–4944. [Google Scholar] [CrossRef] [PubMed]
- Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [PubMed]
- MahmoudianDehkordi, S.; Arnold, M.; Nho, K.; Ahmad, S.; Jia, W.; Xie, G.; Louie, G.; Kueider-Paisley, A.; Moseley, M.A.; Thompson, J.W.; et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease—An emerging role for gut microbiome. Alzheimer’s Dement. 2019, 15, 76–92. [Google Scholar] [CrossRef]
- Baj, A.; Moro, E.; Bistoletti, M.; Orlandi, V.; Crema, F.; Giaroni, C. Glutamatergic Signaling Along The Microbiota-Gut-Brain Axis. Int. J. Mol. Sci. 2019, 20, 1482. [Google Scholar] [CrossRef]
- Tracey, K.J.; Chavan, S.S. Nerve Stimulation for Treatment of Diseases and Disorders. Patent Application No 15/677,080, 25 Ene 2018. [Google Scholar]
- Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-Gut Microbiota Metabolic Interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef]
- Brown, G.C. The endotoxin hypothesis of neurodegeneration. J. Neuroinflammation 2019, 16, 180. [Google Scholar] [CrossRef]
- Larsson, J.M.H.; Karlsson, H.; Crespo, J.G.; Johansson, M.E.V.; Eklund, L.; Sjövall, H.; Hansson, G.C. Altered O-glycosylation profile of MUC2 mucin occurs in active ulcerative colitis and is associated with increased inflammation. Inflamm. Bowel Dis. 2011, 17, 2299–2307. [Google Scholar] [CrossRef]
- Rausch, P.; Rehman, A.; Künzel, S.; Häsler, R.; Ott, S.J.; Schreiber, S.; Rosenstiel, P.; Franke, A.; Baines, J.F. Colonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (Secretor) genotype. Proc. Natl. Acad. Sci. USA 2011, 108, 19030–19035. [Google Scholar] [CrossRef] [PubMed]
- Tong, M.; McHardy, I.; Ruegger, P.; Goudarzi, M.; Kashyap, P.C.; Haritunians, T.; Li, X.; Graeber, T.G.; Schwager, E.; Huttenhower, C.; et al. Reprograming of gut microbiome energy metabolism by the FUT2 Crohn’s disease risk polymorphism. ISME J. 2014, 8, 2193–2206. [Google Scholar] [CrossRef] [PubMed]
- Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The Central Nervous System and the Gut Microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef] [PubMed]
- Friedland, R.P.; Chapman, M.R. The role of microbial amyloid in neurodegeneration. PLoS Pathog. 2017, 13, e1006654. [Google Scholar] [CrossRef]
- Willyard, C. How gut bacteria alter the brain. Nature 2021, 590, 22–25. [Google Scholar] [CrossRef]
- Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T. Human Microbiome Project Consortium: Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar]
- Nelson, W.C.; Weinstock, G.M.; Highlander, S.K.; Worley, K.C.; Creasy, H.H.; Wortman, J.R.; Rusch, D.B.; Mitreva, M.; Sodergren, E.; Chinwalla, A.T.; et al. The Human Microbiome Jumpstart Reference Strains Consortium. A catalog of reference genomes from the human microbiome. Science 2010, 328, 994–999. [Google Scholar] [CrossRef]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
- Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef]
- Hugon, P.; Dufour, J.-C.; Colson, P.; Fournier, P.-E.; Sallah, K.; Raoult, D. A comprehensive repertoire of prokaryotic species identified in human beings. Lancet Infect. Dis. 2015, 15, 1211–1219. [Google Scholar] [CrossRef]
- Bilen, M.; Dufour, J.-C.; Lagier, J.-C.; Cadoret, F.; Daoud, Z.; Dubourg, G.; Raoult, D. The contribution of culturomics to the repertoire of isolated human bacterial and archaeal species. Microbiome 2018, 6, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giovannini, M.; Lana, D.; Traini, C.; Vannucchi, M. The Microbiota–Gut–Brain Axis and Alzheimer Disease. From Dysbiosis to Neurodegeneration: Focus on the Central Nervous System Glial Cells. J. Clin. Med. 2021, 10, 2358. [Google Scholar] [CrossRef] [PubMed]
- Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the Human Intestinal Microbial Flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed]
- Begley, M.; Hill, C.; Gahan, C.G.M. Bile Salt Hydrolase Activity in Probiotics. Appl. Environ. Microbiol. 2006, 72, 1729–1738. [Google Scholar] [CrossRef]
- Dicks, L.; Geldenhuys, J.; Mikkelsen, L.; Brandsborg, E.; Marcotte, H. Our gut microbiota: A long walk to homeostasis. Benef. Microbes 2018, 9, 3–20. [Google Scholar] [CrossRef] [PubMed]
- Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef]
- Gevers, D.; Pop, M.; Schloss, P.D.; Huttenhower, C. Bioinformatics for the Human Microbiome Project. PLoS Comput. Biol. 2012, 8, e1002779. [Google Scholar] [CrossRef]
- Yassour, M.; Jason, E.; Hogstrom, L.J.; Arthur, T.D.; Tripathi, S.; Siljander, H.; Selvenius, J.; Oikarinen, S.; Hyöty, H.; Virtanen, S.M.; et al. Strain-Level Analysis of Mother-to-Child Bacterial Transmission during the First Few Months of Life. Cell Host Microbe 2018, 24, 146–154.e4. [Google Scholar] [CrossRef]
- Garud, N.R.; Pollard, K.S. Population Genetics in the Human Microbiome. Trends Genet. 2019, 36, 53–67. [Google Scholar] [CrossRef]
- Browne, H.P.; Forster, S.C.; Anonye, B.O.; Kumar, N.; Neville, B.A.; Stares, M.D.; Goulding, D.; Lawley, T.D. Culturing of ‘‘unculturable’’ human microbiota reveals novel taxa and extensive sporulation. Nature 2016, 533, 543–546. [Google Scholar] [CrossRef]
- Schloissnig, S.; Arumugam, M.; Sunagawa, S.; Mitreva, M.; Tap, J.; Zhu, A.; Waller, A.; Mende, D.R.; Kultima, J.R.; Martin, J.; et al. Genomic variation landscape of the human gut microbiome. Nature 2012, 493, 45–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- I Costea, P.; Coelho, L.P.; Sunagawa, S.; Munch, R.; Huerta-Cepas, J.; Forslund, K.; Hildebrand, F.; Kushugulova, A.; Zeller, G.; Bork, P. Subspecies in the global human gut microbiome. Mol. Syst. Biol. 2017, 13, 960. [Google Scholar] [CrossRef] [PubMed]
- Truong, D.T.; Tett, A.; Pasolli, E.; Huttenhower, C.; Segata, N. Microbial strain-level population structure and genetic diversity from metagenomes. Genome Res. 2017, 27, 626–638. [Google Scholar] [CrossRef] [PubMed]
- Garud, N.R.; Good, B.H.; Hallatschek, O.; Pollard, K.S. Evolutionary dynamics of bacteria in the gut microbiome within and across hosts. PLoS Biol. 2019, 17, e3000102. [Google Scholar] [CrossRef] [PubMed]
- Maurice, C.F.; Haiser, H.J.; Turnbaugh, P.J. Xenobiotics Shape the Physiology and Gene Expression of the Active Human Gut Microbiome. Cell 2013, 152, 39–50. [Google Scholar] [CrossRef] [PubMed]
- Verster, A.J.; Ross, B.D.; Radey, M.C.; Bao, Y.; Goodman, A.L.; Mougous, J.D.; Borenstein, E. The Landscape of Type VI Secretion across Human Gut Microbiomes Reveals Its Role in Community Composition. Cell Host Microbe 2017, 22, 411–419.e4. [Google Scholar] [CrossRef] [PubMed]
- Linz, B.; Balloux, F.; Moodley, Y.; Manica, A.; Liu, H.; Roumagnac, P.; Falush, D.; Stamer, C.; Prugnolle, F.; Van der Merwe, S.; et al. An African origin for the intimate association between humans and Helicobacter pylori. Nature 2007, 445, 915–918. [Google Scholar] [CrossRef]
- Falush, D.; Wirth, T.; Linz, B.; Pritchard, J.K.; Stephens, M.; Kidd, M.; Blaser, M.J.; Graham, D.Y.; Vacher, S.; Perez-Perez, G.I.; et al. Traces of Human Migrations in Helicobacter pylori Populations. Science 2003, 299, 1582–1585. [Google Scholar] [CrossRef]
- Comas, I.; Coscolla, M.; Luo, T.; Borrell, S.; Holt, K.; Kato-Maeda, M.; Parkhill, J.; Malla, B.; Berg, S.; Thwaites, G.; et al. Out-of-Africa migration and Neolithic coexpansion of Mycobacterium tuberculosis with modern humans. Nat. Genet. 2013, 45, 1176–1182. [Google Scholar] [CrossRef]
- Karcher, N.; Pasolli, E.; Asnicar, F.; Huang, K.D.; Tett, A.; Manara, S.; Armanini, F.; Bain, D.; Duncan, S.; Louis, P.; et al. Analysis of 1321 Eubacterium rectale genomes from metagenomes uncovers complex phylogeographic population structure and subspecies functional adaptations. Genome Biol. 2020, 21, 138. [Google Scholar] [CrossRef]
- Tett, A.; Huang, K.D.; Asnicar, F.; Fehlner-Peach, H.; Pasolli, E.; Karscher, N.; Armanini, F.; Manghi, P.; Bonham, K.; Zolfo, M.; et al. The Prevotella copri complex comprises four distinct clades that are underrepresented in Westernised populations. Cell Host Microbe 2019, 26, 666–679.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghalayini, M.; Launay, A.; Bridier-Nahmias, A.; Clermont, O.; Denamur, E.; Lescat, M.; Tenaillon, O. Evolution of a Dominant Natural Isolate of Escherichia coli in the Human Gut over the Course of a Year Suggests a Neutral Evolution with Reduced Effective Population Size. Appl. Environ. Microbiol. 2018, 84, e02377-17. [Google Scholar] [CrossRef] [PubMed]
- Roodgar, M.; Good, B.H.; Garud, N.R.; Martis, S.; Avula, M.; Zhou, W.; Lancaster, S.M.; Lee, H.; Babveyh, A.; Nesamoney, S.; et al. Longitudinal linked read sequencing reveals ecological and evolutionary responses of a human gutmicrobiome during antibiotic treatment. Genome Res. 2022, 31, 1433–1446. [Google Scholar] [CrossRef] [PubMed]
- Mukudai, S.; Sugiyama, Y.; Hisa, Y. Dorsal motor nucleus of the vagus. In Neuroanatomy and Neurophysiology of the Larynx; Springer: Tokyo, Japan, 2016; pp. 97–102. [Google Scholar]
- Berthoud, H.-R.; Neuhuber, W.L. Functional and chemical anatomy of the afferent vagal system. Auton. Neurosci. 2000, 85, 1–17. [Google Scholar] [CrossRef]
- Bonaz, B.; Bazin, T.; Pellissier, S. The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front. Neurosci. 2018, 12, 49. [Google Scholar] [CrossRef] [PubMed]
- Collins, S.M. A role for the gut microbiota in IBS. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 497–505. [Google Scholar] [CrossRef] [PubMed]
- Breit, S.; Kupferberg, A.; Rogler, G.; Hasler, G. Vagus Nerve as Modulator of the Brain–Gut Axis in Psychiatric and Inflammatory Disorders. Front. Psychiatry 2018, 9, 44. [Google Scholar] [CrossRef]
- Rao, M.; Gershon, M.D. Neurogastroenterology: The dynamic cycle of life in the enteric nervous system. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 453–454. [Google Scholar] [CrossRef]
- Brun, P.; Gobbo, S.; Caputi, V.; Spagnol, L.; Schirato, G.; Pasqualin, M.; Levorato, E.; Palù, G.; Giron, M.C.; Castagliuolo, I. Toll like receptor-2 regulates production of glial-derived neurotrophic factors in murine intestinal smooth muscle cells. Mol. Cell. Neurosci. 2015, 68, 24–35. [Google Scholar] [CrossRef]
- Rodrigues, D.M.; Li, A.Y.; Nair, D.G.; Blennerhassett, M.G. Glial cell line-derived neurotrophic factor is a key neurotrophin in the postnatal enteric nervous system. Neurogastroenterol. Motil. 2010, 23, e44–e56. [Google Scholar] [CrossRef]
- Gwak, M.-G.; Chang, S.-Y. Gut-Brain Connection: Microbiome, Gut Barrier, and Environmental Sensors. Immune Netw. 2021, 21, e20. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.; Yang, W.; Li, Y.; Cong, Y. Enteroendocrine Cells: Sensing Gut Microbiota and Regulating Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2019, 26, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Gribble, F.M.; Reimann, F. Enteroendocrine Cells: Chemosensors in the Intestinal Epithelium. Annu. Rev. Physiol. 2016, 78, 277–299. [Google Scholar] [CrossRef]
- De Silva, A.; Bloom, S.R. Gut Hormones and Appetite Control: A Focus on PYY and GLP-1 as Therapeutic Targets in Obesity. Gut Liver 2012, 6, 10–20. [Google Scholar] [CrossRef]
- Richards, P.; Parker, H.E.; Adriaenssens, A.E.; Hodgson, J.M.; Cork, S.C.; Trapp, S.; Gribble, F.M.; Reimann, F. Identification and Characterization of GLP-1 Receptor–Expressing Cells Using a New Transgenic Mouse Model. Diabetes 2014, 63, 1224–1233. [Google Scholar] [CrossRef]
- Sun, L.-J.; Li, J.-N.; Nie, Y.-Z. Gut hormones in microbiota-gut-brain cross-talk. Chin. Med. J. 2020, 133, 826–833. [Google Scholar] [CrossRef] [PubMed]
- Gurda, G.T.; Guo, L.; Lee, S.-H.; Molkentin, J.D.; Williams, J.A. Cholecystokinin Activates Pancreatic Calcineurin-NFAT Signaling In Vitro and In Vivo. Mol. Biol. Cell 2008, 19, 198–206. [Google Scholar] [CrossRef] [PubMed]
- Xu, D.; Yu, B.-P.; Luo, H.-S.; Chen, L.-D. Control of gallbladder contractions by cholecystokinin through cholecystokinin-A receptors on gallbladder interstitial cells of cajal. World J. Gastroenterol. 2008, 14, 2882–2887. [Google Scholar] [CrossRef]
- Howick, K.; Griffin, B.T.; Cryan, J.F.; Schellekens, H. From Belly to Brain: Targeting the Ghrelin Receptor in Appetite and Food Intake Regulation. Int. J. Mol. Sci. 2017, 18, 273. [Google Scholar] [CrossRef]
- Willesen, M.G.; Kristensen, P.; Rømer, J. Co-Localization of Growth Hormone Secretagogue Receptor and NPY mRNA in the Arcuate Nucleus of the Rat. Neuroendocrinology 1999, 70, 306–316. [Google Scholar] [CrossRef]
- Robinson, S.L.; Thiele, T.E. The Role of Neuropeptide Y (NPY) in Alcohol and Drug Abuse Disorders. Int. Rev. Neurobiol. 2017, 136, 177–197. [Google Scholar] [CrossRef] [PubMed]
- Tatemoto, K. Neuropeptide Y: History and overview. Neuropept. Y Relat. Pept. 2004, 162, 1–21. [Google Scholar] [CrossRef]
- Xiao, E.; Xia-Zhang, L.; Vulliémoz, N.R.; Ferin, M.; Wardlaw, S.L. Agouti-Related Protein Stimulates the Hypothalamic-Pituitary-Adrenal (HPA) Axis and Enhances the HPA Response to Interleukin-1 in the Primate. Endocrinology 2003, 144, 1736–1741. [Google Scholar] [CrossRef] [PubMed]
- Burdyga, G.; Varro, A.; Dimaline, R.; Thompson, D.G.; Dockray, G.J. Ghrelin receptors in rat and human nodose ganglia: Putative role in regulating CB-1 and MCH receptor abundance. Am. J. Physiol. Liver Physiol. 2006, 290, G1289–G1297. [Google Scholar] [CrossRef]
- Kawahara, Y.; Kaneko, F.; Yamada, M.; Kishikawa, Y.; Kawahara, H.; Nishi, A. Food reward-sensitive interaction of ghrelin and opioid receptor pathways in mesolimbic dopamine system. Neuropharmacology 2012, 67, 395–402. [Google Scholar] [CrossRef]
- Abizaid, A.; Liu, Z.-W.; Andrews, Z.B.; Shanabrough, M.; Borok, E.; Elsworth, J.D.; Roth, R.H.; Sleeman, M.W.; Picciotto, M.; Tschöp, M.H.; et al. Ghrelin modulates the activity and synaptic input organization of midbrain dopamine neurons while promoting appetite. J. Clin. Investig. 2006, 116, 3229–3239. [Google Scholar] [CrossRef]
- Cani, P.D.; Dewever, C.; Delzenne, N.M. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. Br. J. Nutr. 2004, 92, 521–526. [Google Scholar] [CrossRef]
- Cho, Y.M.; Fujita, Y.; Kieffer, T.J. Glucagon-Like Peptide-1: Glucose Homeostasis and Beyond. Annu. Rev. Physiol. 2014, 76, 535–559. [Google Scholar] [CrossRef]
- Bäckhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The Gut Microbiota as an Environmental Factor That Regulates Fat Storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef]
- Grasset, E.; Puel, A.; Charpentier, J.; Collet, X.; Christensen, J.E.; Tercé, F.; Burcelin, R. A specific gut microbiota dysbiosis of type 2 diabetic mice induces GLP-1 resistance through an enteric NO-dependent and gut-grain axis mechanism. Cell Metab. 2017, 25, 1075–1090.e5. [Google Scholar] [CrossRef]
- Lebrun, L.J.; Lenaerts, K.; Kiers, D.; de Barros, J.-P.P.; Le Guern, N.; Plesnik, J.; Thomas, C.; Bourgeois, T.; Dejong, C.H.; Kox, M.; et al. Enteroendocrine L Cells Sense LPS after Gut Barrier Injury to Enhance GLP-1 Secretion. Cell Rep. 2017, 21, 1160–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holst, J.J. The Physiology of Glucagon-like Peptide 1. Physiol. Rev. 2007, 87, 1409–1439. [Google Scholar] [CrossRef] [PubMed]
- Schalla, M.A.; Stengel, A. Effects of microbiome changes on endocrine ghrelin signaling—A systematic review. Peptides 2020, 133, 170388. [Google Scholar] [CrossRef]
- Fetissov, S.O.; Hamze Sinno, M.; Coëffier, M.; Bole-Feysot, C.; Ducrotté, P.; Hökfelt, T.; Déchelotte, P. Autoantibodies against appetite-regulating peptide hormones and neuropeptides: Putative modulation by gut microflora. Nutrition 2008, 24, 348–359. [Google Scholar] [CrossRef] [PubMed]
- Chervonsky, A.V. Microbiota and Autoimmunity. Cold Spring Harb. Perspect. Biol. 2013, 5, a007294. [Google Scholar] [CrossRef] [PubMed]
- Tennoune, N.; Chan, P.; Breton, J.; Legrand, R.; Chabane, Y.N.; Akkermann, K.; Järv, A.; Ouelaa, W.; Takagi, K.; Ghouzali, I.; et al. Bacterial ClpB heat-shock protein, an antigen-mimetic of the anorexigenic peptide α-MSH, at the origin of eating disorders. Transl. Psychiatry 2014, 4, e458. [Google Scholar] [CrossRef] [PubMed]
- Mabbott, N.A.; Donaldson, D.S.; Ohno, H.; Williams, I.R.; Mahajan, A. Microfold (M) cells: Important immunosurveillance posts in the intestinal epithelium. Mucosal Immunol. 2013, 6, 666–677. [Google Scholar] [CrossRef] [PubMed]
- Seo, K.; Seo, J.; Yeun, J.; Choi, H.; Kim, Y.-I.; Chang, S.-Y. The role of mucosal barriers in human gut health. Arch. Pharmacal Res. 2021, 44, 325–341. [Google Scholar] [CrossRef]
- Szakál, D.N.; Győrffy, H.; Arato, A.; Cseh, A.; Molnár, K.; Papp, M.; Dezsőfi, A.; Veres, G. Mucosal expression of claudins 2, 3 and 4 in proximal and distal part of duodenum in children with coeliac disease. Virchows Arch. 2010, 456, 245–250. [Google Scholar] [CrossRef]
- Davis, M.A.; Ireton, R.C.; Reynolds, A.B. A core function for p120-catenin in cadherin turnover. J. Cell Biol. 2003, 163, 525–534. [Google Scholar] [CrossRef]
- Goldstein, A.M.; Hofstra, R.; Burns, A.J. Building a brain in the gut: Development of the enteric nervous system. Clin. Genet. 2013, 83, 307–316. [Google Scholar] [CrossRef]
- Furness, J.B.; Callaghan, B.P.; Rivera, L.R.; Cho, H.-J. Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease Advances in Experimental Medicine and Biology; Springer: New York, NY, USA, 2014; pp. 39–71. [Google Scholar]
- Joseph, N.M.; He, S.; Quintana, E.; Kim, Y.-G.; Núñez, G.; Morrison, S.J. Enteric glia are multipotent in culture but primarily form glia in the adult rodent gut. J. Clin. Investig. 2011, 121, 3398–3411. [Google Scholar] [CrossRef] [PubMed]
- Laranjeira, C.; Sandgren, K.; Kessaris, N.; Richardson, W.; Potocnik, A.; Berghe, P.V.; Pachnis, V. Glial cells in the mouse enteric nervous system can undergo neurogenesis in response to injury. J. Clin. Investig. 2011, 121, 3412–3424. [Google Scholar] [CrossRef] [PubMed]
- Memic, F.; Knoflach, V.; Morarach, K.; Sadler, R.; Laranjeira, C.; Leffler, J.H.; Sundström, E.; Pachnis, V.; Marklund, U. Transcription and Signaling Regulators in Developing Neuronal Subtypes of Mouse and Human Enteric Nervous System. Gastroenterology 2018, 154, 624–636. [Google Scholar] [CrossRef] [PubMed]
- De Vadder, F.; Grasset, E.; Holm, L.M.; Karsenty, G.; MacPherson, A.J.; Olofsson, L.E.; Bäckhed, F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc. Natl. Acad. Sci. USA 2018, 115, 6458–6463. [Google Scholar] [CrossRef]
- Kulkarni, S.; Micci, M.-A.; Leser, J.; Shin, C.; Tang, S.-C.; Fu, Y.-Y.; Liu, L.; Li, Q.; Saha, M.; Li, C.; et al. Adult enteric nervous system in health is maintained by a dynamic balance between neuronal apoptosis and neurogenesis. Proc. Natl. Acad. Sci. USA 2017, 114, E3709–E3718. [Google Scholar] [CrossRef]
- Sun, X.; Kaufman, P.D. Ki-67: More than a proliferation marker. Chromosoma 2018, 127, 175–186. [Google Scholar] [CrossRef]
- Park, D.; Xiang, A.P.; Mao, F.F.; Zhang, L.; Di, C.-G.; Liu, X.-M.; Shao, Y.; Ma, B.-F.; Lee, J.-H.; Ha, K.-S.; et al. Nestin Is Required for the Proper Self-Renewal of Neural Stem Cells. Stem Cells 2010, 28, 2162–2171. [Google Scholar] [CrossRef]
- Suzuki, S.; Namiki, J.; Shibata, S.; Mastuzaki, Y.; Okano, H. The Neural Stem/Progenitor Cell Marker Nestin Is Expressed in Proliferative Endothelial Cells, but Not in Mature Vasculature. J. Histochem. Cytochem. 2010, 58, 721–730. [Google Scholar] [CrossRef]
- Vicentini, F.A.; Keenan, C.M.; Wallace, L.E.; Woods, C.; Cavin, J.-B.; Flockton, A.R.; Macklin, W.B.; Belkind-Gerson, J.; Hirota, S.A.; Sharkey, K.A. Intestinal microbiota shapes gut physiology and regulates enteric neurons and glia. Microbiome 2021, 9, 210. [Google Scholar] [CrossRef]
- Kruse, A.C.; Kobilka, B.K.; Gautam, D.; Sexton, P.; Christopoulos, A.; Wess, J. Muscarinic acetylcholine receptors: Novel opportunities for drug development. Nat. Rev. Drug Discov. 2014, 13, 549–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jie, W.U. Understanding of nicotinic acetylcholine receptors. Acta Pharmacol. Sin. 2009, 6, 653–655. [Google Scholar]
- Wolosker, H.; Dumin, E.; Balan, L.; Foltyn, V.N. d-Amino acids in the brain: D-serine in neurotransmission and neurodegeneration. FEBS J. 2008, 275, 3514–3526. [Google Scholar] [CrossRef] [PubMed]
- Ohgi, Y.; Futamura, T.; Kikuchi, T.; Hashimoto, K. Effects of antidepressants on alternations in serum cytokines and depressive-like behavior in mice after lipopolysaccharide administration. Pharmacol. Biochem. Behav. 2012, 103, 853–859. [Google Scholar] [CrossRef]
- Qiu, W.; Wu, M.; Liu, S.; Chen, B.; Pan, C.; Yang, M.; Wang, K.J. Suppressive immunoregulatory effects of three antidepressants via inhibiton of the nuclear factor-κB activation assessed using primary macrophages of carp (Cyprinus carpio). Toxicol. Appl. Pharmacol. 2017, 322, 1–8. [Google Scholar] [CrossRef]
- Singh, V.; Roth, S.; Llovera, G.; Sadler, R.; Garzetti, D.; Stecher, B.; Dichgans, M.; Liesz, A. Microbiota Dysbiosis Controls the Neuroinflammatory Response after Stroke. J. Neurosci. 2016, 36, 7428–7440. [Google Scholar] [CrossRef]
- Stanley, D.; Mason, L.J.; E Mackin, K.; Srikhanta, Y.N.; Lyras, D.; Prakash, M.D.; Nurgali, K.; Venegas, A.; Hill, M.D.; Moore, R.J.; et al. Translocation and dissemination of commensal bacteria in post-stroke infection. Nat. Med. 2016, 22, 1277–1284. [Google Scholar] [CrossRef]
- Houlden, A.; Goldrick, M.; Brough, D.; Vizi, E.S.; Lénárt, N.; Martinecz, B.; Roberts, I.S.; Denes, A. Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production. Brain Behav. Immun. 2016, 57, 10–20. [Google Scholar] [CrossRef]
- Zhou, Y.; Danbolt, N.C. Glutamate as a neurotransmitter in the healthy brain. J. Neural Transm. 2014, 121, 799–817. [Google Scholar] [CrossRef]
- Vyleta, N.P.; Smith, S.M. Spontaneous Glutamate Release Is Independent of Calcium Influx and Tonically Activated by the Calcium-Sensing Receptor. J. Neurosci. 2011, 31, 4593–4606. [Google Scholar] [CrossRef]
- Lau, A.; Tymianski, M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 2010, 460, 525–542. [Google Scholar] [CrossRef] [PubMed]
- Esposito, Z.; Belli, L.; Toniolo, S.; Sancesario, G.; Bianconi, C.; Martorana, A. Amyloid β, glutamate, excitotoxicity in Alzheimer’s disease: Are we on the right track? CNS Neurosci. Ther. 2013, 19, 549–555. [Google Scholar] [CrossRef] [PubMed]
- Bertoldi, M.; Cellini, B.; Paiardini, A.; Di Salvo, M.; Borrivoltattorni, C. Treponema denticola cystalysin exhibits significant alanine racemase activity accompanied by transamination: Mechanistic implications. Biochem. J. 2003, 371, 473–483. [Google Scholar] [CrossRef] [PubMed]
- Nakayama, Y.; Hashimoto, K.-I.; Sawada, Y.; Sokabe, M.; Kawasaki, H.; Martinac, B. Corynebacterium glutamicum mechanosensitive channels: Towards unpuzzling “glutamate efflux” for amino acid production. Biophys. Rev. 2018, 10, 1359–1369. [Google Scholar] [CrossRef] [PubMed]
- Malathi, K.C.; Wachi, M.; Nagai, K. Isolation of the murI gene from Brevibacterium lactofermentum ATCC 13869 encoding D-glutamate racemase. FEMS Microbiol. Lett. 1999, 175, 193–196. [Google Scholar] [CrossRef]
- Tsai, M.-F.; Miller, C. Substrate selectivity in arginine-dependent acid resistance in enteric bacteria. Proc. Natl. Acad. Sci. USA 2013, 110, 5893–5897. [Google Scholar] [CrossRef]
- Kawase, T.; Nagasawa, M.; Ikeda, H.; Yasuo, S.; Koga, Y.; Furuse, M. Gut microbiota of mice putatively modifies amino acid metabolism in the host brain. Br. J. Nutr. 2017, 117, 775–783. [Google Scholar] [CrossRef]
- Chang, C.-H.; Lin, C.-H.; Lane, H.-Y. d-glutamate and Gut Microbiota in Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 2676. [Google Scholar] [CrossRef]
- Olsen, R.W.; DeLorey, T.M. Glycine receptor physiology and pharmacology. In Basic Neurochemistry: Molecular, Cellular and Medical Aspects, 6th ed.; American Society for Neurochemistry: Philadelphia, PA, USA, 1999; pp. 335–346. [Google Scholar]
- Araque, A.; Parpura, V.; Sanzgiri, R.P.; Haydon, P.G. Tripartite synapses: Glia, the unacknowledged partner. Trends Neurosci. 1999, 22, 208–215. [Google Scholar] [CrossRef]
- Yoon, B.-E.; Lee, C.J. GABA as a rising gliotransmitter. Front. Neural Circuits 2014, 8, 141. [Google Scholar] [CrossRef]
- Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2019, 4, 396–403. [Google Scholar] [CrossRef] [PubMed]
- Dover, S.; Halpern, Y.S. Utilization of γ-Aminobutyric Acid as the Sole Carbon and Nitrogen Source by Escherichia coli K-12 Mutants. J. Bacteriol. 1972, 109, 835–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Quillin, S.J.; Tran, P.; Prindle, A. Potential Roles for Gamma-Aminobutyric Acid Signaling in Bacterial Communities. Bioelectricity 2021, 3, 120–125. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Trombley, P.Q.; Pol, A.N.V.D. GABA receptors precede glutamate receptors in hypothalamic development; differential regulation by astrocytes. J. Neurophysiol. 1995, 74, 1473–1484. [Google Scholar] [CrossRef]
- Wu, C.; Sun, D. GABA receptors in brain development, function, and injury. Metab. Brain Dis. 2015, 30, 367–379. [Google Scholar] [CrossRef]
- Couve, A.; Moss, S.J.; Pangalos, M.N. GABAB Receptors: A New Paradigm in G Protein Signaling. Mol. Cell. Neurosci. 2000, 16, 296–312. [Google Scholar] [CrossRef]
- Misgeld, U.; Bijak, M.; Jarolimek, W. A physiological role for GABAB receptors and the effects of baclofen in the mammalian central nervous system. Prog. Neurobiol. 1995, 46, 423–462. [Google Scholar] [CrossRef]
- Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef]
- Janik, R.; Thomason, L.A.; Stanisz, A.M.; Forsythe, P.; Bienenstock, J.; Stanisz, G.J. Magnetic resonance spectroscopy reveals oral Lactobacillus promotion of increases in brain GABA, N-acetyl aspartate and glutamate. NeuroImage 2016, 125, 988–995. [Google Scholar] [CrossRef]
- Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
- Takanaga, H.; Ohtsuki, S.; Hosoya, K.-I.; Terasaki, T. GAT2/BGT-1 as a system responsible for the transport of γ-aminobutyric acid at the mouse blood-brain barrier. J. Cereb. Blood Flow Metabol. 2001, 21, 1232–1239. [Google Scholar] [CrossRef]
- Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J.; et al. The Orphan G Protein-coupled Receptors GPR41 and GPR43 Are Activated by Propionate and Other Short Chain Carboxylic Acids. J. Biol. Chem. 2003, 278, 11312–11319. [Google Scholar] [CrossRef] [Green Version]
- Olson, C.A.; Vuong, H.E.; Yano, J.M.; Liang, Q.Y.; Nusbaum, D.J.; Hsiao, E.Y. The Gut Microbiota Mediates the Anti-Seizure Effects of the Ketogenic Diet. Cell 2018, 173, 1728–1741.e13. [Google Scholar] [CrossRef]
- Bowery, N.G.; Smart, T.G. GABA and glycine as neurotransmitters: A brief history. Br. J. Pharmacol. 2006, 147 (Suppl. 1), S109–S119. [Google Scholar] [CrossRef]
- De Bartolomeis, A.; Manchia, M.; Marmo, F.; Vellucci, L.; Iasevoli, F.; Barone, A. Glycine Signaling in the Framework of Dopamine-Glutamate Interaction and Postsynaptic Density. Implications for Treatment-Resistant Schizophrenia. Front. Psychiatry 2020, 11, 369. [Google Scholar] [CrossRef]
- López-Corcuera, B.; Geerlings, A.; Aragón, C. Glycine neurotransmitter transporters: An update. Mol. Membr. Biol. 2001, 18, 13–20. [Google Scholar] [CrossRef] [PubMed]
- Ko, J.H.; Strafella, A.P. Dopaminergic neurotransmission in the human brain: New lessons from perturbation and imaging. Neuroscientist 2012, 18, 149–168. [Google Scholar] [CrossRef] [PubMed]
- Olguín, H.J.; Guzmán, D.C.; García, E.H.; Mejía, G.B. The Role of Dopamine and Its Dysfunction as a Consequence of Oxidative Stress. Oxidative Med. Cell. Longev. 2015, 2016, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Asano, Y.; Hiramoto, T.; Nishino, R.; Aiba, Y.; Kimura, T.; Yoshihara, K.; Koga, Y.; Sudo, N. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am. J. Physiol. Liver Physiol. 2012, 303, G1288–G1295. [Google Scholar] [CrossRef] [PubMed]
- Rekdal, V.M.; Bess, E.N.; Bisanz, J.E.; Turnbaugh, P.J.; Balskus, E.P. Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science 2019, 364, 6323. [Google Scholar] [CrossRef]
- Borodovitsyna, O.; Flamini, M.; Chandler, D. Noradrenergic Modulation of Cognition in Health and Disease. Neural Plast. 2017, 2017, 1–14. [Google Scholar] [CrossRef] [PubMed]
- O’Donnell, J.; Zeppenfeld, D.; McConnell, E.; Pena, S.; Nedergaard, M. Norepinephrine: A Neuromodulator That Boosts the Function of Multiple Cell Types to Optimize CNS Performance. Neurochem. Res. 2012, 37, 2496–2512. [Google Scholar] [CrossRef] [Green Version]
- Clarke, M.B.; Hughes, D.T.; Zhu, C.; Boedeker, E.C.; Sperandio, V. The QseC sensor kinase: A bacterial adrenergic receptor. Proc. Natl. Acad. Sci. USA 2006, 103, 10420–10425. [Google Scholar] [CrossRef] [PubMed]
- Hughes, D.T.; Sperandio, V. Inter-kingdom signalling: Communication between bacteria and their hosts. Nat. Rev. Genet. 2008, 6, 111–120. [Google Scholar] [CrossRef] [PubMed]
- O’Donnell, P.M.; Aviles, H.; Lyte, M.; Sonnenfeld, G. Enhancement of In Vitro Growth of Pathogenic Bacteria by Norepinephrine: Importance of Inoculum Density and Role of Transferrin. Appl. Environ. Microbiol. 2006, 72, 5097–5099. [Google Scholar] [CrossRef]
- Tsavkelova, E.A.; Botvinko, I.V.; Kudrin, V.S.; Oleskin, A.V. Detection of neurotransmitter amines in microorganisms with the use of high-performance liquid chromatography. Dokl. Biochem. 2000, 372, 115–117. [Google Scholar]
- Sperandio, V.; Torres, A.G.; Jarvis, B.; Nataro, J.P.; Kaper, J.B. Bacteria–host communication: The language of hormones. Proc. Natl. Acad. Sci. USA 2003, 100, 8951–8956. [Google Scholar] [CrossRef]
- Stefano, G.B.; Pilonis, N.; Ptacek, R.; Raboch, J.; Vnukova, M.; Kream, R.M. Gut, Microbiome, and Brain Regulatory Axis: Relevance to Neurodegenerative and Psychiatric Disorders. Cell. Mol. Neurobiol. 2018, 38, 1197–1206. [Google Scholar] [CrossRef]
- Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef]
- Kelly, J.R.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbiota axis: Challenges for translation in psychiatry. Ann. Epidemiol. 2016, 26, 366–372. [Google Scholar] [CrossRef]
- Israelyan, N.; Margolis, K.G. Reprint of: Serotonin as a link between the gut-brain-microbiome axis in autism spectrum disorders. Pharmacol. Res. 2019, 140, 115–120. [Google Scholar] [CrossRef] [PubMed]
- Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous Bacteria from the Gut Microbiota Regulate Host Serotonin Biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed]
- Bellono, N.W.; Bayrer, J.R.; Leitch, D.B.; Castro, J.; Zhang, C.; O’Donnell, T.A.; Brierley, S.M.; Ingraham, H.A.; Julius, D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell 2017, 170, 185–198.e16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef]
- Sgritta, M.; Dooling, S.W.; Buffington, S.A.; Momin, E.N.; Francis, M.B.; Britton, R.A.; Costa-Mattioli, M. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019, 101, 246–259.e6. [Google Scholar] [CrossRef]
- Liu, M.-T.; Kuan, Y.-H.; Wang, J.; Hen, R.; Gershon, M.D. 5-HT4 Receptor-Mediated Neuroprotection and Neurogenesis in the Enteric Nervous System of Adult Mice. J. Neurosci. 2009, 29, 9683–9699. [Google Scholar] [CrossRef]
- McVey Neufeld, K.A.; Perez-Burgos, A.; Mao, Y.K.; Bienenstock, J.; Kunze, W.A. The gut microbiome restores intrinsic and extrinsic nerve function in germ-free mice accompanied by changes in calbindin. Neurogastroenterol. Motil. 2015, 27, 627–636. [Google Scholar] [CrossRef]
- Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F., III; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 1395–1403. [Google Scholar] [CrossRef]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
- El-Salhy, M.; Danielsson, Å.; Stenling, R.; Grimelius, L. Colonic endocrine cells in inflammatory bowel disease. J. Intern. Med. 1997, 242, 413–419. [Google Scholar] [CrossRef]
- Rindi, G.; Leiter, A.B.; Kopin, A.S.; Bordi, C.; Solcia, E. The “normal” endocrine cell of the gut: Changing concepts and new evidences. Ann. N. Y. Acad. Sci. 2004, 1014, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Strid, H.; Simrén, M.; Lasson, A.; Isaksson, S.; Stridsberg, M.; Öhman, L. Fecal chromogranins and secretogranins are increased in patients with ulcerative colitis but are not associated with disease activity. J. Crohn’s Colitis 2013, 7, e615–e622. [Google Scholar] [CrossRef] [PubMed]
- Zhernakova, A.; Kurilshikov, A.; Bonder, M.J.; Tigchelaar, E.F.; Schirmer, M.; Vatanen, T.; Mujagic, Z.; Vila, A.V.; Falony, G.; Vieira-Silva, S.; et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 2016, 352, 565–569. [Google Scholar] [CrossRef] [Green Version]
- Koch, T.R.; Roddy, D.R.; Go, V.L. Abnormalities of fasting serum concentrations of peptide YY in the idiopathic inflammatory bowel diseases. Am. J. Gastroenterol. 1987, 82, 321–326. [Google Scholar] [PubMed]
- Ghia, J.-E.; Li, N.; Wang, H.; Collins, M.; Deng, Y.; El-Sharkawy, R.T.; Côté, F.; Mallet, J.; Khan, W.I. Serotonin Has a Key Role in Pathogenesis of Experimental Colitis. Gastroenterology 2009, 137, 1649–1660. [Google Scholar] [CrossRef]
- Stavely, R.; Fraser, S.; Sharma, M.S.; Rahman, A.A.; Stojanovska, V.; Sakkal, S.; Apostolopoulos, V.; Bertrand, P.; Nurgali, K. The Onset and Progression of Chronic Colitis Parallels Increased Mucosal Serotonin Release via Enterochromaffin Cell Hyperplasia and Downregulation of the Serotonin Reuptake Transporter. Inflamm. Bowel Dis. 2018, 24, 1021–1034. [Google Scholar] [CrossRef]
- Guseva, D.; Holst, K.; Kaune, B.; Meier, M.; Keubler, L.; Glage, S.; Buettner, M.; Bleich, A.; Pabst, O.; Bachmann, O.; et al. Serotonin 5-HT7 Receptor Is Critically Involved in Acute and Chronic Inflammation of the Gastrointestinal Tract. Inflamm. Bowel Dis. 2014, 20, 1516–1529. [Google Scholar] [CrossRef]
- Kim, J.J.; Bridle, B.W.; Ghia, J.-E.; Wang, H.; Syed, S.N.; Manocha, M.M.; Rengasamy, P.; Shajib, M.S.; Wan, Y.; Hedlund, P.B.; et al. Targeted Inhibition of Serotonin Type 7 (5-HT7) Receptor Function Modulates Immune Responses and Reduces the Severity of Intestinal Inflammation. J. Immunol. 2013, 190, 4795–4804. [Google Scholar] [CrossRef]
- Matsumoto, M.; Kibe, R.; Ooga, T.; Aiba, Y.; Sawaki, E.; Koga, Y.; Benno, Y. Cerebral Low-Molecular Metabolites Influenced by Intestinal Microbiota: A Pilot Study. Front. Syst. Neurosci. 2013, 7, 9. [Google Scholar] [CrossRef]
- Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef]
- Hata, T.; Asano, Y.; Yoshihara, K.; Kimura-Todani, T.; Miyata, N.; Zhang, X.-T.; Takakura, S.; Aiba, Y.; Koga, Y.; Sudo, N. Regulation of gut luminal serotonin by commensal microbiota in mice. PLoS ONE 2017, 12, e0180745. [Google Scholar] [CrossRef]
- Hoang, T.K.; He, B.; Wang, T.; Tran, D.Q.; Rhoads, J.M.; Liu, Y. Protective effect of Lactobacillus reuteri DSM 17938 against experimental necrotizing enterocolitis is mediated by Toll-like receptor 2. Am. J. Physiol. Liver Physiol. 2018, 315, G231–G240. [Google Scholar] [CrossRef] [PubMed]
- Margolis, K.G.; Stevanovic, K.; Li, Z.; Yang, Q.M.; Oravecz, T.; Zambrowicz, B.; Jhaver, K.G.; Diacou, A.; Gershon, M.D. Pharmacological reduction of mucosal but not neuronal serotonin opposes inflammation in mouse intestine. Gut 2013, 63, 928–937. [Google Scholar] [CrossRef]
- Bianco, F.; Bonora, E.; Natarajan, D.; Vargiolu, M.; Thapar, N.; Torresan, F.; Giancola, F.; Boschetti, E.; Volta, U.; Bazzoli, F.; et al. Prucalopride exerts neuroprotection in human enteric neurons. Am. J. Physiol. Liver Physiol. 2016, 310, G768–G775. [Google Scholar] [CrossRef] [PubMed]
- Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef] [PubMed]
- Hubbard, T.D.; Murray, I.A.; Perdew, G.H. Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation. Drug Metab. Dispos. 2015, 43, 1522–1535. [Google Scholar] [CrossRef]
- Cervantes-Barragan, L.; Chai, J.N.; Tianero, M.D.; Di Luccia, B.; Ahern, P.P.; Merriman, J.; Cortez, V.S.; . Caparon, M.G.; Donia, M.S.; Gilfillan, S.; et al. Lactobacillus reuteri induces gut intraepithelial CD4+ CD8αα+ T cells. Science 2017, 357, 806–810. [Google Scholar] [CrossRef] [PubMed]
- Satyanarayana, M. Serotonin helps gut microbes thrive. The antidepressant Prozac may interrupt this gut-microbe communication. Chem. Eng. News. 2019. ISSN 0009-2347. Available online: https://cen.acs.org/biological-chemistry/microbiome/Serotonin-helps-gut-microbes-thrive/97/i35 (accessed on 19 April 2022).
- Chitnis, T.; Weiner, H.L. CNS inflammation and neurodegeneration. J. Clin. Investig. 2017, 127, 3577–3587. [Google Scholar] [CrossRef]
- Fung, T.C.; Vuong, H.E.; Luna, C.D.G.; Pronovost, G.N.; Aleksandrova, A.A.; Riley, N.G.; Vavilina, A.; McGinn, J.; Rendon, T.; Forrest, L.R.; et al. Intestinal serotonin and fluoxetine exposure modulate bacterial colonization in the gut. Nat. Microbiol. 2019, 4, 2064–2073. [Google Scholar] [CrossRef]
- Castro-Nallar, E.; Bendall, M.; Perez-Losada, M.; Sabuncyan, S.; Severance, E.G.; Dickerson, F.B.; Schroeder, J.R.; Yolken, R.H.; Crandall, K.A. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ 2015, 3, e1140. [Google Scholar] [CrossRef]
- Lyte, M.; Varcoe, J.J.; Bailey, M.T. Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiol. Behav. 1998, 65, 63–68. [Google Scholar] [CrossRef]
- Passani, M.B.; Panula, P.; Lin, J.S. Histamine in the brain. Front. Syst. Neurosci. 2014, 8, 64. [Google Scholar] [CrossRef]
- Chen, H.; Nwe, P.-K.; Yang, Y.; Rosen, C.E.; Bielecka, A.A.; Kuchroo, M.; Cline, G.W.; Kruse, A.C.; Ring, A.M.; Crawford, J.M.; et al. A Forward Chemical Genetic Screen Reveals Gut Microbiota Metabolites That Modulate Host Physiology. Cell 2019, 177, 1217–1231.e18. [Google Scholar] [CrossRef] [PubMed]
- Colosimo, D.A.; Kohn, J.A.; Luo, P.M.; Piscotta, F.J.; Han, S.M.; Pickard, A.J.; Rao, A.; Cross, J.R.; Cohen, L.J.; Brady, S.F. Mapping Interactions of Microbial Metabolites with Human G-Protein-Coupled Receptors. Cell Host Microbe 2019, 26, 273–282.e7. [Google Scholar] [CrossRef] [PubMed]
- Hsu, B.B.; Gibson, T.E.; Yeliseyev, V.; Liu, Q.; Lyon, L.; Bry, L.; Silver, P.A.; Gerber, G.K. Dynamic Modulation of the Gut Microbiota and Metabolome by Bacteriophages in a Mouse Model. Cell Host Microbe 2019, 25, 803–814.e5. [Google Scholar] [CrossRef] [PubMed]
- Cushing, K.; Alvarado, D.; A Ciorba, M. Butyrate and Mucosal Inflammation: New Scientific Evidence Supports Clinical Observation. Clin. Transl. Gastroenterol. 2015, 6, e108. [Google Scholar] [CrossRef] [PubMed]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Nohr, M.K.; Pedersen, M.H.; Gille, A.; Egerod, K.L.; Engelstoft, M.S.; Husted, A.S.; Sichlau, R.M.; Grunddal, K.V.; Poulsen, S.S.; Han, S.; et al. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for shortchain fatty acids in enteroendocrine cells vs. FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology 2013, 154, 3552–3564. [Google Scholar] [CrossRef]
- Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef]
- Hase, K.; Eckmann, L.; Leopard, J.D.; Varki, N.; Kagnoff, M.F. Cell differentiation is a key determinant of cathelicidin LL-37/human cationic antimicrobial protein 18 expression by human colon epithelium. Infect. Immunol. 2002, 70, 953–963. [Google Scholar] [CrossRef]
- Treem, W.R.; Ahsan, N.; Shoup, M.; Hyams, J.S. Fecal Short-Chain Fatty Acids in Children with Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 1994, 18, 159–164. [Google Scholar] [CrossRef] [PubMed]
- Frank, D.N.; St Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [PubMed]
- Kekuda, R.; Manoharan, P.; Baseler, W.; Sundaram, U. Monocarboxylate 4 Mediated Butyrate Transport in a Rat Intestinal Epithelial Cell Line. Am. J. Dig. Dis. 2013, 58, 660–667. [Google Scholar] [CrossRef] [PubMed]
- Tamai, I.; Takanaga, H.; Ogihara, T.; Higashida, H.; Maeda, H.; Sai, Y.; Tsuji, A. Participation of a Proton-Cotransporter, MCT1, in the Intestinal Transport of Monocarboxylic Acids. Biochem. Biophys. Res. Commun. 1995, 214, 482–489. [Google Scholar] [CrossRef] [PubMed]
- Fleischer, J.; Bumbalo, R.; Bautze, V.; Strotmann, J.; Breer, H. Expression of odorant receptor Olfr78 in enteroendocrine cells of the colon. Cell Tissue Res. 2015, 361, 697–710. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.-B.; Wang, P.-Y.; Wang, X.; Wan, Y.-L.; Liu, Y.-C. Butyrate Enhances Intestinal Epithelial Barrier Function via Up-Regulation of Tight Junction Protein Claudin-1 Transcription. Am. J. Dig. Dis. 2012, 57, 3126–3135. [Google Scholar] [CrossRef]
- De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef]
- Liu, J.; Fu, Y.; Zhang, H.; Wang, J.; Zhu, J.; Wang, Y.; Guo, Y.; Wang, G.; Xu, T.; Chu, M.; et al. The hepatoprotective effect of the probiotic Clostridium butyricum against carbon tetrachloride-induced acute liver damage in mice. Food Funct. 2017, 8, 4042–4052. [Google Scholar] [CrossRef]
- Jin, U.-H.; Cheng, Y.; Park, H.; Davidson, L.A.; Callaway, E.S.; Chapkin, R.S.; Jayaraman, A.; Asante, A.; Allred, C.; Weaver, E.A.; et al. Short Chain Fatty Acids Enhance Aryl Hydrocarbon (Ah) Responsiveness in Mouse Colonocytes and Caco-2 Human Colon Cancer Cells. Sci. Rep. 2017, 7, 10163. [Google Scholar] [CrossRef]
- Zhang, Q.; Yang, F.; Li, X.; Zhang, H.-Y.; Chu, X.-G.; Zhang, H.; Wang, L.-W.; Gong, Z.-J. Trichostatin A protects against intestinal injury in rats with acute liver failure. J. Surg. Res. 2016, 205, 1–10. [Google Scholar] [CrossRef]
- Xu, W.S.; Parmigiani, R.B.; Marks, P.A. Histone deacetylase inhibitors: Molecular mechanisms of action. Oncogene 2007, 26, 5541–5552. [Google Scholar] [CrossRef] [PubMed]
- Roger, T.; Lugrin, J.; Le Roy, D.; Goy, G.; Mombelli, M.; Koessler, T.; Ding, X.C.; Chanson, A.L.; Reymond, M.K.; Miconnet, I.; et al. Histone deacetylase inhibitors impair innate immune responses to Toll-like receptor agonists and to infection. Blood 2011, 117, 1205–1217. [Google Scholar] [CrossRef] [PubMed]
- Yanhai, F.; Yu, W.; Pei, W.; Huang, Y.; Wang, F. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell Physiol. Biochem. 2018, 49, 190–205. [Google Scholar]
- Priyadarshini, M.; Kotlo, K.U.; Dudeja, P.K.; Layden, B.T. Role of Short Chain Fatty Acid Receptors in Intestinal Physiology and Pathophysiology. Compr. Physiol. 2011, 8, 1091–1115. [Google Scholar] [CrossRef]
- Esaxena, M.; Eyeretssian, G. NOD-Like Receptors: Master Regulators of Inflammation and Cancer. Front. Immunol. 2014, 5, 327. [Google Scholar] [CrossRef]
- Galland, L. The Gut Microbiome and the Brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef]
- Collison, L.W.; Workman, C.J.; Kuo, T.T.; Boyd, K.; Wang, Y.; Vignali, K.M.; Cross, R.; Sehy, D.; Blumberg, R.S.; Vignali, D.A.A. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 2007, 450, 566–569. [Google Scholar] [CrossRef]
- Sitkin, S.; Vakhitov, T.; Pokrotnieks, J. Oral butyrate modulates the gut microbiota in patients with inflammatory bowel disease, most likely by reversing proinflammatory metabolic reprogramming of colonocytes. Neurogastroenterol. Motil. 2020, 33, e14038. [Google Scholar] [CrossRef]
- Sun, M.-F.; Shen, Y.-Q. Dysbiosis of gut microbiota and microbial metabolites in Parkinson’s Disease. Ageing Res. Rev. 2018, 45, 53–61. [Google Scholar] [CrossRef]
- Erny, D.; Hrabě de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
- Schroeder, F.A.; Lin, C.L.; Crusio, W.E.; Akbarian, S. Antidepressant-Like Effects of the Histone Deacetylase Inhibitor, Sodium Butyrate, in the Mouse. Biol. Psychiatry 2007, 62, 55–64. [Google Scholar] [CrossRef] [PubMed]
- Mayer, E.A.; Knight, R.; Mazmanian, S.K.; Cryan, J.F.; Tillisch, K. Gut Microbes and the Brain: Paradigm Shift in Neuroscience. J. Neurosci. 2014, 34, 15490–15496. [Google Scholar] [CrossRef] [PubMed]
- Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
- Bercik, P.; Denou, E.; Collins, J.; Jackson, W.; Lu, J.; Jury, J.; Deng, Y.; Blennerhassett, P.; Macri, J.; McCoy, K.D.; et al. The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice. Gastroenterology 2011, 141, 599–609.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef]
- Nankova, B.B.; Agarwal, R.; Macfabe, D.F.; La Gamma, E.F. Enteric Bacterial Metabolites Propionic and Butyric Acid Modulate Gene Expression, Including CREB-Dependent Catecholaminergic Neurotransmission, in PC12 Cells—Possible Relevance to Autism Spectrum Disorders. PLoS ONE 2014, 9, e103740. [Google Scholar] [CrossRef]
- Shah, P.; Nankova, B.B.; Parab, S.; La Gamma, E.F. Short chain fatty acids induce TH gene expression via ERK-dependent phosphorylation of CREB protein. Brain Res. 2006, 1107, 13–23. [Google Scholar] [CrossRef]
- DeCastro, M.; Nankova, B.B.; Shah, P.; Patel, P.; Mally, P.V.; Mishra, R.; La Gamma, E.F. Short chain fatty acids regulate tyrosine hydroxylase gene expression through a cAMP-dependent signaling pathway. Mol. Brain Res. 2005, 142, 28–38. [Google Scholar] [CrossRef]
- Mally, P.; Mishra, R.; Gandhi, S.; Decastro, M.H.; Nankova, B.B.; Lagamma, E.F. Stereospecific Regulation of Tyrosine Hydroxylase and Proenkephalin Genes by Short-Chain Fatty Acids in Rat PC12 Cells. Pediatr. Res. 2004, 55, 847–854. [Google Scholar] [CrossRef]
- Joseph, J.; Depp, C.; Shih, P.B.; Cadenhead, K.S.; Schmid-Schönbein, G. Modified mediterranean diet for enrichment of short chain fatty acids: Potential adjunctive therapeutic to target immune and metabolic dysfunction in schizophrenia? Front. Neurosci. 2017, 11, 155. [Google Scholar] [CrossRef]
- MedlinePlus. Available online: https://medlineplus.gov/genetics/condition/dopamine-beta-hydroxylase-deficiency/ (accessed on 24 March 2022).
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
- Hoyles, L.; Fernández-Real, J.-M.; Federici, M.; Serino, M.; Abbott, J.; Charpentier, J.; Heymes, C.; Luque, J.L.; Anthony, E.; Barton, R.H.; et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat. Med. 2018, 24, 1070–1080. [Google Scholar] [CrossRef] [PubMed]
- Nohr, M.K.; Egerod, K.L.; Christiansen, S.H.; Gille, A.; Offermanns, S.; Schwartz, T.W.; Møller, M. Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience 2015, 290, 126–137. [Google Scholar] [CrossRef] [PubMed]
- Won, Y.J.; Lu, V.B.; Puhl, H.L., 3rd; Ikeda, S.R. β-Hydroxybutyrate modulates N-type calcium channels in rat sympathetic neurons by acting as an agonist for the G-protein-coupled receptor FFA3. J. Neurosci. 2013, 33, 19314–19325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Specian, R.D.; Neutra, M.R. Mechanism of rapid mucus secretion in goblet cells stimulated by acetylcholine. J. Cell Biol. 1980, 85, 626–640. [Google Scholar] [CrossRef] [PubMed]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; Van Der Veeken, J.; DeRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
- Erny, D.; De Angelis, A.L.H.; Prinz, M. Communicating systems in the body: How microbiota and microglia cooperate. Immunology 2016, 150, 7–15. [Google Scholar] [CrossRef]
- Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.M.; Reimann, F.; Blottiere, H.M. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74. [Google Scholar] [CrossRef]
- Mertens, K.L.; Kalsbeek, A.; Soeters, M.R.; Eggink, H.M. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front. Neurosci. 2017, 11, 617. [Google Scholar] [CrossRef]
- Scheperjans, F.; Aho, V.; Pereira, P.A.B.; Koskinen, K.; Paulin, L.; Pekkonen, E.; Haapaniemi, E.; Kaakkola, S.; Eerola-Rautio, J.; Pohja, M.; et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov. Disord. 2015, 30, 350–358. [Google Scholar] [CrossRef]
- Unger, M.M.; Spiegel, J.; Dillmann, K.-U.; Grundmann, D.; Philippeit, H.; Bürmann, J.; Faßbender, K.; Schwiertz, A.; Schäfer, K.H. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef] [PubMed]
- Wu, W.; Sun, M.; Chen, F.; Cao, A.T.; Liu, H.; Zhao, Y.; Huang, X.; Xiao, Y.; Yao, S.; Zhao, Q.; et al. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol. 2016, 10, 946–956. [Google Scholar] [CrossRef] [PubMed]
- Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.K.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef] [Green Version]
- Egerod, K.L.; Petersen, N.; Timshel, P.N.; Rekling, J.C.; Wang, Y.; Liu, Q.; Schwartz, T.W.; Gautron, L. Profiling of G protein-coupled receptors in vagal afferents reveals novel gut-to-brain sensing mechanisms. Mol. Metab. 2018, 12, 62–75. [Google Scholar] [CrossRef]
- Ganesh, B.; Nelson, J.W.; Eskew, J.R.; Ganesan, A.; Ajami, N.J.; Petrosino, J.F.; Bryan, R.M., Jr.; Durgan, D.J. Prebiotics, Probiotics, and Acetate Supplementation Prevent Hypertension in a Model of Obstructive Sleep Apnea. Hypertension 2018, 72, 1141–1150. [Google Scholar] [CrossRef] [PubMed]
- Durgan, D.J.; Lee, J.; McCullough, L.D.; Bryan, R.M., Jr. Examining the Role of the Microbiota-Gut-Brain Axis in Stroke. Stroke 2019, 50, 2270–2277. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dicks, L.M.T. Gut Bacteria and Neurotransmitters. Microorganisms 2022, 10, 1838. https://doi.org/10.3390/microorganisms10091838
Dicks LMT. Gut Bacteria and Neurotransmitters. Microorganisms. 2022; 10(9):1838. https://doi.org/10.3390/microorganisms10091838
Chicago/Turabian StyleDicks, Leon M. T. 2022. "Gut Bacteria and Neurotransmitters" Microorganisms 10, no. 9: 1838. https://doi.org/10.3390/microorganisms10091838