Our Mental Health Is Determined by an Intrinsic Interplay between the Central Nervous System, Enteric Nerves, and Gut Microbiota
Abstract
:1. Introduction
2. Maintenance of the ENS
3. The Influence of Gut Bacteria on the ENS
4. Enteroendocrine Signaling and Immune Response
5. The Role of Gut Bacteria in Mental Health
6. Conclusions
Funding
Conflicts of Interest
References
- 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]
- 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] [PubMed]
- Corpening, J.C.; Cantrell, V.A.; Deal, K.K.; Southard-Smith, E.M. A Histone2BCerulean BAC transgene identifies differential expression of Phox2b in migrating enteric neural crest derivatives and enteric glia. Dev. Dyn. 2008, 237, 1119–1132. [Google Scholar] [CrossRef] [PubMed]
- 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] [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] [PubMed]
- Sun, X.; Kaufman, P.D. Ki-67: More than a proliferation marker. Chromosoma 2018, 127, 175–186. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Furness, J.B. The Enteric Nervous System; Blackwell Publishing: Oxford, UK, 2006. [Google Scholar]
- Bennett, M.R.; Burnstock, G.; Holman, M.E. Transmission from perivascular inhibitory nerves to the smooth muscle of the guinea-pig taenia coli. J. Physiol. 1966, 182, 527–540. [Google Scholar] [CrossRef]
- Bulbring, E.; Tomita, T. Properties of the inhibitory potential of smooth muscle as observed in the response to field stimulation of the guinea-pig taenia coli. J. Physiol. 1967, 189, 299–315. [Google Scholar] [CrossRef]
- Furness, J.B. Types of neurons in the enteric nervous system. J. Auton. Nerv. Syst. 2001, 81, 87–96. [Google Scholar] [CrossRef]
- Berthoud, H.-R.; Neuhuber, W.L. Functional and chemical anatomy of the afferent vagal system. Auton. Neurosci. 2000, 85, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Mahmoudian Dehkordi, 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] [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]
- Brookes, S.J.; Song, Z.M.; Ramsay, G.A.; Costa, M. Long aboral projections of Dogiel type II, AH neurons within the myenteric plexus of the guinea pig small intestine. J. Neurosci. 1995, 15, 4013–4022. [Google Scholar] [CrossRef] [PubMed]
- Furness, J.B.; Kunze, W.A.; Bertrand, P.P.; Clerc, N.; Bornstein, J.C. Intrinsic primary afferent neurons of the intestine. Prog. Neurobiol. 1998, 54, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Dicks, L.M.T. Gut Bacteria and neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef] [PubMed]
- Kugler, E.M.; Michel, K.; Zeller, F.; Demir, I.E.; Ceyhan, G.O.; Schemann, M.; Mazzuoli-Weber, G. Mechanical stress activates neurites and somata of myenteric neurons. Front. Cell Neurosci. 2015, 9, 342. [Google Scholar] [CrossRef]
- Brehmer, A. Classification of human enteric neurons. Histochem. Cell Biol. 2021, 156, 95–108. [Google Scholar] [CrossRef]
- Spencer, N.J.; Smith, T.K. Mechanosensory S-neurons rather than AH-neurons appear to generate a rhythmic motor pattern in guinea-pig distal colon. J. Physiol. 2004, 558, 577–596. [Google Scholar] [CrossRef]
- Mazzuoli-Weber, G.; Schemann, M. Mechanosensitive enteric neurons in the guinea pig gastric corpus. Front Cell Neurosci. 2015, 9, 430. [Google Scholar] [CrossRef]
- Kunze, W.A.; Furness, J.B.; Bertrand, P.P.; Bornstein, J.C. Intracellular recording from myenteric neurons of the guinea-pig ileum that respond to stretch. J. Physiol. 1998, 506 Pt 3, 827–842. [Google Scholar] [CrossRef]
- Bertrand, P.P.; Kunze, W.A.; Bornstein, J.C.; Furness, J.B.; Smith, M.L. Analysis of the responses of myenteric neurons in the small intestine to chemical stimulation of the mucosa. Am. J. Physiol. 1997, 273, G422–G435. [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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- Spencer, N.J.; Hu, H. Enteric nervous system: Sensory transduction, neural circuits and gastrointestinal motility. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 338–351. [Google Scholar] [CrossRef] [PubMed]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
- 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]
- 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]
- Dicks, L.M.T. How does quorum sensing of intestinal bacteria affect our health and mental status? Microorganisms 2022, 10, 1969. [Google Scholar] [CrossRef]
- Huizinga, J.D.; Thuneberg, L.; Klűppel, M.; Malysz, J.; Mikkelsen, H.B.; Bernstein, A. W/kit gene required for interstitial cells of Cajal and for intestinal pacemaker activity. Nature 1995, 373, 347–349. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- 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]
- 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]
- 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-ƙB 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] [PubMed]
- Stanley, D.; Mason, L.J.; Mackin, E.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] [PubMed]
- 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] [PubMed]
- 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. The human microbiome jumpstart reference strains consortium. A catalog of reference genomes from the human microbiome. Science 2010, 328, 994–999. [Google Scholar] [PubMed]
- 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] [PubMed]
- 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]
- 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]
- 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]
- 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] [PubMed]
- Tracey, K.J.; Chavan, S.S. Nerve Stimulation for Treatment of Diseases and Disorders. International Patent PCT/US2016/018573, 19 February 2016. [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] [PubMed]
- 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] [PubMed]
- 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]
- Brown, G.C. The endotoxin hypothesis of neurodegeneration. J. Neuroinflamm. 2019, 16, 180. [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]
- 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]
- Cushing, K.; Alvarado, D.; Ciorba, A.M. Butyrate and mucosal inflammation: New scientific evidence supports clinical observation. Clin. Transl. Gastroenterol. 2015, 6, e108. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Nohr, M.K.; Pedersen, M.H.; Gille, A.; Egerod, K.L.; Engelstoft, M.S.; Husted, A.S.; Sichlau, R.M.; Grunddal, K.V.; Seier Poulsen, 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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- Lau, A.; Tymianski, M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflug. Arch. 2010, 460, 525–542. [Google Scholar] [CrossRef]
- 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]
- 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]
- 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]
- 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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- Takanaga, H.; Ohtsuki, S.; Hosoya, K.-I.; Terasaki, T. GAT2/BGT-1 as a system responsible for the transport of -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]
- 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]
- 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]
- 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, 9730467. [Google Scholar]
- 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] [PubMed]
- Borodovitsyna, O.; Flamini, M.; Chandler, D. Noradrenergic modulation of cognition in health and disease. Neural Plast. 2017, 2017, 6031478. [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] [PubMed]
- 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] [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]
- 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]
- 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]
- 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]
- Collins, S.M. A role for the gut microbiota in IBS. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 497–505. [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, 567–650. [Google Scholar] [CrossRef] [PubMed]
- Worthington, J.J. The intestinal immunoendocrine axis: Novel cross-talk between enteroendocrine cells and the immune system during infection and inflammatory disease. Biochem. Soc. Trans. 2015, 43, 727–733. [Google Scholar] [CrossRef]
- Gribble, F.M. The gut endocrine system as a coordinator of postprandial nutrient homoeostasis. Proc. Nutr. Soc. 2012, 71, 456–462. [Google Scholar] [CrossRef]
- Genton, L.; Kudsk, K.A. Interactions between the enteric nervous system and the immune system: Role of neuropeptides and nutrition. Am. J. Surg. 2003, 186, 253–258. [Google Scholar] [CrossRef]
- Harrison, E.; Lal, S.; McLaughlin, J.T. Enteroendocrine cells in gastrointestinal pathophysiology. Curr. Opin. Pharmacol. 2013, 13, 941–945. [Google Scholar] [CrossRef]
- Moran, G.W.; Leslie, F.C.; Levison, S.E.; McLaughlin, J.T. Enteroendocrine cells: Neglected players in gastrointestinal disorders? Therap. Adv. Gastroenterol. 2008, 1, 51–60. [Google Scholar] [CrossRef]
- Shajib, M.S.; Khan, W.I. The role of serotonin and its receptors in activation of immune responses and inflammation. Acta Physiol. 2015, 213, 561–574. [Google Scholar] [CrossRef]
- Bar, F.; Foh, B.; Pagel, R.; Schroder, T.; Schlichting, H.; Hirose, M.; Lemcke, S.; Klinger, A.; König, P.; Karsten, C.M.; et al. Carboxypeptidase E modulates intestinal immune homeostasis and protects against experimental colitis in mice. PLoS ONE 2014, 9, e102347. [Google Scholar] [CrossRef]
- Jia, X.; Cong, B.; Zhang, J.; Li, H.; Liu, W.; Chang, H.; Dong, M.; Ma, C. CCK8 negatively regulates the TLR9-induced activation of human peripheral blood pDCs by targeting TRAF6 signaling. Eur. J. Immunol. 2014, 44, 489–499. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Han, D.; Cong, B.; Shan, B.; Zhang, J.; Chen, H.; Ma, C.; Liyanage, S.S. Cholecystokinin octapeptide significantly suppresses collagen-induced arthritis in mice by inhibiting Th17 polarization primed by dendritic cells. Cell Immunol. 2011, 272, 53–60. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.G.; Liu, J.X.; Jia, X.X.; Geng, J.; Yu, F.; Cong, B. Cholecystokinin octapeptide regulates the differentiation and effector cytokine production of CD4(+) T cells in vitro. Int. Immunopharmacol. 2014, 20, 307–315. [Google Scholar] [CrossRef]
- Oiry, C.; Gagne, D.; Cottin, E.; Bernad, N.; Galleyrand, J.C.; Berge, G.; Lignon, M.F.; Eldin, P.; Le Cunff, M.; Léger, J.; et al. Cholecystokinin B receptor from human Jurkat lymphoblastic T cells is involved in activator protein-1-responsive gene activation. Mol. Pharmacol. 1997, 52, 292–299. [Google Scholar] [CrossRef] [PubMed]
- Reardon, C.; Duncan, G.S.; Brustle, A.; Brenner, D.; Tusche, M.W.; Olofsson, P.S.; Rosas-Ballina, M.; Tracey, K.J.; Mak, T.W. Lymphocyte-derived ACh regulates local innate but not adaptive immunity. Proc. Natl. Acad. Sci. USA 2013, 110, 1410–1415. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Patel, K.; Tae, H.J.; Lustig, A.; Kim, J.W.; Mattson, M.P.; Taub, D.D. Ghrelin augments murine T-cell proliferation by activation of the phosphatidylinositol-3-kinase, extracellular signal-regulated kinase and protein kinase C signaling pathways. FEBS Lett. 2014, 588, 4708–4719. [Google Scholar] [CrossRef]
- Delgado, M.; Anderson, P.; Garcia-Salcedo, J.A.; Caro, M.; Gonzalez-Rey, E. Neuropeptides kill African trypanosomes by targeting intracellular compartments and inducing autophagic-like cell death. Cell Death Differ. 2009, 16, 406–416. [Google Scholar] [CrossRef]
- Chorny, A.; Anderson, P.; Gonzalez-Rey, E.; Delgado, M. Ghrelin protects against experimental sepsis by inhibiting high-mobility group box 1 release and by killing bacteria. J. Immunol. 2008, 180, 8369–8377. [Google Scholar] [CrossRef]
- Dixit, V.D.; Yang, H.; Cooper-Jenkins, A.; Giri, B.B.; Patel, K.; Taub, D.D. Reduction of T cell-derived ghrelin enhances proinflammatory cytokine expression: Implications for age-associated increases in inflammation. Blood 2009, 113, 5202–5205. [Google Scholar] [CrossRef]
- Friedrich, M.; Diegelmann, J.; Schauber, J.; Auernhammer, C.J.; Brand, S. Intestinal neuroendocrine cells and goblet cells are mediators of IL-17A-amplified epithelial IL-17C production in human inflammatory bowel disease. Mucosal. Immunol. 2014, 8, 943–958. [Google Scholar] [CrossRef]
- Bogunovic, M.; Dave, S.H.; Tilstra, J.S.; Chang, D.T.W.; Harpaz, N.; Xiong, H.B.; Mayer, L.F.; Plevy, S.E.; Margolis, K.G.; Vittorio, J.; et al. Enteroendocrine cells express functional Toll-like receptors. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G1770–G1783. [Google Scholar] [CrossRef] [PubMed]
- Selleri, S.; Palazzo, M.; Deola, S.; Wang, E.; Balsari, A.; Marincola, F.M.; Rumio, C. Induction of pro-inflammatory programs in enteroendocrine cells by the Toll-like receptor agonists flagellin and bacterial LPS. Int. Immunol. 2008, 20, 961–970. [Google Scholar] [CrossRef] [PubMed]
- Dicks, L.M.T.; Hurn, D.; Hermanus, D. Gut bacteria and neuropsychiatric disorders. Microorganisms 2021, 9, 2583. [Google Scholar] [CrossRef] [PubMed]
- Galland, L. The gut microbiome and the brain. J. Med. Food 2014, 17, 1261–1272. [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. [Google Scholar] [CrossRef]
- 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]
- Liang, S.; Wu, X.; Hu, X.; Wang, T.; Jin, F. Recognizing depression from the microbiota–gut–brain axis. Int. J. Mol. Sci. 2018, 19, 1592. [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]
- 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] [PubMed]
- Ong, I.M.; Gonzalez, J.G.; McIlwain, S.J.; Sawin, E.A.; Schoen, A.J.; Adluru, N.; Alexander, A.L.; Yu, J.-P.J. Gut microbiome populations are associated with structure-specific changes in white matter architecture. Transl. Psychiatry 2018, 8, 6. [Google Scholar] [CrossRef] [PubMed]
- Erny, D.; Hrabê 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] [PubMed]
- Jewett, B.E.; Sharma, S. Physiology, GABA; StatPearls: Treasure Island, FL, USA, 2021. Available online: https://pubmed.ncbi.nlm.nih.gov/30020683/ (accessed on 14 October 2021).
- Dinan, T.G.; Stanton, C.; Cryan, J.F. Psychobiotics: A novel class of psychotropic. Biol. Psychiatry 2013, 74, 720–726. [Google Scholar] [CrossRef] [PubMed]
- Roshchina, V.V. Evolutionary considerations of neurotransmitters in microbial, plant, and animal cells. In Microbial Endocrinology: Interkingdom Signaling in Infectious Disease and Health, 1st ed.; Lyte, M., Freestone, P.P.E., Eds.; Springer: New York, NY, USA, 2010; pp. 17–52. [Google Scholar]
- 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]
- Mayer, E.A. Gut feelings: The emerging biology of gut–brain communication. Nat. Rev. Neurosci. 2011, 12, 453–466. [Google Scholar] [CrossRef]
- Hyland, N.P.; Cryan, J.F. Microbe-host interactions: Influence of the gut microbiota on the enteric nervous system. Dev. Biol. 2016, 417, 182–187. [Google Scholar] [CrossRef]
- Tavakoli, P.; Vollmer-Conna, U.; Hadzi-Pavlovic, D.; Grimm, M.C. A Review of inflammatory bowel disease: A model of microbial, immune and neuropsychological integration. Public Health Rev. 2021, 42, 1603990. [Google Scholar] [CrossRef]
- Kim, Y.S.; Kim, J.; Park, S.-J. High-throughput 16S rRNA gene sequencing reveals alterations of mouse intestinal microbiota after radiotherapy. Anaerobe 2015, 33, 1–7. [Google Scholar] [CrossRef]
- Saulnier, D.M.; Riehle, K.; Mistretta, T.-A.; Diaz, M.-A.; Mandal, D.; Raza, S.; Weidler, E.M.; Qin, X.; Coarfa, C.; Milosavljevic, A.; et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology 2011, 141, 1782–1791. [Google Scholar] [CrossRef]
- Ley, R.E. Gut microbiota in 2015: Prevotella in the gut: Choose carefully. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 69–70. [Google Scholar] [CrossRef] [PubMed]
- Hao, Z.; Wang, W.; Guo, R.; Liu, H. Faecalibacterium prausnitzii (ATCC 27766) has preventive and therapeutic effects on chronic unpredictable mild stress-induced depression-like and anxiety-like behavior in rats. Psychoneuroendocrinology 2019, 104, 132–142. [Google Scholar] [CrossRef] [PubMed]
- Molteni, R.; Macchi, F.; Zecchillo, C.; Dell’Agli, M.; Colombo, E.; Calabrese, F.; Guidotti, G.; Racagni, G.; Riva, M.A. Modulation of the inflammatory response in rats chronically treated with the antidepressant agomelatine. Eur. Neuropsychopharmacol. 2013, 23, 1645–1655. [Google Scholar] [CrossRef] [PubMed]
- Luki’c, I.; Getselter, D.; Ziv, O.; Oron, O.; Reuveni, E.; Koren, O.; Elliott, E. Antidepressants affect gut microbiota and Ruminococcus flavefaciens is able to abolish their effects on depressive-like behavior. Transl. Psychiatry 2019, 9, 133. [Google Scholar] [CrossRef] [PubMed]
- Shen, Y.; Xu, J.; Li, Z.; Huang, Y.; Yuan, Y.; Wang, J.; Zhang, M.; Hu, S.; Liang, Y. Analysis of gut microbiota diversity and auxiliary diagnosis as a biomarker in patients with schizophrenia: A cross-sectional study. Schizophr. Res. 2018, 197, 470–477. [Google Scholar] [CrossRef]
- Qin, P.; Zou, Y.; Dai, Y.; Luo, G.; Zhang, X.; Xiao, L. Characterization a novel butyric acid-producing bacterium Collinsella aerofaciens subsp. shenzhenensis subsp. nov. Microorganisms 2019, 7, 78. [Google Scholar] [CrossRef]
- Ezaki, T.; Kawamura, Y.; Li, N.; Li, Z.Y.; Zhao, L.; Shu, S. Proposal of the genera Anaerococcus gen. nov., Peptoniphilus gen. nov. and Gallicola gen. nov. for members of the genus Peptostreptococcus. Int. J. Syst. Evol. Microbiol. 2001, 51, 1521–1528. [Google Scholar] [CrossRef]
- Qing, Y.; Xu, L.; Cui, G.; Sun, L.; Hu, X.; Yang, X.; Jiang, J.; Zhang, J.; Zhang, T.; Wang, T.; et al. Salivary microbiome profiling reveals a dysbiotic schizophrenia-associated microbiota. NPJ Schizophr. 2021, 7, 51. [Google Scholar] [CrossRef]
- Zhou, S.; Cai, Y.; Wang, M.; Yang, W.D.; Duan, N. Oral microbial flora of patients with Sicca syndrome. Mol. Med. Rep. 2018, 18, 4895–4903. [Google Scholar] [CrossRef]
- Castro-Nallar, E.; Bendall, M.L.; Pérez-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. Peer J. 2015, 3, e1140. [Google Scholar] [CrossRef]
- De Vuyst, L.; Leroy, F. Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int. J. Food Microbiol. 2011, 149, 73–80. [Google Scholar] [CrossRef] [PubMed]
- Dickerson, F.; Severance, E.; Yolken, R. The microbiome, immunity, and schizophrenia and bipolar disorder. Brain Behav. Immun. 2017, 62, 46–52. [Google Scholar] [CrossRef] [PubMed]
- Carlier, J.P.; Bedora-Faure, M.; K’ouas, G.; Alauzet, C.; Mory, F. Proposal to unify Clostridium orbiscindens Winter et al. 1991 and Eubacterium plautii (Séguin 1928) Hofstad and Aasjord 1982, with description of Flavonifractor plautii gen. nov., comb. nov. and reassignment of Bacteroides capillosus to Pseudoflavonifractor capiillosus gen. nov., comb. nov. Int. J. Syst. Evol. Microbiol. 2010, 60, 585–590. [Google Scholar] [PubMed]
- Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: From antioxidant to nutraceutical. Eur. J. Pharmacol. 2008, 585, 325–337. [Google Scholar] [CrossRef] [PubMed]
- Painold, A.; Mörkl, S.; Kashofer, K.; Halwachs, B.; Dalkner, N.; Bengesser, S.; Birner, A.; Fellendorf, F.; Platzer, M.; Queissner, R.; et al. A step ahead: Exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode. Bipolar Disord. 2019, 21, 40–49. [Google Scholar] [CrossRef] [PubMed]
- Evans, S.J.; Bassis, C.M.; Hein, R.; Assari, S.; Flowers, S.A.; Kelly, M.B.; Young, V.B.; Ellingrod, V.E.; McInnis, M.G. The gut microbiome composition associates with bipolar disorder and illness severity. J. Psychiatr. Res. 2017, 87, 23–29. [Google Scholar] [CrossRef]
- Qiu, X.; Zhang, M.; Yang, X.; Hong, N.; Yu, C. Faecalibacterium prausnitzii upregulates regulatory T cells and anti-inflammatory cytokines in treating TNBS-induced colitis. J. Crohns Colitis 2013, 7, e558–e568. [Google Scholar] [CrossRef]
- Mccartney, A.L.; Parracho, H.M.R.T.; Bingham, M.O.; Gibson, G.R. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J. Med. Microbiol. 2005, 54, 987–991. [Google Scholar]
- Finegold, S.M.; Dowd, S.E.; Gontcharova, V.; Liu, C.; Henley, K.E.; Wolcott, R.D.; Youn, E.; Summanen, P.H.; Granpeesheh, D.; Dixon, D.; et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010, 16, 444–453. [Google Scholar] [CrossRef]
- Ruscio, A.M.; Stein, D.J.; Chiu, W.T.; Kessler, R.C. The epidemiology of obsessive-compulsive disorder in the National Comorbidity Survey Replication. Mol. Psychiatry 2008, 15, 53–63. [Google Scholar] [CrossRef]
- Turna, J.; Kaplan, K.G.; Anglin, R.; Van Ameringen, M. “What’s bugging the gut in OCD?” A review of the gut microbiome in obsessive-compulsive disorder. Depress. Anxiety 2016, 33, 171–178. [Google Scholar] [CrossRef]
- Göker, M.; Gronow, S.; Zeytun, A.; Nolan, M.; Lucas, S.; Lapidus, A.; Hammon, N.; Deshpande, S.; Cheng, J.F.; Pitluck, S.; et al. Complete genome sequence of Odoribacter splanchnicus type strain (1651/6 T). Stand. Genom. Sci. 2011, 4, 200–209. [Google Scholar] [CrossRef]
- Messaoudi, M.; Violle, N.; Bisson, J.-F.; Desor, D.; Javelot, H.; Rougeot, C. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes 2011, 2, 256–261. [Google Scholar] [CrossRef]
- Kantak, P.A.; Bobrow, D.N.; Nyby, J.G. Obsessive-compulsive-like behaviors in house mice are attenuated by a probiotic (Lactobacillus rhamnosus GG). Behav. Pharmacol. 2014, 25, 71–79. [Google Scholar] [CrossRef]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
- Marcobal, A.; de las Rivas, B.; Muñoz, R. First genetic characterization of a bacterial beta-phenylethylamine biosynthetic enzyme in Enterococcus faecium RM58. FEMS Microbiol. 2006, 258, 144–149. [Google Scholar] [CrossRef]
- Irsfeld, M.; Spadafore, M.; Prüß, B.M. β-phenylethylamine, a small molecule with a large impact. Webmedcentral 2013, 4, 4409. [Google Scholar]
- Yang, Y.X.; Mu, C.L.; Luo, Z.; Zhu, W.Y. Bromochloromethane, a methane analogue, affects the microbiota and metabolic profiles of the rat gastrointestinal tract. Appl. Environ. Microbiol. 2015, 82, 778–787. [Google Scholar] [CrossRef]
- Dyck, L.E.; Yang, C.R.; Boulton, A.A. The biosynthesis of p-tyramine, m-tyramine, and beta-phenylethylamine by rat striatal slices. J. Neurosci. Res. 1983, 10, 211–220. [Google Scholar] [CrossRef] [PubMed]
- Berry, M.D.; Shitut, M.R.; Almousa, A.; Alcorn, J.; Tomberli, B. Membrane permeability of trace amines: Evidence for a regulated, activity-dependent, nonexocytotic, synaptic release. Synapse 2013, 67, 656–667. [Google Scholar] [CrossRef] [PubMed]
- Mosnaim, A.D.; Callaghan, O.H.; Hudzik, T.; Wolf, M.E. Rat brain-uptake index for phenylethylamine and various monomethylated derivatives. Neurochem. Res. 2013, 38, 842–846. [Google Scholar] [CrossRef]
- Baker, G.B.; Raiteri, M.; Bertollini, A.; del Carmine, R. Interaction of betaphenethylamine with dopamine and noradrenaline in the central nervous system of the rat. J. Pharm. Pharmacol. 1976, 28, 456–457. [Google Scholar] [CrossRef]
- Gainetdinov, R.R.; Hoener, M.C.; Berry, M.D. Trace amines and their receptors. Pharmacol. Rev. 2018, 70, 549–620. [Google Scholar] [CrossRef]
- Del Rio, J.A.; Soriano, E.; Ferrer, I. Development of GABA-immunoreactivity in the neocortex of the mouse. J. Comp. Neurol. 1992, 326, 501–526. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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. Our Mental Health Is Determined by an Intrinsic Interplay between the Central Nervous System, Enteric Nerves, and Gut Microbiota. Int. J. Mol. Sci. 2024, 25, 38. https://doi.org/10.3390/ijms25010038
Dicks LMT. Our Mental Health Is Determined by an Intrinsic Interplay between the Central Nervous System, Enteric Nerves, and Gut Microbiota. International Journal of Molecular Sciences. 2024; 25(1):38. https://doi.org/10.3390/ijms25010038
Chicago/Turabian StyleDicks, Leon M. T. 2024. "Our Mental Health Is Determined by an Intrinsic Interplay between the Central Nervous System, Enteric Nerves, and Gut Microbiota" International Journal of Molecular Sciences 25, no. 1: 38. https://doi.org/10.3390/ijms25010038
APA StyleDicks, L. M. T. (2024). Our Mental Health Is Determined by an Intrinsic Interplay between the Central Nervous System, Enteric Nerves, and Gut Microbiota. International Journal of Molecular Sciences, 25(1), 38. https://doi.org/10.3390/ijms25010038