The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome
Abstract
:1. Introduction
1.1. The Instestinal Metaboloma’s Concept
1.2. Metabolome Analysis Techniques
2. The Metabolic Processes of Gut Microorganisms
3. The Metabolic Activity of Gut Microorganisms
4. The Role of the Intestinal Microbiota in the Gut/Brain Axis
4.1. The Bio-Molecular Pathways of ENS and Brain
4.2. The Effect of Gut Microbiota on the CNS
4.3. The Effect of the CNS on the Gut Microbiota
5. Gut/Brain Axis and Diseases
5.1. Neurological Disorders
5.2. Neuro-Psychopathological Diseases
6. The Modulation of the Gut-Brain Axis via Probiotics, Psychobiotics, and Prebiotics
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Wegierska, A.E.; Charitos, I.A.; Topi, S.; Potenza, M.A.; Montagnani, M.; Santacroce, L. The Connection Between Physical Exercise and Gut Microbiota: Implications for Competitive Sports Athletes. Sports Med. 2022, 52, 2355–2369. [Google Scholar] [CrossRef] [PubMed]
- Bottalico, L.; Castellaneta, F.; Charitos, I.A. From Hydrotherapy to the Discovery of The Gut Microbiota: The Historical Gastrointestinal Health Concept. Pharmacophore 2020, 11, 82–90. [Google Scholar]
- Santacroce, L.; Topi, S.; Haxhirexha, K.; Hidri, S.; Charitos, I.A.; Bottalico, L. Medicine and Healing in the Pre-Socratic Thought—A Brief Analysis of Magic and Rationalism in Ancient Herbal Therapy. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 282–287. [Google Scholar] [CrossRef] [PubMed]
- 74Ballini, A.; Charitos, I.A.; Cantore, S.; Topi, S.; Bottalico, L.; Santacroce, L. About Functional Foods: The Probiotics and Prebiotics State of Art. Antibiotics 2023, 12, 635. [Google Scholar] [CrossRef]
- Santacroce, L.; Man, A.; Charitos, I.A.; Haxhirexha, K.; Topi, S. Current knowledge about the connection between health status and gut microbiota from birth to elderly. A narrative review. Front. Biosci. 2021, 26, 135–148. [Google Scholar] [CrossRef]
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stilling, R.M.; Dinan, T.G.; Cryan, J.F. Microbial genes, brain & behaviour—Epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014, 13, 69–86. [Google Scholar] [CrossRef]
- Puchades-Carrasco, L.; Pineda-Lucena, A. Metabolomics in pharmaceutical research and development. Curr. Opin. Biotechnol. 2015, 35, 73–77. [Google Scholar] [CrossRef]
- Onuh, J.O.; Qiu, H. Metabolic Profiling and Metabolites Fingerprints in Human Hypertension: Discovery and Potential. Metabolites 2021, 11, 687. [Google Scholar] [CrossRef] [PubMed]
- Manzoni, C.; Kia, D.A.; Vandrovcova, J.; Hardy, J.; Wood, N.W.; Lewis, P.A.; Ferrari, R. Genome, transcriptome and proteome: The rise of omics data and their integration in biomedical sciences. Briefings Bioinform. 2018, 19, 286–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loo, R.L.; Lodge, S.; Kimhofer, T.; Bong, S.-H.; Begum, S.; Whiley, L.; Gray, N.; Lindon, J.C.; Nitschke, P.; Lawler, N.G.; et al. Quantitative In-Vitro Diagnostic NMR Spectroscopy for Lipoprotein and Metabolite Measurements in Plasma and Serum: Recommendations for Analytical Artifact Minimization with Special Reference to COVID-19/SARS-CoV-2 Samples. J. Proteome Res. 2020, 19, 4428–4441. [Google Scholar] [CrossRef]
- Lee, D.-K.; Yoon, M.H.; Kang, Y.P.; Yu, J.; Park, J.H.; Lee, J.; Kwon, S.W. Comparison of primary and secondary metabolites for suitability to discriminate the origins of Schisandra chinensis by GC/MS and LC/MS. Food Chem. 2013, 141, 3931–3937. [Google Scholar] [CrossRef] [PubMed]
- Fiehn, O. Metabolomics—The link between genotypes and phenotypes. Plant Mol. Biol. 2002, 48, 155–171. [Google Scholar] [CrossRef] [PubMed]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Almeida, A.; Mitchell, A.L.; Boland, M.; Forster, S.C.; Gloor, G.B.; Tarkowska, A.; Lawley, T.D.; Finn, R.D. A new genomic blueprint of the human gut microbiota. Nature 2019, 568, 499–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Psychogios, N.; Hau, D.D.; Peng, J.; Guo, A.C.; Mandal, R.; Bouatra, S.; Sinelnikov, I.; Krishnamurthy, R.; Eisner, R.; Gautam, B.; et al. The Human Serum Metabolome. PLoS ONE 2011, 6, e16957. [Google Scholar] [CrossRef] [Green Version]
- Pinu, F.R.; Goldansaz, S.A.; Jaine, J. Translational Metabolomics: Current Challenges and Future Opportunities. Metabolites 2019, 9, 108. [Google Scholar] [CrossRef] [Green Version]
- Deschamps, E.; Calabrese, V.; Schmitz, I.; Hubert-Roux, M.; Castagnos, D.; Afonso, C. Advances in Ultra-High-Resolution Mass Spectrometry for Pharmaceutical Analysis. Molecules 2023, 28, 2061. [Google Scholar] [CrossRef] [PubMed]
- Gathungu, R.M.; Kautz, R.; Kristal, B.S.; Bird, S.S.; Vouros, P. The integration of LC-MS and NMR for the analysis of low molecular weight trace analytes in complex matrices. Mass Spectrom. Rev. 2020, 39, 35–54. [Google Scholar] [CrossRef]
- Letertre, M.P.M.; Giraudeau, P.; de Tullio, P. Nuclear Magnetic Resonance Spectroscopy in Clinical Metabolomics and Personalized Medicine: Current Challenges and Perspectives. Front. Mol. Biosci. 2021, 8, 698337. [Google Scholar] [CrossRef] [PubMed]
- Garg, E.; Zubair, M. Mass Spectrometer. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
- Dettmer, K.; Aronov, P.A.; Hammock, B.D. Mass spectrometry-based metabolomics. Mass Spectrom. Rev. 2007, 26, 51–78. [Google Scholar] [CrossRef] [PubMed]
- Sanz-Nebot, V.; Benavente, F.; Barbosa, J. Liquid chromatography–mass spectrometry and capillary electrophoresis combined approach for separation and characterization of multicomponent peptide mixtures: Application to crude products of leuprolide synthesis. J. Chromatogr. A 2002, 950, 99–111. [Google Scholar] [CrossRef] [PubMed]
- Ho, C.S.; Lam, C.W.; Chan, M.H.; Cheung, R.C.; Law, L.K.; Lit, L.C.; Ng, K.F.; Suen, M.W.; Tai, H.L. Electrospray ionisation mass spectrometry: Principles and clinical applications. Clin. Biochem. Rev. 2003, 24, 3–12. [Google Scholar]
- Langridge, J.I.; Claude, E. Matrix-Assisted Laser Desorption and Desorption Electrospray Ionization Mass Spectrometry Coupled to Ion Mobility. Methods Mol. Biol. 2020, 2084, 245–265. [Google Scholar] [CrossRef]
- Neagu, A.N.; Jayathirtha, M.; Baxter, E.; Donnelly, M.; Petre, B.A.; Darie, C.C. Applications of Tandem Mass Spectrometry (MS/MS) in Protein Analysis for Biomedical Research. Molecules 2022, 27, 2411. [Google Scholar] [CrossRef] [PubMed]
- Heiles, S. Advanced tandem mass spectrometry in metabolomics and lipidomics—Methods and applications. Anal. Bioanal. Chem. 2021, 413, 5927–5948. [Google Scholar] [CrossRef]
- Schwartz, J.C.; Senko, M.W.; Syka, J.E.P. A two-dimensional quadrupole ion trap mass spectrometer. J. Am. Soc. Mass Spectrom. 2002, 13, 659–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marshall, A.G.; Hendrickson, C.L.; Jackson, G.S. Fourier transform ion cyclotron resonance mass spectrometry: A primer. Mass Spectrom. Rev. 1998, 17, 1–35. [Google Scholar] [CrossRef]
- Donaldson, G.P.; Lee, S.M.; Mazmanian, S.K. Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 2016, 14, 20–32. [Google Scholar] [CrossRef] [Green Version]
- Oren, A.; Garrity, G.M. Valid publication of the names of forty-two phyla of prokaryotes. Int. J. Syst. Evol. Microbiol. 2021, 71, 005056. [Google Scholar] [CrossRef]
- Flint, H.J.; Scott, K.P.; Duncan, S.H.; Louis, P.; Forano, E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 2012, 3, 289–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krautkramer, K.A.; Fan, J.; Bäckhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Genet. 2020, 19, 77–94. [Google Scholar] [CrossRef] [PubMed]
- Ballini, A.; Scacco, S.; Boccellino, M.; Santacroce, L.; Arrigoni, R. Microbiota and Obesity: Where Are We Now? Biology 2020, 9, 415. [Google Scholar] [CrossRef]
- Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef] [Green Version]
- Costea, P.I.; Hildebrand, F.; Arumugam, M.; Bäckhed, F.; Blaser, M.J.; Bushman, F.D.; de Vos, W.M.; Ehrlich, S.D.; Fraser, C.M.; Hattori, M.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seitz, J.; Trinh, S.; Herpertz-Dahlmann, B. The Microbiome and Eating Disorders. Psychiatr. Clin. N. Am. 2019, 42, 93–103. [Google Scholar] [CrossRef] [PubMed]
- Topi, S.; Bottalico, L.; Charitos, I.A.; Colella, M.; Di Domenico, M.; Palmirotta, R.; Santacroce, L. Biomolecular Mechanisms of Autoimmune Diseases and Their Relationship with the Resident Microbiota: Friend or Foe? Pathophysiology 2022, 29, 507–536. [Google Scholar] [CrossRef]
- Cheng, Y.; Ling, Z.; Li, L. The Intestinal Microbiota and Colorectal Cancer. Front. Immunol. 2020, 11, 615056. [Google Scholar] [CrossRef]
- Charitos, I.A.; Topi, S.; Gagliano-Candela, R.; De Nitto, E.; Polimeno, L.; Montagnani, M.; Santacroce, L. The toxic effects of endocrine disrupting chemicals (EDCs) on gut microbiota: Bisphenol A (BPA). A review. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 716–727. [Google Scholar] [CrossRef]
- Li, D.; Wang, P.; Wang, P.; Hu, X.; Chen, F. The gut microbiota: A treasure for human health. Biotechnol. Adv. 2016, 34, 1210–1224. [Google Scholar] [CrossRef]
- Aranda, F.; Bloy, N.; Galluzzi, L.; Kroemer, G.; Senovilla, L. Vitamin B6 improves the immunogenicity of cisplatin-induced cell death. Oncoimmunology 2014, 3, e955685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cerf-Bensussan, N.; Gaboriau-Routhiau, V. The immune system and the gut microbiota: Friends or foes? Nat. Rev. Immunol. 2010, 10, 735–744. [Google Scholar] [CrossRef] [PubMed]
- Mändar, R.; Mikelsaar, M. Transmission of Mother’s Microflora to the Newborn at Birth. Biol. Neonate 1996, 69, 30–35. [Google Scholar] [CrossRef] [PubMed]
- Penders, J.; Thijs, C.; Vink, C.; Stelma, F.F.; Snijders, B.; Kummeling, I.; Van den Brandt, P.A.; Stobberingh, E.E. Factors Influencing the Composition of the Intestinal Microbiota in Early Infancy. Pediatrics 2006, 118, 511–521. [Google Scholar] [CrossRef] [Green Version]
- Ballini, A.; Dipalma, G.; Isacco, C.G.; Boccellino, M.; Di Domenico, M.; Santacroce, L.; Nguyễn, K.C.D.; Scacco, S.; Calvani, M.; Boddi, A.; et al. Oral Microbiota and Immune System Crosstalk: A Translational Research. Biology 2020, 9, 131. [Google Scholar] [CrossRef]
- Ghosh, S.; Pramanik, S. Structural diversity, functional aspects and future therapeutic applications of human gut microbiome. Arch. Microbiol. 2021, 203, 5281–5308. [Google Scholar] [CrossRef]
- Isacco, C.G.; Ballini, A.; De Vito, D.; Nguyen, K.C.D.; Cantore, S.; Bottalico, L.; Quagliuolo, L.; Boccellino, M.; Di Domenico, M.; Santacroce, L.; et al. Rebalancing the Oral Microbiota as an Efficient Tool in Endocrine, Metabolic and Immune Disorders. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 777–784. [Google Scholar] [CrossRef]
- Gogou, M.; Kolios, G. The effect of dietary supplements on clinical aspects of autism spectrum disorder: A systematic review of the literature. Brain Dev. 2017, 39, 656–664. [Google Scholar] [CrossRef]
- Santacroce, L. Comment on “Could health only be defined by an equilibrated microbiome? A COVID-19 reappraisal” by P. Charlier. Ethic-Med. Public Health 2021, 19, 100720. [Google Scholar] [CrossRef]
- Santacroce, L.; Inchingolo, F.; Topi, S.; Del Prete, R.; Di Cosola, M.; Charitos, I.A.; Montagnani, M. Potential beneficial role of probiotics on the outcome of COVID-19 patients: An evolving perspective. Diabetes Metab. Syndr. 2021, 15, 295–301. [Google Scholar] [CrossRef]
- Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [Green Version]
- Torres-Sánchez, A.; Ruiz-Rodríguez, A.; Ortiz, P.; Aguilera, M. Key Stratification of Microbiota Taxa and Metabolites in the Host Metabolic Health–Disease Balance. Int. J. Mol. Sci. 2023, 24, 4519. [Google Scholar] [CrossRef]
- Flint, H.J.; Bayer, E.A.; Rincon, M.T.; Lamed, R.; White, B.A. Polysaccharide utilization by gut bacteria: Potential for new insights from genomic analysis. Nat. Rev. Microbiol. 2008, 6, 121–131. [Google Scholar] [CrossRef] [PubMed]
- Shortt, C.; Hasselwander, O.; Meynier, A.; Nauta, A.; Fernández, E.N.; Putz, P.; Rowland, I.; Swann, J.; Türk, J.; Vermeiren, J.; et al. Systematic review of the effects of the intestinal microbiota on selected nutrients and non-nutrients. Eur J Nutr. 2018, 57, 25–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leentjens, J.; Quintin, J.; Gerretsen, J.; Kox, M.; Pickkers, P.; Netea, M.G. The Effects of Orally Administered Beta-Glucan on Innate Immune Responses in Humans, a Randomized Open-Label Intervention Pilot-Study. PLoS ONE 2014, 9, e108794. [Google Scholar] [CrossRef] [Green Version]
- Hirsch, I.; Janovec, V.; Stranska, R.; Bendriss-Vermare, N. Cross Talk between Inhibitory Immunoreceptor Tyrosine-Based Activation Motif-Signaling and Toll-Like Receptor Pathways in Macrophages and Dendritic Cells. Front. Immunol. 2017, 8, 394. [Google Scholar] [CrossRef] [Green Version]
- Skodje, G.I.; Sarna, V.K.; Minelle, I.H.; Rolfsen, K.L.; Muir, J.G.; Gibson, P.R.; Veierød, M.B.; Henriksen, C.; Lundin, K.E. Fructan, Rather than Gluten, Induces Symptoms in Patients with Self-Reported Non-Celiac Gluten Sensitivity. Gastroenterology 2018, 154, 529–539.e2. [Google Scholar] [CrossRef] [Green Version]
- Riesbeck, S.; Petruschke, H.; Rolle-Kampczyk, U.; Schori, C.; Ahrens, C.H.; Eberlein, C.; Heipieper, H.J.; von Bergen, M.; Jehmlich, N. Adaptation and Resistance: How Bacteroides thetaiotaomicron Copes with the Bisphenol A Substitute Bisphenol F. Microorganisms 2022, 10, 1610. [Google Scholar] [CrossRef]
- Masi, A.C.; Stewart, C.J. Untangling human milk oligosaccharides and infant gut microbiome. iScience 2022, 25, 103542. [Google Scholar] [CrossRef]
- Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L.; et al. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef]
- Egan, M.; Dempsey, E.; Ryan, C.A.; Ross, R.P.; Stanton, C. The Sporobiota of the Human Gut. Gut Microbes 2021, 13, 1–17. [Google Scholar] [CrossRef]
- Ilhan, Z.E.; Marcus, A.K.; Kang, D.-W.; Rittmann, B.E.; Krajmalnik-Brown, R. pH-Mediated Microbial and Metabolic Interactions in Fecal Enrichment Cultures. Msphere 2017, 2, e00047-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Y.; He, H.; Cheng, Z.; Bai, Y.; Ma, X. The Role of Neuropeptide Y and Peptide YY in the Development of Obesity via Gut-brain Axis. Curr. Protein Pept. Sci. 2019, 20, 750–758. [Google Scholar] [CrossRef]
- Wang, C.; Zhang, Y.; Deng, M.; Wang, X.; Tu, W.; Fu, Z.; Jin, Y. Bioaccumulation in the gut and liver causes gut barrier dysfunction and hepatic metabolism disorder in mice after exposure to low doses of OBS. Environ. Int. 2019, 129, 279–290. [Google Scholar] [CrossRef]
- Lin, P.; Li, Q. Can gut flora changes be new biomarkers for depression? Front. Lab. Med. 2017, 1, 129–134. [Google Scholar] [CrossRef]
- Konturek, P.C.; Brzozowski, T.; Konturek, S.J. Stress and the gut: Pathophysiology, clinical consequences, diagnostic approach and treatment options. J. Physiol. Pharmacol. 2011, 62, 591–599. [Google Scholar] [PubMed]
- Furness, J.B.; Callaghan, B.P.; Rivera, L.R.; Cho, H.-J. The Enteric Nervous System and Gastrointestinal Innervation: Integrated Local and Central Control. Adv. Exp. Med. Biol. 2014, 817, 39–71. [Google Scholar] [CrossRef]
- Sasselli, V.; Pachnis, V.; Burns, A.J. The enteric nervous system. Dev. Biol. 2012, 366, 64–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shahrestani, J.; Das, J.M. Neuroanatomy, Auerbach Plexus. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
- Waxenbaum, J.A.; Reddy, V.; Varacallo, M. Anatomy, Autonomic Nervous System. 2021 Jul 29. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
- Natale, G.; Ryskalin, L.; Morucci, G.; Lazzeri, G.; Frati, A.; Fornai, F. The Baseline Structure of the Enteric Nervous System and Its Role in Parkinson’s Disease. Life 2021, 11, 732. [Google Scholar] [CrossRef] [PubMed]
- Costa, M.; Brookes, S.J.; Hennig, G.W. Anatomy and physiology of the enteric nervous system. Gut 2000, 47 (Suppl. S4), iv15–iv26. [Google Scholar] [CrossRef] [Green Version]
- Schemann, M.; Neunlist, M. The human enteric nervous system. Neurogastroenterol. Motil. 2004, 16 (Suppl. S1), 55–59. [Google Scholar] [CrossRef]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693 Pt B, 128–133. [Google Scholar] [CrossRef]
- O’donnell, M.P.; Fox, B.W.; Chao, P.H.; Schroeder, F.C.; Sengupta, P. A neurotransmitter produced by gut bacteria modulates host sensory behaviour. Nature 2020, 583, 415–420. [Google Scholar] [CrossRef]
- Fattorusso, A.; Di Genova, L.; Dell’Isola, G.B.; Mencaroni, E.; Esposito, S. Autism Spectrum Disorders and the Gut Microbiota. Nutrients 2019, 11, 521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giuffrè, M.; Moretti, R.; Campisciano, G.; da Silveira, A.B.M.; Monda, V.M.; Comar, M.; Di Bella, S.; Antonello, R.M.; Luzzati, R.; Crocè, L.S. You Talking to Me? Says the Enteric Nervous System (ENS) to the Microbe. How Intestinal Microbes Interact with the ENS. J. Clin. Med. 2020, 9, 3705. [Google Scholar] [CrossRef]
- Riegler, M.; Castagliuolo, I.; Wang, C.; Wlk, M.; Sogukoglu, T.; Wenzl, E.; Matthews, J.B.; Pothoulakis, C. Neurotensin stimulates Cl(−) secretion in human colonic mucosa in vitro: Role of adenosine. Gastroenterology 2000, 119, 348–357. [Google Scholar] [CrossRef] [PubMed]
- Björnsson, E.S.; Chey, W.D.; Ladabaum, U.; Woods, M.L.; Hooper, F.G.; Owyang, C.; Hasler, W.L. Differential 5-HT3 mediation of human gastrocolonic response and colonic peristaltic reflex. Am. J. Physiol. 1998, 275, G498–G505. [Google Scholar] [CrossRef]
- Foster, J.A.; Rinaman, L.; Cryan, J.F. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol. Stress 2017, 7, 124–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Geng, Z.H.; Zhu, Y.; Li, Q.L.; Zhao, C.; Zhou, P.H. Enteric Nervous System: The Bridge Between the Gut Microbiota and Neurological Disorders. Front. Aging Neurosci. 2022, 14, 810483. [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] [PubMed]
- Goehler, L.E.; Park, S.M.; Opitz, N.; Lyte, M.; Gaykema, R.P. Campylobacter jejuni infection increases anxiety-like behavior in the holeboard: Possible anatomical substrates for viscerosensory modulation of exploratory behavior. Brain Behav. Immun. 2008, 22, 354–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Lartigue, G.; Xu, C. Mechanisms of vagal plasticity influencing feeding behavior. Brain Res. 2018, 1693 Pt B, 146–150. [Google Scholar] [CrossRef]
- MacFabe, D.F.; Cain, N.E.; Boon, F.; Ossenkopp, K.-P.; Cain, D.P. Effects of the enteric bacterial metabolic product propionic acid on object-directed behavior, social behavior, cognition, and neuroinflammation in adolescent rats: Relevance to autism spectrum disorder. Behav. Brain Res. 2011, 217, 47–54. [Google Scholar] [CrossRef]
- Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kendall, M.M.; Rasko, D.A.; Sperandio, V. Global Effects of the Cell-to-Cell Signaling Molecules Autoinducer-2, Autoinducer-3, and Epinephrine in a luxS Mutant of Enterohemorrhagic Escherichia coli. Infect. Immun. 2007, 75, 4875–4884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freestone, P.P.; Sandrini, S.M.; Haigh, R.D.; Lyte, M. Microbial endocrinology: How stress influences susceptibility to infection. Trends Microbiol. 2008, 16, 55–64. [Google Scholar] [CrossRef] [PubMed]
- Collins, S.M.; Bercik, P. The Relationship Between Intestinal Microbiota and the Central Nervous System in Normal Gastrointestinal Function and Disease. Gastroenterology 2009, 136, 2003–2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madison, A.; Kiecolt-Glaser, J.K. Stress, depression, diet, and the gut microbiota: Human–bacteria interactions at the core of psychoneuroimmunology and nutrition. Curr. Opin. Behav. Sci. 2019, 28, 105–110. [Google Scholar] [CrossRef]
- Kunze, W.A.; Mao, Y.K.; Wang, B.; Huizinga, J.D.; Ma, X.; Forsythe, P.; Bienenstock, J. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. J. Cell. Mol. Med. 2009, 13, 2261–2270. [Google Scholar] [CrossRef]
- Huang, F.; Wu, X. Brain Neurotransmitter Modulation by Gut Microbiota in Anxiety and Depression. Front. Cell Dev. Biol. 2021, 9, 649103. [Google Scholar] [CrossRef] [PubMed]
- Barandouzi, Z.A.; Lee, J.; Del Carmen Rosas, M.; Chen, J.; Henderson, W.A.; Starkweather, A.R.; Cong, X.S. Associations of neurotransmitters and the gut microbiome with emotional distress in mixed type of irritable bowel syndrome. Sci. Rep. 2022, 12, 1648. [Google Scholar] [CrossRef] [PubMed]
- Yong, S.J.; Tong, T.; Chew, J.; Lim, W.L. Antidepressive Mechanisms of Probiotics and Their Therapeutic Potential. Front. Neurosci. 2020, 13, 1361. [Google Scholar] [CrossRef] [Green Version]
- Gao, K.; Mu, C.L.; Farzi, A.; Zhu, W.Y. Tryptophan Metabolism: A Link between the Gut Microbiota and Brain. Adv. Nutr. 2020, 11, 709–723. [Google Scholar] [CrossRef]
- Barrett, E.; Ross, R.P.; O’Toole, P.W.; Fitzgerald, G.F.; Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef] [PubMed]
- Cui, Y.; Miao, K.; Niyaphorn, S.; Qu, X. Production of Gamma-Aminobutyric Acid from Lactic Acid Bacteria: A Systematic Review. Int. J. Mol. Sci. 2020, 21, 995. [Google Scholar] [CrossRef] [Green Version]
- Baral, P.; Umans, B.D.; Li, L.; Wallrapp, A.; Bist, M.; Kirschbaum, T.; Wei, Y.; Zhou, Y.; Kuchroo, V.K.; Burkett, P.R.; et al. Author Correction: Nociceptor sensory neurons suppress neutrophil and γδ T cell responses in bacterial lung infections and lethal pneumonia. Nat. Med. 2018, 24, 1625–1626. [Google Scholar] [CrossRef]
- Harvard Medical School. “Nerve Cells Found to Suppress Immune Response during Deadly Lung Infections: Findings Highlight Importance of Nervous System as Target for Non-Antibiotic Therapies That Boost Lung Immunity, Promote Healing.” Science Daily. Available online: www.sciencedaily.com/releases/2018/03/180305111520.htm (accessed on 12 April 2023).
- 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]
- Scott, G.A.; Terstege, D.J.; Vu, A.P.; Law, S.; Evans, A.; Epp, J.R. Disrupted Neurogenesis in Germ-Free Mice: Effects of Age and Sex. Front. Cell Dev. Biol. 2020, 8, 407. [Google Scholar] [CrossRef] [PubMed]
- Sampson, T.R.; Mazmanian, S.K. Control of Brain Development, Function, and Behavior by the Microbiome. Cell Host Microbe 2015, 17, 565–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mayer, E.A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Investig. 2015, 125, 926–938. [Google Scholar] [CrossRef] [Green Version]
- Barbara, G.; Stanghellini, V.; Brandi, G.; Cremon, C.; Di Nardo, G.; De Giorgio, R.; Corinaldesi, R. Interactions Between Commensal Bacteria and Gut Sensorimotor Function in Health and Disease. Am. J. Gastroenterol. 2005, 100, 2560–2568. [Google Scholar] [CrossRef]
- Ward, J.B.; Keely, S.J.; Keely, S.J. Oxygen in the regulation of intestinal epithelial transport. J. Physiol. 2014, 592, 2473–2489. [Google Scholar] [CrossRef]
- Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
- Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular Analysis of Commensal Host-Microbial Relationships in the Intestine. Science 2001, 291, 881–884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neufeld, K.A.; Kang, N.; Bienenstock, J.; Foster, J.A. Effects of intestinal microbiota on anxiety-like behavior. Commun. Integr. Biol. 2011, 4, 492–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bercik, P.; Park, A.J.; Sinclair, D.; Khoshdel, A.; Lu, J.; Huang, X.; Deng, Y.; Blennerhassett, P.A.; Fahnestock, M.; Moine, D.; et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol. Motil. 2011, 23, 1132–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tian, P.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Bifidobacterium with the role of 5-hydroxytryptophan synthesis regulation alleviates the symptom of depression and related microbiota dysbiosis. J. Nutr. Biochem. 2019, 66, 43–51. [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] [Green Version]
- Yunes, R.A.; Poluektova, E.U.; Vasileva, E.V.; Odorskaya, M.V.; Marsova, M.V.; Kovalev, G.I.; Danilenko, V.N. A Multi-strain Potential Probiotic Formulation of GABA-Producing Lactobacillus plantarum 90sk and Bifidobacterium adolescentis 150 with Antidepressant Effects. Probiotics Antimicrob. Proteins 2020, 12, 973–979. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Xu, F.; Nie, Z.; Shao, L. Gut Microbiota Approach—A New Strategy to Treat Parkinson’s Disease. Front. Cell. Infect. Microbiol. 2020, 10, 570658. [Google Scholar] [CrossRef]
- Villageliú, D.; Lyte, M. Dopamine production in Enterococcus faecium: A microbial endocrinology-based mechanism for the selection of probiotics based on neurochemical-producing potential. PLoS ONE 2018, 13, e0207038. [Google Scholar] [CrossRef] [PubMed]
- Johnson, D.; Thurairajasingam, S.; Letchumanan, V.; Chan, K.G.; Lee, L.H. Exploring the Role and Potential of Probiotics in the Field of Mental Health: Major Depressive Disorder. Nutrients 2021, 13, 1728. [Google Scholar] [CrossRef] [PubMed]
- Ait-Belgnaoui, A.; Colom, A.; Braniste, V.; Ramalho, L.; Marrot, A.; Cartier, C.; Houdeau, E.; Theodorou, V.; Tompkins, T. Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterol. Motil. 2014, 26, 510–520. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Dowd, S.E.; Scurlock, B.; Acosta-Martinez, V.; Lyte, M. Memory and learning behavior in mice is temporally associated with diet-induced alterations in gut bacteria. Physiol. Behav. 2009, 96, 557–567. [Google Scholar] [CrossRef]
- Charitos, I.A.; D’agostino, D.; Topi, S.; Bottalico, L. 40 Years of Helicobacter pylori: A Revolution in Biomedical Thought. Gastroenterol. Insights 2021, 12, 111–135. [Google Scholar] [CrossRef]
- Uribe, A.; Alam, M.; Johansson, O.; Midtvedt, T.; Theodorsson, E. Microflora modulates endocrine cells in the gastrointestinal mucosa of the rat. Gastroenterology 1994, 107, 1259–1269. [Google Scholar] [CrossRef]
- Taché, Y.; Stephens, R.L., Jr.; Ishikawa, T. Central Nervous System Action of TRH to Influence Gastrointestinal Function and Ulceration. Ann. N. Y. Acad. Sci. 1989, 553, 269–285. [Google Scholar] [CrossRef]
- Lee, S.H.; Kwon, J.E.; Cho, M.L. Immunological pathogenesis of inflammatory bowel disease. Intest. Res. 2018, 16, 26–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verdu, E.F.; Bercik, P.; Verma-Gandhu, M.; Huang, X.X.; Blennerhassett, P.; Jackson, W.; Mao, Y.; Wang, L.; Rochat, F.; Collins, S.M. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut 2006, 55, 182–190. [Google Scholar] [CrossRef] [Green Version]
- Altaf, M.A.; Sood, M.R. The nervous system and gastrointestinal function. Dev. Disabil. Res. Rev. 2008, 14, 87–95. [Google Scholar] [CrossRef] [PubMed]
- Ntranos, A.; Casaccia, P. The Microbiome–Gut–Behavior Axis: Crosstalk between the Gut Microbiome and Oligodendrocytes Modulates Behavioral Responses. Neurotherapeutics 2018, 15, 31–35. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Sun, K.; Wu, Y.; Yang, Y.; Tso, P.; Wu, Z. Interactions between Intestinal Microbiota and Host Immune Response in Inflammatory Bowel Disease. Front. Immunol. 2017, 8, 942. [Google Scholar] [CrossRef] [Green Version]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
- Montiel-Castro, A.J.; González-Cervantes, R.M.; Bravo-Ruiseco, G.; Pacheco-López, G. The microbiota-gut-brain axis: Neurobehavioral correlates, health and sociality. Front. Integr. Neurosci. 2013, 7, 70. [Google Scholar] [CrossRef] [Green Version]
- Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The microbiome, cancer, and cancer therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef]
- Sarubbo, F.; Cavallucci, V.; Pani, G. The Influence of Gut Microbiota on Neurogenesis: Evidence and Hopes. Cells 2022, 11, 382. [Google Scholar] [CrossRef] [PubMed]
- Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr. Res. 2019, 85, 216–224. [Google Scholar] [CrossRef] [Green Version]
- Costantino, C.M.; Baecher-Allan, C.; Hafler, D.A. Multiple Sclerosis and Regulatory T Cells. J. Clin. Immunol. 2008, 28, 697–706. [Google Scholar] [CrossRef]
- Fletcher, J.M.; Lalor, S.J.; Sweeney, C.M.; Tubridy, N.; Mills, K.H. T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin. Exp. Immunol. 2010, 162, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Duncan, I.D.; Radcliff, A.B. Inherited and acquired disorders of myelin: The underlying myelin pathology. Exp. Neurol. 2016, 283 Pt B, 452–475. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Schepici, G.; Silvestro, S.; Bramanti, P.; Mazzon, E. The Gut Microbiota in Multiple Sclerosis: An Overview of Clinical Trials. Cell Transplant. 2019, 28, 1507–1527. [Google Scholar] [CrossRef] [Green Version]
- Fretheim, H.; Chung, B.K.; Didriksen, H.; Bækkevold, E.S.; Midtvedt, Ø.; Brunborg, C.; Holm, K.; Valeur, J.; Tennøe, A.H.; Garen, T.; et al. Fecal microbiota transplantation in systemic sclerosis: A double-blind, placebo-controlled randomized pilot trial. PLoS ONE 2020, 15, e0232739. [Google Scholar] [CrossRef]
- Demaude, J.; Salvador-Cartier, C.; Fioramonti, J.; Ferrier, L.; Bueno, L. Phenotypic changes in colonocytes following acute stress or activation of mast cells in mice: Implications for delayed epithelial barrier dysfunction. Gut 2006, 55, 655–661. [Google Scholar] [CrossRef] [PubMed]
- Sambucci, M.; Gargano, F.; Guerrera, G.; Battistini, L.; Borsellino, G. One, No One, and One Hundred Thousand: T Regulatory Cells’ Multiple Identities in Neuroimmunity. Front. Immunol. 2019, 10, 2947. [Google Scholar] [CrossRef]
- Varghese, A.K.; Verdú, E.F.; Bercik, P.; Khan, W.I.; Blennerhassett, P.A.; Szechtman, H.; Collins, S.M. Antidepressants Attenuate Increased Susceptibility to Colitis in a Murine Model of Depression. Gastroenterology 2006, 130, 1743–1753. [Google Scholar] [CrossRef] [Green Version]
- Tremlett, H.; Fadrosh, D.W.; Faruqi, A.A.; Hart, J.; Roalstad, S.; Graves, J.; Spencer, C.M.; Lynch, S.V.; Zamvil, S.S.; Waubant, E.; et al. Associations between the gut microbiota and host immune markers in pediatric multiple sclerosis and controls. BMC Neurol. 2016, 16, 182. [Google Scholar] [CrossRef] [Green Version]
- Brahic, M. Multiple sclerosis and viruses. Ann. Neurol. 2010, 68, 6–8. [Google Scholar] [CrossRef]
- Van den Hoogen, W.J.; Laman, J.D.; Hart, B.A. Modulation of Multiple Sclerosis and Its Animal Model Experimental Autoimmune Encephalomyelitis by Food and Gut Microbiota. Front. Immunol. 2017, 8, 1081. [Google Scholar] [CrossRef] [Green Version]
- Sharma, M.; Li, Y.; Stoll, M.L.; Tollefsbol, T.O. The Epigenetic Connection Between the Gut Microbiome in Obesity and Diabetes. Front. Genet. 2020, 10, 1329. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Kasper, L.H. The role of microbiome in central nervous system disorders. Brain, Behav. Immun. 2014, 38, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Ochoa-Repáraz, J.; Mielcarz, D.W.; Ditrio, L.E.; Burroughs, A.R.; Begum-Haque, S.; Dasgupta, S.; Kasper, D.L.; Kasper, L.H. Central Nervous System Demyelinating Disease Protection by the Human Commensal Bacteroides fragilis Depends on Polysaccharide A Expression. J. Immunol. 2010, 185, 4101–4108. [Google Scholar] [CrossRef] [Green Version]
- Banati, M.; Csecsei, P.; Koszegi, E.; Nielsen, H.H.; Suto, G.; Bors, L.; Trauninger, A.; Csepany, T.; Rozsa, C.; Jakab, G.; et al. Antibody response against gastrointestinal antigens in demyelinating diseases of the central nervous system. Eur. J. Neurol. 2013, 20, 1492–1495. [Google Scholar] [CrossRef]
- Preiningerova, J.L.; Zakostelska, Z.J.; Srinivasan, A.; Ticha, V.; Kovarova, I.; Kleinova, P.; Tlaskalova-Hogenova, H.; Havrdova, E.K. Multiple Sclerosis and Microbiome. Biomolecules 2022, 12, 433. [Google Scholar] [CrossRef]
- Bhargava, P.; Anthony, D.C. Metabolomics in multiple sclerosis disease course and progression. Mult. Scler. J. 2020, 26, 591–598. [Google Scholar] [CrossRef] [PubMed]
- Wilkins, J.M.; Trushina, E. Application of Metabolomics in Alzheimer’s Disease. Front. Neurol. 2018, 8, 719. [Google Scholar] [CrossRef] [Green Version]
- Sarlus, H.; Heneka, M.T. Microglia in Alzheimer’s disease. J. Clin. Investig. 2017, 127, 3240–3249. [Google Scholar] [CrossRef] [Green Version]
- Rajmohan, R.; Reddy, P.H. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer’s disease Neurons. J. Alzheimer’s Dis. 2017, 57, 975–999. [Google Scholar] [CrossRef] [Green Version]
- Bachurin, S.O.; Gavrilova, S.I.; Samsonova, A.; Barreto, G.E.; Aliev, G. Mild cognitive impairment due to Alzheimer disease: Contemporary approaches to diagnostics and pharmacological intervention. Pharmacol. Res. 2018, 129, 216–226. [Google Scholar] [CrossRef]
- Papassotiropoulos, A.; Lambert, J.-C.; Vrièze, F.W.-D.; Wollmer, M.A.; von der Kammer, H.; Streffer, J.R.; Maddalena, A.; Huynh, K.-D.; Wolleb, S.; Lütjohann, D.; et al. Cholesterol 25-Hydroxylase on Chromosome 10q Is a Susceptibility Gene for Sporadic Alzheimer’s Disease. Neurodegener. Dis. 2005, 2, 233–241. [Google Scholar] [CrossRef] [Green Version]
- Lim, C.; Hammond, C.J.; Hingley, S.T.; Balin, B.J. Chlamydia pneumoniae infection of monocytes in vitro stimulates innate and adaptive immune responses relevant to those in Alzheimer’s disease. J. Neuroinflamm. 2014, 11, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bulgart, H.R.; Neczypor, E.W.; Wold, L.E.; Mackos, A.R. Microbial involvement in Alzheimer disease development and progression. Mol. Neurodegener. 2020, 15, 42. [Google Scholar] [CrossRef]
- Sun, P.; Su, L.; Zhu, H.; Li, X.; Guo, Y.; Du, X.; Zhang, L.; Qin, C. Gut Microbiota Regulation and Their Implication in the Development of Neurodegenerative Disease. Microorganisms 2021, 9, 2281. [Google Scholar] [CrossRef] [PubMed]
- Zhan, X.; Stamova, B.; Sharp, F.R. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer’s Disease Brain: A Review. Front. Aging Neurosci. 2018, 10, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leblhuber, F.; Ehrlich, D.; Steiner, K.; Geisler, S.; Fuchs, D.; Lanser, L.; Kurz, K. The Immunopathogenesis of Alzheimer’s Disease Is Related to the Composition of Gut Microbiota. Nutrients 2021, 13, 361. [Google Scholar] [CrossRef] [PubMed]
- Oriach, C.S.; Robertson, R.C.; Stanton, C.; Cryan, G.F.; Dinan, T.G. Food for thought: The role of nutrition in the microbiota-gut–brain axis. Clin. Nutr. Exp. 2016, 6, 25–38. [Google Scholar] [CrossRef] [Green Version]
- Arrigoni, R.; Ballini, A.; Santacroce, L.; Cantore, S.; Inchingolo, A.; Inchingolo, F.; Di Domenico, M.; Quagliuolo, L.; Boccellino, M. Another look at dietary polyphenols: Challenges in cancer prevention and treatment. Curr. Med. Chem. 2022, 29, 1061–1082. [Google Scholar] [CrossRef]
- Harach, T.; Marungruang, N.; Duthilleul, N.; Cheatham, V.; Mc Coy, K.D.; Frisoni, G.; Neher, J.J.; Fåk, F.; Jucker, M.; Lasser, T.; et al. Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci. Rep. 2017, 7, 41802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, Z.; Hussain, M.D.; Yan, L.J. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 2014, 124, 307–321. [Google Scholar] [CrossRef] [PubMed]
- Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609–10620. [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]
- LeWitt, P.A.; Li, J.; Lu, M.; Guo, L.; Auinger, P. Metabolomic biomarkers as strong correlates of Parkinson disease progression. Neurology 2017, 88, 862–869. [Google Scholar] [CrossRef] [Green Version]
- Samii, A.; Nutt, J.G.; Ransom, B.R. Parkinson’s disease. Lancet 2004, 363, 1783–1793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schneider, S.A.; Alcalay, R.N. Neuropathology of genetic synucleinopathies with parkinsonism: Review of the literature. Mov. Disord. 2017, 32, 1504–1523. [Google Scholar] [CrossRef]
- Lebouvier, T.; Chaumette, T.; Paillusson, S.; Duyckaerts, C.; Bruley des Varannes, S.; Neunlist, M.; Derkinderen, P. The second brain and Parkinson’s disease. Eur. J. Neurosci. 2009, 30, 735–741. [Google Scholar] [CrossRef]
- Natale, G.; Pasquali, L.; Paparelli, A.; Fornai, F. Parallel manifestations of neuropathologies in the enteric and central nervous systems. Neurogastroenterol. Motil. 2011, 23, 1056–1065. [Google Scholar] [CrossRef]
- Di Domenico, M.; Ballini, A.; Boccellino, M.; Scacco, S.; Lovero, R.; Charitos, I.A.; Santacroce, L. The Intestinal Microbiota May Be a Potential Theranostic Tool for Personalized Medicine. J. Pers. Med. 2022, 12, 523. [Google Scholar] [CrossRef]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e12. [Google Scholar] [CrossRef] [Green Version]
- Lubomski, M.; Tan, A.H.; Lim, S.Y.; Holmes, A.J.; Davis, R.L.; Sue, C.M. Parkinson’s disease and the gastrointestinal microbiome. J. Neurol. 2020, 267, 2507–2523. [Google Scholar] [CrossRef]
- Maes, M.; Yirmyia, R.; Noraberg, J.; Brene, S.; Hibbeln, J.; Perini, G.; Kubera, M.; Bob, P.; Lerer, B.; Maj, M. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: Leads for future research and new drug developments in depression. Metab. Brain Dis. 2009, 24, 27–53. [Google Scholar] [CrossRef]
- Daniele, S.G.; Béraud, D.; Davenport, C.; Cheng, K.; Yin, H.; Maguire-Zeiss, K.A. Activation of MyD88-dependent TLR1/2 signaling by misfolded α-synuclein, a protein linked to neurodegenerative disorders. Sci. Signal. 2015, 8, ra45. [Google Scholar] [CrossRef] [Green Version]
- Giloteaux, L.; Goodrich, J.K.; Walters, W.A.; Levine, S.M.; Ley, R.E.; Hanson, M.R. Reduced diversity and altered composition of the gut microbiome in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome 2016, 4, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Barichella, M.; Severgnini, M.; Cilia, R.; Cassani, E.; Bolliri, C.; Caronni, S.; Ferri, V.; Cancello, R.; Ceccarani, C.; Faierman, S.; et al. Unraveling gut microbiota in Parkinson’s disease and atypical parkinsonism. Mov. Disord. 2019, 34, 396–405. [Google Scholar] [CrossRef]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sullivan, A.; Nord, C.E.; Evengård, B. Effect of supplement with lactic-acid producing bacteria on fatigue and physical activity in patients with chronic fatigue syndrome. Nutr. J. 2009, 8, 4. [Google Scholar] [CrossRef] [Green Version]
- Twisk, F.N. Accurate diagnosis of myalgic encephalomyelitis and chronic fatigue syndrome based upon objective test methods for characteristic symptoms. World J. Methodol. 2015, 5, 68–87. [Google Scholar] [CrossRef] [Green Version]
- Aaron, L.A.; Burke, M.M.; Buchwald, D. Overlapping Conditions Among Patients with Chronic Fatigue Syndrome, Fibromyalgia, and Temporomandibular Disorder. Arch. Intern. Med. 2000, 160, 221–227. [Google Scholar] [CrossRef]
- Naviaux, R.K.; Naviaux, J.C.; Li, K.; Bright, A.T.; Alaynick, W.A.; Wang, L.; Baxter, A.; Nathan, N.; Anderson, W.; Gordon, E. Metabolic features of chronic fatigue syndrome. Proc. Natl. Acad. Sci. USA 2016, 113, E5472–E5480. [Google Scholar] [CrossRef] [Green Version]
- Vandeputte, D.; Falony, G.; Vieira-Silva, S.; Tito, R.Y.; Joossens, M.; Raes, J. Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut 2016, 65, 57–62. [Google Scholar] [CrossRef] [Green Version]
- Sperber, A.D.; Dekel, R. Irritable Bowel Syndrome and Co-morbid Gastrointestinal and Extra-gastrointestinal Functional Syndromes. J. Neurogastroenterol. Motil. 2010, 16, 113–119. [Google Scholar] [CrossRef] [Green Version]
- Yang, T.; Yang, Y.; Wang, D.; Li, C.; Qu, Y.; Guo, J.; Shi, T.; Bo, W.; Sun, Z.; Asakawa, T. The clinical value of cytokines in chronic fatigue syndrome. J. Transl. Med. 2019, 17, 213. [Google Scholar] [CrossRef]
- Lupo, G.; Rocchetti, G.; Lucini, L.; Lorusso, L.; Manara, E.; Bertelli, M.; Puglisi, E.; Capelli, E. Potential role of microbiome in Chronic Fatigue Syndrome/Myalgic Encephalomyelits (CFS/ME). Sci. Rep. 2021, 11, 7043. [Google Scholar] [CrossRef]
- Berstad, A.; Hauso, O.; Berstad, K.; Berstad, J.E. From IBS to ME—The dysbiotic march hypothesis. Med. Hypotheses 2020, 140, 109648. [Google Scholar] [CrossRef]
- Ríos-Covián, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; De Los Reyes-Gavilán, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef] [Green Version]
- König, R.S.; Albrich, W.C.; Kahlert, C.R.; Bahr, L.S.; Löber, U.; Vernazza, P.; Scheibenbogen, C.; Forslund, S.K. The Gut Microbiome in Myalgic Encephalomyelitis (ME)/Chronic Fatigue Syndrome (CFS). Front. Immunol. 2022, 12, 628741. [Google Scholar] [CrossRef]
- Varesi, A.; Deumer, U.S.; Ananth, S.; Ricevuti, G. The Emerging Role of Gut Microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Current Evidence and Potential Therapeutic Applications. J. Clin. Med. 2021, 10, 5077. [Google Scholar] [CrossRef]
- Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr. Med. 2018, 17, 28–32. [Google Scholar]
- Bay-Richter, C.; Janelidze, S.; Hallberg, L.; Brundin, L. Changes in behaviour and cytokine expression upon a peripheral immune challenge. Behav Brain Res. 2011, 222, 193–199. [Google Scholar] [CrossRef]
- Costa, L.N.F.G.; Carneiro, B.A.; Alves, G.S.; Silva, D.H.L.; Guimaraes, D.F.; Souza, L.S.; Bandeira, I.D.; Beanes, G.; Scippa, A.M.; Quarantini, L.C. Metabolomics of Major Depressive Disorder: A Systematic Review of Clinical Studies. Cureus 2022, 14, e23009. [Google Scholar] [CrossRef]
- Lukić, 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] [Green Version]
- Holingue, C.; Budavari, A.C.; Rodriguez, K.M.; Zisman, C.R.; Windheim, G.; Fallin, M.D. Sex Differences in the Gut-Brain Axis: Implications for Mental Health. Curr. Psychiatry Rep. 2020, 22, 83. [Google Scholar] [CrossRef] [PubMed]
- Peirce, J.M.; Alviña, K. The role of inflammation and the gut microbiome in depression and anxiety. J. Neurosci. Res. 2019, 97, 1223–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ortega, M.A.; Alvarez-Mon, M.A.; García-Montero, C.; Fraile-Martinez, O.; Guijarro, L.G.; Lahera, G.; Monserrat, J.; Valls, P.; Mora, F.; Rodríguez-Jiménez, R.; et al. Gut Microbiota Metabolites in Major Depressive Disorder—Deep Insights into Their Pathophysiological Role and Potential Translational Applications. Metabolites 2022, 12, 50. [Google Scholar] [CrossRef]
- Charitos, I.A.; Castellaneta, F.; Santacroce, L.; Bottalico, L. Historical Anecdotes and Breakthroughs of Histamine: From Discovery to Date. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 801–814. [Google Scholar] [CrossRef]
- Perrone, M.G.; Centonze, A.; Miciaccia, M.; Ferorelli, S.; Scilimati, A. Cyclooxygenase Inhibition Safety and Efficacy in Inflammation-Based Psychiatric Disorders. Molecules 2020, 25, 5388. [Google Scholar] [CrossRef]
- Galland, L. The Gut Microbiome and the Brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef] [Green Version]
- Odenwald, M.A.; Turner, J.R. Intestinal Permeability Defects: Is It Time to Treat? Clin. Gastroenterol. Hepatol. 2013, 11, 1075–1083. [Google Scholar] [CrossRef] [Green Version]
- Bystritsky, A.; Khalsa, S.S.; Cameron, M.E.; Schiffman, J. Current diagnosis and treatment of anxiety disorders. Pharm. Ther. 2013, 38, 30–57. [Google Scholar]
- Qin, H.-Y.; Cheng, C.-W.; Tang, X.-D.; Bian, Z.-X. Impact of psychological stress on irritable bowel syndrome. World J. Gastroenterol. 2014, 20, 14126–14131. [Google Scholar] [CrossRef] [PubMed]
- Morris, G.; Berk, M.; Carvalho, A.; Caso, J.R.; Sanz, Y.; Walder, K.; Maes, M. The Role of the Microbial Metabolites Including Tryptophan Catabolites and Short Chain Fatty Acids in the Pathophysiology of Immune-Inflammatory and Neuroimmune Disease. Mol. Neurobiol. 2017, 54, 4432–4451. [Google Scholar] [CrossRef]
- Smith, A.M.; Natowicz, M.R.; Braas, D.; Ludwig, M.A.; Ney, D.M.; Donley, E.L.R.; Burrier, R.E.; Amaral, D.G. A Metabolomics Approach to Screening for Autism Risk in the Children’s Autism Metabolome Project. Autism Res. 2020, 13, 1270–1285. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Han, Y.; Dy, A.B.C.; Hagerman, R.J. The Gut Microbiota and Autism Spectrum Disorders. Front. Cell. Neurosci. 2017, 11, 120. [Google Scholar] [CrossRef] [Green Version]
- Iglesias-Vázquez, L.; Van Ginkel Riba, G.; Arija, V.; Canals, J. Composition of Gut Microbiota in Children with Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macfabe, D. Autism: Metabolism, Mitochondria, and the Microbiome. Glob. Adv. Health Med. 2013, 2, 52–66. [Google Scholar] [CrossRef] [Green Version]
- Israelyan, N.; Margolis, K.G. Serotonin as a link between the gut-brain-microbiome axis in autism spectrum disorders. Pharmacol. Res. 2018, 132, 1–6. [Google Scholar] [CrossRef]
- Gabriele, S.; Sacco, R.; Persico, A.M. Blood serotonin levels in autism spectrum disorder: A systematic review and meta-analysis. Eur. Neuropsychopharmacol. 2014, 24, 919–929. [Google Scholar] [CrossRef]
- Louis, P. Does the Human Gut Microbiota Contribute to the Etiology of Autism Spectrum Disorders? Dig. Dis. Sci. 2012, 57, 1987–1989. [Google Scholar] [CrossRef]
- Adams, J.B.; Johansen, L.J.; Powell, L.D.; Quig, D.; Rubin, R.A. Gastrointestinal flora and gastrointestinal status in children with autism–Comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011, 11, 22. [Google Scholar] [CrossRef] [Green Version]
- Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota Modulate Behavioral and Physiological Abnormalities Associated with Neurodevelopmental Disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mitchison, D.; Hay, P.; Slewa-Younan, S.; Mond, J. Time Trends in Population Prevalence of Eating Disorder Behaviors and Their Relationship to Quality of Life. PLoS ONE 2012, 7, e48450. [Google Scholar] [CrossRef] [Green Version]
- Morita, C.; Tsuji, H.; Hata, T.; Gondo, M.; Takakura, S.; Kawai, K.; Yoshihara, K.; Ogata, K.; Nomoto, K.; Miyazaki, K.; et al. Gut Dysbiosis in Patients with Anorexia Nervosa. PLoS ONE 2015, 10, e0145274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wisse, B.E.; Ogimoto, K.; Tang, J.; Harris, M.K., Jr.; Raines, E.W.; Schwartz, M.W. Evidence that Lipopolysaccharide-Induced Anorexia Depends upon Central, Rather than Peripheral, Inflammatory Signals. Endocrinology 2007, 148, 5230–5237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santacroce, L.; Charitos, I.A.; Bottalico, L. A successful history: Probiotics and their potential as antimicrobials. Expert Rev. Anti-Infect. Ther. 2019, 17, 635–645. [Google Scholar] [CrossRef] [PubMed]
- Santacroce, L.; Charitos, I.A.; Gagliano-Candela, R.; Bottalico, L. Venoms and Poisonings during the Centuries: A Narrative Review. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 558–570. [Google Scholar] [CrossRef]
- Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. Int. Rev. J. 2019, 10 (Suppl. S1), S49–S66, Erratum in Adv Nutr. 2020, 11, 1054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, N.; Ju, Z.; Zuo, T. Time for food: The impact of diet on gut microbiota and human health. Nutrition 2018, 51–52, 80–85. [Google Scholar] [CrossRef]
- Bottalico, L.; Charitos, I.A.; Potenza, M.A.; Montagnani, M.; Santacroce, L. The war against bacteria, from the past to present and beyond. Expert Rev. Anti-Infect. Ther. 2021, 20, 681–706. [Google Scholar] [CrossRef]
- Charitos, I.A.; Gagliano-Candela, R.; Santacroce, L.; Bottalico, L. The Cannabis Spread throughout the Continents and its Therapeutic Use in History. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 407–417. [Google Scholar] [CrossRef]
- Crocetto, F.; Boccellino, M.; Barone, B.; Di Zazzo, E.; Sciarra, A.; Galasso, G.; Settembre, G.; Quagliuolo, L.; Imbimbo, C.; Boffo, S.; et al. The Crosstalk between Prostate Cancer and Microbiota Inflammation: Nutraceutical Products Are Useful to Balance This Interplay? Nutrients 2020, 12, 2648. [Google Scholar] [CrossRef]
- Polimeno, L.; Barone, M.; Mosca, A.; Viggiani, M.T.; Joukar, F.; Mansour-Ghanaei, F.; Mavaddati, S.; Daniele, A.; Debellis, L.; Bilancia, M.; et al. Soy Metabolism by Gut Microbiota from Patients with Precancerous Intestinal Lesions. Microorganisms 2020, 8, 469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allen, M.M.; Pike, O.A.; Kenealey, J.D.; Dunn, M.L. Metabolomics of acid whey derived from Greek yogurt. J. Dairy Sci. 2021, 104, 11401–11412. [Google Scholar] [CrossRef] [PubMed]
- Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef] [Green Version]
- Santacroce, L. Letter in response to the article “Enhancing immunity in viral infections, with special emphasis on COVID-19: A review” (Jayawardena et al.). Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 927. [Google Scholar] [CrossRef]
- Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The Pervasive Effects of an Antibiotic on the Human Gut Microbiota, as Revealed by Deep 16S rRNA Sequencing. PLoS Biol. 2008, 6, e280. [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 andBifidobacterium longum R0175) in healthy human volunteers. Gut Microbes 2011, 2, 256–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X.; et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786–796. [Google Scholar] [CrossRef]
- Gareau, M.G.; Jury, J.; MacQueen, G.; Sherman, P.M.; Perdue, M.H. Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut 2007, 56, 1522–1528. [Google Scholar] [CrossRef] [Green Version]
- de Moreno de LeBlanc, A.; LeBlanc, J.G. Effect of probiotic administration on the intestinal microbiota, current knowledge and potential applications. World J. Gastroenterol. 2014, 20, 16518–16528. [Google Scholar] [CrossRef]
- Johnson, D.; Letchumanan, V.; Thum, C.C.; Thurairajasingam, S.; Lee, L.H. A Microbial-Based Approach to Mental Health: The Potential of Probiotics in the Treatment of Depression. Nutrients 2023, 15, 1382. [Google Scholar] [CrossRef]
- Cohen Kadosh, K.; Basso, M.; Knytl, P.; Johnstone, N.; Lau, J.Y.F.; Gibson, G.R. Psychobiotic interventions for anxiety in young people: A systematic review and meta-analysis, with youth consultation. Transl. Psychiatry 2021, 11, 352. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W. Psychobiotics and the Manipulation of Bacteria–Gut–Brain Signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
- Manning, T.S.; Gibson, G.R. Prebiotics. Best Pract. Res. Clin. Gastroenterol. 2004, 18, 287–298. [Google Scholar] [CrossRef]
- Ford, A.C.; Quigley, E.M.; Lacy, B.E.; Lembo, A.J.; Saito, Y.A.; Schiller, L.R.; Soffer, E.E.; Spiegel, B.M.; Moayyedi, P. Efficacy of Prebiotics, Probiotics, and Synbiotics in Irritable Bowel Syndrome and Chronic Idiopathic Constipation: Systematic Review and Meta-analysis. Am. J. Gastroenterol. 2014, 109, 1547–1561. [Google Scholar] [CrossRef] [PubMed]
- Borody, T.J.; Brandt, L.J.; Paramsothy, S. Therapeutic faecal microbiota transplantation: Current status and future developments. Curr. Opin. Gastroenterol. 2014, 30, 97–105. [Google Scholar] [CrossRef] [Green Version]
- Paramsothy, S.; Kamm, M.A.; Kaakoush, N.O.; Walsh, A.J.; van den Bogaerde, J.; Samuel, D.; Leong, R.W.L.; Connor, S.; Ng, W.; Paramsothy, R.; et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: A randomised placebo-controlled trial. Lancet 2017, 389, 1218–1228. [Google Scholar] [CrossRef]
- Santacroce, L.; Mavaddati, S.; Hamedi, J.; Zeinali, B.; Ballini, A.; Bilancia, M. Expressive Analysis of Gut Microbiota in Pre- and Post-Solid Organ Transplantation Using Bayesian Topic Models Computational Science and Its Applications—ICCSA 2020; Springer: Cham, Switzerland, 2020; pp. 150–165. [Google Scholar] [CrossRef]
- Legette, L.L.; Lee, W.; Martin, B.R.; Story, J.A.; Campbell, J.K.; Weaver, C.M. Prebiotics Enhance Magnesium Absorption and Inulin-based Fibers Exert Chronic Effects on Calcium Utilization in a Postmenopausal Rodent Model. J. Food Sci. 2012, 77, 88–94. [Google Scholar] [CrossRef] [PubMed]
- Scholz-Ahrens, K.E.; Ade, P.; Marten, B.; Weber, P.; Timm, W.; Asil, Y.; Glüer, C.-C.; Schrezenmeir, J. Prebiotics, Probiotics, and Synbiotics Affect Mineral Absorption, Bone Mineral Content, and Bone Structure. J. Nutr. 2007, 137 (Suppl. S2), 838S–846S. [Google Scholar] [CrossRef]
- Man, A.; Mare, A.; Toma, F.; Curticăpean, A.; Santacroce, L. Health Threats from Contamination of Spices Commercialized in Romania: Risks of Fungal and Bacterial Infections. Endocr. Metab. Immune Disord. Drug Targets 2016, 16, 197–204. [Google Scholar] [CrossRef]
- Yoo, J.Y.; Kim, S.S. Probiotics and Prebiotics: Present Status and Future Perspectives on Metabolic Disorders. Nutrients 2016, 8, 173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klaenhammer, T.R.; Kullen, M.J. Selection and design of probiotics. Int. J. Food Microbiol. 1999, 50, 45–57. [Google Scholar] [CrossRef] [PubMed]
- Dey, G.; Mookherjee, S. Probiotics-targeting new milestones from gut health to mental health. FEMS Microbiol. Lett. 2021, 368, fnab096. [Google Scholar] [CrossRef] [PubMed]
Species/Phyla | Neurotransmitters |
---|---|
Bacillus, Escherichia, Saccharomyces | Serotonin, Noradrenaline [76,94] |
Candida, Bacillus, Escherichia spp., Enterococcus, Streptococcus | Dopamine, Noradrenaline, Serotonin [95] |
Lactobacillaceae | Acetylcholine [96] |
Bifidobacterium infantis | Tryptophan (5-HT) [97] |
Bifidobacterium, Lactobacillaceae | γ-aminobutyrate (GABA) [98,99] |
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 authors. 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
Montagnani, M.; Bottalico, L.; Potenza, M.A.; Charitos, I.A.; Topi, S.; Colella, M.; Santacroce, L. The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome. Int. J. Mol. Sci. 2023, 24, 10322. https://doi.org/10.3390/ijms241210322
Montagnani M, Bottalico L, Potenza MA, Charitos IA, Topi S, Colella M, Santacroce L. The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome. International Journal of Molecular Sciences. 2023; 24(12):10322. https://doi.org/10.3390/ijms241210322
Chicago/Turabian StyleMontagnani, Monica, Lucrezia Bottalico, Maria Assunta Potenza, Ioannis Alexandros Charitos, Skender Topi, Marica Colella, and Luigi Santacroce. 2023. "The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome" International Journal of Molecular Sciences 24, no. 12: 10322. https://doi.org/10.3390/ijms241210322
APA StyleMontagnani, M., Bottalico, L., Potenza, M. A., Charitos, I. A., Topi, S., Colella, M., & Santacroce, L. (2023). The Crosstalk between Gut Microbiota and Nervous System: A Bidirectional Interaction between Microorganisms and Metabolome. International Journal of Molecular Sciences, 24(12), 10322. https://doi.org/10.3390/ijms241210322