Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain–Gut–Microbiome Axis: A Focus on Sex Differences
Abstract
:1. Introduction
1.1. Overview of Tryptophan Metabolism
1.2. Overview of the Brain–Gut–Microbiome Axis
1.2.1. Microbiome
1.2.2. Communication Using Neural and Circulating Signals
Neural Signals
- Efferent neural signals
- Afferent neural signals
Circulating Signals
- Efferent circulating signals
- Afferent circulating signals
1.2.3. Experimental Strategies in Tryptophan Metabolite and Gut Microbiome Research
1.3. Aim and Focus of This Review
2. Tryptophan Metabolism in the Brain–Gut–Microbiome Axis
2.1. Overview of Tryptophan and Tryptophan Metabolism
2.2. Tryptophan Metabolism Within the Microbiota–Gut–Brain Axis
2.2.1. Various Tryptophan Metabolites Synthesized from Multiple Pathways
2.2.2. Various Neuroactive Tryptophan Metabolites
2.2.3. 5-HT Synthesis
2.2.4. 5-HT Synthesis in the Gut by Gut Microbiota
2.2.5. 5-HT Synthesis in the Gut Regulated by Microbial Metabolites
2.3. Sex Differences and Roles of Estrogen in Tryptophan Metabolism
2.3.1. Hormone Regulation of Tryptophan Metabolism
2.3.2. Hormone Regulation of 5-HT Synthesis
2.4. Sex Differences and Roles of Estrogen in Gut Microbiota
2.4.1. Sex Specific Alterations in Gut Microbiota in Response to Obesogenic Diet
2.4.2. Sex Specific Alterations in Gut Microbiota Due to Estrogens
3. Functional Roles of Tryptophan Metabolites in the Brain–Gut–Microbiome Axis
3.1. Brain Tryptophan Metabolites Regulate Physiology and Behavior
3.1.1. Brain 5-HT and Its Receptors and Transporter
3.1.2. Sex Differences and Estrogen Modulation in Brain Serotonin Neurotransmission
5-HT1A Receptor
5-HT2A Receptor
5-HT2C Receptor
5-HT3 Receptor
SERT
3.2. Gut Microbial Tryptophan Metabolites Regulate Gut Function and Metabolism
3.2.1. 5-HT Regulates Gut Functions
3.2.2. Tryptamine Regulates Gut Functions
3.3. Diseases Related to Tryptophan Metabolism and 5-HT Neurotransmission in the Brain–Gut–Microbiome Axis
3.3.1. Brain–Gut–Microbiome Axis Dysfunction in Eating Disorders
3.3.2. Brain–Gut–Microbiome Axis Dysfunction in Anxiety and Depression
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Schwarcz, R.; Bruno, J.P.; Muchowski, P.J.; Wu, H.-Q. Kynurenines in the mammalian brain: When physiology meets pathology. Nat. Rev. Neurosci. 2012, 13, 465–477. [Google Scholar] [CrossRef]
- Xue, C.; Li, G.; Zheng, Q.; Gu, X.; Shi, Q.; Su, Y.; Chu, Q.; Yuan, X.; Bao, Z.; Lu, J.; et al. Tryptophan metabolism in health and disease. Cell Metabolism 2023, 35, 1304–1326. [Google Scholar] [CrossRef]
- Burokas, A.; Moloney, R.D.; Dinan, T.G.; Cryan, J.F. Microbiota regulation of the mammalian gut-brain axis. Adv. Appl. Microbiol. 2015, 91, 1–62. [Google Scholar] [CrossRef]
- Mayer, E.A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Investig. 2015, 125, 926–938. [Google Scholar] [CrossRef]
- 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]
- Bastiaanssen, T.F.S.; Cowan, C.S.M.; Claesson, M.J.; Dinan, T.G.; Cryan, J.F. Making sense of the microbiome in psychiatry. Int. J. Neuropsychopharmacol. 2019, 22, 37–52. [Google Scholar] [CrossRef]
- Kelly, J.R.; Minuto, C.; Cryan, J.F.; Clarke, G.; Dinan, T.G. Cross talk: The microbiota and neurodevelopmental disorders. Front. Neurosci. 2017, 11, 490. [Google Scholar] [CrossRef]
- Osadchiy, V.; Martin, C.R.; Mayer, E.A. The gut-brain axis and the microbiome: Mechanisms and clinical implications. Clin. Gastroenterol. Hepatol. 2019, 17, 322–332. [Google Scholar] [CrossRef]
- Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef]
- Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [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]
- The Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef]
- Costello, E.K.; Lauber, C.L.; Hamady, M.; Fierer, N.; Gordon, J.I.; Knight, R. Bacterial community variation in human body habitats across space and time. Science 2009, 326, 1694–1697. [Google Scholar] [CrossRef]
- Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
- Benson, A.K.; Kelly, S.A.; Legge, R.; Ma, F.; Low, S.J.; Kim, J.; Zhang, M.; Oh, P.L.; Nehrenberg, D.; Hua, K.; et al. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc. Natl. Acad. Sci. USA 2010, 107, 18933–18938. [Google Scholar] [CrossRef]
- Schloss, P.D.; Iverson, K.D.; Petrosino, J.F.; Schloss, S.J. The dynamics of a family’s gut microbiota reveal variations on a theme. Microbiome 2014, 2, 25. [Google Scholar] [CrossRef]
- Murphy, E.F.; Cotter, P.D.; Healy, S.; Marques, T.M.; O’Sullivan, O.; Fouhy, F.; Clarke, S.F.; O’Toole, P.W.; Quigley, E.M.; Stanton, C.; et al. Composition and energy harvesting capacity of the gut microbiota: Relationship to diet, obesity and time in mouse models. Gut 2010, 59, 1635–1642. [Google Scholar] [CrossRef]
- Markle, J.G.; Frank, D.N.; Mortin-Toth, S.; Robertson, C.E.; Feazel, L.M.; Rolle-Kampczyk, U.; von Bergen, M.; McCoy, K.D.; Macpherson, A.J.; Danska, J.S. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 2013, 339, 1084–1088. [Google Scholar] [CrossRef]
- Gomez, A.; Luckey, D.; Yeoman, C.J.; Marietta, E.V.; Berg Miller, M.E.; Murray, J.A.; White, B.A.; Taneja, V. Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible 0401 mice but not arthritis-resistant 0402 mice. PLoS ONE 2012, 7, e36095. [Google Scholar] [CrossRef]
- Bhargava, A.; Fan, S.; Lujan, C.R.; Fiehn, O.; Neylan, T.C.; Inslicht, S.S. Chemical set enrichment analysis: Novel insights into sex-specific alterations in primary metabolites in posttraumatic stress and disturbed sleep. Clin. Transl. Med. 2021, 11, e511. [Google Scholar] [CrossRef]
- Suzuki, T.A.; Nachman, M.W. Spatial heterogeneity of gut microbial composition along the gastrointestinal tract in natural populations of house mice. PLoS ONE 2016, 11, e0163720. [Google Scholar] [CrossRef]
- Welch, J.L.M.; Hasegawa, Y.; McNulty, N.P.; Gordon, J.I.; Borisy, G.G. Spatial organization of a model 15-member human gut microbiota established in gnotobiotic mice. Proc. Natl. Acad. Sci. USA 2017, 114, E9105–E9114. [Google Scholar] [CrossRef]
- Borre, Y.E.; O’Keeffe, G.W.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends Mol. Med. 2014, 20, 509–518. [Google Scholar] [CrossRef]
- 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]
- Lloyd-Price, J.; Abu-Ali, G.; Huttenhower, C. The healthy human microbiome. Genome Med. 2016, 8, 51. [Google Scholar] [CrossRef]
- Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
- Winer, D.A.; Luck, H.; Tsai, S.; Winer, S. The intestinal immune system in obesity and insulin resistance. Cell Metab. 2016, 23, 413–426. [Google Scholar] [CrossRef]
- Qin, Y.; Roberts, J.D.; Grimm, S.A.; Lih, F.B.; Deterding, L.J.; Li, R.; Chrysovergis, K.; Wade, P.A. An obesity-associated gut microbiome reprograms the intestinal epigenome and leads to altered colonic gene expression. Genome Biol. 2018, 19, 7. [Google Scholar] [CrossRef]
- Bäckhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef]
- Greco, E.A.; Lenzi, A.; Migliaccio, S.; Gessani, S. Epigenetic modifications induced by nutrients in early life phases: Gender differences in metabolic alteration in adulthood. Front. Genet. 2019, 10, 795. [Google Scholar] [CrossRef]
- Wang, J.; Tang, H.; Zhang, C.; Zhao, Y.; Derrien, M.; Rocher, E.; van-Hylckama Vlieg, J.E.T.; Strissel, K.; Zhao, L.; Obin, M.; et al. Modulation of gut microbiota during probiotic-mediated attenuation of metabolic syndrome in high fat diet-fed mice. ISME J. 2015, 9, 1–15. [Google Scholar] [CrossRef]
- Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143, 913–916.e917. [Google Scholar] [CrossRef]
- Rogers, G.B.; Keating, D.J.; Young, R.L.; Wong, M.L.; Licinio, J.; Wesselingh, S. From gut dysbiosis to altered brain function and mental illness: Mechanisms and pathways. Mol. Psychiatry 2016, 21, 738–748. [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]
- O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef]
- Furness, J.B. Integrated neural and endocrine control of gastrointestinal function. Adv. Exp. Med. Biol. 2016, 891, 159–173. [Google Scholar] [CrossRef]
- 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]
- Furness, J.B. The enteric nervous system and neurogastroenterology. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 286–294. [Google Scholar] [CrossRef]
- Furness, J.B.; Jones, C.; Nurgali, K.; Clerc, N. Intrinsic primary afferent neurons and nerve circuits within the intestine. Prog. Neurobiol. 2004, 72, 143–164. [Google Scholar] [CrossRef]
- Mawe, G.M.; Hoffman, J.M. Serotonin signalling in the gut—Functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473–486. [Google Scholar] [CrossRef]
- Wood, J.D.; Alpers, D.H.; Andrews, P.L. Fundamentals of neurogastroenterology. Gut 1999, 45 (Suppl. 2), ii6–ii16. [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]
- 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]
- O’Mahony, S.M.; Hyland, N.P.; Dinan, T.G.; Cryan, J.F. Maternal separation as a model of brain–gut axis dysfunction. Psychopharmacology 2011, 214, 71–88. [Google Scholar] [CrossRef]
- Popoff, M.R.; Poulain, B. Bacterial toxins and the nervous system: Neurotoxins and multipotential toxins interacting with neuronal cells. Toxins 2010, 2, 683–737. [Google Scholar] [CrossRef]
- Peregrin, A.T.; Ahlman, H.; Jodal, M.; Lundgren, O. Involvement of serotonin and calcium channels in the intestinal fluid secretion evoked by bile salt and cholera toxin. Br. J. Pharmacol. 1999, 127, 887–894. [Google Scholar] [CrossRef]
- Engelstoft, M.S.; Egerod, K.L.; Holst, B.; Schwartz, T.W. A gut feeling for obesity: 7TM sensors on enteroendocrine cells. Cell Metab. 2008, 8, 447–449. [Google Scholar] [CrossRef]
- Stephens, R.L.; Tache, Y. Intracisternal injection of a TRH analogue stimulates gastric luminal serotonin release in rats. Am. J. Physiol. Gastrointest. Liver Physiol. 1989, 256, G377–G383. [Google Scholar] [CrossRef]
- Fothergill, L.J.; Furness, J.B. Diversity of enteroendocrine cells investigated at cellular and subcellular levels: The need for a new classification scheme. Histochem. Cell Biol. 2018, 150, 693–702. [Google Scholar] [CrossRef]
- Gunawardene, A.R.; Corfe, B.M.; Staton, C.A. Classification and functions of enteroendocrine cells of the lower gastrointestinal tract. Int. J. Exp. Pathol. 2011, 92, 219–231. [Google Scholar] [CrossRef]
- 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]
- Cani, P.D.; Everard, A.; Duparc, T. Gut microbiota, enteroendocrine functions and metabolism. Curr. Opin. Pharmacol. 2013, 13, 935–940. [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.e116. [Google Scholar] [CrossRef]
- Tsubouchi, S.; Leblond, C.P. Migration and turnover of entero-endocrine and caveolated cells in the epithelium of the descending colon, as shown by radioautography after continuous infusion of 3H-thymidine into mice. Am. J. Anat. 1979, 156, 431–451. [Google Scholar] [CrossRef]
- Sternini, C.; Anselmi, L.; Rozengurt, E. Enteroendocrine cells: A site of ‘taste’ in gastrointestinal chemosensing. Curr. Opin. Endocrinol. Diabetes Obes. 2008, 15, 73–78. [Google Scholar] [CrossRef]
- Gribble, F.M.; Reimann, F. Enteroendocrine cells: Chemosensors in the intestinal epithelium. Annu. Rev. Physiol. 2016, 78, 277–299. [Google Scholar] [CrossRef]
- Habib, A.M.; Richards, P.; Rogers, G.J.; Reimann, F.; Gribble, F.M. Co-localisation and secretion of glucagon-like peptide 1 and peptide YY from primary cultured human L cells. Diabetologia 2013, 56, 1413–1416. [Google Scholar] [CrossRef]
- Steinert, R.E.; Feinle-Bisset, C.; Asarian, L.; Horowitz, M.; Beglinger, C.; Geary, N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory controls and physiological roles in eating and Glycemia in health, obesity, and after RYGB. Physiol. Rev. 2017, 97, 411–463. [Google Scholar] [CrossRef]
- Habib, A.M.; Richards, P.; Cairns, L.S.; Rogers, G.J.; Bannon, C.A.M.; Parker, H.E.; Morley, T.C.E.; Yeo, G.S.H.; Reimann, F.; Gribble, F.M. Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry. Endocrinology 2012, 153, 3054–3065. [Google Scholar] [CrossRef]
- De Silva, A.; Bloom, S.R. Gut hormones and appetite control: A focus on PYY and GLP-1 as therapeutic targets in obesity. Gut Liver 2012, 6, 10–20. [Google Scholar] [CrossRef]
- Richards, P.; Parker, H.E.; Adriaenssens, A.E.; Hodgson, J.M.; Cork, S.C.; Trapp, S.; Gribble, F.M.; Reimann, F. Identification and characterization of GLP-1 receptor-expressing cells using a new transgenic mouse model. Diabetes 2014, 63, 1224–1233. [Google Scholar] [CrossRef]
- Kaelberer, M.M.; Buchanan, K.L.; Klein, M.E.; Barth, B.B.; Montoya, M.M.; Shen, X.; Bohórquez, D.V. A gut-brain neural circuit for nutrient sensory transduction. Science 2018, 361, eaat5236. [Google Scholar] [CrossRef]
- Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The brain-gut-microbiome axis. Cell Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
- Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef]
- Weinstock, G.M. Genomic approaches to studying the human microbiota. Nature 2012, 489, 250–256. [Google Scholar] [CrossRef]
- McFall-Ngai, M.; Hadfield, M.G.; Bosch, T.C.; Carey, H.V.; Domazet-Lošo, T.; Douglas, A.E.; Dubilier, N.; Eberl, G.; Fukami, T.; Gilbert, S.F.; et al. Animals in a bacterial world, a new imperative for the life sciences. Proc. Natl. Acad. Sci. USA 2013, 110, 3229–3236. [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]
- Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef]
- 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.e593. [Google Scholar] [CrossRef]
- Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef]
- Verma, A.; Inslicht, S.S.; Bhargava, A. Gut-brain axis: Role of microbiome, metabolomics, hormones, and stress in mental health disorders. Cells 2024, 13, 1436. [Google Scholar] [CrossRef]
- Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef]
- Baothman, O.A.; Zamzami, M.A.; Taher, I.; Abubaker, J.; Abu-Farha, M. The role of gut microbiota in the development of obesity and diabetes. Lipids Health Dis. 2016, 15, 108. [Google Scholar] [CrossRef]
- Radwanski, E.R.; Last, R.L. Tryptophan biosynthesis and metabolism: Biochemical and molecular genetics. Plant Cell 1995, 7, 921–934. [Google Scholar] [CrossRef]
- Le Floc’h, N.; Otten, W.; Merlot, E. Tryptophan metabolism, from nutrition to potential therapeutic applications. Amino Acids 2011, 41, 1195–1205. [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]
- Ruddick, J.P.; Evans, A.K.; Nutt, D.J.; Lightman, S.L.; Rook, G.A.W.; Lowry, C.A. Tryptophan metabolism in the central nervous system: Medical implications. Expert. RevMol. Med. 2006, 8, 1–27. [Google Scholar] [CrossRef]
- Maes, M.; Leonard, B.E.; Myint, A.M.; Kubera, M.; Verkerk, R. The new ‘5-HT’ hypothesis of depression: Cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2011, 35, 702–721. [Google Scholar] [CrossRef]
- Kaur, H.; Bose, C.; Mande, S.S. Tryptophan metabolism by gut microbiome and gut-brain-axis: An in silico analysis. Front. Neurosci. 2019, 13, 1365. [Google Scholar] [CrossRef]
- Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
- Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology 2017, 112, 399–412. [Google Scholar] [CrossRef]
- Williams, B.B.; Van Benschoten, A.H.; Cimermancic, P.; Donia, M.S.; Zimmermann, M.; Taketani, M.; Ishihara, A.; Kashyap, P.C.; Fraser, J.S.; Fischbach, M.A. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe 2014, 16, 495–503. [Google Scholar] [CrossRef]
- Gao, J.; Xu, K.; Liu, H.; Liu, G.; Bai, M.; Peng, C.; Li, T.; Yin, Y. Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front. Cell Infect. Microbiol. 2018, 8, 13. [Google Scholar] [CrossRef]
- Galland, L. The gut microbiome and the brain. J. Med. Food 2014, 17, 1261–1272. [Google Scholar] [CrossRef]
- Jones, S.P.; Franco, N.F.; Varney, B.; Sundaram, G.; Brown, D.A.; de Bie, J.; Lim, C.K.; Guillemin, G.J.; Brew, B.J. Expression of the kynurenine pathway in human peripheral blood mononuclear cells: Implications for inflammatory and neurodegenerative disease. PLoS ONE 2015, 10, e0131389. [Google Scholar] [CrossRef]
- Chen, Y.; Guillemin, G.J. Kynurenine pathway metabolites in humans: Disease and healthy States. Int. J. Tryptophan Res. 2009, 2, IJTR.S2097. [Google Scholar] [CrossRef]
- Dehhaghi, M.; Kazemi Shariat Panahi, H.; Guillemin, G.J. Microorganisms, tryptophan metabolism, and kynurenine pathway: A complex interconnected loop influencing human health status. Int. J. Tryptophan Res. 2019, 12, 1178646919852996. [Google Scholar] [CrossRef]
- Raison, C.L.; Dantzer, R.; Kelley, K.W.; Lawson, M.A.; Woolwine, B.J.; Vogt, G.; Spivey, J.R.; Saito, K.; Miller, A.H. CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFN-α: Relationship to CNS immune responses and depression. Mol. Psychiatry 2010, 15, 393–403. [Google Scholar] [CrossRef]
- Lapin, I.P.; Mutovkina, L.G.; Ryzov, I.V.; Mirzaev, S. Anxiogenic activity of quinolinic acid and kynurenine in the social interaction test in mice. J. Psychopharmacol. 1996, 10, 246–249. [Google Scholar] [CrossRef]
- Kanchanatawan, B.; Thika, S.; Sirivichayakul, S.; Carvalho, A.F.; Geffard, M.; Maes, M. In schizophrenia, depression, anxiety, and physiosomatic symptoms are strongly related to psychotic symptoms and excitation, impairments in episodic memory, and increased production of neurotoxic typtophan catabolites: A multivariate and machine learning study. Neurotox. Res. 2018, 33, 641–655. [Google Scholar] [CrossRef]
- Roomruangwong, C.; Kanchanatawan, B.; Carvalho, A.F.; Sirivichayakul, S.; Duleu, S.; Geffard, M.; Maes, M. Body image dissatisfaction in pregnant and non-pregnant females is strongly predicted by immune activation and mucosa-derived activation of the tryptophan catabolite (TRYCAT) pathway. World J. Biol. Psychiatry 2018, 19, 200–209. [Google Scholar] [CrossRef]
- Perna, G.; Iannone, G.; Alciati, A.; Caldirola, D. Are anxiety disorders associated with accelerated aging? A focus on neuroprogression. Neural Plast. 2016, 2016, 8457612. [Google Scholar] [CrossRef] [PubMed]
- Németh, H.; Toldi, J.; Vécsei, L. Kynurenines, Parkinson’s disease and other neurodegenerative disorders: Preclinical and clinical studies. J. Neural Transm. Suppl. 2006, 70, 285–304. [Google Scholar] [CrossRef]
- Roomruangwong, C.; Kanchanatawan, B.; Sirivichayakul, S.; Anderson, G.; Carvalho, A.F.; Duleu, S.; Geffard, M.; Maes, M. IgA/IgM Responses to Gram-Negative Bacteria are not Associated with Perinatal Depression, but with Physio-somatic Symptoms and Activation of the Tryptophan Catabolite Pathway at the End of Term and Postnatal Anxiety. CNS Neurol. Disord. Drug Targets 2017, 16, 472–483. [Google Scholar] [CrossRef]
- Orlikov, A.B.; Prakhye, I.B.; Ryzov, I.V. Kynurenine in blood plasma and DST in patients with endogenous anxiety and endogenous depression. Biol. Psychiatry 1994, 36, 97–102. [Google Scholar] [CrossRef]
- Linan-Rico, A.; Ochoa-Cortes, F.; Beyder, A.; Soghomonyan, S.; Zuleta-Alarcon, A.; Coppola, V.; Christofi, F.L. Mechanosensory signaling in enterochromaffin cells and 5-HT release: Potential implications for gut inflammation. Front. Neurosci. 2016, 10, 564. [Google Scholar] [CrossRef]
- Gershon, M.D. 5-Hydroxytryptamine (serotonin) in the gastrointestinal tract. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 14–21. [Google Scholar] [CrossRef]
- Richard, D.M.; Dawes, M.A.; Mathias, C.W.; Acheson, A.; Hill-Kapturczak, N.; Dougherty, D.M. L-tryptophan: Basic metabolic functions, behavioral research and therapeutic indications. Int. J. Tryptophan Res. 2009, 2, IJTR.S2129. [Google Scholar] [CrossRef]
- Waclawiková, B.; El Aidy, S. Role of microbiota and tryptophan metabolites in the remote effect of intestinal inflammation on brain and depression. Pharmaceuticals 2018, 11, 63. [Google Scholar] [CrossRef]
- El-Merahbi, R.; Löffler, M.; Mayer, A.; Sumara, G. The roles of peripheral serotonin in metabolic homeostasis. FEBS Lett. 2015, 589, 1728–1734. [Google Scholar] [CrossRef] [PubMed]
- Fernstrom, J.D.; Langham, K.A.; Marcelino, L.M.; Irvine, Z.L.E.; Fernstrom, M.H.; Kaye, W.H. The ingestion of different dietary proteins by humans induces large changes in the plasma tryptophan ratio, a predictor of brain tryptophan uptake and serotonin synthesis. Clin. Nutr. 2013, 32, 1073–1076. [Google Scholar] [CrossRef] [PubMed]
- Fernstrom, J.D.; Fernstrom, M.H. Exercise, serum free tryptophan, and central fatigue. J Nutr 2006, 136, 553S–559S. [Google Scholar] [CrossRef] [PubMed]
- Evrensel, A.; Ceylan, M.E. The gut-brain axis: The missing link in depression. Clin. Psychopharmacol. Neurosci. 2015, 13, 239–244. [Google Scholar] [CrossRef]
- Knecht, L.D.; O’Connor, G.; Mittal, R.; Liu, X.Z.; Daftarian, P.; Deo, S.K.; Daunert, S. Serotonin activates bacterial quorum sensing and enhances the virulence of Pseudomonas aeruginosa in the host. EBioMedicine 2016, 9, 161–169. [Google Scholar] [CrossRef]
- Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F., III; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 1395–1403. [Google Scholar] [CrossRef]
- Hata, T.; Asano, Y.; Yoshihara, K.; Kimura-Todani, T.; Miyata, N.; Zhang, X.-T.; Takakura, S.; Aiba, Y.; Koga, Y.; Sudo, N. Regulation of gut luminal serotonin by commensal microbiota in mice. PLoS ONE 2017, 12, e0180745. [Google Scholar] [CrossRef]
- Cao, Y.-N.; Feng, L.-J.; Liu, Y.-Y.; Jiang, K.; Zhang, M.-J.; Gu, Y.-X.; Wang, B.-M.; Gao, J.; Wang, Z.-L.; Wang, Y.-M. Effect of Lactobacillus rhamnosus GG supernatant on serotonin transporter expression in rats with post-infectious irritable bowel syndrome. World J. Gastroenterol. 2018, 24, 338–350. [Google Scholar] [CrossRef]
- Grøndahl, M.L.; Unmack, M.A.; Ragnarsdóttir, H.B.; Hansen, M.B.; Olsen, J.E.; Skadhauge, E. Effects of nitric oxide in 5-hydroxytryptamine-, cholera toxin-, enterotoxigenic Escherichia coli- and Salmonella Typhimurium-induced secretion in the porcine small intestine. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2005, 141, 476–484. [Google Scholar] [CrossRef]
- Louis, P.; Hold, G.L.; Flint, H.J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 2014, 12, 661–672. [Google Scholar] [CrossRef] [PubMed]
- Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed]
- Hansen, M.B.; Witte, A.-B. The role of serotonin in intestinal luminal sensing and secretion. Acta Physiol. 2008, 193, 311–323. [Google Scholar] [CrossRef]
- Bogunovic, M.; Davé, S.H.; Tilstra, J.S.; Chang, D.T.W.; Harpaz, N.; Xiong, H.; Mayer, L.F.; Plevy, S.E. Enteroendocrine cells express functional Toll-like receptors. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G1770–G1783. [Google Scholar] [CrossRef]
- Kidd, M.; Gustafsson, B.I.; Drozdov, I.; Modlin, I. IL1β- and LPS-induced serotonin secretion is increased in EC cells derived from Crohn’s disease. Neurogastroenterol. Motil. 2008, 21, 439–450. [Google Scholar] [CrossRef] [PubMed]
- Gershon, M.D.; Tack, J. The serotonin signaling system: From basic understanding to drug development for functional GI disorders. Gastroenterology 2007, 132, 397–414. [Google Scholar] [CrossRef]
- Zucchi, R.; Chiellini, G.; Scanlan, T.S.; Grandy, D.K. Trace amine-associated receptors and their ligands. Br. J. Pharmacol. 2006, 149, 967–978. [Google Scholar] [CrossRef] [PubMed]
- Holzer, P.; Farzi, A. Neuropeptides and the microbiota-gut-brain axis. In Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease, Lyte, M., Cryan, J.F., Eds.; Springer: New York, NY, USA, 2014; pp. 195–219. [Google Scholar]
- Gareau, M.G.; Silva, M.A.; Perdue, M.H. Pathophysiological mechanisms of stress-induced intestinal damage. Curr. Mol. Med. 2008, 8, 274–281. [Google Scholar] [CrossRef]
- Maes, M.; Leunis, J.C. Normalization of leaky gut in chronic fatigue syndrome (CFS) is accompanied by a clinical improvement: Effects of age, duration of illness and the translocation of LPS from gram-negative bacteria. Neuro Endocrinol. Lett. 2008, 29, 902–910. [Google Scholar]
- Furness, J.B.; Rivera, L.R.; Cho, H.J.; Bravo, D.M.; Callaghan, B. The gut as a sensory organ. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 729–740. [Google Scholar] [CrossRef]
- Badawy, A.A.; Dougherty, D.M. Assessment of the Human Kynurenine Pathway: Comparisons and Clinical Implications of Ethnic and Gender Differences in Plasma Tryptophan, Kynurenine Metabolites, and Enzyme Expressions at Baseline and After Acute Tryptophan Loading and Depletion. Int. J. Tryptophan Res. 2016, 9, 31–49. [Google Scholar] [CrossRef]
- Monteiro-dos-Santos, P.C.; Graeff, F.G.; dos-Santos, J.E.; Ribeiro, R.P.; Guimarães, F.S.; Zuardi, A.W. Effects of tryptophan depletion on anxiety induced by simulated public speaking. Braz. J. Med. Biol. Res. 2000, 33, 581–587. [Google Scholar] [CrossRef] [PubMed]
- Maes, M.; Verkerk, R.; Bonaccorso, S.; Ombelet, W.; Bosmans, E.; Scharpé, S. Depressive and anxiety symptoms in the early puerperium are related to increased degradation of tryptophan into kynurenine, a phenomenon which is related to immune activation. Life Sci. 2002, 71, 1837–1848. [Google Scholar] [CrossRef] [PubMed]
- Doornbos, B.; Dijck-Brouwer, D.A.; Kema, I.P.; Tanke, M.A.; van Goor, S.A.; Muskiet, F.A.; Korf, J. The development of peripartum depressive symptoms is associated with gene polymorphisms of MAOA, 5-HTT and COMT. Prog. Neuropsychopharmacol. Biol. Psychiatry 2009, 33, 1250–1254. [Google Scholar] [CrossRef] [PubMed]
- Roomruangwong, C.; Kanchanatawan, B.; Sirivichayakul, S.; Anderson, G.; Carvalho, A.F.; Duleu, S.; Geffard, M.; Maes, M. IgA/IgM responses to tryptophan and tryptophan catabolites (TRYCATs) are differently associated with prenatal depression, physio-somatic symptoms at the end of term and premenstrual syndrome. Mol. Neurobiol. 2017, 54, 3038–3049. [Google Scholar] [CrossRef]
- Roomruangwong, C.; Kanchanatawan, B.; Sirivichayakul, S.; Mahieu, B.; Nowak, G.; Maes, M. Lower serum zinc and higher crp strongly predict prenatal depression and physio-somatic symptoms, which all together predict postnatal depressive symptoms. Mol. Neurobiol. 2017, 54, 1500–1512. [Google Scholar] [CrossRef]
- Roomruangwong, C.; Anderson, G.; Berk, M.; Stoyanov, D.; Carvalho, A.F.; Maes, M. A neuro-immune, neuro-oxidative and neuro-nitrosative model of prenatal and postpartum depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 81, 262–274. [Google Scholar] [CrossRef]
- Roomruangwong, C.; Barbosa, D.S.; de Farias, C.C.; Matsumoto, A.K.; Baltus, T.H.L.; Morelli, N.R.; Kanchanatawan, B.; Duleu, S.; Geffard, M.; Maes, M. Natural regulatory IgM-mediated autoimmune responses directed against malondialdehyde regulate oxidative and nitrosative pathways and coupled with IgM responses to nitroso adducts attenuate depressive and physiosomatic symptoms at the end of term pregnancy. Psychiatry Clin. Neurosci. 2018, 72, 116–130. [Google Scholar] [CrossRef]
- Kuhn, D.M.; Arthur, R.; States, J.C. Phosphorylation and activation of brain tryptophan hydroxylase: Identification of serine-58 as a substrate site for protein kinase A. J. Neurochem. 1997, 68, 2220–2223. [Google Scholar] [CrossRef]
- Banik, U.; Wang, G.-A.; Wagner, P.D.; Kaufman, S. Interaction of phosphorylated tryptophan hydroxylase with 14-3-3 proteins. J. Biol. Chem. 1997, 272, 26219–26225. [Google Scholar] [CrossRef]
- Cao, X.; Xu, P.; Oyola, M.G.; Xia, Y.; Yan, X.; Saito, K.; Zou, F.; Wang, C.; Yang, Y.; Hinton, A., Jr.; et al. Estrogens stimulate serotonin neurons to inhibit binge-like eating in mice. J. Clin. Investig. 2014, 124, 4351–4362. [Google Scholar] [CrossRef]
- Lu, N.Z.; Shlaes, T.A.; Gundlah, C.; Dziennis, S.E.; Lyle, R.E.; Bethea, C.L. Ovarian steroid action on tryptophan hydroxylase protein and serotonin compared to localization of ovarian steroid receptors in midbrain of guinea pigs. Endocrine 1999, 11, 257–267. [Google Scholar] [CrossRef]
- Hiroi, R.; McDevitt, R.A.; Neumaier, J.F. Estrogen selectively increases tryptophan hydroxylase-2 mRNA expression in distinct subregions of rat midbrain raphe nucleus: Association between gene expression and anxiety behavior in the open field. Biol. Psychiatry 2006, 60, 288–295. [Google Scholar] [CrossRef]
- Baker, J.H.; Peterson, C.M.; Thornton, L.M.; Brownley, K.A.; Bulik, C.M.; Girdler, S.S.; Marcus, M.D.; Bromberger, J.T. Reproductive and appetite hormones and bulimic symptoms during midlife. Eur. Eat. Disord. Rev. 2017, 25, 188–194. [Google Scholar] [CrossRef] [PubMed]
- Mueller, S.; Saunier, K.; Hanisch, C.; Norin, E.; Alm, L.; Midtvedt, T.; Cresci, A.; Silvi, S.; Orpianesi, C.; Verdenelli, M.C.; et al. Differences in fecal microbiota in different European study populations in relation to age, gender, and country: A cross-sectional study. Appl. Environ. Microbiol. 2006, 72, 1027–1033. [Google Scholar] [CrossRef] [PubMed]
- Shi, H.; Seeley, R.J.; Clegg, D.J. Sexual differences in the control of energy homeostasis. Front. Neuroendocrinol. 2009, 30, 396–404. [Google Scholar] [CrossRef] [PubMed]
- Jeffery, E.; Wing, A.; Holtrup, B.; Sebo, Z.; Kaplan, J.L.; Saavedra-Peña, R.; Church, C.D.; Colman, L.; Berry, R.; Rodeheffer, M.S. The adipose tissue microenvironment regulates depot-specific adipogenesis in obesity. Cell Metab. 2016, 24, 142–150. [Google Scholar] [CrossRef]
- Ewaschuk, J.B.; Diaz, H.; Meddings, L.; Diederichs, B.; Dmytrash, A.; Backer, J.; Looijer-van Langen, M.; Madsen, K.L. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G1025–G1034. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Yu, D.H.; Gadkari, M.; Zhou, Q.; Yu, S.; Gao, N.; Guan, Y.; Schady, D.; Roshan, T.N.; Chen, M.H.; Laritsky, E.; et al. Postnatal epigenetic regulation of intestinal stem cells requires DNA methylation and is guided by the microbiome. Genome Biol. 2015, 16, 211. [Google Scholar] [CrossRef]
- Semenkovich, N.P.; Planer, J.D.; Ahern, P.P.; Griffin, N.W.; Lin, C.Y.; Gordon, J.I. Impact of the gut microbiota on enhancer accessibility in gut intraepithelial lymphocytes. Proc. Natl. Acad. Sci. USA 2016, 113, 14805–14810. [Google Scholar] [CrossRef] [PubMed]
- Krautkramer, K.A.; Kreznar, J.H.; Romano, K.A.; Vivas, E.I.; Barrett-Wilt, G.A.; Rabaglia, M.E.; Keller, M.P.; Attie, A.D.; Rey, F.E.; Denu, J.M. Diet-microbiota interactions mediate global epigenetic programming in multiple host tissues. Mol. Cell 2016, 64, 982–992. [Google Scholar] [CrossRef]
- Li, R.; Grimm, S.A.; Chrysovergis, K.; Kosak, J.; Wang, X.; Du, Y.; Burkholder, A.; Janardhan, K.; Mav, D.; Shah, R.; et al. Obesity, rather than diet, drives epigenomic alterations in colonic epithelium resembling cancer progression. Cell Metab. 2014, 19, 702–711. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.M.; Kim, N.; Yoon, H.; Nam, R.H.; Lee, D.H. Microbial changes and host response in F344 rat colon depending on sex and age following a high-fat diet. Front. Microbiol. 2018, 9, 2236. [Google Scholar] [CrossRef] [PubMed]
- Matanoski, G.; Tao, X.; Almon, L.; Adade, A.A.; Davies-Cole, J.O. Demographics and tumor characteristics of colorectal cancers in the United States, 1998–2001. Cancer 2006, 107, 1112–1120. [Google Scholar] [CrossRef]
- Wu, X.C.; Chen, V.W.; Steele, B.; Ruiz, B.; Fulton, J.; Liu, L.; Carozza, S.E.; Greenlee, R. Subsite-specific incidence rate and stage of disease in colorectal cancer by race, gender, and age group in the United States, 1992–1997. Cancer 2001, 92, 2547–2554. [Google Scholar] [CrossRef]
- Wei, E.K.; Giovannucci, E.; Wu, K.; Rosner, B.; Fuchs, C.S.; Willett, W.C.; Colditz, G.A. Comparison of risk factors for colon and rectal cancer. Int. J. Cancer 2004, 108, 433–442. [Google Scholar] [CrossRef]
- White, A.; Ironmonger, L.; Steele, R.J.C.; Ormiston-Smith, N.; Crawford, C.; Seims, A. A review of sex-related differences in colorectal cancer incidence, screening uptake, routes to diagnosis, cancer stage and survival in the UK. BMC Cancer 2018, 18, 906. [Google Scholar] [CrossRef]
- Gao, R.-N.; Neutel, C.I.; Wai, E. Gender differences in colorectal cancer incidence, mortality, hospitalizations and surgical procedures in Canada. J. Public Health 2008, 30, 194–201. [Google Scholar] [CrossRef]
- Garcia, H.; Song, M. Early-life obesity and adulthood colorectal cancer risk: A meta-analysis. Rev. Panam. Salud Publica 2019, 43, e3. [Google Scholar] [CrossRef]
- Chen, K.L.; Madak-Erdogan, Z. Estrogen and microbiota crosstalk: Should we pay attention? Trends Endocrinol. Metab. 2016, 27, 752–755. [Google Scholar] [CrossRef]
- Mauvais-Jarvis, F. Sex differences in metabolic homeostasis, diabetes, and obesity. Biol. Sex. Differ. 2015, 6, 14. [Google Scholar] [CrossRef] [PubMed]
- Chella Krishnan, K.; Mehrabian, M.; Lusis, A.J. Sex differences in metabolism and cardiometabolic disorders. Curr. Opin. Lipidol. 2018, 29, 404–410. [Google Scholar] [CrossRef] [PubMed]
- Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J.; et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef]
- Zackular, J.P.; Rogers, M.A.; Ruffin, M.T.t.; Schloss, P.D. The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev. Res. 2014, 7, 1112–1121. [Google Scholar] [CrossRef] [PubMed]
- Kwa, M.; Plottel, C.S.; Blaser, M.J.; Adams, S. The intestinal microbiome and estrogen receptor-positive female breast cancer. J. Natl. Cancer Inst. 2016, 108, djw029. [Google Scholar] [CrossRef]
- Flores, R.; Shi, J.; Fuhrman, B.; Xu, X.; Veenstra, T.D.; Gail, M.H.; Gajer, P.; Ravel, J.; Goedert, J.J. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: A cross-sectional study. J. Transl. Med. 2012, 10, 253. [Google Scholar] [CrossRef]
- Leibowitz, S.F.; Alexander, J.T. Hypothalamic serotonin in control of eating behavior, meal size, and body weight. Biol. Psychiatry 1998, 44, 851–864. [Google Scholar] [CrossRef]
- Riva, G. Neurobiology of anorexia nervosa: Serotonin dysfunctions link self-starvation with body image disturbances through an impaired body memory. Front. Hum. Neurosci. 2016, 10, 600. [Google Scholar] [CrossRef]
- Hayes, D.J.; Greenshaw, A.J. 5-HT receptors and reward-related behaviour: A review. Neurosci. Biobehav. Rev. 2011, 35, 1419–1449. [Google Scholar] [CrossRef]
- Haahr, M.E.; Rasmussen, P.M.; Madsen, K.; Marner, L.; Ratner, C.; Gillings, N.; Baaré, W.F.C.; Knudsen, G.M. Obesity is associated with high serotonin 4 receptor availability in the brain reward circuitry. NeuroImage 2012, 61, 884–888. [Google Scholar] [CrossRef] [PubMed]
- Pollak Dorocic, I.; Fürth, D.; Xuan, Y.; Johansson, Y.; Pozzi, L.; Silberberg, G.; Carlén, M.; Meletis, K. A whole-brain atlas of inputs to serotonergic neurons of the dorsal and median raphe nuclei. Neuron 2014, 83, 663–678. [Google Scholar] [CrossRef] [PubMed]
- Duan, K.M.; Ma, J.H.; Wang, S.Y.; Huang, Z.; Zhou, Y.; Yu, H. The role of tryptophan metabolism in postpartum depression. Metab. Brain Dis. 2018, 33, 647–660. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, B.L.; Azmitia, E.C. Structure and function of the brain serotonin system. Physiol. Rev. 1992, 72, 165–229. [Google Scholar] [CrossRef]
- Krolick, K.N.; Zhu, Q.; Shi, H. Effects of estrogens on central nervous system neurotransmission: Implications for sex differences in mental disorders. Prog. Mol. Biol. Transl. Sci. 2018, 160, 105–171. [Google Scholar]
- Golubeva, A.V.; Joyce, S.A.; Moloney, G.; Burokas, A.; Sherwin, E.; Arboleya, S.; Flynn, I.; Khochanskiy, D.; Moya-Pérez, A.; Peterson, V.; et al. Microbiota-related changes in bile acid & tryptophan metabolism are associated with gastrointestinal dysfunction in a mouse model of autism. EBioMedicine 2017, 24, 166–178. [Google Scholar] [CrossRef]
- Geldenhuys, W.J.; Van der Schyf, C.J. Role of serotonin in Alzheimer’s disease: A new therapeutic target? CNS Drugs 2011, 25, 765–781. [Google Scholar] [CrossRef]
- Chigome, A.K.; Matsangaise, M.M.; Chukwu, B.O.; Matlala, M.; Sibanda, M.; Meyer, J.C. Review of selective serotonin reuptake inhibitors. S. Afr. Pharm. J. 2017, 84, 52–59. [Google Scholar]
- Young, S.N. Acute tryptophan depletion in humans: A review of theoretical, practical and ethical aspects. J. Psychiatry Neurosci. 2013, 38, 294–305. [Google Scholar] [CrossRef]
- Nichols, D.E.; Nichols, C.D. Serotonin receptors. Chem. Rev. 2008, 108, 1614–1641. [Google Scholar] [CrossRef]
- Hoffman, B.J.; Hansson, S.R.; Mezey, É.; Palkovits, M. Localization and dynamic regulation of biogenic amine transporters in the mammalian central nervous system. Front. Neuroendocrinol. 1998, 19, 187–231. [Google Scholar] [CrossRef] [PubMed]
- Bethea, C.L.; Brown, N.A.; Kohama, S.G. Steroid regulation of estrogen and progestin receptor messenger ribonucleic acid in monkey hypothalamus and pituitary. Endocrinology 1996, 137, 4372–4383. [Google Scholar] [CrossRef]
- Bethea, C.L.; Lu, N.Z.; Gundlah, C.; Streicher, J.M. Diverse actions of ovarian steroids in the serotonin neural system. Front. Neuroendocrinol. 2002, 23, 41–100. [Google Scholar] [CrossRef]
- Dalmasso, C.; Amigone, J.L.; Vivas, L. Serotonergic system involvement in the inhibitory action of estrogen on induced sodium appetite in female rats. Physiol. Behav. 2011, 104, 398–407. [Google Scholar] [CrossRef] [PubMed]
- Robichaud, M.; Debonnel, G. Oestrogen and testosterone modulate the firing activity of dorsal raphe nucleus serotonergic neurones in both male and female rats. J. Neuroendocrinol. 2005, 17, 179–185. [Google Scholar] [CrossRef]
- Walf, A.A.; Rhodes, M.E.; Frye, C.A. Antidepressant effects of ERβ-selective estrogen receptor modulators in the forced swim test. Pharmacol. Biochem. Behav. 2004, 78, 523–529. [Google Scholar] [CrossRef]
- Lund, T.D.; Rovis, T.; Chung, W.C.J.; Handa, R.J. Novel actions of estrogen receptor-beta on anxiety-related behaviors. Endocrinology 2005, 146, 797–807. [Google Scholar] [CrossRef]
- Walf, A.A.; Frye, C.A. ERβ-selective estrogen receptor modulators produce antianxiety behavior when administered systemically to ovariectomized rats. Neuropsychopharmacology 2005, 30, 1598. [Google Scholar] [CrossRef] [PubMed]
- Inoue, S.; Shikanai, H.; Matsumoto, M.; Hiraide, S.; Saito, Y.; Yanagawa, Y.; Yoshioka, M.; Shimamura, K.; Togashi, H. Metaplastic regulation of the median raphe nucleus via serotonin 5-HT1A receptor on hippocampal synaptic plasticity is associated with gender-specific emotional expression in rats. J. Pharmacol. Sci. 2014, 124, 394–407. [Google Scholar] [CrossRef]
- Barnes, N.M.; Sharp, T. A review of central 5-HT receptors and their function. Neuropharmacology 1999, 38, 1083–1152. [Google Scholar] [CrossRef]
- Sprouse, J.S.; Aghajanian, G.K. (-)-Propranolol blocks the inhibition of serotonergic dorsal raphe cell firing by 5-HT1A selective agonists. Eur. J. Pharmacol. 1986, 128, 295–298. [Google Scholar] [CrossRef]
- Higgins, E.S.; George, M.S. The Neuroscience of Clinical Psychiatry: The Pathophysiology of Behavior and Mental Illness; Wolters Kluwer: Philadelphia, PA, USA, 2018. [Google Scholar]
- Sharp, T.; McQuade, R.; Bramwell, S.; Hjorth, S. Effect of acute and repeated administration of 5-HT1A receptor agonists on 5-HT release in rat brain in vivo. Naunyn Schmiedebergs Arch. Pharmacol. 1993, 348, 339–346. [Google Scholar] [CrossRef]
- Bohmaker, K.; Eison, A.S.; Yocca, F.D.; Meller, E. Comparative effects of chronic 8-OH-DPAT, gepirone and ipsapirone treatment on the sensitivity of somatodendritic 5-HT1A autoreceptors. Neuropharmacology 1993, 32, 527–534. [Google Scholar] [CrossRef]
- Birzniece, V.; Johansson, I.M.; Wang, M.D.; Seckl, J.R.; Bäckström, T.; Olsson, T. Serotonin 5-HT(1A) receptor mRNA expression in dorsal hippocampus and raphe nuclei after gonadal hormone manipulation in female rats. Neuroendocrinology 2001, 74, 135–142. [Google Scholar] [CrossRef] [PubMed]
- Pecins-Thompson, M.; Bethea, C.L. Ovarian steroid regulation of serotonin-1A autoreceptor messenger RNA expression in the dorsal raphe of rhesus macaques. Neuroscience 1999, 89, 267–277. [Google Scholar] [CrossRef] [PubMed]
- Osterlund, M.K.; Halldin, C.; Hurd, Y.L. Effects of chronic 17beta-estradiol treatment on the serotonin 5-HT(1A) receptor mRNA and binding levels in the rat brain. Synapse 2000, 35, 39–44. [Google Scholar] [CrossRef]
- Hjorth, S.; Magnusson, T. The 5-HT1A receptor agonist, 8-OH-DPAT, preferentially activates cell body 5-HT autoreceptors in rat brain in vivo. Naunyn Schmiedebergs Arch. Pharmacol. 1988, 338, 463–471. [Google Scholar] [CrossRef] [PubMed]
- Currie, P.J.; Braver, M.; Mirza, A.; Sricharoon, K. Sex differences in the reversal of fluoxetine-induced anorexia following raphe injections of 8-OH-DPAT. Psychopharmacology 2004, 172, 359–364. [Google Scholar] [CrossRef]
- Salamanca, S.; Uphouse, L. Estradiol modulation of the hyperphagia induced by the 5-HT1A agonist, 8-OH-DPAT. Pharmacol. Biochem. Behav. 1992, 43, 953–955. [Google Scholar] [CrossRef]
- Weisstaub, N.V.; Zhou, M.; Lira, A.; Lambe, E.; González-Maeso, J.; Hornung, J.P.; Sibille, E.; Underwood, M.; Itohara, S.; Dauer, W.T.; et al. Cortical 5-HT2A receptor signaling modulates anxiety-like behaviors in mice. Science 2006, 313, 536–540. [Google Scholar] [CrossRef]
- Greenwood, B.N.; Strong, P.V.; Loughridge, A.B.; Day, H.E.; Clark, P.J.; Mika, A.; Hellwinkel, J.E.; Spence, K.G.; Fleshner, M. 5-HT2C receptors in the basolateral amygdala and dorsal striatum are a novel target for the anxiolytic and antidepressant effects of exercise. PLoS ONE 2012, 7, e46118. [Google Scholar] [CrossRef]
- Strong, P.V.; Christianson, J.P.; Loughridge, A.B.; Amat, J.; Maier, S.F.; Fleshner, M.; Greenwood, B.N. 5-hydroxytryptamine 2C receptors in the dorsal striatum mediate stress-induced interference with negatively reinforced instrumental escape behavior. Neuroscience 2011, 197, 132–144. [Google Scholar] [CrossRef]
- Kimura, A.; Stevenson, P.L.; Carter, R.N.; Maccoll, G.; French, K.L.; Simons, J.P.; Al-Shawi, R.; Kelly, V.; Chapman, K.E.; Holmes, M.C. Overexpression of 5-HT2C receptors in forebrain leads to elevated anxiety and hypoactivity. Eur. J. Neurosci. 2009, 30, 299–306. [Google Scholar] [CrossRef]
- Heisler, L.K.; Zhou, L.; Bajwa, P.; Hsu, J.; Tecott, L.H. Serotonin 5-HT(2C) receptors regulate anxiety-like behavior. Genes Brain Behav. 2007, 6, 491–496. [Google Scholar] [CrossRef] [PubMed]
- Stam, N.J.; Vanderheyden, P.; van Alebeek, C.; Klomp, J.; de Boer, T.; van Delft, A.M.L.; Olijve, W. Genomic organisation and functional expression of the gene encoding the human serotonin 5-HT2C receptor. Eur. J. Pharmacol. Mol. Pharmacol. 1994, 269, 339–348. [Google Scholar] [CrossRef]
- Tecott, L.H.; Sun, L.M.; Akana, S.F.; Strack, A.M.; Lowenstein, D.H.; Dallman, M.F.; Julius, D. Eating disorder and epilepsy in mice lacking 5-HT2C serotonin receptors. Nature 1995, 374, 542. [Google Scholar] [CrossRef] [PubMed]
- Nonogaki, K.; Strack, A.M.; Dallman, M.F.; Tecott, L.H. Leptin-independent hyperphagia and type 2 diabetes in mice with a mutated serotonin 5-HT2C receptor gene. Nat. Med. 1998, 4, 1152. [Google Scholar] [CrossRef] [PubMed]
- Voigt, J.-P.; Fink, H. Serotonin controlling feeding and satiety. Behav. Brain Res. 2015, 277, 14–31. [Google Scholar] [CrossRef]
- Gundlah, C.; Pecins-Thompson, M.; Schutzer, W.E.; Bethea, C.L. Ovarian steroid effects on serotonin 1A, 2A and 2C receptor mRNA in macaque hypothalamus. Brain Res. Mol. Brain Res. 1999, 63, 325–339. [Google Scholar] [CrossRef]
- Rivera, H.M.; Santollo, J.; Nikonova, L.V.; Eckel, L.A. Estradiol increases the anorexia associated with increased 5-HT2C receptor activation in ovariectomized rats. Physiol. Behav. 2012, 105, 188–194. [Google Scholar] [CrossRef]
- de Souza Silva, M.; Guimarães, F.S.; Graeff, F.G.; Tomaz, C. Absence of amnestic effect of an anxiolytic 5-HT3 antagonist (BRL 46470A) injected into basolateral amygdala, as opposed to diazepam. Behav. Brain Res. 1993, 59, 141–145. [Google Scholar] [CrossRef]
- Bhatnagar, S.; Nowak, N.; Babich, L.; Bok, L. Deletion of the 5-HT3 receptor differentially affects behavior of males and females in the Porsolt forced swim and defensive withdrawal tests. Behav. Brain Res. 2004, 153, 527–535. [Google Scholar] [CrossRef]
- Carlsson, M.; Carlsson, A. A regional study of sex differences in rat brain serotonin. Prog. Neuropsychopharmacol. Biol. Psychiatry 1988, 12, 53–61. [Google Scholar] [CrossRef] [PubMed]
- Pecins-Thompson, M.; Brown, N.A.; Bethea, C.L. Regulation of serotonin re-uptake transporter mRNA expression by ovarian steroids in rhesus macaques. Brain Res. Mol. Brain Res. 1998, 53, 120–129. [Google Scholar] [CrossRef] [PubMed]
- Mendelson, S.D.; McKittrick, C.R.; McEwen, B.S. Autoradiographic analyses of the effects of estradiol benzoate on [3H]paroxetine binding in the cerebral cortex and dorsal hippocampus of gonadectomized male and female rats. Brain Res. 1993, 601, 299–302. [Google Scholar] [CrossRef]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
- Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [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]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
- Bistoletti, M.; Caputi, V.; Baranzini, N.; Marchesi, N.; Filpa, V.; Marsilio, I.; Cerantola, S.; Terova, G.; Baj, A.; Grimaldi, A.; et al. Antibiotic treatment-induced dysbiosis differently affects BDNF and TrkB expression in the brain and in the gut of juvenile mice. PLoS ONE 2019, 14, e0212856. [Google Scholar] [CrossRef]
- Caputi, V.; Marsilio, I.; Filpa, V.; Cerantola, S.; Orso, G.; Bistoletti, M.; Paccagnella, N.; De Martin, S.; Montopoli, M.; Dall’Acqua, S.; et al. Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice. Br. J. Pharmacol. 2017, 174, 3623–3639. [Google Scholar] [CrossRef]
- Xue, J.; Askwith, C.; Javed, N.H.; Cooke, H.J. Autonomic nervous system and secretion across the intestinal mucosal surface. Auton. Neurosci. 2007, 133, 55–63. [Google Scholar] [CrossRef] [PubMed]
- Camilleri, M. Serotonin in the gastrointestinal tract. Curr. Opin. Endocrinol. Diabetes Obes. 2009, 16, 53–59. [Google Scholar] [CrossRef]
- Ceccotti, C.; Giaroni, C.; Bistoletti, M.; Viola, M.; Crema, F.; Terova, G. Neurochemical characterization of myenteric neurons in the juvenile gilthead sea bream (Sparus aurata) intestine. PLoS ONE 2018, 13, e0201760. [Google Scholar] [CrossRef]
- Kendig, D.M.; Grider, J.R. Serotonin and colonic motility. Neurogastroenterol. Motil. 2015, 27, 899–905. [Google Scholar] [CrossRef] [PubMed]
- Spencer, N.J. Constitutively active 5-HT receptors: An explanation of how 5-HT antagonists inhibit gut motility in species where 5-HT is not an enteric neurotransmitter? Front. Cell Neurosci. 2015, 9, 487. [Google Scholar] [CrossRef]
- De Vadder, F.; Grasset, E.; Mannerås Holm, L.; 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]
- Takaki, M.; Mawe, G.M.; Barasch, J.M.; Gershon, M.D.; Gershon, M.D. Physiological responses of guinea-pig myenteric neurons secondary to the release of endogenous serotonin by tryptamine. Neuroscience 1985, 16, 223–240. [Google Scholar] [CrossRef]
- Forsythe, P.; Kunze, W.A. Voices from within: Gut microbes and the CNS. Cell Mol. Life Sci. 2013, 70, 55–69. [Google Scholar] [CrossRef]
- McVey Neufeld, K.A.; Mao, Y.K.; Bienenstock, J.; Foster, J.A.; Kunze, W.A. The microbiome is essential for normal gut intrinsic primary afferent neuron excitability in the mouse. Neurogastroenterol. Motil. 2013, 25, 183–e188. [Google Scholar] [CrossRef]
- Husebye, E.; Hellström, P.M.; Sundler, F.; Chen, J.; Midtvedt, T. Influence of microbial species on small intestinal myoelectric activity and transit in germ-free rats. Am. J. Physiol. Gastrointest. Liver Physiol. 2001, 280, G368–G380. [Google Scholar] [CrossRef] [PubMed]
- Walecka-Kapica, E.; Chojnacki, J.; Stępień, A.; Wachowska-Kelly, P.; Klupińska, G.; Chojnacki, C. Melatonin and female hormone secretion in postmenopausal overweight women. Int. J. Mol. Sci. 2015, 16, 1030–1042. [Google Scholar] [CrossRef] [PubMed]
- Vitetta, L.; Bambling, M.; Alford, H. The gastrointestinal tract microbiome, probiotics, and mood. Inflammopharmacology 2014, 22, 333–339. [Google Scholar] [CrossRef] [PubMed]
- Lyte, M. Microbial endocrinology and infectious disease in the 21st century. Trends Microbiol. 2004, 12, 14–20. [Google Scholar] [CrossRef]
- Kali, A. Psychobiotics: An emerging probiotic in psychiatric practice. Biomed. J. 2016, 39, 223–224. [Google Scholar] [CrossRef]
- Crumeyrolle-Arias, M.; Jaglin, M.; Bruneau, A.; Vancassel, S.; Cardona, A.; Daugé, V.; Naudon, L.; Rabot, S. Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 2014, 42, 207–217. [Google Scholar] [CrossRef]
- Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G.; et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109–118. [Google Scholar] [CrossRef]
- Dryden, S.; Frankish, H.M.; Wang, Q.; Pickavance, L.; Williams, G. The serotonergic agent fluoxetine reduces neuropeptide Y levels and neuropeptide Y secretion in the hypothalamus of lean and obese rats. Neuroscience 1996, 72, 557–566. [Google Scholar] [CrossRef]
- Mennigen, J.A.; Harris, E.A.; Chang, J.P.; Moon, T.W.; Trudeau, V.L. Fluoxetine affects weight gain and expression of feeding peptides in the female goldfish brain. Regul. Pept. 2009, 155, 99–104. [Google Scholar] [CrossRef]
- Eckel, L.A.; Rivera, H.M.; Atchley, D.P.D. The anorectic effect of fenfluramine is influenced by sex and stage of the estrous cycle in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R1486–R1491. [Google Scholar] [CrossRef]
- Hodes, G.E.; Hill-Smith, T.E.; Suckow, R.F.; Cooper, T.B.; Lucki, I. Sex-specific effects of chronic fluoxetine treatment on neuroplasticity and pharmacokinetics in mice. J. Pharmacol. Exp. Ther. 2010, 332, 266–273. [Google Scholar] [CrossRef]
- Shor-Posner, G.; Grinker, J.A.; Marinescu, C.; Brown, O.; Leibowitz, S.F. Hypothalamic serotonin in the control of meal patterns and macronutrient selection. Brain Res. Bull. 1986, 17, 663–671. [Google Scholar] [CrossRef] [PubMed]
- Kanarek, R.B.; Dushkin, H. Peripheral serotonin administration selectively reduces fat intake in rats. Pharmacol. Biochem. Behav. 1988, 31, 113–122. [Google Scholar] [CrossRef] [PubMed]
- Weiss, G.F.; Rogacki, N.; Fueg, A.; Buchen, D.; Suh, J.S.; Wong, D.T.; Leibowitz, S.F. Effect of hypothalamic and peripheral fluoxetine injection on natural patterns of macronutrient intake in the rat. Psychopharmacology 1991, 105, 467–476. [Google Scholar] [CrossRef] [PubMed]
- Kennett, G.A.; Curzon, G. Evidence that hypophagia induced by mCPP and TFMPP requires 5-HT1C and 5-HT1B receptors; hypophagia induced by RU 24969 only requires 5-HT1B receptors. Psychopharmacology 1988, 96, 93–100. [Google Scholar] [CrossRef]
- Leibowitz, S.F.; Alexander, J.T.; Cheung, W.K.; Weiss, G.F. Effects of serotonin and the serotonin blocker metergoline on meal patterns and macronutrient selection. Pharmacol. Biochem. Behav. 1993, 45, 185–194. [Google Scholar] [CrossRef]
- Heisler, L.K.; Kanarek, R.B.; Homoleski, B. Reduction of fat and protein intakes but not carbohydrate intake following acute and chronic fluoxetine in female rats. Pharmacol. Biochem. Behav. 1999, 63, 377–385. [Google Scholar] [CrossRef]
- Dinan, T.G.; Stanton, C.; Cryan, J.F. Psychobiotics: A novel class of psychotropic. Biol. Psychiatry 2013, 74, 720–726. [Google Scholar] [CrossRef]
- Sun, X.; Zhang, H.F.; Ma, C.L.; Wei, H.; Li, B.M.; Luo, J. Alleviation of anxiety/depressive-like behaviors and improvement of cognitive functions by Lactobacillus plantarum WLPL04 in chronically stressed mice. Can. J. Infect. Dis. Med. Microbiol. 2021, 2021, 6613903. [Google Scholar] [CrossRef]
- Eutamene, H.; Bueno, L. Role of probiotics in correcting abnormalities of colonic flora induced by stress. Gut 2007, 56, 1495–1497. [Google Scholar] [CrossRef]
- Xu, M.; Wang, C.; Krolick, K.N.; Shi, H.; Zhu, J. Difference in post-stress recovery of the gut microbiome and its altered metabolism after chronic adolescent stress in rats. Sci. Rep. 2020, 10, 3950. [Google Scholar] [CrossRef] [PubMed]
- Vataeva, L.A.; Khozhay, L.I.; Makukhina, G.V.; Otellin, V.A. Behavior of male and female mice submitted to action of p-chlorophenylalanine in prenatal ontogenesis. J. Evol. Biochem. Physiol. 2007, 43, 415–420. [Google Scholar] [CrossRef]
- Mendelson, S.D.; McEwen, B.S. Autoradiographic analyses of the effects of restraint-induced stress on 5-HT1A, 5-HT1C and 5-HT2 receptors in the dorsal hippocampus of male and female rats. Neuroendocrinology 1991, 54, 454–461. [Google Scholar] [CrossRef]
- Goel, N.; Innala, L.; Viau, V. Sex differences in serotonin (5-HT) 1A receptor regulation of HPA axis and dorsal raphe responses to acute restraint. Psychoneuroendocrinology 2014, 40, 232–241. [Google Scholar] [CrossRef]
- Maes, M.; Vandewoude, M.; Schotte, C.; Maes, L.; Martin, M.; Blockx, P. Sex-linked differences in cortisol, ACTH and prolactin responses to 5-hydroxy-tryptophan in healthy controls and minor and major depressed patients. Acta Psychiatr. Scand. 1989, 80, 584–590. [Google Scholar] [CrossRef] [PubMed]
- McEuen, J.G.; Semsar, K.A.; Lim, M.A.; Bale, T.L. Influence of sex and corticotropin-releasing factor pathways as determinants in serotonin sensitivity. Endocrinology 2009, 150, 3709–3716. [Google Scholar] [CrossRef]
- Mihm, M.; Gangooly, S.; Muttukrishna, S. The normal menstrual cycle in women. Anim. Reprod. Sci. 2011, 124, 229–236. [Google Scholar] [CrossRef]
- Krolick, K.N.; Shi, H. Estrogenic action in stress-induced neuroendocrine regulation of energy homeostasis. Cells 2022, 11, 879. [Google Scholar] [CrossRef]
Sex Differences | Sex Hormone Effects | |
---|---|---|
Systemic Tryptophan Metabolism | Tryptophan: male > female [122] 5-HT synthesis: male > female Tryptophan metabolites: male < female [80,122,124] | Low estrogen level reduces 5-HT synthesis and 5-HT levels [130,131,132]. Estrogen increases 5-HT level [133]. |
Brain 5-HT Metabolism | Levels of 5-HT in the CNS: male < female [205] Activation of 5-HT1A autoreceptor by 8-OH-DPAT: male > female [188,190,191] | Estrogen suppresses 5-HT1A autoreceptor expression [186,187,188]. Estrogen activates 5-HT neurons [175,176]. Effects of estrogen on 5-HT2C receptor are brain region-specific [201,202]. Estrogen decreases SERT expression [207]. |
5-HT-Related Neuropsychiatric Disorders | Eating disorders: male < female Mood disorders: male < female Anxiety: male < female [123] | Eating disorders are high in pregnant and parturient women [94,124,125,126] and also high in some postmenopausal women [135,224]. |
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. |
© 2025 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
Xu, M.; Zhou, E.Y.; Shi, H. Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain–Gut–Microbiome Axis: A Focus on Sex Differences. Cells 2025, 14, 384. https://doi.org/10.3390/cells14050384
Xu M, Zhou EY, Shi H. Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain–Gut–Microbiome Axis: A Focus on Sex Differences. Cells. 2025; 14(5):384. https://doi.org/10.3390/cells14050384
Chicago/Turabian StyleXu, Mengyang, Ethan Y. Zhou, and Haifei Shi. 2025. "Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain–Gut–Microbiome Axis: A Focus on Sex Differences" Cells 14, no. 5: 384. https://doi.org/10.3390/cells14050384
APA StyleXu, M., Zhou, E. Y., & Shi, H. (2025). Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain–Gut–Microbiome Axis: A Focus on Sex Differences. Cells, 14(5), 384. https://doi.org/10.3390/cells14050384