Nutraceuticals and Physical Activity as Antidepressants: The Central Role of the Gut Microbiota
Abstract
:1. Introduction
2. Microbiota–Gut–Brain Axis
Tryptophan Metabolism and Gut Microbiota–Brain Axis
3. Oxidative Stress
4. Dysbiosis and Depression
5. Antidepressant Effects of Nutraceuticals
5.1. Psychobiotics
5.2. Prebiotics
5.3. Polyphenols
5.4. Short-Chain Fatty Acids
5.5. Omega-3 Poly-Unsaturated Fatty Acids (PUFAs)
5.6. Vitamin A and Carotenoids
6. Exercise as an Antidepressant through the Microbiota–Gut–Brain Axis
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Kennedy, S.H. Core symptoms of major depressive disorder: Relevance to diagnosis and treatment. Dialogues Clin. Neurosci. 2008, 10, 271–277. [Google Scholar] [PubMed]
- Frazier, P.; Liu, Y.; Asplund, A.; Meredith, L.; Nguyen-Feng, V.N. US college student mental health and COVID-19: Comparing pre-pandemic and pandemic timepoints. J. Am. Coll. Health 2021, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Maes, M.; Galecki, P.; Chang, Y.S.; Berk, M. A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness. Prog. Neuropsychopharmacol. Biol. Psychiatry 2011, 35, 676–692. [Google Scholar] [CrossRef]
- Moylan, S.; Berk, M.; Dean, O.M.; Samuni, Y.; Williams, L.J.; O’Neil, A.; Hayley, A.C.; Pasco, J.A.; Anderson, G.; Jacka, F.N.; et al. Oxidative & nitrosative stress in depression: Why so much stress? Neurosci. Biobehav. Rev. 2014, 45, 46–62. [Google Scholar] [CrossRef] [PubMed]
- Salim, S. Oxidative stress and psychological disorders. Curr. Neuropharmacol. 2014, 12, 140–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berthold-Losleben, M.; Himmerich, H. The TNF-alpha system: Functional aspects in depression, narcolepsy and psychopharmacology. Curr. Neuropharmacol. 2008, 6, 193–202. [Google Scholar] [CrossRef] [Green Version]
- Clapp, M.; Aurora, N.; Herrera, L.; Bhatia, M.; Wilen, E.; Wakefield, S. Gut microbiota’s effect on mental health: The gut-brain axis. Clin. Pract. 2017, 7, 987. [Google Scholar] [CrossRef]
- Foster, J.A.; McVey Neufeld, K.A. Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef]
- Mello, B.S.; Monte, A.S.; McIntyre, R.S.; Soczynska, J.K.; Custodio, C.S.; Cordeiro, R.C.; Chaves, J.H.; Vasconcelos, S.M.; Nobre, H.V., Jr.; Florenco de Sousa, F.C.; et al. Effects of doxycycline on depressive-like behavior in mice after lipopolysaccharide (LPS) administration. J. Psychiatr. Res. 2013, 47, 1521–1529. [Google Scholar] [CrossRef]
- Miyaoka, T.; Wake, R.; Furuya, M.; Liaury, K.; Ieda, M.; Kawakami, K.; Tsuchie, K.; Taki, M.; Ishihara, K.; Araki, T.; et al. Minocycline as adjunctive therapy for patients with unipolar psychotic depression: An open-label study. Prog. Neuropsychopharmacol. Biol. Psychiatry 2012, 37, 222–226. [Google Scholar] [CrossRef]
- Filipovic, B.R.; Filipovic, B.F. Psychiatric comorbidity in the treatment of patients with inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 3552–3563. [Google Scholar] [CrossRef] [PubMed]
- Morshedi, M.; Valenlia, K.B.; Hosseinifard, E.S.; Shahabi, P.; Abbasi, M.M.; Ghorbani, M.; Barzegari, A.; Sadigh-Eteghad, S.; Saghafi-Asl, M. Beneficial psychological effects of novel psychobiotics in diabetic rats: The interaction among the gut, blood and amygdala. J. Nutr. Biochem. 2018, 57, 145–152. [Google Scholar] [CrossRef] [PubMed]
- Donati Zeppa, S.; Agostini, D.; Gervasi, M.; Annibalini, G.; Amatori, S.; Ferrini, F.; Sisti, D.; Piccoli, G.; Barbieri, E.; Sestili, P.; et al. Mutual Interactions among Exercise, Sport Supplements and Microbiota. Nutrients 2019, 12, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef]
- Godos, J.; Currenti, W.; Angelino, D.; Mena, P.; Castellano, S.; Caraci, F.; Galvano, F.; Del Rio, D.; Ferri, R.; Grosso, G. Diet and Mental Health: Review of the Recent Updates on Molecular Mechanisms. Antioxidants 2020, 9, 346. [Google Scholar] [CrossRef] [Green Version]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; 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] [Green Version]
- Fava, F.; Rizzetto, L.; Tuohy, K.M. Gut microbiota and health: Connecting actors across the metabolic system. Proc. Nutr. Soc. 2019, 78, 177–188. [Google Scholar] [CrossRef]
- Hills, R.D., Jr.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef] [Green Version]
- Dinan, T.G.; Cryan, J.F. The Microbiome-Gut-Brain Axis in Health and Disease. Gastroenterol. Clin. N. Am. 2017, 46, 77–89. [Google Scholar] [CrossRef] [Green Version]
- Simpson, C.A.; Diaz-Arteche, C.; Eliby, D.; Schwartz, O.S.; Simmons, J.G.; Cowan, C.S.M. The gut microbiota in anxiety and depression—A systematic review. Clin. Psychol. Rev. 2021, 83, 101943. [Google Scholar] [CrossRef] [PubMed]
- Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linlokken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 2014, 26, 1155–1162. [Google Scholar] [CrossRef] [PubMed]
- Qu, W.; Liu, S.; Zhang, W.; Zhu, H.; Tao, Q.; Wang, H.; Yan, H. Impact of traditional Chinese medicine treatment on chronic unpredictable mild stress-induced depression-like behaviors: Intestinal microbiota and gut microbiome function. Food Funct. 2019, 10, 5886–5897. [Google Scholar] [CrossRef]
- Baj, A.; Moro, E.; Bistoletti, M.; Orlandi, V.; Crema, F.; Giaroni, C. Glutamatergic Signaling along the Microbiota-Gut-Brain Axis. Int. J. Mol. Sci. 2019, 20, 1482. [Google Scholar] [CrossRef] [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] [PubMed]
- Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dash, S.; Clarke, G.; Berk, M.; Jacka, F.N. The gut microbiome and diet in psychiatry: Focus on depression. Curr. Opin. Psychiatry 2015, 28, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Lach, G.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. Anxiety, Depression, and the Microbiome: A Role for Gut Peptides. Neurotherapeutics 2018, 15, 36–59. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Zheng, P.; Li, Y.; Wu, J.; Tan, X.; Zhou, J.; Sun, Z.; Chen, X.; Zhang, G.; Zhang, H.; et al. Landscapes of bacterial and metabolic signatures and their interaction in major depressive disorders. Sci. Adv. 2020, 6, eaba8555. [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]
- 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] [PubMed]
- Tuohy, K.M.; Venuti, P.; Cuva, S.; Furlanello, C.; Gasperotti, M.; Mancini, A.; Ceppa, F.; Cavalieri, D.; De Filippo, C.; Vrhovsek, U.; et al. Diet and the Gut Microbiota—How the Gut: Brain Axis Impacts on Autism. In Diet-Microbe Interactions in the Gut; Tuohy, K.M., Del Rio, D., Eds.; Elsevier: Amsterdam, The Netherlands, 2014; pp. 225–245. [Google Scholar]
- Houghton, D.; Hardy, T.; Stewart, C.; Errington, L.; Day, C.P.; Trenell, M.I.; Avery, L. Systematic review assessing the effectiveness of dietary intervention on gut microbiota in adults with type 2 diabetes. Diabetologia 2018, 61, 1700–1711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gershon, M.D. 5-Hydroxytryptamine (serotonin) in the gastrointestinal tract. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 14–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Wang, H.; Gui, S.; Zeng, B.; Pu, J.; Zheng, P.; Zeng, L.; Luo, Y.; Wu, Y.; Zhou, C.; et al. Proteomics analysis of the gut-brain axis in a gut microbiota-dysbiosis model of depression. Transl. Psychiatry 2021, 11, 568. [Google Scholar] [CrossRef]
- Bosi, A.; Banfi, D.; Bistoletti, M.; Giaroni, C.; Baj, A. Tryptophan Metabolites Along the Microbiota-Gut-Brain Axis: An Interkingdom Communication System Influencing the Gut in Health and Disease. Int. J. Tryptophan. Res. 2020, 13, 1178646920928984. [Google Scholar] [CrossRef] [PubMed]
- Donner, N.C.; Johnson, P.L.; Fitz, S.D.; Kellen, K.E.; Shekhar, A.; Lowry, C.A. Elevated tph2 mRNA expression in a rat model of chronic anxiety. Depress. Anxiety 2012, 29, 307–319. [Google Scholar] [CrossRef] [Green Version]
- Kerckhoffs, A.P.; ter Linde, J.J.; Akkermans, L.M.; Samsom, M. SERT and TPH-1 mRNA expression are reduced in irritable bowel syndrome patients regardless of visceral sensitivity state in large intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, G1053–G1060. [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]
- 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] [Green Version]
- Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef] [Green Version]
- Behl, T.; Kaur, I.; Sehgal, A.; Singh, S.; Bhatia, S.; Al-Harrasi, A.; Zengin, G.; Bumbu, A.G.; Andronie-Cioara, F.L.; Nechifor, A.C.; et al. The Footprint of Kynurenine Pathway in Neurodegeneration: Janus-Faced Role in Parkinson’s Disorder and Therapeutic Implications. Int. J. Mol. Sci. 2021, 22, 6737. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Martin-Gallausiaux, C.; Larraufie, P.; Jarry, A.; Beguet-Crespel, F.; Marinelli, L.; Ledue, F.; Reimann, F.; Blottiere, H.M.; Lapaque, N. Butyrate Produced by Commensal Bacteria Down-Regulates Indolamine 2,3-Dioxygenase 1 (IDO-1) Expression via a Dual Mechanism in Human Intestinal Epithelial Cells. Front. Immunol. 2018, 9, 2838. [Google Scholar] [CrossRef] [PubMed]
- Jaglin, M.; Rhimi, M.; Philippe, C.; Pons, N.; Bruneau, A.; Goustard, B.; Dauge, V.; Maguin, E.; Naudon, L.; Rabot, S. Indole, a Signaling Molecule Produced by the Gut Microbiota, Negatively Impacts Emotional Behaviors in Rats. Front. Neurosci. 2018, 12, 216. [Google Scholar] [CrossRef] [PubMed]
- Rothhammer, V.; Mascanfroni, I.D.; Bunse, L.; Takenaka, M.C.; Kenison, J.E.; Mayo, L.; Chao, C.C.; Patel, B.; Yan, R.; Blain, M.; et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 2016, 22, 586–597. [Google Scholar] [CrossRef]
- Messaoud, A.; Mensi, R.; Douki, W.; Neffati, F.; Najjar, M.F.; Gobbi, G.; Valtorta, F.; Gaha, L.; Comai, S. Reduced peripheral availability of tryptophan and increased activation of the kynurenine pathway and cortisol correlate with major depression and suicide. World J. Biol. Psychiatry 2019, 20, 703–711. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Ding, L.; Zhang, H.; Mellor, D.; Wu, H.; Zhao, D.; Wu, C.; Lin, Z.; Yuan, J.; Peng, D. The Metabolic Factor Kynurenic Acid of Kynurenine Pathway Predicts Major Depressive Disorder. Front. Psychiatry 2018, 9, 552. [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]
- Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [Green Version]
- Barbieri, E.; Sestili, P. Reactive oxygen species in skeletal muscle signaling. J. Signal Transduct. 2012, 2012, 982794. [Google Scholar] [CrossRef] [Green Version]
- Cortie, C.H.; Hulbert, A.J.; Hancock, S.E.; Mitchell, T.W.; McAndrew, D.; Else, P.L. Of mice, pigs and humans: An analysis of mitochondrial phospholipids from mammals with very different maximal lifespans. Exp. Gerontol. 2015, 70, 135–143. [Google Scholar] [CrossRef] [PubMed]
- Calabrese, E.J.; Baldwin, L.A. Defining hormesis. Hum. Exp. Toxicol. 2002, 21, 91–97. [Google Scholar] [CrossRef] [PubMed]
- Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef] [PubMed]
- Cortese-Krott, M.M.; Koning, A.; Kuhnle, G.G.C.; Nagy, P.; Bianco, C.L.; Pasch, A.; Wink, D.A.; Fukuto, J.M.; Jackson, A.A.; van Goor, H.; et al. The Reactive Species Interactome: Evolutionary Emergence, Biological Significance, and Opportunities for Redox Metabolomics and Personalized Medicine. Antioxid. Redox Signal. 2017, 27, 684–712. [Google Scholar] [CrossRef] [Green Version]
- da Rocha, A.L.; Pinto, A.P.; Kohama, E.B.; Pauli, J.R.; de Moura, L.P.; Cintra, D.E.; Ropelle, E.R.; da Silva, A.S.R. The proinflammatory effects of chronic excessive exercise. Cytokine 2019, 119, 57–61. [Google Scholar] [CrossRef]
- Tapia, P.C. Sublethal mitochondrial stress with an attendant stoichiometric augmentation of reactive oxygen species may precipitate many of the beneficial alterations in cellular physiology produced by caloric restriction, intermittent fasting, exercise and dietary phytonutrients: “Mitohormesis” for health and vitality. Med. Hypotheses 2006, 66, 832–843. [Google Scholar] [CrossRef]
- Mottawea, W.; Chiang, C.K.; Muhlbauer, M.; Starr, A.E.; Butcher, J.; Abujamel, T.; Deeke, S.A.; Brandel, A.; Zhou, H.; Shokralla, S.; et al. Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn’s disease. Nat. Commun. 2016, 7, 13419. [Google Scholar] [CrossRef]
- Clark, A.; Mach, N. The Crosstalk between the Gut Microbiota and Mitochondria during Exercise. Front. Physiol. 2017, 8, 319. [Google Scholar] [CrossRef]
- Ribeiro, M.F.; Santos, A.A.; Afonso, M.B.; Rodrigues, P.M.; Sa Santos, S.; Castro, R.E.; Rodrigues, C.M.P.; Sola, S. Diet-dependent gut microbiota impacts on adult neurogenesis through mitochondrial stress modulation. Brain Commun. 2020, 2, fcaa165. [Google Scholar] [CrossRef] [PubMed]
- Schluter, J.; Peled, J.U.; Taylor, B.P.; Markey, K.A.; Smith, M.; Taur, Y.; Niehus, R.; Staffas, A.; Dai, A.; Fontana, E.; et al. The gut microbiota is associated with immune cell dynamics in humans. Nature 2020, 588, 303–307. [Google Scholar] [CrossRef]
- Stacy, A.; Andrade-Oliveira, V.; McCulloch, J.A.; Hild, B.; Oh, J.H.; Perez-Chaparro, P.J.; Sim, C.K.; Lim, A.I.; Link, V.M.; Enamorado, M.; et al. Infection trains the host for microbiota-enhanced resistance to pathogens. Cell 2021, 184, 615–627. [Google Scholar] [CrossRef] [PubMed]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Toth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marizzoni, M.; Cattaneo, A.; Mirabelli, P.; Festari, C.; Lopizzo, N.; Nicolosi, V.; Mombelli, E.; Mazzelli, M.; Luongo, D.; Naviglio, D.; et al. Short-Chain Fatty Acids and Lipopolysaccharide as Mediators Between Gut Dysbiosis and Amyloid Pathology in Alzheimer’s Disease. J. Alzheimer’s Dis. 2020, 78, 683–697. [Google Scholar] [CrossRef] [PubMed]
- Huie, R.E.; Padmaja, S. The reaction of no with superoxide. Free Radic. Res. Commun. 1993, 18, 195–199. [Google Scholar] [CrossRef]
- Tiso, M.; Schechter, A.N. Nitrate reduction to nitrite, nitric oxide and ammonia by gut bacteria under physiological conditions. PLoS ONE 2015, 10, e0119712. [Google Scholar] [CrossRef] [Green Version]
- Leclerc, M.; Bedu-Ferrari, C.; Etienne-Mesmin, L.; Mariadassou, M.; Lebreuilly, L.; Tran, S.L.; Brazeau, L.; Mayeur, C.; Delmas, J.; Rue, O.; et al. Nitric Oxide Impacts Human Gut Microbiota Diversity and Functionalities. mSystems 2021, 6, e0055821. [Google Scholar] [CrossRef]
- Saint-Georges-Chaumet, Y.; Edeas, M. Microbiota-mitochondria inter-talk: Consequence for microbiota-host interaction. Pathog. Dis. 2016, 74, ftv096. [Google Scholar] [CrossRef] [Green Version]
- Leschelle, X.; Goubern, M.; Andriamihaja, M.; Blottiere, H.M.; Couplan, E.; Gonzalez-Barroso, M.D.; Petit, C.; Pagniez, A.; Chaumontet, C.; Mignotte, B.; et al. Adaptative metabolic response of human colonic epithelial cells to the adverse effects of the luminal compound sulfide. Biochim. Biophys. Acta 2005, 1725, 201–212. [Google Scholar] [CrossRef]
- Fu, Y.; Ito, M.; Fujita, Y.; Ito, M.; Ichihara, M.; Masuda, A.; Suzuki, Y.; Maesawa, S.; Kajita, Y.; Hirayama, M.; et al. Molecular hydrogen is protective against 6-hydroxydopamine-induced nigrostriatal degeneration in a rat model of Parkinson’s disease. Neurosci. Lett. 2009, 453, 81–85. [Google Scholar] [CrossRef]
- Rose, S.; Bennuri, S.C.; Davis, J.E.; Wynne, R.; Slattery, J.C.; Tippett, M.; Delhey, L.; Melnyk, S.; Kahler, S.G.; MacFabe, D.F.; et al. Butyrate enhances mitochondrial function during oxidative stress in cell lines from boys with autism. Transl. Psychiatry 2018, 8, 42. [Google Scholar] [CrossRef]
- Reese, A.T.; Cho, E.H.; Klitzman, B.; Nichols, S.P.; Wisniewski, N.A.; Villa, M.M.; Durand, H.K.; Jiang, S.; Midani, F.S.; Nimmagadda, S.N.; et al. Antibiotic-induced changes in the microbiota disrupt redox dynamics in the gut. eLife 2018, 7. [Google Scholar] [CrossRef] [PubMed]
- Teckentrup, V.; Neubert, S.; Santiago, J.C.P.; Hallschmid, M.; Walter, M.; Kroemer, N.B. Non-invasive stimulation of vagal afferents reduces gastric frequency. Brain Stimul. 2020, 13, 470–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luca, M.; Luca, A.; Calandra, C. Accelerated aging in major depression: The role of nitro-oxidative stress. Oxid. Med. Cell. Longev. 2013, 2013, 230797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anderson, G.; Maes, M. Oxidative/nitrosative stress and immuno-inflammatory pathways in depression: Treatment implications. Curr. Pharm. Des. 2014, 20, 3812–3847. [Google Scholar] [CrossRef]
- Dobashi, Y.; Miyakawa, Y.; Yamamoto, I.; Amao, H. Effects of intestinal microflora on superoxide dismutase activity in the mouse cecum. Exp. Anim. 2011, 60, 133–139. [Google Scholar] [CrossRef] [Green Version]
- Szczesniak, O.; Hestad, K.A.; Hanssen, J.F.; Rudi, K. Isovaleric acid in stool correlates with human depression. Nutr. Neurosci. 2016, 19, 279–283. [Google Scholar] [CrossRef]
- Han, L.; Zhao, L.; Zhou, Y.; Yang, C.; Xiong, T.; Lu, L.; Deng, Y.; Luo, W.; Chen, Y.; Qiu, Q.; et al. Altered metabolome and microbiome features provide clues in understanding irritable bowel syndrome and depression comorbidity. ISME J. 2021. [Google Scholar] [CrossRef]
- Lai, J.; Li, A.; Jiang, J.; Yuan, X.; Zhang, P.; Xi, C.; Wu, L.; Wang, Z.; Chen, J.; Lu, J.; et al. Metagenomic analysis reveals gut bacterial signatures for diagnosis and treatment outcome prediction in bipolar depression. Psychiatry Res. 2021, 307, 114326. [Google Scholar] [CrossRef]
- Meng, C.; Feng, S.; Hao, Z.; Dong, C.; Liu, H. Antibiotics exposure attenuates chronic unpredictable mild stress-induced anxiety-like and depression-like behavior. Psychoneuroendocrinology 2021, 136, 105620. [Google Scholar] [CrossRef]
- Gao, X.; Cao, Q.; Cheng, Y.; Zhao, D.; Wang, Z.; Yang, H.; Wu, Q.; You, L.; Wang, Y.; Lin, Y.; et al. Chronic stress promotes colitis by disturbing the gut microbiota and triggering immune system response. Proc. Natl. Acad. Sci. USA 2018, 115, E2960–E2969. [Google Scholar] [CrossRef] [Green Version]
- Martine, P.; Rebe, C. Heat Shock Proteins and Inflammasomes. Int. J. Mol. Sci. 2019, 20, 4508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pasquali, M.A.; Harlow, B.L.; Soares, C.N.; Otto, M.W.; Cohen, L.S.; Minuzzi, L.; Gelain, D.P.; Moreira, J.C.F.; Frey, B.N. A longitudinal study of neurotrophic, oxidative, and inflammatory markers in first-onset depression in midlife women. Eur. Arch. Psychiatry Clin. Neurosci. 2018, 268, 771–781. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.Y.; Roos, S.; Jonsson, H.; Ahl, D.; Dicksved, J.; Lindberg, J.E.; Lundh, T. Effects of Lactobacillus johnsonii and Lactobacillus reuteri on gut barrier function and heat shock proteins in intestinal porcine epithelial cells. Physiol. Rep. 2015, 3, e12355. [Google Scholar] [CrossRef] [PubMed]
- Song, J.; Zhou, N.; Ma, W.; Gu, X.; Chen, B.; Zeng, Y.; Yang, L.; Zhou, M. Modulation of gut microbiota by chlorogenic acid pretreatment on rats with adrenocorticotropic hormone induced depression-like behavior. Food Funct. 2019, 10, 2947–2957. [Google Scholar] [CrossRef] [PubMed]
- Chi, L.; Khan, I.; Lin, Z.; Zhang, J.; Lee, M.Y.S.; Leong, W.; Hsiao, W.L.W.; Zheng, Y. Fructo-oligosaccharides from Morinda officinalis remodeled gut microbiota and alleviated depression features in a stress rat model. Phytomedicine 2020, 67, 153157. [Google Scholar] [CrossRef] [PubMed]
- Maes, M.; Kubera, M.; Leunis, J.C.; Berk, M. Increased IgA and IgM responses against gut commensals in chronic depression: Further evidence for increased bacterial translocation or leaky gut. J. Affect. Disord. 2012, 141, 55–62. [Google Scholar] [CrossRef]
- Ceppa, F.; Mancini, A.; Tuohy, K. Current evidence linking diet to gut microbiota and brain development and function. Int. J. Food Sci. Nutr. 2019, 70, 1–19. [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]
- Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W.J. Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
- Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef]
- Roshchina, V.V. New Trends and Perspectives in the Evolution of Neurotransmitters in Microbial, Plant, and Animal Cells. Adv. Exp. Med. Biol. 2016, 874, 25–77. [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] [Green Version]
- Liu, Y.W.; Liu, W.H.; Wu, C.C.; Juan, Y.C.; Wu, Y.C.; Tsai, H.P.; Wang, S.; Tsai, Y.C. Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naive adult mice. Brain Res. 2016, 1631, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, S.; Wang, T.; Hu, X.; Luo, J.; Li, W.; Wu, X.; Duan, Y.; Jin, F. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015, 310, 561–577. [Google Scholar] [CrossRef] [PubMed]
- Savignac, H.M.; Kiely, B.; Dinan, T.G.; Cryan, J.F. Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol. Motil. 2014, 26, 1615–1627. [Google Scholar] [CrossRef] [PubMed]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bercik, P.; Verdu, E.F.; Foster, J.A.; Macri, J.; Potter, M.; Huang, X.; Malinowski, P.; Jackson, W.; Blennerhassett, P.; Neufeld, K.A.; et al. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology 2010, 139, 2102–2112. [Google Scholar] [CrossRef] [Green Version]
- Desbonnet, L.; Garrett, L.; Clarke, G.; Kiely, B.; Cryan, J.F.; Dinan, T.G. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience 2010, 170, 1179–1188. [Google Scholar] [CrossRef]
- Wang, H.; He, S.; Xin, J.; Zhang, T.; Sun, N.; Li, L.; Ni, X.; Zeng, D.; Ma, H.; Bai, Y. Psychoactive Effects of Lactobacillus johnsonii against Restraint Stress-Induced Memory Dysfunction in Mice through Modulating Intestinal Inflammation and permeability—A Study Based on the Gut-Brain Axis Hypothesis. Front. Pharmacol. 2021, 12, 662148. [Google Scholar] [CrossRef]
- Allen, A.P.; Hutch, W.; Borre, Y.E.; Kennedy, P.J.; Temko, A.; Boylan, G.; Murphy, E.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Bifidobacterium longum 1714 as a translational psychobiotic: Modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl. Psychiatry 2016, 6, e939. [Google Scholar] [CrossRef] [Green Version]
- Mohammadi, A.A.; Jazayeri, S.; Khosravi-Darani, K.; Solati, Z.; Mohammadpour, N.; Asemi, Z.; Adab, Z.; Djalali, M.; Tehrani-Doost, M.; Hosseini, M.; et al. The effects of probiotics on mental health and hypothalamic-pituitary-adrenal axis: A randomized, double-blind, placebo-controlled trial in petrochemical workers. Nutr. Neurosci. 2016, 19, 387–395. [Google Scholar] [CrossRef] [PubMed]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, L.H.; Liu, Y.W.; Wu, C.C.; Wang, S.; Tsai, Y.C. Psychobiotics in mental health, neurodegenerative and neurodevelopmental disorders. J. Food Drug Anal. 2019, 27, 632–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kazemi, A.; Noorbala, A.A.; Azam, K.; Eskandari, M.H.; Djafarian, K. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin. Nutr. 2019, 38, 522–528. [Google Scholar] [CrossRef] [PubMed]
- Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef]
- Liu, Y.; Wu, Z.; Cheng, L.; Zhang, X.; Yang, H. The role of the intestinal microbiota in the pathogenesis of host depression and mechanism of TPs relieving depression. Food Funct. 2021, 12, 7651–7663. [Google Scholar] [CrossRef]
- Zhou, N.; Gu, X.; Zhuang, T.; Xu, Y.; Yang, L.; Zhou, M. Gut Microbiota: A Pivotal Hub for Polyphenols as Antidepressants. J. Agric. Food Chem. 2020, 68, 6007–6020. [Google Scholar] [CrossRef]
- Walsh, C.J.; Guinane, C.M.; O’Toole, P.W.; Cotter, P.D. Beneficial modulation of the gut microbiota. FEBS Lett. 2014, 588, 4120–4130. [Google Scholar] [CrossRef] [Green Version]
- Lavefve, L.; Howard, L.R.; Carbonero, F. Berry polyphenols metabolism and impact on human gut microbiota and health. Food Funct. 2020, 11, 45–65. [Google Scholar] [CrossRef]
- Liu, X.; Cao, S.; Zhang, X. Modulation of Gut Microbiota-Brain Axis by Probiotics, Prebiotics, and Diet. J. Agric. Food Chem. 2015, 63, 7885–7895. [Google Scholar] [CrossRef]
- Zhou, J.; Tang, L.; Shen, C.L.; Wang, J.S. Green tea polyphenols modify gut-microbiota dependent metabolisms of energy, bile constituents and micronutrients in female Sprague-Dawley rats. J. Nutr. Biochem. 2018, 61, 68–81. [Google Scholar] [CrossRef] [PubMed]
- Rothenberg, D.O.; Zhang, L. Mechanisms Underlying the Anti-Depressive Effects of Regular Tea Consumption. Nutrients 2019, 11, 1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kelly, J.R.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbiota axis: Challenges for translation in psychiatry. Ann. Epidemiol. 2016, 26, 366–372. [Google Scholar] [CrossRef] [PubMed]
- Kawabata, K.; Kawai, Y.; Terao, J. Suppressive effect of quercetin on acute stress-induced hypothalamic-pituitary-adrenal axis response in Wistar rats. J. Nutr. Biochem. 2010, 21, 374–380. [Google Scholar] [CrossRef]
- Ge, J.F.; Peng, L.; Cheng, J.Q.; Pan, C.X.; Tang, J.; Chen, F.H.; Li, J. Antidepressant-like effect of resveratrol: Involvement of antioxidant effect and peripheral regulation on HPA axis. Pharmacol. Biochem. Behav. 2013, 114, 64–69. [Google Scholar] [CrossRef]
- Yrondi, A.; Sporer, M.; Peran, P.; Schmitt, L.; Arbus, C.; Sauvaget, A. Electroconvulsive therapy, depression, the immune system and inflammation: A systematic review. Brain Stimul. 2018, 11, 29–51. [Google Scholar] [CrossRef]
- Karasawa, K.; Uzuhashi, Y.; Hirota, M.; Otani, H. A matured fruit extract of date palm tree (Phoenix dactylifera L.) stimulates the cellular immune system in mice. J. Agric. Food Chem. 2011, 59, 11287–11293. [Google Scholar] [CrossRef]
- Perez-Berezo, T.; Franch, A.; Castellote, C.; Castell, M.; Perez-Cano, F.J. Mechanisms involved in down-regulation of intestinal IgA in rats by high cocoa intake. J. Nutr. Biochem. 2012, 23, 838–844. [Google Scholar] [CrossRef] [Green Version]
- Cuevas, A.; Saavedra, N.; Salazar, L.A.; Abdalla, D.S. Modulation of immune function by polyphenols: Possible contribution of epigenetic factors. Nutrients 2013, 5, 2314–2332. [Google Scholar] [CrossRef] [Green Version]
- Borutinskaite, V.; Virksaite, A.; Gudelyte, G.; Navakauskiene, R. Green tea polyphenol EGCG causes anti-cancerous epigenetic modulations in acute promyelocytic leukemia cells. Leuk. Lymphoma 2018, 59, 469–478. [Google Scholar] [CrossRef]
- Inserra, A.; Rogers, G.B.; Licinio, J.; Wong, M.L. The Microbiota-Inflammasome Hypothesis of Major Depression. BioEssays 2018, 40, e1800027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moreno-Indias, I.; Sanchez-Alcoholado, L.; Perez-Martinez, P.; Andres-Lacueva, C.; Cardona, F.; Tinahones, F.; Queipo-Ortuno, M.I. Red wine polyphenols modulate fecal microbiota and reduce markers of the metabolic syndrome in obese patients. Food Funct. 2016, 7, 1775–1787. [Google Scholar] [CrossRef] [Green Version]
- Barrett, E.; Ross, R.P.; O’Toole, P.W.; Fitzgerald, G.F.; Stanton, C. Gamma-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef]
- Gonthier, M.P.; Verny, M.A.; Besson, C.; Remesy, C.; Scalbert, A. Chlorogenic acid bioavailability largely depends on its metabolism by the gut microflora in rats. J. Nutr. 2003, 133, 1853–1859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Lin, D.; Zhang, C.; Li, G.; Zhang, N.; Ruan, L.; Yan, Q.; Li, J.; Yu, X.; Xie, X.; et al. Antidepressant-like effects of ferulic acid: Involvement of serotonergic and norepinergic systems. Metab. Brain Dis. 2015, 30, 129–136. [Google Scholar] [CrossRef] [PubMed]
- Zeni, A.L.B.; Camargo, A.; Dalmagro, A.P. Ferulic acid reverses depression-like behavior and oxidative stress induced by chronic corticosterone treatment in mice. Steroids 2017, 125, 131–136. [Google Scholar] [CrossRef] [PubMed]
- Takeda, H.; Tsuji, M.; Inazu, M.; Egashira, T.; Matsumiya, T. Rosmarinic acid and caffeic acid produce antidepressive-like effect in the forced swimming test in mice. Eur. J. Pharmacol. 2002, 449, 261–267. [Google Scholar] [CrossRef]
- Tomas-Barberan, F.A.; Selma, M.V.; Espin, J.C. Interactions of gut microbiota with dietary polyphenols and consequences to human health. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 471–476. [Google Scholar] [CrossRef]
- Parkar, S.G.; Trower, T.M.; Stevenson, D.E. Fecal microbial metabolism of polyphenols and its effects on human gut microbiota. Anaerobe 2013, 23, 12–19. [Google Scholar] [CrossRef]
- Caspani, G.; Kennedy, S.; Foster, J.A.; Swann, J. Gut microbial metabolites in depression: Understanding the biochemical mechanisms. Microb. Cell 2019, 6, 454–481. [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]
- Skonieczna-Zydecka, K.; Grochans, E.; Maciejewska, D.; Szkup, M.; Schneider-Matyka, D.; Jurczak, A.; Loniewski, I.; Kaczmarczyk, M.; Marlicz, W.; Czerwinska-Rogowska, M.; et al. Faecal Short Chain Fatty Acids Profile is Changed in Polish Depressive Women. Nutrients 2018, 10, 1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Hou, L.; Wang, C.; Jia, X.; Qin, X.; Wu, C. Short Term Intrarectal Administration of Sodium Propionate Induces Antidepressant-Like Effects in Rats Exposed to Chronic Unpredictable Mild Stress. Front. Psychiatry 2018, 9, 454. [Google Scholar] [CrossRef] [PubMed]
- Nankova, B.B.; Agarwal, R.; MacFabe, D.F.; La Gamma, E.F. Enteric bacterial metabolites propionic and butyric acid modulate gene expression, including CREB-dependent catecholaminergic neurotransmission, in PC12 cells—Possible relevance to autism spectrum disorders. PLoS ONE 2014, 9, e103740. [Google Scholar] [CrossRef] [PubMed]
- Graff, J.; Tsai, L.H. The potential of HDAC inhibitors as cognitive enhancers. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 311–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef]
- Appleton, K.M.; Sallis, H.M.; Perry, R.; Ness, A.R.; Churchill, R. Omega-3 fatty acids for depression in adults. Cochrane Database Syst. Rev. 2015, 11, CD004692. [Google Scholar] [CrossRef]
- Mocking, R.J.; Harmsen, I.; Assies, J.; Koeter, M.W.; Ruhe, H.G.; Schene, A.H. Meta-analysis and meta-regression of omega-3 polyunsaturated fatty acid supplementation for major depressive disorder. Transl. Psychiatry 2016, 6, e756. [Google Scholar] [CrossRef]
- Mozaffari-Khosravi, H.; Yassini-Ardakani, M.; Karamati, M.; Shariati-Bafghi, S.E. Eicosapentaenoic acid versus docosahexaenoic acid in mild-to-moderate depression: A randomized, double-blind, placebo-controlled trial. Eur. Neuropsychopharmacol. 2013, 23, 636–644. [Google Scholar] [CrossRef]
- Mischoulon, D.; Best-Popescu, C.; Laposata, M.; Merens, W.; Murakami, J.L.; Wu, S.L.; Papakostas, G.I.; Dording, C.M.; Sonawalla, S.B.; Nierenberg, A.A.; et al. A double-blind dose-finding pilot study of docosahexaenoic acid (DHA) for major depressive disorder. Eur. Neuropsychopharmacol. 2008, 18, 639–645. [Google Scholar] [CrossRef]
- Malaspina, A.; Michael-Titus, A.T. Is the modulation of retinoid and retinoid-associated signaling a future therapeutic strategy in neurological trauma and neurodegeneration? J. Neurochem. 2008, 104, 584–595. [Google Scholar] [CrossRef]
- Sherwin, J.C.; Reacher, M.H.; Dean, W.H.; Ngondi, J. Epidemiology of vitamin A deficiency and xerophthalmia in at-risk populations. Trans. R. Soc. Trop. Med. Hyg. 2012, 106, 205–214. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.W.; Zhang, K.W.; Chen, S.J.; Yang, C.; Li, P.G. Vitamin A Deficiency Exacerbates Gut Microbiota Dysbiosis and Cognitive Deficits in Amyloid Precursor Protein/Presenilin 1 Transgenic Mice. Front. Aging Neurosci. 2021, 13, 753351. [Google Scholar] [CrossRef] [PubMed]
- Lyu, Y.; Wu, L.; Wang, F.; Shen, X.; Lin, D. Carotenoid supplementation and retinoic acid in immunoglobulin A regulation of the gut microbiota dysbiosis. Exp. Biol. Med. 2018, 243, 613–620. [Google Scholar] [CrossRef] [Green Version]
- Elsherbiny, N.M.; Salama, M.F.; Said, E.; El-Sherbiny, M.; Al-Gayyar, M.M. Crocin protects against doxorubicin-induced myocardial toxicity in rats through down-regulation of inflammatory and apoptic pathways. Chem. Biol. Interact. 2016, 247, 39–48. [Google Scholar] [CrossRef] [PubMed]
- Hosseinzadeh, H.; Sadeghnia, H.R.; Ghaeni, F.A.; Motamedshariaty, V.S.; Mohajeri, S.A. Effects of saffron (Crocus sativus L.) and its active constituent, crocin, on recognition and spatial memory after chronic cerebral hypoperfusion in rats. Phytother. Res. 2012, 26, 381–386. [Google Scholar] [CrossRef] [PubMed]
- Shafiee, M.; Arekhi, S.; Omranzadeh, A.; Sahebkar, A. Saffron in the treatment of depression, anxiety and other mental disorders: Current evidence and potential mechanisms of action. J. Affect. Disord. 2018, 227, 330–337. [Google Scholar] [CrossRef] [PubMed]
- Vahdati Hassani, F.; Naseri, V.; Razavi, B.M.; Mehri, S.; Abnous, K.; Hosseinzadeh, H. Antidepressant effects of crocin and its effects on transcript and protein levels of CREB, BDNF, and VGF in rat hippocampus. DARU J. Pharm. Sci. 2014, 22, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, Q.; Shu, R.; Wu, C.; Tong, Y.; Xiong, Z.; Zhou, J.; Yu, C.; Xie, X.; Fu, Z. Crocin-I alleviates the depression-like behaviors probably via modulating “microbiota-gut-brain” axis in mice exposed to chronic restraint stress. J. Affect. Disord. 2020, 276, 476–486. [Google Scholar] [CrossRef]
- Zhao, B.; Wu, J.; Li, J.; Bai, Y.; Luo, Y.; Ji, B.; Xia, B.; Liu, Z.; Tan, X.; Lv, J.; et al. Lycopene Alleviates DSS-Induced Colitis and Behavioral Disorders via Mediating Microbes-Gut-Brain Axis Balance. J. Agric. Food Chem. 2020, 68, 3963–3975. [Google Scholar] [CrossRef]
- Ge, H.; Yang, T.; Sun, J.; Zhang, D. Associations between dietary carotenoid intakes and the risk of depressive symptoms. Food Nutr. Res. 2020, 64, 3920. [Google Scholar] [CrossRef] [PubMed]
- Kondo, S.; El Omri, A.; Han, J.; Isoda, H. Antidepressant-like effects of rosmarinic acid through mitogen-activated protein kinase phosphatase-1 and brain-derived neurotrophic factor modulation. J. Funct. Foods 2015, 14, 758–766. [Google Scholar] [CrossRef] [Green Version]
- Girish, C.; Raj, V.; Arya, J.; Balakrishnan, S. Evidence for the involvement of the monoaminergic system, but not the opioid system in the antidepressant-like activity of ellagic acid in mice. Eur. J. Pharmacol. 2012, 682, 118–125. [Google Scholar] [CrossRef] [PubMed]
- Gujral, S.; Aizenstein, H.; Reynolds, C.F., 3rd; Butters, M.A.; Erickson, K.I. Exercise effects on depression: Possible neural mechanisms. Gen. Hosp. Psychiatry 2017, 49, 2–10. [Google Scholar] [CrossRef]
- Ernst, C.; Olson, A.K.; Pinel, J.P.; Lam, R.W.; Christie, B.R. Antidepressant effects of exercise: Evidence for an adult-neurogenesis hypothesis? J. Psychiatry Neurosci. 2006, 31, 84–92. [Google Scholar]
- Dalton, A.; Mermier, C.; Zuhl, M. Exercise influence on the microbiome-gut-brain axis. Gut Microbes 2019, 10, 555–568. [Google Scholar] [CrossRef]
- Sheng, L.; Wang, Y.; Jiang, A.; Zhou, Y.; Zhou, H. Effect of Regular Physical Exercise on Gut Microbiota and Depressive Behaviors in Rats. J. Food Qual. 2021, 2021, 1–8. [Google Scholar] [CrossRef]
- Nay, K.; Smiles, W.J.; Kaiser, J.; McAloon, L.M.; Loh, K.; Galic, S.; Oakhill, J.S.; Gundlach, A.L.; Scott, J.W. Molecular Mechanisms Underlying the Beneficial Effects of Exercise on Brain Function and Neurological Disorders. Int. J. Mol. Sci. 2021, 22, 4052. [Google Scholar] [CrossRef]
- Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [Green Version]
- Mailing, L.J.; Allen, J.M.; Buford, T.W.; Fields, C.J.; Woods, J.A. Exercise and the Gut Microbiome: A Review of the Evidence, Potential Mechanisms, and Implications for Human Health. Exerc. Sport Sci. Rev. 2019, 47, 75–85. [Google Scholar] [CrossRef]
- Donati Zeppa, S.; Amatori, S.; Sisti, D.; Gervasi, M.; Agostini, D.; Piccoli, G.; Pazienza, V.; Gobbi, P.; Rocchi, M.B.L.; Sestili, P.; et al. Nine weeks of high-intensity indoor cycling training induced changes in the microbiota composition in non-athlete healthy male college students. J. Int. Soc. Sports Nutr. 2021, 18, 74. [Google Scholar] [CrossRef] [PubMed]
- Queipo-Ortuno, M.I.; Seoane, L.M.; Murri, M.; Pardo, M.; Gomez-Zumaquero, J.M.; Cardona, F.; Casanueva, F.; Tinahones, F.J. Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 2013, 8, e65465. [Google Scholar] [CrossRef] [PubMed]
- Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sports Exerc. 2018, 50, 747–757. [Google Scholar] [CrossRef] [PubMed]
- McFadzean, R. Exercise Can Help Modulate Human Gut Microbiota. Bachelor’s Thesis, University of Colorado Boulder, Boulder, CO, USA, 2014. [Google Scholar]
- Petersen, L.M.; Bautista, E.J.; Nguyen, H.; Hanson, B.M.; Chen, L.; Lek, S.H.; Sodergren, E.; Weinstock, G.M. Community characteristics of the gut microbiomes of competitive cyclists. Microbiome 2017, 5, 98. [Google Scholar] [CrossRef]
- Clark, A.; Mach, N. Exercise-induced stress behavior, gut-microbiota-brain axis and diet: A systematic review for athletes. J. Int. Soc. Sports Nutr. 2016, 13, 43. [Google Scholar] [CrossRef] [Green Version]
- Claes, S.; Myint, A.M.; Domschke, K.; Del-Favero, J.; Entrich, K.; Engelborghs, S.; De Deyn, P.; Mueller, N.; Baune, B.; Rothermundt, M. The kynurenine pathway in major depression: Haplotype analysis of three related functional candidate genes. Psychiatry Res. 2011, 188, 355–360. [Google Scholar] [CrossRef]
Category | Treatment | Possible Mechanism of Action | Model | Reference |
---|---|---|---|---|
Probiotics | Lactobacillus brevis | ↑ GABA | Human | [31] |
Bifidobacterium dentium | ↑ GABA ↑ 5-HT | Mice | [31] | |
Lactobacillus plantarum | ↑ GABA ↑ 5-HT ↑ ACh | Mice | [31] | |
Lactobacillus odontolyticus | ↑ ACh | Mice | [92] | |
Lactobacillus plantarum PS128 | ↓ Pro-inflammatory cytokine ↑ Anti-inflammatory cytokine ↑ DA ↑ 5-HT | Mice | [94] | |
Lactobacillus helveticus NS8 | ↑ BDNF ↑ NE ↑ 5-HT | Sprague–Dawley rats | [95] | |
Lactobacillus rhamnosus (JB-1) | ↑ GABA B 1b (cortical regions) ↓ GABA B 1b (hippocampus, amygdala, and locus coeruleus) ↓ GABA A α2 (prefrontal cortex and amygdala) ↑ GABA A α2 (hippocampus) | Adult male BALB/c mice | [97] | |
Lactobacillus johnsonii BS15 | ↑ Anti-inflammatory cytokine | C57BL/6J male mice | [100] | |
Bifidobacterium longum 1714 | - | Healthy human males | [101] | |
Probiotic yogurt (Lactobacillus acidophilus LA5 and Bifidobacterium lactis BB12) | ↑ Mental health (DASS, GHQ) | Human | [102] | |
Probiotic capsule (Lactobacillus casei, L. acidophilus, and L. rhamnosus) | ↑ Mental health (DASS, GHQ) | Human | [102] | |
Prebiotics | Fructo-oligosaccharides (FOS) Galacto-oligosaccharides (GOS) | ↑ Acetate ↑ Propionate ↓ Isobutyrate ↓ Pro-inflammatory cytokine | C57BL/6J male mice | [106] |
PUFAs | Eicosapentaenoic acid (EPA) Docosahexaenoic acid (DHA) | - | Human | [139] |
SCFAs | Propionate | ↑ NE ↑ DA ↑ TPH | Sprague–Dawley rats | [134,135] |
Polyphenols | Quercetin | ↓ CRF ↓ HPA activation ↑ SCFAs | Wistar rats | [115] |
Resveratrol | Neuroprotective | Sprague–Dawley rats | [116] | |
Ferulic acid | ↓ MAO-A ↑ Antioxidant defense ↑ Monoaminergic system ↑ SCFAs | Mice | [126,127] | |
Caffeic acid | ↓ MAO ↓ Monoammine uptake ↑ SCFAs | ICR Mice | [128] | |
Rosmarinic acid | ↓ Corticosterone ↑ Dopamine ↓ Mkp-1 ↑ BDNF | Mice | [153] | |
Ellagic acid | ↑ 5-HT ↑ NE | Mice | [154] | |
Quercetin, Chlorogenic acid, and Caffeic acid | Bifidobacterium Bacteroidetes Firmicutes | Human | [130] | |
Tea | Tea saponin, l-theanine, Epigallocatechin gallate, and Catechine | ↓ NF-kB ↑ DA | - | [113] |
Lycopene | - | ↓ Neuroinflammation ↑ Neurotrophic factor ↑ SCFAs ↑ Bifidobacterium ↑ Lactobacillus | Mice | [151] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Donati Zeppa, S.; Ferrini, F.; Agostini, D.; Amatori, S.; Barbieri, E.; Piccoli, G.; Sestili, P.; Stocchi, V. Nutraceuticals and Physical Activity as Antidepressants: The Central Role of the Gut Microbiota. Antioxidants 2022, 11, 236. https://doi.org/10.3390/antiox11020236
Donati Zeppa S, Ferrini F, Agostini D, Amatori S, Barbieri E, Piccoli G, Sestili P, Stocchi V. Nutraceuticals and Physical Activity as Antidepressants: The Central Role of the Gut Microbiota. Antioxidants. 2022; 11(2):236. https://doi.org/10.3390/antiox11020236
Chicago/Turabian StyleDonati Zeppa, Sabrina, Fabio Ferrini, Deborah Agostini, Stefano Amatori, Elena Barbieri, Giovanni Piccoli, Piero Sestili, and Vilberto Stocchi. 2022. "Nutraceuticals and Physical Activity as Antidepressants: The Central Role of the Gut Microbiota" Antioxidants 11, no. 2: 236. https://doi.org/10.3390/antiox11020236