Exercise-induced stress behavior, gut-microbiota-brain axis and diet: a systematic review for athletes

Allison Clark, Núria Mach, Allison Clark, Núria Mach

Abstract

Fatigue, mood disturbances, under performance and gastrointestinal distress are common among athletes during training and competition. The psychosocial and physical demands during intense exercise can initiate a stress response activating the sympathetic-adrenomedullary and hypothalamus-pituitary-adrenal (HPA) axes, resulting in the release of stress and catabolic hormones, inflammatory cytokines and microbial molecules. The gut is home to trillions of microorganisms that have fundamental roles in many aspects of human biology, including metabolism, endocrine, neuronal and immune function. The gut microbiome and its influence on host behavior, intestinal barrier and immune function are believed to be a critical aspect of the brain-gut axis. Recent evidence in murine models shows that there is a high correlation between physical and emotional stress during exercise and changes in gastrointestinal microbiota composition. For instance, induced exercise-stress decreased cecal levels of Turicibacter spp and increased Ruminococcus gnavus, which have well defined roles in intestinal mucus degradation and immune function. Diet is known to dramatically modulate the composition of the gut microbiota. Due to the considerable complexity of stress responses in elite athletes (from leaky gut to increased catabolism and depression), defining standard diet regimes is difficult. However, some preliminary experimental data obtained from studies using probiotics and prebiotics studies show some interesting results, indicating that the microbiota acts like an endocrine organ (e.g. secreting serotonin, dopamine or other neurotransmitters) and may control the HPA axis in athletes. What is troubling is that dietary recommendations for elite athletes are primarily based on a low consumption of plant polysaccharides, which is associated with reduced microbiota diversity and functionality (e.g. less synthesis of byproducts such as short chain fatty acids and neurotransmitters). As more elite athletes suffer from psychological and gastrointestinal conditions that can be linked to the gut, targeting the microbiota therapeutically may need to be incorporated in athletes' diets that take into consideration dietary fiber as well as microbial taxa not currently present in athlete's gut.

Keywords: Athlete; Behaviour; Diet; Exercise; Microbiota; Neurotransmitters; Stress.

Figures

Fig. 1
Fig. 1
Stress hormones released during high intense exercise. Stress responses to intense exercise are mediated by largely overlapping circuits in the limbic forebrain, the hypothalamus and the brainstem, so that the respective contributions on the neuroendocrine and autonomic systems are tuned in accordance with stressor and intensity [6]. When brainstem receives inputs that signal major homeostatic perturbations, such as respiratory distress, energy imbalance, desydration, visceral or somatic pain, inflammation or exteroceptive factors respond through a coordinated modulation of the HPA axis and the sympathetic and parasympathetic branch of the autonomic nervous system (ANS). By contrast, forebrain limbic regions have no direct connections with the HPA axis or the ANS and thus require intervening synapses before they can access autonomic or neuroendocrine neurons (top-down regulation) [6]. Briefly, exercise-induced stress results in activation of preganglionic sympathetic neurons in the intermediolateral cell column of the thoracolumbar spinal cord (shown in purple and clear grey). This sympathetic activation represents the classic 'fight or flight' response and it generally increases circulating levels of catecholamines. Parasympathetic tone can also be modulated during stress (shown in dark grey color). Parasympathetic actions are generally opposite to those of the sympathetic system and alter the vagal tone to the heart and lungs. Within the HPA axis, stress activates hypophysiotropic neurons in the paraventricular nucleus of the hypothalamus (PVN) that secrete releasing hormones, such as corticotrophin-releasing hormone (CRH) and arginine vasopressin (AVP), into the portal circulation of the median eminence. These releasing hormones act on the anterior pituitary to promote the secretion of adrenocorticotropic hormone (ACTH), which in turn acts on the inner adrenal cortex to initiate the synthesis and release of glucocorticoid hormones. Moreover, the adrenal cortex is directly innervated by the sympathetic nervous system, which can also regulate corticosteroid release. Additionally, gastrointestinal tract responds to stress in an endocrine manner by releasing hormones such as Gamma-amino butyric acid (GABA), neuropeptide Y and dopamine that have been purported to be involved in the gastrointestinal disturbances, anxiety, depression, reduced food intake and less stress coping. Microorganisms that colonize the digestive tract can be involved in the regulation of the HPA axis through the regulation or production of short chain fatty acids and neurotransmitters such as GABA, dopamine and serotonin, as well as cytokines. The neuroendocrine stress response to exercise is determined not only by the emotional stress but the volume of physical exposure, where volume consists of the intensity and/or duration of the exercise session. As exercise intensity is increased, there are approximately proportional increases in circulating concentrations of ACTH and cortisol. There is a critical threshold of exercise intensity that must be reached (~50–60% of maximal oxygen uptake [VO2max]) before circulating levels increase in response to exercise [170, 171]
Fig. 2
Fig. 2
Gastrointestinal disruption during high intensity exercise. Proper intestinal barrier function is crucial for maintaining health and immunity. During intense exercise, athletes’ body temperature increases and blood pools away from the gastrointestinal tract to periphery muscles and organs such as the heart and lungs during intense physical activity [62]. The redistribution of blood flow away from the intestines together with thermal damage to the intestinal mucosa can cause intestinal barrier disruption, followed by an inflammatory response [63]. Additionally, intense exercise over a prolonged period of time increase stress hormones and lipopolysaccharides (LPS) translocation in the gastrointestinal tract, which triggers immune responses that often results in increased pro-inflammatory cytokines and intestinal permeability. Additionally, intestinal permeability may be made worse by the increased production of reactive oxygen species (ROS) and by the alteration of gut-microbiota composition and activity (the so-called dysbiosis). Furthermore, gastrointestinal tract responds to stress by releasing hormones such as GABA, neuropeptide Y (NPY) and dopamine that have been purported to cause GI disturbances, anxiety, depression, reduced food intake and less stress coping [9]. Conversely, the microbiota’s production of butyrate and propionate can increase transepithelial resistance, which improves intestinal barrier function and decreases inflammation.
Fig. 3
Fig. 3
Gut microbiota effects on mood disturbance, fatigue, insomnia and risk of depression during exercise. The putative mechanisms by which bacteria connects with the brain and influence behavior during exercise include bacterial subproducts that gain access to the brain via the bloodstream and the area postrema, via cytokine release from mucosal immune cells, via the release of gut hormones such as 5-hydroxytryptamine (5-HT) from enteroendocrine cells, or via afferent neural pathways, including the vagus nerve. Stress during intense period of training and competitions can influence the microbial composition of the gut through the release of stress hormones or sympathetic neurotransmitters that influence gut physiology and alter the habitat of the microbiota (reviewed by Mach [23]). Alternatively, host stress hormones such as noradrenaline might influence bacterial gene expression or signaling between bacteria, and this might change the microbial composition and activity of the microbiota.

References

    1. Galley JD, Nelson MC, Yu Z, et al. Exposure to a social stressor disrupts the community structure of the colonic mucosa-associated microbiota. BMC Microbiol. 2014;14:189. doi: 10.1186/1471-2180-14-189.
    1. Morgan JA, Corrigan F, Baune BT. Effects of physical exercise on central nervous system functions: A review of brain region specific adaptations. J Mol Psychiatry. 2015;3:3. doi: 10.1186/s40303-015-0010-8.
    1. Lin TW, Chen SJ, Huang TY, et al. Different types of exercise induce differential effects on neuronal adaptations and memory performance. Neurobiol Learn Mem. 2012;97:140–7. doi: 10.1016/j.nlm.2011.10.006.
    1. Purvis D, Gonsalves S, Deuster PA. Physiological and psychological fatigue in extreme conditions: Overtraining and elite athletes. PM R. 2010;2:442–50. doi: 10.1016/j.pmrj.2010.03.025.
    1. Mackinnon LT. Overtraining effects on immunity and performance in athletes. Immun Cell Biol. 2000;78:502–509. doi: 10.1111/j.1440-1711.2000.t01-7-.x.
    1. Ulrich-Lai YM, Herman JP. Neural regulation of endocrine and autonomic stress responses. Nat Rev Neurosci. 2009;10:397–409. doi: 10.1038/nrn2647.
    1. Martins AS, Crescenzi A, Stern JE, et al. Hypertension and exercise training differentially affect oxytocin and oxytocin receptor expression in the brain. Hypertension. 2005;46:1004–9. doi: 10.1161/01.HYP.0000175812.03322.59.
    1. Eisenstein M. Microbiome: Bacterial broadband. Nature. 2016;533:104–6. doi: 10.1038/533S104a.
    1. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol. 2009;6:306–14. doi: 10.1038/nrgastro.2009.35.
    1. Lyte M, Vulchanova L, Brown DR. Stress at the intestinal surface: Catecholamines and mucosa-bacteria interactions. Cell Tissue Res. 2011;343:23–32. doi: 10.1007/s00441-010-1050-0.
    1. Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014;13:69–86. doi: 10.1111/gbb.12109.
    1. Li J, Jia H, Cai X, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–41. doi: 10.1038/nbt.2942.
    1. Rajilic-Stojanovic M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. Fems Microbiology Reviews. 2014;38:996–1047. doi: 10.1111/1574-6976.12075.
    1. Hsu YJ, Chiu CC, Li YP, et al. Effect of intestinal microbiota on exercise performance in mice. J Strength Cond Res. 2015;29:552–8. doi: 10.1519/JSC.0000000000000644.
    1. Mach N, Berri M, Estelle J, et al. Early-life establishment of the swine gut microbiome and impact on host phenotypes. Environ Microbiol Rep. 2015;7:554–69. doi: 10.1111/1758-2229.12285.
    1. Ramayo-Caldas Y, Mach N, Lepage P, et al.: Phylogenetic network analysis applied to pig gut microbiota identifies an ecosystem structure linked with growth traits. ISME 2016 (In press).
    1. Sugahara H, Odamaki T, Hashikura N, et al. Differences in folate production by bifidobacteria of different origins. Biosci Microbiota Food Health. 2015;34:87–93. doi: 10.12938/bmfh.2015-003.
    1. Marley MG, Meganathan R, Bentley R. Menaquinone (vitamin k2) biosynthesis in escherichia coli: Synthesis of o-succinylbenzoate does not require the decarboxylase activity of the ketoglutarate dehydrogenase complex. Biochemistry. 1986;25:1304–7. doi: 10.1021/bi00354a017.
    1. Flint HJ, Bayer EA, Rincon MT, et al. Polysaccharide utilization by gut bacteria: Potential for new insights from genomic analysis. Nat Rev Microbiol. 2008;6:121–31. doi: 10.1038/nrmicro1817.
    1. Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7. doi: 10.1126/science.1223813.
    1. Clarke G, Stilling RM, Kennedy PJ, et al. Minireview: Gut microbiota: The neglected endocrine organ. Mol Endocrinol. 2014;28:1221–38. doi: 10.1210/me.2014-1108.
    1. Moloney RD, Desbonnet L, Clarke G, et al. The microbiome: Stress, health and disease. Mamm Genome. 2014;25:49–74. doi: 10.1007/s00335-013-9488-5.
    1. Mach N and Fuster-Botella D: Endurance exercise and gut microbiota: A review. J Sport Health Sci 2016 (In press).
    1. Fasano A. Leaky gut and autoimmune diseases. Clin Rev Allergy Immunol. 2012;42:71–8. doi: 10.1007/s12016-011-8291-x.
    1. Zhang C, Zhang M, Wang S, et al. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 2010;4:232–41. doi: 10.1038/ismej.2009.112.
    1. David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. doi: 10.1038/nature12820.
    1. Aguirre M, Eck A, Koenen ME, et al. Diet drives quick changes in the metabolic activity and composition of human gut microbiota in a validated in vitro gut model. Res Microbiol. 2016;167:114–25. doi: 10.1016/j.resmic.2015.09.006.
    1. Rodriguez NR, Di Marco NM, Langley S. American college of sports medicine position stand. Nutrition and athletic performance. Med Sci Sports Exerc. 2009;41:709–31. doi: 10.1249/MSS.0b013e31890eb86.
    1. Herman JP. Neural control of chronic stress adaptation. Front Behav Neurosci. 2013;7:61. doi: 10.3389/fnbeh.2013.00061.
    1. Angeli A, Minetto M, Dovio A, et al. The overtraining syndrome in athletes: A stress-related disorder. J Endocrinol Invest. 2004;27:603–12. doi: 10.1007/BF03347487.
    1. Hill EE, Zack E, Battaglini C, et al. Exercise and circulating cortisol levels: The intensity threshold effect. J Endocrinol Invest. 2008;31:587–91. doi: 10.1007/BF03345606.
    1. Lehmann M, Foster C, Dickhuth HH, et al. Autonomic imbalance hypothesis and overtraining syndrome. Med Sci Sports Exerc. 1998;30:1140–5. doi: 10.1097/00005768-199807000-00019.
    1. Lehmann M, Schnee W, Scheu R, et al. Decreased nocturnal catecholamine excretion: Parameter for an overtraining syndrome in athletes? Int J Sports Med. 1992;13:236–42. doi: 10.1055/s-2007-1021260.
    1. Cronin O, Molloy MG, Shanahan F. Exercise, fitness, and the gut. Curr Opin Gastroenterol. 2016;32:67–73. doi: 10.1097/MOG.0000000000000240.
    1. Carabotti M, Scirocco A, Maselli MA, et al. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28:203–209.
    1. Marchesi JR, Adams DH, Fava F, et al. The gut microbiota and host health: A new clinical frontier. Gut. 2016;65:330–9. doi: 10.1136/gutjnl-2015-309990.
    1. Holzer P, Farzi A. Neuropeptides and the microbiota-gut-brain axis. Adv Exp Med Biol. 2014;817:195–219. doi: 10.1007/978-1-4939-0897-4_9.
    1. Barrett E, Ross RP, O'Toole PW, et al. Gamma-aminobutyric acid production by culturable bacteria from the human intestine. J Appl Microbiol. 2012;113:411–7. doi: 10.1111/j.1365-2672.2012.05344.x.
    1. Hsu YC, Chen HI, Kuo YM, et al. Chronic treadmill running in normotensive rats resets the resting blood pressure to lower levels by upregulating the hypothalamic gabaergic system. J Hypertens. 2011;29:2339–48. doi: 10.1097/HJH.0b013e32834c628f.
    1. de Groote L, Linthorst AC. Exposure to novelty and forced swimming evoke stressor-dependent changes in extracellular gaba in the rat hippocampus. Neuroscience. 2007;148:794–805. doi: 10.1016/j.neuroscience.2007.06.030.
    1. Ramson R, Jurimae J, Jurimae T, et al. The effect of 4-week training period on plasma neuropeptide y, leptin and ghrelin responses in male rowers. Eur J Appl Physiol. 2012;112:1873–80. doi: 10.1007/s00421-011-2166-y.
    1. Saunders PR, Santos J, Hanssen NP, et al. Physical and psychological stress in rats enhances colonic epithelial permeability via peripheral crh. Dig Dis Sci. 2002;47:208–15. doi: 10.1023/A:1013204612762.
    1. Eisenhofer G, Aneman A, Friberg P, et al. Substantial production of dopamine in the human gastrointestinal tract. J Clin Endocrinol Metab. 1997;82:3864–71. doi: 10.1210/jcem.82.11.4339.
    1. Heijnen S, Hommel B, Kibele A, et al. Neuromodulation of aerobic exercise-a review. Front Psychol. 2015;6:1890.
    1. Foster J. Gut-brain communication: How the microbiome influences anxiety and depression. Europ Neuro Psycho Pharmacol. 2015;25:14.
    1. Crumeyrolle-Arias M, Jaglin M, Bruneau A, et al. Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psycho Neuro Endocrinol. 2014;42:207–217. doi: 10.1016/j.psyneuen.2014.01.014.
    1. Sudo N, Chida Y, Aiba Y, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558:263–75. doi: 10.1113/jphysiol.2004.063388.
    1. Asano Y, Hiramoto T, Nishino R, et al. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am J Physiol Gastrointest Liver Physiol. 2012;303:1288–95. doi: 10.1152/ajpgi.00341.2012.
    1. Bravo JA, Forsythe P, Chew MV, et al. Ingestion of lactobacillus strain regulates emotional behavior and central gaba receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011;108:16050–5. doi: 10.1073/pnas.1102999108.
    1. Mirescu C, Peters JD, Gould E. Early life experience alters response of adult neurogenesis to stress. Nat Neurosci. 2004;7:841–6. doi: 10.1038/nn1290.
    1. Bermon S, Petriz B, Kajeniene A, et al. The microbiota: An exercise immunology perspective. Exerc Immunol Rev. 2015;21:70–9.
    1. Allen JM, Berg Miller ME, Pence BD, et al. Voluntary and forced exercise differentially alters the gut microbiome in c57bl/6j mice. J Appl Physiol (1985) 2015;118:1059–66. doi: 10.1152/japplphysiol.01077.2014.
    1. de Oliveira EP, Burini RC, Jeukendrup A. Gastrointestinal complaints during exercise: Prevalence, etiology, and nutritional recommendations. Sports Med. 2014;44(Suppl 1):S79–85. doi: 10.1007/s40279-014-0153-2.
    1. Gareau MG, Silva MA, Perdue MH. Pathophysiological mechanisms of stress-induced intestinal damage. Curr Mol Med. 2008;8:274–81. doi: 10.2174/156652408784533760.
    1. Rao R, Samak G. Role of glutamine in protection of intestinal epithelial tight junctions. J Epithel Biol Pharmacol. 2012;5:47–54. doi: 10.2174/1875044301205010047.
    1. Brown WM, Davison GW, McClean CM, et al. A systematic review of the acute effects of exercise on immune and inflammatory indices in untrained adults. Sports Med Open. 2015;1:35. doi: 10.1186/s40798-015-0032-x.
    1. Zuhl M, Schneider S, Lanphere K, et al. Exercise regulation of intestinal tight junction proteins. Br J Sports Med. 2014;48:980–6. doi: 10.1136/bjsports-2012-091585.
    1. Ferrier L. Significance of increased human colonic permeability in response to corticotrophin-releasing hormone (crh) Gut. 2008;57:7–9. doi: 10.1136/gut.2007.129841.
    1. Wallon C, Yang PC, Keita AV, et al. Corticotropin-releasing hormone (crh) regulates macromolecular permeability via mast cells in normal human colonic biopsies in vitro. Gut. 2008;57:50–8. doi: 10.1136/gut.2006.117549.
    1. Lamprecht M, Bogner S, Schippinger G, et al. Probiotic supplementation affects markers of intestinal barrier, oxidation, and inflammation in trained men; a randomized, double-blinded, placebo-controlled trial. J Int Soc Sports Nutr. 2012;9:45. doi: 10.1186/1550-2783-9-45.
    1. Jeukendrup AE, Vet-Joop K, Sturk A, et al. Relationship between gastro-intestinal complaints and endotoxaemia, cytokine release and the acute-phase reaction during and after a long-distance triathlon in highly trained men. Clin Sci (Lond) 2000;98:47–55. doi: 10.1042/cs0980047.
    1. Qamar MI, Read AE. Effects of exercise on mesenteric blood flow in man. Gut. 1987;28:583–7. doi: 10.1136/gut.28.5.583.
    1. Lambert GP. Stress-induced gastrointestinal barrier dysfunction and its inflammatory effects. J Anim Sci. 2009;87:101–8. doi: 10.2527/jas.2008-1339.
    1. van Wijck K, Lenaerts K, Grootjans J, et al. Physiology and pathophysiology of splanchnic hypoperfusion and intestinal injury during exercise: Strategies for evaluation and prevention. Am J Physiol Gastrointest Liver Physiol. 2012;303:155–68. doi: 10.1152/ajpgi.00066.2012.
    1. Holland AM, Hyatt HW, Smuder AJ, et al. Influence of endurance exercise training on antioxidant enzymes, tight junction proteins, and inflammatory markers in the rat ileum. BMC Res Notes. 2015;8:514. doi: 10.1186/s13104-015-1500-6.
    1. Brock-Utne JG, Gaffin SL, Wells MT, et al. Endotoxaemia in exhausted runners after a long-distance race. S Afr Med J. 1988;73:533–6.
    1. Starkie RL, Hargreaves M, Rolland J, et al. Heat stress, cytokines, and the immune response to exercise. Brain Behav Immun. 2005;19:404–12. doi: 10.1016/j.bbi.2005.03.005.
    1. Gleeson M. Immune function in sport and exercise. J Appl Physiol (1985) 2007;103:693–9. doi: 10.1152/japplphysiol.00008.2007.
    1. Bosenberg AT, Brock-Utne JG, Gaffin SL, et al. Strenuous exercise causes systemic endotoxemia. J Appl Physiol (1985) 1988;65:106–8.
    1. Neish AS. Mucosal immunity and the microbiome. Ann Am Thorac Soc. 2014;11:28–32. doi: 10.1513/AnnalsATS.201306-161MG.
    1. Lambert JE, Myslicki JP, Bomhof MR, et al. Exercise training modifies gut microbiota in normal and diabetic mice. Appl Physiol Nutr Metab. 2015;40:749–52. doi: 10.1139/apnm-2014-0452.
    1. Xu J, Xu C, Chen X, et al. Regulation of an antioxidant blend on intestinal redox status and major microbiota in early weaned piglets. Nutrition. 2014;30:584–9. doi: 10.1016/j.nut.2013.10.018.
    1. Redondo N, Gheorghe A, Serrano R, et al. Hydragut study: Influence of hydration status on the gut microbiota and their impact on the immune system. The FASEB J. 2015;29:1.
    1. den Besten G, van Eunen K, Groen AK, et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54:2325–40. doi: 10.1194/jlr.R036012.
    1. Cook MD, Martin SA, Williams C, et al. Forced treadmill exercise training exacerbates inflammation and causes mortality while voluntary wheel training is protective in a mouse model of colitis. Brain Behav Immun. 2013;33:46–56. doi: 10.1016/j.bbi.2013.05.005.
    1. Costa KA, Soares AD, Wanner SP, et al. L-arginine supplementation prevents increases in intestinal permeability and bacterial translocation in male swiss mice subjected to physical exercise under environmental heat stress. J Nutr. 2014;144:218–23. doi: 10.3945/jn.113.183186.
    1. Musso G, Gambino R, Cassader M. Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes. Ann Rev Med. 2011;62:361–380. doi: 10.1146/annurev-med-012510-175505.
    1. Puddu A, Sanguineti R, Montecucco F, et al. Evidence for the gut microbiota short-chain fatty acids as key pathophysiological molecules improving diabetes. Mediators Inflamm. 2014;2014:1.
    1. Canani RB, Costanzo MD, Leone L, et al. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. 2011;17:1519–28. doi: 10.3748/wjg.v17.i12.1519.
    1. Matsumoto M, Inoue R, Tsukahara T, et al. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. Biosci Biotechnol Biochem. 2008;72:572–6. doi: 10.1271/bbb.70474.
    1. Gleeson M. Immune function in sport and exercise. J Appl Physiol. 2007;103:693–699. doi: 10.1152/japplphysiol.00008.2007.
    1. Best J, Nijhout HF, Reed M. Serotonin synthesis, release and reuptake in terminals: A mathematical model. Theor Biol Med Model. 2010;7:34. doi: 10.1186/1742-4682-7-34.
    1. Evans JM, Morris LS, Marchesi JR. The gut microbiome: The role of a virtual organ in the endocrinology of the host. J Endocrinol. 2013;218:37–47. doi: 10.1530/JOE-13-0131.
    1. Stasi C, Rosselli M, Bellini M, et al. Altered neuro-endocrine-immune pathways in the irritable bowel syndrome: The top-down and the bottom-up model. J Gastroenterol. 2012;47:1177–85. doi: 10.1007/s00535-012-0627-7.
    1. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol. 2012;10:735–742. doi: 10.1038/nrmicro2876.
    1. El Aidy S, Dinan TG, Cryan JF. Gut microbiota: The conductor in the orchestra of immune-neuroendocrine communication. Clin Ther. 2015;37:954–67. doi: 10.1016/j.clinthera.2015.03.002.
    1. McKinney J, Knappskog PM, Haavik J. Different properties of the central and peripheral forms of human tryptophan hydroxylase. J Neurochem. 2005;92:311–20. doi: 10.1111/j.1471-4159.2004.02850.x.
    1. Walther DJ, Peter JU, Bashammakh S, et al. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science. 2003;299:76. doi: 10.1126/science.1078197.
    1. Xie W, Cai L, Yu Y, et al. Activation of brain indoleamine 2,3-dioxygenase contributes to epilepsy-associated depressive-like behavior in rats with chronic temporal lobe epilepsy. J Neuroinflammation. 2014;11:41. doi: 10.1186/1742-2094-11-41.
    1. Otsuka T, Nishii A, Amemiya S, et al. Effects of acute treadmill running at different intensities on activities of serotonin and corticotropin-releasing factor neurons, and anxiety- and depressive-like behaviors in rats. Beh Brain Res. 2016;298:44–51. doi: 10.1016/j.bbr.2015.10.055.
    1. Cryan JF, Dinan TG. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012;13:701–12. doi: 10.1038/nrn3346.
    1. Yano JM, Yu K, Donaldson GP, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161:264–76. doi: 10.1016/j.cell.2015.02.047.
    1. Otsuka T, Nishii A, Amemiya S, et al. Effects of acute treadmill running at different intensities on activities of serotonin and corticotropin-releasing factor neurons, and anxiety- and depressive-like behaviors in rats. Beh Brain Res. 2016;1:44–51. doi: 10.1016/j.bbr.2015.10.055.
    1. Dey S, Singh RH, Dey PK. Exercise training: Significance of regional alterations in serotonin metabolism of rat brain in relation to antidepressant effect of exercise. Physiol Behav. 1992;52:1095–9. doi: 10.1016/0031-9384(92)90465-E.
    1. Fernstrom JD, Fernstrom MH. Exercise, serum free tryptophan, and central fatigue. J Nutr. 2006;136:553–59.
    1. Pechlivanis A, Kostidis S, Saraslanidis P, et al. 1 h nmr study on the short- and long-term impact of two training programs of sprint running on the metabolic fingerprint of human serum. J Proteome Res. 2013;12:470–80. doi: 10.1021/pr300846x.
    1. Foley TE, Fleshner M. Neuroplasticity of dopamine circuits after exercise: Implications for central fatigue. Neuromolecular Med. 2008;10:67–80. doi: 10.1007/s12017-008-8032-3.
    1. Bailey SP, Davis JM, Ahlborn EN. Brain serotonergic activity affects endurance performance. Int J Sports Med. 1993;6:330–33. doi: 10.1055/s-2007-1021187.
    1. Sutton EE, Coill MR, Deuster PA. Ingestion of tyrosine: Effects on endurance, muscle strength, and anaerobic performance. Int J Sport Nutr Exerc Metab. 2005;15:173–85.
    1. Dunn AJ. Effects of cytokines and infections on brain neurochemistry. Clin Neurosci Res. 2006;6:52–68. doi: 10.1016/j.cnr.2006.04.002.
    1. Guezennec CY, Abdelmalki A, Serrurier B, et al. Effects of prolonged exercise on brain ammonia and amino acids. Int J Sports Med. 1998;19:323–7.
    1. Desvergne B, Michalik L, Wahli W. Transcriptional regulation of metabolism. Physiol Rev. 2006;86:465–514. doi: 10.1152/physrev.00025.2005.
    1. Curi R, Newsholme P, Procopio J, et al. Glutamine, gene expression, and cell function. Front Biosci. 2007;12:344-57.
    1. Mul JD, Stanford KI, Hirshman MF, et al. Exercise and regulation of carbohydrate metabolism. Prog Mol Biol Transl Sci. 2015;135:17–37. doi: 10.1016/bs.pmbts.2015.07.020.
    1. Gleeson M. Dosing and efficacy of glutamine supplementation in human exercise and sport training. J Nutr. 2008;138:2045–49.
    1. Clarke G, Grenham S, Scully P, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18:666–73. doi: 10.1038/mp.2012.77.
    1. Desbonnet L, Garrett L, Clarke G, et al. The probiotic bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J Psychiatr Res. 2008;43:164–74. doi: 10.1016/j.jpsychires.2008.03.009.
    1. Reigstad CS, Salmonson CE, Rainey JF, et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. Faseb J. 2015;29:1395–1403. doi: 10.1096/fj.14-259598.
    1. Desborough JP. The stress response to trauma and surgery. Br J Anaesth. 2000;85:109–17. doi: 10.1093/bja/85.1.109.
    1. Norheim F, Gjelstad IM, Hjorth M, et al. Molecular nutrition research: The modern way of performing nutritional science. Nutrients. 2012;4:1898–944. doi: 10.3390/nu4121898.
    1. Antonio J, Stout JR, Willoughby D, et al.: Essentials of sports nutrition and supplements. International society of sports nutrition, Human Press 2008
    1. Sonnenburg ED, Smits SA, Tikhonov M, et al. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212–5. doi: 10.1038/nature16504.
    1. Gleeson M, Bishop NC. Special feature for the olympics: Effects of exercise on the immune system: Modification of immune responses to exercise by carbohydrate, glutamine and anti-oxidant supplements. Immunol Cell Biol. 2000;78:554–61. doi: 10.1111/j.1440-1711.2000.t01-6-.x.
    1. Achten J, Halson SL, Moseley L, et al. Higher dietary carbohydrate content during intensified running training results in better maintenance of performance and mood state. J Appl Physiol (1985) 2004;96:1331–40. doi: 10.1152/japplphysiol.00973.2003.
    1. Morgan WP, Costill DL, Flynn MG, et al. Mood disturbance following increased training in swimmers. Med Sci Sports Exerc. 1988;20:408–14. doi: 10.1249/00005768-198808000-00014.
    1. De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from europe and rural africa. Proc Natl Acad Sci U S A. 2010;107:14691–6. doi: 10.1073/pnas.1005963107.
    1. Tipton KD, Witard OC. Protein requirements and recommendations for athletes: Relevance of ivory tower arguments for practical recommendations. Clin Sports Med. 2007;26:17–36. doi: 10.1016/j.csm.2006.11.003.
    1. Rodriguez NR, Vislocky LM, Gaine PC. Dietary protein, endurance exercise, and human skeletal-muscle protein turnover. Curr Opin Clin Nutr Metab Care. 2007;10:40–5. doi: 10.1097/MCO.0b013e3280115e3b.
    1. Helms ER, Zinn C, Rowlands DS, et al. High-protein, low-fat, short-term diet results in less stress and fatigue than moderate-protein, moderate-fat diet during weight loss in male weightlifters: A pilot study. Int J Sport Nutr Exer Metab. 2015;25:163–170. doi: 10.1123/ijsnem.2014-0056.
    1. Krzwkowski K, Wolsk E, Ostrowski K, et al. Effects of glutamine and protein supplementation on exercise-induced decrease in lymphocyte function and salivary iga. J Appl Physiol. 2001;91:832–838.
    1. Kingsbury KJ, Kay L, Hjelm M. Contrasting plasma free amino acid patterns in elite athletes: Association with fatigue and infection. Br J Sports Med. 1998;32:25–32. doi: 10.1136/bjsm.32.1.25.
    1. Bruunsgaard H, Pedersen BK. Special feature for the olympics: Effects of exercise on the immune system: Effects of exercise on the immune system in the elderly population. Immunol Cell Biol. 2000;78:523–31. doi: 10.1111/j.1440-1711.2000.t01-14-.x.
    1. Pasiakos SM, McClung HL, McClung JP, et al. Leucine-enriched essential amino acid supplementation during moderate steady state exercise enhances postexercise muscle protein synthesis. Am J Clin Nutr. 2011;94:809–18. doi: 10.3945/ajcn.111.017061.
    1. Crowe MJ, Weatherson JN, Bowden BF. Effects of dietary leucine supplementation on exercise performance. Eur J Appl Physiol. 2006;97:664–72. doi: 10.1007/s00421-005-0036-1.
    1. Meeusen R. Exercise, nutrition and the brain. Sports Med. 2014;44:47–56. doi: 10.1007/s40279-014-0150-5.
    1. Tumilty L, Davison G, Beckmann M, et al. Oral tyrosine supplementation improves exercise capacity in the heat. Eur J Appl Physiol. 2011;111:2941–50. doi: 10.1007/s00421-011-1921-4.
    1. van Wijck K, Lenaerts K, Grootjans J, et al. Physiology and pathophysiology of splanchnic hypoperfusion and intestinal injury during exercise: Strategies for evaluation and prevention. Am J Physiol Gastroint Liver Physiol. 2012;303:155–68. doi: 10.1152/ajpgi.00066.2012.
    1. Watson P, Enever S, Page A, et al. Tyrosine supplementation does not influence the capacity to perform prolonged exercise in a warm environment. Int J Sport Nutr Exerc Metab. 2012;22:363–373. doi: 10.1123/ijsnem.22.5.363.
    1. Windey K, De Preter V, Verbeke K. Relevance of protein fermentation to gut health. Mol Nutr Food Res. 2012;56:184–96. doi: 10.1002/mnfr.201100542.
    1. Drygas W, Rebowska E, Stepien E, et al. Biochemical and hematological changes following the 120-km open-water marathon swim. J Sports Sci Med. 2014;13:632–7.
    1. Blachier F, Mariotti F, Huneau JF, et al. Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences. Amino Acids. 2007;33:547–62. doi: 10.1007/s00726-006-0477-9.
    1. Smith EA, Macfarlane GT. Dissimilatory amino acid metabolism in human colonic bacteria. Anaerobe. 1997;3:327–337. doi: 10.1006/anae.1997.0121.
    1. Hughes R, Magee EA, Bingham S. Protein degradation in the large intestine: Relevance to colorectal cancer. Curr Issues Intest Microbiol. 2000;1:51–8.
    1. Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–85. doi: 10.1038/nm.3145.
    1. Toden S, Bird AR, Topping DL, et al. Resistant starch prevents colonic DNA damage induced by high dietary cooked red meat or casein in rats. Cancer Biol Ther. 2006;5:267–72. doi: 10.4161/cbt.5.3.2382.
    1. Taciak M, Barszcz M, Tusnio A, et al. Interactive effects of indigestible carbohydrates, protein type, and protein level on biomarkers of large intestine health in rats. PloS One. 2015;10:e0142176. doi: 10.1371/journal.pone.0142176.
    1. Zimmer J, Lange B, Frick JS, et al. A vegan or vegetarian diet substantially alters the human colonic faecal microbiota. Eur J Clin Nutr. 2012;66:53–60. doi: 10.1038/ejcn.2011.141.
    1. Ferrocino I, Di Cagno R, De Angelis M, et al. Fecal microbiota in healthy subjects following omnivore, vegetarian and vegan diets: Culturable populations and rrna dgge profiling. PloS One. 2015;10:e0128669. doi: 10.1371/journal.pone.0128669.
    1. Williams C, Chryssanthopoulos C. Pre-exercise food intake and performance. World Rev Nutr Diet. 1997;82:33–45. doi: 10.1159/000059648.
    1. Bjorntorp P. Importance of fat as a support nutrient for energy: Metabolism of athletes. J Sports Sci. 1991;9:71–76. doi: 10.1080/02640419108729867.
    1. Pendergast DR, Leddy JJ, Venkatraman JT. A perspective on fat intake in athletes. J Am College Nutr. 2000;19:345–350. doi: 10.1080/07315724.2000.10718930.
    1. de Haan JJ, Lubbers T, Hadfoune M, et al. Postshock intervention with high-lipid enteral nutrition reduces inflammation and tissue damage. Ann Surg. 2008;248:842–8. doi: 10.1097/SLA.0b013e318188752c.
    1. Bruce-Keller AJ, Salbaum JM, Luo M, et al. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol Psychiatry. 2015;77:607–15. doi: 10.1016/j.biopsych.2014.07.012.
    1. Pedersen BK, Helge JW, Richter EA, et al. Training and natural immunity: Effects of diets rich in fat or carbohydrate. Eur J Appl Physiol. 2000;82:98–102. doi: 10.1007/s004210050657.
    1. Pedersen L, Idorn M, Olofsson GH, et al. Voluntary running suppresses tumor growth through epinephrine- and il-6-dependent nk cell mobilization and redistribution. Cell Metab. 2016;23:554–62. doi: 10.1016/j.cmet.2016.01.011.
    1. Konig D, Berg A, Weinstock C, et al. Essential fatty acids, immune function, and exercise. Exerc Immunol Rev. 1997;3:1–31.
    1. Mickleborough TD. Omega-3 polyunsaturated fatty acids in physical performance optimization. Int J Sport Nutr Exerc Metab. 2013;23:83–96. doi: 10.1123/ijsnem.23.1.83.
    1. Ormsbee MJ, Bach CW, Baur DA. Pre-exercise nutrition: The role of macronutrients, modified starches and supplements on metabolism and endurance performance. Nutrients. 2014;6:1782–808. doi: 10.3390/nu6051782.
    1. de La Serre CB, Ellis CL, Lee J, et al. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am J Physiol Gastrointest Liver Physiol. 2010;299:440–48. doi: 10.1152/ajpgi.00098.2010.
    1. Zhang M, Izumi I, Kagamimori S, et al. Role of taurine supplementation to prevent exercise-induced oxidative stress in healthy young men. Amino Acids. 2004;26:203–7. doi: 10.1007/s00726-003-0002-3.
    1. Nieman DC, Gillitt ND, Knab AM, et al. Influence of a polyphenol-enriched protein powder on exercise-induced inflammation and oxidative stress in athletes: A randomized trial using a metabolomics approach. PloS One. 2013;8:e72215. doi: 10.1371/journal.pone.0072215.
    1. Dahl WJ, Stewart ML. Position of the academy of nutrition and dietetics: Health implications of dietary fiber. J Acad Nutr Diet. 2015;115:1861–70. doi: 10.1016/j.jand.2015.09.003.
    1. Hold GL. The gut microbiota, dietary extremes and exercise. Gut. 2014;63:1838–9. doi: 10.1136/gutjnl-2014-307305.
    1. Del Chierico F, Vernocchi P, Dallapiccola B, et al. Mediterranean diet and health: Food effects on gut microbiota and disease control. Intern J Mol Scien. 2014;15:11678–99. doi: 10.3390/ijms150711678.
    1. Kimura I, Inoue D, Maeda T, et al. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via g protein-coupled receptor 41 (gpr41) Proc Natl Acad Sci USA. 2011;108:8030–5. doi: 10.1073/pnas.1016088108.
    1. Grider JR, Piland BE. The peristaltic reflex induced by short-chain fatty acids is mediated by sequential release of 5-ht and neuronal cgrp but not bdnf. Am J Physiol Gastrointest Liver Physiol. 2007;292:429–37. doi: 10.1152/ajpgi.00376.2006.
    1. Drenowatz C, Eisenmann JC, Carlson JJ, et al. Energy expenditure and dietary intake during high-volume and low-volume training periods among male endurance athletes. Appl Physiol Nutr Metab. 2012;37:199–205. doi: 10.1139/h11-155.
    1. Caccialanza R, Cameletti B, Cavallaro G. Nutritional intake of young italian high-level soccer players: Under-reporting is the essential outcome. J Sports Sci Med. 2007;6:538–42.
    1. Aerenhouts D, Hebbelinck M, Poortmans JR, et al. Nutritional habits of flemish adolescent sprint athletes. Int J Sport Nutr Exerc Metab. 2008;18:509–23. doi: 10.1123/ijsnem.18.5.509.
    1. de Oliveira EP, Burini R, Jeukendrup A. Gastrointestinal complaints during exercise: Prevalence, etiology, and nutritional recommendations. Sports Med. 2014;44:79–85. doi: 10.1007/s40279-014-0153-2.
    1. Mackos AR, Eubank TD, Parry NM, et al. Probiotic lactobacillus reuteri attenuates the stressor-enhanced severity of citrobacter rodentium infection. Infect Immun. 2013;81:3253–63. doi: 10.1128/IAI.00278-13.
    1. Selhub EM, Logan AC, Bested AC. Fermented foods, microbiota, and mental health: Ancient practice meets nutritional psychiatry. J Physiol Anthropol. 2014;33:2. doi: 10.1186/1880-6805-33-2.
    1. Jacouton E, Mach N, Cadiou J, et al. Lactobacillus rhamnosus cncmi-4317 modulates fiaf/angptl4 in intestinal epithelial cells and circulating level in mice. PloS One. 2015;10:e0138880. doi: 10.1371/journal.pone.0138880.
    1. Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, et al. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition. 2016;32:315–20. doi: 10.1016/j.nut.2015.09.003.
    1. Messaoudi M, Violle N, Bisson JF, et al. Beneficial psychological effects of a probiotic formulation (lactobacillus helveticus r0052 and bifidobacterium longum r0175) in healthy human volunteers. Gut Microbes. 2011;2:256–61. doi: 10.4161/gmic.2.4.16108.
    1. Schmidt C. Mental health: Thinking from the gut. Nature. 2015;518:S12–5. doi: 10.1038/518S13a.
    1. Ait-Belgnaoui A, Durand H, Cartier C, et al. Prevention of gut leakiness by a probiotic treatment leads to attenuated hpa response to an acute psychological stress in rats. Psycho Neuro Endocrinol. 2012;37:1885–95. doi: 10.1016/j.psyneuen.2012.03.024.
    1. Liang S, Wang T, Hu X, et al. Administration of lactobacillus helveticus ns8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience. 2015;310:561–77. doi: 10.1016/j.neuroscience.2015.09.033.
    1. Ait-Belgnaoui A, Colom A, Braniste V, et al. Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neuro Gastroenterol Motil. 2014;26:510–20. doi: 10.1111/nmo.12295.
    1. Brooks K, Carter J. Overtraining, exercise, and adrenal insufficiency. J Nov Physiother. 2013;3:1. doi: 10.4172/2165-7025.1000126.
    1. Luger A, Deuster PA, Kyle SB, et al. Acute hypothalamic-pituitary-adrenal responses to the stress of treadmill exercise. Physiologic adaptations to physical training. N Engl J Med. 1987;316:1309–15. doi: 10.1056/NEJM198705213162105.

Source: PubMed

3
Iratkozz fel