Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals

Amar Sarkar, Soili M Lehto, Siobhán Harty, Timothy G Dinan, John F Cryan, Philip W J Burnet, Amar Sarkar, Soili M Lehto, Siobhán Harty, Timothy G Dinan, John F Cryan, Philip W J Burnet

Abstract

Psychobiotics were previously defined as live bacteria (probiotics) which, when ingested, confer mental health benefits through interactions with commensal gut bacteria. We expand this definition to encompass prebiotics, which enhance the growth of beneficial gut bacteria. We review probiotic and prebiotic effects on emotional, cognitive, systemic, and neural variables relevant to health and disease. We discuss gut-brain signalling mechanisms enabling psychobiotic effects, such as metabolite production. Overall, knowledge of how the microbiome responds to exogenous influence remains limited. We tabulate several important research questions and issues, exploration of which will generate both mechanistic insights and facilitate future psychobiotic development. We suggest the definition of psychobiotics be expanded beyond probiotics and prebiotics to include other means of influencing the microbiome.

Keywords: gut–brain axis; interkingdom signalling; microbiome; microbiota; prebiotics; probiotics.

Copyright © 2016 The Authors. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Systems-Level Overview of Psychobiotic Action. Blue arrows indicate psychobiotic processes and effects, while red arrows indicate processes associated with leaky gut and inflammation. Probiotics directly introduce beneficial bacteria such as Lactobacilli and Bifidobacteria into the gut. Prebiotics (e.g., galacto-oligosaccharides) support the growth of such bacteria. SCFAs and gut hormones: Both probiotics and prebiotics increase production of short-chain fatty acids (SCFAs), which interact with gut mucosal enteroendocrine cells and catalyse the release of gut hormones such as cholecystokinin (CCK), peptide tyrosine tyrosine (PYY) and glucagon-like peptide- 1 (GLP-1). Prebiotics may have stronger effects in this regard in comparison to probiotics. SCFAs and gut hormones enter circulation and can migrate into the central nervous system. Gut hormones are also secreted by tissues other than enteroendocrine cells. Neurotransmitters: psychobiotics enhance neurotransmitter production in the gut, including dopamine (DA), serotonin (5-HT), noradrenaline (NA), and γ-aminobutyric acid (GABA), which likely modulate neurotransmission in the proximal synapses of the enteric nervous system. Vagal connections: the vagus nerve synapses on enteric neurons and enables gut–brain communication. Stress, barrier function, and cytokines: barrier dysfunction is exacerbated through stress-induced glucocorticoid exposure. This enables migration of bacteria with pro-inflammatory components, increasing inflammation directly and also triggering a rise in pro-inflammatory cytokines via the immunogenic response. These cytokines impair the integrity of the blood–brain barrier and permit access to potentially pathogenic or inflammatory elements. Pro-inflammatory cytokines (red circles) also reduce the integrity of the gut barrier. Psychobiotic action restores gut barrier function and decreases circulating concentrations of glucocorticoids and pro-inflammatory cytokines. They also increase concentrations of anti-inflammatory cytokines (blue circles), which enhance integrity of the blood–brain barrier, the gut barrier, and reduce overall inflammation. Cytokines clustering at the brain represent cytokine interaction with the blood–brain barrier. Central lymphatic vessels: cytokines may interact more directly with the brain than previously appreciated through the recently discovered central lymphatic vessels.

References

    1. Sudo N. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J. Physiol. 2004;558:263–275.
    1. Hooper L.V. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273.
    1. Kau A.L. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474:327–336.
    1. Le Chatelier E. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–546.
    1. Turnbaugh P.J. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031.
    1. Bravo J.A. Communication between gastrointestinal bacteria and the nervous system. Curr. Opin. Pharmacol. 2012;12:667–672.
    1. Foster J.A., Neufeld K.A.M. Gut–brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013;36:305–312.
    1. Dinan T.G. Psychobiotics: a novel class of psychotropic. Biol. Psychiatry. 2013;74:720–726.
    1. Mayer E.A. Gut microbes and the brain: paradigm shift in neuroscience. J. Neurosci. 2014;34:15490–15496.
    1. Burnet P.W., Cowen P.J. Psychobiotics highlight the pathways to happiness. Biol. Psychiatry. 2013;74:708–709.
    1. Sansonetti P.J., Medzhitov R. Learning tolerance while fighting ignorance. Cell. 2009;138:416–420.
    1. Gibson G.R. Dietary prebiotics: current status and new definition. Food Sci. Technol. Bull. Funct. Foods. 2010;7:1–19.
    1. Dowlati Y. A meta-analysis of cytokines in major depression. Biol. Psychiatry. 2010;67:446–457.
    1. Udina M. Interferon-induced depression in chronic hepatitis C: a systematic review and meta-analysis. J. Clin. Psychiatry. 2012;73:1128–1138.
    1. McNutt M.D. Neurobehavioral effects of interferon-α in patients with hepatitis C: symptom dimensions and responsiveness to paroxetine. Neuropsychopharmacology. 2012;37:1444–1454.
    1. Lu Y. BDNF: a key regulator for protein synthesis-dependent LTP and long-term memory? Neurobiol. Learn. Mem. 2008;89:312–323.
    1. Heldt S.A. Hippocampus-specific deletion of BDNF in adult mice impairs spatial memory and extinction of aversive memories. Mol. Psychiatry. 2007;12:656–670.
    1. Martinowich K., Lu B. Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology. 2008;33:73–83.
    1. Desbonnet L. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience. 2010;170:1179–1188.
    1. Gareau M.G. Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut. 2007;56:1522–1528.
    1. Bravo J.A. 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;108:16050–16055.
    1. Matthews D.M., Jenks S.M. Ingestion of Mycobacterium vaccae decreases anxiety-related behavior and improves learning in mice. Behav. Processes. 2013;96:27–35.
    1. Desbonnet L. The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J. Psychiatr. Res. 2008;43:164–174.
    1. Liang S. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience. 2015;310:561–577.
    1. Janik R. Magnetic resonance spectroscopy reveals oral Lactobacillus promotion of increases in brain GABA, N-acetyl aspartate and glutamate. NeuroImage. 2016;125:988–995.
    1. Alander M. Persistence of colonization of human colonic mucosa by a probiotic strain, Lactobacillus rhamnosus GG, after oral consumption. Appl. Environ. Microbiol. 1999;65:351–354.
    1. Savignac H.M. Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-d-aspartate receptor subunits and d-serine. Neurochem. Int. 2013;63:756–764.
    1. Li F., Tsien J.Z. Memory and the NMDA receptors. N. Engl. J. Med. 2009;361:302–303.
    1. Vázquez E. Effects of a human milk oligosaccharide, 2′-fucosyllactose, on hippocampal long-term potentiation and learning capabilities in rodents. J. Nutr. Biochem. 2015;26:455–465.
    1. Williams S. Neonatal prebiotic (BGOS) supplementation increases the levels of synaptophysin, GluN2A-subunits and BDNF proteins in the adult rat hippocampus. Synapse. 2016;70:121–124.
    1. Oliveros E. Oral supplementation of 2′-fucosyllactose during lactation improves memory and learning in rats. J. Nutr. Biochem. 2016;31:20–27.
    1. Benton D. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007;61:355–361.
    1. Messaoudi M. 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.
    1. Messaoudi M. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes. 2011;2:256–261.
    1. Steenbergen L. A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav. Immun. 2015;48:258–264.
    1. Kato-Kataoka A. Fermented milk containing Lactobacillus casei strain Shirota preserves the diversity of the gut microbiota and relieves abdominal dysfunction in healthy medical students exposed to academic stress. Appl. Environ. Microbiol. 2016;82:3649–3658.
    1. Sashihara T. Effects of Lactobacillus gasseri OLL2809 and α-lactalbumin on university-student athletes: a randomized, double-blind, placebo-controlled clinical trial. Appl. Physiol. Nutr. Metab. 2013;38:1228–1235.
    1. O’Mahony L. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541–551.
    1. Mayer E.A., Tillisch K. The brain–gut axis in abdominal pain syndromes. Annu. Rev. Med. 2011;62:381–396.
    1. Kassinen A. The fecal microbiota of irritable bowel syndrome patients differs significantly from that of healthy subjects. Gastroenterology. 2007;133:24–33.
    1. Clarke G. Irritable bowel syndrome: towards biomarker identification. Trends Mol. Med. 2009;15:478–489.
    1. Tillisch K. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology. 2013;144:1394–1401.
    1. Britton J.C. Facial expressions and complex IAPS pictures: common and differential networks. Neuroimage. 2006;31:906–919.
    1. Schmidt K. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology (Berl) 2015;232:1793–1801.
    1. Bhagwagar Z. Increased salivary cortisol after waking in depression. Psychopharmacology (Berl) 2005;182:54–57.
    1. Mannie Z.N. Increased waking salivary cortisol levels in young people at familial risk of depression. Am. J. Psychiatry. 2007;164:617–621.
    1. Beck A.T. Penguin; 1979. Cognitive Therapy and the Emotional Disorders.
    1. Ironside M. Frontal cortex stimulation reduces vigilance to threat: implications for the treatment of depression and anxiety. Biol. Psychiatry. 2016;79:823–830.
    1. Bercik P. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterol. Motil. 2011;23:1132–1139.
    1. Kunze W.A. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. J. Cell. Mol. Med. 2009;13:2261–2270.
    1. Ma X. Lactobacillus reuteri ingestion prevents hyperexcitability of colonic DRG neurons induced by noxious stimuli. Am. J. Physiol. Gastrointest. Liver Physiol. 2009;296:G868–G875.
    1. 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.
    1. McVey Neufeld The microbiome is essential for normal gut intrinsic primary afferent neuron excitability in the mouse. Neurogastroenterol. Motil. 2013;25:183–188.
    1. Collins J. Intestinal microbiota influence the early postnatal development of the enteric nervous system. Neurogastroenterol. Motil. 2014;26:98–107.
    1. Lomasney K.W. Selective influence of host microbiota on cAMP-mediated ion transport in mouse colon. Neurogastroenterol. Motil. 2014;26:887–888.
    1. Kabouridis P.S. Microbiota controls the homeostasis of glial cells in the gut lamina propria. Neuron. 2015;85:289–295.
    1. Barrett E. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012;113:411–417.
    1. Dinan T.G. Collective unconscious: how gut microbes shape human behavior. J. Psychiatr. Res. 2015;63:1–9.
    1. Lyte M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: microbial endocrinology in the design and use of probiotics. Bioessays. 2011;33:574–581.
    1. Thayer J.F., Sternberg E.M. Neural concomitants of immunity: focus on the vagus nerve. Neuroimage. 2009;47:908–910.
    1. de Haan J.J. Lipid-rich enteral nutrition regulates mucosal mast cell activation via the vagal anti-inflammatory reflex. Am. J. Physiol. Gastrointest. Liver Physiol. 2013;305:G383–G391.
    1. Mezzacappa E.S. Vagal rebound and recovery from psychological stress. Psychosom. Med. 2001;63:650–657.
    1. Spalding T.W. Vagal and cardiac reactivity to psychological stressors in trained and untrained men. Med. Sci. Sports Exerc. 2000;32:581–591.
    1. Borovikova L.V. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–462.
    1. Kirchner A. Left vagus nerve stimulation suppresses experimentally induced pain. Neurology. 2000;55:1167–1171.
    1. Morris G.L., Mueller W.M. Long-term treatment with vagus nerve stimulation in patients with refractory epilepsy. Neurology. 1999;53:1731–1735.
    1. Sackeim H.A. Vagus nerve stimulation (VNS™) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25:713–728.
    1. Rush A.J. Vagus nerve stimulation for treatment-resistant depression: a randomized, controlled acute phase trial. Biol. Psychiatry. 2005;58:347–354.
    1. Adinoff B. Vagal tone decreases following intravenous diazepam. Psychiatry Res. 1992;41:89–97.
    1. Salam O.M.A. Fluoxetine and sertraline stimulate gastric acid secretion via a vagal pathway in anaesthetised rats. Pharmacol. Res. 2004;50:309–316.
    1. Smith R. Increased association over time between regional frontal lobe BOLD change magnitude and cardiac vagal control with sertraline treatment for major depression. Psychiatry Res. Neuroimag. 2014;224:225–233.
    1. De Lartigue G. Vagal afferent neurons in high fat diet-induced obesity; intestinal microflora, gut inflammation and cholecystokinin. Physiol. Behav. 2011;105:100–105.
    1. Bercik P. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141:599–609.
    1. Qin J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65.
    1. Horn T., Klein J. Neuroprotective effects of lactate in brain ischemia: dependence on anesthetic drugs. Neurochem. Int. 2013;62:251–257.
    1. Moretti M. Behavioral and neurochemical effects of sodium butyrate in an animal model of mania. Behav. Pharmacol. 2011;22:766–772.
    1. Overduin J. Dietary galacto-oligosaccharides and calcium: effects on energy intake, fat-pad weight and satiety-related, gastrointestinal hormones in rats. Br. J. Nutr. 2013;109:1338–1348.
    1. Sun J. Antidepressant-like effects of sodium butyrate and its possible mechanisms of action in mice exposed to chronic unpredictable mild stress. Neurosci. Lett. 2016;618:159–166.
    1. Stilling R.M. Microbial genes, brain & behaviour–epigenetic regulation of the gut–brain axis. Genes Brain Behav. 2014;13:69–86.
    1. Erny D. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015;18:965–977.
    1. Gagliano H. High doses of the histone deacetylase inhibitor sodium butyrate trigger a stress-like response. Neuropharmacology. 2014;79:75–82.
    1. Alenghat T., Artis D. Epigenomic regulation of host–microbiota interactions. Trends Immunol. 2014;35:518–525.
    1. Perry R.J. Acetate mediates a microbiome–brain–β-cell axis to promote metabolic syndrome. Nature. 2016;534:213–217.
    1. Nøhr M.K. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology. 2013;154:3552–3564.
    1. Psichas A. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015;39:424–429.
    1. Cani P.D. Oligofructose promotes satiety in rats fed a high-fat diet: involvement of glucagon-like peptide-1. Obes. Res. 2005;13:1000–1007.
    1. Dockray G.J. Gastrointestinal hormones and the dialogue between gut and brain. J. Physiol. 2014;592:2927–2941.
    1. Anderberg R.H. Dopamine signaling in the amygdala, increased by food ingestion and GLP-1, regulates feeding behavior. Physiol. Behav. 2014;136:135–144.
    1. Stadlbauer U. PYY 3-36: Beyond food intake. Front. Neuroendocrinol. 2015;38:1–11.
    1. Morris G. The role of the microbial metabolites including tryptophan catabolites and short chain fatty acids in the pathophysiology of immune-inflammatory and neuroimmune disease. Mol. Neurobiol. 2016 Published online June 27, 2016.
    1. Wikoff W.R. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U.S.A. 2009;106:3698–3703.
    1. Yano J.M. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161:264–276.
    1. Mackey D., McFall A.J. MAMPs and MIMPs: proposed classifications for inducers of innate immunity. Mol. Microbiol. 2006;61:1365–1371.
    1. Chu H., Mazmanian S.K. Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat. Immunol. 2013;14:668–675.
    1. Zhou W. Protective effects of bifidobacteria on intestines in newborn rats with necrotizing enterocolitis and its regulation on TLR2 and TLR4. Genet Mol. Res. 2015;14:11505–11514.
    1. Bode L. Inhibition of monocyte, lymphocyte, and neutrophil adhesion to endothelial cells by human milk oligosaccharides. Thromb. Haemost. 2004;92:1402–1410.
    1. Eiwegger T. Prebiotic oligosaccharides: in vitro evidence for gastrointestinal epithelial transfer and immunomodulatory properties. Pediatr. Allergy Immunol. 2010;21:1179–1188.
    1. McCusker R.H., Kelley K.W. Immune–neural connections: How the immune system's response to infectious agents influences behavior. J. Exp. Biol. 2013;216:84–98.
    1. Miller A.H. Cytokine targets in the brain: impact on neurotransmitters and neurocircuits. Depress. Anxiety. 2013;30:297–306.
    1. Felger J.C., Lotrich F.E. Inflammatory cytokines in depression: neurobiological mechanisms and therapeutic implications. Neuroscience. 2013;246:199–229.
    1. Louveau A. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–341.
    1. Pessi T. Interleukin-10 generation in atopic children following oral Lactobacillus rhamnosus GG. Clin. Exp. Allergy. 2000;30:1804–1808.
    1. Banks W.A., Erickson M.A. The blood–brain barrier and immune function and dysfunction. Neurobiol. Dis. 2010;37:26–32.
    1. de Vries H.E. The influence of cytokines on the integrity of the blood–brain barrier in vitro. J. Neuroimmunol. 1996;64:37–43.
    1. Ek M. Inflammatory response: pathway across the blood–brain barrier. Nature. 2001;410:430–431.
    1. Bercik P. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology. 2010;139:2102–2112.
    1. Wang F. Interferon-γ and tumor necrosis factor-α synergize to induce intestinal epithelial barrier dysfunction by up-regulating myosin light chain kinase expression. Am. J. Pathol. 2005;166:409–419.
    1. Wang F. IFN-γ-induced TNFR2 expression is required for TNF-dependent intestinal epithelial barrier dysfunction. Gastroenterology. 2006;131:1153–1163.
    1. Donato K.A. Lactobacillus rhamnosus GG attenuates interferon-γ and tumour necrosis factor-α-induced barrier dysfunction and pro-inflammatory signalling. Microbiology. 2010;156:3288–3297.
    1. O’Hara A.M. Functional modulation of human intestinal epithelial cell responses by Bifidobacterium infantis and Lactobacillus salivarius. Immunology. 2006;118:202–215.
    1. Redhu N. O-005 YI microbiota drives inflammation by altering intestinal lamina propria macrophage phenotype in a novel IL10R-deficient model of very early onset IBD. Inflamm. Bowe. Dis. 2016:22.
    1. Ray A. Gut microbial dysbiosis due to helicobacter drives an increase in marginal zone B cells in the absence of IL-10 signaling in macrophages. J. Immunol. 2015;195:3071–3085.
    1. Kim K.S. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science. 2016;351:858–863.
    1. Bharwani A. Structural & functional consequences of chronic psychosocial stress on the microbiome & host. Psychoneuroendocrinology. 2016;63:217–227.
    1. Söderholm J.D., Perdue M.H. II. Stress and intestinal barrier function. Am. J. Physiol. Gastrointest. Liver Physiol. 2001;280:G7–G13.
    1. Santos J. Role of mast cells in chronic stress induced colonic epithelial barrier dysfunction in the rat. Gut. 2001;48:630–636.
    1. Mass M. The gut–brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol. Lett. 2008;29:117–124.
    1. Maes 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.
    1. Ait-Belgnaoui A. Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology. 2012;37:1885–1895.
    1. Zareie M. Probiotics prevent bacterial translocation and improve intestinal barrier function in rats following chronic psychological stress. Gut. 2006;55:1553–1560.
    1. Romijn A.R., Rucklidge J.J. Systematic review of evidence to support the theory of psychobiotics. Nutr. Rev. 2015;73:675–693.
    1. Forsythe P. Moody microbes or fecal phrenology: what do we know about the microbiota–gut–brain axis? BMC Med. 2016;14:58.
    1. Davey K.J. Olanzapine induced weight gain in the rat: Impact on inflammatory, metabolic and microbiota parameters. Psychopharmacology (Berl) 2013;221:155–169.
    1. Davey K.J. Antipsychotics and the gut microbiome: olanzapine-induced metabolic dysfunction is attenuated by antibiotic administration in the rat. Transl. Psychiatry. 2013;3:e309.
    1. Cho I. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012;488:621–626.
    1. El Rakaiby M. Pharmacomicrobiomics: the impact of human microbiome variations on systems pharmacology and personalized therapeutics. OMICS. 2014;18:402–414.
    1. David L.A. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563.
    1. Hildebrandt M.A. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology. 2009;137:1716–1724.
    1. Kang S.S. Diet and exercise orthogonally alter the gut microbiome and reveal independent associations with anxiety and cognition. Mol. Neurodegener. 2014;9:36.
    1. Goldsmith R.L. Physical fitness as a determinant of vagal modulation. Med. Sci. Sports Exerc. 1997;29:812–817.
    1. Sugawara J. Change in post-exercise vagal reactivation with exercise training and detraining in young men. Eur. J. Appl. Physiol. 2001;85:259–263.
    1. De Filippo C. 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;107:14691–14696.
    1. Fallani M. Intestinal microbiota of 6-week-old infants across Europe: geographic influence beyond delivery mode, breast-feeding, and antibiotics. J. Pediatr. Gastroenterol. Nutr. 2010;51:77–84.
    1. Yatsunenko T. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–227.
    1. Jia S. Chitosan oligosaccharides alleviate cognitive deficits in an amyloid-β 1-42-induced rat model of Alzheimer disease. Int. J. Biol. Macromol. 2016;83:416–425.
    1. Song L. Galactooligosaccharide improves the animal survival and alleviates motor neuron death in SOD1 G93A mouse model of amyotrophic lateral sclerosis. Neuroscience. 2013;246:281–290.
    1. Hsiao E.Y. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–1463.
    1. Rodrigues D.M. Probiotics are effective for the prevention and treatment of Citrobacter rodentium–induced colitis in mice. J. Infect. Dis. 2012;206:99–109.
    1. Gareau M.G. Probiotics prevent death caused by Citrobacter rodentium infection in neonatal mice. J. Infect. Dis. 2010;201:81–91.
    1. Mackos A.R. Probiotic Lactobacillus reuteri attenuates the stressor-enhanced severity of Citrobacter rodentium infection. Infect. Immun. 2013;81:3253–3263.
    1. Mackos A.R. Social stress-enhanced severity of Citrobacter rodentium-induced colitis is CCL2-dependent and attenuated by probiotic Lactobacillus reuteri. Mucosal. Immunol. 2015;9:515–526.
    1. Davari S. Probiotics treatment improves diabetes-induced impairment of synaptic activity and cognitive function: behavioral and electrophysiological proofs for microbiome–gut–brain axis. Neuroscience. 2013;240:287–296.
    1. Marques T.M. Influence of GABA and GABA-producing Lactobacillus brevis DPC 6108 on the development of diabetes in a streptozotocin rat model. Benef. Microbes. 2016:1–12.
    1. Luo J. Ingestion of Lactobacillus strain reduces anxiety and improves cognitive function in the hyperammonemia rat. Sci. China Life Sci. 2014;57:327–335.
    1. Savignac H.M. Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice. Brain Behav. Immun. 2016;52:120–131.
    1. McNulty N.P. The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Sci. Transl. Med. 2011;3:106ra106.
    1. Kristensen N.B. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials. Genome Med. 2016;8:52.
    1. Claesson M.J. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. U.S.A. 2011;108:4586–4591.
    1. Claesson M.J. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488:178–184.
    1. Distrutti E. Modulation of intestinal microbiota by the probiotic VSL# 3 resets brain gene expression and ameliorates the age-related deficit in LTP. PLoS One. 2014;9:e106503.
    1. Lahtinen S.J. Probiotic cheese containing Lactobacillus rhamnosus HN001 and Lactobacillus acidophilus NCFM® modifies subpopulations of fecal lactobacilli and Clostridium difficile in the elderly. Age. 2012;34:133–143.
    1. Rampelli S. A probiotics-containing biscuit modulates the intestinal microbiota in the elderly. J. Nutr. Health Aging. 2013;17:166–172.
    1. Savignac H.M. Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol. Motil. 2014;26:1615–1627.
    1. Savignac H.M. Bifidobacteria modulate cognitive processes in an anxious mouse strain. Behav. Brain Res. 2015;287:59–72.
    1. Ohland C.L. Effects of Lactobacillus helveticus on murine behavior are dependent on diet and genotype and correlate with alterations in the gut microbiome. Psychoneuroendocrinology. 2013;38:1738–1747.

Source: PubMed

3
Prenumerera