Skip to main content

Advertisement

Log in

Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The gut microbiota is essential to health and has recently become a target for live bacterial cell biotherapies for various chronic diseases including metabolic syndrome, diabetes, obesity and neurodegenerative disease. Probiotic biotherapies are known to create a healthy gut environment by balancing bacterial populations and promoting their favorable metabolic action. The microbiota and its respective metabolites communicate to the host through a series of biochemical and functional links thereby affecting host homeostasis and health. In particular, the gastrointestinal tract communicates with the central nervous system through the gut–brain axis to support neuronal development and maintenance while gut dysbiosis manifests in neurological disease. There are three basic mechanisms that mediate the communication between the gut and the brain: direct neuronal communication, endocrine signaling mediators and the immune system. Together, these systems create a highly integrated molecular communication network that link systemic imbalances with the development of neurodegeneration including insulin regulation, fat metabolism, oxidative markers and immune signaling. Age is a common factor in the development of neurodegenerative disease and probiotics prevent many harmful effects of aging such as decreased neurotransmitter levels, chronic inflammation, oxidative stress and apoptosis—all factors that are proven aggravators of neurodegenerative disease. Indeed patients with Parkinson’s and Alzheimer’s diseases have a high rate of gastrointestinal comorbidities and it has be proposed by some the management of the gut microbiota may prevent or alleviate the symptoms of these chronic diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Burokas A, Moloney RD, Dinan TG, Cryan JF (2015) Microbiota regulation of the mammalian gut–brain axis. Adv Appl Microbiol 91:1–62. doi:10.1016/bs.aambs.2015.02.001

    Article  PubMed  Google Scholar 

  2. Forsythe P, Bienenstock J, Kunze WA (2014) Vagal pathways for microbiome–brain–gut axis communication. Microbial endocrinology: the microbiota–gut–brain axis in health and disease. Springer, New York, pp 115–133

    Google Scholar 

  3. Bauer KC, Huus KE, Finlay BB (2016) Microbes and the mind: emerging hallmarks of the gut microbiota–brain axis. Cell Microbiol 18:632–644. doi:10.1111/cmi.12585

    Article  CAS  PubMed  Google Scholar 

  4. Noble EE, Hsu TM, Kanoski SE (2017) Gut to brain dysbiosis: mechanisms linking western diet consumption, the microbiome, and cognitive impairment. Front Behav Neurosci 11:9. doi:10.3389/fnbeh.2017.00009

    Article  PubMed  PubMed Central  Google Scholar 

  5. Mulak A, Bonaz B (2015) Brain–gut–microbiota axis in Parkinson’s disease. WJG 21:10609–10620. doi:10.3748/wjg.v21.i37.10609

    CAS  PubMed  Google Scholar 

  6. Friedland RP (2015) Mechanisms of molecular mimicry involving the microbiota in neurodegeneration. J Alzheimers Dis 45:349–362. doi:10.3233/JAD-142841

    CAS  PubMed  Google Scholar 

  7. Qin J, Li R, Raes J et al (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464:59–65. doi:10.1038/nature08821

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rajilic-Stojanovic M, Smidt H, de Vos WM (2007) Diversity of the human gastrointestinal tract microbiota revisited. Environ Microbiol 9:2125–2136. doi:10.1111/j.1462-2920.2007.01369.x

    Article  PubMed  Google Scholar 

  9. The Human Microbiome Project Consortium (2012) Structure, function and diversity of the healthy human microbiome. Nature 486:207–214. doi:10.1038/nature11234

    Article  PubMed Central  CAS  Google Scholar 

  10. Mandal RS, Saha S, Das S (2015) Metagenomic surveys of gut microbiota. Genom Proteom Bioinform 13:148–158. doi:10.1016/j.gpb.2015.02.005

    Article  Google Scholar 

  11. Everard A, Cani PD (2014) Gut microbiota and GLP-1. Rev Endocr Metab Disord 15:189–196. doi:10.1007/s11154-014-9288-6

    Article  CAS  PubMed  Google Scholar 

  12. Burns AJ (2005) Migration of neural crest-derived enteric nervous system precursor cells to and within the gastrointestinal tract. Int J Dev Biol 49:143–150. doi:10.1387/ijdb.041935ab

    Article  PubMed  Google Scholar 

  13. O’Mahony SM, Clarke G, Borre YE et al (2014) Serotonin, tryptophan metabolism and the brain–gut–microbiome axis. Behav Brain Res. doi:10.1016/j.bbr.2014.07.027

    PubMed  Google Scholar 

  14. Ochoa-Reparaz J, Mielcarz DW, Begum-Haque S, Kasper LH (2011) Gut, bugs, and brain: role of commensal bacteria in the control of central nervous system disease. Ann Neurol 69:240–247. doi:10.1002/ana.22344

    Article  CAS  PubMed  Google Scholar 

  15. Luczynski P, McVey Neufeld K-A, Oriach CS et al (2016) Growing up in a bubble: using germ-free animals to assess the influence of the gut microbiota on brain and behavior. Int J Neuropsychopharmacol 19(8):234–248. doi:10.1093/ijnp/pyw020

    Article  CAS  Google Scholar 

  16. Houlden A, Goldrick M, Brough D et al (2016) Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production. Brain Behav Immun 57:10–20. doi:10.1016/j.bbi.2016.04.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Catanzaro R, Anzalone MG, Calabrese F et al (2014) The gut microbiota and its correlations with the central nervous system disorders. Panminerva Med 57(3):127–143

    PubMed  Google Scholar 

  18. Dinan TG, Cryan JF (2017) Gut instincts: microbiota as a key regulator of brain development, ageing and neurodegeneration. J Physiol 595:489–503. doi:10.1113/JP273106

    Article  CAS  PubMed  Google Scholar 

  19. Kohler CA, Maes M, Slyepchenko A et al (2016) The gut–brain axis, including the microbiome, leaky gut and bacterial translocation: mechanisms and pathophysiological role in Alzheimer’s disease. Curr Pharm Des 22:6152–6166

    Article  CAS  PubMed  Google Scholar 

  20. Jyothi HJ, Vidyadhara DJ, Mahadevan A et al (2015) Aging causes morphological alterations in astrocytes and microglia in human substantia nigra pars compacta. Neurobiol Aging 36:3321–3333. doi:10.1016/j.neurobiolaging.2015.08.024

    Article  CAS  PubMed  Google Scholar 

  21. Erny D, Hrabe de Angelis AL, Jaitin D et al (2015) Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18:965–977. doi:10.1038/nn.4030

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sampson TR, Debelius JW, Thron T et al (2016) Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell 167(1469–1480):e12. doi:10.1016/j.cell.2016.11.018

    Google Scholar 

  23. Alam MZ, Alam Q, Kamal MA et al (2014) A possible link of gut microbiota alteration in type 2 diabetes and Alzheimer’s disease pathogenicity: an update. CNS Neurol Disord Drug Targets 13:383–390

    Article  CAS  PubMed  Google Scholar 

  24. Naseer MI, Bibi F, Alqahtani MH et al (2014) Role of gut microbiota in obesity, type 2 diabetes and Alzheimer’s disease. CNS Neurol Disord Drug Targets 13:305–311

    Article  CAS  PubMed  Google Scholar 

  25. Bekkering P, Jafri I, van Overveld FJ, Rijkers GT (2013) The intricate association between gut microbiota and development of type 1, type 2 and type 3 diabetes. Expert Rev Clin Immunol 9:1031–1041. doi:10.1586/1744666X.2013.848793

    Article  CAS  PubMed  Google Scholar 

  26. Rao M, Gershon MD (2016) The bowel and beyond: the enteric nervous system in neurological disorders. Nat Rev Gastroenterol Hepatol 13:517–528. doi:10.1038/nrgastro.2016.107

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Baltazar MT, Dinis-Oliveira RJ, de Lourdes Bastos M et al (2014) Pesticides exposure as etiological factors of Parkinson’s disease and other neurodegenerative diseases—a mechanistic approach. Toxicol Lett 230:85–103. doi:10.1016/j.toxlet.2014.01.039

    Article  CAS  PubMed  Google Scholar 

  28. Virmani A, Pinto L, Binienda Z, Ali S (2013) Food, nutrigenomics, and neurodegeneration–neuroprotection by what you eat! Mol Neurobiol 48:353–362. doi:10.1007/s12035-013-8498-3

    Article  CAS  PubMed  Google Scholar 

  29. Ley RE, Hamady M, Lozupone C et al (2008) Evolution of mammals and their gut microbes. Science 320:1647–1651. doi:10.1126/science.1155725

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Goldman JG, Postuma R (2014) Premotor and nonmotor features of Parkinson’s disease. Curr Opin Neurol 27:434–441. doi:10.1097/WCO.0000000000000112

    Article  PubMed  PubMed Central  Google Scholar 

  31. Murphy MP (2009) How mitochondria produce reactive oxygen species. Biochem J 417:1–13. doi:10.1042/BJ20081386

    Article  CAS  PubMed  Google Scholar 

  32. Mitsuma T, Odajima H, Momiyama Z et al (2008) Enhancement of gene expression by a peptide p(CHWPR) produced by Bifidobacterium lactis BB-12. Microbiol Immunol 52:144–155. doi:10.1111/j.1348-0421.2008.00022.x

    Article  CAS  PubMed  Google Scholar 

  33. Wang X, Michaelis EK (2010) Selective neuronal vulnerability to oxidative stress in the brain. Front Aging Neurosci 2:12. doi:10.3389/fnagi.2010.00012

    PubMed  PubMed Central  Google Scholar 

  34. Zhu X, Raina AK, Lee H-G et al (2004) Oxidative stress signalling in Alzheimer’s disease. Brain Res 1000:32–39

    Article  CAS  PubMed  Google Scholar 

  35. Franceschi C, Campisi J (2014) Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci 69(Suppl 1):S4–S9. doi:10.1093/gerona/glu057

    Article  PubMed  Google Scholar 

  36. Biagi E, Nylund L, Candela M et al (2010) Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One 5:e10667. doi:10.1371/journal.pone.0010667

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Steen E, Terry BM, Rivera EJ et al (2005) Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease—is this type 3 diabetes? J Alzheimers Dis 7:63–80

    Article  CAS  PubMed  Google Scholar 

  38. Cheng J, Palva AM, de Vos WM, Satokari R (2013) Contribution of the intestinal microbiota to human health: from birth to 100 years of age. In: Dobrindt U, Hacker JH, Svanborg C (eds) Between pathogenicity and commensalism. Springer, Berlin, pp 323–346

    Google Scholar 

  39. Claesson MJ, Cusack S, O’Sullivan O et al (2011) Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci USA 108(Suppl 1):4586–4591. doi:10.1073/pnas.1000097107

    Article  CAS  PubMed  Google Scholar 

  40. Cassani E, Barichella M, Cancello R et al (2015) Increased urinary indoxyl sulfate (indican): new insights into gut dysbiosis in Parkinson’s disease. Parkinsonism Relat Disord 21:389–393. doi:10.1016/j.parkreldis.2015.02.004

    Article  PubMed  Google Scholar 

  41. Scheperjans F, Aho V, Pereira PAB et al (2014) Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord 30(3):350–358. doi:10.1002/mds.26069

    Article  PubMed  Google Scholar 

  42. Keshavarzian A, Green SJ, Engen PA et al (2015) Colonic bacterial composition in Parkinson’s disease. Mov Disord 30:1351–1360. doi:10.1002/mds.26307

    Article  CAS  PubMed  Google Scholar 

  43. Shannon KM, Keshavarzian A, Dodiya HB et al (2012) Is alpha-synuclein in the colon a biomarker for premotor Parkinson’s disease? Evidence from 3 cases. Mov Disord 27:716–719. doi:10.1002/mds.25020

    Article  PubMed  Google Scholar 

  44. Unger MM, Spiegel J, Dillmann K-U et al (2016) Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat Disord 32:66–72. doi:10.1016/j.parkreldis.2016.08.019

    Article  PubMed  Google Scholar 

  45. Kidd SK, Schneider JS (2010) Protection of dopaminergic cells from MPP+-mediated toxicity by histone deacetylase inhibition. Brain Res 1354:172–178. doi:10.1016/j.brainres.2010.07.041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wu X, Chen PS, Dallas S et al (2008) Histone deacetylase inhibitors up-regulate astrocyte GDNF and BDNF gene transcription and protect dopaminergic neurons. Int J Neuropsychopharmacol 11:1123–1134. doi:10.1017/S1461145708009024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Guseo A (2012) The Parkinson puzzle. Orv Hetil 153:2060–2069. doi:10.1556/OH.2012.29461

    Article  PubMed  Google Scholar 

  48. Smith PD, Smythies LE, Shen R et al (2011) Intestinal macrophages and response to microbial encroachment. Mucosal Immunol 4:31–42. doi:10.1038/mi.2010.66

    Article  PubMed  CAS  Google Scholar 

  49. Faden AI, Loane DJ (2015) Chronic neurodegeneration after traumatic brain injury: Alzheimer disease, chronic traumatic encephalopathy, or persistent neuroinflammation? Neurotherapeutics 12:143–150. doi:10.1007/s13311-014-0319-5

    Article  CAS  PubMed  Google Scholar 

  50. Vivekanantham S, Shah S, Dewji R et al (2015) Neuroinflammation in Parkinson’s disease: role in neurodegeneration and tissue repair. Int J Neurosci 125:717–725. doi:10.3109/00207454.2014.982795

    Article  CAS  PubMed  Google Scholar 

  51. Clarke G, Stilling RM, Kennedy PJ et al (2014) Minireview: gut microbiota: the neglected endocrine organ. Mol Endocrinol 28:1221–1238. doi:10.1210/me.2014-1108

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Mancuso C, Santangelo R (2014) Ferulic acid: pharmacological and toxicological aspects. Food Chem Toxicol 65:185–195. doi:10.1016/j.fct.2013.12.024

    Article  CAS  PubMed  Google Scholar 

  53. Trombino S, Cassano R, Ferrarelli T et al (2013) Trans-ferulic acid-based solid lipid nanoparticles and their antioxidant effect in rat brain microsomes. Colloids Surf B Biointerfaces 109:273–279. doi:10.1016/j.colsurfb.2013.04.005

    Article  CAS  PubMed  Google Scholar 

  54. Hu C-T, Wu J-R, Cheng C-C et al (2011) Reactive oxygen species-mediated PKC and integrin signaling promotes tumor progression of human hepatoma HepG2. Clin Exp Metastasis 28:851–863. doi:10.1007/s10585-011-9416-6

    Article  CAS  PubMed  Google Scholar 

  55. Yabe T, Hirahara H, Harada N et al (2010) Ferulic acid induces neural progenitor cell proliferation in vitro and in vivo. Neuroscience 165:515–524. doi:10.1016/j.neuroscience.2009.10.023

    Article  CAS  PubMed  Google Scholar 

  56. Yu L, Zhang Y, Liao M et al (2011) Neurogenesis-enhancing effect of sodium ferulate and its role in repair following stress-induced neuronal damage. WJNS 1(2):9–18. doi:10.4236/wjns.2011.12002

    Article  CAS  Google Scholar 

  57. Tomaro-Duchesneau C, Saha S, Malhotra M et al (2012) Probiotic ferulic acid esterase active Lactobacillus fermentum NCIMB 5221 APA microcapsules for oral delivery: preparation and in vitro characterization. Pharmaceuticals (Basel) 5:236–248. doi:10.3390/ph5020236

    Article  CAS  Google Scholar 

  58. Szwajgier D, Dmowska K (2010) Novel ferulic acid esterases from Bifidobacterium sp. produced on selected synthetic and natural carbon sources. Acta Sci Pol Technol Aliment 9:305–318

    CAS  Google Scholar 

  59. Bhathena J, Martoni C, Kulamarva A et al (2009) Orally delivered microencapsulated live probiotic formulation lowers serum lipids in hypercholesterolemic hamsters. J Med Food 12:310–319. doi:10.1089/jmf.2008.0166

    Article  CAS  PubMed  Google Scholar 

  60. Durairajan SSK, Yuan Q, Xie L et al (2008) Salvianolic acid B inhibits Abeta fibril formation and disaggregates preformed fibrils and protects against Abeta-induced cytotoxicty. Neurochem Int 52:741–750. doi:10.1016/j.neuint.2007.09.006

    Article  CAS  PubMed  Google Scholar 

  61. Mori T, Koyama N, Guillot-Sestier M-V et al (2013) Ferulic acid is a nutraceutical β-secretase modulator that improves behavioral impairment and Alzheimer-like pathology in transgenic mice. PLoS One 8:e55774. doi:10.1371/journal.pone.0055774

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yan J-J, Jung J-S, Kim T-K et al (2013) Protective effects of ferulic acid in amyloid precursor protein plus presenilin-1 transgenic mouse model of Alzheimer disease. Biol Pharm Bull 36:140–143

    Article  CAS  PubMed  Google Scholar 

  63. Zhang L, Wang H, Wang T et al (2015) Ferulic acid ameliorates nerve injury induced by cerebral ischemia in rats. Exp Ther Med 9:972–976

    Article  CAS  PubMed  Google Scholar 

  64. Srinivasan M, Sudheer AR, Menon VP (2007) Ferulic acid: therapeutic potential through its antioxidant property. J Clin Biochem Nutr 40:92–100. doi:10.3164/jcbn.40.92

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Cheng C-Y, Tang N-Y, Kao S-T, Hsieh C-L (2016) Ferulic acid administered at various time points protects against cerebral infarction by activating p38 MAPK/p90RSK/CREB/Bcl-2 anti-apoptotic signaling in the subacute phase of cerebral ischemia–reperfusion injury in rats. PLoS One 11:e0155748

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Sung J-H, Gim S-A, Koh P-O (2014) Ferulic acid attenuates the cerebral ischemic injury-induced decrease in peroxiredoxin-2 and thioredoxin expression. Neurosci Lett 566:88–92. doi:10.1016/j.neulet.2014.02.040

    Article  CAS  PubMed  Google Scholar 

  67. Patenaude A, Murthy MRV, Mirault M-E (2005) Emerging roles of thioredoxin cycle enzymes in the central nervous system. Cell Mol Life Sci 62:1063–1080. doi:10.1007/s00018-005-4541-5

    Article  CAS  PubMed  Google Scholar 

  68. Gan Y, Ji X, Hu X et al (2012) Transgenic overexpression of peroxiredoxin-2 attenuates ischemic neuronal injury via suppression of a redox-sensitive pro-death signaling pathway. Antioxid Redox Signal 17:719–732. doi:10.1089/ars.2011.4298

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Saitoh M, Nishitoh H, Fujii M et al (1998) Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. EMBO J 17:2596–2606. doi:10.1093/emboj/17.9.2596

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ma YH, Su N, Chao XD et al (2012) Thioredoxin-1 attenuates post-ischemic neuronal apoptosis via reducing oxidative/nitrative stress. Neurochem Int 60:475–483. doi:10.1016/j.neuint.2012.01.029

    Article  CAS  PubMed  Google Scholar 

  71. Gim S-A, Sung J-H, Shah F-A et al (2013) Ferulic acid regulates the AKT/GSK-3β/CRMP-2 signaling pathway in a middle cerebral artery occlusion animal model. Lab Anim Res 29:63. doi:10.5625/lar.2013.29.2.63

    Article  PubMed  PubMed Central  Google Scholar 

  72. Gao C, Holscher C, Liu Y, Li L (2012) GSK3: a key target for the development of novel treatments for type 2 diabetes mellitus and Alzheimer disease. Rev Neurosci 23:1–11

    Article  CAS  Google Scholar 

  73. Yoshimura T, Kawano Y, Arimura N et al (2005) GSK-3beta regulates phosphorylation of CRMP-2 and neuronal polarity. Cell 120:137–149. doi:10.1016/j.cell.2004.11.012

    Article  CAS  PubMed  Google Scholar 

  74. Gu Y, Hamajima N, Ihara Y (2000) Neurofibrillary tangle-associated collapsin response mediator protein-2 (CRMP-2) is highly phosphorylated on Thr-509, Ser-518, and Ser-522. Biochemistry 39:4267–4275

    Article  CAS  PubMed  Google Scholar 

  75. Petratos S, Li Q-X, George AJ et al (2008) The beta-amyloid protein of Alzheimer’s disease increases neuronal CRMP-2 phosphorylation by a Rho-GTP mechanism. Brain 131:90–108. doi:10.1093/brain/awm260

    Article  PubMed  Google Scholar 

  76. Soutar MPM, Thornhill P, Cole AR, Sutherland C (2009) Increased CRMP2 phosphorylation is observed in Alzheimer’s disease; does this tell us anything about disease development? Curr Alzheimer Res 6:269–278

    Article  CAS  PubMed  Google Scholar 

  77. Verbeke KA, Boobis AR, Chiodini A et al (2015) Towards microbial fermentation metabolites as markers for health benefits of prebiotics. Nutr Res Rev 28:42–66. doi:10.1017/S0954422415000037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Tagliabue A, Elli M (2013) The role of gut microbiota in human obesity: recent findings and future perspectives. Nutr Metab Cardiovasc Dis 23:160–168. doi:10.1016/j.numecd.2012.09.002

    Article  CAS  PubMed  Google Scholar 

  79. Kasubuchi M, Hasegawa S, Hiramatsu T et al (2015) Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation. Nutrients 7:2839–2849. doi:10.3390/nu7042839

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. den Besten G, van Eunen K, Groen AK et al (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 54:2325–2340. doi:10.1194/jlr.R036012

    Article  CAS  Google Scholar 

  81. Gao Z, Yin J, Zhang J et al (2009) Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58:1509–1517. doi:10.2337/db08-1637

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Fushimi T, Suruga K, Oshima Y et al (2006) Dietary acetic acid reduces serum cholesterol and triacylglycerols in rats fed a cholesterol-rich diet. Br J Nutr 95:916–924

    Article  CAS  PubMed  Google Scholar 

  83. Todesco T, Rao AV, Bosello O, Jenkins DJ (1991) Propionate lowers blood glucose and alters lipid metabolism in healthy subjects. Am J Clin Nutr 54:860–865

    CAS  PubMed  Google Scholar 

  84. Saad MJA, Santos A, Prada PO (2016) Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology (Bethesda) 31:283–293. doi:10.1152/physiol.00041.2015

    CAS  Google Scholar 

  85. Soldavini J, Kaunitz JD (2013) Pathobiology and potential therapeutic value of intestinal short-chain fatty acids in gut inflammation and obesity. Dig Dis Sci 58:2756–2766. doi:10.1007/s10620-013-2744-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Brahe LK, Astrup A, Larsen LH (2013) Is butyrate the link between diet, intestinal microbiota and obesity-related metabolic diseases? Obes Rev 14:950–959. doi:10.1111/obr.12068

    Article  CAS  PubMed  Google Scholar 

  87. De Vadder F, Kovatcheva-Datchary P, Goncalves D et al (2014) Microbiota-generated metabolites promote metabolic benefits via gut–brain neural circuits. Cell 156:84–96. doi:10.1016/j.cell.2013.12.016

    Article  PubMed  CAS  Google Scholar 

  88. Delaere F, Duchampt A, Mounien L et al (2012) The role of sodium-coupled glucose co-transporter 3 in the satiety effect of portal glucose sensing. Mol Metab 2:47–53

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Lacassagne O, Kessler JP (2000) Cellular and subcellular distribution of the amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor subunit GluR2 in the rat dorsal vagal complex. Neuroscience 99:557–563

    Article  CAS  PubMed  Google Scholar 

  90. Greene JG (2014) Causes and consequences of degeneration of the dorsal motor nucleus of the vagus nerve in Parkinson’s disease. Antioxid Redox Signal 21:649–667. doi:10.1089/ars.2014.5859

    Article  CAS  PubMed  Google Scholar 

  91. Lal S, Kirkup AJ, Brunsden AM et al (2001) Vagal afferent responses to fatty acids of different chain length in the rat. AJP: Gastrointest Liver Physiol 281:G907–G915

    CAS  Google Scholar 

  92. Sjogren MJC, Hellstrom PTO, Jonsson MAG et al (2002) Cognition-enhancing effect of vagus nerve stimulation in patients with Alzheimer’s disease: a pilot study. J Clin Psychiatry 63:972–980

    Article  PubMed  Google Scholar 

  93. Merrill CA, Jonsson MAG, Minthon L et al (2006) Vagus nerve stimulation in patients with Alzheimer’s disease: additional follow-up results of a pilot study through 1 year. J Clin Psychiatry 67:1171–1178

    Article  CAS  PubMed  Google Scholar 

  94. Vijay N, Morris ME (2014) Role of monocarboxylate transporters in drug delivery to the brain. Curr Pharm Des 20:1487–1498

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Braniste V, Al-Asmakh M, Kowal C et al (2014) The gut microbiota influences blood–brain barrier permeability in mice. Sci Transl Med 6:263ra158. doi:10.1126/scitranslmed.3009759

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Brown AJ, Goldsworthy SM, Barnes AA et al (2003) The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 278:11312–11319

    Article  CAS  PubMed  Google Scholar 

  97. Beharry AW, Sandesara PB, Roberts BM et al (2014) HDAC1 activates FoxO and is both sufficient and required for skeletal muscle atrophy. J Cell Sci 127:1441–1453. doi:10.1242/jcs.136390

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Martins R, Lithgow GJ, Link W (2016) Long live FOXO: unraveling the role of FOXO proteins in aging and longevity. Aging Cell 15:196–207. doi:10.1111/acel.12427

    Article  CAS  PubMed  Google Scholar 

  99. Pino E, Amamoto R, Zheng L et al (2014) FOXO3 determines the accumulation of alpha-synuclein and controls the fate of dopaminergic neurons in the substantia nigra. Hum Mol Genet 23:1435–1452. doi:10.1093/hmg/ddt530

    Article  CAS  PubMed  Google Scholar 

  100. Xu P, Das M, Reilly J, Davis RJ (2011) JNK regulates FoxO-dependent autophagy in neurons. Genes Dev 25:310–322. doi:10.1101/gad.1984311

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Bahia PK, Pugh V, Hoyland K et al (2012) Neuroprotective effects of phenolic antioxidant tBHQ associate with inhibition of FoxO3a nuclear translocation and activity. J Neurochem 123:182–191. doi:10.1111/j.1471-4159.2012.07877.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Edwards C, Canfield J, Copes N et al (2014) D-beta-hydroxybutyrate extends lifespan in C. elegans. Aging (Albany NY) 6:621–644. doi:10.18632/aging.100683

    Article  CAS  Google Scholar 

  103. Zhang M, Poplawski M, Yen K et al (2009) Role of CBP and SATB-1 in aging, dietary restriction, and insulin-like signaling. PLoS Biol 7:e1000245. doi:10.1371/journal.pbio.1000245

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Cohen E, Bieschke J, Perciavalle RM et al (2006) Opposing activities protect against age-onset proteotoxicity. Science 313:1604–1610. doi:10.1126/science.1124646

    Article  CAS  PubMed  Google Scholar 

  105. Koh H, Kim H, Kim MJ et al (2012) Silent information regulator 2 (Sir2) and Forkhead box O (FOXO) complement mitochondrial dysfunction and dopaminergic neuron loss in Drosophila PTEN-induced kinase 1 (PINK1) null mutant. J Biol Chem 287:12750–12758. doi:10.1074/jbc.M111.337907

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Nankova BB, Agarwal R, MacFabe DF, La Gamma EF (2014) 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 9:e103740

    Article  PubMed  PubMed Central  Google Scholar 

  107. Bourassa MW, Alim I, Bultman SJ, Ratan RR (2016) Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? Neurosci Lett 625:56–63. doi:10.1016/j.neulet.2016.02.009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Reigstad CS, Salmonson CE, Rainey JF et al (2015) Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J 29:1395–1403. doi:10.1096/fj.14-259598

    Article  CAS  PubMed  Google Scholar 

  109. Fukumoto S, Tatewaki M, Yamada T et al (2003) Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats. Am J Physiol Regul Integr Comp Physiol 284:R1269–R1276

    Article  CAS  PubMed  Google Scholar 

  110. Gershon MD (2013) 5-Hydroxytryptamine (serotonin) in the gastrointestinal tract. Curr Opin Endocrinol Diabetes Obes 20:14–21. doi:10.1097/MED.0b013e32835bc703

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. DeCastro M, Nankova BB, Shah P et al (2005) Short chain fatty acids regulate tyrosine hydroxylase gene expression through a cAMP-dependent signaling pathway. Brain Res Mol Brain Res 142:28–38. doi:10.1016/j.molbrainres.2005.09.002

    Article  CAS  PubMed  Google Scholar 

  112. Govindarajan N, Agis-Balboa RC, Walter J et al (2011) Sodium butyrate improves memory function in an Alzheimer’s disease mouse model when administered at an advanced stage of disease progression. J Alzheimers Dis 26:187–197. doi:10.3233/JAD-2011-110080

    CAS  PubMed  Google Scholar 

  113. del Rio R, Noubade R, Saligrama N et al (2012) Histamine H4 receptor optimizes T regulatory cell frequency and facilitates anti-inflammatory responses within the central nervous system. J Immunol 188:541–547. doi:10.4049/jimmunol.1101498

    Article  PubMed  Google Scholar 

  114. Naddafi F, Mirshafiey A (2013) The neglected role of histamine in Alzheimer’s disease. Am J Alzheimers Dis Other Demen 28:327–336. doi:10.1177/1533317513488925

    Article  PubMed  Google Scholar 

  115. Dong H, Zhang W, Zeng X et al (2014) Histamine induces upregulated expression of histamine receptors and increases release of inflammatory mediators from microglia. Mol Neurobiol 49:1487–1500. doi:10.1007/s12035-014-8697-6

    Article  CAS  PubMed  Google Scholar 

  116. Schneider E, Rolli-Derkinderen M, Arock M, Dy M (2002) Trends in histamine research: new functions during immune responses and hematopoiesis. Trends Immunol 23:255–263

    Article  CAS  PubMed  Google Scholar 

  117. Morgan RK, McAllister B, Cross L et al (2007) Histamine 4 receptor activation induces recruitment of FoxP3+ T cells and inhibits allergic asthma in a murine model. J Immunol 178:8081–8089

    Article  CAS  PubMed  Google Scholar 

  118. Landete JM, de las Rivas B, Marcobal A, Munoz R (2008) Updated molecular knowledge about histamine biosynthesis by bacteria. Crit Rev Food Sci Nutr 48:697–714. doi:10.1080/10408390701639041

    Article  CAS  PubMed  Google Scholar 

  119. Thomas CM, Hong T, van Pijkeren JP et al (2012) Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling. PLoS One 7:e31951

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Alvarez XA, Franco A, Fernandez-Novoa L, Cacabelos R (1996) Blood levels of histamine, IL-1 beta, and TNF-alpha in patients with mild to moderate Alzheimer disease. Mol Chem Neuropathol 29:237–252. doi:10.1007/BF02815005

    Article  CAS  PubMed  Google Scholar 

  121. Stasiak A, Mussur M, Unzeta M et al (2011) The central histamine level in rat model of vascular dementia. J Physiol Pharmacol 62:549–558

    CAS  PubMed  Google Scholar 

  122. Chen L, Xing T, Wang M et al (2011) Local infusion of ghrelin enhanced hippocampal synaptic plasticity and spatial memory through activation of phosphoinositide 3-kinase in the dentate gyrus of adult rats. Eur J Neurosci 33:266–275. doi:10.1111/j.1460-9568.2010.07491.x

    Article  PubMed  Google Scholar 

  123. Gahete MD, Cordoba-Chacon J, Kineman RD et al (2011) Role of ghrelin system in neuroprotection and cognitive functions: implications in Alzheimer’s disease. Peptides 32:2225–2228. doi:10.1016/j.peptides.2011.09.019

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Steinert RE, Feinle-Bisset C, Asarian L et al (2017) Ghrelin, CCK, GLP-1, and PYY(3-36): secretory controls and physiological roles in eating and glycemia in health, obesity, and after RYGB. Physiol Rev 97:411–463. doi:10.1152/physrev.00031.2014

    Article  PubMed  Google Scholar 

  125. Guan XM, Yu H, Palyha OC et al (1997) Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res Mol Brain Res 48:23–29

    Article  CAS  PubMed  Google Scholar 

  126. Bauer PV, Hamr SC, Duca FA (2016) Regulation of energy balance by a gut–brain axis and involvement of the gut microbiota. Cell Mol Life Sci 73:737–755. doi:10.1007/s00018-015-2083-z

    Article  CAS  PubMed  Google Scholar 

  127. Fallucca F, Porrata C, Fallucca S, Pianesi M (2014) Influence of diet on gut microbiota, inflammation and type 2 diabetes mellitus. First experience with macrobiotic Ma-Pi 2 diet. Diabetes Metab Res Rev 30(Suppl 1):48–54. doi:10.1002/dmrr.2518

    Article  CAS  PubMed  Google Scholar 

  128. Queipo-Ortuno MI, Seoane LM, Murri M et al (2013) 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 8:e65465

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Dos Santos VV, Rodrigues ALS, De Lima TC et al (2013) Ghrelin as a neuroprotective and palliative agent in Alzheime’s and Parkinson’s disease. Curr Pharm Des 19:6773–6790

    Article  PubMed  CAS  Google Scholar 

  130. Liu Y, Chen L, Xu X et al (2009) Both ischemic preconditioning and ghrelin administration protect hippocampus from ischemia/reperfusion and upregulate uncoupling protein-2. BMC Physiol 9:17. doi:10.1186/1472-6793-9-17

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Moon M, Choi JG, Nam DW et al (2011) Ghrelin ameliorates cognitive dysfunction and neurodegeneration in intrahippocampal amyloid-beta1-42 oligomer-injected mice. J Alzheimers Dis 23:147–159. doi:10.3233/JAD-2010-101263

    CAS  PubMed  Google Scholar 

  132. Andrews ZB, Erion D, Beiler R et al (2009) Ghrelin promotes and protects nigrostriatal dopamine function via a UCP2-dependent mitochondrial mechanism. J Neurosci 29:14057–14065. doi:10.1523/JNEUROSCI.3890-09.2009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bayliss JA, Lemus M, Santos VV et al (2016) Acylated but not des-acyl ghrelin is neuroprotective in an MPTP mouse model of Parkinson’s disease. J Neurochem 137:460–471. doi:10.1111/jnc.13576

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Schwarcz R, Bruno JP, Muchowski PJ, Wu H-Q (2012) Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci 13:465–477. doi:10.1038/nrn3257

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Ruddick JP, Evans AK, Nutt DJ et al (2006) Tryptophan metabolism in the central nervous system: medical implications. Expert Rev Mol Med 8:1–27. doi:10.1017/S1462399406000068

    Article  PubMed  Google Scholar 

  136. Desbonnet L, Garrett L, Clarke G et al (2008) The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J Psychiatr Res 43:164–174. doi:10.1016/j.jpsychires.2008.03.009

    Article  PubMed  Google Scholar 

  137. Braidy N, Grant R, Adams S et al (2009) Mechanism for quinolinic acid cytotoxicity in human astrocytes and neurons. Neurotox Res 16:77–86. doi:10.1007/s12640-009-9051-z

    Article  CAS  PubMed  Google Scholar 

  138. Vamos E, Pardutz A, Klivenyi P et al (2009) The role of kynurenines in disorders of the central nervous system: possibilities for neuroprotection. J Neurol Sci 283:21–27. doi:10.1016/j.jnd.2009.02.326

    Article  CAS  PubMed  Google Scholar 

  139. Guillemin GJ, Williams KR, Smith DG et al (2003) Quinolinic acid in the pathogenesis of Alzheimer’s disease. Adv Exp Med Biol 527:167–176

    Article  CAS  PubMed  Google Scholar 

  140. Widner B, Leblhuber F, Walli J et al (2000) Tryptophan degradation and immune activation in Alzheimer’s disease. J Neural Transm 107:343–353

    Article  CAS  PubMed  Google Scholar 

  141. Clarke G, Grenham S, Scully P et al (2013) The microbiome–gut–brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 18:666–673. doi:10.1038/mp.2012.77

    Article  CAS  PubMed  Google Scholar 

  142. Wikoff WR, Anfora AT, Liu J et al (2009) Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci USA 106:3698–3703. doi:10.1073/pnas.0812874106

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Freewan M, Rees MD, Plaza TSS et al (2013) Human indoleamine 2,3-dioxygenase is a catalyst of physiological heme peroxidase reactions: implications for the inhibition of dioxygenase activity by hydrogen peroxide. J Biol Chem 288:1548–1567. doi:10.1074/jbc.M112.410993

    Article  CAS  PubMed  Google Scholar 

  144. Tomas MSJ, Claudia Otero M, Ocana V, Elena Nader-Macias M (2004) Production of antimicrobial substances by lactic acid bacteria I: determination of hydrogen peroxide. Methods Mol Biol 268:337–346. doi:10.1385/1-59259-766-1:337

    CAS  PubMed  Google Scholar 

  145. Rahman A, Ting K, Cullen KM et al (2009) The excitotoxin quinolinic acid induces tau phosphorylation in human neurons. PLoS One 4:e6344. doi:10.1371/journal.pone.0006344

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Guillemin GJ, Brew BJ, Noonan CE et al (2005) Indoleamine 2,3 dioxygenase and quinolinic acid immunoreactivity in Alzheimer’s disease hippocampus. Neuropathol Appl Neurobiol 31:395–404. doi:10.1111/j.1365-2990.2005.00655.x

    Article  CAS  PubMed  Google Scholar 

  147. Yamada A, Akimoto H, Kagawa S et al (2009) Proinflammatory cytokine interferon-gamma increases induction of indoleamine 2,3-dioxygenase in monocytic cells primed with amyloid beta peptide 1–42: implications for the pathogenesis of Alzheimer’s disease. J Neurochem 110:791–800. doi:10.1111/j.1471-4159.2009.06175.x

    Article  CAS  PubMed  Google Scholar 

  148. Giorgini F, Guidetti P, Nguyen Q et al (2005) A genomic screen in yeast implicates kynurenine 3-monooxygenase as a therapeutic target for Huntington disease. Nat Genet 37:526–531. doi:10.1038/ng1542

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Forsythe P, Kunze WA, Bienenstock J (2012) On communication between gut microbes and the brain. Curr Opin Gastroenterol 28:557–562. doi:10.1097/MOG.0b013e3283572ffa

    Article  PubMed  Google Scholar 

  150. Lyte M (2014) Microbial endocrinology: host–microbiota neuroendocrine interactions influencing brain and behavior. Gut Microbes 5:381–389. doi:10.4161/gmic.28682

    Article  PubMed  PubMed Central  Google Scholar 

  151. Barrett E, Ross RP, O’Toole PW et al (2012) gamma-Aminobutyric acid production by culturable bacteria from the human intestine. J Appl Microbiol 113:411–417. doi:10.1111/j.1365-2672.2012.05344.x

    Article  CAS  PubMed  Google Scholar 

  152. Sun J, Ling Z, Wang F et al (2016) Clostridium butyricum pretreatment attenuates cerebral ischemia/reperfusion injury in mice via anti-oxidation and anti-apoptosis. Neurosci Lett 613:30–35. doi:10.1016/j.neulet.2015.12.047

    Article  CAS  PubMed  Google Scholar 

  153. Ait-Belgnaoui A, Colom A, Braniste V et al (2014) Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterol Motil 26:510–520. doi:10.1111/nmo.12295

    Article  CAS  PubMed  Google Scholar 

  154. Ezendam J, de Klerk A, Gremmer ER, van Loveren H (2008) Effects of Bifidobacterium animalis administered during lactation on allergic and autoimmune responses in rodents. Clin Exp Immunol 154:424–431. doi:10.1111/j.1365-2249.2008.03788.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Ochoa-Reparaz J, Mielcarz DW, Wang Y et al (2010) A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol 3:487–495. doi:10.1038/mi.2010.29

    Article  CAS  PubMed  Google Scholar 

  156. Kobayashi T, Kato I, Nanno M et al (2010) Oral administration of probiotic bacteria, Lactobacillus casei and Bifidobacterium breve, does not exacerbate neurological symptoms in experimental autoimmune encephalomyelitis. Immunopharmacol Immunotoxicol 32:116–124. doi:10.3109/08923970903200716

    Article  CAS  PubMed  Google Scholar 

  157. Kobayashi T, Suzuki T, Kaji R et al (2012) Probiotic upregulation of peripheral IL-17 responses does not exacerbate neurological symptoms in experimental autoimmune encephalomyelitis mouse models. Immunopharmacol Immunotoxicol 34:423–433. doi:10.3109/08923973.2010.617755

    Article  CAS  PubMed  Google Scholar 

  158. Kwon H-K, Kim G-C, Kim Y et al (2013) Amelioration of experimental autoimmune encephalomyelitis by probiotic mixture is mediated by a shift in T helper cell immune response. Clin Immunol 146:217–227. doi:10.1016/j.clim.2013.01.001

    Article  CAS  PubMed  Google Scholar 

  159. Lavasani S, Dzhambazov B, Nouri M et al (2010) A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS One 5:e9009. doi:10.1371/journal.pone.0009009

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  160. Ohland CL, Kish L, Bell H et al (2013) Effects of Lactobacillus helveticus on murine behavior are dependent on diet and genotype and correlate with alterations in the gut microbiome. Psychoneuroendocrinology 38:1738–1747. doi:10.1016/j.psyneuen.2013.02.008

    Article  CAS  PubMed  Google Scholar 

  161. Davari S, Talaei SA, Alaei H, Salami M (2013) Probiotics treatment improves diabetes-induced impairment of synaptic activity and cognitive function: behavioral and electrophysiological proofs for microbiome–gut–brain axis. Neuroscience 240:287–296. doi:10.1016/j.neuroscience.2013.02.055

    Article  CAS  PubMed  Google Scholar 

  162. Gareau MG, Wine E, Rodrigues DM et al (2011) Bacterial infection causes stress-induced memory dysfunction in mice. Gut 60:307–317. doi:10.1136/gut.2009.202515

    Article  PubMed  Google Scholar 

  163. Rao AV, Bested AC, Beaulne TM et al (2009) A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog 1:6. doi:10.1186/1757-4749-1-6

    Article  PubMed  PubMed Central  Google Scholar 

  164. Yan MH, Wang X, Zhu X (2013) Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic Biol Med 62:90–101. doi:10.1016/j.freeradbiomed.2012.11.014

    Article  CAS  PubMed  Google Scholar 

  165. Distrutti E, O’Reilly J-A, McDonald C et al (2014) Modulation of intestinal microbiota by the probiotic VSL#3 resets brain gene expression and ameliorates the age-related deficit in LTP. PLoS One 9:e106503

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Girard S-A, Bah TM, Kaloustian S et al (2009) Lactobacillus helveticus and Bifidobacterium longum taken in combination reduce the apoptosis propensity in the limbic system after myocardial infarction in a rat model. Br J Nutr 102:1420–1425. doi:10.1017/S0007114509990766

    Article  CAS  PubMed  Google Scholar 

  167. Liu J, Sun J, Wang F et al (2015) Neuroprotective effects of Clostridium butyricum against vascular dementia in mice via metabolic butyrate. Biomed Res Int 2015:412946. doi:10.1155/2015/412946

    PubMed  PubMed Central  Google Scholar 

  168. O’Sullivan E, Barrett E, Grenham S et al (2011) BDNF expression in the hippocampus of maternally separated rats: does Bifidobacterium breve 6330 alter BDNF levels? Benef Microbes 2:199–207. doi:10.3920/BM2011.0015

    Article  PubMed  CAS  Google Scholar 

  169. Bercik P, Park AJ, Sinclair D et al (2011) The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterol Motil 23:1132–1139. doi:10.1111/j.1365-2982.2011.01796.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Le TK, Hosaka T, Le TT et al (2014) Oral administration of Bifidobacterium spp. improves insulin resistance, induces adiponectin, and prevents inflammatory adipokine expressions. Biomed Res Int 35:303–310

    Article  CAS  Google Scholar 

  171. Bomhof MR, Saha DC, Reid DT et al (2014) Combined effects of oligofructose and Bifidobacterium animalis on gut microbiota and glycemia in obese rats. Obesity (Silver Spring) 22:763–771. doi:10.1002/oby.20632

    Article  CAS  Google Scholar 

  172. Hoarau C, Martin L, Faugaret D et al (2008) Supernatant from bifidobacterium differentially modulates transduction signaling pathways for biological functions of human dendritic cells. PLoS One 3:e2753. doi:10.1371/journal.pone.0002753

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Chang C-Y, Ke D-S, Chen J-Y (2009) Essential fatty acids and human brain. Acta Neurol Taiwan 18:231–241

    PubMed  Google Scholar 

  174. Wall R, Marques TM, O’Sullivan O et al (2012) Contrasting effects of Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330 on the composition of murine brain fatty acids and gut microbiota. Am J Clin Nutr 95:1278–1287. doi:10.3945/ajcn.111.026435

    Article  CAS  PubMed  Google Scholar 

  175. McCarthy J, O’Mahony L, O’Callaghan L et al (2003) Double blind, placebo controlled trial of two probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. Gut 52:975–980

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Rodes L, Khan A, Paul A et al (2013) Effect of probiotics Lactobacillus and Bifidobacterium on gut-derived lipopolysaccharides and inflammatory cytokines: an in vitro study using a human colonic microbiota model. J Microbiol Biotechnol 23:518–526

    Article  CAS  PubMed  Google Scholar 

  177. Sudo N, Chida Y, Aiba Y et al (2004) Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J Physiol 558:263–275. doi:10.1113/jphysiol.2004.063388

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Stone TW, Stoy N, Darlington LG (2013) An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol Sci 34:136–143. doi:10.1016/j.tips.2012.09.006

    Article  CAS  PubMed  Google Scholar 

  179. Mao Y-K, Kasper DL, Wang B et al (2013) Bacteroides fragilis polysaccharide A is necessary and sufficient for acute activation of intestinal sensory neurons. Nat Commun 4:1465. doi:10.1038/ncomms2478

    Article  PubMed  CAS  Google Scholar 

  180. Kuley E, Ozogul F, Ozogul Y, Akyol I (2011) The function of lactic acid bacteria and brine solutions on biogenic amine formation by foodborne pathogens in trout fillets. Food Chem 129:1211–1216. doi:10.1016/j.foodchem.2011.05.113

    Article  CAS  PubMed  Google Scholar 

  181. Lin Q (2013) Submerged fermentation of Lactobacillus rhamnosus YS9 for gamma-aminobutyric acid (GABA) production. Braz J Microbiol 44:183–187

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Stanaszek PM, Snell JF, O’Neill JJ (1977) Isolation, extraction, and measurement of acetylcholine from Lactobacillus plantarum. Appl Environ Microbiol 34:237–239

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Sobko T, Huang L, Midtvedt T et al (2006) Generation of NO by probiotic bacteria in the gastrointestinal tract. Free Radic Biol Med 41:985–991. doi:10.1016/j.freeradbiomed.2006.06.020

    Article  CAS  PubMed  Google Scholar 

  184. Bajaj JS, Heuman DM, Hylemon PB et al (2014) Randomised clinical trial: Lactobacillus GG modulates gut microbiome, metabolome and endotoxemia in patients with cirrhosis. Aliment Pharmacol Ther 39:1113–1125. doi:10.1111/apt.12695

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Fetissov SO, Hamze Sinno M, Coeffier M et al (2008) Autoantibodies against appetite-regulating peptide hormones and neuropeptides: putative modulation by gut microflora. Nutrition 24:348–359. doi:10.1016/j.nut.2007.12.006

    Article  CAS  PubMed  Google Scholar 

  186. Konstantinov SR, Smidt H, de Vos WM et al (2008) S layer protein A of Lactobacillus acidophilus NCFM regulates immature dendritic cell and T cell functions. Proc Natl Acad Sci USA 105:19474–19479. doi:10.1073/pnas.0810305105

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Amdekar S, Singh V, Kumar A et al (2013) Lactobacillus casei and Lactobacillus acidophilus regulate inflammatory pathway and improve antioxidant status in collagen-induced arthritic rats. J Interferon Cytokine Res 33:1–8. doi:10.1089/jir.2012.0034

    Article  CAS  PubMed  Google Scholar 

  188. Bravo JA, Julio-Pieper M, Forsythe P et al (2012) Communication between gastrointestinal bacteria and the nervous system. Curr Opin Pharmacol 12:667–672. doi:10.1016/j.coph.2012.09.010

    Article  CAS  PubMed  Google Scholar 

  189. Bhathena J, Duchesneau CT (2012) Effect of orally administered microencapsulated FA-producing L. fermentum on markers of metabolic syndrome: an in vivo analysis. J Diabetes. doi:10.4172/2155-6156.S2-009

    Google Scholar 

  190. Tomaro-Duchesneau C, Saha S, Malhotra M et al (2012) Lactobacillus fermentum NCIMB 5221 has a greater ferulic acid production compared to other ferulic acid esterase producing Lactobacilli. Int J Probiotics Prebiotics 7(1):23–32

    Google Scholar 

  191. Grompone G, Martorell P, Llopis S et al (2012) Anti-inflammatory Lactobacillus rhamnosus CNCM I-3690 strain protects against oxidative stress and increases lifespan in Caenorhabditis elegans. PLoS One 7:e52493

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Yan F, Cao H, Cover TL et al (2007) Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 132:562–575. doi:10.1053/j.gastro.2006.11.022

    Article  CAS  PubMed  Google Scholar 

  193. Sanchez B, Urdaci MC, Margolles A (2010) Extracellular proteins secreted by probiotic bacteria as mediators of effects that promote mucosa–bacteria interactions. Microbiology 156:3232–3242. doi:10.1099/mic.0.044057-0

    Article  CAS  PubMed  Google Scholar 

  194. Claes IJJ, Lebeer S, Shen C et al (2010) Impact of lipoteichoic acid modification on the performance of the probiotic Lactobacillus rhamnosus GG in experimental colitis. Clin Exp Immunol 162:306–314. doi:10.1111/j.1365-2249.2010.04228.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Sagar S, Morgan ME, Chen S et al (2014) Bifidobacterium breve and Lactobacillus rhamnosus treatment is as effective as budesonide at reducing inflammation in a murine model for chronic asthma. Respir Res 15:46. doi:10.1186/1465-9921-15-46

    Article  PubMed  PubMed Central  Google Scholar 

  196. Ushakova G, Fed’kiv O, Prykhod’ko O et al (2009) The effect of long-term lactobacilli (lactic acid bacteria) enteral treatment on the central nervous system of growing rats. J Nutr Biochem 20:677–684. doi:10.1016/j.jnutbio.2008.06.010

    Article  CAS  PubMed  Google Scholar 

  197. Pereira DIA, McCartney AL, Gibson GR (2003) An in vitro study of the probiotic potential of a bile-salt-hydrolyzing Lactobacillus fermentum strain, and determination of its cholesterol-lowering properties. Appl Environ Microbiol 69:4743–4752

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Duary RK, Bhausaheb MA, Batish VK, Grover S (2012) Anti-inflammatory and immunomodulatory efficacy of indigenous probiotic Lactobacillus plantarum Lp91 in colitis mouse model. Mol Biol Rep 39:4765–4775. doi:10.1007/s11033-011-1269-1

    Article  CAS  PubMed  Google Scholar 

  199. Tsavkelova EA, Botvinko IV, Kudrin VS, Oleskin AV (2000) Detection of neurotransmitter amines in microorganisms with the use of high-performance liquid chromatography. Dokl Biochem 372:115–117

    CAS  PubMed  Google Scholar 

  200. Chen Z, Guo L, Zhang Y et al (2014) Incorporation of therapeutically modified bacteria into gut microbiota inhibits obesity. J Clin Invest 124:3391–3406. doi:10.1172/JCI72517

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Shishov VA, Kirovskaia TA, Kudrin VS, Oleskin AV (2009) Amine neuromediators, their precursors, and oxidation products in the culture of Escherichia coli K-12. Prikl Biokhim Mikrobiol 45:550–554

    CAS  PubMed  Google Scholar 

  202. Pryde S, Duncan S, Hold G et al (2002) The microbiology of butyrate formation in the human colon. FEMS Microbiol Lett 217:133–139

    Article  CAS  PubMed  Google Scholar 

  203. Macfarlane S, Macfarlane GT (2003) Regulation of short-chain fatty acid production. Proc Nutr Soc 62:67–72

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

This review was conceptualized and written by SW with supporting sections written and edited by NL and SYD. Advisement and manuscript suggestions were provided by SPS and SP.

Corresponding author

Correspondence to Satya Prakash.

Ethics declarations

Conflict of interest

The authors declare that there is no conflict of interest or competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Westfall, S., Lomis, N., Kahouli, I. et al. Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis. Cell. Mol. Life Sci. 74, 3769–3787 (2017). https://doi.org/10.1007/s00018-017-2550-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-017-2550-9

Keywords

Navigation