Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour

Key Points

  • There is a growing appreciation that the gut microbiota plays a key role in maintaining homeostasis and that a disruption in its composition contributes to various disease states, including CNS disorders.

  • The concept of a microbiota–gut–brain axis, although debated, is emerging to capture the importance that the microbiota has on regulating bidirectional gut–brain communication pathways.

  • It is clear that stress, including stress in early life, can alter microbiota composition and this can have marked consequences on physiology in adulthood.

  • Studies in germ-free animals and in animals exposed to pathogenic bacterial infections, probiotic bacteria or antibiotic drugs suggest a role for the gut microbiota in the regulation of anxiety, mood, cognition and pain.

  • Although not as conceptually or empirically developed, the gut microbiota has also been implicated in obesity, autism and multiple sclerosis.

  • Mechanisms as to how the microbiota are affecting gut–brain signalling are only now being unravelled. These mechanisms may include alterations in microbial composition, immune activation, vagus nerve signalling, alterations in tryptophan metabolism, production of specific microbial neuroactive metabolites and bacterial cell wall sugars.

  • Harnessing such mechanisms may pave the way for microbial-based therapeutics for various CNS disorders.

Abstract

Recent years have witnessed the rise of the gut microbiota as a major topic of research interest in biology. Studies are revealing how variations and changes in the composition of the gut microbiota influence normal physiology and contribute to diseases ranging from inflammation to obesity. Accumulating data now indicate that the gut microbiota also communicates with the CNS — possibly through neural, endocrine and immune pathways — and thereby influences brain function and behaviour. Studies in germ-free animals and in animals exposed to pathogenic bacterial infections, probiotic bacteria or antibiotic drugs suggest a role for the gut microbiota in the regulation of anxiety, mood, cognition and pain. Thus, the emerging concept of a microbiota–gut–brain axis suggests that modulation of the gut microbiota may be a tractable strategy for developing novel therapeutics for complex CNS disorders.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Pathways involved in bidirectional communication between the gut microbiota and the brain.
Figure 2: Strategies used to investigate the role of the microbiota–gut–brain axis in health and disease.
Figure 3: Impact of the gut microbiota on the gut–brain axis in health and disease.

Similar content being viewed by others

References

  1. Sekirov, I., Russell, S. L., Antunes, L. C. & Finlay, B. B. Gut microbiota in health and disease. Physiol. Rev. 90, 859–904 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Clemente, J. C., Ursell, L. K., Parfrey, L. W. & Knight, R. The impact of the gut microbiota on human health: an integrative view. Cell 148, 1258–1270 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Human Microbiome Project Consortium. A framework for human microbiome research. Nature 486, 215–221 (2012).

  4. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214 (2012).

  5. Banks, W. A. The blood–brain barrier: connecting the gut and the brain. Regul. Pept. 149, 11–14 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mayer, E. A. Gut feelings: the emerging biology of gut–brain communication. Nature Rev. Neurosci. 12, 453–466 (2011). A comprehensive recent review of the underlying neurobiology and bidirectional nature of the gut–brain axis.

    Article  CAS  Google Scholar 

  7. Aziz, Q. & Thompson, D. G. Brain–gut axis in health and disease. Gastroenterology 114, 559–578 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Tache, Y., Vale, W., Rivier, J. & Brown, M. Brain regulation of gastric secretion: influence of neuropeptides. Proc. Natl Acad. Sci. USA 77, 5515–5519 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Konturek, S. J., Konturek, J. W., Pawlik, T. & Brzozowki, T. Brain–gut axis and its role in the control of food intake. J. Physiol. Pharmacol. 55, 137–154 (2004).

    CAS  PubMed  Google Scholar 

  10. Rhee, S. H., Pothoulakis, C. & Mayer, E. A. Principles and clinical implications of the brain–gut–enteric microbiota axis. Nature Rev. Gastroenterol. Hepatol. 6, 306–314 (2009). One of the first papers to formalize the concept of a microbiota–gut–brain axis.

    Article  CAS  Google Scholar 

  11. Reber, S. O. Stress and animal models of inflammatory bowel disease — an update on the role of the hypothalamo–pituitary–adrenal axis. Psychoneuroendocrinology 37, 1–19 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Gill, S. R. et al. Metagenomic analysis of the human distal gut microbiome. Science 312, 1355–1359 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Qin, J. et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 59–65 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. O'Hara, A. M. & Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 7, 688–693 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Backhed, F., Ley, R. E., Sonnenburg, J. L., Peterson, D. A. & Gordon, J. I. Host–bacterial mutualism in the human intestine. Science 307, 1915–1920 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Round, J. L., O'Connell, R. M. & Mazmanian, S. K. Coordination of tolerogenic immune responses by the commensal microbiota. J. Autoimmun. 34, J220–J225 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Olszak, T. et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489–493 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Backhed, F. et al. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl Acad. Sci. USA 101, 15718–15723 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bercik, P., Collins, S. M. & Verdu, E. F. Microbes and the gut–brain axis. Neurogastroenterol. Motil. 24, 405–413 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Fraher, M. H., O'Toole, P. W. & Quigley, E. M. Techniques used to characterize the gut microbiota: a guide for the clinician. Nature Rev. Gastroenterol. Hepatol. 9, 312–322 (2012).

    Article  CAS  Google Scholar 

  21. Ley, R. E., Peterson, D. A. & Gordon, J. I. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell 124, 837–848 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Eckburg, P. B. et al. Diversity of the human intestinal microbial flora. Science 308, 1635–1638 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Grenham, S., Clarke, G., Cryan, J. & Dinan, T. G. Brain–gut–microbe communication in health and disease. Front. Physiol. 2, 94 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Mackie, R. I., Sghir, A. & Gaskins, H. R. Developmental microbial ecology of the neonatal gastrointestinal tract. Am. J. Clin. Nutr. 69, 1035S–1045S (1999).

    Article  CAS  PubMed  Google Scholar 

  25. Palmer, C., Bik, E. M., DiGiulio, D. B., Relman, D. A. & Brown, P. O. Development of the human infant intestinal microbiota. PLoS Biol. 5, e177 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gulati, A. S. et al. Mouse background strain profoundly influences Paneth cell function and intestinal microbial composition. PLoS ONE 7, e32403 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Arumugam, M. et al. Enterotypes of the human gut microbiome. Nature 473, 174–180 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cryan, J. F. & O'Mahony, S. M. The microbiome–gut–brain axis: from bowel to behavior. Neurogastroenterol. Motil. 23, 187–192 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Wu, S. V. & Hui, H. Treat your bug right. Front. Physiol. 2, 9 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Forsythe, P., Sudo, N., Dinan, T., Taylor, V. H. & Bienenstock, J. Mood and gut feelings. Brain Behav. Immun. 24, 9–16 (2010).

    Article  PubMed  Google Scholar 

  31. Claesson, M. J. et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl Acad. Sci. USA 108, 4586–4591 (2011).

    Article  PubMed  Google Scholar 

  32. Claesson, M. J. et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 488, 178–184 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Collins, S. M. & Bercik, P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology 136, 2003–2014 (2009).

    Article  PubMed  Google Scholar 

  34. Tannock, G. W. & Savage, D. C. Influences of dietary and environmental stress on microbial populations in the murine gastrointestinal tract. Infect. Immun. 9, 591–598 (1974).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Dinan, T. G. & Cryan, J. F. Regulation of the stress response by the gut microbiota: implications for psychoneuroendocrinology. Psychoneuroendocrinology 37, 1369–1378 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. O'Mahony, S. M., Hyland, N. P., Dinan, T. G. & Cryan, J. F. Maternal separation as a model of brain–gut axis dysfunction. Psychopharmacology 214, 71–88 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Bailey, M. T. & Coe, C. L. Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev. Psychobiol. 35, 146–155 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. O'Mahony, S. M. et al. Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol. Psychiatry 65, 263–267 (2009). An important study demonstrating that stress early in life alters brain–gut axis function and also modifies the relative diversity of the gut microbiota.

    Article  PubMed  Google Scholar 

  39. Bailey, M. T. et al. Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain Behav. Immun. 25, 397–407 (2011). This study is one of the first to show that stress in adulthood modifies the composition of the gut microbiota.

    Article  CAS  PubMed  Google Scholar 

  40. Santos, J., Yang, P. C., Soderholm, J. D., Benjamin, M. & Perdue, M. H. Role of mast cells in chronic stress induced colonic epithelial barrier dysfunction in the rat. Gut 48, 630–636 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Soderholm, J. D. & Perdue, M. H. Stress and gastrointestinal tract. II. Stress and intestinal barrier function. Am. J. Physiol. Gastrointest. Liver Physiol. 280, G7–G13 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Zareie, M. et al. Probiotics prevent bacterial translocation and improve intestinal barrier function in rats following chronic psychological stress. Gut 55, 1553–1560 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ait-Belgnaoui, A. et al. Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology 25 April 2012 (doi:10.1016/j.psyneuen.2012.03.02).

  44. Maes, M., Kubera, M., Leunis, J. C. & Berk, M. Increased IgA and IgM responses against gut commensals in chronic depression: further evidence for increased bacterial translocation or leaky gut. J. Affect. Disord. 141, 55–62 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Gems, D. & Partridge, L. Stress-response hormesis and aging: “that which does not kill us makes us stronger”. Cell. Metab. 7, 200–203 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Franceschi, C. et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. NY Acad. Sci. 908, 244–254 (2000).

    Article  CAS  PubMed  Google Scholar 

  47. Sudo, N. et al. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J. Physiol. 558, 263–275 (2004). A landmark study showing that germ-free mice have altered HPA axis function, which can be reversed by colonization with specific bacterial strains early in life.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Clarke, G. et al. The microbiome–gut–brain axis during early-life regulates the hippocampal serotonergic system in a gender-dependent manner. Mol. Psychiatry 12 Jun 2012 (doi:10.1038/mp.2012.77).

  49. Heijtz, R. D. et al. Normal gut microbiota modulates brain development and behavior. Proc. Natl Acad. Sci. USA 108, 3047–3052 (2011).

    Article  PubMed Central  Google Scholar 

  50. Neufeld, K. M., Kang, N., Bienenstock, J. & Foster, J. A. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol. Motil. 23, 255–264 (2010). References 48–50 are important studies linking the gut microbiota to neurodevelopmental processes and behaviour. They independently show that germ-free mice have alterations in concentrations of neurotransmitters and neurotrophic factors in the brain, and have reduced anxiety-like behaviour.

    Article  PubMed  Google Scholar 

  51. Gareau, M. G. et al. Bacterial infection causes stress-induced memory dysfunction in mice. Gut 60, 307–317 (2011). One of the first studies to assess cognitive function in germ-free mice, therefore showing that the gut microbiota may be a therapeutic target for cognitive enhancement.

    Article  PubMed  Google Scholar 

  52. Cryan, J. F. & Sweeney, F. F. The age of anxiety: role of animal models of anxiolytic action in drug discovery. Br. J. Pharmacol. 164, 1129–1161 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Bergami, M. et al. Deletion of TrkB in adult progenitors alters newborn neuron integration into hippocampal circuits and increases anxiety-like behavior. Proc. Natl Acad. Sci. USA 105, 15570–15575 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Akimova, E., Lanzenberger, R. & Kasper, S. The serotonin-1A receptor in anxiety disorders. Biol. Psychiatry 66, 627–635 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Barkus, C. et al. Hippocampal NMDA receptors and anxiety: at the interface between cognition and emotion. Eur. J. Pharmacol. 626, 49–56 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Jacobson, L. H. & Cryan, J. F. Feeling strained? Influence of genetic background on depression-related behavior in mice: a review. Behav. Genet. 37, 171–213 (2007).

    Article  CAS  PubMed  Google Scholar 

  57. Benson, A. K. et al. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc. Natl Acad. Sci. USA 107, 18933–18938 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Esworthy, R. S., Smith, D. D. & Chu, F. F. A. Strong impact of genetic background on gut microflora in mice. Int. J. Inflam. 2010, 986046 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kovacs, A. et al. Genotype is a stronger determinant than sex of the mouse gut microbiota. Microb. Ecol. 61, 423–428 (2011).

    Article  PubMed  Google Scholar 

  60. Bercik, P. et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 141, 599–609.e3 (2011). A key study showing the utility of microbiota transplantation in mice to examine the microbiota–gut–brain axis.

    Article  CAS  PubMed  Google Scholar 

  61. Bercik, P. et al. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology 139, 2102–2112.e1 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Lyte, M., Li, W., Opitz, N., Gaykema, R. & Goehler, L. E. Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium. Physiol. Behav. 89, 350–357 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Kennedy, P. J. et al. Gut memories: towards a cognitive neurobiology of irritable bowel syndrome. Neurosci. Biobehav. Rev. 36, 310–340 (2012).

    Article  PubMed  Google Scholar 

  64. O'Malley, D., Quigley, E. M., Dinan, T. G. & Cryan, J. F. Do interactions between stress and immune responses lead to symptom exacerbations in irritable bowel syndrome? Brain Behav. Immun. 25, 1333–1341 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Gaykema, R. P., Goehler, L. E. & Lyte, M. Brain response to cecal infection with Campylobacter jejuni: analysis with Fos immunohistochemistry. Brain Behav. Immun. 18, 238–245 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Goehler, L. E., Park, S. M., Opitz, N., Lyte, M. & Gaykema, R. P. Campylobacter jejuni infection increases anxiety-like behavior in the holeboard: possible anatomical substrates for viscerosensory modulation of exploratory behavior. Brain Behav. Immun. 22, 354–366 (2008).

    Article  CAS  PubMed  Google Scholar 

  67. Wang, X. et al. Evidences for vagus nerve in maintenance of immune balance and transmission of immune information from gut to brain in STM-infected rats. World J. Gastroenterol. 8, 540–545 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Gareau, M. G., Sherman, P. M. & Walker, W. A. Probiotics and the gut microbiota in intestinal health and disease. Nature Rev. Gastroenterol. Hepatol. 7, 503–514 (2010).

    Article  Google Scholar 

  69. Quigley, E. M. Probiotics in functional gastrointestinal disorders: what are the facts? Curr. Opin. Pharmacol. 8, 704–708 (2008).

    Article  CAS  PubMed  Google Scholar 

  70. Clarke, G., Cryan, J. F., Dinan, T. G. & Quigley, E. M. Review article: probiotics for the treatment of irritable bowel syndrome — focus on lactic acid bacteria. Aliment. Pharmacol. Ther. 35, 403–413 (2012).

    Article  CAS  PubMed  Google Scholar 

  71. Logan, A. C. & Katzman, M. Major depressive disorder: probiotics may be an adjuvant therapy. Med. Hypotheses 64, 533–538 (2005).

    Article  PubMed  Google Scholar 

  72. Rao, S., Srinivasjois, R. & Patole, S. Prebiotic supplementation in full-term neonates: a systematic review of randomized controlled trials. Arch. Pediatr. Adolesc. Med. 163, 755–764 (2009).

    Article  PubMed  Google Scholar 

  73. Messaoudi, M. et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 105, 755–764 (2011). One of the first human studies assessing the psychotropic-like effects of probiotics.

    Article  CAS  PubMed  Google Scholar 

  74. Arseneault-Breard, J. et al. Combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces post-myocardial infarction depression symptoms and restores intestinal permeability in a rat model. Br. J. Nutr. 107, 1793–1799 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Bravo, J. A. 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. USA 108, 16050–16055 (2011). An important study demonstrating the ability of a potential probiotic to modify the stress response, behaviours relevant to anxiety, depression and cognition and alter central levels of GABA receptors. Moreover, it demonstrates that these effects are dependent on the vagus nerve.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Bercik, P. et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterol. Motil. 23, 1132–1139 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ma, X. et al. Lactobacillus reuteri ingestion prevents hyperexcitability of colonic DRG neurons induced by noxious stimuli. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G868–G875 (2009).

    Article  CAS  PubMed  Google Scholar 

  78. Kunze, W. A. et al. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. J. Cell. Mol. Med. 13, 2261–2270 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Tanida, M. et al. Effects of intraduodenal injection of Lactobacillus johnsonii La1 on renal sympathetic nerve activity and blood pressure in urethane-anesthetized rats. Neurosci. Lett. 389, 109–114 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Maes, M. et al. Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med. 10, 66 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Desbonnet, L. et al. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience 170, 1179–1188 (2010).

    Article  CAS  PubMed  Google Scholar 

  82. Desbonnet, L., Garrett, L., Clarke, G., Bienenstock, J. & Dinan, T. G. The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J. Psychiatr. Res. 43, 164–174 (2008).

    Article  PubMed  Google Scholar 

  83. Wall, R. et al. 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 (2012).

    Article  CAS  PubMed  Google Scholar 

  84. Innis, S. M. Dietary (n-3) fatty acids and brain development. J. Nutr. 137, 855–859 (2007).

    Article  CAS  PubMed  Google Scholar 

  85. Rapoport, S. I. Brain arachidonic and docosahexaenoic acid cascades are selectively altered by drugs, diet and disease. Prostaglandins Leukot. Essent. Fatty Acids 79, 153–156 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Luchtman, D. W. & Song, C. Cognitive enhancement by omega-3 fatty acids from child-hood to old age: findings from animal and clinical studies. Neuropharmacology 27 Jul 2012 (doi:10.1016/j.neuropharm.2012.07.019).

  87. Tillisch, K. et al. Modulation of the brain–gut axis after 4-week intervention with a probiotic fermented dairy product. Gastroenterology 142, S-115 (2012).

    Article  Google Scholar 

  88. Craig, A. D. How do you feel — now? The anterior insula and human awareness. Nature Rev. Neurosci. 10, 59–70 (2009).

    Article  CAS  Google Scholar 

  89. Paulus, M. P. & Stein, M. B. An insular view of anxiety. Biol. Psychiatry 60, 383–387 (2006).

    Article  PubMed  Google Scholar 

  90. Verdu, E. F. et al. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut 55, 182–190 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Larauche, M., Mulak, A. & Tache, Y. Stress and visceral pain: from animal models to clinical therapies. Exp. Neurol. 233, 49–67 (2012).

    Article  PubMed  Google Scholar 

  92. Mayer, E. A. et al. Functional GI disorders: from animal models to drug development. Gut 57, 384–404 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Mertz, H. et al. Regional cerebral activation in irritable bowel syndrome and control subjects with painful and nonpainful rectal distention. Gastroenterology 118, 842–848 (2000).

    Article  CAS  PubMed  Google Scholar 

  94. Gibney, S. M., Gosselin, R. D., Dinan, T. G. & Cryan, J. F. Colorectal distension-induced prefrontal cortex activation in the Wistar–Kyoto rat: implications for irritable bowel syndrome. Neuroscience 165, 675–683 (2010).

    Article  CAS  PubMed  Google Scholar 

  95. O'Mahony, C. M., Sweeney, F. F., Daly, E., Dinan, T. G. & Cryan, J. F. Restraint stress-induced brain activation patterns in two strains of mice differing in their anxiety behaviour. Behav. Brain Res. 213, 148–154 (2010).

    Article  PubMed  Google Scholar 

  96. Wang, Z. et al. Regional brain activation in conscious, nonrestrained rats in response to noxious visceral stimulation. Pain 138, 233–243 (2008).

    Article  PubMed  Google Scholar 

  97. Gareau, M. G., Jury, J., MacQueen, G., Sherman, P. M. & Perdue, M. H. Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut 56, 1522–1528 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. McKernan, D. P., Fitzgerald, P., Dinan, T. G. & Cryan, J. F. The probiotic Bifidobacterium infantis 35624 displays visceral antinociceptive effects in the rat. Neurogastroenterol. Motil. 22, 1029–1035 (2010).

    Article  CAS  PubMed  Google Scholar 

  99. Rousseaux, C. et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nature Med. 13, 35–37 (2007).

    Article  CAS  PubMed  Google Scholar 

  100. Ait-Belgnaoui, A. et al. Lactobacillus farciminis treatment suppresses stress induced visceral hypersensitivity: a possible action through interaction with epithelial cell cytoskeleton contraction. Gut 55, 1090–1094 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Johnson, A. C., Greenwood- Van Meerveld, B. & McRorie, J. Effects of Bifidobacterium infantis 35624 on post-inflammatory visceral hypersensitivity in the rat. Dig. Dis. Sci. 56, 3179–3186 (2011).

    Article  PubMed  Google Scholar 

  102. Wang, B. et al. Lactobacillus reuteri ingestion and IKCa channel blockade have similar effects on rat colon motility and myenteric neurones. Neurogastroenterol. Motil. 22, 98–107 (2010).

    Article  CAS  PubMed  Google Scholar 

  103. de Theije, C.G. et al. Pathways underlying the gut-to-brain connection in autism spectrum disorders as future targets for disease management. Eur. J. Pharmacol. 668, S70–S80 (2011).

    Article  CAS  PubMed  Google Scholar 

  104. Williams, B. L., Hornig, M., Parekh, T. & Lipkin, W. I. Application of novel PCR-based methods for detection, quantitation, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances. MBio 3, e00261–e00211 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Finegold, S. M. et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 16, 444–453 (2010).

    Article  CAS  PubMed  Google Scholar 

  106. Finegold, S. M. et al. Gastrointestinal microflora studies in late-onset autism. Clin. Infect. Dis. 35, S6–S16 (2002).

    Article  PubMed  Google Scholar 

  107. Parracho, H. M., Bingham, M. O., Gibson, G. R. & McCartney, A. L. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J. Med. Microbiol. 54, 987–991 (2005).

    Article  PubMed  Google Scholar 

  108. Adams, J. B., Johansen, L. J., Powell, L. D., Quig, D. & Rubin, R. A. Gastrointestinal flora and gastrointestinal status in children with autism — comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 11, 22 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  109. Wang, L. et al. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig. Dis. Sci. 57, 2096–2102 (2012).

    Article  CAS  PubMed  Google Scholar 

  110. Thomas, R. H. et al. The enteric bacterial metabolite propionic acid alters brain and plasma phospholipid molecular species: further development of a rodent model of autism spectrum disorders. J. Neuroinflamm. 9, 153 (2012).

    Article  CAS  Google Scholar 

  111. MacFabe, D. F., Cain, N. E., Boon, F., Ossenkopp, K. P. & Cain, D. P. Effects of the enteric bacterial metabolic product propionic acid on object-directed behavior, social behavior, cognition, and neuroinflammation in adolescent rats: relevance to autism spectrum disorder. Behav. Brain Res. 217, 47–54 (2011).

    Article  CAS  PubMed  Google Scholar 

  112. Sandler, R. H. et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J. Child Neurol. 15, 429–435 (2000).

    Article  CAS  PubMed  Google Scholar 

  113. Turnbaugh, P. J. & Gordon, J. I. The core gut microbiome, energy balance and obesity. J. Physiol. 587, 4153–4158 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Backhed, F., Manchester, J. K., Semenkovich, C. F. & Gordon, J. I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl Acad. Sci. USA 104, 979–984 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Morton, G. J., Cummings, D. E., Baskin, D. G., Barsh, G. S. & Schwartz, M. W. Central nervous system control of food intake and body weight. Nature 443, 289–295 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Schellekens, H., Finger, B. C., Dinan, T. G. & Cryan, J. F. Ghrelin signalling and obesity: at the interface of stress, mood and food reward. Pharmacol. Ther. 135, 316–326 (2012).

    Article  CAS  PubMed  Google Scholar 

  117. Manco, M. Gut microbiota and developmental programming of the brain: from evidence in behavioral endophenotypes to novel perspective in obesity. Front. Cell. Infect. Microbiol. 2, 109 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Davey, K. J. et al. Gender-dependent consequences of chronic olanzapine in the rat: effects on body weight, inflammatory, metabolic and microbiota parameters. Psychopharmacology 221, 155–169 (2012).

    Article  CAS  PubMed  Google Scholar 

  119. Berer, K. et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541 (2011).

    Article  CAS  PubMed  Google Scholar 

  120. Lee, Y. K., Menezes, J. S., Umesaki, Y. & Mazmanian, S. K. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 108, 4615–4622 (2011).

    Article  PubMed  Google Scholar 

  121. O'Toole, P. W. & Cooney, J. C. Probiotic bacteria influence the composition and function of the intestinal microbiota. Interdiscip. Perspect. Infect. Dis. 2008, 175285 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Forsythe, P. & Bienenstock, J. Immunomodulation by commensal and probiotic bacteria. Immunol. Invest. 39, 429–448 (2010).

    Article  CAS  PubMed  Google Scholar 

  123. Duerkop, B. A., Vaishnava, S. & Hooper, L. V. Immune responses to the microbiota at the intestinal mucosal surface. Immunity 31, 368–376 (2009).

    Article  CAS  PubMed  Google Scholar 

  124. Sternberg, E. M. Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nature Rev. Immunol. 6, 318–328 (2006).

    Article  CAS  Google Scholar 

  125. Dantzer, R., O'Connor, J. C., Freund, G. G., Johnson, R. W. & Kelley, K. W. From inflammation to sickness and depression: when the immune system subjugates the brain. Nature Rev. Neurosci. 9, 46–56 (2008).

    Article  CAS  Google Scholar 

  126. Wang, H. et al. Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation. Nature 421, 384–388 (2003).

    Article  CAS  PubMed  Google Scholar 

  127. Thayer, J. F. & Sternberg, E. M. Neural concomitants of immunity-focus on the vagus nerve. Neuroimage 47, 908–910 (2009).

    Article  PubMed  Google Scholar 

  128. de Lartigue, G., de La Serre, C. B. & Raybould, H. E. Vagal afferent neurons in high fat diet-induced obesity; intestinal microflora, gut inflammation and cholecystokinin. Physiol. Behav. 105, 100–105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Ruddick, J. P. et al. Tryptophan metabolism in the central nervous system: medical implications. Expert Rev. Mol. Med. 8, 1–27 (2006).

    Article  PubMed  Google Scholar 

  130. Clarke, G. et al. Tryptophan degradation in irritable bowel syndrome: evidence of indoleamine 2,3-dioxygenase activation in a male cohort. BMC Gastroenterol. 9, 6 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Nicholson, J. K. et al. Host–gut microbiota metabolic interactions. Science 336, 1262–1267 (2012).

    Article  CAS  PubMed  Google Scholar 

  132. Gundersen, B. B. & Blendy, J. A. Effects of the histone deacetylase inhibitor sodium butyrate in models of depression and anxiety. Neuropharmacology 57, 67–74 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: microbial endocrinology in the design and use of probiotics. Bioessays 33, 574–581 (2011).

    Article  CAS  PubMed  Google Scholar 

  134. Matur, E. & Eraslan, E. in New Advances in the Basic and Clinical Gastroenterology (ed. Brzozowski, T. ) (InTech, 2012).

    Google Scholar 

  135. Barrett, E., Ross, R. P., O'Toole, P. W., Fitzgerald, G. F. & Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 113, 411–417 (2012).

    Article  CAS  PubMed  Google Scholar 

  136. Forsythe, P. & Kunze, W. A. Voices from within: gut microbes and the CNS. Cell. Mol. Life Sci. 26 May 2012 (doI:10.1007/s00018-012-1028-z).

  137. Fanning, S. et al. Bifidobacterial surface-exopolysaccharide facilitates commensal–host interaction through immune modulation and pathogen protection. Proc. Natl Acad. Sci. USA 109, 2108–2113 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Julio-Pieper at Imágenes Ciencia for assistance with figures, and G. Clarke and L. Desbonnet for helpful comments on the paper. The Alimentary Pharmabiotic Centre is a research centre funded by Science Foundation Ireland (SFI), through the Irish Government's National Development Plan. The authors and their work were supported by SFI (grant numbers 02/CE/B124 and 07/CE/B1368).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John F. Cryan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

John F. Cryan's homepage

Timothy (Ted) G. Dinan's homepage

Human Microbiome Project

Glossary

Microbiota

The collection of microorganisms in a particular habitat, such as the microbiota of the skin or gut.

Stress response

The name given to the hormonal and metabolic changes that follow exposure to a threat. It involves the activation of the hypothalamus–pituitary–adrenal axis.

Microbiome

The collective genomes of all of the microorganisms in a microbiota.

Hypothalamus–pituitary–adrenal (HPA) axis

The HPA axis is the endocrine core of the stress system. Its activation results in the release of corticotropin-releasing factor from the hypothalamus, adrenocorticotropic hormone from the pituitary and cortisol (corticosterone in rats and mice) from the adrenal glands.

Maternal separation

A model of stress in early life. Isolation of pups from their mother in early life alters maternal behaviour upon being reunited and results in permanent changes in brain and behaviour in the offspring.

Probiotic

A living microorganism that, when ingested by humans or animals, can beneficially influence health.

Inflamm-ageing

A neologism to reflect the concept that ageing is accompanied by a global reduction in the capacity to cope with various stressors and a concomitant progressive increase in pro-inflammatory status.

Mono-association

The inoculation of germ-free animals with a specific bacterium.

Bacteriocins

Proteinaceous toxins produced by bacteria to inhibit the growth of similar or closely related bacterial strain(s).

Colonic AH neurons

The major intrinsic sensory neurons in the colon. They are termed AH owing to their common electrophysiological properties whereby action potentials are followed by prolonged and substantial after-hyperpolarizing (AH) potentials.

Dysbiosis

A microbial imbalance on or within the body, often localized to the gut.

Colorectal distension

A method for assessing visceral hypersensitivity. It is a noxious visceral stimulus that can be used in studies performed in animals and humans.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cryan, J., Dinan, T. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci 13, 701–712 (2012). https://doi.org/10.1038/nrn3346

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn3346

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing