The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health (2024)

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health (1)

Link to Publisher's site

Integr Med (Encinitas). 2018 Aug; 17(4): 28–32.

PMCID: PMC6469458

PMID: 31043907

Copyright and License information PMC Disclaimer

Abstract

The gut-brain axis is a bidirectional communication network that links the enteric and central nervous systems. This network is not only anatomical, but it extends to include endocrine, humoral, metabolic, and immune routes of communication as well. The autonomic nervous system, hypothalamic-pituitary-adrenal (HPA) axis, and nerves within the gastrointestinal tract, all link the gut and the brain, allowing the brain to influence intestinal activities, including activity of functional immune effector cells; and the gut to influence mood, cognition, and mental health.

The gut-brain axis is a bidirectional communication network that links the enteric and central nervous systems. This network is not only anatomical, but it extends to include endocrine, humoral, metabolic, and immune routes of communication as well. The autonomic nervous system, hypothalamic-pituitary-adrenal (HPA) axis, and nerves within the gastrointestinal (GI) tract, all link the gut and the brain, allowing the brain to influence intestinal activities, including activity of functional immune effector cells; and the gut to influence mood, cognition, and mental health.

Clinical, epidemiological, and immunological evidence suggest that enteric microbiota extensively and profoundly influences the gut-brain relationship (ie, mental state, emotional regulation, neuromuscular function, and regulation of the HPA). Research continues to elucidate mechanisms of action to explain the effects of microbiota, both directly and indirectly, on emotional and cognitive centres of the brain1 and has demonstrated that fluctuations of the microbiota are linked to changes within these systems of communication.2

For example, several mood disorders, such as anxiety, depression, and autism spectrum disorders now have well-established links to functional GI disruptions, whereas GI disease (eg, irritable bowel syndrome, irritable bowel disease) often involve psychological comorbidities associated with alteration of the gut microbiome.3-9 In addition, research has demonstrated that the composition of gut bacteria appears to be influential in fetal and neonatal neurologic development.10 And, not surprising, diet has also been shown to influence the gut microbiome’s impact on cognitive function.11

Pathways of the Gut-Brain Axis

As early as 1998, oral administration of a single, unique bacterium (Campylobacter jejuni) to rats in subclinical doses was found to lead to anxiety-like behavior, without an accompanying immune response.12 Later research confirmed that introduction C jejuni caused anxiety-like behavior in mice, with concomitant activation of neuronal regions in the brain that were dependent on information received from the gut via the vagus nerve.13

The seminal first studies establishing mechanisms of the gut-brain axis made use of animals raised in a sterile environment. Sudo et al14 sought to answer the question of whether postnatal microbial colonization could affect the development of brain plasticity and subsequent physiological response. To test the idea that gut microbes might affect the development of neural systems that govern the endocrine response to stress, they studied the HPA axis reaction to stress by comparing germ-free (GF), specific pathogen free (SPF) and gnotobiotic mice. They found that colonizing microbes altered the HPA response to restraint stress, indicating that the interaction of gut bacteria with the brain is also bidirectional, just like the brain-gut axis. This was the first report to show commensal microbes affecting the neural network responsible for controlling stress responsiveness. In this study, the HPA response of the GF mice was more sensitive to restraint stress than that of the SPF mice, whereas both groups of mice failed to show any difference in the sensitivity to ether stress. In addition, GF mice exhibited reduced brain-derived neurotrophic factor (BDNF) expression levels in the cortex and hippocampus relative to SPF mice. The authors concluded:

Because the HPA response to restraint stress is affected by the limbic system, including the prefrontal cortex, hippocampus and amygdala, and requires assembly and processing of signals from multiple sensory modalities before initiation of a stress response, whereas ether stress does not…, these results indicate that cognitive processing in the limbic system may be involved in an exaggerated HPA response by GF mice.

Their study showed that commensal microbiota are an environmental determinant that regulates the development of the HPA stress response, and that the series of events in the GI tract following postnatal microbial colonization can have a long-lasting influence on the neural processing of sensory information regarding the endocrine stress axis.

In the decade since this groundbreaking research, these findings have been corroborated and expanded. For example, administration of oral antimicrobials to SPF mice was shown to transiently alter the composition of the microbiota and increase exploratory behavior and hippocampal expression of brain-derived neurotropic factor (BDNF), a key protein involved in neuronal plasticity and cognition.15 These changes were independent of inflammatory activity, changes in levels of GI neurotransmitters, and vagal or sympathetic integrity, leading the authors to conclude that intestinal microbiota influences brain chemistry and behavior independently. Other related research has shown that administration of Lactobacillus rhamnosus beneficially altered brain expression levels of BDNF and of genes involved in serotonin signaling and metabolism in zebrafish.16

A great deal of research, well beyond the scope of this review, has further elucidated 4 major pathways of the gut-brain axis: neurologic, endocrine, humoral/metabolic, and immune.

Neurologic Pathway

The neurologic pathway includes the vagus nerve, the enteric nervous system, and the activity of neurotransmitters within the GI tract. Neurologic modulation of afferent sensory nerves directly produces molecules that can act as local neurotransmitters, such as GABA, serotonin, melatonin, histamine, and acetylcholine; this pathway also generates biologically active forms of catecholamines in the lumen of the gut.2 The autonomic nervous system also influences immune system activation in the gut, for example by directly modulating macrophage and mast cell responses to luminal bacteria. In addition, the gut microbiome appears to be essential for normal gut intrinsic primary afferent neuron excitability.17

Endocrine Pathway

Gut microbiota alters nutrient availability and thus influences the release of biologically active peptides from enteroendocrine cells, which in turn can affect the gut-brain axis. For example, the neuropeptide galanin is thought to be involved in many critical neurobiological functions including nociception, sleep/wake cycle regulation, feeding, mood, blood pressure regulation, parental behavior, and neurotrophic functions. Galanin stimulates the activity of the central branch of the HPA axis (influencing release of corticotropin-releasing factor and adrenocorticotropic hormone), and thus enhances glucocorticoid secretion from the adrenal cortex.18 In addition, it is able to directly stimulate cortisol secretion from the adrenal cortex, as well as norepinephrine release from the adrenal medulla, demonstrating its involvement in the HPA axis response to stress.19

Humoral/Metabolic Pathway

Bacterial metabolites (most importantly short-chain fatty acids [SCFAs], produced by the bacterial fermentation of dietary carbohydrates) are decisive humoral influencers. Best known to affect the nutrition of enterocytes, they also exert significant hormone-like activity, have immunomodulatory properties, and interact with nerve cells by stimulating the sympathetic branch of the autonomic nervous system. Furthermore, microbiota-derived SCFAs are able to cross the blood-brain barrier and have been shown to regulate microglia hom*oeostasis, which is required for proper brain development and brain tissue hom*oeostasis, and is involved in behavior modulation.20 Of important note, disruptions to SCFA metabolism have been implicated in the development of autism through the disruption of microglial communication and function.21-24 SCFAs also regulate the release of gut peptides from enteroendocrine cells and have been shown to regulate the synthesis of gut-derived serotonin from enterochromaffin cells, both of which in turn affect gut–brain hormonal communication.25 The gut provides approximately 95% of total body serotonin, most of which exists in plasma. Although serotonin has intrinsic roles in the intestines and peripheral metabolism, it is capable of locally activating afferent nerve endings that are connected directly to the central nerve system. 22 Although most physicians are familiar with the fluoxetine’s mechanism of blocking the transport of gut serotonin into plasma, many are unaware that elevated plasma serotonin has been observed in children with autism.26,27 An inverse correlation between high plasma serotonin and low serotonergic neurotransmission has been demonstrated in young male adults with autism spectrum disorder. 22

Another decisive bacterial metabolite is lipopolysaccharide (LPS), which is principally derived from the cell walls of Gram-negative enterobacteria. LPS gains entry to the systemic circulation via intestinal epithelial tight junction permeability defects, colloquially known as leaky gut syndrome. The human body produces antibodies against LPS, and their levels are known to be higher in patients with major depression than in controls.28

Immune Pathway

Inflammation metabolism within the GI tract is influenced by the gut microbiome, principally via the immune systems release of cytokines (such as interleukin [IL]-10 and IL-4) and other cellular communication mediators, such as interferon-gamma, during times of dysbiosis. In irritable bowel syndrome (IBS), as an example, abnormal microbiota populations activate mucosal innate immune responses, which increases gut epithelial permeability, activates gut pain sensory pathways, and dysregulates the enteric nervous system2,29,30; both brain-gut and gut-brain dysfunctions occur, the former being dominant.31 Disruptions in the gut-brain axis affect intestinal motility and secretion, contribute to visceral hypersensitivity and lead to cellular alterations of the entero-endocrine and immune systems.2

Epithelial Barrier Structure and Function

Stress is now well demonstrated to alter intestinal epithelial permeability, permitting bacterial antigens and LPS to enter circulation and become humoral influencers, with wide-ranging effects.32-36 In vivo experiments have shown that acute stress acts on the GI tract by inducing changes in colonocyte differentiation and decreased expression of mRNA encoding tight junction proteins.37

Intestinal permeability defects have been associated with a number of gut-related disorders, such as irritable bowel syndrome (IBD), necrotizing enterocolitis, and the low-level inflammation commonly seen in metabolic syndrome, obesity, and diabetes.38 So-called leaky gut is seen in some subtypes of IBS, such as that which commonly follows an infectious etiology,39 as well as in some forms of allergy.40 It should come as no surprise, then, that supplementation with probiotics could influence intestinal tight junction integrity, and thus support resolution of conditions caused or exacerbated by gut barrier dysfunction. Few clinical trials of probiotics have directly investigated epithelial barrier function in humans, but evidence is emerging in support of using lactobacilli for this purpose.41,42

For example, a multistrain combination of lactobacilli, lactococci, and bifidobacteria (Ecologic BARRIER, Winclove, Amsterdam, Netherlands) has demonstrated in vitro strengthening of the epithelial barrier after a pathogenic bacterial and inflammatory stressors, inhibition of mast-cell activation, stimulation of anti-inflammatory cytokine IL-10, and decreased LPS load.43 This preclinical work formed the basis for later clinical evidence of efficacy in preventing depression, discussed in the next section.

Inflammation, Depression, and the Microbiome

Studies from animal models conducted by independent research groups have corroborated findings of gut dysbiosis and its relation to monoamine disruptions seen in clinical depression, connecting gut microbiota with mood.44-48 In addition, intestinal permeability defects are thought to underlie the chronic low-grade inflammation observed in stress-related psychiatric disorders.49 Those with symptoms of depression frequently exhibit increased expression of proinflammatory cytokines, such as IL-1β, IL-6, tumor necrosis factor-α, as well as interferon gamma, and C-reactive protein.50-51,52 Gut microbiota influence transcription of these same cytokines, with dysbiosis triggering the so-called inflammasome pathway, whereas beneficial metabolites (SCFAs in particular) reduce production of proinflammatory cytokines, such as NF-κB.53

Gut microbiota are well known to support tight junction integrity between enterocytes. It should therefore come as no surprise that dysbiosis and associated increases in intestinal permeability are now recognized features of rheumatoid arthritis, Alzheimer’s disease, asthma, autism spectrum disorders, and other systemic conditions both inflammatory and otherwise. In recent years, there has been a tremendous amount research validating the mechanisms and role of the microbiome and probiotics in managing inflammatory conditions, particularly IBD.54-59

Depression is increasingly recognized as having an inflammatory component; indeed, anti-inflammatory drugs, such as COX-2 inhibitors, have previously demonstrated efficacy in major depression.60 Research has demonstrated that an inflammatory phenotype alters neurotransmitter metabolism by reducing the availability of neurotransmitter precursors and activating the HPA axis, all of which contribute to the pathogenesis of clinical depression.61,62 Although introduced as early as 1910,63 it has taken over a century for converging aspects of research to establish the gut-brain axis as a critical pathway to the effective prevention and treatment of clinical depression.64,65 Preclinical research laid the groundwork to investigate the use of probiotics for the treatment of mood disorders in humans.

Clinical Trials in Humans

A new class of probiotics, known as psychobiotics or psychomicrobiotics, has emerged in the last decade and is being fervently embraced by many health care practitioners as a nontoxic intervention for various psychiatric conditions.66,67 Several clinical trials have now documented effects, or lack thereof, of certain probiotics for depression and anxiety.

2002: In a pre- and postintervention assessment of adults suffering from stress or exhaustion (N = 34), a combination of L acidophilus, B bifidum and B longum improved subjects’ general condition by 40.7% after 6 months.68

2004: In a prospective, randomized, controlled, parallel study of healthy students (N = 136), researchers found no significant effects of L casei supplementation on anxiety levels, although beneficial alterations of lymphocyte and CD56 cell counts were observed.69

2007: In randomized, double-blind trial (N = 124), consumption of probiotic-containing yogurt had no effect on profile of mood states results, although there was improved self-reported mood of those whose mood was initially poor.70

2009: In a double-blind, randomized, placebo-controlled pilot study (N = 35, chronic fatigue syndrome patients), 2 months’ Anxiety Inventory supplementation with L casei significantly improved Beck scores. There was no effect on Beck depression inventory scores.71

2011: In subjects with reduced urinary free cortisol, consumption of the probiotics reduced anxiety and depression scores.72

2011: In a double-blind, randomized, controlled, parallel study (N = 55), consumption of L helveticus and B longum reduced somatization, depression, and anger-hostility as well as hospital anxiety and depression scale global scores; self-blame score on coping checklist and increased focus on problem solving, but there was no effect on perceived stress.73

2014: In a randomized, double-blind study (N = 36), L helveticus supplementation for 12 weeks had no significant effects on the perceived stress scale or geriatric depression scale; improvements were noted, however, on the digit span test, story recall test, verbal learning test, rapid visual information-processing, and stroop tasks scores.74

2015: In self-report questionnaires on fermented food consumption, neurosis, and social anxiety in young adults (N = 710), consumption of fermented foods containing probiotic bacteria was inversely associated with social anxiety and neurosis. Those at higher genetic risk for social anxiety disorder (indexed by higher tendency to neurosis) exhibited fewer social anxiety symptoms when they consumed more fermented foods.75

2015: The effects of a multistrain probiotic formula (Ecologic BARRIER, Winclove, Amsterdam) was tested in a randomized, triple-blind, placebo-controlled trial (N = 40 nonsmoking healthy young adults, mean age 20 y).76 The formula contained specific strains of B bifidum, B lactis, L acidophilus, L brevis, L casei, L salivarius, and L lactis at a dose of 5 billion colony-forming units (CFUs) per day. Consumption of this multispecies probiotic significantly reduced overall cognitive reactivity to depression, in particular aggressive and ruminative thoughts, as assessed by the Leiden index of depression sensitivity. This study is noteworthy because many patients, especially in young people with no prior history of depression, would prefer nonpharmaceutical interventions as a first-line treatment.77

2016: In randomized, double-blind study (N = 40), administration of a combination of L acidophilus, L casei, and Bifidobacterium bifidum for 8 weeks improved scores on the beck depression inventory.78

In a 2017 systematic review by Wallace and Milev of 10 clinical trials, most of the studies found positive results on measures of depressive symptoms.61 Because clinical trials on probiotics for depression and anxiety have been heterogeneous in terms of dosing, probiotic strain selection, and length of treatment, further randomized controlled clinical trials are warranted to validate the efficacy of this promising intervention.

Biography

Jeremy Appleton, ND, is a writer and speaker on topics in natural medicine and dietary supplements. An alumnus and former faculty member of the National University of Naturopathic Medicine, Dr Appleton is a licensed naturopathic physician, author, educator, and vice president of science and education for SFI USA, which manufactures dietary supplements under the Klaire Labs brand.

References

1. Carabotti M, Scirocco A, Maselli MA, Severi C.The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28(2):203-209. [PMC free article] [PubMed] [Google Scholar]

2. Mayer EA, Savidge T, Shulman RJ.Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology. 2014;146:1500-1512. [PMC free article] [PubMed] [Google Scholar]

3. Kennedy PJ, Clarke G, Quigley EM, Groeger JA, Dinan TG, Cryan JF.Gut memories: Towards a cognitive neurobiology of irritable bowel syndrome. Neurosci Biobehav Rev. 2012;36:310-340. [PubMed] [Google Scholar]

4. Mayer EA.Gut feelings: The emerging biology of gut-brain communication. Nat Rev Neurosci. 2011;12:453-466. [PMC free article] [PubMed] [Google Scholar]

5. Moloney RD, Desbonnet L, Clarke G, et al.The microbiome: stress, health and disease. Mamm Genome. 2014;25(1-2):49-74. [PubMed] [Google Scholar]

6. Cryan JF, Dinan TG.Mind-altering microorganisms: The impact of the gut microbiota on brain and Behavior. Nat Rev Neurosci. 2012;13:701-712. [PubMed] [Google Scholar]

7. Collins SM, Kassam Z, Bercik P.The adoptive transfer of behavioral phenotype via the intestinal microbiota: Experimental evidence and clinical implications. Curr Opin Microbiol. 2013;16:240-245. [PubMed] [Google Scholar]

8. Foster JA, McVey Neufeld KA.Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013;36:305-312. [PubMed] [Google Scholar]

9. Lyte M.Microbial endocrinology in the microbiome-gut-brain axis: How bacterial production and utilization of neurochemicals influence behavior. PLOS Pathog. 2013;9:e1003726. [PMC free article] [PubMed] [Google Scholar]

10. Douglas-Escobar M, Elliott E, Neu J.Effect of intestinal microbial ecology on the developing brain. JAMA Pediatr. 2013;167:374-379. [PubMed] [Google Scholar]

11. Noble EE, Hsu TM, Kanoski SE.Gut to brain dysbiosis: Mechanisms linking Western diet consumption, the microbiome, and cognitive impairment. Front Behav Neurosci. 2017;11-19. [PMC free article] [PubMed] [Google Scholar]

12. Lyte M, Varcoe JJ, Bailey MT.Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiol Behav. 1998;65:63-68. [PubMed] [Google Scholar]

13. Goehler LE, Gaykema RP, Opitz N, Reddaway R, Badr N, Lyte M.Activation in vagal afferents and central autonomic pathways: Early responses to intestinal infection with Campylobacter jejuni. Brain Behav Immun. 2005;19:334-344. [PubMed] [Google Scholar]

14. Sudo N, Chida Y, Aiba Y, et al.Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558(Pt 1):263-275. [PMC free article] [PubMed] [Google Scholar]

15. Bercik P, Denou E, Collins J, et al.The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141(2):599-609. [PubMed] [Google Scholar]

16. Borrelli L, Aceto S, Agnisola C., et al.Probiotic modulation of the microbiota-gut-brain axis and behaviour in zebrafish. Sci Rep. 2016;6:30046. [PMC free article] [PubMed] [Google Scholar]

17. McVey Neufeld KA, Mao YK, Bienenstock J, Foster JA, Kunze WA.The microbiome is essential for normal gut intrinsic primary afferent neuron excitability in the mouse. Neurogastroenterol Motil. 2013;25(2):183-188. [PubMed] [Google Scholar]

18. Azzam I, Gilad S, Limor R, Stern N, Greenman Y.Ghrelin stimulation by hypothalamic-pituitary-adrenal axis activation depends on increasing cortisol levels. Endocr Connect. 2017;6(8):847-855. [PMC free article] [PubMed] [Google Scholar]

19. Piccioto MR.Galanin: 25 years with a multitalented neuropeptide. Cell Mol Life Sci2008;65(12):1872-1879. [PMC free article] [PubMed] [Google Scholar]

20. Mayer EA, Tillisch K, Gupta A.Gut/brain axis and the microbiota. J Clin Invest. 2015; 125(3):926-938. [PMC free article] [PubMed] [Google Scholar]

21. Rogers GB, Keating DJ, Young RL, et al.From gut dysbiosis to altered brain function and mental illness: Mechanisms and pathways. Mol Psychiatry. 2016;21(6):738-748. [PMC free article] [PubMed] [Google Scholar]

22. MacFabe D.Autism: Metabolism, mitochondria, and the microbiome. Glob Adv Health Med. 2013;2(6):52-66. [PMC free article] [PubMed] [Google Scholar]

23. Frye RE, Rose S, Chacko J, et al.Modulation of mitochondrial function by the microbiome metabolite propionic 2. Transl Psychiatry. 2016;6(10):e927. [PMC free article] [PubMed] [Google Scholar]

24. MacFabe D.Enteric short-chain fatty acids: Microbial messengers of metabolism, mitochondria, and mind: implications in autism spectrum disorders. Microb Ecol Health Dis. 2015;26:10. [PMC free article] [PubMed] [Google Scholar]

25. Wang Y, Kasper LH.The role of microbiome in central nervous system disorders. Brain Behav Immun. 2014; 38:1-12. [PMC free article] [PubMed] [Google Scholar]

26. Marler S, Ferguson BJ, Lee EB, et al.Brief report: Whole blood serotonin levels and gastrointestinal symptoms in autism spectrum disorder. J Autism Dev Disord. 2016;46(3):1124-1130. [PMC free article] [PubMed] [Google Scholar]

27. Gabriele S, Sacco R, Persico AM.Blood serotonin levels in autism spectrum disorder: A systematic review and meta-analysis. Eur Neuropsychopharmacol. 2014;24(6):1. [PubMed] [Google Scholar]

28. Maes M, Kubera M, Leunis JC.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(1):117-124. [PubMed] [Google Scholar]

29. Dupont HL.Review article: Evidence for the role of gut microbiota in irritable bowel syndrome and its potential influence on therapeutic targets. Aliment Pharmacol Ther. 2014;39:1033-1042. [PubMed] [Google Scholar]

30. Kennedy PJ, Cryan JF, Dinan TG, Clarke G.Irritable bowel syndrome: A microbiome-gut-brain axis disorder?World J Gastroenterol2014;20:14105-14125. [PMC free article] [PubMed] [Google Scholar]

31. Koloski NA, Jones M, Kalantar J, et al.The brain-gut pathway in functional gastrointestinal disorders is bidirectional: A 12-year prospective population-based study. Gut. 2012;61:1284-1290. [PubMed] [Google Scholar]

32. Kiliaan AJ, Saunders PR, Bijlsma PB, et al.Stress stimulates transepithelial macromolecular uptake in rat jejunum. Am J Physiol. 1998;275:G1037-G1044. [PubMed] [Google Scholar]

33. Groot J, Bijlsma P, Van Kalkeren A, Kiliaan A, Saunders P, Perdue M.Stress-induced decrease of the intestinal barrier function. The role of muscarinic receptor activation. Ann NY Acad Sci. 2000;915:237-246. [PubMed] [Google Scholar]

34. Yates DA, Santos J, Soderholm JD, Perdue MH.Adaptation of stress-induced mucosal pathophysiology in rat colon involves opioid pathways. Am J Physiol Gastrointest Liver Physiol. 2001;281:G124-G128. [PubMed] [Google Scholar]

35. Soderholm JD, Yates DA, Gareau MG, Yang PC, MacQueen G, Perdue MH.Neonatal maternal separation predisposes adult rats to colonic barrier dysfunction in response to mild stress. Am J Physiol Gastrointest Liver Physiol. 2002. 283:G1257-G1263. [PubMed] [Google Scholar]

36. Jacob C, Yang PC, Darmoul D, et al.Mast cell tryptase controls paracellular permeability of the intestine: Role of protease-activated receptor 2 and beta-arrestins. J Biol Chem. 2005;280:31936-31948. [PubMed] [Google Scholar]

37. Demaude J, Salvador-Cartier C, Fioramonti J, Ferrier L, Bueno L.Phenotypic changes in colonocytes following acute stress or activation of mast cells in mice: Implications for delayed epithelial barrier dysfunction. Gut. 2006;55:655-661. [PMC free article] [PubMed] [Google Scholar]

38. Bron PA, Kleerebezem M, Brummer R-J, Cani PD.Can probiotics modulate human disease by impacting intestinal barrier function?Br J Nutr. 2017;117(1):93-107. [PMC free article] [PubMed] [Google Scholar]

39. Spiller RC, Jenkins D, Thornley JP, et al.Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post-dysenteric irritable bowel syndrome. Gut. 2000;47(6):804-811. [PMC free article] [PubMed] [Google Scholar]

40. Panzer AR, Lynch SV.Influence and effect of the human microbiome in allergy and asthma. Curr Opin Rheumatol. 2015;27(4):373-380. [PubMed] [Google Scholar]

41. Karczewski J, Troost FJ, Konings I, et al.Regulation of human epithelial tight junction proteins by Lactobacillus plantarum in vivo and protective effects on the epithelial barrier. Am J Physiol Gastrointest Liver Physiol. 2010;298(6):G851-G859. [PubMed] [Google Scholar]

42. Gotteland M, Cruchet S, Verbeke S.Effect of Lactobacillus ingestion on the gastrointestinal mucosal barrier alterations induced by indometacin in humans. Aliment Pharmacol Ther. 2001; 15(1):11-17. [PubMed] [Google Scholar]

43. Van Hemert S, Ormel G.Influence of the Multispecies Probiotic Ecologic BARRIER on parameters of intestinal barrier function. Food Nutr Sci. 2014;5:1739-1745. [Google Scholar]

44. Diaz Heijtz R, Wang S, Anuar F, et al.Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108:3047-3052. [PMC free article] [PubMed] [Google Scholar]

45. Neufeld KA, Kang N, Bienenstock J, Foster JA.Effects of intestinal microbiota on anxiety-like behavior. Commun Integr Biol. 2011;4:492-494. [PMC free article] [PubMed] [Google Scholar]

46. Clarke G, Grenham S, Scully P, et al.The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18:666-673 [PubMed] [Google Scholar]

47. Yirmiya R.Behavioral and psychological effects of immune activation: Implications for ‘depression due to a general medical condition. Curr Opin Psychiatry. 1997;10(6):470-476. [Google Scholar]

48. Anisman H, Ravindran A, Griffiths J, et al.Endocrine and cytokine correlates of major depression and dysthymia with typical or atypical. Mol Psychiatry. 1999;4:182-188. [PubMed] [Google Scholar]

49. Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP.Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci. 2015;9:392. [PMC free article] [PubMed] [Google Scholar]

50. Howren MB, Lamkin DM, Suls J.Associations of depression with C-reactive protein, IL-1, and IL-6: A meta-analysis. Psychosom Med. 2009;71(2):171-186. [PubMed] [Google Scholar]

51. Owen BM, Eccleston D, Ferrier IN, et al.Raised levels of plasma interleukin-1β in major and postviral depression. Acta Psychiatr Scand. 2001;103(3):226-228. [PubMed] [Google Scholar]

52. Maes M, Scharpé S, Meltzer HY, et al.Increased neopterin and interferon-gamma secretion and lower availability of l-tryptophan in major depression: further evidence for an immune response. Psychiatry Res. 1994;54(2):143-160. [PubMed] [Google Scholar]

53. van den Elsen LW, Poyntz HC, Weyrich LS, Young W, Forbes-Blom EE.Embracing the gut microbiota: the new frontier for inflammatory and infectious diseases. Clin Transl Immunol. 2017;6(1):e125. [PMC free article] [PubMed] [Google Scholar]

54. Ahmed I, Roy BC, Khan SA, Umar S.Microbiome, metabolome and inflammatory bowel disease. Microorganisms. 2016;4(2):1. [PMC free article] [PubMed] [Google Scholar]

55. Dong J, Teng G, Wei T, Gao W, Wang H.Methodological quality assessment of meta-analyses and systematic reviews of probiotics in inflammatory bowel disease and pouchitis. PLoS One. 2016;11(12):e0168785. [PMC free article] [PubMed] [Google Scholar]

56. Gong D, Gong X, Wang L, Yu X, Dong Q.Involvement of reduced microbial diversity in inflammatory bowel disease. Gastroenterol Res Pract. 2016;2016:6951091. [PMC free article] [PubMed] [Google Scholar]

57. Plaza-Díaz J, Ruiz-Ojeda FJ, Vilchez-Padial LM, Gil A.Evidence of the anti-inflammatory effects of probiotics and synbiotics in intestinal chronic diseases. Nutrients. 2017;9(6):1. [PMC free article] [PubMed] [Google Scholar]

58. Souza DG, Vieira AT, Soares AC, et al.The essential role of the intestinal microbiota in facilitating acute inflammatory responses. J Immunol. 2004;173(6):4137-4146. [PubMed] [Google Scholar]

59. Hörmannsperger G, Haller D.Molecular crosstalk of probiotic bacteria with the intestinal immune system: Clinical relevance in the context of inflammatory bowel disease. Int J Med Microbiol. 2010;300(1):63-73. [PubMed] [Google Scholar]

60. Müller N, Schwartz MJ, Douhe A, et al.The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: Results of a double-blind, randomized, placebo controlled, add-on pilot study to reboxetine. Molecular Psych. 2006;11:680-684. [PubMed] [Google Scholar]

61. Wallace CJK, Milev R.The effects of probiotics on depressive symptoms in humans: A systematic review. Ann Gen Psychiatry. 2017;16:14. [PMC free article] [PubMed] [Google Scholar]

62. Clarke G, Stilling RM, Kennedy PJ, et al.Minireview: Gut microbiota: The neglected endocrine organ. Mol Endocrinol. 2014;28(8):1221-1238. [PMC free article] [PubMed] [Google Scholar]

63. Phillips JGP.The treatment of melancholia by the lactic acid bacillus. Br J Psychiatry. 1910;56:422-431. [Google Scholar]

64. Dinan TG, Cryan JF.Melancholic microbes: A link between gut microbiota and depression?Neurogastroenterol Motil. 2013;25(9):713-719. [PubMed] [Google Scholar]

65. Evrensel A, Ceylan ME.Gut-brain axis: The role of gut microbiota in the psychiatric disorders. Curr Approach Psychiatry. 2015;7:461-472. [Google Scholar]

66. Fond G, Boukouaci W, Chevalier G, et al.The “psychomicrobiotic”: Targeting microbiota in major psychiatric disorders: A systematic review. Pathol Biol (Paris). 2015;63:35-42. [PubMed] [Google Scholar]

67. Evrensel A, Ceylan ME.The gut-brain axis: The missing link in depression. Clin Psychopharmacol Neurosci. 2015;13(3):239-244. [PMC free article] [PubMed] [Google Scholar]

68. Gruenwald J, Graubaum HJ, Harde A.Effect of a probiotic multivitamin compound on stress and exhaustion. Adv Ther. 2002;19(3):141-150 [PubMed] [Google Scholar]

69. Marcos A, Wärnberg J, Nova E, et al.The effect of milk fermented by yogurt cultures plus Lactobacillus casei DN-114001 on the immune response of subjects under academic examination stress. Eur J Nutr. 2004;43(6):381-389. [PubMed] [Google Scholar]

70. Benton D, Williams C, Brown A.Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur J Clin Nutr. 2007;61(3):355-361. [PubMed] [Google Scholar]

71. Rao AV, Bested AC, Beaulne TM, et al.A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathogens. 2009;1(1):1. [PMC free article] [PubMed] [Google Scholar]

72. Messaoudi M, Lalonde R, Violle N, et al.Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr. 2011;105(05):755-764 [PubMed] [Google Scholar]

73. Messaoudi M, Violle N, Bisson JF, et al.Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut Microbes. 2011;2(4):256-261. [PubMed] [Google Scholar]

74. Chung YC, Jin HM, Cui Y, et al.Fermented milk of Lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J Funct Foods. 2014;10:465-474. [Google Scholar]

75. Hilimire MR, DeVylder JE, Forestell CA.Fermented foods, neuroticism, and social anxiety: An interaction model. Psychiatry Res. 2015;228(2):203-208. [PubMed] [Google Scholar]

76. Steenbergen L, Sellaro R, van Hemert S, et al.A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav Immun. 2015;48:258-264. [PubMed] [Google Scholar]

77. Hughes-Morley A, Young B, Waheed W, Small N, Bower P.Factors affecting recruitment into depression trials: Systematic review, meta-synthesis and conceptual framework. J Affect Disorders2015;172:274-290. [PubMed] [Google Scholar]

78. Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, et al.Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition. 2016;32(3):315-320. [PubMed] [Google Scholar]

Articles from Integrative Medicine: A Clinician's Journal are provided here courtesy of InnoVision Media

The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health (2024)
Top Articles
Latest Posts
Article information

Author: Arline Emard IV

Last Updated:

Views: 5502

Rating: 4.1 / 5 (72 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Arline Emard IV

Birthday: 1996-07-10

Address: 8912 Hintz Shore, West Louie, AZ 69363-0747

Phone: +13454700762376

Job: Administration Technician

Hobby: Paintball, Horseback riding, Cycling, Running, Macrame, Playing musical instruments, Soapmaking

Introduction: My name is Arline Emard IV, I am a cheerful, gorgeous, colorful, joyous, excited, super, inquisitive person who loves writing and wants to share my knowledge and understanding with you.