Gut-brain Axis
10th Jul, 2019

This post is specifically for health practitioners. Create a free account

Alternatively, view general public info.

Learn more about vital.ly

Gut-Brain Axis Essentials

Gut-brain Axis

The gut microbiota consists of a diverse community of bacterial species in the gastrointestinal (GI) tract which exists symbiotically with the human host. A healthy and stable gut microbiota community plays a vital role in maintaining motility, permeability, secretion and immunity of the gut. In recent years, the gut microbiome has also been associated with many psychiatric and neurodevelopmental disorders via a system known as the gut-brain axis (Table 1).

Table 1. Conditions influenced by the gut-brain axis (1,2,3,4)

Gut-Brain Axis Essentials Table 1The gut-brain axis incorporates the central nervous system (CNS), autonomic nervous system (ANS) and enteric nervous system (ENS) in addition to the neuroendocrine and the neuroimmune systems.

The gut microbiome communicates via multiple pathways, including (1,4):

  • vagus nerve
  • neurotransmitters (e.g. serotonin)
  • immune system
  • hormonal system
  • bacterial metabolites such as short-chain fatty acids (SCFAs)

 

Figure 1. A systems model of brain-gut-microbiome interactions (5) CC BY-NC-ND 4.0

 Gut-Brain Axis Essentials Fig 1

The communication between the CNS and the GI tract is bidirectional and is responsible for the modulation of digestive processes, immune function and responses to visceral stimuli. Disturbance of these pathways results in disruption to neurotransmitter balance, increases in chronic inflammation or exacerbated hypothalamic-pituitary-adrenal (HPA) axis activity (2,6).

The trigger that causes blood-brain barrier leakage, immune cell activation and inflammation, and ultimately neuroinflammation in the CNS is believed to be due to disturbance of the microbiome-gut-axis (2,4). An imbalance of the gut microbiota results in dysbiosis and disruption of the gut microbiota homeostasis. Signalling between the gut-brain axis is also impacted potentially leading to neurological and neuroimmune abnormalities. Dysbiosis is commonly associated with a compromised gut epithelium, and the subsequent bacterial translocation (‘leaky-gut’) may result in innate immune activation and systemic inflammation which can promote neuroinflammation across the blood-brain barrier (7).

 

Figure 2. Key communication pathways of the microbiota-gut-brain axis (8) CC BY-NC-ND 4.0

Gut-Brain Axis Essentials Fig 2

 

Traditional Understanding

Historically, the microbiota was known to consist of commensal bacteria that do not harm the host nor necessarily impart any significant benefit. The exception was the knowledge based on observations that commensal bacteria compete with pathogenic species for nutrients and sites for colonization and, therefore, a compromised gut microbiota can lead to pathogenic intestinal infection (7). More recently the microbiome has been associated with gut functions and gut barrier integrity. Neurological and psychological conditions were assumed to be related to dysfunction in brain and neurotransmitter processes. The revelation of the bidirectional communication between the brain and the gut microbiome is helping to solve many unanswered questions about the aetiology and the possible treatment of both neurological, psychological and GIT conditions.

 

Latest Research

Recent studies have shown that the gut microbiota is involved in neurodevelopment and diverse brain functions by regulating the gut-brain axis. GI symptoms, such as constipation, diarrhoea, and abdominal pain are common comorbidities in many neurological diseases. In addition, dysregulation in the composition of gut microbiota (gut dysbiosis) is present in a variety of neurological diseases. Therefore, the importance of maintaining a healthy microbiota community (gut symbiosis) in the regulation of the gut-brain axis is of utmost importance. For example, certain species of Bifidobacterium, which are decreased in Alzheimer’s Disease (AD), are associated with anti-inflammatory properties and decreased intestinal permeability. Similarly, gram-negative intestinal bacteria of Bacteroides, which are increased in patients with AD, may result in increased translocation of lipopolysaccharides (LPS) from the gut to the systemic circulation, which in turn may contribute to or exacerbate AD pathology through neuroinflammation (9).

Bacteria have been shown to produce and/or consume a wide range of neurotransmitters, including dopamine, norepinephrine, serotonin and or gamma-aminobutyric acid (GABA). Accumulating preclinical evidence suggests that manipulation of these neurotransmitters by bacteria may have an impact in host physiology, and preliminary human studies are showing that microbiota-based interventions can also alter neurotransmitter levels (10).

The term microbiota-gut-brain axis has been introduced to highlight the role of the microbiota in the gut-brain axis (11).

 

Parkinson's Disease

Growing evidence shows that alterations in gut microbiota composition and microbial metabolites may be involved in the pathogenesis and clinical phenotype of Parkinson's disease (PD) (12,13).

  • PD is a neurodegenerative disorder that affects predominately dopamine-producing (‘dopaminergic’) neurons. Dopamine synthesis in the brain is induced by dopamine-producing enzymes that are controlled by gut microbiota (14).
  • The prevalence of small intestinal bacterial overgrowth (SIBO) is 25-54% in PD patients compared to 8-20% in normal controls and has been associated with a greater degree of impairment in motor control in PD (15).
  • Early involvement of GI dysfunction is considered a possible pre-symptomatic stage of PD making the gut microbiota a promising source of information for diagnosis, prognosis, and, potentially, pathogenesis (4,5). The risk of PD development increases with infrequency of bowel movement and constipation severity, and there is a significant comorbidity of PD and IBS-like symptoms (16).
  • The manipulation of gut microbiota and microbial metabolites may have a positive impact on PD patients through a reduction in non-motor symptoms and potential stemming of classical motor symptoms. However, how the microbiota directly or indirectly exerts effects on brain neurons or immune cells, such as microglia and astrocytes in PD, has yet to be uncovered (4).
  • To date, little is known about the molecular causes of altered gut homeostasis in PD. A reduction in the level of neurotrophins, including brain-derived neurotrophic factor (BDNF), may contribute to deregulated gut homeostasis and constipation in PD (7).

 

Figure 3. Possible pathways involved in PD pathogenesis (17) CC BY 4.0

Gut-Brain Axis Essentials Fig 3

 

Alzheimer's Disease

Recent evidence suggests that alterations in the microbiome of those with Alzheimer’s disease (AD) may be associated with or contribute to the pathophysiology of AD (18,19,20).

  • An association between an altered microbiome and the presentation of AD has been demonstrated in humans. The gut microbiome of AD cases contained less microbial diversity and was compositionally distinct from age-and sex-matched controls (9).
  • A significant reduction in BDNF and of neuroprotective signalling is observed in post-mortem brain tissue as well as in vivo models of AD. This has been suggested to contribute to the overt and progressive neurodegeneration of hippocampal neurons (21). BDNF confers a neuroprotective role, and recent evidence shows that BDNF levels are regulated by the gut microbiome. A reduction in BDNF can also exacerbate oxidative stress and alter gut homeostasis in AD cases (7).
  • The progression and presentation of AD may be modified through the modification of the microbiota. It has been suggested that AD pathology commences as early as 20 years prior to the manifestation of overt symptoms, therefore modifying AD progression after symptoms are manifest may be a challenge. It is possible that the greatest opportunities in modifying the microbiota will be in the proactive prevention of AD for those with a hereditary predisposition (7).
  • Gut bacteria synthesize and release amyloid peptides and lipopolysaccharides, which in turn contribute to the regulation of signalling pathways and activate inflammatory signalling through the release of cytokines, with potential effects on the pathophysiological cascade of AD (22).
  • An imbalance of the gut microbiota is closely related to other factors involved in the pathogenesis of AD, such as obesity and type 2 diabetes (23).

Although promising, this is an emerging field in neuroscience and the available evidence requires confirmation by further rigorous studies (22).

 

Figure 4. Gut microbiota dysbiosis and Alzheimer’s disease. Mechanism and potential therapeutic interventions. Adapted from (24) CC BY 4.0

Gut-Brain Axis Essentials Fig 4 

 

Autism Spectrum Disorder

As with other neurological disorders, such as PD and AD, GI dysfunction is among the most common medical co-morbidities associated with autism spectrum disorder (ASD), presenting with symptoms such as constipation, bloating and diarrhoea. Researchers have discovered that a gene mutation that affects neuron communication in the brain, the first identified as a cause of ASD, also causes dysfunction in the gut (25). A correlation has been found between GI dysfunction and the degree of social impairment in ASD. Studies have reported that ASD is often associated with an altered intestinal microbiome (26,27). Currently, however, there is no distinct microbiome profile characteristic of ASD or a specific profile that is associated with its neurobehavioral manifestations. Whether changes in the microbiome are caused by GI dysfunction or whether changes in the microbiome cause GI problems is still unclear (28). It should be noted that children with ASD are also characterized by alterations in the oral microbiome, raising the possibility that more than just the gut microbiota may be involved (29).

Alterations of the gut microbiome might promote the symptoms of ASD through microbial metabolites, immune regulation, HPA axis modulation and direct interaction with the brain (30).

  • ASD-associated alterations in eating behaviour and diet are likely to play a role in altered intestinal microbiome composition (31)
  • Compared with ASD-unaffected children, children with ASD and GI dysfunction have increased mucosal tissue levels of select microbial taxa, predominantly Clostridium, that correlate with cytokine and tryptophan homeostasis (32)
  • A recent clinical trial investigating the potential benefits of a modified faecal transplantation therapy in children with ASD resulted in a significant improvement in GI symptoms (80% reduction) as well as behavioural symptoms, which were maintained at 8 weeks post-treatment. The treatment combined antibiotics, a bowel cleanse, a stomach-acid suppressant and FMT (33). A follow-up 2 years later found that GI improvements and increased gut microbiome diversity had been maintained. ASD related symptoms improved even more after the end of treatment. The proportion of children with severe symptoms dropped from 83% at baseline to 17% at the two-year follow-up, while 44% had dropped below the diagnostic cut-off score (34)
  • Accumulating evidence demonstrates the beneficial effects of probiotics treatment in the improvement of comorbid symptoms seen in ASD (30). Research also suggests that the administration of probiotics might have a preventive effect on ASD. In a study of Lactobacillus rhamnosus given at early postnatal times and followed for 13 years, the group receiving no probiotics was more affected by Asperger syndrome and attention-deficit hyperactivity disorder than the group receiving the probiotics, which was not prone to develop those disorders (35). However, the detailed roles of the gut microbiome in the pathology of ASD are still unclear
  • Many epidemiological, clinical, and in vivo studies have shown that maternal infections primarily influence the compositional and functional changes of gut microbiota in the offspring and are associated with the development of ASD symptoms. Evidence indicates that the maternal diet and inflammatory events result in systemic products such as cytokines and serotonin that alter CNS neurodevelopment of regions critical to the cognition of social interaction. Select intestinal commensal microbiota can induce or attenuate production of these molecules, therefore the intestinal mucosa and microbiome may be a modifier of ASD occurrence (36)
  • The gut microbiota of infants in the first 2-3 years of life is associated with brain development and brain function in early life (37). The microbial composition of the human gut at 1 year of age predicts cognitive performance at 2 years of age, particularly in the area of communicative behaviour. Results may have implications for developmental disorders characterized by cognitive or language delay (38)

 

Figure 5. The potential mechanisms by which the gut microbiome is involved in ASD (31)

Gut-Brain Axis Essentials Fig 5

Other Conditions

Recent research in other CNS conditions associated with the gut-brain axis is summarised in Table 2.

 

Table 2. Other conditions influenced by the gut-brain axis

Gut-Brain Axis Essentials Table 2

 

Loading...
References
1Ming X, Chen N, Ray C, Brewer G, Kornitzer J, Steer RA. A Gut Feeling: A Hypothesis of the Role of the Microbiome in Attention-Deficit/Hyperactivity Disorders. Child Neurol Open. 2018;5:2329048X18786799.
2Nair AT, Ramachandran V, Joghee NM, Antony S, Ramalingam G. Gut Microbiota Dysfunction as Reliable Non-invasive Early Diagnostic Biomarkers in the Pathophysiology of Parkinson’s Disease: A Critical Review. J Neurogastroenterol Motil. 2018 Jan 30;24(1):30–42.
3Sarkar A, Harty S, Lehto SM, Moeller AH, Dinan TG, Dunbar RIM, et al. The Microbiome in Psychology and Cognitive Neuroscience. Trends in Cognitive Sciences. 2018 Jul;22(7):611–36.
4Sun M-F, Shen Y-Q. Dysbiosis of gut microbiota and microbial metabolites in Parkinson’s Disease. Ageing Research Reviews. 2018 Aug;45:53–61.
5Martin CR, Osadchiy V, Kalani A, Mayer EA. The Brain-Gut-Microbiome Axis. Cellular and Molecular Gastroenterology and Hepatology. 2018;6(2):133–48.
6Bruce-Keller AJ, Salbaum JM, Berthoud H-R. Harnessing Gut Microbes for Mental Health: Getting From Here to There. Biological Psychiatry. 2018 Feb;83(3):214–23.
7Lombardi VC, De Meirleir KL, Subramanian K, Nourani SM, Dagda RK, Delaney SL, et al. Nutritional modulation of the intestinal microbiota; future opportunities for the prevention and treatment of neuroimmune and neuroinflammatory disease. The Journal of Nutritional Biochemistry. 2018 Nov;61:1–16.
8Foster JA, Rinaman L, Cryan JF. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiology of Stress. 2017 Dec;7:124–36.
9Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017 Oct 19;7(1):13537.
10Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Research. 2018 Aug;1693:128–33.
11Kim N, Yun M, Oh YJ, Choi H-J. Mind-altering with the gut: Modulation of the gut-brain axis with probiotics. J Microbiol. 2018 Mar;56(3):172–82.
12Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, Pekkonen E, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 2015 Mar;30(3):350–8.
13Unger MM, Spiegel J, Dillmann K-U, Grundmann D, Philippeit H, Bürmann J, et al. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism & Related Disorders. 2016 Nov;32:66–72.
14Thakur AK, Shakya A, Mohammed G, Emerald M, Kumar V. Gut-Microbiota and Mental Health: Current and Future Perspectives. 2014;15.
15Tan AH, Mahadeva S, Thalha AM, Gibson PR, Kiew CK, Yeat CM, et al. Small intestinal bacterial overgrowth in Parkinson’s disease. Parkinsonism Relat Disord. 2014 May;20(5):535–40.
16Mertsalmi TH, Aho VTE, Pereira P a. B, Paulin L, Pekkonen E, Auvinen P, et al. More than constipation - bowel symptoms in Parkinson’s disease and their connection to gut microbiota. Eur J Neurol. 2017;24(11):1375–83.
17Perez-Pardo P, Hartog M, Garssen J, Kraneveld AD. Microbes Tickling Your Tummy: the Importance of the Gut-Brain Axis in Parkinson’s Disease. Curr Behav Neurosci Rep. 2017;4(4):361–8.
18Kobayashi Y, Sugahara H, Shimada K, Mitsuyama E, Kuhara T, Yasuoka A, et al. Therapeutic potential of Bifidobacterium breve strain A1 for preventing cognitive impairment in Alzheimer’s disease. Sci Rep. 2017 Oct 18;7.
19Köhler CA, Maes M, Slyepchenko A, Berk M, Solmi M, Lanctôt KL, et al. The Gut-Brain Axis, Including the Microbiome, Leaky Gut and Bacterial Translocation: Mechanisms and Pathophysiological Role in Alzheimer’s Disease. Curr Pharm Des. 2016;22(40):6152–66.
20Minter MR, Zhang C, Leone V, Ringus DL, Zhang X, Oyler-Castrillo P, et al. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease. Scientific Reports. 2016 Sep;6(1).
21Dagda RK, Banerjee TD. Role of PKA in regulating mitochondrial function and neuronal development: implications to neurodegenerative diseases. Rev Neurosci. 2015 Jun 1;26(3):359–70.
22de J.R. De-Paula V, Forlenza AS, Forlenza OV. Relevance of gutmicrobiota in cognition, behaviour and Alzheimer’s disease. Pharmacological Research. 2018 Oct;136:29–34.
23Kim Y-K, Shin C. The Microbiota-Gut-Brain Axis in Neuropsychiatric Disorders: Pathophysiological Mechanisms and Novel Treatments. Curr Neuropharmacol. 2018;16(5):559–73.
24Sochocka M, Donskow-Łysoniewska K, Diniz BS, Kurpas D, Brzozowska E, Leszek J. The Gut Microbiome Alterations and Inflammation-Driven Pathogenesis of Alzheimer’s Disease-a Critical Review. Mol Neurobiol. 2018 Jun 23;
25Hosie S, Ellis M, Swaminathan M, Ramalhosa F, Seger GO, Balasuriya GK, et al. Gastrointestinal dysfunction in patients and mice expressing the autism-associated R451C mutation in neuroligin-3. Autism Res. 2019 May 22;
26Finegold SM, Dowd SE, Gontcharova V, Liu C, Henley KE, Wolcott RD, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010 Aug;16(4):444–53.
27Kang D-W, Ilhan ZE, Isern NG, Hoyt DW, Howsmon DP, Shaffer M, et al. Differences in fecal microbial metabolites and microbiota of children with autism spectrum disorders. Anaerobe. 2018 Feb;49:121–31.
28Israelyan N, Margolis KG. Serotonin as a link between the gut-brain-microbiome axis in autism spectrum disorders. Pharmacological Research. 2018 Jun;132:1–6.
29Qiao Y, Wu M, Feng Y, Zhou Z, Chen L, Chen F. Alterations of oral microbiota distinguish children with autism spectrum disorders from healthy controls. Sci Rep. 2018 Jan 25;8.
30Yang Y, Tian J, Yang B. Targeting gut microbiome: A novel and potential therapy for autism. Life Sciences. 2018 Feb;194:111–9.
31Vuong HE, Hsiao EY. Emerging Roles for the Gut Microbiome in Autism Spectrum Disorder. Biological Psychiatry. 2017 Mar;81(5):411–23.
32Luna RA, Oezguen N, Balderas M, Venkatachalam A, Runge JK, Versalovic J, et al. Distinct Microbiome-Neuroimmune Signatures Correlate With Functional Abdominal Pain in Children With Autism Spectrum Disorder. Cellular and Molecular Gastroenterology and Hepatology. 2017 Mar;3(2):218–30.
33Kang D-W, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017 Jan 23;5.
34Kang D-W, Adams JB, Coleman DM, Pollard EL, Maldonado J, McDonough-Means S, et al. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Sci Rep. 2019 Apr 9;9(1):5821.
35Pärtty A, Kalliomäki M, Wacklin P, Salminen S, Isolauri E. A possible link between early probiotic intervention and the risk of neuropsychiatric disorders later in childhood: a randomized trial. Pediatr Res. 2015 Jun;77(6):823–8.
36Braun J. Tightening the Case for Gut Microbiota in Autism-Spectrum Disorder. Cellular and Molecular Gastroenterology and Hepatology. 2017 Mar;3(2):131–2.
37Wang S, Harvey L, Martin R, van der Beek EM, Knol J, Cryan JF, et al. Targeting the gut microbiota to influence brain development and function in early life. Neuroscience & Biobehavioral Reviews. 2018 Dec;95:191–201.
38Carlson AL, Xia K, Azcarate-Peril MA, Goldman BD, Ahn M, Styner MA, et al. Infant Gut Microbiome Associated With Cognitive Development. Biological Psychiatry. 2018 Jan;83(2):148–59.
39Yang B, Wei J, Ju P, Chen J. Effects of regulating intestinal microbiota on anxiety symptoms: A systematic review. Gen Psych. 2019 Apr;32(2):e100056.
40Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang J, Tito RY, et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. 2019 Feb 4;
41Cussotto S, Sandhu KV, Dinan TG, Cryan JF. The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Frontiers in Neuroendocrinology. 2018 Oct;51:80–101.