Chapter 40
Impact of the Gut Microbiome on Behavior and Emotions Ingrid Rivera-In˜iguez1, 2, Sonia Roman1, 2, Claudia Ojeda-Granados1, 2 and Panduro Arturo1, 2 1
Department of Molecular Biology in Medicine, Civil Hospital of Guadalajara, Guadalajara, Jalisco, Mexico; 2Health Sciences Center,
University of Guadalajara, Guadalajara, Jalisco, Mexico
INTRODUCTION Behavior and emotions have allowed humans to adapt to the environment, by detecting or reacting toward harmful or dangerous situations. However, chronic incidences of negative emotions or behavioral disorders may interact with genetic and environmental factors to detonate serious diseases. Patients with gastrointestinal disorders are a good example of recurrence of feelings of depression, anxiety, and stressful personalities. Therefore, it has been suspected that behavior and emotions could have a more gastrointestinal origin. The gastrointestinal tract contains a wide variety of microorganisms that sustain essential and beneficial interactions with the host [1]. Interestingly, gut microbiota communicates with the central nervous system, synthesizes neurotransmitters, and influences neurogenesis. As a result, microbiota modulates human behavior and emotion [2]. Understanding the communication pathways between the gut microbiota and central nervous system via distinct mediators, and the mechanisms of such pathways, could be a promising strategy in the treatment of behavioral and gastrointestinal disorders.
BRAIN STRUCTURES INVOLVED IN BEHAVIOR AND EMOTIONS Behavior and emotions are regulated in the brain reward circuit, localized in the mesolimbic region [3]. In this region, different brain structures are interconnected by neurons and neurotransmitters to regulate memory, learning, emotions, cognition, and behavior. The amygdala serves as a warning system that detects stimuli inputs related to emotions and sends this information to the reticular activating system [4]. Furthermore, the amygdala regulates the endocrine response to stress, by interacting with the hypothalamicepituitaryeadrenal (HPA) axis [5]. Physiological alterations of the amygdala have been associated with behavioral disturbances in germ-free mice [6]. In humans, an impaired function of the amygdala may interfere with social behavior, as observed in patients with schizophrenia and autism spectrum disorders (ASD) [7]. Furthermore, the amygdala and the prefrontal cortex (PFC) evaluates the stimuli and establishes a memory. This region also controls motor functions that give feedback information to the hippocampal septum. This information is known as working memory [8,9]. The PFC can also suppress negative responses, such as anxiety and fear [9,10]. The orbital frontal cortex (OFC) connects to the nucleus accumbens (NAC), hypothalamus, insula, dorsal striatum area, and medial frontal cortex, and regulates the emotional value to the memorized information [11].
GUTeBRAIN AXIS The brain maintains a bidirectional communication with the gastrointestinal system by neural, immune, and endocrine pathways that ensure a healthy behavior, as seen in Fig. 40.1. The brain interacts with the gut via sympathetic and parasympathetic fibers, vagus nerve, adrenergic nerve, and portal vein [12]. In fact, the vagus nerve is considered as the principal route of communication because it connects close to 100 million of enteric neurons to the brain.
Microbiome and Metabolome in Diagnosis, Therapy, and other Strategic Applications. https://doi.org/10.1016/B978-0-12-815249-2.00040-3 Copyright © 2019 Elsevier Inc. All rights reserved.
379
380 BLOCK | IV Diagnostic and Therapeutical Applications
CC
PFC
PUT
OFC
Nac VTA SN AMY
Normal signals
Immune
Neural Vagus nerve
Adrenergic nerve
Endocrine
Neuro-hormones
Neurotransmitters
HYP
Cortisol Dendritic cell
5-HT
SCFA
FIGURE 40.1 Healthy behavior and emotions. A healthy gut microbiota is characterized by being diverse and rich in beneficial bacteria that contributes to a normal communication with the brain. This includes a normal neurotransmitter signaling, cytokine profile, and endocrine signals. 5-HT, Serotonin; AMY, Amygdala; CC, cingulate cortex; HYP, Hypothalamus; Nac, Nucleus accumbens; OFC, Orbitofrontal cortex; PFC, prefrontal cortex; PUT, Putamen; SCFA, Short-chain fatty acids; SN, Substantia nigra; VTA, Ventral tegmental área.
Efferent signals from the brain (sympathetic and parasympathetic) control different gastrointestinal functions, like motility and the secretion of hormones and enzymes [13]. Bacteria in the gut produce neurotransmitters, like dopamine and serotonin, from the metabolism of tryptophan and neuropeptides, including P substance, calcitonin, neuropeptide Y (NPY), peptide YY, corticotrophin-releasing factor, pancreatic polypeptide, vasoactive intestinal polypeptide, glucagon-like peptide 1 (GLP-1), and somatostatin, among others [14,15](Fig. 40.1). Fig. 40.2 highlights how neuroendocrine signals affect mood and behavior. Hormones mediate the secretion of neurotransmitters by gut microbiota [16]. In the presence of physiological or psychological stress, the HPA axis activates the release of corticosterone by the hypothalamus, adrenocorticotropic hormone by the pituitary gland, and cortisol by the adrenal gland [13]. The release of systemic cortisol induces a proinflammatory response, which is capable of modifying gut microbiota composition [17,18]. Also, stress hormones such as adrenaline and noradrenaline promote overgrowth of pathogenic bacteria such as Escherichia coli (E. coli O157), Yersinia enterocolitica, and Pseudomonas aeruginosa [16]. Beneficial and pathogenic bacteria interact with the immune system, by secreting antimicrobial peptides (defensins, mucin, and angiogenin 4), mucus secretion, and releasing immunoglobulin A [19]. Bacteria associated with antigen
Gut Microbiome, Behavior, and Emotions Chapter | 40
PFC
CC PUT Nac
SN AMY
Neural
Immune Inflammatory signaling
Adrenergic nerve
Neuro-hormones
Neurotransmitters
HYP
VTA
Vagus nerve
OFC
Endocrine
381
ROS
Cortisol LPS 5-HT
TNF-α, IL-6, IL-1β, CRP
Dendritic cell
SCFA
FIGURE 40.2 Behavioral and emotional disorders. Gut microbiota dysbiosis is characterized by low diversity and overgrowth of pathogenic bacteria that contributes to an abnormal communication with the brain. This conditions includes impaired neurotransmitter signaling, a proinflammatory profile (TNFa, INFg, IL-6, IL-1), and altered endocrine signals. 5-HT, Serotonin; CC, cingulate cortex; HYP, Hypothalamus; LPS, Lipopolysaccharides; MY, Amygdala; Nac, Nucleus accumbens; OFC, Orbitofrontal cortex; PFC, prefrontal cortex; PUT, Putamen; SCFA, Short-chain fatty acids; SN, Substantia nigra; VTA, Ventral tegmental area.
recognition of Pathogen-Associated Molecular Patterns (PAMPS), by toll-like receptors (TLRs), lead to the subsequent activation of the immune response against pathogenic bacteria [20]. Environmental factors such as stress and diet can jeopardize the integrity of the intestinal mucosa [21]. This scenario facilitates that lipopolysaccharides and pathogenic bacteria translocate into the circulation resulting in metabolic endotoxemia, which is associated with chronic disease progression [22,23]. At the same time, the fermentation of nondigestible carbohydrates by gut bacteria generates short-chain fatty acids (SCFA), such as butyrate, acetate, and propionate that modulate the immune response and can also interact with the brain [24] (Fig. 40.2).
THE LINK BETWEEN MICROBIOTA, BEHAVIOR, AND EMOTIONS Initially, the hypothesis that the brain could communicate with the gut was formulated by observing patients with hepatic encephalopathy, who experienced mood changes and cognitive impairments. Later on, the gut microbiota-brain
382 BLOCK | IV Diagnostic and Therapeutical Applications
communication was confirmed in animal models. Studies in germ-free mice have revealed that neurogenesis depends mostly on gut microbiota composition [25]. NIH Swiss mice exhibit a normal behavior, whereas germ-free mice have an anxiety-like behavior [26]. Interestingly, when feces from NIH Swiss mice were transferred to germ-free BALB/c mice, this anxiety-like behavior was reduced, and levels of brain-derived neurotrophic factor (BDNF) increased [27]. Furthermore, the interactions between gut microbiota and brain activity vary according to sex, as shown by male/female differences in BDNF expression in germfree mice [28]. Also, gut bacteria produce neurotransmitters such as serotonin from tryptophan metabolism. This finding was verified by Liu and colleagues, who administered Lactobacillus plantarum PS128 and found increased levels of neurotransmitters dopamine and serotonin in the brain, which were associated with behavior [29].
Gastrointestinal Disorders and Emotions The relevance of the gutebrain axis is generally accepted in irritable bowel syndrome (IBS), colitis, and other experimental mood disorders [30]. Intrauterine stress in animal models causes gut microbial dysbiosis and this, in turn, affects both gastrointestinal and central nervous system development [27]. Furthermore, IBS patients tend to have more anxiety, depression, and neurotic type personality [31]. In fact, neuroticism is a personality trait that is related to IBS, colitis, Crohn’s disease, and dyspepsia [32]. Negative emotions exacerbate symptoms like altered motility, abdominal pain, and hyperreactivity among IBS patients [33]. It is proposed that these negative feelings influence the gastrointestinal system, through signals that arise from the central nervous, endocrine, and immune systems. In fact, stress alters cortisol levels via HPA axis dysregulation, and this is associated with IBS reactivity episodes, especially in women [34]. Cortisol also causes gut dysbiosis and exacerbates the inflammation profiles related to different gastrointestinal diseases, including Crohn’s disease and colitis [35]. Moreover, high levels of anxiety and depression induce the overexpression of the heat shock protein 70 (HSP70) in polymorphonuclear cells of patients with severe ulcerative colitis, destroying the intestinal mucosa [36]. In addition, patients with colitis have a poor microbiota diversity, including reductions in Akkermansia, which promotes protection against toxins [37]. Patients with inflammatory bowel disease also present reduced abundance of Firmicutes and a higher presence of Proteobacteria, Tenericutes, and Escherichia coli [38]. Patients with Crohn’s disease have a decreased number of Firmicutes and increased number of Bacteroidetes, when compared with healthy controls [39]. Patients with depression or anxiety are at higher risk of chronic diseases such as cardiovascular disease, diabetes, cerebral vascular disease, and obesity [40e43].
Psychiatric and Social Behavior Disorders The etiology of mental disorders such as schizophrenia, attention deficit hyperactivity disorder (ADHD), and autism spectrum disorders (ASD) is complex. However, autoimmunity is highlighted as a common key player [44]. In this sense, the gut immune system could detect beneficial bacteria as pathogenic and trigger an autoimmune environment by facilitating the activity of Th17 cells and suppressing the role of the regulatory T cells (Tregs) [45]. In Table 40.1, the gut microbiota composition in different behavioral and emotional disorders is depicted. As noted before, early exposure to adverse events triggers changes in the gut microbiota. Patients with schizophrenia present immunity activation for food protein antigens and pathogens and have a detrimental bacteria profile when compared to healthy control [46].
Gut Microbiota and Emotional Disorders Neurodevelopmental programming in the offspring depends largely on gut microbiota composition, which could be affected by stressful events [47]. Intrauterine stress in animal models causes a gut dysbiosis and this, in turn, affects central nervous system development [25]. The HPA axis is affected by microbiota alterations and early exposure to stress. Changes occurring in the microbiota composition influence anxiety-type behavior in stress-induced animal models [48]. Gram negative-derived lipopolysaccharides generate a systemic low-grade inflammatory profile, during stress and depression [49]. Also, neuroinflammation provoked by an imbalance in gut microbiota seems to affect the host’s behavior [50]. Oxidative stress can also be facilitated and plausibly promote neurodegeneration. Therefore, the impact of gut microbiota dysbiosis on cognition and learning can contribute to Alzheimer’s and Parkinson’s disease [51].
Gut Microbiome, Behavior, and Emotions Chapter | 40
383
TABLE 40.1 Gut Microbiota Composition in Behavioral and Emotional Disorders Disease
Experimental Model (Reference)
Measurements
Bacteria Composition
Irritable bowel syndrome (IBS) with anxiety or depression or low quality of life
37 patients with IBS [80]
Fecal microbiota by pyrosequencing
High Firmicutes/Bacteroidetes ration in IBS patients
Major depressive disorder (MDD)
46 MDD patients and 30 healthy controls [81]
Hamilton’s depression scale (HAMDS) Montgomery-Asberg Depression Rating Scale (MADRS), fecal gut microbiota by pyrosequencing, cytokine, and BDNF levels
Increments in fecal bacteria a-diversity in MDD patients related to high levels of Bacteroidetes, Proteobacteria, and Actinobacteria and reductions in Firmicutes Reductions in Faecalibacterium were negatively correlated with symptom severity
Depression
34 depressive patients and 33 healthy controls [82] Fecal microbiota transplanted (FMT) adult male SpragueeDawley rats from depressed patients [82]
Fecal microbiota by next generation sequencing, SCFA concentration, salivary cortisol, kynurenine/ tryptophan determination, cytokine levels, lipopolysaccharidebinding protein Rats: Behavioral test, corticosterone, CRP, cytokine, LPS, SCFA levels
Reductions in phylogenetic diversity in depressed group from phylum Prevotellaceae and increases in Thermoanaerobacteriaceae FMT rats: presented anhedonia-like behaviors, anxiety-like behaviors, increased plasma kynurenine and kynurenine/tryptophan ratio, reductions in microbiota diversity and increments in acetate levels
In toddlers, a higher phylogenetic diversity was observed among subjects with more social active temperaments, than those with fear temperaments [52]. Patients with anorexia nervosa had lower bacteria diversity when compared to healthy controls, in the inpatient environment, and these results were related to high levels of depression and anxiety [53].
Eating Behavior It has been proposed that the gutebrain axis interacts with the hunger-reward system, which is modulated by both genes and environmental factors that influence emotionally driven food decisions [54]. Several human genes that encode brain and gut receptors of the hunger-satiety circuit, and also in the brain reward system, express polymorphic alleles. Some of these alleles increase the risk for altered eating behaviors and excess weight, thus indirectly altering the gut microbiota [54e56]. Metabolic syndrome is related to impaired synthesis of neurotransmitters and deficiency in melatonin and tryptophan [57], which contribute to damage in the hippocampus [58]. The hypothesis that microbiota dysbiosis may affect eating behavior was proposed by Dr. Joe Alcok in 2014. He proposed that cravings and eating without control be elicited by microbes through the neuroendocrine system, to optimize their functioning [59]. Later on, Perry and colleagues documented that gut microbiota dysbiosis generates an increased production of acetate, which in turn could elicit overeating behaviors, by affecting the secretion of ghrelin, insulin, and glucose [60]. Postabsorptive signals seem to influence dopamine levels, which is a major neurotransmitter that is implicated in rewarding addictive behaviors [61]. An important single nucleotide polymorphism is the DRD2/ANKK1 A1 polymorphism, caused by an amino acid substitution (Glu713Lys), within the 11th ankyrin repeat of the ANKK1 gene. Carriers of the DRD2/ANKK1A1 allele are at risk of alcoholism and other addictive behaviors. Recently in a Mexican-Mestizo population, the highest prevalence of the A1 allele known to date worldwide was reported that coincides with the cultural history of alcohol consumption [62].
PSYCHOBIOTICS: FROM PREBIOTICS AND PROBIOTICS TO DIETARY INTERVENTIONS Table 40.2 summarizes the different strategies aimed to modify the gut microbiota, for the treatment of behavioral and emotional disorders. Animal and clinical studies have demonstrated that probiotic supplementation in autism, personality
384 BLOCK | IV Diagnostic and Therapeutical Applications
TABLE 40.2 Modulation of Gut Microbiota in the Treatment of Behavioral and Emotional Disorders Component
Experimental Model (Reference)
Measurements
Key Outcomes
Probiotics L. Rhamnosus JB-1
Corticosterone-induced stress and anxiety [64]
Anxiety, depression
Reductions in anxiety and stress increases in hippocampal GABAb1b receptor expression and cortical GABAAa2 receptor expression
Fermented milk product (Bifidobacterium animalis, Streptococcus thermophiles, Lactobacillus bulgaricus, Lactococcus lactis)
Healthy women [65] Twice/day for 4 weeks
MRI: Brain activity and response to face emotions
Reductions in task response in affective viscerosensory and somatosensory networks. Improvements in brain connectivity
Lactobacillus helveticus R0052
IL-10/ mice and wildtype 129/SvEv mice [83] 21 days of Western diet
Barnes maze: Memory and exploratory and anxiety-like behavior IFNY, IL-1b, IL-2, Il-4, IL-5, keratinocyte-derived chemokine, IL-10, IL-12, TNFa and corticosterone levels Fecal microbiota composition
Reductions in weight increases, inflammatory markers and corticosterone. Increased microbiota diversity associated with improvements in memory and reductions in anxietylike behavior
L. helveticusNS8
21 days of stress [84]
Cognition and behavior tests
Reductions in corticosterone levels and anxiety and increases in 5-HT, IL-10, and cognition
Bifidobacterium bifidum W23, Bifidobacterium lactis W52, Lactobacillus acidophilus W37, Lactobacillus brevis W63, Lactobacillus casei W56, Lactobacillus salivarius W24, Lactococcus lactis
Triple-blind randomized control study [85] Healthy volunteers Four weeks
Dysphoria assessed with Leiden index of depression sensitivity scale (LEIDS-r)
Reductions in cognitive reaction to sadness
VSL3 Streptococcus salivarius subsp., thermophilis, Bifidobacterium (B. breve, B. infanti, B. longum), Lactobacillus acidophilus, L. planarum, L. casei, and L. delbrueekisubsp. Bulgaricus
Inflammatory murine model of liver inflammation [86] C57BL/6 mice
Body weight, sickness social behavior, gut permeability, fecal microbiota, intravital microscopy cerebral vasculature, cerebral monocyte infiltration
Reductions in TNF-a, intestinal permeability, monocyte recruitment and improvements in social behavior
Probiotic yogurt (Lactobacillus acidophilus LA5 and Bifidobacterium lactisBB12) þ placebo capsule or Probiotic capsule (Actobacilluscasei, L. Acidophilus, L. Rhamnosus, L. Bulgaricus, Bifidobacterium breve, B. Longum, S. thermophilus) þ conventional yogurt (Streptococcus thermophilus and Lactobacillus bulgaricus starters) or conventional yogurt þ placebo capsule
Double-blind randomized study [87] 75 healthy petrochemical workers (male and female)
Health questionnaire (GHQ-28) Depression anxiety and stress scale (DASS), in kynurenine, tryptophan, neuropeptide Y, cortisol and ACTH levels
Improvements in health, depression, and anxiety with probiotic yogurt and probiotic capsule
Lactobacillus acidophilus, Lactobacillus casei, and Bifidobacterium bifidum
Double-blind randomized study [88] 40 patients with major depressive disorder (MDD) 8-week intervention
Depression with Beck depression inventory (BDI), fasting plasma glucose, insulin metabolism markers, lipids, c-reactive protein, oxidative stress markers, antioxidant capacity, and glutathione GSH levels
Reductions in depression scores and serum metabolic, inflammatory and oxidative stress improvements
Continued
Gut Microbiome, Behavior, and Emotions Chapter | 40
385
TABLE 40.2 Modulation of Gut Microbiota in the Treatment of Behavioral and Emotional Disordersdcont’d Component
Experimental Model (Reference)
Measurements
Key Outcomes
Randomized, doubleblind, placebocontrolled, pilot study [89] 44 patients with Inflammatory bowel syndrome with anxiety and depression
HAD scale: Anxiety and/or Depression, State-Trait Anxiety Inventory, (STAI), IBS global adequate relief, IBS symptoms, somatization, quality of life, changes in brain activation patterns (functional Magnetic Resonance Imaging, fMRI), serum inflammatory markers, neurotransmitters and BDNF, and fecal microbiota
Reduction in depression scores, in responses to negative emotional stimuli in amygdala and frontolimbic regions. Improvements in quality of life scores
Fructooligosaccharides or Oligosaccharides
Adult male Sprague eDawley Rats [90] Five weeks
Plasma glucose, PPY, GLP-1 levels, cortex and hippocampus BDNF and NMDARs protein levels and expression, Fecal microbiota
Increments in BDNF hippocampal regions, NR1 expression in hippocampal regions and frontal cortex, and NR2 in hippocampal regions, increments in PYY levels Growth of Lactobacilli and Bifidobacteria
Fructooligosaccharides (FOS) or Bimuno-galactooligosaccharides (B-GOS)
Double-blind randomized study [91] Healthy volunteers (22 males, 23 females) Three-week supplementation
Levels of salivary cortisol Computer-based Emotional Test Battery IQ test Personality, stress response, mood questionnaires, Beck depression inventory, and state-trait anxiety inventory
B-GOS reduced salivary cortisol levels and attentional vigilance to negative versus positive words
Resistant dextrin (Nutriose06) or maltodextrin
Females with type 2 Diabetes Mellitus [92]
General health questionnaire (GHQ) Depression, anxiety, and stress scale (DASS) White blood cell count, CD4, CD8, IFNg, IL12, IL4, IL10, cortisol, tryptophan, Adrenocorticotropic hormone, kynurenine, and plasma lipopolysaccharide (LPS)
Reductions in cortisol, kynurenine, IFNg, IL-12, IL-10, IL-4, LPS, CD8, GHQ, DASS
Randomized, doubleblind, placebocontrolled trial [93] Overweight and obese
Visual analogue scale for appetite Satiety quotient Three-factor eating questionnaire State-trait food cravings questionnaire trait Beck depression inventory Body esteem scale Binge eating scale Perceived stress scale State-trait anxiety inventory Energy intake, physical activity
Reductions in weight associated with improvements in food behavior and sad feelings in women. Improvements in food behavior and cognition in males.
B. longum NCC3001 subspecies longum strain (BL)
Prebiotics
Synbiotics Lactobacillus rhamnosus CGMCC1.3724 (LPR) þ oligofructose and inulin þ magnesium or Placebo
386 BLOCK | IV Diagnostic and Therapeutical Applications
disorders, depression, and cognitive impairment are a promising treatment strategy [15,63]. Bravo and colleagues achieved to reduce anxiety and stress, by giving L. rhamnosus JB-1 to mice, showing improvements in gamma-aminobutyric acid (GABA) receptor activity in brain structures related to emotion and behaviors [64]. In fact, the use of probiotics ameliorates brain connectivity, as shown in the supplementation of a fermented product containing Bifidobacterium animalis, Streptococcus thermophilus, Lactobacillus bulgaricus, and Lactococcus lactis to healthy women, finding signs of healthy behavior and emotions [64]. Prebiotics are another alternative, potentially enhancing SCFA such as butyrate, which activates gluconeogenesis, improves insulin sensitivity and energy efficiency and therefore is implicated in appetite and satiety. However, one of the most used prebiotics is resistant starch, with controversial changes in gut microbiota and behavior [66,67].
Healthy Diets and Supplements A westernized diet is deleterious for gut microbiota and behaviors [50]. In contrast, good sources of fats such as u-3-rich oils can modify gut microbiota and improve behavior [69]. Other dietary strategies include phytochemicals, which are rich in crucial nutrients for mental health such as magnesium, potassium, zinc, vitamin c, folic acid, and vitamin B12. By adding more fiber through the consumption of fruits and vegetables, depressive symptoms could be reduced [70].
Ethnic and Geographical Considerations Gut bacteria in the Japanese population, specifically Bacteroides plebeius, have a unique gene signature that codifies carbohydrate-active enzymes (CAZymes) porphyranases and agarases. These genes were acquired from the marine Bacteroidetes, Zobellia galactanivorans, which naturally degrade seaweeds [71]. The Hazda hunter-gatherers of Tanzania have a distinctive gut microbiota composition, enriched in Prevotella, Treponema, and unclassified Bacteroidetes that gives them the ability to digest fibrous vegetable foods and benefit from their nutrients [72]. Another example is the gut microbiome of the Inuit in the Canadian Arctic, with significantly reduced genetic diversity within the Prevotella, a genus related to their traditional diet, low in fiber and rich in animal fats and protein [73]. Furthermore, it has been documented that population groups living in communal lifestyle and consuming their seasonally traditional diets have a healthier microbiome and less microbiome-associated diseases [74]. In agreement with the genetic adaptations to traditional diets, and bacteria coevolving with humans, recommendations of the dietary pattern should include regionally available prebiotic foods, to promote a beneficial gut bacteria [75]. For example, dietary strategies based on the food components of the Mediterranean diet have proven beneficial for mental health [76]. Genetic adaptations to traditional and prehispanic foods of the Mexican populations seem to have influenced a healthier and fitter status [77,78]. Mexican prehispanic foods that have shown significant prebiotic and probiotic activities, include a healthy combination of maize-derived products and legumes, prickly pear, amaranth, chia seeds, fiber, and low-degree alcohol fermented beverages [77e79]. In other cultures, traditional foods such as soy, fermented products, turmeric, cocoa, and green tea that are rich in polyphenols, phytochemicals, and nondigestible fibers that could improve altered gut microbiota and general health.
CONCLUSION Personalized-medicine and genome-based nutrition strategies are necessary to balance the host’s genes, emotions, and diet, to prevent and alleviate gut microbiota-related diseases.
LIST OF ACRONYMS AND ABBREVIATIONS ADHD Attention deficit hyperactivity disorder ASD Autism spectrum disorders BDNF Brain-derived neurotrophic factor CNS Central nervous system GABA Gamma-aminobutyric acid GLP-1 Glucagon-like peptide 1 HPA Hypothalamic pituitary adrenal HSP70 Heat shock protein IBS Irritable bowel syndrome
Gut Microbiome, Behavior, and Emotions Chapter | 40
387
NAC Nucleus accumbens NCCDs Noncommunicable chronic diseases NPY Neuropeptide Y OFC Orbital frontal cortex PAMPS Pathogen-associated molecular patterns PFC Prefrontal cortex PYY Peptide YY SCFA Short-chain fatty acids TLRs Toll-like receptors Tregs Regulatory T cells
REFERENCES [1] Sommer F, Backhed F. The gut microbiota-masters of host development and physiology. Nat Rev Microbiol 2013;11:227e38. [2] Cani PD. Crosstalk between the gut microbiota and the endocannabinoid system: impact on the gut barrier function and the adipose tissue. Clin Microbiol Infect 2012;18(Suppl. 4):50e3. [3] Olds J, Milner P. Positive reinforcement produced by electrical stimulation of septal area and other regions of rat brain. J Comp Physiol Psychol 1954;47:419e27. [4] Schoenbaum G, Roesch M. Orbitofrontal cortex, associative learning, and expectancies. Neuron 2005;47:633e6. [5] McEwen BS, Nasca C, Gray JD. Stress effects on neuronal structure: hippocampus, amygdala, and prefrontal cortex. Neuropsychopharmacology 2016;41:3e23. [6] Price JL, Drevets WC. Neurocircuitry of mood disorders. Neuropsychopharmacology 2010;35:192e216. [7] Rutishauser U, Mamelak AN, Adolphs R. The primate amygdala in social perception - insights from electrophysiological recordings and stimulation. Trends Neurosci 2015;38:295e306. [8] Arana FS, Parkinson JA, Hinton E, Holland AJ, Owen AM, Roberts AC. Dissociable contributions of the human amygdala and orbitofrontal cortex to incentive motivation and goal selection. J Neurosci 2003;23:9632e8. [9] Riley MR, Constantinidis C. Role of prefrontal persistent activity in working memory. Front Syst Neurosci 2015;9:181. [10] Richard JM, Berridge KC. Prefrontal cortex modulates desire and dread generated by nucleus accumbens glutamate disruption. Biol Psychiatry 2013;73:360e70. [11] Sandoval-Salazar C, Ramirez-Emiliano J, Trejo-Bahena A, Oviedo-Solis CI, Solis-Ortiz MS. A high-fat diet decreases GABA concentration in the frontal cortex and hippocampus of rats. Biol Res 2016;49:15. [12] Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, Mu J, Foufelle F, Ferré P, Birnbaum MJ, Stuck BJ, Kahn BB. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature 2004;428:569e74. [13] Grenham S, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbe communication in health and disease. Front Physiol 2011;2:94. [14] Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y, Blennerhassett P, Macri J, McCoy KD, Verdu EF, Collins SM. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 2011;141:599e609. 609.e1-3. [15] Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J Psychiatr Res 2008;43:164e74. [16] Holzer P, Farzi A. Neuropeptides and the microbiota-gut-brain axis. Adv Exp Med Biol 2014;817:195e219. [17] Ando T, Brown RF, Berg RD, Dunn AJ. Bacterial translocation can increase plasma corticosterone and brain catecholamine and indoleamine metabolism. Am J Physiol Regul Integr Comp Physiol 2000;279:R2164e72. [18] Dunn AJ, Ando T, Brown RF, Berg RD. HPA axis activation and neurochemical responses to bacterial translocation from the gastrointestinal tract. Ann NY Acad Sci 2003;992:21e9. [19] Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013;5:1465e83. [20] Kalliomaki MA, Walker WA. Physiologic and pathologic interactions of bacteria with gastrointestinal epithelium. Gastroenterol Clin North Am 2005;34:383e99. vii. [21] Galley JD, Nelson MC, Yu Z, Dowd SE, Walter J, Kumar PS, Lyte M, Bailey MT. Exposure to a social stressor disrupts the community structure of the colonic mucosa-associated microbiota. BMC Microbiol 2014;14:189. [22] Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology 2011;140:1729e37. [23] Sharma V, Garg S, Aggarwal S. Probiotics and liver disease. Perm J 2013;17:62e7. [24] Cani PD, Everard A, Duparc T. Gut microbiota, enteroendocrine functions and metabolism. Curr Opin Pharmacol 2013;13:935e40. [25] Selkrig J, Wong P, Zhang X, Pettersson S. Metabolic tinkering by the gut microbiome: implications for brain development and function. Gut Microb 2014;5:369e80. [26] Crumeyrolle-Arias M, Jaglin M, Bruneau A, Vancassel S, Cardona A, Daugé V, Naudon L, Rabot S. Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 2014;42:207e17. [27] Neufeld K-AM, Kang N, Bienenstock J, Foster JA. Effects of intestinal microbiota on anxiety-like behavior. Commun Integr Biol 2011;4:492e4. [28] Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, Dinan TG, Cryan JF. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 2013;18:666e73.
388 BLOCK | IV Diagnostic and Therapeutical Applications
[29] Liu YW, Liu W-H, Wu CC, Juan YC, Wu YC, Tsai HP, Wang S, Tsai YC. Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naive adult mice. Brain Res 2016;1631:1e12. [30] Muscatello MRA, Bruno A, Scimeca G, Pandolfo G, Zoccali RA. Role of negative affects in pathophysiology and clinical expression of irritable bowel syndrome. World J Gastroenterol 2014;20:7570e86. [31] Tayama J, Nakaya N, Hamaguchi T, Tomile T, Shinozaki M, Saigo T, Shirabe S, Fukudo S. Effects of personality traits on the manifestations of irritable bowel syndrome. Biopsychosoc Med 2012;6:20. [32] Lackner JM, Gudleski GD, Ma C-X, Dewanwala A, Naliboff B. Fear of GI symptoms has an important impact on quality of life in patients with moderate-to-severe IBS. Am J Gastroenterol 2014;109:1815e23. [33] Zhu L, Huang D, Shi L, Liang L, Xu T, Chang M, Chen W, Wu D, Zhang F, Fang X. Intestinal symptoms and psychological factors jointly affect quality of life of patients with irritable bowel syndrome with diarrhea. Health Qual Life Outcomes 2015;13:49. [34] Suarez-Hitz KA, Otto B, Bidlingmaier M, Schwizer W, Fried M, Ehlert U. Altered psychobiological responsiveness in women with irritable bowel syndrome. Psychosom Med 2012;74:221e31. [35] Strober W. Impact of the gut microbiome on mucosal inflammation. Trends Immunol 2013;34:423e30. [36] Vlachos II, Barbatis C, Tsopanomichalou M, Abou-Assabeh L, Goumas K, Ginieri-Coccossis M, Economou M, Papadimitriou GN, Patsouris E, Nicolopoulou-Stamati P. Correlation between depression, anxiety, and polymorphonuclear cells’ resilience in ulcerative colitis: the mediating role of heat shock protein 70. BMC Gastroenterol 2014;14:77. [37] Vigsnaes LK, Brynskov J, Steenholdt C, Wilcks A, Licht TR. Gram-negative bacteria account for main differences between faecal microbiota from patients with ulcerative colitis and healthy controls. Benef Microbes 2012;3:287e97. [38] Tong M, Li X, Wegener Parfrey L, Roth B, Ippoliti A, Wei B, Borneman J, McGovern DP, Frank DN, Li E, Horvarth S, Knight R, Braun J. A modular organization of the human intestinal mucosal microbiota and its association with inflammatory bowel disease. PLoS One 2013;8:e80702. [39] Erickson AR, Cantarel BL, Lamendella R, Darzi Y, Mongodin EF, Pan C, Shah M, Halfvarson J, Tsyk C, Henrissat B, Raes J, Verberkmoes NC, Fraser CM, Hettich RL, Jansson JK. Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PLoS One 2012;7:e49138. [40] Kurth B-M, Ellert U. Perceived or true obesity: which causes more suffering in adolescents? Findings of the German health interview and examination survey for children and adolescents (KiGGS). Dtsch Arzteblatt Int 2008;105:406e12. [41] Penninx BWJH, Milaneschi Y, Lamers F, Vogelzangs N. Understanding the somatic consequences of depression: biological mechanisms and the role of depression symptom profile. BMC Med 2013;11:129. [42] Bjorntorp P. Do stress reactions cause abdominal obesity and comorbidities? Obes Rev 2001;2:73e86. [43] Weber-Hamann B, Werner M, Hentschel F, Bindeballe N, Lederbogen F, Deuschle M, Heuser I. Metabolic changes in elderly patients with major depression: evidence for increased accumulation of visceral fat at follow-up. Psychoneuroendocrinology 2006;31:347e54. [44] Benros ME, Eaton WW, Mortensen PB. The epidemiologic evidence linking autoimmune diseases and psychosis. Biol Psychiatry 2014;75:300e6. [45] Severance EG, Tveiten D, Lindstrom LH, Yolken RH, Reichelt KL. The gut microbiota and the emergence of autoimmunity: relevance to major psychiatric disorders. Curr Pharm Des 2016;22:6076e86. [46] Castro-Nallar E, Bendall ML, Perez-Losada M, Sabuncyan S, Severance EG, Dickerson FB, Schroeder JR, Yolken RH, Crandall KA. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ 2015;3:e1140. [47] Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, Kubo C, Koga Y. Postnatal microbial colonization programs the hypothalamic-pituitaryadrenal system for stress response in mice. J Physiol 2004;558:263e75. [48] Bangsgaard Bendtsen KM, Krych L, Sorensen DB, Pang W, Nielsen DS, Josefsen K, Hansen LH, Sorensen SJ, Hansen AK. Gut microbiota composition is correlated to grid floor induced stress and behavior in the BALB/c mouse. PLoS One 2012;7:e46231. [49] De Punder K, Pruimboom L. Stress induces endotoxemia and low-grade inflammation by increasing barrier permeability. Front Immunol 2015;6:223. [50] Pyndt Jorgensen B, Hansen JT, Krych L, Larsen C, Klein AB, Nielsen DS, Josefsen K, Hansen AK, Sorensen DB. A possible link between food and mood: dietary impact on gut microbiota and behavior in BALB/c mice. PLoS One 2014;9:e103398. [51] Mulak A, Bonaz B. Brain-gut-microbiota axis in Parkinson’s disease. World J Gastroenterol 2015;21:10609e20. [52] Christian LM, Galley JD, Hade EM, Schoppe-Sullivan S, Kamp Dush C, Bailey MT. Gut microbiome composition is associated with temperament during early childhood. Brain Behav Immun 2015;45:118e27. [53] Kleiman SC, Watson HJ, Bulik-Sullivan EC, Huh EY, Tarantino LM, Bulik CM, Carroll IM. The intestinal microbiota in acute anorexia nervosa and during renourishment: relationship to depression, anxiety, and eating disorder psychopathology. Psychosom Med 2015;77:969e81. [54] Panduro A, Rivera-Iniguez I, Sepulveda-Villegas M, Roman S. Genes, emotions and gut microbiota: the next frontier for the gastroenterologist. World J Gastroenterol 2017;23:3030e42. [55] Foley DL, Morley KI, Madden PAF, Heath AC, Whitfield JB, Martin NG. Major depression and the metabolic syndrome. Twin Res Hum Genet 2010;13:347e58. [56] Wiltink J, Michal M, Wild PS, Schneider A, Konig J, Blettner M, Munzel T, Schulz A, Weber M, Fottner C, Pfeiffer N, Lackner K, Beutel ME. Associations between depression and diabetes in the community: do symptom dimensions matter? Results from the Gutenberg Health Study. PLoS One 2014;9:e105499. [57] Elovainio M, Hurme M, Jokela M, Pulkki-Raback L, Kivimaki M, Hintsanen M, Hintsa T, Lehtimaki T, Vikari J, Raitakari OT, KeltikangasJarvinen L. Indoleamine 2,3-dioxygenase activation and depressive symptoms: results from the Young Finns Study. Psychosom Med 2012;74:675e81.
Gut Microbiome, Behavior, and Emotions Chapter | 40
389
[58] Schiepers OJG, Wichers MC, Maes M. Cytokines and major depression. Prog Neuro-Psychopharmacol Biol Psychiatry 2005;29:201e17. [59] Alcock J, Maley CC, Aktipis CA. Is eating behavior manipulated by the gastrointestinal microbiota? Evolutionary pressures and potential mechanisms. BioEssays News Rev Mol Cell Dev Biol 2014;36:940e9. [60] Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL, Petersen KF, Kibbey RG, Goodman AL, Shulman GI. Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic syndrome. Nature 2016;534:213e7. [61] Sustkova-Fiserova M, Jerabek P, Havlickova T, Kacer P, Krsiak M. Ghrelin receptor antagonism of morphine-induced accumbens dopamine release and behavioral stimulation in rats. Psychopharmacol (Berl) 2014;231:2899e908. [62] Panduro A, Ramos-Lopez OM, Campollo O, Zepeda-Carrillo EA, Gonzalez-Aldaco K, Torres-Valadez R, Roman S. High frequency of the DRD2/ ANKK1 A1 allele in Mexican Native Amerindians and Mestizos and its associations with alcohol consumption. Drug Alcohol Depend 2017;172:66e72. [63] Distrutti E, O’Reilly J-A, McDonald C, Cipriani S, Renga B, Lynch MA, Fiorucci S. Modulation of intestinal microbiota by the probiotic VSL#3 resets brain gene expression and ameliorates the age-related deficit in LTP. PLoS One 2014;9:e106503. [64] Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, Bienenstock J, Cryan JF. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA 2011;108:16050e5. [65] Tillisch K, Labus J, Kilpatrick L, Jiang Z, Stains J, Ebrat B, Guyonnet D, Legrain-Raspaud S, Trotin B, Naliboff B, Mayer EA. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013;144:1394e401. 1401.e1-4. [66] Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garret WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013;341:569e73. [67] Lyte M, Chapel A, Lyte JM, Ai Y, Proctor A, Jane JL, Philips GJ. Resistant starch alters the microbiota-gut brain axis: implications for dietary modulation of behavior. PLoS One 2016;11:e0146406. [68] Xu Z, Knight R. Dietary effects on human gut microbiome diversity. Br J Nutr 2015;113(Suppl: S1e5). [69] Robertson RC, Seira Oriach C, Murphy K, Moloney GM, Cryan JF, Dinan TG, Paul Ross R, Stanton C. Omega-3 polyunsaturated fatty acids critically regulate behaviour and gut microbiota development in adolescence and adulthood. Brain Behav Immun 2017;59:21e37. [70] Miki T, Eguchi M, Kurotani K, Kochi T, Kuwahara K, Ito R, Tsuruoka H, Akter S, Kashino I, Kabe I, Kawakami N, Mizoue T. Dietary fiber intake and depressive symptoms in Japanese employees: the Furukawa Nutrition and Health Study. Nutrition 2016;32:584e9. [71] Hehemann J-H, Correc G, Barbeyron T, Helbert W, Czjzek M, Michel G. Transfer of carbohydrate-active enzymes from marine bacteria to Japanese gut microbiota. Nature 2010;464:908e12. [72] Schnorr SL, Candela M, Rampelli S, Centanni M, Consolandi C, Basaglia G, Turroni S, Biagi E, Peano C, Severgnini M, Fiori J, Gotti R, De Bellis G, Luiselli D, Brigidi P, Mabulla A, Marlowe F, Henry AG, Crittenden AN. Gut microbiome of the Hadza hunter-gatherers. Nat Commun 2014;5:3654. [73] Girard C, Tromas N, Amyot M, Shapiro BJ. Gut microbiome of the canadian arctic inuit. mSphere 2017;2. [74] Smits SA, Leach J, Sonnenburg ED, Gonzalez CG, Lichtman JS, Reid G, Knight R, Manjurano A, Changalucha J, Elias JE, Dominguez-Bello MG, Sonnenburg JL. Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science 2017;357:802e6. [75] Kang JX. Gut microbiota and personalized nutrition. J Nutr Nutr 2013;6:IeII. [76] Logan AC, Jacka FN. Nutritional psychiatry research: an emerging discipline and its intersection with global urbanization, environmental challenges and the evolutionary mismatch. J Physiol Anthropol 2014;33:22. [77] Roman S, Ojeda-Granados C, Ramos-Lopez O, Panduro A. Genome-based nutrition: an intervention strategy for the prevention and treatment of obesity and nonalcoholic steatohepatitis. World J Gastroenterol 2015;21:3449e61. [78] Ojeda-Granados C, Panduro A, Gonzalez-Aldaco K, Sepulveda-Villegas M, Rivera-Iniguez I, Roman S. Tailoring nutritional advice for Mexicans based on prevalence profiles of diet-related adaptive gene polymorphisms. J Pers Med 2017;7. [79] Avila-Nava A, Noriega LG, Tovar AR, Granados O, Perez-Cruz C, Pedraza-Chaverri J, Torres N. Food combination based on a pre-hispanic Mexican diet decreases metabolic and cognitive abnormalities and gut microbiota dysbiosis caused by a sucrose-enriched high-fat diet in rats. Mol Nutr Food Res 2017;61. [80] Jeffery IB, O’Toole PW, Ohman L, Claesson MJ, Deane J, Quigley EM, Simrén M. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut 2012;61:997e1006. [81] Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J, Li L, Ruan B. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun 2015;48:186e94. [82] Kelly JR, Borre Y, O’ Brien C, Patterson E, EL Aidy S, Deane J, Kennedy PJ, Beers S, Scott K, Moloney G, Hoban AE, Scott L, Fitzgerald P, Ross P, Stanton C, Clarke G, Cryan JF, Dinan TG. Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res 2016;82:109e18. [83] Ohland CL, Kish L, Bell H, Thiesen S, Hotte N, Pankiv E, Madsen KL. Effects of Lactobacillus helveticus on murine behavior are dependent on diet and genotype and correlate with alterations in the gut microbiome. Psychoneuroendocrinology 2013;38:1738e47. [84] Liang S, Wang T, Hu X, Luo J, Li W, Wu X, Duan Y, Jin F. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015;310:561e77. [85] Steenbergen L, Sellaro R, van Hemert S, Bosch JA, Colzato LS. A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav Immun 2015;48:258e64. [86] D’Mello C, Ronaghan N, Zaheer R,Dicay M, Le T, MacNaughton WK, Surrette MG, Swain MG. Probiotics improve inflammation-associated sickness behavior by altering communication between the peripheral immune system and the brain. J Neurosci 2015;35:10821e30.
390 BLOCK | IV Diagnostic and Therapeutical Applications
[87] Mohammadi AA, Jazayeri S, Khosravi-Darani K, Solati Z, Mohammadpour N, Asemi Z, Adab Z, Djalali M, Tehrani-Doost M, Hosseini M, Eghtesadi S. The effects of probiotics on mental health and hypothalamic-pituitary-adrenal axis: a randomized, double-blind, placebo-controlled trial in petrochemical workers. Nutr Neurosci 2016;19:387e95. [88] Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, Jafari P, Akbari H, Taghizadeh M, Memarzadeh MR, Asemi Z, Esmaillzadeh A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: a randomized, double-blind, placebo-controlled trial. Nutrition 2016;32:315e20. [89] Pinto-Sanchez MI, Hall GB, Ghajar K, NArdelli A, Bolino C, Lau JT, Martin FP, Cominetti O, Welsh C, Rieder A, Traynor J, Gregory C, De Palma G, Pigrau M, Ford AC, Macri J, Berger B, Bergonzelli G, Surette MG, Collins SM, Moayyedi P, Bercik P. Probiotic Bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: a pilot study in patients with irritable bowel syndrome. Gastroenterology 2017;153:e8448e59. [90] Savignac HM, Corona G, Mills H, Chen L, Spencer JP, Tzortzis G, Burnet PW. Prebiotic feeding elevates central brain derived neurotrophic factor. Neurochem Int 2013;63:756e64. [91] Schmidt K, Cowen PJ, Harmer CJ, Tzortzis G, Errington S, Burnet PWJ. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacol (Berl) 2015;232:1793e801. [92] Farhangi MA, Javid AZ, Sarmadi B, Karimi P, Dehghan P. A randomized controlled trial on the efficacy of resistant dextrin, as functional food, in women with type 2 diabetes: targeting the hypothalamic-pituitary-adrenal axis and immune system. Clin Nutr 2017. published online June 10. [93] Sanchez M, Darimont C, Panahi S, Drapeau V, Marette A, Taylor VH, Doré J, Tremblay A. Effects of a diet-based weight-reducing program with probiotic supplementation on satiety efficiency, eating behavior traits, and psychosocial behaviors in obese individuals. Nutrients 2017;9.
FURTHER READING [1] Roman S, Panduro A. Genomic medicine in gastroenterology: a new approach or a new specialty? World J Gastroenterol 2015;21(27):8227e37.