IMR Press / FBL / Volume 25 / Issue 2 / DOI: 10.2741/4810
Review
The neurobehavioral implications of the brain and microbiota interaction
Show Less
1 Behavioral Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
2 Behavioral Neuroscience/Neurobiology Unit, Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
3 Molecular Bacteriology Unit, Department of Microbiology and Parasitology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
Send correspondence to: Adejoke Y. Onaolapo, Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria, Tel:2348062240434, E-mail: ayonaolapo@lautech.edu.ng
Front. Biosci. (Landmark Ed) 2020, 25(2), 363–397; https://doi.org/10.2741/4810
Published: 1 January 2020
Abstract

In the past, microorganisms were not considered to be particularly important in brain development and functioning. However, recent evidence shows the existence of a bidirectional, and possibly multidimensional relationship between the body microbiota and the brain. The microbiota influence brain behavior in health or disease, by utilizing endocrine, neurocrine and immunologic signaling pathways. Also, the chemical mediators involved range from known neurotransmitters to small peptide molecules. Here, we discuss the evidence that currently exists in experimental animals and/or humans in support of the existence of a relationship involving the skin/gut microbiome, the brain, and behavior; and the mechanisms involved in such interactions. The implications of such interactions for shifts in behaviors, and the pathogenesis of behavioral and neurodegenerative disorders are also discussed. Finally, the possible clinical applications of deliberate manipulations of the microbiota composition and density for the management or prevention of behavioral and neurodegenerative disorders is discussed.

Keywords
Brain
Gastrointestinal tract
Neurobehavior
Skin
microbiota
Microbial colonies
Probiotics
Review
2. INTRODUCTION

Humans and animals share a life-long relationship with a number of microbial species resident on or within their body. A number of these microbes maintain a symbiotic relationship with the host, and have been shown to regulate host’s nutrition/ metabolism, ensure the development/functioning of the immune system, maintain general health/wellbeing (1-2), and modulate the development of diseases (3).

The mammalian microbiota is a unique ensemble of microorganisms that include bacteria, fungi, and viruses resident in the different niches (skin, gut, genitalia) on and within the body (4). The diversity of these microbes, and the significant inter-individual variations in their composition in health (1) and with aging (5) is reshaping our understanding of the extent of these relationships, as well as the implications for the development of chronic diseases (6) and brain disorders (4-5).

In recent times, the term gut microbiota has been used to describe the microbial population resident in the gastrointestinal tract. Research exploring the gut microbiota has continued to expand our understanding of the widespread systemic implications of gut microbiota dysbiosis as it relates to the development of obesity, diabetes mellitus (7), inflammatory bowel diseases (8), skin diseases (9), and neurodevelopmental/neuropsychiatric (10-11) disorders.

Recent advances demonstrate the importance of the microbiota (especially the gut microbiota) in the maintenance of brain structure and function, and the development of neurological disorders (5). The local and systemic benefits of the skin in regulating the homeostasis of epidermal keratinocytes and the host immune network are also being revealed (12). Ongoing research into the dynamics and molecular mechanisms of the gut-brain axis suggests the existence of a skin-gut-brain axis (13-14), which may also (directly or indirectly) influence the development of the immune system and/or the brain (15-17). In this review, we discuss the evidence that currently exists in experimental animals and/or humans in support of the existence of a relationship involving the skin and/or gut microbiome, the brain, and behavior; and mechanisms involved in such interactions.

2.1. The microbiota
2.1.1. The microbiota in health

The human body contains about a thousand different species of bacteria carrying at least 150 times more genes than exists in the entire human genome (18). The bacterial composition in and on the human body varies across subjects, and with multiple health conditions. Studies have shown that these variations are influenced by environmental factors including geographical locations, sex, race (19), dietary factors (20), social interaction (21-22) and genetics (23). The evolution of the microbiota with advancing age, as well as the important role it plays in human health maintenance and the development of diseases (24–26) have also been reported.

In humans, earlier studies had reported that bacterial colonization of the ‘germ-free’ infant begins at birth, and gut microbial communities become demonstrable as early as the 1st week of life (27). Thereafter, the bacterial composition continues to fluctuate until about 1-3 years of age when it begins to approach the adult microbial composition (27-28) This notion that human microbial colonization begins at birth (in the birth canal for vaginally-delivered babies) since the uterus was considered sterile (29) was entertained for years. However, in the last few years, researchers have begun to question the sterility of the uterus (30-31), with suggestions that microbial colonization in humans (like in a number of other organisms) could possibly begin in-utero (29, 32). The presence of bacteria in close to a third of the placentas obtained following life-birth (33), evidence of bacteria in meconium (34), the demonstration of microbial DNA in the placenta (32) and amniotic fluid (31, 35) are evidence in support of this recent view. Evaluation of the microbiome of human placentas revealed the presence of microbial colonies consisting of non-pathogenic microbiota including Fusobacteria, Tenericutes, Firmicutes, Bacteroidetes and Proteobacteria. This microbial composition was closely-related to the microbiota of the human oral cavity (32). Also, similarities between the microbial populations in the placenta, amniotic fluid and neonate’s meconium are all accumulating evidence supporting the prenatal colonization of the infant.

An initial contact with microbial colonies provides a nidus for a gradual evolution of these communities into a diverse eco-system, which is maintained throughout the host’s lifetime (36). A symbiotic relationship develops between the host and the microbial community, and this relationship may be critical for the sustenance of health and the prevention of disease. The rapid colonization of the infant skin within days of delivery has been shown to coincide with functional changes, including maturation of skin structure and function (37). Also, while the composition of microbial communities on the skin evolves with age and environment (39), proper and timely establishment of a healthy skin microbiome may be crucial in preventing the invasion of the body by potentially-harmful microbes (37). Failure of this could predispose to the development of cutaenous diseases, allergies, and inflammatory non-communicable diseases (12). Contact with microbial communities also contributes to the development of cutaenous homeostasis and immune system (37,39-40). The proper development of the immune system is also crucial to the development and proper functioning of most organ systems, including the human brain (12). The skin microbiome contributes significantly to the development of host immunity by influencing the host cells to produce endogenous antimicrobial peptides. It also modulates the host’s innate and adaptive defense system. In healthy skin, Staphylococcus epidermidis is a commonly-isolated microbe that protects humans from colonization by pathogenic bacteria (41). This shows that a balance between the host cells and resident bacteria is crucial for optimal skin defense and body health (42).

The gut microbiota includes a diverse community of microbes that inhabit the bowel (8). Over 100 trillion bacteria (made up of about one thousand species) reside in the human gastrointestinal tract (43-44). The gut microbiota is fairly more stable than the skin microbiota, although, like the skin microbiota, it shows significant variations amongst healthy subjects (45). It varies with age, aging, nutrition, antibiotic therapy and illness (43). The gut microbiota is also important in the development and proper functioning of the immune system, and the synthesis of vitamins (44). Bacterial colonies in the gut also aid in metabolizing indigestible fibers and in the defense against colonization by infectious and/or opportunistic pathogens. They also contribute to the formation of the architecture of the intestine (46).

The crucial role played by the microbiota in proper development and functioning of body organs have also been highlighted by studies that have evaluated the beneficial effects of prebiotics or probiotics supplements in humans (47-48). Supplementation of the milk of formula-fed infants with prebiotics oligosaccharides have been shown to promote the growth of probiotics members of the Bifidobacterium and Lactobacillus species; and inhibit the growth of pathogenic members of the Clostridium species (48). This has been associated with a decreased incidence of colic in these children (48). In rats, probiotic supplementation with Lactobacillus reuteri has been shown to increase the number of action potentials and the excitability of the enteric nervous system; with suggestions that this could modulate gut motility and pain perception (49). Prenatal probiotics interventions have also been shown to modulate host-microbe interactions via their ability to influence the expression of Toll–like receptor-related genes in the placenta and fetal microbiome, thereby reducing the risk of the development of diseases like atopic dermatitis (50-51).

2.1.2. The microbiota in disease

A complex symbiosis develops between the microbiota and the human body; its disruption could be detrimental to health and well-being. Microbial dysbiosis has been linked to a myriad of causes, including physiological alterations that occur due to antibiotic therapy, diet, and hygiene (44, 52). Also, physical activity (53) and environmental factors like temperature, humidity, ethnicity and cultural habits (54-55) can significantly alter the microbial composition of the human microbiota.

A number of diseases have been linked to gut microbiota dysbiosis (Table 1), including metabolic diseases (obesity, type 2 diabetes mellitus and cardiovascular disease), digestive system disorders (inflammatory bowel disease, coeliac disease and colorectal cancers), skin conditions (acne, dermatitis and eczema), respiratory system diseases (cystic fibrosis, chronic obstructive pulmonary disease and asthma), and neurological/neurodegenerative disorders such as autism spectrum disorders, schizophrenia, bipolar disorder, Parkinson’s disease, and Alzheimer’s disease (56–61). Studies in obese rodents and humans have demonstrated a predominance or relative abundance of certain microbes like the Firmicutes, or a reduction in the colonization by Bacteroidetes (62-63). Also, studies in germ-free mice have demonstrated the role of intestinal microbiota in fat storage (64–66). The absence of intestinal microbes in these mice was protective against the development of diet-induced obesity (64–66).

Table 1 Chronic diseases and alterations in bodily microbial density
Disorder Host Microbial composition Reference
Obesity Humans FirmicutesBacteroidetes (62,63)
Type 2 diabetes mellitus Human Faecalibacterium prausnitzii and Roseburia intestinalis (67)
Dysmetabolism Murine administration of Akkermansia muciniphila improved glucose control (68)
Coeliac disease Human a decrease in the ratio of gram-positive to gram-negative bacteria, lower concentrations of Clostridium histolyticum, Faecalibacterium prausnitzii, Bifidobacterium and C. lituseburense with coeliac disease (69)
Psoriasis Human high concentrations of Actinobacteria and Firmicutes (39)
Leprosy Human Proteobacteria with a reduction in Actinobacteria (72)
Atopic dermatitis Human The decrease in microbiome diversity, alteration in microbial diversity correlates with disease severity and linked to mutations in the fillaggrin genes. ↑Firmicutes and staphylococci, Staphylococcus aureus and Staphylococcus epidermidis (42)
Acne Human ↑Propionibacterium acnes (73)

In humans, metagenome-wide association studies have shown that specific intestinal bacterial colonies or bacterial genes correlate significantly with type 2 diabetes mellitus. These studies also demonstrated low concentrations of certain bacteria, especially butyrate- and or short-chain fatty acid producing bacteria like Faecalibacterium prausnitzii and Roseburia intestinalis in subjects with type 2 diabetes mellitus (67). The importance of gut microbiota in the control of blood glucose levels in diet-induced dysmetabolism was demonstrated in mice that were fed high-fat diet, in which oral administration of Akkermansia muciniphila was associated with improved glucose tolerance and attenuation of inflammation within the adipose tissue (68).The administration of metformin to these mice was also associated with an increase in the concentration of Akkermansia, a mucin-degrading bacterium, suggesting that modulation of the gut microbiota could impact the antidiabetic effects of metformin (68).

Gastrointestinal diseases like coeliac disease and inflammatory bowel disease have also been linked to dysbiosis of the gut microbiota. De Palma et al. (69) reported a decrease in the levels of immunoglobulin (Ig) A-coated fecal bacteria in treated and untreated subjects with celiac disease (CD) compared to healthy controls. Levels of Ig G and Ig M-coated bacteria were also significantly lesser in treated CD compared to untreated CD subjects. Bacterial counts also revealed a decrease in the ratio of gram-positive to gram-negative bacteria in treated and untreated CD patients compared to healthy controls; while lower concentrations of Clostridium histolyticum, Faecalibacterium prausnitzii, Bifidobacterium and C. lituseburense were observed in untreated CD patients compared to healthy controls (69).

Dysbiosis of the skin microbiome has also been observed in a few systemic diseases (70); however, how this contributes to disease pathogenesis and/or pathophysiology remains unclear (39,71). In patients with psoriasis, the skin microbiome has been reported to have high concentrations of Actinobacteria and Firmicutes (37). Also, in subjects with leprosy, (specifically lesional forms) the microbiome contained an abundance of Proteobacteria, with a reduction in Actinobacteria (72). Patients with atopic dermatitis (AD) have a decrease in microbiome diversity, compared to healthy controls (42). This loss of diversity correlated with disease severity, and was also linked to mutations in the fillaggrin genes (42). In patients with AD, the skin microbiome has also been shown to harbor higher concentrations of Firmicutes and staphylococci such as Staphylococcus aureus and Staphylococcus epidermidis (42). Acne is another skin condition which is associated with the presence of Propionibacterium acnes (73).

2.1.2.1. Antibiotic use and the microbiota

The use of antibiotics has extended man’s life expectancy by an average of 20 years (74). However, it is very unfortunate that these agents have been indiscriminately used and/or abused. As a result of this, microbes are becoming increasingly-resistant to antibiotics through a number of mechanisms, including enzymatic inactivation of the antibiotics, modification of the targets of the antibiotics and prevention of the accumulation of lethal intra-cellular concentrations of the antibiotic through efflux pumps (75). Antibiotic resistance may also occur through mutations and horizontal transfer of resistance genes.

Uncontrolled or unguarded use of antibiotics has been shown to cause dysbiosis within the human microbiota. Gut dysbiosis has been associated with significant reduction in taxonomic richness, diversity, and evenness (76,77). Depletion of bacterial diversity, loss of potential competitors, lower expression of natural antibacterial, as well as decreased neutrophil-mediated killing, may enhance host’s susceptibility to exogenous pathogens or opportunistic members of the microbiota (78,79). Beyond alteration of composition, antibiotics can also affect gene expression, protein activity, and overall metabolism of the gut microbiome (80); allowing changes in the nutritional landscape of the gut which directs the expansion of pathogenic bacteria (81). Alterations in the gut microbiome that occur as a result of antibiotics use or abuse predisposes the host to infections that are caused by newly-acquired or opportunistic pathogens. The accumulation of resistance genes by members of the gut microbiota can also follow abuse of antibiotics.

Antibiotic-induced dysbiosis may also adversely impact brain function. Antibiotics that are strong enough to kill off gut bacteria can also impede the growth of new brain cells in the hippocampus, a section of the brain associated with memory (82).

3. THE BRAIN AND THE MICROBIOTA
3.1. Brain development and the microbiota

In recent times, researchers have begun to ask questions about the presence of a microbiome in the brain, especially following reports demonstrating the presence of microbes in the brain specimens of HIV-infected subjects (83). While the healthy human brain continues to be considered a sterile environment, the role of the microbiome in other regions of the body in modulating brain development, neurochemistry and behaviors (Table 2) have been examined (84–88).

Table 2 Brain development and the microbiota
Model Neurodevelopmental changes References
Mice Exposure of germ-free mice to normal gut microbiota early in life influenced brain development, motor activity and anxiety-like behaviours by modulating canonical signal pathways, and the expression of genes synaptic-related proteins like synaptophysin and PSD-95 in the striatum. (86)
Mice Germ-free mice had alteration in levels of noradrenaline, dopamine and serotonin (86)
Mice Post-natal reconstitution of germ-free mice with normal gut microbiota flora containing Bifidobacterium infantis improved the stress response of the germ-free mice by influencing adrenocorticotrophic hormone levels and development of the hypothalomopituitary axis (90)
Mice Absence of normal gut microbial flora as observed in germ-free mice was associated with a decrease in the expression of the mRNA of N-methyl-D-aspartate receptor subunit in the amygdala (86,87)
Mice A decrease in the expression of serotonin receptor in the hippocampus and increased hippocampal expression of brain-derived neurotrophic factor has also been reported in germ- free mice (102, 103)
Rat Bifdobacterium infantis colonization was associated with antidepressant effects and a ↓serontonin levels in frontal cortex and an ↑ kynurenic acid and tryptophan (91-92)
Mice Antibiotic-induced depletion of gut microbiota from early adolescence caused cognitive deficits, alteration in tryptophan metabolism and ↓oxytocin, ↓brain derived neurotropic factor and ↓vasopressin in the adult brain (106)

Human brain development begins in-utero and extends through adolescence into early adulthood. It is an intricate process that involves the migration of cells over long distances (in-utero), with extensions of their cell processes over even greater distances (4,89). The protracted course of pre- and post-natal brain development and the complexity of the process increase its vulnerability, providing a window of opportunity for environmental factors to exert influences that modulate brain structure, function and neurotransmitter/receptor expression (4,76, 87). In the last decade or more, there have been suggestions that the microbiota (especially the gut) influences brain development and maturation (86,90).

While examining the impact of normal gut microbiota on brain development in mice, Diaz Heijtz et al. (86) concluded that early-life but not late-life exposure of germ-free mice to normal gut microbiota was associated with the modulation of motor activity and anxiety-like behaviors (86). These modulatory effects on behaviors occurred via their ability to influence canonical signal pathways, and release of brain neurotransmitter (including serotonin, dopamine and noradrenaline) and synaptic-related proteins like synaptophysin (86). Sudo et al (90) reported that post-natal reconstitution of germ-free mice with normal gut microbiota flora containing Bifidobacterium infantis improved the response of germ-free mice to stress by influencing adrenocorticotrophic hormone levels in mice (90). Studies in rodents have also demonstrated the antidepressant effects of Bifidobacterium infantis via its ability to normalize the concentration of pro-inflammatory cytokines and tryptophan (91-92). Gut microbiota have also been reported to influence the expression and synthesis of receptors and neurotransmitters (including serotonin) which are important in brain development (93). Desbonnet et al. (91) also reported that in rat pups of dams colonized with Bifidobacterium infantis a decrease in frontal cortex concentration of serotonin, and an increase in plasma levels of kynurenic acid and tryptophan were observed, when compared to pups of non-colonized controls (91).

During the first three years of life, microbial colonization of the different niches on and within the human body occurs rapidly. There is an initial domination of the microbiota by members of the Actinobacteria and Proteobacteria phyla, and then a gradual shift towards Firmicutes and Bacteroidetes domination as adulthood is reached (94-95). This process also coincides with the period of brain development (spanning from early-life to adolescence) during which synaptogenesis and pruning goes on in the brain (94-97). In the growing infant, there is increasing advocacy for the consideration of the gut microbial communities as modifiable influencers of brain development and host behavior (98–101) The involvement of the gut microbiota in the programming of the brain circuitries that modulate a number of behavioural responses including stress, anxiety, cognition, motor coordination and social interaction during early brain maturation (90, 99,102–105) have also been reported.

Gut microbiota depletion (due to use of antibiotics from early adolescence) was associated with cognitive deficits, alteration in tryptophan metabolism, and a significant decrease in the expression of oxytocin, brain derived neurotropic factor and vasopressin in the adult brain (106). There have been suggestions that gut microbial depletion or dysbiosis may play a role in the pathogenesis of attention-deficit-hyperactivity and autism spectrum disorders. A number of studies have also implicated the central nervous system in the pathogenesis and pathophysiology of some functional gastrointestinal tract diseases including inflammatory bowel disease and irritable bowel syndrome (107-108). Studies have also demonstrated the beneficial effects of prenatal prebiotics and probiotic interventions in disease prevention (50-51,109) suggesting that these could be effective novel methods in preventing and/or managing neurodevelopmental disorders like autism and attention deficit hyperactivity disorder. Studies demonstrating the concurrence of postnatal neurodevelopment and the establishment of the gut microbiota provide evidence of a possible bi-directional modulation or regulation of the maturation of one another (110).

The constitution of the skin microbiome (like that of the gut) is a reflection of the synergy of the functional metabolic activity of the host tissues and resident microbes, which are greatly influenced by host behavior and the surrounding environment. In the last decade, there have been suggestions that the brain interacts with the skin, like in a “skin-brain” axis. Also, the extensive innervation of the skin has led researchers to consider it a sort of “diffuse brain” (111). The occurrence of skin disorders like psoriasis and atopic dermatitis is now being associated with the development of behavioral disorders like posttraumatic stress disorder, anxiety, and depression (112–114). Some studies have also shown that the colonization, virulence, and adhesion of skin microbes could be modulated by peptide neurohormone, neurotransmitters and steroid hormones (14). These neuropeptides such as vasoactive intestinal peptide, catecholamines, calcitonin gene-related peptide, nerve growth factor and substance P, can be secreted by some skin cells. However, while the idea of a skin-brain or brain–skin axis is plausible, the full extent of this relationship (unidirectional or bidirectional) remains largely speculative.

3.2. Neurobehavioral phenotypes and the microbiome

Gut microbiota influences on the central nervous system were first reported following observations that laxatives and/or oral antibiotics could mitigate the progression of hepatic encephalopathy (87). In the symbiotic relationship between man and microbes, the human body provides lodgings and nutrition, while microbes repay this favor by ensuring the maintenance of health/well-being, and prevention of outsider invasion (115). The roles of microbes in ensuring optimal brain development and functioning have been reported (86, 87, 90,103,1116). Also, current research is beginning to report the importance of the microbiota in modulating host behaviors (Table 3).

Table 3 Neurobehavioral phenotypes and the microbiota
Model Microbial composition/procedure Behavior Phenotype reference
Drosophila melanogaster Germ-free(GF) antibiotic-treated flies Motor ↑locomotion ↑walking speed (117)
Drosophila melanogaster Lactobacillus brevis Motor Reversal of ↑locomotion ↑walking speed (117)
Bactrocera dorsalis Enterobactereacea Foraging behavior Basal Increase in foraging (118)
Mice High-fat diet microbiota Exploratory activity, cognition ↑Locomotor activity, and ↓cognition (122)
Mice GF Locomotor activity ↑Locomotor and rearing (86)
Mice GF Stereotypy, social interaction ↑grooming, ↓social investigation, avoidance of conspecifics, (104)
Mice GF Memory ↓short term recognition memory and working memory 124
Mice Oral antibiotic depletion of microbiota Exploratory activity ↑exploratory activity (84)
Mice GF anxiety ↓Basal anxiety (87, 102)
Mice GF depression ↓ time spent immobile (125)
Mice Lactobacillus rhamnosus Anxiety, depression Increased open-arm time and decreased immobility time (128)
Mice Colonization with Citrobacter rodentum cognition ↑Stress-induced memory loss (124)
Human Age-associated dysbiosis (↑Bacteroidetes ↓Firmicutes) cognition ↓cognition (126)
Humans ↑Alcaligeneceae and Porphyromonadaceae, Veillonellaceae in cirrhotics with hepatic encephalopathy cognition Memory loss (127)
Mice Maternal high-fat diet-induced microbiota low in Social interaction ↓social interaction, ↓preference for social novelty (132)
Mice Lactobacilus reuteri Social interaction ↑social interaction, (132)
Rat Peri-conceptional use of antibiotic Social interaction, anxiety ↓social interaction, ↑anxiety (133)
Mice Use of antibiotics to deplete microbiota during adolescence Social memory ↓social memory in adulthood (106)
Mice Bacteroides fragilis Cognition, social interaction and anxiety ↑social interactions, ↓stereotypy, ↓anxiety and ↓sensorimotor deficits (3)
Rat Bifidobacterium infantis Depression decreased immobility time (92)
Mice Bifidobacterium breve Cognition ↑cognitive abilities (135)
Rats Lactobacillus helveticus and Bifidobacterium longus anxiety ↓anxiety in the conditioned defensive burying test (136)
Mice Prebiotic administration of human milk oligosaccharides 3′Sialyllactose or 6′Sialyllactose anxiety ↓stress-induced anxiety (137)
Human Lactobacillus casei Anxiety and depressive-like behaviors ↓anxiety and depressive-like behaviours (138,139)
Human Bifidobacterium longum and Lactobacillus helveticus Psychological stress indices ↓ somatization, anger, anxiety and depression (136)
Human Administration of prebiotic trans-galactooligosaccharide to irritable bowel syndrome sufferers anxiety ↓ anxiety (140)

Studies in germ-free or antibiotic-treated fruit-fly (Drosophila melanogaster) showed hyperlocomotion and increased walking speed, which was reversed by mono-colonization with the fruit-fly commensal Lactobacillus brevis (117). In another study by Akami et al (118), increase in foraging behaviors of Oriental flies (Bactrocera dorsalis) was determined by the intestinal microbiota consisting mainly of members of the Enterobacteriaceae family (118). In mammals, the importance of the microbiota in modulating dietary behaviors and metabolic profile been reported. In a study in which cultured and uncultured human fecal microbiota were transferred from weight-discordant twins (obese and lean) to germ-free mice, it was observed that the germ-free mice colonized with obese fecal microbiota showed significantly-higher weight increases compared to mice colonized with microbiota from the lean twin (66). Also, co-habitation of these two mouse groups was associated with a transformation of the metabolic profile of the obese microbiota mouse groups to a lean-like state (66). There have been reports associating obesity and related dysmetabolism with an increase in the incidence of mood disorders, depression and cognitive decline (119-120). The possibility of replicating these features in murine models of obesity (121) has allowed researchers to examine the role of diet and dysmetabolism on neurobehavior (122-123). Bruce-Keller and colleagues (122) reported that mice colonized with high-fat diet microbiota (with a preponderance of bacteria from the phylum Firmicutes and a depletion of Bacteroidetes) had alteration in exploratory activity, cognition and stereotypic behavior compared to controls (122).

Diaz Heijtz et al. (86) reported increased locomotor activity and rearing behaviours in germ-free mice, while Desbonnet et al (104) reported evidence of increased self-grooming and alteration in social interaction evidenced by avoidance of conspecifics, decreased preferences for novel conspecifics, and decreased time spent in social investigation (104). In another study using germ-free mice Gareau et al (124) reported impairment of short-term recognition memory and working memory (124), while Zheng et al (125) reported antidepressant-like response (125). Gareau et al (124) also observed that colonization of mice with pathogenic bacteria Citrobacter rodentum was associated with accentuation of stress–induced memory loss (124).

In humans, there have been reports of a loss of microbial diversity with aging (with a predominance of Bacteroidetes and a decrease in Firmicutes) and the presence of an age-associated cognitive decline in the same age group (126). In a study examining gut microbial diversity of cirrhotics with or without hepatic encephalopathy (HE), it was reported that (compared to healthy controls), cirrhotics had a preponderance of commensals from the phyla Enterobacteriaceae, Alcaligeneceae, and Fusobacteriaceae, with low concentrations of Lachnospiraceae and Ruminococcaceae (127). However, in cirrhotics (with HE) a higher concentration of Alcaligeneceae and Porphyromonadaceae Veillonellaceae was observed, which correlated positively with cognitive-deficits, high levels of pro-inflammatory markers (TNF-α, IL-2, IL=6 and IL-13) and endotoxemia (127).

Oral antibiotics–induced microbiota depletion in mice have also been reported to transiently alter the microbial composition of the gut, resulting in an increase in exploratory behavior and an increase in the hippocampal expression of brain- derived neurotropic factor (84). These changes did not occur with intraperitoneal administration of the antibiotics (84). A few studies have also reported that in germ-free mice, the basal behavior was anxiolysis (86, 102) compared to the expected aversion for open spaces (a protective feature) in mice with normal gut microbiota. In the germ-free mice, this alteration in anxiety-related behaviors was accompanied by an increase in the expression of brain derived neurotropic factor (BDNF) in the hippocampus (dentate gyrus), a decrease in the expression N-methyl-D-aspartate receptor subunit in the central amygdala, and a decrease in serotonin receptor expression by the dentate gyrus region of the hippocampus (102). In another study, Bravo e al (128) reported that long-term treatment of healthy mice with Lactobacillus rhamnosus probiotic was associated with anxiolysis in the elevated plus-maze and decreased immobility time in the forced swim test; there was also a significant decrease in the levels of stress-induced corticosterone levels and region-specific increase in the expression of γ-amino butyric acid receptor mRNA. These features were however abolished with vagotomy (128), suggesting that these effects were vagus nerve dependent.

Currently, the role of the gut microbiota in social interaction and the development of social behavior has being studied extensively (123,129–132), using either germ free mice, or following colonisation with probiotic (132). Peri-conceptional exposure to antibiotics has also been shown to alter social behavior and induce anxiety (133). The use of antibiotics to deplete mouse gut microbiota during adolescence was associated with impairment of social memory in adulthood (106), features which corresponded with a reduction in mRNA levels of vasopressin and oxytocin in the mouse hypothalamus (106). In general, alterations in social memory and neurobehavior could also be attributed to the ability of gut microbiota to modulate myelination of neurons in the prefrontal cortex (134). Hoban et al. (134) also reported evidence of abnormal hypermyelination of axons in male germ-free mice (134). Studies in the mouse model of autism have also revealed that oral treatments with Bacteroides fragilis were associated with improvements in social interactions, stereotypic behaviors, anxiety-related behaviors and sensorimotor deficits (3).

The role of the microbiota in the modulation of neurobehavior and/or brain neurochemistry has also been demonstrated by studies using prebiotics or probiotic interventions. In rodents, probiotic intervention with Bacteroides infantis was associated with decreased immobility time in the forced swim paradigm (92). Buffington et al. (132) demonstrated that a maternal diet that is high in fat caused microbiota dysbiosis, deficits in social interaction, preference for social novelty and a decrease in the neuronal oxytocin immunoreactivity in the paraventricular nucleus of the hypothalamus in the maternal high-fat diet offspring, compared to normal diet fed controls. However, there was restoration of behavioral deficits, microbiota dysbiosis and oxytocin immunoreactivity following reconstitution with Lactobacillus reuteri probiotic (132), demonstrating that these deficits were possibly due to low-concentrations of Lactobacillus reuteri in mouse pups (132). Colonization with specific members of the Bifidobacterium breve species have been associated with alteration of the fatty-acid composition of the brain in favor of fatty-acids that improve of cognitive abilities (135). Also, in rats, probiotic formulations of Lactobacillus helveticus and Bifidobacterium longus were associated with a decrease in anxiety-related behaviors measured in the conditioned defensive burying test (136). The use of prebiotic intervention to promote selective proliferation of commensal bacteria has also been reported to modulate neurobehavior. Tarr et al (137) reported that a diet rich in human milk oligosaccharides 3′Sialyllactose or 6′Sialyllactose reduced stress-induced anxiety-like behavior in the open field and light/dark preference paradigms (137).

In humans, the benefits of probiotic intervention on the modulation of behavior have also been demonstrated. In healthy adults with baseline depressive symptoms or patients with chronic fatigue syndrome, the probiotic administration of Lactobacillus casei was associated with improvement in symptoms of depression and anxiety respectively (138-139). Probiotic formulations of Bifidobacterium longum and Lactobacillus helveticus administered to otherwise healthy subjects was also associated with a decrease in psychological distress including somatization, anger, anxiety and depression, using relevant clinical indices (136). Administration of prebiotic trans-galactooligosaccharide to irritable bowel syndrome sufferers was associated with significant reduction in anxiety and also an increase in the concentration of bifidobacteria in fecal samples (140).

A number of other studies have also reported the possible influences behavioral modification can exert on microbial density or diversity (141-142). Exposure to social disruption stress in rodents was associated with a decrease in the relative abundance of bacteria of the genus Bacteroides, and an increase in those belonging to the genus Clostridium. There was also an associated increase in the circulating levels of interleukin 6 (IL-6) and monocyte chemo-attractant protein (MCP)-1 (142) Increased levels of IL-6 and MCP-1 have also been observed to correlate positively with increased concentration of bacteria from 3 genera including Coprococcus spp, Pseudobutyrivibrio sp, and Dorea spp (142). In mice exposed to chronic restraint stress, a significant increase in Citrobacter rodentium colonization with an associated increase in the gene expression for tumor necrosis factor alpha in colonic tissue has been demonstrated (141). Accumulating evidence of the ability of gut microbiota to influence neurobehaviour and the alteration of microbiota diversity and density due to influences from neurobehavioural modifications further confirm the possible existence of a bidirectional communication that allows brain signals to influence sensory, motor and secretory functions of the gut, while visceral messages from the gut can in turn influence neurobehavior, social behavior and brain chemistry (123).

Also the relationships that link the gut microbiome, skin microbiome, and the brain could be crucial in understanding the contributions of bacteria to the development of odor cues, which is known to contain important information about the host’s social relationships and social behaviors. Studies have shown that the intensity of the human odor and/or its chemical composition is closely-linked to the skin’s microbial diversity and composition (143–145). The interaction of the skin microbiota, the sweat glands, and the brain is important in the creation of human odors that allows kith recognition of kin, the detection of fear in animal prey, and sexual behaviors in animals (146). There have been reports suggesting that Staphylococcus epidermidis which is part of the normal skin microbial flora has the ability to degrade leucine content of sweat causing foot odor. Also, colonization of the plantar skin by Bacillus subtilis has been associated with strong foot odor (143). Absence of axillary odor has also been linked to paucity of gram positive bacteria (145).

3.2.1. Signaling pathways/mediators and molecular alterations involved in behavior/microbiota interactions

The communication between the gut microbiota and the brain affects behavior. This communications occurs via neural, immune and endocrine pathways that connect the microbiota to the central nervous system (147). The interactions (Table 4) between microbiota and brain alter neurobehavior, signal pathways, neurotransmitters and hormones (Figure 1); it also modulates the expression of receptor mRNA in different regions of the brain (86, 90, 102, 124, 147).

Figure 1

Showing how the gut microbiota via neural, homonal or endocrine mechanisms modulatesbrain function and chemistry.

Table 4 Microbiota-mediated neurochemical and molecular alterations
Model Microbial Composition/Mediators Brain region/tissue Neurochemical/Molecular alteration Reference
Mice Lactobacillus rhamnosus Hippocampus, amygdala, LC Prefrontal cortex, cingulated gyrus GABAAα2 mRNA ↓ GABAB1b mRNA ↓ GABAAα2 mRNA ↑GABAB1b mRNA (128)
Mice GF Hippocampus serotonin and 5-hydroxyindoleacetic (103)
Mice GF Dentate gyrus serotonin receptor mRNA (102)
Mice GF Dentate gyrus Cornus ammonis 1 region of the hippocampus, amygdala and cingulate cortex Hippocampus BDNF ↑BDNF ↑neurogenesis (102) (86, 90, 124) (149)
Mice GF Cortex Cortex and hippocampus Central amygdala N-methyl-D-aspartate receptor; the NR1 ↓NR2a ↓NR2b (90) (90) (102)
Mice GF Striatum Hippocampus ↓ nerve growth factor-inducible clone A ↓Gopamine receptor ↑dopamine receptor (86) (86)
Mice GF Cerebral cortex ↑Dopamine (150)
Mice Antibiotic-induced depletion of microbiota Hippocampus ↑BDNF (84)
Mice Trichuris muri Hippocampus ↓ BDNF mRNA (151)
Mice Citrobacter roduntum CA1 region of the hippocampus ↓BDNF expression (124)
Mice Bifidobacterium infantis Frontal cortex Amygdala Paraventricular nucleus ↓5–hydroxyl indole acetic acid ↓3,4-Dihydroxyphenylacetic acid ↓cFos expression (92) (90)
Mice GF Hypothalamus ↑corticotropin-releasing factor gene expression and protein levels ↑Plasma ACTH (90)
Rats Bifidobacterium pseudocatenulatum Bifidobacterium animalis subsp lactis and Propionibacterium jensenii Hypothalamus ↓dopamine and adrenaline ↑ACTH (reversal of maternal-separation stress induced derangements) reversal of maternal-separation stress induced biochemical derangements and microbial density (157) (155)
Rats IBS microbiota Colonic colonic genes involved in glucocorticoid receptor signaling (156).
Mice GF or mice deficient of SCFAs receptor FFAR2a (GPR43) Microglia Fcgr2β, Mapk8, IL-1α, and Cd86., Janus kinase 3 and the signal transducer and activator of transcription 1 (Stat1Nfkbiα) and the central microglia transcription and survival factors (Sfpi1 and Csf1r) (166)
Mice GF Hippocampus,hypothalamus ↑Apoptosis, (153)
Mice GF Prefrontal cortex Hypermyelination, overexpression of myelin-related genes (134)

Neural pathways involve efferents from the central nervous system to the gut, and afferents from the gut to the central nervous system. Efferent signals that are conducted from the central nervous system to the gut modulate gut motility, secretion, and permeability, which in turn affect microbiota composition. The efferent pathway relies on neurotransmitters such as acetylcholine. The afferent signals are transmitted from the gut to the central nervous system via the vagus nerve which is capable of recognizing microbial products and cellular components. The changes that had been observed in the brain include region-dependent alterations in levels of gamma-aminobutyric acid (GABA) mRNA gene expression (128). Bravo et al (128) reported that colonization with Lactobacillus rhamnosus was associated with a decrease in the expression of GABAB1b mRNA in the amygdala, hippocampus and locus coeruleus; with an increase in the prefrontal cortex and cingulate cortex. The expression of GABAAα2 mRNA was decreased in the cortical region, and increased in the hippocampus/amygdala (128). The role of GABA in maintaining central excitation/inhibition balance has been reported (148). Studies examining the impact of gut microbiota on neurotransmitters concentration in germ-free mice have reported that hippocampal concentrations of serotonin and 5-hydroxyindoleacetic acid (metabolite of serotonin) were elevated compared to control (103); while a decrease in the expression of the serotonin receptor mRNA was observed in the dentate gyrus (102). The expression of brain derived neurotropic factor (BDNF) increased in the dentate gyrus (102), and decreased in the cornus ammonis (CA) 1 region of the hippocampus, amygdale and cingulated cortex (86, 90, 124). While the content and expression of BDNF is modulated by the gut microbiota, brain derived neurotropic factor itself regulates neurogenesis, supporting the result of a recent study that demonstrated increased hippocampal neurogenesis in adult germ-free mice (149). There was also reduced expression of different subunits of the N-methyl-D-aspartate receptor; the NR1 in the cortex (90), NR2a in the cortex and hippocampus (90) and NR2b in the central amygdale (102). Diaz Heijtz et al (86) reported a decrease in the expression of nerve growth factor-inducible clone A and dopamine receptor in the striatum, and an increase expression of dopamine receptor in the hippocampus of germ–free mice (86).

Antibiotic-induced depletion of gut microbiota was associated with increased expression of BDNF in the hippocampus (84). Matsumoto et al (150) also reported increase in dopamine concentration in the cerebral cortex of germ-free mice compared to mice colonized by normal microbial flora (150), which supports the increased exploratory activity reported by previous studies (86, 102). Colonization of mice with non-invasive parasite Trichuris muri was associated with colonic inflammation and anxiety-like behavior, while in-situ hybridization revealed a decrease in hippocampal expression of BDNF mRNA (151). Gareau et al (124) also reported decreased BDNF expression in mice colonized with citrobacter roduntum following induction of stress (124). Intervention with probiotic Bifidobacterium infantis was associated with a decrease in the frontal cortex concentration of 5– hydroxyl indole acetic acid and amygdaloid cortex concentration of 3, 4-Dihydroxyphenylacetic acid, metabolites of serotonin and dopamine respectively (92); and a decrease in the expression of cFos in the paraventricular nucleus (92), neurotransmitter modulations that could be related to the antidepressant effects observed in the forced swim test. A number of these studies confirm previous reports associating hippocampal depletion of neurotransmitters with the development of depression and depressive-like symptoms (152). Neuronal apoptosis has also been linked to the gut microbiota. In the hypothalamus and some regions of the hippocampus, increased apoptosis was observed in neonatal germ-free mice when compared with control mice, this was also accompanied by increase microbial count and density (153). Neuron myelination and myelin plasticity have also been reported to be impacted by the gut microbiota; in germ-free mice, evidence of hypermyelination of the axons in the prefrontal cortex as well as over-expression of myelin-related genes have been reported (134).

The endocrine pathway involves the hypothalamo-pituitary-adrenal (HPA) axis, and the secretion of hormonally-active peptides such as peptide YY. Changes in the HPA axis response can be triggered by impulses originating from the brain (e.g. due to stress), or from the gut microbes who can positively or negatively regulate the HPA axis (90). In germ-free mice, restraint stress has been observed to cause increased HPA response, leading to an elevation of hypothalamic corticotropin-releasing factor gene expression/protein levels, a decrease in cortical/hippocampal brain-derived neurotrophic factor, and an elevation of plasma adrenocorticotropic hormone (ACTH)/corticosteron (90). However, colonization by specific microbiota at certain periods in their life can either normalize their HPA response to stress, or make them to continue to show exaggerated responses (90). This influence has been suggested to profoundly impact behaviors like anxiety, attention, and certain aspects of memory.

Probiotic intervention (from postnatal day 2 to 21) with a strain of Bifidobacterium pseudocatenulatum reversed neuroendocrine and neurobehavioural changes induced by maternal deprivation induced stress (154). The neuroendocrine response included an attenuation of stress-induced increase in corticosterone levels, and catecholamine levels in the hypothalamus. A down-regulation of maternal-deprivation stress–induced increase in intestinal levels of interferon gamma and catecholamine (dopamine and adrenaline) was also demonstrated (154).

Maternal probiotic supplementation with Bifidobacterium animalis subsp lactis and Propionibacterium jensenii has also been shown to reverse the changes induced by maternal-separation stress or the adult-stress protocol on biochemical parameters and microbial density (155). In rats pups exposed to maternal-separation stress, an increase in plasma levels of ACTH and fecal concentration of enterococci, Escherichia coli, and clostridia has been shown. In adult animals exposed to stress, an increase in both ACTH and corticosterone levels was observed; there was also a decrease in the density of Bifidobacteria, and an increase in Escherichia coli and Bacteroides (155). Transplantation of fecal microbiota from patients with irritable bowel syndrome into germ free mice has also associated with anxiety-related behavior, increased gastrointestinal transit time, alteration in the intestinal barrier (156). There was also increased expression of colonic genes involved in glucocorticoid receptor signaling (156).

In line with the bidirectional communication that occurs in the gut-brain axis, the neurons of the autonomic nervous system and neuroendocrine mediators carry outputs from the brain to the gut. Biologically active peptides including peptide YY (PYY), neuropeptide Y and pancreatic polypeptide are expressed at distinct levels of the gut–brain axis (157). Peptide YY and pancreatic polypeptide are expressed exclusively by the endocrine cells of the digestive system, while NPY is expressed at all levels of the gut–brain and brain–gut axis (157). PYY is synthesized in the gut by enteroendocrine cells with the G protein-coupled receptor FFAR3 (Gpr41) which sense dietary proteins/fats, and microbial-derived short-chain fatty acids (158). The PYY that is transported to the brain affects feeding behavior, while in the GIT, it by triggers satiety, reduces food intake, and slows the GI motility (158).

The immune pathway involves the influence of microbial antigens and metabolites on the immune system, which in turn affects the central nervous system. It is known that specific microbes and their products can use different mechanisms to influence the development and functions of particular subsets of immune cells (159). Microbial antigens might trigger the immune system and contribute to the pathogenesis of neurodegenerative disorders such as multiple sclerosis which has profound behavioral components (160-161). On the other hand, the microbial metabolites that affect immune function include chemical mediators such as small chain fatty-acids (acetic acid, propionic acid and butyric acid) and tryptophan metabolites. The short chain fatty acids (SCFAs) are mainly produced by large-intestinal bacterial fermentation of the complex carbohydrates. They modulate gut motility, enhance intestinal epithelium integrity, increase mucus production, promote regulatory T-cells, and inactivate nuclear factor kappa B (162-164). In the gastrointestinal system, SCFAs induce the release of neuropeptides and hormones including glucagon-like peptide 1 and PYY from intestinal enteroendocrine cells (165), via the activation of the G protein-coupled receptors (GPRs; GPR41, GPR43, and GPR109A), and also by acting as epigenetic regulators. SCFAs modulate gene expression through the inhibition of histone deacetylases.

In the central nervous system, SCFAs are important in the modulation of microglial maturation and homeostasis (166). The microglia in germ-free mice exhibited global defects including altered cell size, immature phenotypes and impaired innate immune response. These features were also evident in mice deficient of SCFAs receptor FFAR2a (GPR43) (166). Genome-wide mRNA studies revealed significant differences in mRNA profiles of microglia genes of germ-free compared to specific-pathogen (SPF) mice (166). A total of 198 genes were observed to be downregulated while about 173 genes were unregulated in microglia from germ-free mice, compared controls (166).A number of the downregulated genes were genes that had been linked to cell activation including Fcgr2β, Mapk8, IL-1α, and Cd86. Also diminished were genes such as Janus kinase 3 and the signal transducer and activator of transcription 1 (Stat1), which are linked to signaling of type I IFN receptors (166). Upregulated genes in the germ-free mice microglia include a number of genes responsible for the inhibition of transcription (Nfkbiα) and the central microglia transcription and survival factors (Sfpi1 and Csf1r) (166), which are usually downregulated in microglia (167-168).

Bacterial populations in the gut have also been reported to be capable of producing hormones and/or neurotransmitters similar to that produced by humans (169). Members of the Bifidobacterium species produce gamma-amino butyrate (GABA), Lactobacillus species secrete acetylcholine and GABA, while Escherichia produce norepinephrine, dopamine and serotonin. Serotonin is also produced by the Streptococcus and Enterococcus spp, while dopamine and norepinephrine is produced by members of the Bacillus species. These neurotransmitters that are produced by microbes could also impact neurobehaviour (169-170).

3.3. Brain disorders and the microbiota

The microbiota contributes to the maintenance of the central nervous system structure and function through direct interactions between the enteric nervous system and the gut microbiota (171-172), or indirectly through the modulation of endocrine, immunological, and neural pathways (173). Gut microbiome dysbiosis has been associated with the development of neuropsychiatric disorders, including bipolar disorder, anxiety disorder, schizophrenia, autism, chronic fatigue syndrome, major depressive disorder and stress (174–176). Neurodegenerative diseases like Alzheimer’s disease, Parkinson’s disease, and stroke have also been linked to microbiome dysbiosis (177).

A number of studies (Table 5) have demonstrated the presence of alterations in specific microbes (or their metabolites) with the occurrence of central nervous system disorders (55,178–182). Following the examination of fecal samples of children with autism, Finegold and colleagues (183-184) reported low levels of microbes within the phyla Actinobacteria and Firmicutes, and an increase in the concentration of Proteobacteria and Bacteroidetes, when compared to healthy controls (183-184). The presence of high loads of Bacteroidetes, Bacteroides vulgatus, and Desulfovibrio species were observed in the feces of severely- autistic children (183). Song et al. (178) reported an abundance of Clostridia in children with autism who had gastrointestinal disorders (178), while Kang et al (185) reported evidence of a decrease in microbial diversity and the presence of lower levels of Coprococcus, Prevotella, and Veillonellaceae in stool samples of autistic children (185). In the valproic acid model of autism in rodents, alterations in the concentrations of Firmicutes and Bacteroidetes occurred alongside autism-like social behaviors (186). In children with attention deficit hyperactivity disorder, an increase in fecal Bifidobacterium has also been reported (187). In Rete syndrome, alteration of the gut microbiota has also been described; and examination of stool samples from patients revealed a decrease in microbial diversity and an increase in levels of Bifidobacterium, Bacteroidaceae, Actinomyces, Clostridium Enterococcus, Lactobacillus, Escherichia, and Shigella species (180). In patients with major depressive disorder (MDD), there have been reports suggesting a preponderance of Bacteroidetes phyla and a decrease in Lachnospiraceae (188). Jiang et al. (189) reported an increase in the concentration of Proteobacteria, Bacteroidetes and Actinobacteria in depressed patients; while Zheng et al (125) reported a relative abundance of Actinobacteria and a decrease in Bacteroidetes in patients with MDD compared to healthy controls (125). A cause-effect relationship has also been demonstrated between the microbiota dysbiosis and mood disorders, using fecal transplantation studies (125, 190). Transplantation of fecal microbiota from depressed patients to rodents was associated with the induction of depression-like behavior (125). Anxiety and anhedonia were also reported in rats colonized with fecal microbiota from depressed patients (190).

Table 5 Brain disorders and alterations in microbiota density
Disorder/Behavioural alterations Host Microbial composition Reference
Autism spectrum disorder Humans ↑Clostridia, Bacteroidetes, Bacteroides vulgates, Desulfovibrio species, Proteobacteria ↓ Actinobacteria and Firmicutes ↓microbial diversity ↓Coprococcus, Prevotella and Veillonellaceae species (178,183–185)
Autism spectrum disorder Murine ↑Bacteroidetes, ↓ Firmicutes (186)
Attention deficit hyperactivity disorder Human ↑ Bifidobacterium (187)
Rete syndrome Human ↑Bifidobacterium, Actinomyces, Clostridia Enterococcus, Lactobacillus, Escherichia, and Shigella species (180)
Major depressive disorder Human ↑Bacteroidetes, ↓ Lachnospiraceae (188).
Major depressive disorder Human ↑ Proteobacteria, Bacteroidetes and Actinobacteria (189)
Major depressive disorder Human ↑ Actinobacteria ↓ Bacteroidetes (125)
Parkinson’s disease Human ↓Enterobacteria and Prevotella species (179)
Alzheimer’s disease Murine ↓Akkermansia and Allobaculum ↑Rikenellaceae (182)
Anxiety, anhedonia and depressivelike behaviour Rat Fecal transplantation of microbiota from depressed patients (125, 190)
ASD: Autism spectrum disorders, ADHD: Attention deficit Hyperactivity disorder, MDD: Major depressive disorder, PD: Parkinson’s disease, AD: Alzheimer’s disease.

A number of neurodegenerative disorders have been associated with microbiota dysbiosis. In Parkinson’s disease, it had been suggested that the pathogenesis may have actually arisen from the abnormalities in the gut before spreading via the gut-brain axis to the brain (191). However, what is generally obvious is increasing evidence in support of the existence of a brain-gut interaction (107,110), and possibly the involvement of the brain-skin cross talk (192) in the pathophysiology of a number of neurodegenerative disorders. Examination of fecal samples from Parkinson’s disease patients revealed a decrease in the population of Enterobacteria and Prevotella species (179). The transfer of microbiota from new-onset, treatment-naïve PD patients to groups of germ-free mice has also been shown to lead to worsening motor deficits (193). In Alzheimer’s disease, an association has been established between the presence of beta-amyloid plaques and microbial dysbiosis (182). Alzheimer’s disease has also been associated with alterations in microbial diversity and population. Studies using transgenic mice revealed Alzheimer’s disease-induced alteration of microbial diversity, with alterations in the populations of Bacteroidetes and Firmicutes; specifically lowering the population of Akkermansia and Allobaculum, and increasing the population of Rikenellaceae (182).

In recent times, the brain-skin crosstalk has been proposed, as reported by studies that have demonstrated over-activation of neuropeptides in skin disorders like psoriasis and atopic dermatitis; and revealed skin conditions related to neuroimmunological stress (194–197). Mijouin et al. (113) also reported that exposure of the skin microbe Bacillus cereus to substance P was associated with increased cytotoxicity, increased caspase-1 activity, and morphological changes in the actin cytoskeleton (113). In neurodegenerative disorders like Parkinson’s disease and Alzheimer’s disease, direct alteration of the skin microbial density or diversity is rarely reported. Instead, the evidence of skin involvement would include an increase in melanoma and non-melanoma skin cancers in Parkinson’s disease (198), or altered skin physiology in Alzheimer’s disease (199). However, the precise role of the skin microbiome in all these is still a subject of research.

3.4. Behavioral disorders, the microbiota, and future therapeutic implications

Based on available evidence, it is easy to deduce that manipulation of the microbiota may be a therapeutic tool in the management or prevention of behavioural disorders. Along this line, the study of how this knowledge can be used specifically for the management of behavioral disorders is an advancing field that holds a lot of promise. Human behavioral disorders that have been linked to microbiota dysbiosis include ADHD, ASD, schizophrenia, bipolar disorders, major depression, anxiety disorders, obsessive-compulsive disorder, eating disorders; and specific neurological disorders such as AD, PD, and MS (200). However, beyond the recognition or identification of a link, the questions is to what extent can manipulation of the microbiome be used in the therapy of these disorders, and what the future holds if therapy is steered in this direction. Also, while the global market for agents that may be of benefit in the prevention or management of dysbiosis continues to expand, the extent to which they may be used in the clinical management of behavioral disorders remains largely unknown. Again, the multifactorial origin of a number of disorders implies that targeting the microbiome alone is not likely to be enough for therapy. However, some scientists are in support of the idea of targeting the microbiota as novel therapeutic options in the management of neuropsychiatric and neurodegenerative diseases. Their support for the exploration of this novel approach to therapy has been backed by a number of theories, including: a) the “old friend” hypothesis, b) the gut microbiota hypothesis, and c) the leaky-gut theory.

The old friend theory suggests that a symbiotic relationship has existed between man and microbes from time immemorial (201), this relationship is crucial to the maintenance of the balance of nature as well as the health and well-being of humans. The transformations that have occurred as a result of modernization (leading to changes in lifestyle, diet and healthcare) have greatly reduced man’s exposure to microbes, which has in turn altered man’s immune system and brain development (202). The gut microbiota theory of brain disorders initially arose from the results of a 2002 study that demonstrated resistance to disease and improvement in health and brain function following the feeding of Lactobacillus–fermented fodder to pigs; and subsequently from accumulating evidence linking the gut microbiota to neurobehavioural dysfunction and the development of mood disorders and mental illness (202-203). The leaky gut theory is based on the hypothesis that the human body has two main barriers, the blood brain barrier (BBB) and the gut barrier. The gut barrier regulates the movement of nutrient and signal molecules into the body, and prevents the entry of microorganisms. However, the BBB with the aid of tight junctions regulates the entry and exit of molecules in central nervous system. The maintenance of the integrity of these barriers is critical to health and wellbeing (204-205). The importance of the gut microbiota in the development of the BBB was demonstrated by Braniste et al. (206), when they reported that in germ-free mice an increase in the permeability of the BBB was observed when compared to mice with normal gut flora. This was observed to persist to adulthood and was also associated with a decrease in the expression of tight junction proteins (occludin and claudin-5) (206).

These theories portray the important role the microbiota–gut-brain axis could potentially play in the prevention of and development of drugs for the management of brain disorders; also, accumulating preclinical evidence continues to support these views. A number of methods directed at improving the microbiota such as the transplantation of fecal microbiota, and the use of probiotics or prebiotics have been shown to have significant benefits in health and disease. In children with ASD, seven-to-eight weeks of microbiota transfer therapy altered the gut microbiome and improved both gastrointestinal and behavioral symptoms (207). These improvements were associated with increased overall bacterial diversity and an increase in the abundance of Bifidobacterium, Prevotella, and Desulfovibrio species (207). Also, several systematic reviews have indicated that probiotics can effectively improve mood in humans (208-209), offering promise in the management of depression. In a clinical trial involving patients with mania, 24 weeks of adjunctive probiotics (using Lactobacillus rhamnosus strain GG and Bifidobacterium animalis subsp. lactis strain Bb12) were shown to prevent psychiatric re-hospitalizations after discharge, especially in individuals with elevated levels of baseline systemic inflammation (210).

From the foregoing, while it is becoming increasingly-evident that manipulations of the microbiome can be clinically-beneficial for a limited number of behavioral disorders, for the vast majority, research is yet to substantiate their benefit in humans. Therefore, more effort is needed towards conducting research in this area, especially, considering the fact that orthodox medications have not proven to be fully-satisfactory in the management of a number of such disorders.

4. CONCLUSION

The increase in the knowledge of the microbiota and how they influence brain development and behavior has revolutionized our perception of the microbes that live in and on our bodies. Presently, we are aware that the brain and the gut microbiota can communicate with each other via neural, endocrine and immunological pathways; and that these communications might be important in the pathogenesis of neurological and behavioral disorders. Also, a number of chemical mediators that act as messengers along such pathways are already being identified. However, while novel therapies that target these pathways are being investigated, only limited progress has been made in the area of microbiome manipulation for the prevention and management of behavioral/neurological disorders. As more resources are dedicated to research in this area, the coming years are likely to yield more interesting findings and possible clinical applications that might revolutionize the management of behavioral and neurological disorders.

5. ACKNOWLEDGMENTS

This research did not receive any specific grant from agencies in the public, commercial, or not-for-profit sectors. This article does not contain any studies with human participants or animals performed by any of the authors. The authors declare no conflict of interest.

References
[1]
Consortium Human Microbiome Project Structure, function, and diversity of the healthy human microbiome Nature 2012 486 207 14 DOI: 10.1038/nature11234
[2]
Rodríguez JM Murphy K Stanton RP Ross C Kober OI Juge N Avershina E Rudi K Narbad A Jenmalm MC Marchesi JR Collado MC The composition of the gut microbiota throughout life, with an emphasis on early life Microb Ecol Heal Dis 2015 26 26050 DOI: 10.3402/mehd.v26.26050
[3]
Hsiao EY McBride SW Hsien S Sharon G Hyde ER McCue T Codelli JA Chow J Reisman SE Petrosino JF Patterson PH Mazmanian SK Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders Cell 2013 155 1451 64 DOI: 10.1016/j.cell.2013.11.024
[4]
Sharon G Sampson TR Geschwind DH Mazmanian SK The Central Nervous System and the Gut Microbiome Cell 2016 167 915 32 DOI: 10.1016/j.cell.2016.10.027
[5]
Calvani R Picca A Monaco MR Lo Landi F Bernabei R Microbes E Of Microbes and Minds: A Narrative Review on the Second Brain Aging Front Med (Lausanne) 2018 5 53 DOI: 10.3389/fmed.2018.00053
[6]
Mayer EA Knight R Mazmanian SK Cryan JF Tillisch K Gut microbes, and the brain: a paradigm shift in neuroscience J Neurosci 2014 34 1540 6 DOI: 10.1523/JNEUROSCI.3299-14.2014
[7]
Bull MJ 1 NT Plummer: Part The Human Gut Microbiome in Health and Disease Integr Med Encinitas 2014 13 17 22
[8]
Ahmad OF Akbar A Microbiome, antibiotics, and irritable bowel syndrome Br Med Bull 2016 120 91 9 DOI: 10.1093/bmb/ldw038
[9]
Vaughn AR Notay M Clark AK Sivamani RK Skin-gut axis: The relationship between intestinal bacteria and skin health World J Dermatology 2017 6 52 8 DOI: 10.5314/wjd.v6.i4.52
[10]
Dash S Clarke G Berk M Jacka FN The gut microbiome and diet in psychiatry: focus on depression Curr Opin Psychiatry 2015 28 1 6 DOI: 10.1097/YCO.0000000000000117
[11]
Logan AC Jacka FN Craig JM Microbiome SL Prescott: The Health Mental Looking Back, Moving Forward with Lessons from Allergic Diseases Clin Psychopharmacol Neurosci 2016 14 131 47 DOI: 10.9758/cpn.2016.14.2.131
[12]
Prescott SL Larcombe DL Logan AC West C Burks W Caraballo L Levin M Etten EV Horwitz P Kozyrskyj A Campbell DE The skin microbiome: impact of modern environments on skin ecology, barrier integrity, and systemic immune programming World Allergy Organ J 2017 10 29 DOI: 10.1186/s40413-017-0160-5
[13]
Bowe WP Logan AC Acne vulgaris, probiotics and the gut-brain-skin axis - back to the future? Gut Pathog 2011 31 1 DOI: 10.1186/1757-4749-3-1
[14]
Perspective K Beri Stabilizing the Microbiome Skin-Gut-Brain Axis with Natural Plant Botanical Ingredients in Cosmetics Cosmet 2018 5 37 DOI: 10.3390/cosmetics5020037
[15]
Collado MC Cernada M Baüerl C Vento M Pérez-Martínez G Microbial ecology and host-microbiota interactions during early life stages Gut Microbes 2012 3 352 65 DOI: 10.4161/gmic.21215
[16]
Prince AL Antony KM Ma J Aagaard KM The microbiome and development: a mother's perspective Semin Reprod Med 2014 32 14 22 DOI: 10.1055/s-0033-1361818
[17]
Belkaid Y Tamoutounour S The influence of skin microorganisms on cutaneous immunity Nat Rev Immunol 2016 16 353 366 DOI: 10.1038/nri.2016.48
[18]
Ursell LK Haiser HJ Treuren W Van Garg N Reddivari L Vanamala J Dorrestein PC Turnbaugh PJ Knight R The intestinal metabolome: an intersection between microbiota and host Gastroenterol 2014 146 1470 6 DOI: 10.1053/j.gastro.2014.03.001
[19]
Hollister EB Gao C Versalovic J Compositional and functional features of the gastrointestinal microbiome and their effects on human health Gastroenterol 2014 146 1449 58 DOI: 10.1053/j.gastro.2014.01.052
[20]
David LA Maurice CF Carmody RN Gootenberg DB Button JE Wolfe BE Ling AV Devlin AS Varma Y Fischbach MA Biddinger SB Dutton RJ Turnbaugh PJ Diet rapidly and reproducibly alters the human gut microbiome Nature 2014 505 559 63 DOI: 10.1038/nature12820
[21]
Archie EA Tung J Social behavior and the microbiome Curr Opin Behav Sci 2015 6 28 34 DOI: 10.1016/j.cobeha.2015.07.008
[22]
Tung J Barreiro LB Burns MB Grenier JC Lynch J Grieneisen LE Altmann J Alberts SC Blekhman R Archie EA Social networks predict gut microbiome composition in wild baboons Elife 2015 16 4 DOI: 10.7554/eLife.05224
[23]
Blekhman R Goodrich JK Huang K Sun Q Bukowski R Bell JT Spector TD Keinan A Ley RE Gevers D Clark AG Host genetic variation impacts microbiome composition across human body sites Genome Biol 2015 16 191 DOI: 10.1186/s13059-015-0759-1
[24]
Claesson MJ Jeffery IB Conde S Power SE O'Connor EM Cusack S Harris HM Coakley M Lakshminarayanan B O'Sullivan O Fitzgerald GF Deane J O'Connor M Harnedy N O'Connor K O'Mahony D Sinderen D van Wallace M Brennan L Stanton C Marchesi JR Fitzgerald AP Shanahan F Hill C Ross RP O'Toole PW Gut microbiota composition correlates with diet and health in the elderly Nature 2012 488 178 84 DOI: 10.1038/nature11319
[25]
Wang Y Wang B Wu J Jiang X Tang H Nielsen OH Modulation of Gut Microbiota in Pathological States Engineering 2017 3 83 9 DOI: 10.1016/J. ENG.2017.01.013
[26]
Wang B Yao M Lv L Ling Z Li L The Human Microbiota in Health and Disease Engineering 2017 3 71 82 DOI: 10.1016/J. ENG.2017.01.008
[27]
Favier CF Vaughan EE Vos WM De Akkermans ADL Molecular monitoring of succession of bacterial communities in human neonates Appl Environ Microbiol 2002 68 219 226 DOI: 10.1128/AEM.68.1.219-226.2002
[28]
Palmer C Bik EM DiGiulio DB Relman DA Brown PO Development of the human infant intestinal microbiota PLOS Biol 2007 5 e177 DOI: 10.1371/journal.pbio.0050177
[29]
Funkhouser LJ Bordenstein SR Mom knows best: the universality of maternal microbial transmission PLOS Biol 2013 11 e1001631 DOI: 10.1371/journal.pbio.1001631
[30]
Wassenaar TM Panigrahi P Is a foetus developing in a sterile environment? Lett Appl Microbiol 2014 59 572 9 DOI: 10.1111/lam.12334
[31]
Collado MC Rautava S Aakko J Isolauri E Salminen S Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid Sci Rep 2016 6 23129 DOI: 10.1038/srep23129
[32]
Aagaard K Ma J Antony KM Ganu R Petrosino J Versalovic J The placenta harbors a unique microbiome Sci Transl Med 2014 237 237 65 DOI: 10.1126/scitranslmed.3008599
[33]
Stout MJ Conlon B Landau M Lee I Bower C Zhao Q Roehl KA Nelson DM Macones GA Mysorekar IU Identification of intracellular bacteria in the basal plate of the human placenta in term and preterm gestations Am J Obs Gynecol 2013 208 e1 7 DOI: 10.1016/j.ajog.2013.01.018
[34]
Ardissone AN Cruz DM de la Davis-Richardson AG Rechcigl KT Li N Drew JC Murgas-Torrazza R Sharma R Hudak ML Triplett EW Neu J Meconium microbiome analysis identifies bacteria correlated with premature birth PLoS One 2014 9 e90784 DOI: 10.1371/journal.pone.0090784
[35]
DiGiulio DB Diversity of microbes in amniotic fluid Semin Fetal Neonatal Med 2012 2012 17 2 11 DOI: 10.1016/j.siny.2011.10.001
[36]
Rogier EW Frantz AL Bruno ME Wedlund L Cohen DA Stromberg AJ Lessons from mother: long-term impact of antibodies in breast milk on the gut microbiota and intestinal immune system of breastfed offspring Gut Microbes 2014 5 663 8 DOI: 10.4161/19490976.2014.969984
[37]
Capone KA Dowd SE Stamatas GN Nikolovski J Diversity of the human skin microbiome early in life J Invest Dermatol 2011 131 2026 32 DOI: 10.1038/jid.2011.168
[38]
Lehtimäki J Karkman A Laatikainen T Paalanen L Hertzen L von Haahtela T Hanski I Ruokolainen L Patterns in the skin microbiota differ in children and teenagers between rural and urban environments Sci Rep 7 2017 45651 DOI: 10.1038/srep45651
[39]
Sanford JA Gallo RL Functions of the skin microbiota in health and disease Semin Immunol 2013 25 370 7 DOI: 10.1016/j.smim.2013.09.005
[40]
Blaser MJ Dominguez-Bello MG The human Microbiome before birth Cell Host Microbe 2016 20 558 560 DOI: 10.1016/j.chom.2016.10.014
[41]
Iwase T Uehara Y Shinji H Tajima A Seo H Takada K Agata T Mizunoe Y Staphylococcus epidermidis Esp inhibits Staphylococcus aureus biofilm formation and nasal colonization Nature 2010 465 346 9 DOI: 10.1038/nature09074
[42]
Clausen ML Agner T Lilje B Edslev SM Johannesen TB Andersen PS Association of Disease Severity With Skin Microbiome and Filaggrin Gene Mutations in Adult Atopic Dermatitis JAMA Dermatol 2018 154 293 300 DOI: 10.1001/jamadermatol.2017.5440
[43]
Moschen AR Wieser V Factors H Tilg: Dietary Major Regulators of the Gut's Microbiota Gut Liver 2012 6 411 6 DOI: 10.5009/gnl.2012.6.4.411
[44]
D'Argenio V Salvatore F The role of the gut microbiome in the healthy adult status Clin Chim Acta 2015 451 97 102 DOI: 10.1016/j.cca.2015.01.003
[45]
Shreiner AB Kao JY Young VB The gut microbiome in health and in disease Curr Opin Gastroenterol 2015 31 69 75 DOI: 10.1097/MOG.0000000000000139
[46]
Round JL Mazmanian SK The gut microbiota shapes intestinal immune responses during health and disease Nat Rev Immunol 2009 9 313 23 DOI: 10.1038/nri2515
[47]
Closa-Monasterolo R Gispert-Llaurado M Luque V Ferre N Rubio-Torrents C Zaragoza-Jordana M Escribano J Safety and efficacy of inulin and oligofructose supplementation in infant formula: results from a randomized clinical trial Clin Nutr 2013 32 918 27 DOI: 10.1016/j.clnu.2013.02.009
[48]
Giovannini M Verduci E Gregori D Ballali S Soldi S Ghisleni D Riva E Group PLAGOS Trial Study Prebiotic effect of an infant formula supplemented with galacto-oligosaccharides: randomized multicenter trial J Am Coll Nutr 2014 33 385 93 DOI: 10.1080/07315724.2013.878232
[49]
Kunze WA Mao YK Wang B Huizinga JD X X Ma Forsythe P Bienenstock J Lactobacillus reuteri enhances excitability of colonic ah neurons by inhibiting calcium-dependent potassium channel opening J. Cell. Mol. Med 2009 13 2261 2270 DOI: 10.1111/j.1582-4934.2009.00686.x
[50]
Rautava S Collado MC Salminen S Isolauri E Probiotics modulate host-microbe interaction in the placenta and fetal gut: a randomized, double-blind, placebo-controlled trial Neonatol 2012 102 178 84 DOI: 10.1159/000339182
[51]
Toh ZQ Anzela A Tang ML Licciardi PV Probiotic therapy as a novel approach for allergic disease Front Pharmacol 2012 3 171 DOI: 10.3389/fphar.2012.00171
[52]
Grice EA Segre JA The human microbiome: our second genome Ann Rev Genom Hum Genet 2012 13 151 170 DOI: 10.1146/annurev-genom-090711-163814
[53]
Clarke SF Murphy EF O'Sullivan O Lucey AJ Humphreys M Hogan A Hayes P O'Reilly M Jeffery IB Wood-Martin R Kerins DM Quigley E Ross RP O'Toole PW Molloy MG Falvey E Shanahan F Cotter PD Exercise and associated dietary extremes impact on gut microbial diversity Gut 2014 63 1913 1920 DOI: 10.1136/gutjnl-2013-306541
[54]
McBride ME Duncan WC Knox JM The environment and the microbial ecology of human skin Appl Env Microbiol 1977 33 603 608
[55]
Ghaisas S Maher J Kanthasamy A Gut microbiome in health and disease: linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases Pharmacol Ther 2016 158 52 62 DOI: 10.1016/j.pharmthera.2015.11.012
[56]
Baumgart DC Carding SR Inflammatory bowel disease: cause and immunobiology Lancet 2007 369 1627 40 DOI: 10.1016/S0140-6736(07)60750-8
[57]
Krogius-Kurikka L Lyra A Malinen E Aarnikunnas J Tuimala J Paulin L Mäkivuokko H Kajander K Palva A Microbial community analysis reveals high level phylogenetic alterations in the overall gastrointestinal microbiota of diarrhoea-predominant irritable bowel syndrome sufferers BMC Gastroenterol 2009 9 95 DOI: 10.1186/1471-230X-9-95
[58]
Turnbaugh PJ Hamady M Yatsunenko T Cantarel BL Duncan A Ley RE Sogin ML Jones WJ Roe BA Affourtit JP Egholm M Henrissat B Heath AC Knight R Gordon JI A core gut microbiome in obese and lean twins Nature 2009 457 480 4 DOI: 10.1038/nature07540
[59]
Scotti E Bou'e S Sasso GL Zanetti F Belcastro V Poussin C Sierro N Battey J Gimalac A Ivanov NV Hoeng J Exploring the microbiome in health and disease: implications for toxicology Toxicol Res Appl 2017 1 1 37 DOI: 10.1177/2397847317741884
[60]
Shukla SD Budden KF Neal R Microbiome effects on immunity, health and disease in the lung Clin Trans Immunol 2017 6 e133 DOI: 10.1038/cti.2017.6
[61]
Carding S Verbeke K Vipond DT Corfe BM Owen LJ Dysbiosis of the gut microbiota in disease Microb Ecol Heal Dis 2015 2 26191 DOI: 10.3402/mehd.v26.26191
[62]
Ley RE Turnbaugh PJ Klein S Gordon JI Microbial ecology: human gut microbes associated with obesity Nature 2006 444 1022 3 DOI: 10.1038/4441022a
[63]
Ley RE Backhed F Turnbaugh P Lozupone CA Knight RD Gordon JI Obesity alters gut microbial ecology Proc Natl Acad Sci USA 2005 102 11070 5 DOI: 10.1073/pnas.0504978102
[64]
Backhed F Manchester JK Semenkovich CF Gordon JI Mechanisms underlying the resistance to diet-induced obesity in germ-free mice Proc Natl Acad Sci USA 2007 104 979 84 DOI: 10.1073/pnas.0605374104
[65]
Larsen N Vogensen FK Berg FW van den Nielsen DS Andreasen AS Pedersen BK Al-Soud WA Sørensen SJ Hansen LH Jakobsen M Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults PLoS One 2010 5 e9085 DOI: 10.1371/journal.pone.0009085
[66]
Ridaura VK Faith JJ Rey FE Cheng J Duncan AE Kau AL Griffin NW Lombard V Henrissat B Bain JR Muehlbauer MJ Ilkayeva O Semenkovich CF Funai K Hayashi DK Lyle BJ Martini MC Ursell LK Clemente JC Van Treuren W Walters WA Knight R Newgard CB Heath AC Gordon JI Gut microbiota from twins discordant for obesity modulate metabolism in mice Science 2013 341 1241214 DOI: 10.1126/science.1241214
[67]
Tilg H Moschen AR Microbiota and diabetes: an evolving relationship Gut 2014 63 1513 21 DOI: 10.1136/gutjnl-2014-306928
[68]
Shin NR Lee JC Lee HY Kim MS Whon TW Lee MS Bae JW An increase in the Akkermansia spp population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014 63 727 35 DOI: 10.1136/gutjnl-2012-303839
[69]
Palma G De Nadal I Medina M Donat E Ribes-Koninckx C Calabuig M Sanz Y Intestinal dysbiosis and reduced immunoglobulin-coated bacteria associated with coeliac disease in children BMC Microbiol 2010 10 63 DOI: 10.1186/1471-2180-10-63
[70]
Kong HH Segre JA The Molecular Revolution in Cutaneous Biology: Investigating the Skin Microbiome J Invest Dermatol 2017 137 e119 e122 DOI: 10.1016/j.jid.2016.07.045
[71]
Silva PES Reis MP Ávila MP Dias MF Costa PS Suhadolnik MLS BG BG Kunzmann Carmo AO Kalapotakis E Chartone-Souza E Nascimento AMA Insights into the skin microbiome dynamics of leprosy patients during multi-drug therapy and in healthy individuals from Brazil Sci Rep 2018 8 8793 DOI: 10.1038/s41598-018-27074-0
[72]
Silva PE Costa PS Ávila MP Suhadolnik ML Reis MP Salgado AP Lima MF Chartone-Souza E Nascimento AM Leprous lesion presents enrichment of opportunistic pathogenic bacteria Springerplus 2015 4 187 DOI: 10.1186/s40064-015-0955-1
[73]
McDowell A Patrick S Eishi Y P Lambert Eady A Propionibacterium acnes in human health and disease Biomed Res Int 2013 2013 493564 DOI: 10.1155/2013/493564
[74]
Hawkey PM The growing burden of antimicrobial resistance J Antimicrob Chemother 2008 62 1 9 DOI: 10.1093/jac/dkn241
[75]
Hall BG Barlow M Evolution of the serine beta-lactamases: past, present, and future Drug Resist Updat 2004 7 111 3 DOI: 10.1016/j.drup.2004.02.003
[76]
Ubeda C Taur Y Jenq RR Equinda MJ Son T Samstein M Viale A Socci ND Brink MR van den Kamboj M Pamer EG Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans J Clin Invest 2010 120 4332 41 DOI: 10.1172/JCI43918
[77]
Tyakht AV Kostryukova ES Popenko AS Belenikin MS Pavlenko AV Larin AK Karpova IY Selezneva OV Semashko TA Ospanova EA Babenko VV Maev IV Cheremushkin SV Kucheryavy YA Shcherbakov PL Grinevich VB Efimov OI Sas EI Abdulkhakov RA Abdulkhakov SR Lyalyukova EA Livzan MA Vlassov VV Sagdeev RZ Tsukanov VV Osipenko MF Kozlova IV Tkachev AV Sergienko VI Alexeev DG Govorun VM Human gut microbiota community structures in urban and rural populations in Russia Nat Commun 2013 4 2469 DOI: 10.1038/ncomms3469
[78]
Madan JC Salari RC Saxena D Davidson L O'Toole GA Moore JH Sogin ML Foster JA Edwards WH Palumbo P Hibberd PL Gut microbial colonisation in premature neonates predicts neonatal sepsis Arch Dis Child Fetal Neonatal Ed 2012 97 456 62 DOI: 10.1136/fetalneonatal-2011-301373
[79]
Jernberg C Lofmark S Edlund C Jansson JK Long-term ecological impacts of antibiotic administration on the human intestinal microbiota Isme J 2007 1 55 66 DOI: 10.1038/ismej.2007.3
[80]
Ferreyra JA Wu KJ Hryckowian AJ Bouley DM Weimer BC Sonnenburg JL Gut microbiota-produced succinate promotes C. difficile infection after antibiotic treatment or motility disturbance Cell Host Microbe 2014 16 770 7 DOI: 10.1016/j.chom.2014.11.003
[81]
Ng KM Ferreyra JA Higginbottom SK Lynch JB Kashyap PC Gopinath S Naidu N Choudhury B Weimer BC Monack DM Sonnenburg JL Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens Nature 2013 502 96 9 DOI: 10.1038/nature12503
[82]
Möhle L Mattei D Heimesaat MM Bereswill S Fischer A Alutis M French T Hambardzumyan D Matzinger P Dunay IR Wolf SA Ly6Chi Monocytes Provide a Link between Antibiotic-Induced Changes in Gut Microbiota and Adult Hippocampal Neurogenesis Cell Reports 2016 15 1945 56 DOI: 10.1016/j.celrep.2016.04.074
[83]
Branton WG Ellestad KK Maingat F Wheatley BM Rud E Warren RL Holt RA Surette MG Power C Brain microbial populations in HIV/AIDS: α-proteobacteria predominate independent of host immune status PLoS One 2013 8 e54673 DOI: 10.1371/journal.pone.0054673
[84]
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 Gastroenterol 2011 141 599 DOI: 10.1053/j.gastro.2011.04.052
[85]
Collins S Intestinal microbiota and the brain-gut axis In: Finlay B, editor. Microbiota Agents Heal. Dis. The Biomedical & Life Sciences Collection 2011 London Henry Stewart Talks Ltd
[86]
Heijtz R Daiz Wang S Anuar F Qian Y Björkholm B Samuelsson A Hibberd ML Forssberg H Pettersson S Normal gut microbiota modulates brain development and behavior Proc Natl Acad Sci USA 2011 108 3047 52 DOI: 10.1073/pnas.1010529108
[87]
Al-Asmakh M Anuar F Zadjali F Rafter J Pettersson S Gut microbial communities modulating brain development and function Gut Microbes 2012 3 366 73 DOI: 10.4161/gmic.21287
[88]
Fuente-Nunez C de la Meneguetti BT Franco OL Lu TK Neuromicrobiology: How Microbes Influence the Brain ACS Chem Neurosci 2018 9 141 50 DOI: 10.1021/acschemneuro.7b00373
[89]
Geschwind DH Rakic P Cortical evolution: judge the brain by its cover Neuron 2013 80 633 47 DOI: 10.1016/j.neuron.2013.10.045
[90]
Sudo N Chida Y Aiba Y Sonoda J Oyama N Yu XN Kubo C Koga Y Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice J Physiol 2004 558 263 75 DOI: 10.1113/jphysiol.2004.063388
[91]
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 43 164 174 DOI: 10.1016/j.jpsychires.2008.03.009
[92]
Desbonnet L Garrett L Clarke G Kiely B Cryan J Dinan T Effects of the probiotic bifidobacterium infantis in the maternal separation model of depression Neurosci 2010 170 1179 1188 DOI: 10.1016/j.neuroscience.2010.08.005
[93]
Migliarini S Pacini G Pelosi B Lunardi G Pasqualetti M Lack of brain serotonin affects postnatal development and serotonergic bneuronal circuitry formation Mol. Psychiat 2013 18 1106 1118 DOI: 10.1038/mp.2012.128
[94]
Clemente JC Ursell LK Parfrey LW R R Knight The impact of the gut microbiota on human health: an integrative view Cell. 16 2012 148 6 1258 70 DOI: 10.1016/j.cell.2012.01.035
[95]
Cerdó T Ruíz A Suárez A Campoy C Probiotic, Prebiotic, and Brain Development Nutrients 2017 9 E1247 DOI: 10.3390/nu9111247
[96]
Tau GZ Peterson BS Normal development of brain circuits Neuropsychopharmacol 2010 35 147 68 DOI: 10.1038/npp.2009.115
[97]
Jost T Lacroix C Braegger CP Rochat F Chassard C Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding Environ Microbiol. 2014 16 2891 904 DOI: 10.1111/1462-2920.12238
[98]
Collins SM Surette M Bercik P The interplay between the intestinal microbiota and the brain Nat Rev Microbiol 2012 10 735 42 DOI: 10.1038/nrmicro2876
[99]
Luna RA Foster JA Gut brain axis: diet microbiota interactions and implications for modulation of anxiety and depression Curr Opin Biotechnol 2015 32 35 41 DOI: 10.1016/j.copbio.2014.10.007
[100]
Dinan TG Cryan JF The Microbiome-Gut-Brain Axis in Health and Disease Gastroenterol Clin North Am 2017 46 77 89 DOI: 10.1016/j.gtc.2016.09.007
[101]
Lu J Synowiec S Lu L Yu Y Bretherick T Takada S Yarnykh V Caplan J Caplan M Claud EC Drobyshevsky A Microbiota influence the development of the brain and behaviors in C57BL/6J mice PLoS One 2018 13 e0201829 DOI: 10.1371/journal.pone.0201829
[102]
Neufeld KA Kang N Bienenstock J Foster JA Effects of intestinal microbiota on anxiety-like behavior Commun Integr Biol 2011 4 492 4 DOI: 10.4161/cib.15702
[103]
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 18 666 73 DOI: 10.1038/mp.2012.77
[104]
Desbonnet L Clarke G Shanahan F Dinan TG Cryan JF Microbiota is essential for social development in the mouse Mol Psychiatry 2014 19 146 8 DOI: 10.1038/mp.2013.65
[105]
Vuong HE Yano JM Fung TC Hsiao EY The Microbiome and Host Behavior Annu Rev Neurosci 2017 40 21 49 DOI: 10.1146/annurev-neuro-072116-031347
[106]
Desbonnet L Clarke G Traplin A O'Sullivan O Crispie F Moloney RD Cotter PD Dinan TG Cryan JF Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour Brain Behav Immun 2015 48 165 73 DOI: 10.1016/j.bbi.2015.04.004
[107]
Chang L Sundaresh S Elliott J Anton PA Baldi P Licudine A Mayer M Vuong T Hirano M Naliboff BD Ameen VZ Mayer EA Dysregulation of the hypothalamic-pituitary-adrenal HPA) axis in irritable bowel syndrome Neurogastroenterol Motil 2009 l21 149 159 DOI: 10.1111/j.1365-2982.2008.01171.x
[108]
Stasi C E Orlandelli Role of the brain-gut axis in the pathophysiology of Crohn's disease Dig Dis 2008 26 156 166 DOI: 10.1159/000116774
[109]
Zhang GQ Hu HJ Liu CY Zhang Q Shakya S Li ZY Probiotics for Prevention of Atopy and Food Hypersensitivity in Early Childhood: A PRISMA-Compliant Systematic Review and Meta-Analysis of Randomized Controlled Trials Medicine (Baltimore) 2016 95 e2562 DOI: 10.1097/MD.0000000000002562
[110]
Aziz Q Thompson DG Brain-gut axis in health and disease Gastroenterol 1998 114 559 578 DOI: 10.1016/S0016-5085(98)70540-2
[111]
Urpe M Buggiani G Lotti T Stress and psychoneuroimmunologic factors in dermatology Dermatol Clin 2005 23 609 17 DOI: 10.1016/j.det.2005.05.017
[112]
Pavlovic S Daniltchenko M Tobin DJ Hagen E Hunt SP Klapp BF Arck PC Peters EM Further exploring the brain-skin connection: stress worsens dermatitis via substance P-dependent neurogenic inflammation in mice J Investig Dermatol 2008 128 434 446 DOI: 10.1038/sj.jid.5701079
[113]
Mijouin L Hillion M Ramdani Y Jaouen T Duclairoir-Poc C Follet-Gueye ML Lati E Yvergnaux F Driouich A Lefeuvre L Farmer C Misery L Feuilloley MG Effects of a skin neuropeptide (substance P) on cutaneous microflora PLoS One 2013 8 e78773 DOI: 10.1371/journal.pone.0078773
[114]
Gupta MA Jarosz P Gupta AK Posttraumatic stress disorder PTSD) and the dermatology patient Clin Dermatol 2017 35 260 266 DOI: 10.1016/j.clindermatol.2017.01.005
[115]
Geurts L Neyrinck AM Delzenne NM Knauf C Cani PD Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics Benef Microbes 2013 2 1 15 DOI: 10.3920/BM2012.0065
[116]
Chen X D'souza R Hong ST The role of gut microbiota in the gut-brain axis: current challenges and perspective Protein Cell 2013 4 403 14 DOI: 10.1007/s13238-013-3017-x
[117]
Schretter CE Vielmetter J Bartos I Marka Z Marka S Argade S Mazmanian SK A gut microbial factor modulates locomotor behaviour in Drosophila Nature 2018 563 402 6 DOI: 10.1038/s41586-018-0634-9
[118]
Akami M Andongma AA Zhengzhong C Nan JKK Jurkevitch E Niu CY Diptera B Yuval B Intestinal bacteria modulate the foraging behavior of the oriental fruit fly Bactrocera dorsalis Tephritidae) PLoS One 2019 14 e0210109 DOI: 10.1371/journal.pone.0210109
[119]
Riederer P Korczyn AD Ali SS Bajenaru O Choi MS Chopp M Dermanovic-Dobrota V Grünblatt E Jellinger KA Kamal MA Kamal W Leszek J TM Sheldrick-Michel Mushtaq G Meglic B Natovich R Pirtosek Z Rakusa M Salkovic-Petrisic M Schmidt R Schmitt A Sridhar GR Vécsei L Wojszel ZB Yaman H Zhang ZG The diabetic brain and cognition J Neural Trans 2017 124 1431 54 DOI: 10.1007/s00702-017-1763-2
[120]
Sanderlin AH Todem D Bozoki AC Obesity and co-morbid conditions are associated with specific neuropsychiatric symptoms in mild cognitive impairment Front Aging Neurosci 2017 9 164 DOI: 10.3389/fnagi.2017.00325
[121]
Andre C Dinel AL Ferreira G Laye S cognition N Castanon Diet-induced obesity progressively alters behavior anxiety-like behavior lipopolysaccharide-induced depressive-like focus on brain indoleamine 2,3-dioxygenase activation Brain Behav Immun 2014 41 10 21 DOI: 10.1016/j.bbi.2014.03.012
[122]
Bruce-Keller AJ Salbaum JM Luo M Blanchard ET Taylor CM Welsh DA Bertoud HR Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity Biol Psychiatry 2015 77 607 15 DOI: 10.1016/j.biopsych.2014.07.012
[123]
Montiel-Castro AJ Gonzalez-Cervantes RM Bravo-Ruiseco G Pacheco-Lopez G The microbiota-gut-brain axis: neurobehavioral correlates, health, and sociality Front Integr Neurosci 2013 7 70 DOI: 10.3389/fnint.2013.00070
[124]
Gareau MG Wine E Rodrigues DM Cho JH Whary MT Philpott DJ Macqueen G Sherman PM Bacterial infection causes stress-induced memory dysfunction in mice Gut 2011 60 307 10 DOI: 10.1136/gut.2009.202515
[125]
Zheng P Zeng B Zhou C Liu M Fang Z Xu X Zeng L Chen J Fan S Du X Zhang X Yang D Yang Y Meng H Li W Melgiri ND Licinio J Wei H Xie P Gut microbiome remodeling induces depressive like behaviors through a pathway mediated by the host's metabolism Mol Psychiatr 2016 21 786 96 DOI: 10.1038/mp.2016.44
[126]
Claesson MJ Cusack S O'sullivan O Greene-diniz R DeWeerd H Flannery E Marchesi JR Falush D Dinan T Fitzgerald G Stanton C Sinderen D van O'Connor M Harnedy N O'Connor K Henry C O'Mahon D Fitzgerald AP Shanahan F Twomey C Hill C Ross RP O'Toole PW Composition, variability, and temporal stability of the intestinal microbiota of the elderly Proc Natl Acad Sci U S A 2011 108 4586 91 DOI: 10.1073/pnas.1000097107
[127]
Bajaj JS Ridlon JM Hylemon PB Thacker LR Heuman DM Smith S Sikaroodi M Gillevet PM Linkage of gut microbiome with cognition in hepatic encephalopathy Am J Physiol Gastrointest Liver Physiol 2012 302 G168 75 DOI: 10.1152/ajpgi.00190.2011
[128]
Bravo A 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 16050 16055 DOI: 10.1073/pnas.1102999108
[129]
Troyer K Microbes, herbivory and the evolution of social-behavior J Theor Biol 1984 106 157 69 DOI: 10.1016/0022-5193(84)90016-X
[130]
Lombardo MP Access to mutualistic endosymbiotic microbes: an underappreciated benefit of group living Behav Ecol Sociobiol 2008 62 479 DOI: 10.1007/s00265-007-0428-9
[131]
Dinan TG Stilling RM Stanton C Cryan JF Collective unconscious: how gut microbes shape human behavior J Psychiatr Res 2015 63 1 9 DOI: 10.1016/j.jpsychires.2015.02.021
[132]
Buffington SA Prisco GV Di Auchtung TA Ajami NJ Petrosino JF Costa-Mattioli M Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring Cell 2016 165 1762 75 DOI: 10.1016/j.cell.2016.06.001
[133]
Degroote S Hunting DJ Baccarelli AA Takser L Maternal gut and fetal brain connection: increased anxiety and reduced social interactions in Wistar rat offspring following peri-conceptional antibiotic exposure Prog. Neuropsychopharmacol. Biol Psychiatry 2016 71 76 82 DOI: 10.1016/j.pnpbp.2016.06.010
[134]
Hoban AE Stilling RM Ryan FJ Shanahan F Dinan TG Claesson NJ arke G C Cryan JF Regulation of prefrontal cortex myelination by the microbiota Transl Psychiatry 2016 6 e774 DOI: 10.1038/tp.2016.42
[135]
Wall R Marques TM O'sullivan O Ross RP Shanahan F Quigley EM Dinan TG Kiely B Fitzgerald GF Cotter PD Fouhy F Stanton C Contrasting effects of Bifidobacterium breve NCIMB 702258 and Bifidobacterium breveDPC 6330 on the composition of murine brain fatty acids and gut microbiota Am J Clin Nutr 2012 95 1278 87 DOI: 10.3945/ajcn.111.026435
[136]
Messaoudi M Lalonde R Violle N Javelot H Desor D Nejdi A 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 105 755 764 DOI: 10.1017/S0007114510004319
[137]
Tarr AJ Galley JD Fisher SE Chichlowski M Berg BM Bailey MT The prebiotics 3′ sialyllactose and 6′ sialyllactose diminish stressor-induced anxiety-like behavior and colonic microbiota alterations: Evidence for effects on the gut-brain axis Brain Behav Immun 2015 50 166 177 DOI: 10.1016/j.bbi.2015.06.025
[138]
Benton D Williams C Brown A Impact of consuming a milk drink containing a probiotic on mood and cognition Eur J Clin Nutr 2007 61 355 361 DOI: 10.1038/sj.ejcn.1602546
[139]
Rao AV Bested AC Beaulne TM Katzman MA Iorio C Berardi JM A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome Gut Pathog. 2009 1 6 DOI: 10.1186/1757-4749-1-6
[140]
Silk DB Davis A Vulevic J Tzortzis G Gibson GR Clinical trial: the effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome Aliment Pharmacol Ther 2009 29 508 518 DOI: 10.1111/j.1365-2036.2008.03911.x
[141]
Bailey MT Dowd SE Parry NM Galley JD Schauer DB Lyte M Stressor exposure disrupts commensal microbial populations in the intestines and leads to increased colonization by Citrobacter rodentium Infect Immun 2010 78 1509 19 DOI: 10.1128/IAI.00862-09
[142]
Bailey MT Dowd SE Galley JD Hufnagle AR Allen RG Lyte M Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation Brain Behav Immun 2011 25 397 407 DOI: 10.1016/j.bbi.2010.10.023
[143]
Ara KM Hama S Akiba K Koike K Okisaka T Hagura T Kamiya T Tomita F Foot odour due to microbial metabolism and its control Can J Microbiol 2006 52 357 364 DOI: 10.1139/w05-130
[144]
Taylor D Daulby S Grimshaw G James JM Vaziri A Characterization of the microflora of the human axilla Int J Cosmet Sci 2003 25 137 145 DOI: 10.1046/j.1467-2494.2003.00181.x
[145]
Xu YS Dixon R Brereton H Soini M Novotny K Trebesius I Comparison of human axillary odour profiles obtained by gas chromatography/mass spectrometry and skin microbial profiles obtained by denaturing gradient gel electrophoresis using multivariate pattern recognition Metabolomics 2007 3 427 37 DOI: 10.1007/s11306-007-0054-6
[146]
EA Archie ecology Theis KR. Animal behaviour meets microbial Animal Behav 82: 425e436 2011 DOI: 10.1016/j.anbehav.2011.05.029
[147]
Cox LM Weiner HL Microbiota Signaling Pathways that Influence Neurologic Disease Neurotherapeutics 2018 15 135 145 DOI: 10.1007/s13311-017-0598-8
[148]
Rudolph U Möhler H Analysis of GABAA receptor function and dissection of the pharmacology of benzodiazepines and general anesthetics through mouse genetics Annu Rev Pharmacol Toxicol 2004 44 475 98 DOI: 10.1146/annurev.pharmtox.44.101802.121429
[149]
Ogbonnaya ES Clarke G Shanahan F Dinan TG Cryan JF O O'Leary Adult Hippocampal Neurogenesis Is Regulated by the Microbiome Biol Psychiatry 2015 78 e7 9 DOI: 10.1016/j.biopsych.2014.12.023
[150]
Matsumoto M Kibe R Ooga T Aiba Y Sawaki E Koga Y Benno Y Cerebral low-molecular metabolites influenced by intestinal microbiota: a pilot study Front Syst Neurosci. 2013 7 9 DOI: 10.3389/fnsys.2013.00009
[151]
Bercik P Verdu EF Foster JA Macri J Potter M Huang X Malinowski P Jackson W Blennerhassett P Neufeld KA Lu J Khan WI Corthesy-Theulaz I Cherbut C Bergonzelli GE Collins SM Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice Gastroenterol 2010 139 2102 2112 DOI: 10.1053/j.gastro.2010.06.063
[152]
Campbell S Macqueen G G The role of the hippocampus in the pathophysiology of major depression J Psychiatry Neurosci 2004 29 417 26
[153]
Castillo-Ruiz A Mosley M George AJ Mussaji LF Fullerton EF Ruszkowski EM Jacobs AJ Gewirtz AT Chassaing B Forger NG The microbiota influences cell death and microglial colonization in the perinatal mouse brain Brain Behav Immun 2018 67 218 229 DOI: 10.1016/j.bbi.2017.08.027
[154]
Moya-Pérez A Luczynski P Renes IB Wang S Borre Y Ryan C Anthony Knol J Stanton C Dinan TG Cryan JF Intervention strategies for cesarean section-induced alterations in the microbiota-gut-brain axis Nutr Rev 2017 75 225 240 DOI: 10.1093/nutrit/nuw069
[155]
Barouei J Moussavi M Hodgson DM Effect of maternal probiotic intervention on HPA axis, immunity and gut microbiota in a rat model of irritable bowel syndrome PLOS ONE 2012 7 e46051 DOI: 10.1371/journal.pone.0046051
[156]
Palma G De Lynch MD Lu J Dang VT Deng Y Jury J Umeh G Miranda PM Pastor M Pigrau Sidani S Pinto-Sanchez MI Philip V McLean PG Hagelsieb MG Surette MG Bergonzelli GE Verdu EF Britz-McKibbin P Neufeld JD Collins SM Bercik P Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice Sci Transl Med. 2017 9 379 DOI: 10.1126/scitranslmed.aaf6397
[157]
Holzer P Reichmann F Farzi A Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis Neuropeptides 2012 46 261 74 DOI: 10.1016/j.npep.2012.08.005
[158]
Ichimura A Hirasawa A Hara T Tsujimoto G Free fatty acid receptors act as nutrient sensors to regulate energy homeostasis Prostaglandins Other Lipid Mediat 2009 89 82 8 DOI: 10.1016/j.prostaglandins.2009.05.003
[159]
Kamada N Seo S-U Chen GY Núñez G Role of the gut microbiota in immunity and inflammatory disease Nat Rev Immunol 2013 13 321 335 DOI: 10.1038/nri3430
[160]
Ivanov II Atarashi K Manel N Brodie EL Shima T Karaoz U Wei D Goldfarb KC Santee CA Lynch SV Tanoue T Imaoka A Itoh K Takeda K Umesaki Y Honda K Littman DR Induction of intestinal Th17 cells by segmented filamentous bacteria Cell 2009 139 485 498 DOI: 10.1016/j.cell.2009.09.033
[161]
Ivanov II Littman DR Segmented filamentous bacteria take the stage Mucosal Immunol 2010 3 209 212 DOI: 10.1038/mi.2010.3
[162]
Usami M Kishimoto K Ohata A Miyoshi M Aoyama M Fueda Y Kotani J Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells Nutr Res 2008 28 321 328 DOI: 10.1016/j.nutres.2008.02.012
[163]
Furusawa Y Obata Y Fukuda S Endo TA Nakato G Takahashi D Nakanishi Y Uetake C Kato K Kato T Takahashi M Fukuda NN Murakami S Miyauchi E Hino S Atarashi K Onawa S Fujimura Y Lockett T Clarke JM Topping DL Tomita M Hori S Ohara O Morita T Koseki H Kikuchi J Honda K Hase K Ohno H Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells Nature 2013 504 446 450 DOI: 10.1038/nature12721
[164]
Chang PV Hao L Offermanns S Medzhitov R The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition Proc Natl Acad Sci U S A 2014 111 2247 2252 DOI: 10.1073/pnas.1322269111
[165]
Rooks MG Garrett WS Gut microbiota, metabolites and host immunity Nat Rev Immunol 2016 16 341 352 DOI: 10.1038/nri.2016.42
[166]
Erny D Angelis ALH de Jaitin D Wieghofer P Staszewski O David E Keren-Shaul H Mahlakoiv T Jakobshagen K Buch T Schwierzeck V Utermöhlen O Chun E Garrett WS McCoy KD Diefenbach A Staeheli P Stecher B Amit I Prinz M Host microbiota constantly control maturation and function of microglia in the CNS Nat Neurosci 2015 18 965 977 DOI: 10.1038/nn.4030
[167]
Kierdorf K Prinz M Factors regulating microglia activation Front Cell Neurosci 2013 7 44 DOI: 10.3389/fncel.2013.00044
[168]
Kierdorf K Erny D Goldmann T Sander V Schulz C Perdiguero EG Wieghofer P Heinrich A Riemke P Hölscher C Müller DN Luckow B Brocker T Debowski K Fritz G Opdenakker G Diefenbach A Biber K Heikenwalder M Geissmann F Rosenbauer F Prinz M Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways Nat Neurosci 2013 16 273 80 DOI: 10.1038/nn.3318
[169]
Galland L The gut microbiome and the brain J Med Food 2014 17 1261 72 DOI: 10.1089/jmf.2014.7000
[170]
Cryan JF Dinan TG Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour Nat Rev Neurosci 2012 13 701 12 DOI: 10.1038/nrn3346
[171]
Sudo N Role of microbiome in regulating the HPA axis and its relevance to allergy Chem Immunol Allerg 2012 98 63 175 DOI: 10.1159/000336510
[172]
Dickerson F Severance E Yolken R The microbiome, immunity, and schizophrenia and bipolar disorder Brain Behav Immun 2017 62 46 52 DOI: 10.1016/j.bbi.2016.12.010
[173]
Hyland NP Cryan JF Microbe-host interactions: influence of the gut microbiota on the enteric nervous system Dev Biol 2016 417 182 187 DOI: 10.1016/j.ydbio.2016.06.027
[174]
Slyepchenko A Maes M Jacka FN Gut microbiota, bacterial translocation, and interactions with diet: pathophysiological links between major depressive disorder and non-communicable medical comorbidities Psychother Psychosom 2017 86 31 46 DOI: 10.1159/000448957
[175]
Hsiao EY Gastrointestinal issues in autism spectrum disorder Harvard Rev Psych 2014 22 104 111 DOI: 10.1097/HRP.0000000000000029
[176]
Wang Y Kasper LH The role of microbiome in central nervous system disorders Brain Behav Immun 2014 38 1 12 DOI: 10.1016/j.bbi.2013.12.015
[177]
Lee HU McPherson ZE Tan B Host-microbiome interactions: the aryl hydrocarbon receptor and the central nervous system J Mole Med 2017 95 29 39 DOI: 10.1007/s00109-016-1486-0
[178]
Song Y Liu C Finegold SM Real-time PCR quantitation of clostridia in feces of autistic children Appl Env Microbiol 2004 70 6459 6465 DOI: 10.1128/AEM.70.11.6459-6465.2004
[179]
Scheperjans F Aho V Pereira PA Gut microbiota are related to Parkinson's disease and clinical phenotype Move Dis Off J Move Dis Soc 2015 30 350 358 DOI: 10.1002/mds.26069
[180]
Strati F Cavalieri D Albanese D Altered gut microbiota in Rett syndrome Microbiome 2016 4 41 DOI: 10.1186/s40168-016-0185-y
[181]
Strati F Cavalieri D Albanese D New evidences on the altered gut microbiota in autism spectrum disorders Microbiome 2017 5 24 DOI: 10.1186/s40168-017-0242-1
[182]
Marungruang T Harach N Duthilleul N Reduction of abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota Sci Rep 2017 7 41802 DOI: 10.1038/srep46856
[183]
Finegold SM Dowd SE Gontcharova V Liu C Henley KE Wolcott RD Youn E Summanen PH Granpeesheh D Dixon D Liu M Molitoris DR Green JA 3rd Pyrosequencing study of fecal microflora of autistic and control children Anaerobe 2010 16 444 53 DOI: 10.1016/j.anaerobe.2010.06.008
[184]
Finegold SM Downes J Summanen PH Microbiology of regressive autism Anaerobe 2012 18 260 262 DOI: 10.1016/j.anaerobe.2011.12.018
[185]
Kang DW J JG Park Ilhan ZE Wallstrom G LaBaer J Adams JB Krajmalnik-Brown R Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children PLoS One 2013 8 e68322 DOI: 10.1371/journal.pone.0068322
[186]
Theije CG De Wopereis H Ramadan M Eijndthoven T van Lambert J Knol J Garssen J Kraneveld AD Oozeer R Altered gut microbiota and activity in a murine model of autism spectrum disorders Brain Behav Immun 2014 37 197 206 DOI: 10.1016/j.bbi.2013.12.005
[187]
Aarts E Ederveen THA Naaijen J Gut microbiome in ADHD and its relation to neural reward anticipation PLoS One 2017 12 e0183509 DOI: 10.1371/journal.pone.0183509
[188]
Naseribafrouei A Hestad K Avershina E Sekelja M Linløkken A Wilson R Rudi K Correlation between the human fecal microbiota and depression Neurogastroenterol Motil 2014 26 1155 62 DOI: 10.1111/nmo.12378
[189]
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 48 186 94 DOI: 10.1016/j.bbi.2015.03.016
[190]
Kelly JR Borre Y Brien C O' Patterson E Aidy S El 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 82 109 18 DOI: 10.1016/j.jpsychires.2016.07.019
[191]
Braak H Vos RA de Bohl J Gastric alpha-synuclein immunoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Parkinson's disease-related brain pathology Neurosci Lett 2006 396 67 72 DOI: 10.1016/j.neulet.2005.11.012
[192]
Planken A Kurvits L Reimann E Kadastik-Eerme L Kingo K Kõks S Taba P disease Looking beyond the brain to improve the pathogenic understanding of Parkinson's implications of whole transcriptome profiling of Patients' skin BMC Neurol 2017 17 6 DOI: 10.1186/s12883-016-0784-z
[193]
Sampson TR Debelius JW Thron T Janssen S Shastri GG Ilhan ZE Challis C Schretter CE Rocha S Gradinaru V Chesselet MF Keshavarzian A Shannon KM Krajmalnik-Brown R Wittung-Stafshede P Knight R Mazmanian SK Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease Cell 2016 167 1469 1480 DOI: 10.1016/j.cell.2016.11.018
[194]
Fortune DG Richards HL Griffiths CE Main CJ Psychological stress, distress and disability in patients with psoriasis: consensus and variation in the contribution of illness perceptions, coping and alexithymia Br J Clin Psychol 2002 41 157 74 DOI: 10.1348/014466502163949
[195]
Biró T Ko MC Bromm B Wei ET Bigliardi P Siebenhaar F Hashizume H Misery L Bergasa NV Kamei C Schouenborg J Roostermann D Szabó T Maurer M Bigliardi-Qi M Meingassner JG Hossen MA Schmelz M Steinhoff M How best to fight that nasty itch - from new insights into the neuroimmunological, neuroendocrine, and neurophysiological bases of pruritus to novel therapeutic approaches Exp Dermatol 2005 14 225 40 DOI: 10.1111/j.0906-6705.2005.0321a.x
[196]
Zhan M Zheng W Jiang Q Zhao Z Wang Z Wang J Zhang H He S Upregulated expression of substance P SP) and NK1R in eczema and SP-induced mast cell accumulation Cell Biol Toxicol 2017 33 389 405 DOI: 10.1007/s10565-016-9379-0
[197]
Lee WJ Jung HD Lee HJ Kim BS Lee SJ Kim DW Influence of substance-P on cultured sebocytes Arch Dermatol Res 2008 300 311 316 DOI: 10.1007/s00403-008-0854-1
[198]
Liu R Gao X Lu Y Chen H Meta-analysis of the relationship between Parkinson's disease and melanoma Neurology 2011 76 2002 9 DOI: 10.1212/WNL.0b013e31821e554e
[199]
Cecchi C Fiorillo C Sorbi S Latorraca S Nacmias B Bagnoli S Nassi P Liguri O Oxidative stress and reduced antioxidant defenses in peripheral cells from familial Alzheimer's patients Free Radic Biol Med 2002 33 1372 9 DOI: 10.1016/S0891-5849(02)01049-3
[200]
Bastiaanssen TFS Cowan CSM Claesson MJ Dinan TG Cryan JF Making Sense of … the Microbiome in Psychiatry International J Neuropsychopharmacol 2019 22 37 52 DOI: 10.1093/ijnp/pyy067
[201]
Rook GA Regulation of the immune system by biodiversity from the natural environment: an ecosystem service essential to health Proc Natl Acad Sci U S A. 2013 110 18360 7 DOI: 10.1073/pnas.1313731110
[202]
Liang S Wu X Psychology F Jin: Gut-Brain Rethinking Psychology From the Microbiota-Gut-Brain Axis Front Integr Neurosci 2018 12 33 DOI: 10.3389/fnint.2018.00033
[203]
Liang S Wang T Hu X Li W Jin F Wang L Microorganism and behavior and psychiatric disorders Adv. Psychol. Sci. 2012 20 75 97 DOI: 10.3724/SP. J.1042.2012.00075
[204]
Borre YE O'Keeffe GW Clarke G Stanton C Dinan TG Cryan JF Microbiota and neurodevelopmental windows: implications for brain disorders Trends Mol Med 2014 20 509 18 DOI: 10.1016/j.molmed.2014.05.002
[205]
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 DOI: 10.3389/fncel.2015.00392
[206]
Braniste V Al-Asmakh M Kowal C Anuar F Abbaspour A Tóth M Korecka A Bakocevic N Ng LG Kundu P Gulyás B Halldin C Hultenby K Nilsson H Hebert H Volpe BT Diamond B Pettersson S The gut microbiota influences blood-brain barrier permeability in mice Sci Transl Med 2014 6 263ra158 DOI: 10.1126/scitranslmed.3009759
[207]
Kang D-W Adams JB Gregory AC Borody T Chittick L Fasano A Khoruts A Geis E Maldonado JI McDonough-Means S Pollard EL Roux S Sadowsky MJ Lipson KS Sullivan MB Caporaso JG Krajmalnik-Brown R Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study Microbiome 2017 5 10 DOI: 10.1186/s40168-016-0225-7
[208]
Huang J Wang K Hu J Effect of probiotics on depression: a systematic review and meta-analysis of randomized controlled trials Nutrients 2016 8 483 DOI: 10.3390/nu8080483
[209]
Wallace CJK Milev R The effects of probiotics on depressive symptoms in humans: a systematic review Ann Gen Psychiatry 2017 16 14 DOI: 10.1186/s12991-017-0138-2
[210]
Dickerson F Adamos M Katsafanas E Khushalani S Origoni A Savage C Schweinfurth L Stallings C Sweeney K Goga J Yolken RH Adjunctive probiotic microorganisms to prevent rehospitalisation in patients with acute mania: A randomised controlled trial Bipolar Disord 2018 20 614 21 DOI: 10.1111/bdi.12652
Share
Back to top