Next Article in Journal
Sedimentary Ancient DNA (sedaDNA) Reveals Fungal Diversity and Environmental Drivers of Community Changes throughout the Holocene in the Present Boreal Lake Lielais Svētiņu (Eastern Latvia)
Previous Article in Journal
A Large Spatial Survey of Colistin-Resistant Gene mcr-1-Carrying E. coli in Rivers across Taiwan
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress

1
School of Food and Advanced Technology, Massey University, Palmerston North 4442, New Zealand
2
The New Zealand Institute for Plant and Food Research Limited, Palmerston North 4410, New Zealand
3
Riddet Institute, Massey University, Palmerston North 4442, New Zealand
4
Smart Foods Innovation Centre of Excellence, AgResearch, Palmerston North 4442, New Zealand
5
Department of Human Nutrition, Otago University, Dunedin 9016, New Zealand
6
High-Value Nutrition National Science Challenge, Auckland 1145, New Zealand
*
Author to whom correspondence should be addressed.
Microorganisms 2021, 9(4), 723; https://doi.org/10.3390/microorganisms9040723
Submission received: 23 March 2021 / Revised: 29 March 2021 / Accepted: 29 March 2021 / Published: 31 March 2021
(This article belongs to the Section Gut Microbiota)

Abstract

:
Episodes of depression and anxiety commonly follow the experience of stress, however not everyone who experiences stress develops a mood disorder. Individuals who are able to experience stress without a negative emotional effect are considered stress resilient. Stress-resilience (and its counterpart stress-susceptibility) are influenced by several psychological and biological factors, including the microbiome-gut-brain axis. Emerging research shows that the gut microbiota can influence mood, and that stress is an important variable in this relationship. Stress alters the gut microbiota and plausibly this could contribute to stress-related changes in mood. Most of the reported research has been conducted using animal models and demonstrates a relationship between gut microbiome and mood. The translational evidence from human clinical studies however is rather limited. In this review we examine the microbiome-gut-brain axis research in relation to stress resilience.

1. Introduction

Vulnerability to developing mood disorders such as anxiety disorders and depression depends on a mixture of genetic and environmental factors [1,2,3] and of the environmental factors, stress plays a significant role. Childhood adversity increases susceptibility to developing mood disorders later in life, and episodes of major depressive disorder are commonly preceded by psychosocial stress [4]. Not everyone who experiences stress develops a mood disorder [5]. Stress resilience is the ability to experience stressful events without the development of chronic elevated stress (psychological and/or biological) and associated changes in emotional behavior [6,7]. Stress susceptibility is related to psychological factors such as passive coping skills [7] and high emotional reactivity [8] but is also associated with biological factors such as hypo- or hyper- responsiveness of the stress response system (SRS), gonadal sex hormones, central and peripheral immune activation, and glucocorticoid resistance [6,7]. Interestingly, coping style is influenced by the level of biological stress response and associated neuroendocrine systems [7], and active coping under stress is increased with anti-depressant drugs [7]. This suggests that interventions which affect the biological side of stress resilience may be a useful adjunct to current treatments and preventative care.
The gut microbiome is a biological factor which is emerging as a possible influencer of stress resilience. The broad influence of the gut microbiota on human health, including psychiatric health, has begun to be realised and understood over the last decade [9]. The gut-brain axis is the bidirectional communication between the gut and the central nervous system that plays an important role in maintaining neural, hormonal and immunological homeostasis [10]. With emerging evidence showing that the gut microbiome can influence symptoms of depression and anxiety, the gut microbiome is now seen as a key component of this cross-talk between the gut and the brain and the term has been extended to microbiome-gut-brain axis (MGBA). Stress also alters the gut microbiota [11,12,13,14,15], and the effects of early life stress on the microbiota may extend to adulthood [16]. It is therefore plausible that changes in the gut microbiota due to stress at least partially mediate the onset of stress-related depressive or anxious episodes. This narrative review discusses research published on the interactions between stress and the MGBA and examines the evidence and potential mechanisms of how differences in stress-related changes in the gut microbiota may be associated with stress-resilience.

2. Stress and the Microbiome-Gut-Brain Axis

2.1. The Link between the Gut Microbiota and Behavior

The gut microbiota comprises around 0.2 kg of human body weight and has around the same number of cells as human eukaryotic cells (recently revised estimate) [17]. It also has around 150-fold more genes [18]. Interest in the gut microbiome has flourished in recent decades due to realization of its role in production of metabolic and endocrine products, and interactions with the host nervous and immune systems. The suggestion that the gut microbiota is linked with and may influence mood disorders began with the observation of a high co-morbidity of anxiety and depression disorders in people with gut disorders such as inflammatory bowel disease [19,20] and irritable bowel syndrome [19,20,21,22]. Correlational studies have shown that fecal microbiota composition in individuals with anxiety or depression (including those in remission) differs from that in healthy controls [23,24,25]. Women with a higher fecal Prevotella abundance experienced increased negative emotional response to viewing negative images, and lower brain activity in the hippocampus than those with a higher Bacteroides abundance [26]. Several studies in rodents have experimentally shown that the presence and composition of the gut microbiota can alter emotional behavior. In mice, gut infections or inflammation caused an increase in patterns of behavior thought to represent anxiety, including decreased exploration [27] and increased behavioral inhibition [28,29]. Germ free (GF) rats and mice (born and raised with no microbiota) show either increased or decreased anxiety and depressive-like behaviors compared with counterparts with specific pathogen-free (SPF) gut microbiota [30,31,32,33]. Some probiotics also show an effect on mood. Psychobiotics are probiotics which have been shown to “confer mental health benefits through interactions with commensal gut bacteria”. Not all probiotics or prebiotics are considered psychobiotics [34].

2.2. Inconsistencies and Problems with MGBA Research

The results of MGBA studies do not always agree, and the results from animal studies do not always translate well to human research. This has been a concern with MGBA research. Animal behavioral testing has limitations on how well it reflects anxiety- or depressive-like symptoms in humans, but there are methodological limitations with human studies due to heterogeneity of lifestyles and because it is difficult (or impossible) to collect certain biological samples such as colon microbiota and host tissues.
Probiotic supplementation has shown mixed effects on emotional behavior (refer to Table 1). There are several studies in rodents which show an amelioration of anxiety-like or depressive like behaviors following probiotic supplementation [35,36], inflammation-induced behavior changes [29,37,38,39], and stress-induced behavior changes [40,41]. Other studies have found no difference [42,43]. Translational research has shown variable results. No difference in mood was found in healthy adults [44] or in people with irritable bowel syndrome [45] following a probiotic supplement, but in other studies, depression scores were reduced in people with diagnosed depression [46] and healthy adults [47,48], or emotional reactivity was reduced [49]. Mixed results include a study where anxiety scores but not depression scores were reduced [50], and mood improved only in those who had low baseline mood [51].
Another interesting observation is that the direction of change in anxiety-like behaviors in GF rodents (compared with their SPF counterparts) seems to depend on the strain. Stress-sensitive strains (BALB/c mice and Fischer 344 rats) showed increased anxiety-like behaviors, in contrast to a decrease in anxiety-like behaviors in more resilient strains (NMRI mice, Swiss Webster mice and Wistar rats). Both increased and decreased anxiety-like behaviors occurred in mice when the gut microbiota were depleted with anti-microbial drugs [52,53,54]. A fecal transplant from an anxious-type mouse strain into a GF non-anxious strain caused an increase in anxiety behaviors [53]. The reverse was also observed, with a decrease in anxiety-like behavior in previously GF mice colonized from a non-anxious mouse strain [53].
There are several reasons for the inconsistencies found in MGBA research. Probiotics effects are strain specific, and factors such as variation in survivability, ability to adhere to the gut mucosa, and their capacity to produce bioactive compounds [55]. The dose is also important and the efficacious dose may vary between probiotic strains. The variability in the effect on mood in GF rats suggest that it is the interactions of the microbiome with the host which is important, and that variations in host genotype/phenotype may be a key part of whether changes in the gut microbiota impact mood or not. Proposed mechanisms of the MGBA (Figure 1) are complicated, intertwined and bidirectional. Different hosts, with different life experiences (e.g., diet, stress, exercise), mean that the mechanisms for changes in mood could differ between animals and humans, animal strains, and possibly even individuals. Multiple mechanisms could also act in parallel.

2.3. Stress, the Gut Microbiota, and Behavior

There is interest developing in the interactions of stress with the MGBA. The gut microbiota composition can be altered under stress, as shown in rodent models of psychological stress [11,12,13,15,16,56,57,58,59,60,61,62,63,64]. Stress during pregnancy has also been shown to alter the gut microbiome structure in mouse offspring as well as the dams [65,66]. With the emerging evidence showing that alterations in the gut microbiota can influence mood, it seems plausible that stress-induced changes in the gut microbiota could (at least partially) mediate the development of chronic stress and/or anxiety and depression following a stressful event. Conversely, alleviation of the stress-induced changes in the gut microbiota and/or the physiological effects of this change may help to increase stress resilience.
Compositional changes in the gut microbiota following stress vary widely between studies and most of the evidence comes from rodent studies. Changes include a decrease in relative abundance of the genus Lactobacillus [15,56,58,67,68] and an increase in genera containing opportunistic pathogens such as Odoribacter [12,66], Clostridium [11,15], and Mucisprillum [13,66]. The genus Bifidobacterium has been shown to decrease under stress [15,63], and in one study was found to increase under stress in stress-resilient mice only [62]. The change in the gut microbiota may differ within different gut niches, for example restraint stress in male CD-1 mice caused a decrease in relative abundance of the genus Lactobacillus in the mucosa-associated microbiota but not the luminal microbiota [13].
The timeframe of the effect of stress on the gut microbiota varies. Similarly, recovery of the gut microbiota following stress seems to vary and changes can be persistent. Galley et al. [56] found differences in microbial beta diversity (which is comparison of changes in microbiota between samples rather than within samples) after only two hours of social stress in C57BL/6 mice, but a decrease in absolute abundance and relative abundance of Lactobacillus spp. after six days [56]. Infant rhesus monkeys had an altered gut microbiome three days following the stress of being separated from their mothers and placed instead in individual cages near other infant monkeys. Interestingly, five days post-separation, it was restored to the pre-separation composition [67]. In contrast, differences in fecal microbiota were found in rats seven weeks after maternal separation stress [16]. Bailey et al. [11] found that immediately following social stress, the gut microbiome of mice showed reduced diversity and richness, and clustered differently from the control group. However, after 15 h, the separation of the gut microbiota between groups was no longer as clear, with the stress group showing high variability, suggesting recovery of the gut microbiota following stress may vary between individuals.
Evidence from human studies is sparse. A study looking at the effect of diet and living conditions in (grounded) astronauts found that in fecal samples, the bacterial counts of Bacteroides fragilis subsp. thetaiotaomicron increased following interpersonal conflict in a confined living situation [68]. Prenatal stress in pregnant women was associated with persistently altered microbiota composition in their infants [69]. Increased relative abundances of Proteobacteria and a reduced relative abundances of lactic acid bacteria were found in the infants, which appear to be related to increased reports of gut problems [69].
The gut microbiota composition in humans or rodents is probably affected by a shift in the gut environment due to physiological changes in the gut under stress. Stress activates sympathetic pathways in the gut which regulate water absorption by gut epithelial cells; mucin production from goblet cells; gut permeability and inflammation (increasing mast cell degranulation and cytokine production) [70]. Increased gut motility and mucin secretion occur due to the mast cell degranulation [71,72]. Stress also causes slowed gastric emptying, but an overall decreased transit time, with increased distal colon motility [15,70]. Bacterial composition and function are also likely to be directly affected by circulating stress hormones [73]. Increases in concentrations of the catecholamines norepinephrine and epinephrine have been shown to increase the growth, virulence, and colonization of pathogenic bacteria [14,60,74,75]. Rodent stress models have shown increased colonization by Citrobacter rodentium, a colonic pathogen [60], and increased adherence and penetration of gut bacteria into mucosal cells [59]. Dexamethasone (an anti-inflammatory corticosteroid drug with similar actions to cortisol) administration in rats caused increased bacterial adherence and increased paracellular gut permeability (discussed in Section 3.2. in detail) [74].

3. Mechanisms Associated with Stress-Induced Changes in the Gut

3.1. Gut and Systemic Inflammation

Stress-induced gut inflammation could be a key mechanism for changes in emotional behavior under stress, with the gut microbiome function promoting or decreasing gut and systemic inflammation. Increased plasma inflammatory markers including interleukin (IL)-6 and tumor necrosis factor (TNF)-α have been found in people with anxiety [75] and depression [76,77] particularly in those who fail to respond to classical treatments [77]. Both depression and anxiety are more prevalent in people with inflammatory bowel disease (IBD), and are associated with more frequent IBD flare-ups and more severe IBD symptoms [78].
There is some evidence of the ability of the gut microbiome to promote or ameliorate systemic inflammation. Commensal microbiota seems to be associated with decreased inflammation, whereas potentially pathogenic bacteria are associated with inflammatory gut conditions [79,80,81]. In GF mice, the immune response to LPS is blunted [32,82,83], suggesting that the gut microbiota primes the immune system. Microglial activation, which causes neural inflammation, seems to be affected by the gut microbiota [37,82,83], possibly mediated by free fatty acid receptor 2 (FFAR2) in the gut, activated by short-chain fatty acids (SCFA) produced by the gut microbiota [84]. Both inflammatory and subclinical microbial gut infections, as well as chemically induced colitis increase anxiety and depression behavior or neurobiochemical markers in rodents [27,29,36,82,83,85]. Probiotic supplementation has been shown to attenuate pro-inflammatory markers or responses (Lactobacillus helveticus ns8 [41]; Lactobacillus salivarius UBL S22 [86]; Lactobacillus farciminis [87]; Bifidobacterium infantis 35624 [40,43]) and commercial mix VSL#3 [37]) although not always (Lactobacillus rhamnosus JB-1 [44]; Bifidobacterium longum NCC3001 and L. rhamnosus NCC4007 [29]). The probiotic L. salivarius UBL S22 also decreased the total fecal Escherichia coli count [86], a bacteria linked with gut inflammation [88].
The stress-induced gut microbiota composition, with decreased commensal microbiota and increased opportunistic pathogens, is likely to be inflammatory, but whether it is the cause of stress-induced gut inflammation and whether it mediates emotional behavior is unknown. Stress-induced inflammation also occurs directly via the sympathetic nervous system innervation of lymphoid organs [70] and activation of mast cells [72,73,89] and dendritic cells [90], however antibiotic administration in mice during stress prevented an increase in pro-inflammatory markers, suggesting a key role of the gut microbiota [11]. The probiotic L. rhamnosus JB-1 was also able to ameliorate stress-induced dendritic cell activation as well as reduce the effects of stress-related changes in anxiety-like behavior [90].
Seven days of repeated restraint stress in mice increased the immune response to a colonic pathogen challenge. A fecal transplant from the stressed mice into GF mice caused an increased inflammatory response to colonic pathogen (Citrobacter rodentium) in the GF mice, compared with a fecal transplant from non-stressed mice [63]. Whether the fecal microbiota itself or other molecules within the feces caused the increased immune response is unclear, but it is likely due to an interaction between the gut microbiota and the enteric immune system. The gut microbiota plays a fundamental role in the function and maturation of the gut immune system, including CD4 cells [91]. Susceptibility to colonic inflammation has previously been shown to increase following stress due to sensitization of CD4(+) lymphocytes, with the increased susceptibility able to be transferred to other rats with intravenous transfer of the CD4(+) lymphocytes [92].

3.2. Gut Permeability

Increased gut permeability was found in over 40 % of people with depression in one study [89], and may be a cause of increased systemic inflammation [93]. Gram-negative bacteria, such as Proteobacteria (including E. coli), have endotoxic lipopolysaccharide (LPS) chains on their outer cell wall, and with increased gut permeability, translocation of LPS from the gut lumen into the body occurs. LPS interacts with immune cells and induces the expression of several inflammatory molecules such as pro-inflammatory cytokines, nitric oxide, and eicosanoids, which are also found in those with depression [78,94,95]. Intravenous administration of LPS has been shown to increase anxiety and depression behaviors in people [96] and mice [84], and can induce neuroinflammation, causing microglial cells to become activated [97,98]. Evidence for increased gut permeability and bacterial translocation in people with depression has also been found, with increased concentrations of serum IgA and IgM against LPS [99], and associated increased activation of inflammation, oxidative and nitrosative stress pathways [93].
Increased gut permeability occurs under stress [87,94,95,100,101,102], with decreased expression of tight junction proteins [94] and an increase in translocation of large antigenic molecules [95,100,101]. The release of antigens triggers the inflammatory mechanisms such as activation of CD4+ cells resulting in mast cell mast cell degranulation, neutrophil infiltration and increased cytokine IFN-γ [94,102]. Increased gut permeability to antigenic molecules is likely to contribute to colonic inflammation due to reactivation of sensitized CD4(+) cells [103]. Whether increased gut permeability following stress occurs due to physiological reasons or because of a change in the gut microbiota is unclear. The increased permeability can be induced by dexamethasone or eliminated by adrenalectomy or glucocorticoid receptor blockade, suggesting physiological mechanisms [101]. However, the evidence does suggest that promotion of the growth of commensal bacteria with a decrease in LPS-producing bacteria is likely to alleviate stress-induced increases in gut permeability and associated pro-inflammatory immune activation. Dysbiosis, including a decrease in Bifidobacterium and an increase in LPS-producing bacteria has been associated with increased gut permeability [104], and probiotic supplementation (L. farciminis [87]; mix of L. rhamnosus R0011 and L. helveticus R0052 [59]; L. paracasei NCC2461 [105]) can attenuate stress-induced gut permeability in rodents. Whether supporting and promoting the growth of commensal bacteria under stress increases psychological resilience is the next level of research needed.

3.3. Dysbiosis and Hypothalamic-Pituitary-Adrenal Axis Dysfunction

A dysfunctional stress response and hypothalamic-pituitary-adrenal (HPA) axis may be a contributor to the development of both anxiety and depression [106]. The gut microbiome may also influence the stress response. The markers of the HPA axis such as corticosterone have been shown to be altered in GF mice compared with SPF counterparts [31,32,33,107]. Blunted corticosterone responses have also been found in adult rats exposed to early life stress, and the corticosterone concentrations negatively correlated to fecal Akkermansia and Rikenella [108,109]. A fecal transplant from depressed people into GF rats caused an increase in the rats’ corticosterone response to acute stress alongside increased depression-like and anxiety-like behaviors [110].
How the gut microbiota affects the stress response is uncertain. Immune activation activates the HPA axis [111,112,113], providing an indirect mode of effect of the gut microbiota on the stress response system. It is known that some bacteria produce catecholamines including norepinephrine, epinephrine and dopamine [114,115,116,117,118,119,120]. This is unlikely to cause a direct increase in concentrations of systemic catecholamines, because GF rats have typically been found with increased HPA-axis markers compared to rats with the normal gut microbiota [31,32,33,107]. It is plausible however that microbially produced catecholamines could contribute to baseline concentrations, and therefore HPA-axis programming in early life. Early life dysbiosis could therefore provide artificially high or low basal catecholamine concentrations and cause dysfunctional programming. This has not been tested.
The balance between pathogenic and commensal bacteria in the gut is likely to be important. Elevated plasma ACTH and corticosterone concentrations in GF rats were able to be prevented by the colonization of the GF mice at an early age with B. infantis or a mutant strain of E. coli lacking the translocated intimin receptor gene, neither of which are internalized into the gut epithelial cells. Wild-type E. coli, which do get internalized, did not prevent the heightened stress response [107]. Probiotic supplementation during or following stress has been shown to reduce corticosterone in stressed animals (L. rhamnosus JB-1 [36], L. farciminis [87], L. helveticus ns8 [41], mix of L. rhamnosus R0011 and L. helveticus R0052 [59] or monoassociation with B. infantis [107] or people (probiotic L. helveticus R0052 and B. longum R0175) [47], but B. infantis 35624 did not affect corticosterone in non-stressed animals [43]. However, no changes in systemic corticosterone concentrations were found following stress-induced changes in the gut microbiota and behavior in mice [58], or with amelioration of stress-induced behavioral changes in rats following supplementation with B. infantis 35624 [40]. Interestingly, a synbiotic supplement (L. rhamnosus GG + polydextrose and galactooligosaccharide) increased plasma corticosterone concentration in rats following acute stress compared to that of control animals, whereas the same probiotic and prebiotic mix given separately did not [64].
The HPA axis dysregulation in people with depression is in itself not a straightforward relationship, with both high and low levels of cortisol found, as well as other dysfunction such as delayed return to baseline following acute stress and glucocorticoid resistance. With the gut microbiota able to increase HPA axis activation, stress-induced dysbiosis may increase the physiological stress response higher than that which is required to deal with the stressor effectively. Whether the gut microbiome moderates stress-induced increases in HPA activation in a meaningful way, and whether this affects emotional behavior is unclear. More research is needed in this area.

3.4. Metabolites

Microbially derived metabolites include SCFAs, bile acids, choline and phenolic metabolites, indole derivatives, vitamins, polyamines, and lipids [121]. The metabolites are primary secreted signaling molecules (influenced by host-derived signaling molecules) which cross-talk with other microbes, and the host immune system, or secondary metabolites (produced through the metabolism of food, non-food ingested compounds, such as medication, and metabolites from other microbes).
SCFAs activate several receptors in the gut which have been shown to reduce colonic inflammation [122,123] and microglial neuroinflammation [84]. These receptors can also increase gut epithelial cell barrier integrity by increasing the expression of tight junctions [reviewed in [124]]. There is some evidence of altered SCFA production with depression and stress, although the direction of change is conflicting. No difference in SCFA concentrations was found in the fecal samples of people with depression compared with controls in two studies [110,125]. When fecal samples from depressed people were transplanted into mice, an increase in fecal acetate and total SCFA concentrations was found along with increases in depression-like behavior [110]. Prebiotic supplementation in mice increased SCFA concentrations many of which were negatively correlated with depression-like and anxiety-like behaviors [61].
A decrease in fecal acetate and butyrate, as well as SCFA-producing bacteria, occurred in mice following psychosocial stress, and this was associated with an increase in gut inflammation [126]. A similar study found an increase in cecal acetate, a decrease in propionate, butyrate and valerate, but no change in branched-chain fatty acids. No increase in systemic LPS was found, despite an increase in gut permeability to FITC-dextran, but this was likely due to the colonic mucus layer being unaffected. Interestingly, only minor changes in the gut microbiota composition at the family and genus level were observed following the stress intervention [127]. Functional changes in the gut microbiota have also been seen in mice following stress. KEGG analysis of 16S RNA marker genes in fecal samples predicted reduced pathways for the synthesis and metabolism of neurotransmitter precursors tyrosine and tryptophan, and SCFAs. This finding was positively associated with reduced exploration and sociability in the mice [128]. In contrast, children who had increased self-reported stress showed increased fecal SCFAs (butyrate, valerate, isovalerate and isovalerate), but no increased gut inflammation (based on fecal calprotectin concentrations) was found [129]. A possible explanation for the increased fecal SCFA is stress-induced decrease in gut transit time rather than a change in gut microbiota fermentation. Hair cortisol concentrations (a measure of long-term stress) in the children were not related to SCFAs, but heart rate variability was associated with decreased valerate [129]. Heart rate variability is a proxy for parasympathetic nervous system activation [130], which affects gut transit time [70]. Gut motility likely has bidirectional interactions with SCFAs and microbial composition. Stress induced dysmotility was able to be reversed in vitro with the application of either propionate or L. rhamnosus JB-1 [131].
Supplementation of an SCFA mixture in healthy adult men (174.2 mmol acetate, 13.3 mmol propionate, and 52.4 mmol butyrate), administered daily via the colon, reduced acute corticosterone response to acute stress, and increased serum SCFAs [132]. This result was not, however, associated with a change in subjective mood ratings [132]. In male mice, daily oral supplementation of the SCFAs (67.5 mmol acetate, 25 mmol propionate, and 40 mmol butyrate) decreased stress-related increases in anxiety-like behaviors in the open field test, and increased sucrose preference and decreased urine sniffing, both markers of depression-like behavior. The SCFA supplement was associated with changes in gene expression in the brain related to dopamine receptors, part of the mesolimbic reward pathway which can be altered in depression [127]. Cecal SCFAs also differ between stress-sensitive WKY rat strain compared with the stress-resilient Sprague Dawley rat strain [133]. Anti-inflammatory effects of an increase in SCFA in some of the populations studied could be contributing to stress resilience.
The gut microbiome is a source of vitamins, including vitamin K and B vitamins niacin, biotin, riboflavin, folate and pyroxidine [134,135,136,137,138]. Serum folate (B9) and pyroxidine (B6) are lower in those with depression or an increased risk of depression [139,140,141,142]. Micronutrients may affect depression risk via effects on the production and activity of monoamine neurotransmitters such as serotonin [143,144,145,146,147,148], alterations to the HPA system [149], glutamatergic signaling [149], or inflammatory and oxidative stress [149,150]. They also play a role in the gut, for example, niacin is anti-inflammatory in the gut due to activation of the Gpr109a receptor, the same receptor that is activated by the SCFA butyrate [122]. Folate and biotin are also immunomodulatory [151,152]. Pyroxidine (B6), is an essential co-factor for several enzymes in the kynurenine pathway [153], and a deficiency increases levels of xanthurenate, a kynurenine metabolite which is an antagonist for glutamate receptors. GF rats show increased susceptibility to developing B6 deficiency [136], and an accumulation of xanthurenate [154]. Changes in the gut microbiota could alter the available concentrations of microbially produced vitamins, plausibly contributing to immune and metabolic pathway changes which are related to mood.
Up to 95% of the neurotransmitter serotonin, which has a well-known link to anxiety and depression [155] is produced endogenously in the gut mucosa [156], and secretion of serotonin from enterochromaffin cells is influenced by microbial metabolites [157,158,159,160]. It is also produced by the gut microbiota, along with several other neurotransmitters including dopamine, gamma aminobutyric acid (GABA), acetylcholine and norepinephrine [115,116,117,118,119,120]. Neurotransmitter levels and turnover in the brain differ in GF mice [32,33,161] and reduced levels of circulating GABA and serotonin have been found in GF rats [162,163]. Whether alterations of these metabolites in the gut (endogenous and microbially produced) are related to those in the brain is uncertain, but there is some evidence that they can cross the blood–brain barrier [164], of which the permeability is in itself affected by the gut microbiota [165]. Whether gut metabolites can directly reach the brain or not, they can affect the gastric environment and neural signaling. For example, GABA and acetylcholine are immunomodulatory [166,167], and GABA and serotonin affect gastric motility and acid secretion via enteric neurons [160,161]. Whether stress-related changes in the gut microbiota alter the concentration of microbially produced vitamins or neurotransmitters in the gut is unknown.
Changes in metabolic pathways are another way that neurotransmitters are altered. Chronic stress in mice caused a decrease in the genus Lactobacillus, and a correlated increase in depression-like behavior. Supplementation with the probiotic Lactobacillus reuteri ATCC 23272 decreased the depression-like behavior, seemingly via the production of H2O2 which inhibits the enzyme indoleamine 2,3-dioxygenase 1 (IDO1) and restores the balance of serotonin/kynurenine pathways [15]. IDO1 is activated by inflammation and LPS [168].

3.5. Gut Nervous System—Enteric Nerves and Vagus Nerve

Autonomic nervous system dysfunction, with increased sympathetic tone and decreased parasympathetic (vagal) tone, is proposed to be a contributing factor in the development of depression [169]. People with depression commonly show decreased heart rate variability, a measure of high sympathetic activity [170], and vagal nerve stimulation may be effective for treatment-resistant depression, e.g., [171], although more research is needed [172].
The vagus nerve is linked with both the HPA axis and the immune system. Afferent fibers of the vagus nerve innervate the nucleus of the solitary tract, a brain region that directly regulates the HPA axis. Vagal nerve stimulation therapy can normalize HPA activity [169]. Secondary fibers also innervate brain regions responsible for emotional regulation [169,173]. Vagal sensory endings have receptors for cytokines and relay information about thoracic and gut inflammation to the brain [173]. In response, efferent fibers of the vagus nerve influence inflammation via cholinergic signaling, which inhibits cytokine release from LPS-stimulated macrophages. This negative feedback effect is known as the vagal-immune reflex. Low vagal tone is thought to promote systemic inflammation [174,175]. There may also be an influence on neuroinflammation via receptors located on microglia and astrocytes [169].
Gut infection or systemic immune challenge with the bacterial endotoxin LPS caused vagal ganglia activation (shown by FOS immunoreactivity) [27,82]. Associated sickness and anxiety-like behavior were able to be alleviated with vagotomy [39,88,176], suggesting that the vagal signaling can mediate the development of emotional behaviors. Increased vagal activation also occurs with probiotic supplementation (Lactobacillus johnsonii La1 [177]; B. Infantis [107]), and vagotomy prevented the restorative effect of probiotics on anxiety (B. longum NCC3001 [39]; L. rhamnosus JB-1 [36]). Early life stress in rats increased the cholinergic secretory response of enteric nerves to stimulation [59]. While there is strong evidence for the vagus nerve being a key mediator in the gut-brain axis, some studies have found changes in emotional behavior in mice due to gut infection [29] or anti-microbial treatment [53] despite a vagotomy procedure [29,53]. The vagus nerve may just be one mechanism of transmitting infection information to the brain.

4. Early Life Programming

Childhood adversity and stressful life events are both strongly linked with an increased risk of developing depression [4,5,178]. Early life stress in rodents (during the neonatal period) can cause increased anxiety-like and depressive-like behaviors in adulthood [64,179,180,181]. Stress during adolescence in mice also caused increased anxiety-like behaviors in adulthood [182]. In contrast, a safe and reliable childhood with strong maternal/caregiver attachment decreases the risk of anxiety and depression later in life, even in those with a higher genetic risk [7,183]. The stress response system (SRS) is the biological response to both psychological and physiological (such as illness, injury) stressors. Both a hyper and hypo-responsive SRS are linked with mood disorders [176,184,185]. The SRS is functionally and epigenetically programmed in early life to match the individual’s phenotype to their environment [178,186,187]. Similar epigenetic programming occurs in early life for the immune system. Early life stress causes an inflammatory immune phenotype characterized by increased pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) which are also associated with depression [16,188]. Whether the early life stress causes independent epigenetic changes to the immune system or is mediated through epigenetic modulation of the SRS is debated [188]. The two systems are interlinked [111,112,113]; early exposure to high cortisol levels can cause immune dysfunction later in life [187].
The gut microbiota may play a role in this early life programming. The biological response to physiological and psychological stressors are similar, and it is likely that exposure to the increases in inflammation and alterations in HPA-axis signalling due to stress-induced dysbiosis, maybe a significant part of the environmental signalling which causes epigenetic programming. Injections of LPS in rats in the neonatal period [178] and in early adolescence [189] caused increased anxiety-like behaviors later in life, and changes in GABA, corticotropin-releasing hormone (CRH) and glucocorticoid receptors in the hippocampus and hypothalamus in [178]. Maternal Separation Stress (MSS) has been shown to increase gut permeability [59], and therefore systemic LPS exposure. Whether interventions to manipulate the gut microbiota in early life can prevent or reduce the effects of early life stress is still being elucidated.
Probiotic supplementation with L. rhamnosus strain R0011 (95%) and L. helveticus strain R0052 (5%) during the separation period of MSS was able to prevent the stress-induced increase in serum corticosterone, adherence and penetration of bacteria into mucosal cells, and increase in gut permeability [59]. Additionally, increases in anxiety-like behavior and changes in gene expression in rats following MSS were able to be ameliorated by dietary supplementation of the probiotic L. rhamnosus GG alone or in combination with prebiotics polydextrose and galactooligosaccharide [64]. In contrast, supplementation of rats with omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DPA) following MSS restored the gut microbiota [108] and prevented higher levels of corticosterone in response to stress [109], but caused reduced anxiety and increased cognitive performance in the non-stressed rats only, with no difference in behavior in the stressed rats [109]. These findings may be due to the intervention being after rather than during the stress period, the type of intervention, or an indication that the gut microbiota are not the key mediator of the behavioral effects of early life stress.

5. Therefore, Could the Gut Microbiota Be Key in Stress-Resilience?

The evidence for stress-induced changes in the gut microbiota being a mechanism rather than a covariate of stress-induced changes in mood is limited and sometimes conflicting, but has plausible mechanisms. If stress-induced changes in the gut microbiota do reduce stress resilience then differences between individuals who are stress-resilient and stress-sensitive and/or correlations of the gut microbiota with mood symptoms should be observed. There is some evidence to support this. Studies investigating links between stress, the gut microbiome and mood are summarized in Table 2.
A comparison of stress-resilient mice showed increased Bifidobacterium spp. in the stress-resilient mice compared with control mice or stress-sensitive mice [62]. Some strains of Bifidobacterium are considered psychobiotic so it seems straightforward that an increase in their abundance in some of the rats could promote stress resilience. However, the mechanism for how an increase in Bifidobacterium would occur due to stress was unclear.
Other studies did not compare stress-resilient and stress-sensitive mice, but found some correlations between gut microbiota and behavior. Marin et al. [15] found decreased Lactobacillus in male mice following chronic mild stress and a positive correlation between Lactobacillus and escape behaviors (active swimming) in the forced swim test [15]. Bansgaard Bendtsen et al. [12] found that in female BALB/c mice exposed to two weeks of grid floor stress, the cecal microbiota differed from that of the control group and was correlated with behavior. The time spent in the dark compartment of the light/dark box test (considered as anxiety-like behavior) was positively correlated with Ruminococcaceae spp., which was also negatively correlated with pro-inflammatory cytokine interleukin-2. Time spent in the closed arm of the elevated plus maze (also considered anxiety-like behavior) was negatively correlated with the genus Butyricicoccus (a butyrate producer). In the same study, risk assessment behavior (two paws placed in open arms and then retracted) were positively correlated with Lachnospiraceae, and in the control group only, the relative abundance of Bacterioides (a major propionate producer) positively correlated with the number of immobility episodes in the tail suspension test [12]. Likewise, comparison of stress-sensitive WKY rats and stress-resilient Sprague Dawley rats under acute stress showed an increase in relative abundance of cecal Lactococcus, a lactic acid producer. Lactococcus was positively correlated with brain and plasma lipid metabolites [133].
The evidence for specific microbiota relating to behavior is sparse but suggests that changes in SCFAs and inflammation may be key mechanisms. These could be linked due to SCFAs being immunodulatory. Stress-induced decreases in the genus Lactobacillus have been found in several animal stress studies [11,15,56,57,58,59,60,61,62,63,64,65,66,67,68]. It is plausible that individual differences in stress-induced decrease in Lactobacillus could be the difference between an in individual being stress-resilience or stress-sensitive. Marin et al. [15] found that kynurenine was increased in stressor-exposed mice alongside the lowered Lactobacillus and that while a supplement of the probiotic L. reuteri ameliorated stress-induced behavior, it did not work if L-kynureine was also supplemented alongside. They found that in vitro, a reactive oxygen species produced by Lactobacillus inhibited the enzyme indoleamine 2,3-Dioxygenase 1 (IDO1), a key enzyme which allows tryptophan to be converted to kynurenine. IDO1 is also activated by inflammation and therefore could be a mechanism
Not all studies have found associations between microbiota composition and behavior. Tsilimigras et al. [58] reported stress-induced changes in behavior and the gut microbiota in mice, but there was no correlation between any of the gut microbiota changes with behavior changes. Conflicting results could be due to the site of microbiota sampling: Bangsgaard Bendtsen et al. [12] found that stress-induced behavioral changes correlated with cecal but not fecal microbiota changes.
Correlation of gut microbiota composition with behavior does not show causality. It is equally likely that differing changes in the gut due to different levels of perceived stress in stress-resilient or stress-sensitive individuals are the mediator for changes to the gut microbiota. However, intervention with prebiotic and probiotics have been able to alleviate stress-induced changes in emotional behavior. People given 30 days of a probiotic mix (L. helveticus R0052 and B. longum R0175) had reduced anxiety, depression, and perceived stress scores, as well as a decrease in 24 h urinary free cortisol from baseline concentrations [47]. The baseline anxiety and depression scores of the participants ranged from low to moderately high. In mice, oral supplementation with Bifidobacterium (LAC-B Granular Powder) increased the number of mice resilient to social defeat stress, and prevented a stress-induced decrease in sucrose intake [62]. L. reuteri 23272 given to male mice during chronic, mild stress decreased despair behavior in the forced swim test [15]. A probiotic (L. rhamnosus GG), prebiotic mix (polydextrose and galactooligosaccharide) or combined in a synbiotic mix, following maternal separation stress in male and female Sprague Dawley rats reduced stress-induced increases in anxiety-like behavior. The synbiotic had the greatest effect and was also able to ameliorate stress-induced memory changes [64]. An increase in stress-induced defecation was able to be prevented by prebiotic (fructooligosaccharides and galactooligosaccharides) supplementation [61].
It is possible that the positive action of probiotic and prebiotic supplementation on emotional behaviors may be more effective following stress. Sprague Dawley rats given a probiotic (B. infantis 35624) for 40 days following maternal deprivation stress, had reduced stress-induced immobility in the forced swim test [40] whereas the same daily dose of the same probiotic (although for only 14 days) without the stress intervention did not affect behavior [43]. Similarly, stress-induced increases in anxiety-like and depressive-like behaviors in mice were able to be ameliorated by dietary supplementation with prebiotics fructooligosaccharides and galactooligosaccharides [61]. Basal and acute stress-induced corticosterone levels were also reduced, and the prebiotic supplement prevented a stress-induced decrease in the Actinobacteria: Proteobacteria ratio and the relative abundance of Bifidobacterium and Lactobacillus. The decreased Actinobacteria: Proteobacteria ratio may have reflected a decrease in inflammation-reducing bacteria such as Bifidobacterium and an increase in opportunistic pathogens and bacteria with LPS. This may explain some of the stress-related changes in inflammation and mood. The prebiotic supplementation, given in a prior study with no stress intervention, had a much weaker effect on behavior [61]. Finally, a reduction in anxiety-like behavior was found in BALB/c stress-sensitive mice following supplementation of L. rhamnosus JB-1 [36], but the same probiotic given to healthy men did not alter HPA response or subjective mood or stress measures [44].
It is also possible that stress is one of the reasons why the results of animal intervention studies do not always translate well to human research. Laboratory conditions are neither reflective of real life for humans nor the animals involved in the research. Laboratory conditions can be stressful for animals, for example single housing [191]. This means that many intervention studies in animal may be effective by alleviating stress-induced changes in the gut-microbiota and/or physiology. If the same stress-induced changes are not present in human study participants then there may be no effect.

Considerations for Future Research

Defining stress is not straightforward, and could have an impact on research results.
“Stress” is a broad term. External stress is defined by the environmental conditions, whereas perceived (internal) stress depends on how the individual feels. It is often assumed that under environmental stress all the individuals are also experiencing perceived stress but this may not be true. There is also a difference between how individuals experience perceived stress. “Good stress” is termed eustress and can improve performance and mood. “Bad stress” is what is typically considered to be stress and is sometimes defined as distress. In a similar vein, it is possible that animals in studies where stress is induced are experiencing environmental stress but not perceived stress. A careful definition of the type of stress being measured is important, and the measurement of baseline stress levels (e.g., corticosterone in hair or feces) as well as acute levels is beneficial.
The type of biological processes activated under stress also needs to be differentiated. HPA-axis activation may affect the MGBA differently than sympathetic nervous system activation. It is unclear whether the MGBA increases the risk of chronic perceived stress developing into anxiety and/or depression, or whether it increases the risk of an individual experiencing chronic perceived stress under high environmental stress. It may do both.
There is a reported association between diet and depression but the nature of the relationship is still unclear. Some research indicates that a healthy diet is protective against developing depression, but there are also many studies showing no effect [192]. Dietary manipulation of the gut microbiota composition may be a key variable in the diet-depression relationship, especially in preventing stress-induced changes in the gut microbiota [192]. Taylor, et al. [193] found independent relationships between the gut microbiota and mood (stress, anxiety and depression); and dietary factors and mood, with the microbiota-mood associations were mediated by fiber intake.

6. Conclusions

Stress-induced changes in the gut microbiota are a key variable which needs to be considered more in mood research. The limited research available suggests that promoting the growth of commensal bacteria, particularly those considered to be probiotic, is likely to confer some increase in emotional resilience under stress. Whether this is through direct prevention of effects from stress-altered gut microbiota, or through alleviation of physiological consequences of stress-induced changes, or both, is unknown. More research is needed. The mechanisms are still being elucidated and which individuals are more likely to respond to microbial support under stress, and whether there are ages, types of stress, time points in which interventions may be more or less effective, remain unknown. There is strong evidence for a role of lactobacillus, both as probiotic supplement or as commensal bacteria and research into probiotics and their mechanisms continues. The consequences of stress and interventions in different life periods, especially in early life should continue to be investigated. Dietary manipulation is another key area for research, particularly because diet will affect the gut microbiota composition and function. More nuanced physiological and psychological measurements are needed in order to differentiate between environmental, perceived stress and stress-related physiological processes.

Author Contributions

Writing—original draft preparation, T.B.; writing—review and editing, P.G., J.D., N.R., C.B. and J.C.; funding acquisition and research conceptualization, P.G. and N.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Riddet Institute, a New Zealand Centre of Research Excellence, funded by the Tertiary Education Commission.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Elwenspoek, M.M.C.; Kuehn, A.; Muller, C.P.; Turner, J.D. The effects of early life adversity on the immune system. Psychoneuroendocrinology 2017, 82, 140–154. [Google Scholar] [CrossRef] [PubMed]
  2. Kendler, K.S.; Neale, M.C.; Kessler, R.C.; Heath, A.C.; Eaves, L.J. Major depression and generalized anxiety disorder: Same genes, (partly) different environments? Arch. Gen. Psychiatry 1992, 49, 716–722. [Google Scholar] [CrossRef] [PubMed]
  3. Toomey, R.; Panizzon, M.S.; Kremen, W.S.; Franz, C.E.; Lyons, M.J. A twin-study of genetic contributions to morningness-eveningness and depression. Chronobiol. Int. J. Biol. Med. Rhythm Res. 2015, 32, 303–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Newman, S.C.; Bland, R.C. Life events and the 1-year prevalence of major depressive episode, generalized anxiety disorder, and panic disorder in a community sample. Comp. Psychiatry 1994, 35, 76–82. [Google Scholar]
  5. Mazure, C.M. Life stressors as risk factors in depression. Clin. Psychol. Sci. Pract. 1998, 5, 291–313. [Google Scholar] [CrossRef]
  6. Pfau, M.L.; Russo, S.J. Peripheral and central mechanisms of stress resilience. Neurobiol. Stress 2015, 1, 66–79. [Google Scholar] [CrossRef] [Green Version]
  7. Franklin, T.B.; Saab, B.J.; Mansuy, I.M. Neural mechanisms of stress resilience and vulnerability. Neuron 2012, 75, 747–761. [Google Scholar] [CrossRef] [Green Version]
  8. Smith, J.; Prior, M. Temperament and stress resilience in school-age children: A within-families study. J. Am. Acad. Child. Psychiatry 1995, 34, 168–179. [Google Scholar] [CrossRef]
  9. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar]
  10. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  11. Bailey, M.T.; Dowd, S.E.; Galley, J.D.; Hufnagle, A.R.; Allen, R.G.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bangsgaard Bendtsen, K.M.; Krych, L.; Sørensen, D.B.; Pang, W.; Nielsen, D.S.; Josefsen, K.; Hansen, L.H.; Sørensen, S.J.; Hansen, A.K. Gut microbiota composition is correlated to grid floor induced stress and behavior in the balb/c mouse. PLoS ONE 2012, 7, e46231. [Google Scholar]
  13. Galley, J.D.; Yu, Z.; Kumar, P.; Dowd, S.E.; Lyte, M.; Bailey, M.T. The structures of the colonic mucosa-associated and luminal microbial communities are distinct and differentially affected by a prolonged murine stressor. Gut Microbes 2014, 5, 748–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tannock, G.W.; Savage, D.C. Influences of dietary and environmental stress on microbial populations in the murine gastrointestinal tract. Infect. Immun. 1974, 9, 591–598. [Google Scholar] [CrossRef] [Green Version]
  15. Marin, I.A.; Goertz, J.E.; Ren, T.; Rich, S.S.; Onengut-Gumuscu, S.; Farber, E.; Wu, M.; Overall, C.C.; Kipnis, J.; Gaultier, A. Microbiota alteration is associated with the development of stress-induced despair behavior. Sci. Rep. 2017, 7, 43859. [Google Scholar] [CrossRef] [PubMed]
  16. O’Mahony, S.M.; Marchesi, J.R.; Scully, P.; Codling, C.; Ceolho, A.M.; Quigley, E.M.; Cryan, J.F.; Dinan, T.G. Early life stress alters behavior, immunity, and microbiota in rats: Implications for irritable bowel syndrome and psychiatric illnesses. Biol. Psychiatry 2009, 65, 263–267. [Google Scholar] [CrossRef]
  17. Sender, R.; Fuchs, S.; Milo, R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [Green Version]
  18. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59. [Google Scholar] [CrossRef] [Green Version]
  19. Kurina, L.; Goldacre, M.; Yeates, D.; Gill, L. Depression and anxiety in people with inflammatory bowel disease. J. Epidemiol. Community Health 2001, 55, 716–720. [Google Scholar] [CrossRef] [Green Version]
  20. Addolorato, G.; Capristo, E.; Stefanini, G.F.; Gasbarrini, G. Inflammatory bowel disease: A study of the association between anxiety and depression, physical morbidity, and nutritional status. Scand. J. Gastroenterol. 1997, 32, 1013–1021. [Google Scholar] [CrossRef]
  21. Lydiard, R.B. Irritable bowel syndrome, anxiety, and depression: What are the links? J. Clin. Psychiatry 2001, 62, 38–47. [Google Scholar]
  22. Masand, P.S.; Kaplan, D.S.; Gupta, S.; Bhandary, A.N.; Nasra, G.S.; Kline, M.D.; Margo, K.L. Major depression and irritable bowel syndrome: Is there a relationship? J. Clin. Psychiatry 1995, 56, 363–367. [Google Scholar]
  23. Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linlokken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. J. Neurogastroenterol. Motil. 2014, 26, 1155–1162. [Google Scholar] [CrossRef]
  24. Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jiang, H.-Y.; Zhang, X.; Yu, Z.-H.; Zhang, Z.; Deng, M.; Zhao, J.-H.; Ruan, B. Altered gut microbiota profile in patients with generalized anxiety disorder. J. Psychiatry Res. 2018, 104, 130–136. [Google Scholar] [CrossRef] [PubMed]
  26. Tillisch, K.; Mayer, E.; Gupta, A.; Gill, Z.; Brazeilles, R.; Le Nevé, B.; van Hylckama Vlieg, J.E.; Guyonnet, D.; Derrien, M.; Labus, J.S. Brain structure and response to emotional stimuli as related to gut microbial profiles in healthy women. Psychosom. Med. 2017, 79, 905. [Google Scholar] [CrossRef] [PubMed]
  27. Lyte, M.; Li, W.; Opitz, N.; Gaykema, R.P.; Goehler, L.E. Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium. Physiol. Behav. 2006, 89, 350–357. [Google Scholar] [CrossRef]
  28. Lyte, M.; Varcoe, J.J.; Bailey, M.T. Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiol. Behav. 1998, 65, 63–68. [Google Scholar] [CrossRef]
  29. Bercik, P.; Verdu, E.F.; Foster, J.A.; Macri, J.; Potter, M.; Huang, X.; Malinowski, P.; Jackson, W.; Blennerhassett, P.; Neufeld, K.-A.M.; et al. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology 2010, 139, 2102. [Google Scholar] [CrossRef] [Green Version]
  30. Nishino, R.; Mikami, K.; Takahashi, H.; Tomonaga, S.; Furuse, M.; Hiramoto, T.; Aiba, Y.; Koga, Y.; Sudo, N. Commensal microbiota modulate murine behaviors in a strictly contamination-free environment confirmed by culture-based methods. J. Neurogastroenterol. Motil. 2013, 25, 521. [Google Scholar] [CrossRef] [PubMed]
  31. Crumeyrolle-Arias, M.; Jaglin, M.; Bruneau, A.; Vancassel, S.; Cardona, A.; Dauge, V.; Naudon, L.; Rabot, S. Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 2014, 42, 207–217. [Google Scholar] [CrossRef]
  32. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Neufeld, K.M.; Kang, N.; Bienenstock, J.; Foster, J.A. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. J. Neurogastroenterol. Motil. 2011, 23, 255–264.e119. [Google Scholar] [CrossRef] [PubMed]
  34. Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W.J. Psychobiotics and the manipulation of bacteria–gut–brain signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Abildgaard, A.; Elfving, B.; Hokland, M.; Wegener, G.; Lund, S. Probiotic treatment reduces depressive-like behaviour in rats independently of diet. Psychoneuroendocrinology 2017, 79, 40–48. [Google Scholar] [CrossRef] [PubMed]
  36. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. D’Mello, C.; Ronaghan, N.; Zaheer, R.; Dicay, M.; Le, T.; MacNaughton, W.K.; Surrette, M.G.; Swain, M.G. Probiotics improve inflammation-associated sickness behavior by altering communication between the peripheral immune system and the brain. J. Neurosci. 2015, 35, 10821–10830. [Google Scholar] [CrossRef] [Green Version]
  38. Smith, C.J.; Emge, J.R.; Berzins, K.; Lung, L.; Khamishon, R.; Shah, P.; Rodrigues, D.M.; Sousa, A.J.; Reardon, C.; Sherman, P.M. Probiotics normalize the gut-brain-microbiota axis in immunodeficient mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G793–G802. [Google Scholar] [CrossRef]
  39. Bercik, P.; Park, A.; Sinclair, D.; Khoshdel, A.; Lu, J.; Huang, X.; Deng, Y.; Blennerhassett, P.; Fahnestock, M.; Moine, D. The anxiolytic effect of Bifidobacterium longum ncc3001 involves vagal pathways for gut–brain communication. J. Neurogastroenterol. Motil. 2011, 23, 1132–1139. [Google Scholar] [CrossRef] [Green Version]
  40. Desbonnet, L.; Garrett, L.; Clarke, G.; Kiely, B.; Cryan, J.F.; Dinan, T.G. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience 2010, 170, 1179–1188. [Google Scholar] [CrossRef] [PubMed]
  41. Liang, S.; Wang, T.; Hu, X.; Luo, J.; Li, W.; Wu, X.; Duan, Y.; Jin, F. Administration of Lactobacillus helveticus ns8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015, 310, 561–577. [Google Scholar] [CrossRef] [PubMed]
  42. Barrera-Bugueño, C.; Realini, O.; Escobar-Luna, J.; Sotomayor-Zárate, R.; Gotteland, M.; Julio-Pieper, M.; Bravo, J.A. Anxiogenic effects of a Lactobacillus, inulin and the synbiotic on healthy juvenile rats. Neuroscience 2017, 359, 18–29. [Google Scholar] [CrossRef]
  43. Desbonnet, L.; Garrett, L.; Clarke, G.; Bienenstock, J.; Dinan, T.G. The probiotic bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J. Psychiatry Res. 2009, 43, 164–174. [Google Scholar] [CrossRef]
  44. Kelly, J.R.; Allen, A.P.; Temko, A.; Hutch, W.; Kennedy, P.J.; Farid, N.; Murphy, E.; Boylan, G.; Bienenstock, J.; Cryan, J.F.; et al. Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (jb-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain Behav. Immun. 2017, 61, 50–59. [Google Scholar] [CrossRef] [PubMed]
  45. Simrén, M.; Ohman, L.; Olsson, J.; Svensson, U.; Ohlson, K.; Posserud, I.; Strid, H. Clinical trial: The effects of a fermented milk containing three probiotic bacteria in patients with irritable bowel syndrome—A randomized, double-blind, controlled study. Aliment. Pharmacol. Ther. 2010, 31, 218–227. [Google Scholar] [CrossRef]
  46. Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315–320. [Google Scholar] [CrossRef]
  47. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Messaoudi, M.; Violle, N.; Bisson, J.-F.; Desor, D.; Javelot, H.; Rougeot, C. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus r0052 and Bifidobacterium longum r0175) in healthy human volunteers. Gut Microbes 2011, 2, 256–261. [Google Scholar] [CrossRef] [Green Version]
  49. Steenbergen, L.; Sellaro, R.; van Hemert, S.; Bosch, J.A.; Colzato, L.S. A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav. Immun. 2015, 48, 258–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2006, 61, 355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Leclercq, S.; Mian, F.M.; Stanisz, A.M.; Bindels, L.B.; Cambier, E.; Ben-Amram, H.; Koren, O.; Forsythe, P.; Bienenstock, J. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat. Commun. 2017, 8, 1–12. [Google Scholar] [CrossRef]
  53. Bercik, P.; Denou, E.; Collins, J.; Jackson, W.; Lu, J.; Jury, J.; Deng, Y.; Blennerhassett, P.; Macri, J.; McCoy, K.D. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 2011, 141, 599–609.e593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Desbonnet, L.; Clarke, G.; Traplin, A.; O’Sullivan, O.; Crispie, F.; Moloney, R.D.; Cotter, P.D.; Dinan, T.G.; Cryan, J.F. Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain Behav. Immun. 2015, 48, 165–173. [Google Scholar] [CrossRef]
  55. Pirbaglou, M.; Katz, J.; de Souza, R.J.; Stearns, J.C.; Motamed, M.; Ritvo, P. Probiotic supplementation can positively affect anxiety and depressive symptoms: A systematic review of randomized controlled trials. Nutr. Res. 2016, 36, 889–898. [Google Scholar] [CrossRef] [PubMed]
  56. Galley, J.D.; Nelson, M.C.; Yu, Z.; Dowd, S.E.; Walter, J.; Kumar, P.S.; Lyte, M.; Bailey, M.T. Exposure to a social stressor disrupts the community structure of the colonic mucosa-associated microbiota. BMC Microbiol. 2014, 14, 189. [Google Scholar] [CrossRef] [Green Version]
  57. Gautam, A.; Kumar, R.; Chakraborty, N.; Muhie, S.; Hoke, A.; Hammamieh, R.; Jett, M. Altered fecal microbiota composition in all male aggressor-exposed rodent model simulating features of post-traumatic stress disorder. J. Neurosci. Res. 2018, 96, 1311–1323. [Google Scholar] [CrossRef]
  58. Tsilimigras, M.C.B.; Gharaibeh, R.Z.; Sioda, M.; Gray, L.; Fodor, A.A.; Lyte, M. Interactions between stress and sex in microbial responses within the microbiota-gut-brain axis in a mouse model. Psychosom. Med. 2018, 80, 361–369. [Google Scholar] [CrossRef] [PubMed]
  59. Gareau, M.G.; Jury, J.; MacQueen, G.; Sherman, P.M.; Perdue, M.H. Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut 2007, 56, 1522. [Google Scholar] [CrossRef] [Green Version]
  60. Bailey, M.T.; Dowd, S.E.; Parry, N.M.; Galley, J.D.; Schauer, D.B.; Lyte, M. Stressor exposure disrupts commensal microbial populations in the intestines and leads to increased colonization by Citrobacter rodentium. Infect. Immun. 2010, 78, 1509–1519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the microbiota-gut-brain axis: Prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, C.; Fujita, Y.; Ren, Q.; Ma, M.; Dong, C.; Hashimoto, K. Bifidobacterium in the gut microbiota confer resilience to chronic social defeat stress in mice. Sci. Rep. 2017, 7, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Galley, J.D.; Parry, N.M.; Ahmer, B.M.M.; Fox, J.G.; Bailey, M.T. The commensal microbiota exacerbate infectious colitis in stressor-exposed mice. Brain Behav. Immun. 2017, 60, 44–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. McVey Neufeld, K.-A.; O’Mahony, S.M.; Hoban, A.E.; Waworuntu, R.V.; Berg, B.M.; Dinan, T.G.; Cryan, J.F. Neurobehavioural effects of Lactobacillus rhamnosus gg alone and in combination with prebiotics polydextrose and galactooligosaccharide in male rats exposed to early-life stress. Nutr. Neurosci. 2019, 22, 425–434. [Google Scholar] [CrossRef]
  65. Gur, T.L.; Shay, L.; Palkar, A.V.; Fisher, S.; Varaljay, V.A.; Dowd, S.; Bailey, M.T. Prenatal stress affects placental cytokines and neurotrophins, commensal microbes, and anxiety-like behavior in adult female offspring. Brain Behav. Immun. 2017, 64, 50–58. [Google Scholar] [CrossRef]
  66. Jašarević, E.; Howard, C.D.; Misic, A.M.; Beiting, D.P.; Bale, T.L. Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci. Rep. 2017, 7, 44182. [Google Scholar] [CrossRef] [PubMed]
  67. Bailey, M.T.; Coe, C.L. Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev. Psychobiol. 1999, 35, 146–155. [Google Scholar] [CrossRef]
  68. Holdeman, L.V.; Good, I.J.; Moore, W.E. Human fecal flora: Variation in bacterial composition within individuals and a possible effect of emotional stress. App. Environ. Microbiol. 1976, 31, 359. [Google Scholar] [CrossRef] [Green Version]
  69. Zijlmans, M.A.C.; Korpela, K.; Riksen-Walraven, J.M.; de Vos, W.M.; de Weerth, C. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 2015, 53, 233–245. [Google Scholar] [CrossRef]
  70. Mayer, E.A. The neurobiology of stress and gastrointestinal disease. Gut 2000, 47, 861. [Google Scholar] [CrossRef] [Green Version]
  71. Castagliuolo, I.; Lamont, J.T.; Qiu, B.; Fleming, S.M.; Bhaskar, K.R.; Nikulasson, S.T.; Kornetsky, C.; Pothoulakis, C. Acute stress causes mucin release from rat colon: Role of corticotropin releasing factor and mast cells. Am. J. Physiol. Gastrointest. Liver Physiol. 1996, 271, G884–G892. [Google Scholar] [CrossRef]
  72. Santos, J.; Yang, P.; Soderholm, J.; Benjamin, M.; Perdue, M. Role of mast cells in chronic stress induced colonic epithelial barrier dysfunction in the rat. Gut 2001, 48, 630–636. [Google Scholar] [CrossRef] [Green Version]
  73. Lyte, M.; Vulchanova, L.; Brown, D.R. Stress at the intestinal surface: Catecholamines and mucosa-bacteria interactions. Cell Tissue Res. 2011, 343, 23–32. [Google Scholar] [CrossRef] [PubMed]
  74. Spitz, J.; Hecht, G.; Taveras, M.; Aoys, E.; Alverdy, J. The effect of dexamethasone administration on rat intestinal permeability: The role of bacterial adherence. Gastroenterology 1994, 106, 35–41. [Google Scholar] [CrossRef]
  75. Vogelzangs, N.; Beekman, A.T.F.; de Jonge, P.; Penninx, B.W.J.H. Anxiety disorders and inflammation in a large adult cohort. Transl. Psychiatry 2013, 3, e249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kohler, C.A.; Freitas, T.H.; Maes, M.; de Andrade, N.Q.; Liu, C.S.; Fernandes, B.S.; Stubbs, B.; Solmi, M.; Veronese, N.; Herrmann, N.; et al. Peripheral cytokine and chemokine alterations in depression: A meta-analysis of 82 studies. Acta Psychiatry Scand. 2017, 135, 373–387. [Google Scholar] [CrossRef]
  77. O’Brien, S.M.; Scully, P.; Fitzgerald, P.; Scott, L.V.; Dinan, T.G. Plasma cytokine profiles in depressed patients who fail to respond to selective serotonin reuptake inhibitor therapy. J. Psychiatry Res. 2007, 41, 326–331. [Google Scholar] [CrossRef] [PubMed]
  78. Mikocka-Walus, A.; Knowles, S.R.; Keefer, L.; Graff, L. Controversies revisited: A systematic review of the comorbidity of depression and anxiety with inflammatory bowel diseases. Inflamm. Bowel Dis. 2016, 22, 752–762. [Google Scholar] [CrossRef]
  79. Bajaj, J.S.; Hylemon, P.B.; Ridlon, J.M.; Heuman, D.M.; Daita, K.; White, M.B.; Monteith, P.; Noble, N.A.; Sikaroodi, M.; Gillevet, P.M. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G675–G685. [Google Scholar] [CrossRef]
  80. De Angelis, M.; Francavilla, R.; Piccolo, M.; De Giacomo, A.; Gobbetti, M. Autism spectrum disorders and intestinal microbiota. Gut Microbes 2015, 6, 207–213. [Google Scholar] [CrossRef] [Green Version]
  81. Shen, L.; Liu, L.; Ji, H.-F. Alzheimer’s disease histological and behavioral manifestations in transgenic mice correlate with specific gut microbiome state. J. Alzheimer’s Dis. 2017, 56, 385–390. [Google Scholar] [CrossRef]
  82. Goehler, L.E.; Gaykema, R.P.; Opitz, N.; Reddaway, R.; Badr, N.; Lyte, M. Activation in vagal afferents and central autonomic pathways: Early responses to intestinal infection with campylobacter jejuni. Brain Behav. Immun. 2005, 19, 334–344. [Google Scholar] [CrossRef]
  83. Gaykema, R.P.; Goehler, L.E.; Tilders, F.J.; Bol, J.G.; McGorry, M.; Fleshner, M.; Maier, S.F.; Watkins, L.R. Bacterial endotoxin induces fos immunoreactivity in primary afferent neurons of the vagus nerve. Neuroimmunomodulation 1998, 5, 234–240. [Google Scholar] [CrossRef] [PubMed]
  84. Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the cns. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef]
  85. Gaykema, R.P.; Goehler, L.E.; Lyte, M. Brain response to cecal infection with campylobacter jejuni: Analysis with fos immunohistochemistry. Brain Behav. Immun. 2004, 18, 238–245. [Google Scholar] [CrossRef]
  86. Rajkumar, H.; Kumar, M.; Das, N.; Kumar, S.N.; Challa, H.R.; Nagpal, R. Effect of probiotic Lactobacillus salivarius ubl s22 and prebiotic fructo-oligosaccharide on serum lipids, inflammatory markers, insulin sensitivity, and gut bacteria in healthy young volunteers: A randomized controlled single-blind pilot study. J. Cardiovasc. Pharmacol. Ther. 2015, 20, 289–298. [Google Scholar] [CrossRef] [PubMed]
  87. Ait-Belgnaoui, A.; Durand, H.; Cartier, C.; Chaumaz, G.; Eutamene, H.; Ferrier, L.; Houdeau, E.; Fioramonti, J.; Bueno, L.; Theodorou, V. Prevention of gut leakiness by a probiotic treatment leads to attenuated hpa response to an acute psychological stress in rats. Psychoneuroendocrinology 2012, 37, 1885–1895. [Google Scholar] [CrossRef] [PubMed]
  88. Kittana, H.; Gomes-Neto, J.C.; Heck, K.; Geis, A.L.; Segura Muñoz, R.R.; Cody, L.A.; Schmaltz, R.J.; Bindels, L.B.; Sinha, R.; Hostetter, J.M.; et al. Commensal Escherichia coli strains can promote intestinal inflammation via differential interleukin-6 production. Front. Immunol. 2018, 9, 2318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Maes, M.; Kubera, M.; Leunis, J.-C. The gut-brain barrier in major depression: Intestinal mucosal dysfunction with an increased translocation of lps from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol. Lett. 2008, 29, 117–124. [Google Scholar]
  90. Bharwani, A.; Mian, M.F.; Surette, M.G.; Bienenstock, J.; Forsythe, P. Oral treatment with Lactobacillus rhamnosus attenuates behavioural deficits and immune changes in chronic social stress. BMC Med. 2017, 15, 7. [Google Scholar] [CrossRef] [Green Version]
  91. Smith, P.; Garrett, W. The gut microbiota and mucosal t cells. Front. Microbiol. 2011, 2, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Qiu, B.S.; Vallance, B.A.; Blennerhassett, P.A.; Collins, S.M. The role of cd4+ lymphocytes in the susceptibility of mice to stress-induced reactivation of experimental colitis. Nat. Med. 1999, 5, 1178–1182. [Google Scholar] [CrossRef] [PubMed]
  93. Maes, M.; Kubera, M.; Leunis, J.C.; Berk, M.; Geffard, M.; Bosmans, E. In depression, bacterial translocation may drive inflammatory responses, oxidative and nitrosative stress (o&ns), and autoimmune responses directed against o&ns-damaged neoepitopes. Acta Psychiatry Scand. 2012, 127, 344–354. [Google Scholar]
  94. Demaude, J.; Salvador-Cartier, C.; Fioramonti, J.; Ferrier, L.; Bueno, L. Phenotypic changes in colonocytes following acute stress or activation of mast cells in mice: Implications for delayed epithelial barrier dysfunction. Gut 2006, 55, 655. [Google Scholar] [CrossRef]
  95. Saunders, P.R.; Kosecka, U.; McKay, D.M.; Perdue, M.H. Acute stressors stimulate ion secretion and increase epithelial permeability in rat intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 1994, 267, G794–G799. [Google Scholar] [CrossRef] [PubMed]
  96. Lasselin, J.; Elsenbruch, S.; Lekander, M.; Axelsson, J.; Karshikoff, B.; Grigoleit, J.-S.; Engler, H.; Schedlowski, M.; Benson, S. Mood disturbance during experimental endotoxemia: Predictors of state anxiety as a psychological component of sickness behavior. Brain Behav. Immun. 2016, 57, 30–37. [Google Scholar] [CrossRef]
  97. Zhao, Q.Y.; Wu, X.H.; Yan, S.; Xie, X.F.; Fan, Y.H.; Zhang, J.Q.; Peng, C.; You, Z.L. The antidepressant-like effects of pioglitazone in a chronic mild stress mouse model are associated with ppar gamma-mediated alteration of microglial activation phenotypes. J. Neuroinflamm. 2016, 13, 259. [Google Scholar] [CrossRef] [Green Version]
  98. Qin, L.; Wu, X.; Block, M.L.; Liu, Y.; Breese, G.R.; Hong, J.-S.; Knapp, D.J.; Crews, F.T. Systemic lps causes chronic neuroinflammation and progressive neurodegeneration. Glia 2007, 55, 453–462. [Google Scholar] [CrossRef] [Green Version]
  99. Maes, M.; Kubera, M.; Leunis, J.C.; Berk, M. Increased iga and igm responses against gut commensals in chronic depression: Further evidence for increased bacterial translocation or leaky gut. J. Affect. Disord. 2012, 141, 55–62. [Google Scholar] [CrossRef]
  100. Kiliaan, A.J.; Saunders, P.R.; Bijlsma, P.B.; Berin, M.C.; Taminiau, J.A.; Groot, J.A.; Perdue, M.H. Stress stimulates transepithelial macromolecular uptake in rat jejunum. Am. J. Physiol. Gastrointest. Liver Physiol. 1998, 275, G1037. [Google Scholar] [CrossRef] [PubMed]
  101. Meddings, J.B.; Swain, M.G. Environmental stress–induced gastrointestinal permeability is mediated by endogenous glucocorticoids in the rat. Gastroenterology 2000, 119, 1019–1028. [Google Scholar] [CrossRef]
  102. Soderholm, J.D.; Yang, P.C.; Ceponis, P.; Vohra, A.; Riddell, R.; Sherman, P.M.; Perdue, M.H. Chronic stress induces mast cell-dependent bacterial adherence and initiates mucosal inflammation in rat intestine. Gastroenterology 2002, 123, 1099–1108. [Google Scholar] [CrossRef]
  103. Bhatia, V.; Tandon, R.K. Stress and the gastrointestinal tract. J. Gastroenterol. Hepatol. 2005, 20, 332–339. [Google Scholar] [CrossRef] [PubMed]
  104. Leclercq, S.; Matamoros, S.; Cani, P.D.; Neyrinck, A.M.; Jamar, F.; Starkel, P.; Windey, K.; Tremaroli, V.; Backhed, F.; Verbeke, K.; et al. Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc. Natl. Acad. Sci. USA 2014, 111, E4485–E4493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Eutamene, H.; Lamine, F.; Chabo, C.; Theodorou, V.; Rochat, F.; Bergonzelli, G.E.; Corthésy-Theulaz, I.; Fioramonti, J.; Bueno, L. Synergy between Lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J. Nutr. 2007, 137, 1901–1907. [Google Scholar] [CrossRef]
  106. Arborelius, L.; Owens, M.; Plotsky, P.; Nemeroff, C. The role of corticotropin-releasing factor in depression and anxiety disorders. J. Endocrin. 1999, 160, 1–12. [Google Scholar] [CrossRef] [Green Version]
  107. Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.-N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef] [PubMed]
  108. Pusceddu, M.M.; El Aidy, S.; Crispie, F.; O’Sullivan, O.; Cotter, P.; Stanton, C.; Kelly, P.; Cryan, J.F.; Dinan, T.G. N-3 polyunsaturated fatty acids (pufas) reverse the impact of early-life stress on the gut microbiota. PLoS ONE 2015, 10, e0139721. [Google Scholar] [CrossRef]
  109. Pusceddu, M.M.; Kelly, P.; Ariffin, N.; Cryan, J.F.; Clarke, G.; Dinan, T.G. N-3 pufas have beneficial effects on anxiety and cognition in female rats: Effects of early life stress. Psychoneuroendocrinology 2015, 58, 79–90. [Google Scholar] [CrossRef]
  110. Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatry Res. 2016, 82, 109–118. [Google Scholar] [CrossRef] [PubMed]
  111. Silverman, M.N.; Pearce, B.D.; Biron, C.A.; Miller, A.H. Immune modulation of the hypothalamic-pituitary-adrenal (hpa) axis during viral infection. Viral Immunol. 2005, 18, 41–78. [Google Scholar] [CrossRef]
  112. Turnbull, A.V.; Rivier, C.L. Regulation of the hypothalamic-pituitary-adrenal axis by cytokines: Actions and mechanisms of action. Physiol. Rev. 1999, 79, 1–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Silverman, M.N.; Sternberg, E.M. Glucocorticoid regulation of inflammation and its functional correlates: From hpa axis to glucocorticoid receptor dysfunction. Ann. N. Y. Acad. Sci. 2012, 1261, 55–63. [Google Scholar] [CrossRef]
  114. Asano, Y.; Hiramoto, T.; Nishino, R.; Aiba, Y.; Kimura, T.; Yoshihara, K.; Koga, Y.; Sudo, N. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G1288–G1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Tsavkelova, E.; Klimova, S.Y.; Cherdyntseva, T.; Netrusov, A. Hormones and hormone-like substances of microorganisms: A review. Appl. Biochem. Microbiol. 2006, 42, 229–235. [Google Scholar] [CrossRef]
  116. Roshchina, V.V. Evolutionary considerations of neurotransmitters in microbial, plant, and animal cells. In Microbial Endocrinology; Springer: New York, NY, USA, 2010; pp. 17–52. [Google Scholar]
  117. Ross, R.P.; Mills, S.; Hill, C.; Fitzgerald, G.F.; Stanton, C. Specific metabolite production by gut microbiota as a basis for probiotic function. Int. Dairy J. 2010, 20, 269. [Google Scholar] [CrossRef]
  118. Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. Bioessays 2011, 33, 574–581. [Google Scholar] [CrossRef]
  119. Holzer, P.; Farzi, A. Neuropeptides and the microbiota-gut-brain axis. In Microbial Endocrinology: The microbiota-Gut-Brain Axis in Health and Disease; Lyte, M., Cryan, J.F., Eds.; Springer: New York, NY, USA, 2014; Volume 817, pp. 195–219. [Google Scholar]
  120. Matsumoto, M.; Kibe, R.; Ooga, T.; Aiba, Y.; Kurihara, S.; Sawaki, E.; Koga, Y.; Benno, Y. Impact of intestinal microbiota on intestinal luminal metabolome. Sci. Rep. 2012, 2, 233. [Google Scholar] [CrossRef] [Green Version]
  121. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; David, H.M.; et al. Activation of gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor gpr43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef]
  124. Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef] [PubMed]
  125. Skonieczna-Zydecka, K.; Grochans, E.; Maciejewska, D.; Szkup, M.; Schneider-Matyka, D.; Jurczak, A.; Loniewski, I.; Kaczmarczyk, M.; Marlicz, W.; Czerwinska-Rogowska, M.; et al. Faecal short chain fatty acids profile is changed in polish depressive women. Nutrients 2018, 10, 1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Maltz, R.M.; Keirsey, J.; Kim, S.C.; Mackos, A.R.; Gharaibeh, R.Z.; Moore, C.C.; Xu, J.; Somogyi, A.; Bailey, M.T. Social stress affects colonic inflammation, the gut microbiome, and short-chain fatty acid levels and receptors. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 533–540. [Google Scholar] [CrossRef] [PubMed]
  127. van de Wouw, M.; Boehme, M.; Lyte, J.M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain–gut axis alterations. J. Physiol. 2018, 596, 4923–4944. [Google Scholar] [CrossRef] [PubMed]
  128. Bharwani, A.; Mian, M.F.; Foster, J.A.; Surette, M.G.; Bienenstock, J.; Forsythe, P. Structural & functional consequences of chronic psychosocial stress on the microbiome & host. Psychoneuroendocrinology 2016, 63, 217–227. [Google Scholar] [PubMed] [Green Version]
  129. Michels, N.; Van de Wiele, T.; De Henauw, S. Chronic psychosocial stress and gut health in children: Associations with calprotectin and fecal short-chain fatty acids. Psychosom. Med. 2017, 79, 927–935. [Google Scholar] [CrossRef]
  130. Marques, A.H.; Silverman, M.N.; Sternberg, E.M. Evaluation of stress systems by applying noninvasive methodologies: Measurements of neuroimmune biomarkers in the sweat, heart rate variability and salivary cortisol. Neuroimmunomodulation 2010, 17, 205–208. [Google Scholar] [CrossRef] [Green Version]
  131. West, C.; Wu, R.Y.; Wong, A.; Stanisz, A.M.; Yan, R.; Min, K.K.; Pasyk, M.; McVey Neufeld, K.A.; Karamat, M.I.; Foster, J.A.; et al. Lactobacillus rhamnosus strain jb-1 reverses restraint stress-induced gut dysmotility. Neurogastroenterol. Motil 2017, 29, e12903. [Google Scholar] [CrossRef]
  132. Dalile, B.; Vervliet, B.; Bergonzelli, G.; Verbeke, K.; Van Oudenhove, L. Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: A randomized, placebo-controlled trial. Neuropsychopharmacology 2020, 45, 2257–2266. [Google Scholar] [CrossRef]
  133. Bassett, S.A.; Young, W.; Fraser, K.; Dalziel, J.E.; Webster, J.; Ryan, L.; Fitzgerald, P.; Stanton, C.; Dinan, T.G.; Cryan, J.F.; et al. Metabolome and microbiome profiling of a stress-sensitive rat model of gut-brain axis dysfunction. Sci. Rep. 2019, 9, 14026. [Google Scholar] [CrossRef] [PubMed]
  134. Hill, M. Intestinal flora and endogenous vitamin synthesis. Eur. J. Cancer Prev. 1997, 6, S43–S45. [Google Scholar] [CrossRef]
  135. Burgess, C.M.; Smid, E.J.; Van Sinderen, D. Bacterial vitamin b2, b11 and b12 overproduction: An overview. Int. J. Food. Microbiol. 2009, 133, 1–7. [Google Scholar] [CrossRef] [PubMed]
  136. Sumi, Y.; Miyakawa, M.; Kanzaki, M.; Kotake, Y. Vitamin b-6 deficiency in germfree rats. J. Nutr. 1977, 107, 1707–1714. [Google Scholar] [CrossRef] [Green Version]
  137. LeBlanc, J.G.; Milani, C.; de Giori, G.S.; Sesma, F.; van Sinderen, D.; Ventura, M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Curr. Opin. Biotech. 2013, 24, 160–168. [Google Scholar] [CrossRef]
  138. Rosenberg, J.; Ischebeck, T.; Commichau, F.M. Vitamin b6 metabolism in microbes and approaches for fermentative production. Biotechnol. Adv. 2017, 35, 31–40. [Google Scholar] [CrossRef] [PubMed]
  139. Gougeon, L.; Payette, H.; Morais, J.A.; Gaudreau, P.; Shatenstein, B.; Gray-Donald, K. Intakes of folate, vitamin b6 and b12 and risk of depression in community-dwelling older adults: The quebec longitudinal study on nutrition and aging. Eur. J. Clin. Nutr. 2016, 70, 380–385. [Google Scholar] [CrossRef] [PubMed]
  140. Jacka, F.N.; Maes, M.; Pasco, J.A.; Williams, L.J.; Berk, M. Nutrient intakes and the common mental disorders in women. J. Affect. Disord. 2012, 141, 79–85. [Google Scholar] [CrossRef] [PubMed]
  141. Vulser, H.; Wiernik, E.; Hoertel, N.; Thomas, F.; Pannier, B.; Czernichow, S.; Hanon, O.; Simon, T.; Simon, J.M.; Danchin, N.; et al. Association between depression and anemia in otherwise healthy adults. Acta Psychiatry Scand. 2016, 134, 150–160. [Google Scholar] [CrossRef]
  142. Alpert, J.E.; Fava, M. Nutrition and depression: The role of folate. Nutr. Rev. 1997, 55, 145–149. [Google Scholar] [CrossRef]
  143. Coppen, A.; Bolander-Gouaille, C. Treatment of depression: Time to consider folic acid and vitamin b12. J. Psychopharm. 2005, 19, 59–65. [Google Scholar] [CrossRef]
  144. Paul, R.T.P.; McDonnell, A.P.; Kelly, C.B. Folic acid: Neurochemistry, metabolism and relationship to depression. Hum. Psychopharm. Clin. 2004, 19, 477–488. [Google Scholar] [CrossRef]
  145. Spedding, S. Vitamin d and depression: A systematic review and meta-analysis comparing studies with and without biological flaws. Nutrients 2014, 6, 1501–1518. [Google Scholar] [CrossRef] [PubMed]
  146. Partonen, T. Vitamin d and serotonin in winter. Med. Hypotheses 1998, 51, 267–268. [Google Scholar] [CrossRef]
  147. Hartvig, P.; Lindner, K.J.; Bjurling, P.; Långström, B.; Tedroff, J. Pyridoxine effect on synthesis rate of serotonin in the monkey brain measured with positron emission tomography. J. Neural Transm. 1995, 102, 91–97. [Google Scholar] [CrossRef]
  148. Dakshinamurti, K.; Sharma, S.K.; Bonke, D. Influence of b vitamins on binding properties of serotonin receptors in the cns of rats. Klin. Wochenschr. 1990, 68, 142–145. [Google Scholar] [CrossRef]
  149. Wang, J.; Um, P.; Dickerman, B.A.; Liu, J. Zinc, magnesium, selenium and depression: A review of the evidence, potential mechanisms and implications. Nutrients 2018, 10, 584. [Google Scholar] [CrossRef] [Green Version]
  150. Rybka, J.; Kedziora-Kornatowska, K.; Banas-Lezanska, P.; Majsterek, I.; Carvalho, L.A.; Cattaneo, A.; Anacker, C.; Kedziora, J. Interplay between the pro-oxidant and antioxidant systems and proinflammatory cytokine levels, in relation to iron metabolism and the erythron in depression. Free Radic. Biol. Med. 2013, 63, 187–194. [Google Scholar] [CrossRef] [PubMed]
  151. Agrawal, S.; Agrawal, A.; Said, H.M. Biotin deficiency enhances the inflammatory response of human dendritic cells. Am. J. Physiol. Cell Physiol. 2016, 311, C386–C391. [Google Scholar] [CrossRef] [PubMed]
  152. Pfalzer, A.C.; Choi, S.-W.; Tammen, S.A.; Park, L.K.; Bottiglieri, T.; Parnell, L.D.; Lamon-Fava, S. S-adenosylmethionine mediates inhibition of inflammatory response and changes in DNA methylation in human macrophages. Physiol. Genom. 2014, 46, 617–623. [Google Scholar] [CrossRef]
  153. Myint, A.-M.; Kim, Y.K.; Verkerk, R.; Scharpé, S.; Steinbusch, H.; Leonard, B. Kynurenine pathway in major depression: Evidence of impaired neuroprotection. J. Affect. Disord. 2007, 98, 143–151. [Google Scholar] [CrossRef]
  154. Takeuchi, F.; Shibata, Y. Kynurenine metabolism in vitamin-b-6-deficient rat liver after tryptophan injection. Biochem. J. 1984, 220, 693–699. [Google Scholar] [CrossRef] [Green Version]
  155. Willner, P.; Scheel-Kruger, J.; Belzung, C. The neurobiology of depression and antidepressant action. Neurosci. Biobehav. Rev. 2013, 37, 2331–2371. [Google Scholar] [CrossRef] [PubMed]
  156. Gershon, M.D. 5-hydroxytryptamine (serotonin) in the gastrointestinal tract. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 14–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Sjögren, K.; Engdahl, C.; Henning, P.; Lerner, U.H.; Tremaroli, V.; Lagerquist, M.K.; Bäckhed, F.; Ohlsson, C. The gut microbiota regulates bone mass in mice. J. Bone Miner. Res. 2012, 27, 1357–1367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Mazmanian, S.K.; Hsiao, E.Y.; Ma, L.; Ismagilov, R.F.; Nagler, C.R. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F.; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 1395–1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Fukumoto, S.; Tatewaki, M.; Yamada, T.; Fujimiya, M.; Mantyh, C.; Voss, M.; Eubanks, S.; Harris, M.; Pappas, T.N.; Takahashi, T. Short-chain fatty acids stimulate colonic transit via intraluminal 5-ht release in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2003, 284, R1269. [Google Scholar] [CrossRef] [Green Version]
  161. Krantis, A. Gaba in the mammalian enteric nervous system. Physiology 2000, 15, 284–290. [Google Scholar] [CrossRef]
  162. 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. [Google Scholar] [CrossRef] [Green Version]
  163. Wikoff, W.R.; Anfora, A.T.; Liu, J.; Schultz, P.G.; Lesley, S.A.; Peters, E.C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. USA 2009, 106, 3698. [Google Scholar] [CrossRef] [Green Version]
  164. Young, L.W.; Darios, E.S.; Watts, S.W. An immunohistochemical analysis of sert in the blood–brain barrier of the male rat brain. Histochem. Cell Biol. 2015, 144, 321–329. [Google Scholar] [CrossRef] [Green Version]
  165. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. de Jonge, W.J. The gut’s little brain in control of intestinal immunity. ISRN Gastroenterol. 2013, 2013, 630159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Bjurstöm, H.; Wang, J.; Ericsson, I.; Bengtsson, M.; Liu, Y.; Kumar-Mendu, S.; Issazadeh-Navikas, S.; Birnir, B. Gaba, a natural immunomodulator of t lymphocytes. J. Neuroimmunol. 2008, 205, 44–50. [Google Scholar] [CrossRef]
  168. O’Connor, J.C.; Lawson, M.A.; Andre, C.; Moreau, M.; Lestage, J.; Castanon, N.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Mol. Psychiatry 2008, 14, 511–522. [Google Scholar] [CrossRef] [Green Version]
  169. Ondicova, K.; Pecenak, J.; Mravec, B. The role of the vagus nerve in depression. Neuro Endocrinol. Lett. 2010, 31, 602–608. [Google Scholar]
  170. Agelink, M.W.; Boz, C.; Ullrich, H.; Andrich, J. Relationship between major depression and heart rate variability.: Clinical consequences and implications for antidepressive treatment. Psychiatry Res. 2002, 113, 139–149. [Google Scholar] [CrossRef]
  171. Schlaepfer, T.E.; Frick, C.; Zobel, A.; Maier, W.; Heuser, I.; Bajbouj, M.; O’Keane, V.; Corcoran, C.; Adolfsson, R.; Trimble, M.; et al. Vagus nerve stimulation for depression: Efficacy and safety in a european study. Psychol. Med. 2008, 38, 651–661. [Google Scholar] [CrossRef] [Green Version]
  172. Lv, H.; Zhao, Y.-H.; Chen, J.-G.; Wang, D.-Y.; Chen, H. Vagus nerve stimulation for depression: A systematic review. Front. Psychol. 2019, 10, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Goehler, L.E.; Gaykema, R.P.; Hansen, M.K.; Anderson, K.; Maier, S.F.; Watkins, L.R. Vagal immune-to-brain communication: A visceral chemosensory pathway. Autonom. Neurosci. 2000, 85, 49–59. [Google Scholar] [CrossRef]
  174. Tracey, K.J. The inflammatory reflex. Nature 2002, 420, 853–859. [Google Scholar] [CrossRef] [PubMed]
  175. Pavlov, V.A.; Tracey, K.J. The vagus nerve and the inflammatory reflex--linking immunity and metabolism. Nat. Rev. Endocrinol. 2012, 8, 743–754. [Google Scholar] [CrossRef] [PubMed]
  176. Daskalakis, N.P.; Bagot, R.C.; Parker, K.J.; Vinkers, C.H.; de Kloet, E.R. The three-hit concept of vulnerability and resilience: Toward understanding adaptation to early-life adversity outcome. Psychoneuroendocrinology 2013, 38, 1858–1873. [Google Scholar] [CrossRef] [Green Version]
  177. Tanida, M.; Yamano, T.; Maeda, K.; Okumura, N.; Fukushima, Y.; Nagai, K. Effects of intraduodenal injection of Lactobacillus johnsonii la1 on renal sympathetic nerve activity and blood pressure in urethane-anesthetized rats. Neurosci. Lett. 2005, 389, 109–114. [Google Scholar] [CrossRef]
  178. Sominsky, L.; Fuller, E.A.; Bondarenko, E.; Ong, L.K.; Averell, L.; Nalivaiko, E.; Dunkley, P.R.; Dickson, P.W.; Hodgson, D.M. Functional programming of the autonomic nervous system by early life immune exposure: Implications for anxiety. PLoS ONE 2013, 8, e57700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Réus, G.Z.; Stringari, R.B.; Ribeiro, K.F.; Cipriano, A.L.; Panizzutti, B.S.; Stertz, L.; Lersch, C.; Kapczinski, F.; Quevedo, J. Maternal deprivation induces depressive-like behaviour and alters neurotrophin levels in the rat brain. Neurochem. Res. 2011, 36, 460–466. [Google Scholar] [CrossRef] [PubMed]
  180. Matthews, K.; Robbins, T.W. Early experience as a determinant of adult behavioural responses to reward: The effects of repeated maternal separation in the rat. Neurosci. Biobehav. Rev. 2003, 27, 45–55. [Google Scholar] [CrossRef]
  181. Montalvo-Ortiz, J.L.; Bordner, K.A.; Carlyle, B.C.; Gelernter, J.; Simen, A.A.; Kaufman, J. The role of genes involved in stress, neural plasticity, and brain circuitry in depressive phenotypes: Convergent findings in a mouse model of neglect. Behav. Brain Res. 2016, 315, 71–74. [Google Scholar] [CrossRef] [Green Version]
  182. Yohn, N.L.; Blendy, J.A. Adolescent chronic stress and adult anxiety adolescent chronic unpredictable stress exposure is a sensitive window for long-term changes in adult behavior in mice. Neuropsychopharmacology 2017, 42, 1670–1678. [Google Scholar] [CrossRef] [Green Version]
  183. Jaffee, S.R. Sensitive, stimulating caregiving predicts cognitive and behavioral resilience in neurodevelopmentally at-risk infants. Dev. Psychopathol. 2007, 19, 631–647. [Google Scholar] [CrossRef] [Green Version]
  184. Del Giudice, M.; Ellis, B.J.; Shirtcliff, E.A. The adaptive calibration model of stress responsivity. Neurosci. Biobehav. Rev. 2011, 35, 1562–1592. [Google Scholar] [CrossRef] [Green Version]
  185. Nesse, R.M.; Bhatnagar, S.; Ellis, B. Chapter 11—Evolutionary origins and functions of the stress response system. In Stress: Concepts, Cognition, Emotion, and Behavior; Fink, G., Ed.; Academic Press: San Diego, CA, USA, 2016; pp. 95–101. [Google Scholar]
  186. Heim, C.; Binder, E.B. Current research trends in early life stress and depression: Review of human studies on sensitive periods, gene–environment interactions, and epigenetics. Exp. Neurol. 2012, 233, 102–111. [Google Scholar] [CrossRef] [PubMed]
  187. Schapiro, S. Some physiological, biochemical, and behavioral consequences of neonatal hormone administration: Cortisol and thyroxine. Gen. Comp. Endocrinol. 1968, 10, 214–228. [Google Scholar] [CrossRef]
  188. Tambs, K.; Czajkowsky, N.; Røysamb, E.; Neale, M.C.; Reichborn-Kjennerud, T.; Aggen, S.H.; Harris, J.R.; Ørstavik, R.E.; Kendler, K.S. Structure of genetic and environmental risk factors for dimensional representations of dsm–iv anxiety disorders. Br. J. Psychiatry 2009, 195, 301–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. On Wah, D.T.; Kavaliers, M.; Bishnoi, I.R.; Ossenkopp, K.-P. Lipopolysaccharide (lps) induced sickness in early adolescence alters the behavioral effects of the short-chain fatty acid, propionic acid, in late adolescence and adulthood: Examining anxiety and startle reactivity. Behav. Brain Res. 2019, 360, 312–322. [Google Scholar] [CrossRef]
  190. Aguilera, M.; Vergara, P.; Martínez, V. Stress and antibiotics alter luminal and wall-adhered microbiota and enhance the local expression of visceral sensory-related systems in mice. J. Neurogastroenterol. Motil. 2013, 25, e515–e529. [Google Scholar] [CrossRef]
  191. Brenes, J.C.; Fornaguera, J. Effects of environmental enrichment and social isolation on sucrose consumption and preference: Associations with depressive-like behavior and ventral striatum dopamine. Neurosci. Lett. 2008, 436, 278–282. [Google Scholar] [CrossRef]
  192. Bear, T.L.K.; Dalziel, J.E.; Coad, J.; Roy, N.C.; Butts, C.A.; Gopal, P.K. The role of the gut microbiota in dietary interventions for depression and anxiety. Adv. Nutr. 2020, 11, 890–907. [Google Scholar] [CrossRef] [Green Version]
  193. Taylor, A.M.; Thompson, S.V.; Edwards, C.G.; Musaad, S.M.A.; Khan, N.A.; Holscher, H.D. Associations among diet, the gastrointestinal microbiota, and negative emotional states in adults. Nutr. Neurosci. 2019, 23, 983–992. [Google Scholar] [CrossRef]
Figure 1. The proposed mechanisms of the microbiome-gut-brain axis (MGBA) are complex and intertwined. Emerging research shows that psychological stress interacts not only directly with the brain and mood, but also with many of the MGBA mechanisms thought to contribute to changes in mood with alteration of the gut microbiota. Solid lines indicate strong evidence of an effect, and dotted lines show proposed mechanisms with limited but emerging evidence. Abbreviations: HPA; Hypothalamic-Pituitary-Adrenal.
Figure 1. The proposed mechanisms of the microbiome-gut-brain axis (MGBA) are complex and intertwined. Emerging research shows that psychological stress interacts not only directly with the brain and mood, but also with many of the MGBA mechanisms thought to contribute to changes in mood with alteration of the gut microbiota. Solid lines indicate strong evidence of an effect, and dotted lines show proposed mechanisms with limited but emerging evidence. Abbreviations: HPA; Hypothalamic-Pituitary-Adrenal.
Microorganisms 09 00723 g001
Table 1. Summary of studies testing the effects of probiotic supplements on mood. Effect on mood is indicated with: + for a positive effect on mood, − for a negative effect on mood, and / for no effect on mood. Abbreviations: LDB, Light Dark Box; SDT, Step Down Test; FST, Forced Swim Test; SPT, Sucrose Preference Test; PSS, Perceived Stress Scale; BAI, Beck Depression Inventory; BDI, Beck Anxiety Inventory; HADS, Hospital Anxiety and Depression scale; POMS, Profile of Mood State.
Table 1. Summary of studies testing the effects of probiotic supplements on mood. Effect on mood is indicated with: + for a positive effect on mood, − for a negative effect on mood, and / for no effect on mood. Abbreviations: LDB, Light Dark Box; SDT, Step Down Test; FST, Forced Swim Test; SPT, Sucrose Preference Test; PSS, Perceived Stress Scale; BAI, Beck Depression Inventory; BDI, Beck Anxiety Inventory; HADS, Hospital Anxiety and Depression scale; POMS, Profile of Mood State.
Subject, Study Design and ModelProbioticDose and AdministrationTreatment DurationEffect on Mood Reference
Animal Studies
Male AKR mice with parasite-induced (Trichuris muris) chronic gastrointestinal inflammationBifidobacterium longum NCC3001 and Lactobacillus rhamnosus NCC4007 Gavaged daily, dose not specified10 daysReduction in anxiety-like behaviors in the LDB+[29]
Immunodeficient (B and T cell-deficient) male and female Rag 1−/− miceL. rhamnosus R0011 and Lactobacillus helveticus R0052 109 CFU/mL in drinking water daily4 weeksProbiotic supplement normalized deficits in anxiety in LDB tests+[38]
Male C57BL/6 mice with liver inflammation-induced sickness behavior and brain inflammationCommercial mixture VSL#3: L. casei, L. plantarum, L. acidophilus and L. delbrueckii subsp. Bulgaricus, B. longum, B. breve and Bifidobacterium infantis, Streptococcus salivarius subsp. Thermophiles. Strains unspecified1.7 billion bacteria/day, gavaged daily10 daysPrevention of a decrease in social interaction+[37]
Male AKR mice with chemically induced colitis B. longum NCC3001 -100 µL of 1 × 1010 CFU7 daysA probiotic supplementation reduced anxiety-like behavior in SDT, but only when the vagus nerve was intact+/[39]
Male Sprague Dawley RatsB. bifidum W23, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, L. lactis W19, L. lactis W584.5 g (2.5 × 109 CFU/g) of freeze-dried powder in 30 mL of tap water per cage (2 rats) daily10 weeksA probiotic mix decreased depressive-like behavior in FST+[35]
Male Sprague Dawley rats following maternal separation stressB. infantis 35624 1 × 1010 live bacterial cells/100 mL drinking water55 daysA probiotic supplement ameriorated MSS induced depressive-like behavior in FST+[40]
A probiotic given alongside 3 weeks of restraint stress in male Sprague Dawley ratsL. helveticus ns8109 CFU/mL live bacteria in drinking water3 weeksProbiotic ameliorated stress-induced depressive-like behavior in SPT, and anxiety like behavior in EPM+[41]
Male BALB/c miceL. rhamnosus JB-1 109 CFU, gavaged daily28 daysDecreased anxiety-like behaviors in the EPM+[36]
Male Sprague Dawley ratsL. casei 54-2-33104 CFU/mL in drinking water14 daysIncrease in anxiety-like behavior in the OFT and no difference in anxiety-like behavior in the EPM[42]
Male Sprague Dawley ratsB. infantis 356241 × 1010 live bacterial cells/100 mL drinking water14 daysNo decrease in depressive-like behaviors in FST/[43]
Human Studies
Healthy adult menL. rhamnosus JB-1 109 CFU, probiotic capsule, daily8 weeksNo reduction in subjective stress measure, depression or anxiety scores on the PSS, BAI or BDI scales or improve cognitive measures/[44]
Healthy men and womenL. helveticus R0052 and B. longum R01753 × 109 CFU probiotic capsule daily30 daysReduction in depression and anxiety scores (HADS). In a subset of people with low baseline urinary cortisol, the perceived stress scores were also reduced by the probiotic+[47,48]
Healthy men and womenB. bifidum W23, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, and Lactococcus lactis (W19 and W58)2.5 × 109 CFU probiotic capsule daily4 weeksReduction in participant’s cognitive reactivity to sad mood+[49]
Men and women with chronic fatigue syndromeL. casei Shirota 8 × 109 CFU probiotic capsule daily2 monthsImproved anxiety (BAI) but not depressive (BDI) symptoms+/[50]
Healthy men and womenMilk drink containing probiotic L. casei Shirota6.5 × 109 CFU in a milk drink Improvement in mood in POMS only in those who already had low mood+/[51]
Men and women with irritable bowel syndromeYohgurt containing Lactobacillus paracasei, ssp. paracasei F19, L. acidophilus La5 and B. lactis Bb12 (Cultura; active) 5 × 107 cfu/mL × 200 mL milk drink, daily8 weeksThe probiotic yoghurt drink did not improve mood scores in HADS/[45]
Men and women with diagnosed depression B. bifidum, L. acidophilus, and L. casei (strains not specified)L. acidophilus (2 × 109 CFU/g), L.casei (2 × 109 CFU/g), B. bifidum (2 × 109 CFU/g), amount not specified8 weeksReduction in symptoms of depression I BDI, along with fasting plasma insulin, glutathione, and C-reactive protein+[46]
Table 2. Summary of studies investigating links between stress and the gut microbiota. Microbiota which show an association (not necessarily causality) with stress-resilience and stress-sensitivity are indicated in the columns labeled SR and SS, respectively. This depends on the study but may mean, e.g., a probiotic supplement which increased stress-resilience, or an increase in relative abundance of the microbiota in stress-resilient individuals. Abbreviations: EPM, Elevated Plus Maze; FST, Forced Swim Test; TST, Tail Suspension Test; HbA1c, Hemoglobin A1c; IL, Interleukin; IFN-γ, Interferon gamma; LDB, Light-Dark Box; OFT, Open Field Test; NOR, Novel Object Recognition; MSS, Maternal Separation Stress.
Table 2. Summary of studies investigating links between stress and the gut microbiota. Microbiota which show an association (not necessarily causality) with stress-resilience and stress-sensitivity are indicated in the columns labeled SR and SS, respectively. This depends on the study but may mean, e.g., a probiotic supplement which increased stress-resilience, or an increase in relative abundance of the microbiota in stress-resilient individuals. Abbreviations: EPM, Elevated Plus Maze; FST, Forced Swim Test; TST, Tail Suspension Test; HbA1c, Hemoglobin A1c; IL, Interleukin; IFN-γ, Interferon gamma; LDB, Light-Dark Box; OFT, Open Field Test; NOR, Novel Object Recognition; MSS, Maternal Separation Stress.
Study Design (Stress, Subjects, Intervention) ResultsSRSSReference
  • FST
  • BALB/c mice (M, adult)
  • Probiotic Lactobacillus rhamnosus JB-1, 28 days prior to FST/Vagotomy
  • Increase in anxiety-like (EPM) and depressive like behaviour (FST). Both ameliorated by probiotic
  • Stress-induced increase in corticosterone ameliorated by probiotic. Stress-induced hyperthermia not affected by probiotic.
  • Vagotomy prevented the anxiolytic effects of the probiotic.
  • Changes in gut microbiota not reported
Probiotic: L. rhamnosus JB-1Not applicable[36]
  • Chronic mild stress, 7 w
  • C57BL/6J Mice (M, 7 w)
  • Probiotic: Lactobacillus reuteri ATCC 23272, 2 w during stress and 2 w post-stress
  • Increase in depression-like behaviour (FST): prevented by probiotic.
  • Increase in serum kynurenine following stress. Prevented by probiotic.
  • Inhibition of the enzyme IDO1 by Lactobacillus-produced reactive oxygen species (H202) in vivo
  • Decreased (fecal) class Bacillus, specifically genera Lactobacillus and Turicibacter
Probiotic: L. reuteri ATCC 23272Decrease in (fecal) Lactobacillus[15]
  • Grid floor stress, 15 d
  • BALB/c mice (F, 5 w)
  • No intervention
  • Increase in anxiety-like (triple test) and depressive-like behaviour (TST)
  • Lower blood glucose but highter HbA1c were found. Cytokines were reduced in the control mice but not the stressed mice. Correlations were found between IL-6, IFN-γ, and behavior in the LDB, EPM, and OFT.
  • Increase in (cecal) Odoribacter, Alistipes and an unclassified genus from the Coriobacteriaceae family. Lachnospiraceae correlated to risk assessment behaviors. Bacterioides correlated with immobility in TST; Ruminococccaceae correlated to entries to closed arms in triple test (anxiety/activity)
Not applicableBacterioides; Ruminococccaceae[12]
  • MSS, 15 d
  • Sprague Dawley rats, sex not stated, 4 d neonatal
  • Probiotic L. rhamnosus strain R0011 (95%) and L. helveticus strain R0052 (5%), 15 d during stress
  • Behaviour not measured
  • Increase in serum cortisol and gut permeability. Prevented by probiotic
  • Decrease in genus Lactobacillus. Increase in bacterial adherence and penetration into mucosal cells.
  • Increases in cortisol, gut permeability, and bacterial adherence/penetration was prevented by probiotic supplementation.
Probiotic: L. rhamnosus strain R0011 (95%) and Lactobacillus helveticus strain R0052 (5%)Not applicable[59]
  • MSS, 15 d
  • Sprague Dawley rats (M, 90 d)
  • Probiotic Bifidobacterium infantis 35624, 45 days
  • Increase in depressive-like behviour (FST), ameliorated by probiotic supplementation
  • No difference in plasma corticosterone, L-kynurenine, tryptophan or kynurenic acid. An increase in IL-6 following stimulation with immune stimulant concanavalin A was prevented by the probiotic
  • Gut microbiota not measured
Probiotic: B. infantis 35624Not applicable[40].
  • MSS, 10 d
  • Sprague Dawley rats (M, 7–8 w)
  • No intervention
  • Increased stress-induced faecal boli number in the OFT, but no changes in behavior
  • Increased plasma corticosterone and increased systemic immune response in response to in vitro LPS challenge. Decreased pain threshold
  • Change in microbiota structure (taxa not specified)
Not applicableNot applicable[16]
  • MSS, 1 w
  • Infant rhesus monkeys (M+F, 6–9 m)
  • No intervention
  • Decrease in total abundance of fecal bacteria and Lactobacillus by day 3, but back to normal after a week
Not applicableNot applicable[67]
  • Prenatal stress
  • Human infants
  • No intervention
  • Gastrointestinal symptoms more common in babies from mothers who reported higher stress. Cortisol and stress related questionnaires did not correlate in the mothers.
  • Increased fecal Escherichia-enterobacteria and lower lactic acid bacteria and Actinobacteria
Not applicableNot applicable[69]
  • Prenatal (dam exposed to CMS)
  • Offspring of pregnant C57BL mice exposed to CMS stress
  • No intervention
  • Behavior not measured
  • Increased fecal Rikenellaceae and Odoribacter, Mucispirillum and a decrease in Bacteroides
Not applicable Not applicable [66]
  • Prenatal (dam exposed to restraint stress)
  • Offspring of pregnant C57/B16 mice
  • No intervention
  • Offspring showed increase in anxiey-like behvior (EPM, NOR) in adulthood
  • Increased plasma IL-1β in placenta and fetal brains but did not persist till adulthood. Decreased BDNF found in maternal placenta and in brains of adult offspring.
  • Microbial community composition clustered differently in the stress group from control in both pregnant dams and their offspring
Not applicable Not applicable [65]
  • Restraint Stress (16 h/d × 7 d)
  • Swiss Webster & CD-1 mice (M, 6–8 w)
  • Fecal transplant from stressed mice to germ free mice
  • Behaviour not measured
  • Increased inflammatory response to colonic pathogen in germ free mice with fecal transplant from stressor exposed mice.
  • Increased (fecal) Firmicutes and decreased Actinobacteria and Bifidobacterium.
Not applicable Not applicable [63]
  • Restraint Stress (6 h/d × 3 w)
  • Sprague Dawley rats (M, 220–240 g)
  • Probiotic: L. helveticus ns8, 26 d
  • Increased depressive-like behavior (SPT) and anxiety-like behavior (EPM, OF).
  • Body weight was reduced. Increase in plasma corticosterone and pro-inflammatory cytokines TNF-α and IFN-γ and decrease in plasma IL-10. Decreased BDNF in the hippocampus, prevented by probiotic
  • Stress-induced changes in behaviour, corticosterone, IL-10, BDNF were prevented by the probiotic supplementation
  • Gut microbiota not measured
Probiotic: L. helveticus ns8Not applicable [41]
  • Restraint stress, 12 h/d × 7 d
  • CD1 mice (M, 8 w)
  • No intervention
  • Behaviour not measured
  • Increased TNF-α gene expression in colonic tissue
  • Total bacteria and Gram negative bacteria increased in small intestine, cecum, and large intestine. Decrease in bacterial diversity and richness. Reduced family Porphyromonadaceae, specifically genus Tannerella. Increased colonization by introduced pathogen Citrobacter rodentium
Not applicable Not applicable [60]
  • Restraint Stress, 15 h/d × 7 d
  • CD-1 mice (M, 6–8 w)
  • No intervention
  • In the (colonic) mucosca-associated bacteria, a decrease in the families S24-7 and Lactobacillaceae and genera Lactobacillus spp., were found, and in an increase in the family Ruminococcaceae, and genera Oscillospira. In the luminal bacteria, a decrease in the family S24-7, as well as genera Adlercreutzia, and an unclassified genus in S24-7 were found
Not applicable Not applicable [13]
  • Restraint stress/FST alternated, 19 days
  • CF-1 mice (M+F, 6 w)
  • Distance travelled in EPM and LDB increased, increased rearings in OF. Blood collected after behavioral tests
  • Males had higher corticosterone levels following acute stress (behavioral testing)
  • Increase in family Lachnospiraceae. Decrease in genus Sarcina only in females. Ruminococcus gnavus increased in females but decreased in males
Not applicable Not applicable [58]
  • Social stress: cage in cage aggressor
  • CD1 and C57BL/6 mice (M, 6–8 w)
  • Behaviour not measured
  • No difference in colonic cytokines
  • Decrease in relative abundance of families Porphyromonadaceae and Lactobacilliaceae, and genera Lactobacillus, Parabacteroides, and an unclassified genus from phylum Firmicutes and unclassified genus from class Bacilli. The absolute abundance of lactobacilli was also reduced, specifically L. reuteri, but only in the outbred CD-1, not the inbred C57BL/6 mice
Not applicable Not applicable [56]
  • Social stress: chronic social defeat
  • C57BL/6 mice (M, 8 w)
  • Decrease in social interaction
  • Increase in (fecal) genus Bifidobacterium in the stress resilient group. Not detected in the control group or stress-sensitive group
BifidobacteriumNot applicable [62]
  • Social stress: resident intruder, 6 h/d × 10 d
  • C57BL/6J male, juvenile (5–6 wk)
  • Behaviour not measured
  • Differed across time points. Key changes were a decrease in phylums Bacteroidetes, Firmicutes, Verrucomicrobia; and genera Oscilospira and Anaeroplasma, with a trend in decrease in Lactobacillus. An increase and decrease in Akkermansia were found at different time points. A trend of increase in phylum Proteobacteria was found
Not applicable Not applicable [57]
  • Social stressor (6 d × 2 h/d)
  • CD1 Mice (M, 8 w)
  • Antibiotics (ampicillin (1 mg/mL), vancomycin (0.5 mg/mL), neomycin sulfate (1 mg/mL), and metronidazole (1 mg/mL))
  • Behaviour not measured
  • Increase in proinflammatory markers, particularly IL-6, prevented in antibiotic group
  • Immediately after induced stress, the (cecal) microbiome of mice had consistently altered within the group, and clustered separately from the control group, but after 15 h, the separation was no longer as clear, with variation within the stress group.Within genera, decrease in Bacteroides,increase in Clostridium, trend of decrease in Lactobacillus
  • Stress-induced increases in plasma IL-6 was inversley correlated with relative abundances of genera Coprococcus, Pseudobutyrivibrio and positively correlated with Dorea
Not applicable Not applicable [11]
  • Water Avoidance Stress (1 h/d × 7 d)
  • C57BL/6N mice (F, 6 w)
  • Antibiotics during stress (Bacitracin A, Neomycin, Amphotericin B)
  • Pain related behavior in response to intracolonic capsaicum increased. Slightly mitigated with antibiotics.
  • Increased fecal pellet output, plasma corticosterone, and adrenal gland weight. Increased luminal s-IgA levels. In the colon tissue, cannabanoid receptors increased marginally, and tryptophan hydroxylase (TPH1) expression increased by 40%
  • Antibiotics and stress enhanced bacterial adherence to luminal wall. Fecal Clostridium coccoideds cluster XIVa was increased, and Verrucobactera, Lactobacillus and Enterococcus spp. decreased
Not applicable Not applicable [190]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bear, T.; Dalziel, J.; Coad, J.; Roy, N.; Butts, C.; Gopal, P. The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress. Microorganisms 2021, 9, 723. https://doi.org/10.3390/microorganisms9040723

AMA Style

Bear T, Dalziel J, Coad J, Roy N, Butts C, Gopal P. The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress. Microorganisms. 2021; 9(4):723. https://doi.org/10.3390/microorganisms9040723

Chicago/Turabian Style

Bear, Tracey, Julie Dalziel, Jane Coad, Nicole Roy, Christine Butts, and Pramod Gopal. 2021. "The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress" Microorganisms 9, no. 4: 723. https://doi.org/10.3390/microorganisms9040723

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop