Skip to main content

REVIEW article

Front. Gastroenterol., 12 October 2022
Sec. Gastrointestinal Infection
This article is part of the Research Topic Gut Microbiota in Health and Disease View all 18 articles

Gut microbiota in anxiety and depression: Pathogenesis and therapeutics

  • 1CEMAD Digestive Disease Center, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
  • 2Departement of Neuroscience, Università Cattolica del Sacro Cuore - Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy

Depression and anxiety disorders represent a burdensome clinical issue. Considering the unsatisfactory clinical response of some patients to antidepressant therapy, new personalized approaches are being studied. In recent years, pre-clinical and clinical studies have investigated the role of intestinal microbiota demonstrating the importance of the gut-brain axis in these diseases. Indeed, gut microbes are able to interact with the brain interfering with behavior through some mechanisms such as amino acid metabolism, short–chain fatty acids, vagus nerve, endocrine signaling and immune responses. Experiments of gut microbiota transfer from subjects with major depression to animal models corroborated the causative role of intestinal microbes in mood disorders and anxiety. Furthermore, the incidence of dysbiosis in patients with anxiety and depression suggests a potential role for gut microbiota modulators in the treatment of these disorders. In particular, several probiotics and synbiotics have been shown to be effective in improving clinical symptoms, promising results have emerged also from fecal microbiota transplantation, but the evidence is still limited. These promising results switch on the use of gut microbiota modulators as an adjunctive tool to anti-depressant therapy. Developing pharmaceutical or nutraceutical strategies to modify the composition of gut microbiota may offer novel and personalized therapeutic tools against anxiety and depression.

Introduction

Anxiety and mood disorders represent an alarming clinical issue, as well as cause of disability and mortality worldwide (1). Unfortunately, the mechanisms triggering these diseases have not yet been fully understood. Several factors such as oxidative stress (2), impaired signaling by neurotrophic factor (3) or chronic inflammation (4) have been hypothesized to be involved in the development and susceptibility of mood disorders, which presumably are caused by an interplay between genetics and environmental factors (5, 6). To date, the lack of this knowledge has a negative effect on the efficacy of common therapies, so there is a need for personalized treatment for these patients (7). In this regard, considering the pathophysiological role of the intestinal microbiome, the development of innovative therapies for these disorders can be hypothesized. Gut microbes are able to produce most neurotransmitters, influencing neurochemistry and behavior via the so-called “gut-brain axis” (8). Moreover, the high prevalence of stress-related psychiatric symptoms in patients with gastrointestinal disorders supports the link between gut microbiota changes and psychiatric disorders (9). The functional crosstalk among enteric microorganisms, gut and brain may occur through multiple mechanisms, including metabolic and neuroimmunological pathways. Finally, developing pharmaceutical or nutraceutical strategies to modify the composition of gut microbiota may offer novel and personalized therapeutic tools against anxiety and depression, which we will discuss below.

Gut microbiota regulates anxiety-like and depression-like behavior: Evidences from animal studies

Despite the limitations represented mainly by the difference in the composition of the human and murine microbiota, and the difficulty of translate the findings from experimental models to patients where no complete ablation of the microbiota can be achieved, studies on rodents indicate that gut microbiota influences brain function and may impact on the behavior (10). Experimental approaches used to study the microbiota-gut-brain axis included the treatment with probiotics/antibiotics, the induction of gut inflammation by injection of enteric bacterial pathogens, the use of germ-free (GF)/gnotobiotic animals and the human diseases-related fecal microbiota transplantation (FMT) (11). The main advantages of studies performed on murine experimental models are the efficacy of behavioral tests to reveal changes similar to what observed in patients affected by anxiety or depression (12), and the possibility to analyze the effects of a single bacterial phylum or species on behavior. Animal studies suggested that changes in the microbiota induced brain modifications at both molecular and behavioral level. Mice treated with a cocktail of non-absorbable antibiotics showed changes of intestinal microbiota profile (i.e., a reduction of Shigella, Bacteroides and Klebsiella genera and an increase of Actinobacter and Lactobacillus populations) in parallel with greater exploratory activity (13). This anxiolytic effect was accompanied by an increase of brain-derived neurotrophic factor (BDNF) levels in the hippocampus and amygdala. More importantly, the authors did not observe the same responses in animals intraperitoneally injected with the antibiotics or in germ-free mice to which the drugs were administered by gavage. Moreover, gut microbiota seems to be involved in the diet-induced brain modification. High fat diet (HFD) is a well-established experimental model able to induce changes of both insulin and leptin signaling into the brain, anxiety and memory deficits (1416). Soto and colleagues demonstrated that in HFD-fed mice, oral treatment with antibiotics modified the levels of neuromodulators such as tryptophan, γ-aminobutyric acid (GABA) and BDNF, ameliorated brain insulin signaling and counteracted anxiety and depression (17). In addition, the authors documented that these effects were transferable to germ-free mice by FMT.

Indeed, a large part of these studies based on the transferability of behavioral traits from donor mice to germ-free animals via the intestinal microbiota. For instance, BALB/c mice have anxiety-like behavior but, when they were colonized with the microbiota from Swiss mice, they acquired a more exploratory behavior. Accordingly, germ-free Swiss mice colonized with the intestinal bacteria from BALB/c mice exhibited a more anxious behavior (13). More recently, it has been showed that mice transplanted with fecal microbiota from Irritable bowel syndrome (IBS) patients exhibited intestinal barrier dysfunction, immunological activation, and anxiety-like behavior (18). More generally, intestinal microbiota appears to influence the stress response of rodents. Sudo and colleagues demonstrated that plasma levels of both ACTH and corticosterone were more prone to increase upon restraint stress in GF mice than in microbiota-competent animals (19). Moreover, the colonization by Bifidobacterium infantis of germ free mice was able to fully reverse these effects, revealing a causative role for the gut microbiota in modulating stress responses. Accordingly, the reduced expression of inflammatory interleukins and increased the amount of BDNF in the hippocampus was obtained by oral intake of Bifidobacterium, causing anxiolytic and antidepressant effects in mice (20). Bifidobacterium administration has been also shown to offer resilience to chronic social defeat stress in mice (21). In addition, three independent studies found altered concentrations of neurotransmitters and neurotrophic factors in the brain, and reduced anxiety in GF mice (2224). These neurochemical and behavioral findings are not actually in agreement, because enhanced hypothalamic–pituitary–adrenal (HPA) axis is usually related to increased anxiety-like behavior. Clarke and colleagues also reported elevated concentrations of tryptophan, the precursor of serotonin, in the plasma and a significant increase of serotonin metabolites in the hippocampus of male GF mice compared with control animals (24). Serotonin is an excitatory neurotransmitter produced also in the gut and able to counteract anxiety and depression at central level (25). Metabolomics studies revealed elevated serum tryptophan and less serum serotonin in GF mice compared to controls (26). However, whether changes in serotonin and neurotrophic factors (e.g., BDNF) are involved in the gut microbiota-dependent modification of anxiety-like behavior remains to be elucidated.

Rodent models have provided the mechanisms by which the gut microbiota may modulate depression-like behaviors. Maternal separation is a model of early life stress that induces anxiety and depression by altering HPA axis, immune system and aminoacid metabolism along with affecting microbiota composition (27, 28). More recently, De Palma and colleagues demonstrated that maternal separation of GF mice did not induce depressive or anxiety behavior despite it caused increase of circulating corticosterone (29). This study suggests that gut microbiota is not required for stress-induced changes in HPA axis activity but it is necessary for development of anxiety and depression-like behaviors. Therefore, intestinal microbes appeared to regulate stress responses in the brain of animal models and this evidence stimulated the possibility of using probiotic treatments to modulate brain function in physiological and pathological conditions (30). A plethora of probiotic agents have been tested in rodent models of anxiety and depression. Bifidobacterium and Lactobacillus are the main genera that have provided beneficial effects on neurological disorders (31). Bifidobacterium infantis has been shown to have antidepressant effect promoting antidepressant-like performance in the forced swim test, a widely used test to evaluate the efficacy of antidepressant drugs (32). Supplementation of Bifidobacterium infantis also counteracted the maternal separation-induced increase of both plasma tryptophan and pro-inflammatory cytokines, which have been demonstrated to play a role in the pathophysiology of depression (33). Many studies also clarified the mechanisms underlying the effects of probiotics on brain functions. Several studies focalized the attention on the ability of probiotics to modulate the inflammatory response of the organism. Lactobacillus rhamnosus has been proved to inhibit in vitro the Salmonella enterica-related synthesis of pro-inflammatory interleukin-8 and tumor necrosis factor alpha (34). This bacterial strain has been also found to induce region-dependent changes in GABA receptor expression in the brain. More importantly, Lactobacillus rhamnosus administration reduced in mice the stress-dependent increase of corticosterone levels and counteracted the related anxiety- and depression-like behavior. Moreover, the beneficial effects of this probiotics were abolished in vagotomized animals (35). More recently, Janik and colleagues documented by magnetic resonance spectroscopy that chronic treatment with Lactobacillus rhamnosus induced significant changes in the concentration of neurotransmitters such as glutamate, N-acetyl aspartate, and GABA into the brain (36). It suggests that probiotics could affect brain activity by regulating neurochemical pathways underlying synaptic transmission and plasticity. In addition, administration of Bifidobacterium Infantis enhanced the expression of BDNF and N-methyl-D-aspartate receptor subunit 2a, which are molecules involved in learning and memory (19). Collectively, these studies prompt the idea that probiotics can modulate microbiome-gut-brain axis and influence brain function. Despite significant difference occurs between the human and mouse microbiomes, the evidence from experimental models suggest that changes of gut microbiota composition may affect molecular pathways involved in the onset and progression of anxiety- and depression-related behaviors Figure 1.

FIGURE 1
www.frontiersin.org

Figure 1 Role of the gut microbiota in the development of mood disorders. Some pathophysiological mechanisms underlying the development of anxiety and depression have been proposed, in particular the balance of immunological, neurotransmitter and hormonal mechanisms is the proposed orchestror.

Dysbiosis in depression and anxiety disorders: Evidences from human studies

In recent years, some studies were conducted to investigate how the intestinal microbiota play a role in patients with anxiety and mood disorders. In particular, several data from human studies shown that fecal microbiota often has some variability between patients and healthy controls, considering microbial diversity and taxonomic compositions. Furthermore, was reported that specific bacteria were associated with metabolic or inflammatory profiles and clinical characteristics (37).

Microbial diversity is a fundamental aspect in the study of fecal microbiota that is considered a marker of health, but the reproducibility of data is strongly limited by the interference of many environmental factors (38). To date, few studies reported data about microbial diversity in humans, most of these failed to demonstrate an association between lower microbial diversity and depressive disorders (3941), while only one study reported higher α-diversity (i.e., the number of species detectable in a microbial ecosystem) of gut microbiota in major depressive disorder (MDD) patients compared to healthy subjects (42).

Taxonomic differences are described in several studies involving MDD patients, interesting differences have been reported for the main Phyla represented. Unfortunately, the findings from human studies are often conflicting, probably due to several confounding factors. For instance, several changes in microbial composition were reported in the Phylum of Firmicutes, but as previous discussed, findings were often contradictory. The relative abundance of this phylum appeared to be more represented in MDD according to some studies (41, 43), however this finding it was not confirmed by farther report (42). Moreover, more differences were reported at family level considering that Lachnospiraceae were found increased (4042) or decreased (39) between available studies, likewise Ruminococcaceae had a fluctuating representation, higher (40, 41) or lower (42) among reports. Finally, the genus level showed the most remarkable changes that were described for Faecalibacterium (40, 42) and Ruminococcus (42), these genera were decreased in subjects with depressive disorders. Similarly, changes in microbial composition were described for other phyla such as Bacteroidetes (39, 4143) and Actinobacteria (41, 42), although with sometimes conflicting results among the various studies. Most significative differences were observed at the genus level as a reduced representation of Bifidobacterium (44). Furthermore, correlation between clinical characteristics of patients and microbial signature was reported. Specifically, Fusobacteria and Proteabacteria appeared to be increased or reduced, respectively in active-MDD or recovering-MDD (42).

Above, we have briefly reported the complexity and the divergences between the evidences probably due to methodological differences and environmental variability among different studies. A recent systematic review showed that about 50 bacterial taxa exhibit differences between patients with MDD and controls (45). However, the authors failed to demonstrate the prevalence of a specific bacterial taxa in the development of depression.

In the near future, meta-proteomics studies should add further elements in the understanding the association between microbiota and the development of depression. A pioneering study by Chen and colleagues investigated the metabolomic profile in patients with MDD and it reported several significant differences in the pathway of bacterial proteins that were mainly involved in glucose metabolism and amino acid metabolism (46).

Interesting alterations of the fecal microbiota have also been identified in patients suffering from anxiety disorder. In particular were found a reduction in microbial richness and diversity in patients with generalized anxiety disorder (GAD), associated with reduced short-chain fatty acid producing bacteria such as Eubacterium rectale and Fecalibacterium, and an increase in Escherichia, Shigella, Fusobacterium and Ruminococcus (47). More importantly, these changes were not reversed in remissive GAD. Conversely, another study failed to demonstrate any correlation between intestinal dysbiosis and anxiety in female subjects (48), confirming the variability between human studies.

Potential for therapy

Gut microbiota represents a new frontier in psychiatry. For this reason, antibiotics, probiotics, prebiotics and FMT were investigated for the treatment of anxiety (49) and depression (50). Psychobiotics define these therapeutic tools (51), in particular the main evidences on the modulation of the gut microbiota in depression and anxiety disorders were reported in the next paragraphs.

Antibiotics

Antibiotics are deep modulators of gut microbiota, and consequently they appear to change, in a positive or negative way, the nature of several gastrointestinal or extra-intestinal disorders (52). Therefore, in consideration of their known effect on behavior, they have been proposed as a therapeutic tool also in psychiatry (53). Potential and beneficial effects were described in individual with depression or anxiety related disorders.

For instance, Minocycline has been identified as a potential novel treatment for depression taking into consideration its potent anti-inflammatory and neuroprotective effects (54). In recent years, several clinical trials investigated the potential role of this drug in the scenario of depression; meta-analyses that included three RCTs reported preliminary evidence for a significant antidepressant effect of minocycline. The antidepressant effect size was found to be large (SMD − 0.78; 95%CI; 0.4−1.33; p=0.005) with moderate heterogeneity of the pooled sample. However, the small number of published RCTs and small sample sizes were significant limitations to draw definitive conclusions (55). Furthermore, the broad-spectrum antibiotic Cycloserine was investigated for the treatment of anxiety disorders. A meta-analysis that included 21 studies that involved 1047 individuals with several psychiatrics disorders (phobia, social anxiety disorder, panic disorder, obsessive-compulsive disorder and post-traumatic stress disorder) showed that Cycloserine was associated with a small augmentation effect on exposure-based therapy and suggested that this effect was not modulated by the concurrent use of antidepressants (56). However, antibiotics have also been associated with a negative effect on mood disorders. In particular, recurrent exposure to antibiotics such as penicillins (OR 1.23; 95% CI, 1.18-1.29) or quinolones (OR 1.25; 95% CI, 1.15-1.35) appeared to be associated with increased risk for depression and anxiety (57).

Probiotics

Probiotics are defined as live microorganisms that, upon administration in adequate amounts, confer a health benefit on the host (58). To date, some studies report results on the use of probiotics in the treatment of mood disorders, albeit with some limitations as the heterogeneity of enrolled patients and the variety of the administered mixtures (59). Miyaoka and colleagues investigated the role of Clostridium butyricum (CBM588) as adjunctive therapy in patients with treatment-resistant MDD. In this study was reported a significant improvement in depression scale after 8 weeks of treatment, suggesting a potential therapeutic role for this probiotic strain in combination with antidepressant drugs (60). Another clinical trial reported that a probiotic mixture (L. helveticus R00052 and B. longum R0175) was able to ameliorate the beck depression inventory (BDI) in individuals with mild to moderate MDD compared to placebo (61). Farther, the administration of a mixture of L. acidophilus, L. casei and B. bifidum resulted in a significant reduction of BDI score (62). Sometimes MDD patients experienced gastrointestinal disorders and in particular IBS, in this context Majeed and colleagues reported significant improvement of depression and IBS symptoms in patients treated with Bacillus Coagulans MTCC 5856 (63). Promising results were also reported about stress and anxiety. Indeed, Lactobacillus plantarum DR7 appeared to be beneficial in reducing symptoms and psychological scores (64).

However, not all studies documented positive results, maybe due to probiotic strain, concurrent medications or other unexplored factors. For instance, Romijn and colleagues demonstrated that a probiotic mixture (L. helveticus R0052 and B. longum R0175) failed to improve depressive symptoms in individuals with low mood not currently taking psychotropic medications (65). Finally, another study clearly showed that the probiotic B. Longum NCCC3001 reduced depression but not anxiety scores and increased quality of life in patients with IBS (66). Furthermore, the effects were associated with changes in brain activation patterns demonstrating that this probiotic reduces limbic reactivity (66).

Prebiotics

Prebiotics are selectively fermented compounds promoting changes in both composition and activity of intestinal microbiota that offer benefits to the host (67). Few studies investigated the role of prebiotics in mood disorders. Smith and colleagues failed to demonstrate a significant effect of a prebiotic mixture (oligofructose enriched inulin) on mood scores in a cohort of healthy adults. However, participants reported greater well-being after consumption of inulin (68). Similarly, despite beta-glucan derived from Saccharomyces cerevisiae improved mood in stressed subjects, no significant differences in depression scores were observed compared to placebo (69). Moreover, another clinical trial failed to demonstrate that prebiotic supplementation improved depressive symptoms. Indeed, administration of galacto-oligosaccharides for eight weeks did not significantly modify BDI score in MDD patients compared to placebo and its effect was lower than that of probiotic mixture (61). On the other hand, prebiotics supplementation appeared to be more efficacious on psychiatric symptoms in IBS patients. Short-chain fructo-oligosaccharides (scFOS) showed beneficial effects in a population with gastrointestinal symptoms. Specifically, scFOS supplementation for four weeks resulted in a significantly improvement of depression and anxiety scores, furthermore this effect was associated to changes in microbiota composition including increase of Bifidobacteria in feces (70). However, another prebiotic galacto-oligosaccharide mixture (B-GOS) not improved anxiety and depression scale in individuals with functional bowel disorders, albeit some beneficial effects were reported for gastrointestinal symptoms (71).

These conflicting data confirm the need for further studies to better establish the patient cohorts and compounds more efficacious for this type of intervention.

Synbiotics

Synbiotics are defined as a synergic mixture of probiotics and prebiotics that promote beneficial effects on health, in particular prebiotics are involving in favoring the colonization of the gut by probiotics (72). A small number of clinical trials that investigated the role of synbiotics in mood disorders have been published. A first trial demonstrated that a symbiotic mixture (Lactobacillus casaei, Lactobacillus acidofilus, Lactobacillus bulgarigus, Lactobacillus rhamnosus, Bifidobacterium breve, Bifidobacterium longum, Streptococus thermophiles, and fructo-oligosaccharide) was able to decrease HAM-D score and to improve depressive symptoms in patients with moderate MDD (73).

Afterwards, another clinical trial demonstrated the greater efficacy of symbiotic formulations compared to probiotics mixture alone in the treatment of mood disorder. In the clinical trial designed by Haghighat and colleagues (74), patients were randomly assigned to receive synbiotics (prebiotics: fructo-oligosaccharides, galacto-oligosaccharides, and inulin; probiotics: Lactobacillus acidophilus T16, Bifidobacterium bifidum BIA-6, Bifidobacterium lactis BIA-7, and Bifidobacterium longum BIA-8) or probiotics (the same mixture of synbiotics without prebiotics) or placebo for twelve weeks Table 1.

TABLE 1
www.frontiersin.org

Table 1 Results from clinical trials on modulation of gut microbiota in anxiety and depression.

Fecal microbiota transplantation

Fecal microbiota transplantation is the infusion of a fecal suspension derived from a healthy donor into the intestine of a recipient to restore the imbalanced gut microbiota (75). Some fascinating studies on animal models have supported the idea that the transfer of “good microbes” can represent a new tool in the treatment of depression and anxiety. For example, it has been demonstrated that the transfer of healthy microbiota in an animal model of alcohol-induced anxiety and depression reduced the clinical manifestation in the animal (76). On the other hand, it was reported the “transfer of depression” trough microbiota. Indeed, germ free mice who underwent to FMT derived from MDD patients resulted in depression-like behaviors compared with colonization by microbiota derived from healthy control individuals (40). Furthermore, another study confirmed that FMT from depressed patients to microbiota-deficient rats could induce behavioral and physiological features characteristic of depression in the recipient animals, including anhedonia and anxiety-like behaviors (77). Unfortunately, the evidence for the use of FMT in humans is still limited (78, 79). A small study on 17 patients with functional gastrointestinal disorders treated with FMT reported an improvement of depression and anxiety symptoms independently of gastrointestinal symptom changes (80). A further small clinical study demonstrates that FMT in patients with IBS-D is able to reduce levels of anxiety and depression, as well as gastroenterological symptoms, in particular was associated to the decreased abundance of Faecalibacterium, Eubacterium and Escherichia (81). Further studies are needed to validate the procedure and to identify microbiome more efficacious for FMT.

Final remarks

The microbiota-gut–brain axis is an integrative system that involves metabolic, immunological and neuroendocrine signals, and alterations of these pathways play relevant roles in human neurological diseases. Extensive research has demonstrated that diet, drugs and stress influence both composition and function of gut microbiota, which in turn can modulate neurophysiology and behavior. Therefore, gut microbiota represents a key mechanism underlying the impact of environmental stimuli on brain function and identifying the biological pathways involved in the microbiota-gut-brain axis may be relevant to understand the pathophysiology of human mood disorders. Further, developing therapeutic strategies to modify the composition of gut microbiota may offer novel and personalized therapeutic tools (82). Indeed, several studies have reported that treatments able to modify the intestinal microbiota exerted a significant effect on the symptoms of anxiety disorders and depression in humans. More specifically, treatment with probiotics and synbiotics showed the best results in terms of symptom improvement, suggesting a potential role as adjunctive therapy. Unfortunately, the results about prebiotics alone are not satisfactory in the setting of mood disorders. Results from FMT studies in humans are fascinating but still too weak. Finally, the evidences from antibiotic studies are conflicting (83), because while some drugs such as minocycline and cycloserine have shown to have beneficial effects, other drugs of wide clinical use, as penicillins or quinolones, may increase the risk for depression and anxiety. In this review we have analyzed how some pharmacological approaches can modify the gut microbiota and promote a favorable effect on anxiety and depression. On the other hand, in recent years, “non-pharmacological” treatments are also being considered to regulate microbiota composition. It is known that diet plays a fundamental role in modulating the microbiota (84), this is true both in health and in disease. In particular, several evidences are emerging on how diet can play a role in the treatment of behavioral disorders (85). For instance, it has been shown that a diet rich in fat can favor the development or persistence of anxiety and depression, an effect sometimes reversible with probiotics (86). Furthermore, experimental models have shown how a supplementation diet with psychoactive metabolites, such as tryptophan, can have a protective role on the development of these mood disorders through the reduction of stress-induced gut barrier damage and inflammatory responses in the gut (87). Still reporting on non-pharmacological approaches, in the last year very interesting results have emerged from studies evaluating the role of cognitive behavioral therapy (CBT) in modifying the microbiota. For instance, a small study demonstrate that mindfulness CBT promote changes in gut microbiota of subjects affected by anxiety, in particular the individuals who responded better by reducing anxiety modified the microbiota making it more similar to healthy subjects and interestingly they increased the metabolism of tryptophan (88). The interpretation of these results opens up new frontiers on the modulation of the gut-brain axis, in fact it appears possible to modulate it in both directions (gut-brain and brain-gut) to obtain modifications for therapeutics.

In conclusion, drugs and non-pharmacological approaches regulating the composition of intestinal microbiota represent promising beneficial strategies against anxiety and depression. The study of the crosstalk between microbiota and brain can improve knowledge about the development of mood disorders and help to identify new therapeutic tools for the personalized medicine.

Author contributions

SB, SF, and AG contributed to conception and design of the study. SB and SF wrote the first draft of the manuscript. All authors contributed to manuscript revision, read, and approved the submitted version

Funding

This paper received funding from Fondazione Roma.

Acknowledgments

The authors thank Fondazione Roma for the continuous non-conditioning support.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

MDD, Major depressive disorders; GAD, Generalized anxiety disorder; IBS, Irritable bowel syndrome; SCFA, short chain fatty acid; BDI, Beck Depression Inventory; IBS, irritable bowel syndrome; HAM-D, Hamilton rating scale for depression; HADS, Hospital Anxiety and Depression Scale; CBT, cognitive behavioral therapy.

References

1. Murray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C, et al. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990-2010: a systematic analysis for the global burden of disease study 2010. Lancet (2012) 380(9859):2197–223. doi: 10.1016/S0140-6736(12)61689-4

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Salim S. Oxidative stress and psychological disorders. Curr Neuropharmacol (2014) 12(2):140–7. doi: 10.2174/1570159X11666131120230309

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Castren E, Kojima M. Brain-derived neurotrophic factor in mood disorders and antidepressant treatments. Neurobiol Dis (2017) 97(Pt B):119–26. doi: 10.1016/j.nbd.2016.07.010

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Rosenblat JD, Cha DS, Mansur RB, McIntyre RS. Inflamed moods: a review of the interactions between inflammation and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry (2014) 53:23–34. doi: 10.1016/j.pnpbp.2014.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Lopresti AL, Hood SD, Drummond PD. A review of lifestyle factors that contribute to important pathways associated with major depression: diet, sleep and exercise. J Affect Disord (2013) 148(1):12–27. doi: 10.1016/j.jad.2013.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Sharma S, Powers A, Bradley B, Ressler KJ. Gene x environment determinants of stress- and anxiety-related disorders. Annu Rev Psychol (2016) 67:239–61. doi: 10.1146/annurev-psych-122414-033408

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Cuijpers P, Reynolds CF 3rd, Donker T, Li J, Andersson G, Beekman A. Personalized treatment of adult depression: medication, psychotherapy, or both? a systematic review. Depress Anxiety (2012) 29(10):855–64. doi: 10.1002/da.21985

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Dinan TG, Cryan JF. The microbiome-Gut-Brain axis in health and disease. Gastroenterol Clin North Am (2017) 46(1):77–89. doi: 10.1016/j.gtc.2016.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol (2009) 6(5):306–14. doi: 10.1038/nrgastro.2009.35

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci (2012) 13(10):701–12. doi: 10.1038/nrn3346

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Cryan JF, O'Mahony SM. The microbiome-gut-brain axis: from bowel to behavior. Neurogastroenterol Motil (2011) 23(3):187–92. doi: 10.1111/j.1365-2982.2010.01664.x

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Ellenbroek BA, Cools AR. Animal models with construct validity for schizophrenia. Behav Pharmacol (1990) 1(6):469–90.

PubMed Abstract | Google Scholar

13. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. (2011) 141(2):599–609, e1-3. doi: 10.1053/j.gastro.2011.04.052

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Spinelli M, Fusco S, Mainardi M, Scala F, Natale F, Lapenta R, et al. Brain insulin resistance impairs hippocampal synaptic plasticity and memory by increasing GluA1 palmitoylation through FoxO3a. Nat Commun (2017) 8(1):2009. doi: 10.1038/s41467-017-02221-9

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Sharma S, Fulton S. Diet-induced obesity promotes depressive-like behaviour that is associated with neural adaptations in brain reward circuitry. Int J Obes (2013) 37(3):382–9. doi: 10.1038/ijo.2012.48

CrossRef Full Text | Google Scholar

16. Yamada N, Katsuura G, Ochi Y, Ebihara K, Kusakabe T, Hosoda K, et al. Impaired CNS leptin action is implicated in depression associated with obesity. Endocrinology (2011) 152(7):2634–43. doi: 10.1210/en.2011-0004

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Soto M, Herzog C, Pacheco JA, Fujisaka S, Bullock K, Clish CB, et al. Gut microbiota modulate neurobehavior through changes in brain insulin sensitivity and metabolism. Mol Psychiatry (2018) 23(12):2287–301. doi: 10.1038/s41380-018-0086-5

PubMed Abstract | CrossRef Full Text | Google Scholar

18. De Palma G, Lynch MD, Lu J, Dang VT, Deng Y, Jury J, et al. Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice. Sci Transl Med (2017) 9(379):eaaf6397. doi: 10.1126/scitranslmed.aaf6397

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol (2004) 558(Pt1):263–75. doi: 10.1113/jphysiol.2004.063388

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Guo Y, Xie JP, Deng K, Li X, Yuan Y, Xuan Q, et al. Prophylactic effects of Bifidobacterium adolescentis on anxiety and depression-like phenotypes after chronic stress: A role of the gut microbiota-inflammation axis. Front Behav Neurosci (2019) 13:126. doi: 10.3389/fnbeh.2019.00126

PubMed Abstract | CrossRef Full Text | Google Scholar

21. 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) 7:45942. doi: 10.1038/srep45942

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil (2011) 23(3):255–64, e119. doi: 10.1111/j.1365-2982.2010.01620.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Diaz Heijtz R, Wang S, Anuar F, Qian Y, Bjorkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A (2011) 108(7):3047–52. doi: 10.1073/pnas.1010529108

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry (2013) 18(6):666–73. doi: 10.1038/mp.2012.77

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. (2015) 161(2):264–76. doi: 10.1016/j.cell.2015.02.047

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A (2009) 106(10):3698–703. doi: 10.1073/pnas.0812874106

PubMed Abstract | CrossRef Full Text | Google Scholar

27. O'Mahony SM, Marchesi JR, Scully P, Codling C, Ceolho AM, Quigley EM, et al. Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry (2009) 65(3):263–7. doi: 10.1016/j.biopsych.2008.06.026

PubMed Abstract | CrossRef Full Text | Google Scholar

28. O'Mahony SM, Hyland NP, Dinan TG, Cryan JF. Maternal separation as a model of brain-gut axis dysfunction. Psychopharmacology. (2011) 214(1):71–88. doi: 10.1007/s00213-010-2010-9

PubMed Abstract | CrossRef Full Text | Google Scholar

29. De Palma G, Blennerhassett P, Lu J, Deng Y, Park AJ, Green W, et al. Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nat Commun (2015) 6:7735. doi: 10.1038/ncomms8735

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Bravo JA, Julio-Pieper M, Forsythe P, Kunze W, Dinan TG, Bienenstock J, et al. Communication between gastrointestinal bacteria and the nervous system. Curr Opin Pharmacol (2012) 12(6):667–72. doi: 10.1016/j.coph.2012.09.010

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Kim N, Yun M, Oh YJ, Choi HJ. Mind-altering with the gut: Modulation of the gut-brain axis with probiotics. J Microbiol (2018) 56(3):172–82. doi: 10.1007/s12275-018-8032-4

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The probiotic bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J Psychiatr Res (2008) 43(2):164–74. doi: 10.1016/j.jpsychires.2008.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG. Effects of the probiotic bifidobacterium infantis in the maternal separation model of depression. Neuroscience. (2010) 170(4):1179–88. doi: 10.1016/j.neuroscience.2010.08.005

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Ma D, Forsythe P, Bienenstock J. Live Lactobacillus rhamnosus [corrected] is essential for the inhibitory effect on tumor necrosis factor alpha-induced interleukin-8 expression. Infect Immun (2004) 72(9):5308–14. doi: 10.1128/IAI.72.9.5308-5314.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A (2011) 108(38):16050–5. doi: 10.1073/pnas.1102999108

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Janik R, Thomason LAM, Stanisz AM, Forsythe P, Bienenstock J, Stanisz GJ. Magnetic resonance spectroscopy reveals oral Lactobacillus promotion of increases in brain GABA, n-acetyl aspartate and glutamate. Neuroimage. (2016) 125:988–95. doi: 10.1016/j.neuroimage.2015.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Huang TT, Lai JB, Du YL, Xu Y, Ruan LM, Hu SH. Current understanding of gut microbiota in mood disorders: An update of human studies. Front Genet (2019) 10:98. doi: 10.3389/fgene.2019.00098

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. (2012) 486(7402):207–14. doi: 10.1038/nature11234

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linlokken A, Wilson R, et al. Correlation between the human fecal microbiota and depression. Neurogastroenterol Motil (2014) 26(8):1155–62. doi: 10.1111/nmo.12378

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism. Mol Psychiatry (2016) 21(6):786–96. doi: 10.1038/mp.2016.44

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Chen JJ, Zheng P, Liu YY, Zhong XG, Wang HY, Guo YJ, et al. Sex differences in gut microbiota in patients with major depressive disorder. Neuropsychiatr Dis Treat (2018) 14:647–55. doi: 10.2147/NDT.S159322

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun (2015) 48:186–94. doi: 10.1016/j.bbi.2015.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Lin P, Ding B, Feng C, Yin S, Zhang T, Qi X, et al. Prevotella and Klebsiella proportions in fecal microbial communities are potential characteristic parameters for patients with major depressive disorder. J Affect Disord (2017) 207:300–4. doi: 10.1016/j.jad.2016.09.051

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Aizawa E, Tsuji H, Asahara T, Takahashi T, Teraishi T, Yoshida S, et al. Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J Affect Disord (2016) 202:254–7. doi: 10.1016/j.jad.2016.05.038

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Cheung SG, Goldenthal AR, Uhlemann AC, Mann JJ, Miller JM, Sublette ME. Systematic review of gut microbiota and major depression. Front Psychiatry (2019) 10:34. doi: 10.3389/fpsyt.2019.00034

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Chen Z, Li J, Gui S, Zhou C, Chen J, Yang C, et al. Comparative metaproteomics analysis shows altered fecal microbiota signatures in patients with major depressive disorder. Neuroreport (2018) 29(5):417–25. doi: 10.1097/WNR.0000000000000985

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Jiang HY, Zhang X, Yu ZH, Zhang Z, Deng M, Zhao JH, et al. Altered gut microbiota profile in patients with generalized anxiety disorder. J Psychiatr Res (2018) 104:130–6. doi: 10.1016/j.jpsychires.2018.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Kleiman SC, Bulik-Sullivan EC, Glenny EM, Zerwas SC, Huh EY, Tsilimigras MC, et al. The gut-brain axis in healthy females: Lack of significant association between microbial composition and diversity with psychiatric measures. PLoS One (2017) 12(1):e0170208. doi: 10.1371/journal.pone.0170208

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Yang B, Wei J, Ju P, Chen J. Effects of regulating intestinal microbiota on anxiety symptoms: A systematic review. Gen Psychiatr (2019) 32(2):e100056. doi: 10.1136/gpsych-2019-100056

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Peirce JM, Alvina K. The role of inflammation and the gut microbiome in depression and anxiety. J Neurosci Res (2019) 97(10):1223–41. doi: 10.1002/jnr.24476

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Vaghef-Mehrabany E, Maleki V, Behrooz M, Ranjbar F, Ebrahimi-Mameghani M. Can psychobiotics "mood" ify gut? an update systematic review of randomized controlled trials in healthy and clinical subjects, on anti-depressant effects of probiotics, prebiotics, and synbiotics. Clin Nutr (2019) 39(5):1395–10. doi: 10.1016/j.clnu.2019.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Ianiro G, Tilg H, Gasbarrini A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut (2016) 65(11):1906–15. doi: 10.1136/gutjnl-2016-312297

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Van Ameringen M, Turna J, Patterson B, Pipe A, Mao RQ, Anglin R, et al. The gut microbiome in psychiatry: A primer for clinicians. Depress Anxiety (2019) 36(11):1004–25. doi: 10.1002/da.22936

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Soczynska JK, Mansur RB, Brietzke E, Swardfager W, Kennedy SH, Woldeyohannes HO, et al. Novel therapeutic targets in depression: minocycline as a candidate treatment. Behav Brain Res (2012) 235(2):302–17. doi: 10.1016/j.bbr.2012.07.026

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Rosenblat JD, McIntyre RS. Efficacy and tolerability of minocycline for depression: A systematic review and meta-analysis of clinical trials. J Affect Disord (2018) 227:219–25. doi: 10.1016/j.jad.2017.10.042

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Mataix-Cols D, Fernandez de la Cruz L, Monzani B, Rosenfield D, Andersson E, Perez-Vigil A, et al. D-cycloserine augmentation of exposure-based cognitive behavior therapy for anxiety, obsessive-compulsive, and posttraumatic stress disorders: A systematic review and meta-analysis of individual participant data. JAMA Psychiatry (2017) 74(5):501–10. doi: 10.1001/jamapsychiatry.2016.3955

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Lurie I, Yang YX, Haynes K, Mamtani R, Boursi B. Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. J Clin Psychiatry (2015) 76(11):1522–8. doi: 10.4088/JCP.15m09961

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Reid G. Probiotics: definition, scope and mechanisms of action. Best Pract Res Clin Gastroenterol (2016) 30(1):17–25. doi: 10.1016/j.bpg.2015.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Liu RT, Walsh RFL, Sheehan AE. Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev (2019) 102:13–23. doi: 10.1016/j.neubiorev.2019.03.023

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Miyaoka T, Kanayama M, Wake R, Hashioka S, Hayashida M, Nagahama M, et al. Clostridium butyricum MIYAIRI 588 as adjunctive therapy for treatment-resistant major depressive disorder: A prospective open-label trial. Clin Neuropharmacol (2018) 41(5):151–5. doi: 10.1097/WNF.0000000000000299

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Kazemi A, Noorbala AA, Azam K, Eskandari MH, Djafarian K. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin Nutr (2019) 38(2):522–8. doi: 10.1016/j.clnu.2018.04.010

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, Jafari P, Akbari H, Taghizadeh M, et al. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition. (2016) 32(3):315–20. doi: 10.1016/j.nut.2015.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Majeed M, Nagabhushanam K, Arumugam S, Majeed S, Ali F. Bacillus coagulans MTCC 5856 for the management of major depression with irritable bowel syndrome: a randomised, double-blind, placebo controlled, multi-centre, pilot clinical study. Food Nutr Res (2018) 62. doi: 10.29219/fnr.v62.1218

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Chong HX, Yusoff NAA, Hor YY, Lew LC, Jaafar MH, Choi SB, et al. Lactobacillus plantarum DR7 alleviates stress and anxiety in adults: a randomised, double-blind, placebo-controlled study. Benef Microbes (2019) 10(4):355–73. doi: 10.3920/BM2018.0135

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Romijn AR, Rucklidge JJ, Kuijer RG, Frampton C. A double-blind, randomized, placebo-controlled trial of Lactobacillus helveticus and Bifidobacterium longum for the symptoms of depression. Aust N Z J Psychiatry (2017) 51(8):810–21. doi: 10.1177/0004867416686694

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Pinto-Sanchez MI, Hall GB, Ghajar K, Nardelli A, Bolino C, Lau JT, et al. Probiotic Bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: A pilot study in patients with irritable bowel syndrome. Gastroenterology (2017) 153(2):448–59 e8. doi: 10.1053/j.gastro.2017.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Gibson GR, Probert HM, Loo JV, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev (2004) 17(2):259–75. doi: 10.1079/NRR200479

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Smith AP, Sutherland D, Hewlett P. An investigation of the acute effects of oligofructose-enriched inulin on subjective wellbeing, mood and cognitive performance. Nutrients. (2015) 7(11):8887–96. doi: 10.3390/nu7115441

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Talbott SM, Talbott JA. Baker's yeast beta-glucan supplement reduces upper respiratory symptoms and improves mood state in stressed women. J Am Coll Nutr (2012) 31(4):295–300. doi: 10.1080/07315724.2012.10720441

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Azpiroz F, Dubray C, Bernalier-Donadille A, Cardot JM, Accarino A, Serra J, et al. Effects of scFOS on the composition of fecal microbiota and anxiety in patients with irritable bowel syndrome: a randomized, double blind, placebo controlled study. Neurogastroenterol Motil (2017) 29(2):e12911. doi: 10.1111/nmo.12911

CrossRef Full Text | Google Scholar

71. Vulevic J, Tzortzis G, Juric A, Gibson GR. Effect of a prebiotic galactooligosaccharide mixture (B-GOS(R)) on gastrointestinal symptoms in adults selected from a general population who suffer with bloating, abdominal pain, or flatulence. Neurogastroenterol Motil (2018) 30(11):e13440. doi: 10.1111/nmo.13440

PubMed Abstract | CrossRef Full Text | Google Scholar

72. de Vrese M, Schrezenmeir J. Probiotics, prebiotics, and synbiotics. Adv Biochem Eng Biotechnol (2008) 111:1–66. doi: 10.1007/10_2008_097

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Ghorbani Z., Nazari S., Etesam F., Nourimajd S., Ahmadpanah M., Jahromi S. R.. The effect of synbiotic as an adjuvant therapy to fluoxetine in moderate depression: A randomized multicenter trial. Arch Neurosci (2018) 2(5):e60507. doi: 10.5812/archneurosci.60507

CrossRef Full Text | Google Scholar

74. Haghighat N, Rajabi S, Mohammadshahi M. Effect of synbiotic and probiotic supplementation on serum brain-derived neurotrophic factor level, depression and anxiety symptoms in hemodialysis patients: a randomized, double-blinded, clinical trial. Nutr Neurosci (2019) 24(6):490–9. doi: 10.1080/1028415X.2019.1646975

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Bibbo S, Ianiro G, Gasbarrini A, Cammarota G. Fecal microbiota transplantation: past, present and future perspectives. Minerva Gastroenterol Dietol (2017) 63(4):420–30. doi: 10.23736/S1121-421X.17.02374-1

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Xu Z, Liu Z, Dong X, Hu T, Wang L, Li J, et al. Fecal microbiota transplantation from healthy donors reduced alcohol-induced anxiety and depression in an animal model of chronic alcohol exposure. Chin J Physiol (2018) 61(6):360–71. doi: 10.4077/CJP.2018.BAH633

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Kelly JR, Borre Y, O’Brien C, Patterson E, El Aidy S, Deane J, et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res (2016) 82:109–18. doi: 10.1016/j.jpsychires.2016.07.019

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Settanni CR, Ianiro G, Bibbò S, Cammarota G, Gasbarrini A. Gut microbiota alteration and modulation in psychiatric disorders: Current evidence on fecal microbiota transplantation. Prog Neuropsychopharmacol Biol Psychiatry (2021) 109:110258. doi: 10.1016/j.pnpbp.2021.110258

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Meyyappan AC, Forth E, Wallace C, Milev R. Effect of fecal microbiota transplant on symptoms of psychiatric disorders: a systematic review. BMC Psychiatry (2020) 20:299. doi: 10.1186/s12888-020-02654-5

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Kurokawa S, Kishimoto T, Mizuno S, Masaoka T, Naganuma M, Liang KC, et al. The effect of fecal microbiota transplantation on psychiatric symptoms among patients with irritable bowel syndrome, functional diarrhea and functional constipation: An open-label observational study. J Affect Disord (2018) 235:506–12. doi: 10.1016/j.jad.2018.04.038

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Lin H, Guo Q, Wen Z, Tan S, Chen J, Lin L, et al. The multiple effects of fecal microbiota transplantation on diarrhea-predominant irritable bowel syndrome (IBS-d) patients with anxiety and depression behaviors. Microb Cell Fact (2021) 20:233. doi: 10.1186/s12934-021-01720-1

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Ianiro G, Bibbò S, Gasbarrini A, Cammarota G. Therapeutic modulation of gut microbiota: current clinical applications and future perspectives. Curr Drug Targets (2014) 15:762–70. doi: 10.2174/1389450115666140606111402

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Dinan K, Dinan T. Antibiotics and mental health: The good, the bad and the ugly. J Intern Med (2022). doi: 10.1111/joim.13543

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Bibbò S, Ianiro G, Giorgio V, Scaldaferri F, Masucci L, Gasbarrini A, et al. The role of diet on gut microbiota composition. Eur Rev Med Pharmacol Sci (2016) 20:4742–9.

PubMed Abstract | Google Scholar

85. Luna RA, Foster JA. Gut brain axis: diet microbiota interactions and implications for modulation of anxiety and depression. Curr Opin Biotechnol (2015) 32:35–41. doi: 10.1016/j.copbio.2014.10.007

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Lof J, Smits K, Melotte V, Kuil LE. The health effect of probiotics on high-fat diet-induced cognitive impairment, depression and anxiety: A cross-species systematic review. Neurosci Biobehav Rev (2022) 136:104634. doi: 10.1016/j.neubiorev.2022.104634

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Wang D, Wu J, Zhu P, Xie H, Lu L, Bai W, et al. Tryptophan-rich diet ameliorates chronic unpredictable mild stress induced depression- and anxiety-like behavior in mice: The potential involvement of gut-brain axis. Food Res Int (2022) 157:111289. doi: 10.1016/j.foodres.2022.111289

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Wang Z, Liu S, Xu X, Xiao Y, Yang M, Zhao X, et al. Gut microbiota associated with effectiveness and responsiveness to mindfulness-based cognitive therapy in improving trait anxiety. Front Cell Infect Microbiol (2022) 12:719829. doi: 10.3389/fcimb.2022.719829

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: FMT, gut-brain axis, probiotic, prebiotics, antibiotic, synbiotic

Citation: Bibbò S, Fusco S, Ianiro G, Settanni CR, Ferrarese D, Grassi C, Cammarota G and Gasbarrini A (2022) Gut microbiota in anxiety and depression: Pathogenesis and therapeutics. Front. Gastroenterol. 1:1019578. doi: 10.3389/fgstr.2022.1019578

Received: 15 August 2022; Accepted: 20 September 2022;
Published: 12 October 2022.

Edited by:

Ana Faria, Universidade Nova de Lisboa, Portugal

Reviewed by:

Feng Ji, Zhejiang University, China
Padhmanand Sudhakar, KU Leuven, Belgium
Irena Mladenova, Trakia University, Bulgaria
Quanjiang Dong, Qingdao University Medical College, China
Hossein Dabiri, Shahid Beheshti University of Medical Sciences, Iran

Copyright © 2022 Bibbò, Fusco, Ianiro, Settanni, Ferrarese, Grassi, Cammarota and Gasbarrini. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Antonio Gasbarrini, antonio.gasbarrini@unicatt.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.