Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-20T04:05:04.787Z Has data issue: false hasContentIssue false

Role of the gut microbiome in three major psychiatric disorders

Published online by Cambridge University Press:  04 May 2022

Jenny Borkent*
Affiliation:
Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
Magdalini Ioannou
Affiliation:
Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
Jon D. Laman
Affiliation:
Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
Bartholomeus C. M. Haarman
Affiliation:
Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
Iris E. C. Sommer
Affiliation:
Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
*
Author for correspondence: Jenny Borkent, E-mail: j.borkent@umcg.nl
Rights & Permissions [Opens in a new window]

Abstract

Major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia-spectrum disorders (SSD) are heterogeneous psychiatric disorders, which place significant burden on patient's well-being and global health. Disruptions in the gut-microbiome may play a role in these psychiatric disorders. This review presents current data on composition of the human gastrointestinal microbiota, and its interaction mechanisms in the gut–brain axis in MDD, BD and SSD. Diversity metrics and microbial relative abundance differed across studies. More studies reported inconsistent findings (n = 7) or no differences (n = 8) than studies who reported lower α-diversity in these psychiatric disorders (n = 5). The most consistent findings across studies were higher relative abundances of the genera Streptococcus, Lactobacillus, and Eggerthella and lower relative abundance of the butyrate producing Faecalibacterium in patients with psychiatric disorders. All three increased genera were associated with higher symptom severity. Confounders, such as medication use and life style have not been accounted for. So far, the results of probiotics trials have been inconsistent. Most traditional and widely used probiotics (consisting of Bifidobacterium spp. and Lactobacillus spp.) are safe, however, they do not correct potential microbiota disbalances in these disorders. Findings on prebiotics and faecal microbiota transplantation (FMT) are too limited to draw definitive conclusions. Disease-specific pro/prebiotic treatment or even FMT could be auspicious interventions for prevention and therapy for psychiatric disorders and should be investigated in future trials.

Type
Invited Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2022. Published by Cambridge University Press

Introduction

The human gut microbiome (GM), comprised of trillions of microorganisms, bacteria, viruses, fungi and other life forms, has been implicated in numerous aspects of human health and disease (Lai, Gao, & Zhang, Reference Lai, Gao and Zhang2020). The GM is diverse, personalized, dynamic and can be influenced, especially early in life, by factors such as vaginal/caesarean birth, diet, sleep, contact with other humans and stress (Gacesa et al., Reference Gacesa, Kurilshikov, Vich Vila, Sinha, Klaassen, Bolte and Weersma2022; Szeligowski, Yun, Lennox, & Burnet, Reference Szeligowski, Yun, Lennox and Burnet2020). However, genetic factors, especially immunological background also determine a person's microbiome composition (Thaiss, Zmora, Levy, & Elinav, Reference Thaiss, Zmora, Levy and Elinav2016). The GM partakes in a bidirectional communication with the central nervous system (CNS), called the gut–brain axis (Cryan et al., Reference Cryan, O'riordan, Cowan, Sandhu, Bastiaanssen, Boehme and Dinan2019). Disruptions to the GM have been associated with several neuropsychiatric disorders, including major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia-spectrum disorders (e.g. schizophrenia, schizo-affective disorder and schizophreniform disorder, SSD) (Bastiaanssen et al., Reference Bastiaanssen, Cussotto, Claesson, Clarke, Dinan and Cryan2020; Genedi, Janmaat, Haarman, & Sommer, Reference Genedi, Janmaat, Haarman and Sommer2019; Nguyen, Hathaway, Kosciolek, Knight, & Jeste, Reference Nguyen, Hathaway, Kosciolek, Knight and Jeste2021; Szeligowski et al., Reference Szeligowski, Yun, Lennox and Burnet2020).

MDD, BD and SSD are heterogeneous psychiatric disorders, which place significant burden on a patient's well-being and global health (WHO, 2006). Worldwide, MDD, BD and SSD affect 264 million, 45 million and 20 million people, respectively (James et al., Reference James, Abate, Abate, Abay, Abbafati, Abbasi and Murray2018). Besides the overlap in psychiatric symptomatology, cognitive and biological functions, there is also a large genetic overlap between MDD, BD and SSD (Anttila et al., Reference Anttila, Bulik-Sullivan, Finucane, Walters, Bras, Duncan and Neale2018). Nowadays it is generally assumed that SSD, BD and MDD are disorders that are not entirely separated, but represent different stages of a continuum of clinical pictures (Haarman, Riemersma-Van der Lek, Burger, Drexhage, & Nolen, Reference Haarman, Riemersma-Van der Lek, Burger, Drexhage and Nolen2016).

Research into the role of the GM in MDD, BD and SSD is still in its infancy. This review is a narrative review which we have approached in a structured way and it presents data on the development and composition of the human gastrointestinal microbiota, and its interaction mechanisms in the gut–brain axis in MDD, BD and SSD. Furthermore, the therapeutic potential of pre/probiotics, diet and faecal microbiota transplantation (FMT) in these disorders will also be discussed. We begin by showing the different routes between the gut and brain and will then discuss the microbiome, after that we will discuss gut permeability and the GM in the psychiatric disorders. Finally, we will discuss potential mechanisms on how to adapt the GM to improve the clinical situation of patients.

Gut–brain axis

The gut and the brain communicate bidirectionally via several routes, including the vagal nerve, hypothalamic–pituitary–adrenal (HPA) axis, the production of bacterial metabolites, such as short-chain fatty acids (SCFAs), immune mediators and entero-endocrine signalling (Cryan et al., Reference Cryan, O'riordan, Cowan, Sandhu, Bastiaanssen, Boehme and Dinan2019; Golofast & Vales, Reference Golofast and Vales2020) (Fig. 1).

Fig. 1. (1) Environmental factors known to impinge on the human GM. (2) GM dysbiosis impairs intestinal permeability. (3) Increased intestinal permeability causes translocation of luminal components and reactivity of the intestinal immune system. (4) Bacterial translocation activates the gut–brain axis. (5) The gut and the brain communicate bidirectionally via several routes, including the vagal nerve, the HPA axis, immune mediators such as cytokines, and the production of bacterial metabolites, such as SCFAs. (6) The environmental factors, GM dysbiosis and increased permeability separately and in concert could contribute the development of psychiatric disorders. Created with BioRender.com.

Vagal nerve

The most direct route between the gut and the brain is the vagal nerve, roughly translated as the wandering nerve, and also known as the 10th cranial nerve. In reports from 1953 and 1961, ablation of the vagal nerve, formerly used for the treatment of peptic ulcers, resulted in an increase in the incidence of psychiatric disorders (Browning & Houseworth, Reference Browning and Houseworth1953; Whitlock, Reference Whitlock1961). Interestingly, this procedure recently turned out to reduce the incidence of Parkinson's disease (Svensson et al., Reference Svensson, Horváth-Puhó, Thomsen, Djurhuus, Pedersen, Borghammer and Sørensen2015). In animals, Sgritta et al. (Reference Sgritta, Dooling, Buffington, Momin, Francis, Britton and Costa-Mattioli2019) observed that the effects of Lactobacillus reuteri on social behaviour were dissipated after vagotomy in a genetic mouse model of autism (Shank3B−/− mouse).

Short-chain fatty acids

SCFAs are capable of signalling to the brain indirectly via nerve activation and can therefore influence behaviour (Sherwin, Sandhu, Dinan, & Cryan, Reference Sherwin, Sandhu, Dinan and Cryan2016). Ninety five per cent of SCFAs consist of acetate, propionate and butyrate. The primary source of SCFAs is microbial fermentation of dietary fibres in the gut, which is supported by the fact that germ-free animals and antibiotic treatment results in low SCFA levels (Cryan et al., Reference Cryan, O'riordan, Cowan, Sandhu, Bastiaanssen, Boehme and Dinan2019). SCFAs are capable of modulating neurotransmission. For example, propionic acid increases tryptophan hydroxylase expression which can reduce indoleamine serotonin, thereby influencing serotonergic neurotransmission (El-Ansary, Bacha, & Kotb, Reference El-Ansary, Bacha and Kotb2012; Nankova, Agarwal, MacFabe, & La Gamma, Reference Nankova, Agarwal, MacFabe and La Gamma2014). So far, only a few studies have investigated relations between neuropsychiatric disorders and SCFAs. Skonieczna-żydecka et al. (Reference Skonieczna-żydecka, Grochans, Maciejewska, Szkup, Schneider-Matyka, Jurczak and Stachowska2018) reported lower median content of acetate and higher isocaproic acid concentration in depressed women as compared to healthy women. Negative correlations were yielded between acetic, propionic and isocaproic acids and severity of depression as assessed with the Beck's Depression Inventory (BDI) scores. Szczesniak, Hestad, Hanssen, and Rudi (Reference Szczesniak, Hestad, Hanssen and Rudi2016) reported significantly higher isovaleric levels in the depressed patients as compared to healthy subjects. However, Kelly et al. (Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016) found no differences in acetate, propionate, isobutyrate and butyrate between depressed patients and healthy controls. The limited sample sizes in all three studies could explain these inconsistent results. In animal models of mania, sodium butyrate reversed mania-like, e.g. behavioural hyperactivity, and depressive-like behaviour (Moretti et al., Reference Moretti, Valvassori, Varela, Ferreira, Rochi, Benedet and Quevedo2011; Resende et al., Reference Resende, Valvassori, Réus, Varela, Arent, Ribeiro and Quevedoa2013; Valvassori et al., Reference Valvassori, Resende, Budni, Dal-Pont, Bavaresco, Reus and Quevedo2015)

Immune mediators

Immune mediators are important intermediaries between the gut microbiota and the brain. Cytokines can signal to the brain from the periphery via the vagal nerve or via the circumventricular organs (Sherwin et al., Reference Sherwin, Sandhu, Dinan and Cryan2016). In the blood, bacteria or fragments from bacteria can bind lipopolysaccharide (LPS) binding protein (LBP), which is then connected to TLR-4, expressed on monocytes, macrophages and microglia, by soluble cluster of differentiation 14 (sCD14) (Kiecolt-Glaser et al., Reference Kiecolt-Glaser, Wilson, Bailey, Andridge, Peng, Jaremka and Belury2018; Kitchens & Thompson, Reference Kitchens and Thompson2005; Lim, Chang, & Wu, Reference Lim, Chang and Wu2019), which can lead to nuclear factor kappa-light-chain enhancer of activated B cells (NFκB) activation, inducing pro-inflammatory cytokine production (Genedi et al., Reference Genedi, Janmaat, Haarman and Sommer2019; Sherwin et al., Reference Sherwin, Sandhu, Dinan and Cryan2016). Thus, the presence of bacteria or small parts of bacteria can activate the immune system which also affects the brain. Blood levels of LBP, sCD14 and NFκB can reflect activity of this route. The group of Robert Yolken showed that these levels are increased in patients with BD and SSD, when patients had comorbid gastro-intestinal complaints (Severance et al., Reference Severance, Gressitt, Stallings, Origoni, Khushalani, Leweke and Yolken2013; Severance, Dickerson, & Yolken, Reference Severance, Dickerson and Yolken2020). Moreover, Foster, Baker, and Dursun (Reference Foster, Baker and Dursun2021) recently reviewed the relation of the GM and the immune system in MDD, where they draw attention to the importance of the immune system as an important player in the neurobiology combined with the GM in subtypes of depression.

Tryptophan metabolites

Along with influencing other metabolites, gut-bacteria influence tryptophan metabolism (Carlessi, Borba, Zugno, Quevedo, & Réus, Reference Carlessi, Borba, Zugno, Quevedo and Réus2021). Tryptophan is an essential amino acid, which is metabolized by two main pathways, namely, the serotonin (5-HT) pathway and the kynurenine pathway. According to a meta-analysis of 101 studies tryptophan and kynurenine are decreased across MDD, BD and SSD (Marx et al., Reference Marx, McGuinness, Rocks, Ruusunen, Cleminson, Walker and Fernandes2020). Conventional antidepressants enhance levels of central serotonin to produce a therapeutic effect. Most of the tryptophan is metabolized into kynurenine in the liver by tryptophan-2,3-dioxygenase for energy production, or following an inflammatory stimulus by indoleamine-2,3-dioxygenase. The presence of gut bacteria such as Clostridium perfringens can modulate gut production of 5-HT (Beaver & Wostmann, Reference Beaver and Wostmann1962; Yano et al., Reference Yano, Yu, Donaldson, Shastri, Ann, Ma and Hsiao2015). Studies in rats have shown that probiotic treatment can influence tryptophan, kynurenine and serotonin levels (Sherwin et al., Reference Sherwin, Sandhu, Dinan and Cryan2016). Rats treated with Bifidobacterium infantis 35624 had increased circulating tryptophan levels (Desbonnet, Garrett, Clarke, Bienenstock, & Dinan, Reference Desbonnet, Garrett, Clarke, Bienenstock and Dinan2009). Furthermore, rats treated with Lactobacillus johnsonii had lower circulating kynurenine levels with increased serotonin levels in the ileum and in the circulation (Valladares et al., Reference Valladares, Bojilova, Potts, Cameron, Gardner, Lorca and Gonzalez2013). Moreover, one study in MDD patients found decreases in the kynurenine/tryptophan ratio after treatment with probiotics (Lactobacillus helveticus and Bifidobacterium longum) (Kazemi, Noorbala, Azam, Eskandari, & Djafarian, Reference Kazemi, Noorbala, Azam, Eskandari and Djafarian2019). A meta-analysis studying the effect of probiotic supplementation on the tryptophan–kynurenine pathway observed significantly lower serum kynurenine and a decreased kynurenine/tryptophan ratio after treatment (Purton et al., Reference Purton, Staskova, Lane, Dawson, West, Firth and Marx2021). The results so far provide preliminary evidence that probiotics can modulate the tryptophan–kynurenine pathway.

Gut microbiome

The GM contains 1014 microorganisms, 2–20 million unique genes and over 1000 unique bacterial species. The GM is a perplexing genomic structure with many more genes than the human genome (Golofast & Vales, Reference Golofast and Vales2020). In contrast to the human genome, which is unalterable over lifetime, the GM is highly adaptable (Nguyen et al., Reference Nguyen, Hathaway, Kosciolek, Knight and Jeste2021). The GM is already influenced by the environment at the day of birth. Vaginally delivered neonates' microbiota resembles the maternal vaginal and faecal bacteria, whereas for infants born by caesarean section, their microbiota resembles the maternal skin and hospital environment (Bäckhed et al., Reference Bäckhed, Roswall, Peng, Feng, Jia, Kovatcheva-Datchary and Jun2015; Korpela et al., Reference Korpela, Helve, Kolho, Saisto, Skogberg, Dikareva and de Vos2020; Mitchell et al., Reference Mitchell, Mazzoni, Hogstrom, Bryant, Bergerat, Cher and Yassour2020). In Reference Cannon, Jones and Murray2002 Cannon, Jones and Murray already identified unplanned caesarean section as a risk factor for schizophrenia in their seminal meta-analysis. One epidemiological study found a weak association between birth by planned caesarean section, but not by unplanned emergency caesarean section and the risk of developing psychosis or BD (O'Neill et al., Reference O'Neill, Curran, Dalman, Kenny, Kearney, Clarke and Khashan2016). However, after correcting for matched siblings, this effect was no longer significant. The Finnish birth register data also showed an odds ratio (OR) of 2.5 for BD for birth by caesarean section (Chudal et al., Reference Chudal, Sourander, Polo-Kantola, Hinkka-Yli-Salomäki, Lehti, Sucksdorff and Brown2014).

Associations between antibiotic exposure and psychiatric disorders have been found in multiple large population-based studies (Köhler et al., Reference Köhler, Petersen, Mors, Mortensen, Yolken, Gasse and Benros2017; Liang et al., Reference Liang, Ye, Wen, Li, Cheng, Cheng and Zhang2021; Lurie, Yang, Hayne, Mamtani, & Boursi, Reference Lurie, Yang, Hayne, Mamtani and Boursi2015). Lurie et al. (Reference Lurie, Yang, Hayne, Mamtani and Boursi2015) found that treatment with a single antibiotic course was associated with higher risk for depression (n = 202,974) compared to a healthy control group (n = 803,961), with adjusted ORs of 1.23 [95% confidence level (CL) 1.18–1.29] for penicillin's and 1.25 (95% CL 1.15–1.35) for quinolones. Köhler et al. (Reference Köhler, Petersen, Mors, Mortensen, Yolken, Gasse and Benros2017) performed a large-scale study in individuals born in Denmark in 1985–2002 (n = 1,015,447), of which 5759 individuals were diagnosed with schizophrenia. The association of infections treated with anti-infective agents and the subsequent risk of schizophrenia and affective disorders during 1995–2013 was studied. Antibiotics, with a hazard rate ratio (HRR) of 1.44 (95% CL 1.25–1.66), and especially broad spectrum antibiotics (HRR = 1.53; 95% CL 1.32–1.71) were associated with increased risk for schizophrenia. However, Lurie et al. (Reference Lurie, Yang, Hayne, Mamtani and Boursi2015) did not find an association between antibiotic use and psychosis (n = 8487). Liang et al. (Reference Liang, Ye, Wen, Li, Cheng, Cheng and Zhang2021) observed positive associations between long-term antibiotic use during early life (defined by the UK Biobank as child or teenager) and anxiety and depression. Lastly, there are also studies showing that antibiotic use can induce hypomania or mania, also known as antibiomania (Abouesh, Stone, & Hobbs, Reference Abouesh, Stone and Hobbs2002). In a review of 47 published cases, clarithromycin, a broad spectrum antibiotic, was related to (hypo)mania in 16 out of the 47 cases (Lambrichts, Van Oudenhove, & Sienaert, Reference Lambrichts, Van Oudenhove and Sienaert2017). One explanation could be that the administration of antibiotics can result in changes in the microbiome which could in turn increases the risk of (hypo)mania, for example by lowering the bacterial production of gamma-amino-butyric acid (Dickerson, Severance, & Yolken, Reference Dickerson, Severance and Yolken2017).

Gastrointestinal permeability in patients with severe psychiatric disorders

Gastrointestinal barrier

The gastrointestinal barrier is a dynamic, multilayer system which consists of a physical barrier and a biochemical barrier. The main components of the physical barrier are the epithelial cells sealed by tight junctions and the gut mucosa. The biochemical barrier consists of the gut microbiota and the mucosal immune system. Its main function is to keep pathogens out of the host's internal milieu, while at the same time facilitate the absorption of nutrients, water and electrolytes (Bischoff et al., Reference Bischoff, Barbara, Buurman, Ockhuizen, Schulzke, Serino and Wells2014). For the homoeostasis of the organism, the permeability of the gastrointestinal barrier needs to be maintained within a narrow equilibrium.

Diet and lifestyle factors such as energy-dense food and alcohol can disturb gut permeability and lead to translocation of luminal components and reactivity of the intestinal immune system (Bischoff et al., Reference Bischoff, Barbara, Buurman, Ockhuizen, Schulzke, Serino and Wells2014). Increased intestinal permeability can also be a result of changes in gut microbiota, infections or reduced perfusion of the gut (Bischoff et al., Reference Bischoff, Barbara, Buurman, Ockhuizen, Schulzke, Serino and Wells2014).

Gastrointestinal permeability and brain structure and function

Gastrointestinal permeability and brain structure and function are governed by a bidirectional interaction. On one hand, pre-clinical studies (Keita, Söderholm, & Ericson, Reference Keita, Söderholm and Ericson2010; Kiliaan et al., Reference Kiliaan, Saunders, Bijlsma, Cecilia Berin, Taminiau, Groot and Perdue1998; Söderholm et al., Reference Söderholm, Yang, Ceponis, Vohra, Riddell, Sherman and Perdue2002; Vicario et al., Reference Vicario, Guilarte, Alonso, Yang, Martínez, Ramos and Santos2010) and studies in healthy volunteers (Vanuytsel et al., Reference Vanuytsel, Van Wanrooy, Vanheel, Vanormelingen, Verschueren, Houben and Tack2014) suggest a causal effect of psychosocial stress on gut permeability, likely through corticotropin-releasing hormone-mediated mast cell activation and decreased blood flow to the gut during stressful periods. On the other hand, it is hypothesized that increased gut permeability and abnormal influx of food- and bacteria-derived antigens drives systemic low-grade immune dysregulation, which in turn affects brain structure and function (Genedi et al., Reference Genedi, Janmaat, Haarman and Sommer2019). Lastly, psychiatric comorbidity is common in diseases with known structural and functional abnormalities of the gastrointestinal barrier, such as Crohn's disease and colitis ulcerosa (Bennett, Tennant, Piesse, Badcock, & Kellow, Reference Bennett, Tennant, Piesse, Badcock and Kellow1998; Faresjö et al., Reference Faresjö, Grodzinsky, Johansson, Wallander, Timpka and Åkerlind2007; Nicholl et al., Reference Nicholl, Halder, Macfarlane, Thompson, O'Brien, Musleh and McBeth2008). It is therefore hypothesized that abnormal intestinal permeability is involved both as an effect and as a cause of severe psychiatric disorder, yet, this is still a research area in its infancy.

Evidence of gastrointestinal barrier dysfunction in MDD, BD and SSD

In MDD, BD and SSD, gastrointestinal permeability has been assessed with markers of structural barrier integrity and paracellular permeability such as zonulin and intestinal-type fatty acid-binding protein (I-FABP) (Table 1). In a study in MDD or anxiety disorder, authors reported significantly higher levels of zonulin and I-FABP in patients compared to controls and this was associated with gut dysbiosis (Stevens et al., Reference Stevens, Goel, Seungbum, Richards, Holbert, Pepine and Genomics2018). Alvarez-Mon et al. (Reference Alvarez-Mon, Gómez, Orozco, Lahera, Sosa, Diaz and Alvarez-Mon2019) also reported significantly higher levels of I-FABP in MDD patients compared to controls but no significant difference was observed for zonulin. In another study, patients of different psychiatric diagnoses who had recently attempted suicide had higher levels of I-FABP but lower levels of zonulin compared to both MDD patients without a history of suicide attempt and healthy controls (Ohlsson et al., Reference Ohlsson, Gustafsson, Lavant, Suneson, Brundin, Westrin and Lindqvist2019). In BD, patients in full remission showed higher zonulin and claudin-5 compared to controls, while there were no differences between active manic episode and remission (Kılıç, Işık, Demirdaş, Doğuç, & Bozkurt, Reference Kılıç, Işık, Demirdaş, Doğuç and Bozkurt2020). Interestingly, this observation might suggest that zonulin and claudin-5 are ‘trait’ rather than ‘state’ markers of BD. However, the small sample size of this study precludes firm conclusions, before the findings are confirmed in larger populations. Moreover, there is a certain level of uncertainty surrounding the measurement of zonulin with commercially available ELISA kits. The results should be interpreted with caution until an in depth understanding of the biomarker is acquired (Massier, Chakaroun, Kovacs, & Heiker, Reference Massier, Chakaroun, Kovacs and Heiker2021). In schizophrenia, Maes, Kanchanatawan, Sirivichayakul, and Carvalho (Reference Maes, Kanchanatawan, Sirivichayakul and Carvalho2019a) and Maes, Sirivichayakul, Kanchanatawan, and Vodjani (Reference Maes, Sirivichayakul, Kanchanatawan and Vodjani2019b) showed that immunoglobulin M (IgM) responses to zonulin were higher in patients compared to controls, while IgM response to occludin was significantly associated with deficit schizophrenia. Authors also reported an increased ratio of IgM towards components of the paracellular route (zonulin + occludin)/components of the transcellular route (talin + actin + vinculin) in deficit v. non-deficit schizophrenia and in schizophrenia v. controls. This ratio was significantly associated with increased IgA responses to Gram-negative bacteria. Schizophrenia patients with deficit syndrome (i.e. severe negative and cognitive symptoms) can be expected to have unhealthy dietary habits, which could explain the association with IgM response to occludin. Barber et al. (Reference Barber, Sturgeon, Fasano, Gascella, Eaton, McMahon and Kelly2019) showed that 42.9% of the patients had higher levels of zonulin than the cut-off for elevated levels (>2.33 mg/dl).

Table 1. Main findings of studies assessing gut permeability in MDD, BD and SSD

sCD14, soluble cluster of differentiation 14; LBP, lipopolysaccharide (LPS) binding protein; I-FABP, fatty acid-binding protein.

Markers of bacteria translocation can also reflect abnormal gastrointestinal permeability. LPS and LBP were up-regulated in MDD or anxiety disorder (Alvarez-Mon et al., Reference Alvarez-Mon, Gómez, Orozco, Lahera, Sosa, Diaz and Alvarez-Mon2019; Stevens et al., Reference Stevens, Goel, Seungbum, Richards, Holbert, Pepine and Genomics2018). In BD, sCD14, a co-receptor for the LBP, used as a marker for bacterial translocation was significantly higher in patients compared to controls (Severance et al., Reference Severance, Gressitt, Stallings, Origoni, Khushalani, Leweke and Yolken2013). In SSD, sCD14 was significantly higher in multiple studies (Dzikowski et al., Reference Dzikowski, Juchnowicz, Dzikowska, Rog, Próchnicki, Kozioł and Karakula-Juchnowicz2020; Severance et al., Reference Severance, Gressitt, Stallings, Origoni, Khushalani, Leweke and Yolken2013; Tanaka et al., Reference Tanaka, Matsuda, Hayes, Yang, Rodriguez, Severance and Eaton2017; Weber et al., Reference Weber, Gressitt, Cowan, Niebuhr, Yolken and Severance2018) but not all (Morch et al., Reference Morch, Dieset, Færden, Reponen, Hope, Hoseth and Andreassen2019). In regard to antibodies against gut commensal bacteria, in MDD, Maes et al. (Reference Maes, Kubera, Leunis, Berk, Geffard and Bosmans2013) showed increased IgM/IgA against Gram-negative bacteria. More recently, Simeonova et al. (Reference Simeonova, Stoyanov, Leunis, Carvalho, Kubera, Murdjeva and Maes2020) showed increased immune responses to Gram-negative bacteria in both MDD and BD, especially in the presence of melancholia, compared to healthy controls. Interestingly, IgA/IgM response profiles differ among the two diagnosis, as well as between BD I and II subtypes and patients with melancholia. This might reflect a common underlying disruption of the gut barrier across diagnoses, accompanied however by distinct microbiome profiles and immune susceptibilities, though this remains to be confirmed in future studies. In SSD IgM/IgA against gut commensal bacteria were associated with negative symptoms, neurocognitive impairments and the deficit phenotype of SSD (Maes et al., Reference Maes, Sirivichayakul, Kanchanatawan and Vodjani2019b).

GM in three major psychiatric disorders

Alpha and beta diversity

α-Diversity is the mean species diversity in sites or habitats at a local scale (Whittaker, Reference Whittaker1972). α-Diversity is often measured by the Fisher, Ace, Chao, Simpson and/or Shannon indices. β-Diversity shows differentiation among habitats (Whittaker, Reference Whittaker1972). Multiple studies analysed diversity metrics; the findings however were mixed.

For MDD studies (Table 2), α-diversity was not different from controls in three studies (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019; Zheng et al., Reference Zheng, Zeng, Zhou, Liu, Fang, Xu and Fan2016, Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020). Two studies found lower α-diversity in MDD (Huang et al., Reference Huang, Shi, Li, Shen, Shi, Wang and Liang2018; Liu et al., Reference Liu, Zhang, Wang, Wang, Zhang, Jiang and Duan2016). Four studies found inconsistent findings across indices (Jiang et al., Reference Jiang, Ling, Zhang, Mao, Ma, Yin and Ruan2015; Kelly et al., Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016; Lai et al., Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019). β-Diversity was significantly different between MDD and controls in four studies (Huang et al., Reference Huang, Shi, Li, Shen, Shi, Wang and Liang2018; Kelly et al., Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016; Lai et al., Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021; Zheng et al., Reference Zheng, Zeng, Zhou, Liu, Fang, Xu and Fan2016) and not different in three studies (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019; Jiang et al., Reference Jiang, Ling, Zhang, Mao, Ma, Yin and Ruan2015; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019).

Table 2. Studies of the GM in major depressive disorder

HC, healthy controls; MDD, major depressive disorder; PCoA, principal coordinates analysis; PLS-DA, partial least-squares discriminant analysis; HDRS, Hamilton Depression Rating Scale; MADRS, Montgomery–Åsberg Depression Rating Scale; HAMDS, Hamilton's Depression Scale; PROMIS Depression Score: Patient-Reported Outcomes Measurement Information System Depression Score; OTU, operational taxonomic unit.

For α-diversity in the BD studies (Table 3), three had inconsistent findings (Hu et al., Reference Hu, Li, Huang, Lai, Li, Sublette and Xu2019; Painold et al., Reference Painold, Mörkl, Kashofer, Halwachs, Dalkner, Bengesser and Reininghaus2018; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019) and one found no differences (Zheng et al., Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020). β-Diversity was different in two studies (Hu et al., Reference Hu, Li, Huang, Lai, Li, Sublette and Xu2019; Zheng et al., Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020) and two found no differences (Painold et al., Reference Painold, Mörkl, Kashofer, Halwachs, Dalkner, Bengesser and Reininghaus2018; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019).

Table 3. Studies of the GM in individuals with BD

HC, healthy controls; BD, bipolar disorder; PLS-DA, partial least-squares discriminant analysis; PCoA, principal coordinates analysis; HCL-32, Hypomania Check List-32; HAMD, Hamilton's Depression Scale; BDI, Beck Depression Inventory; MADRS, Montgomery–Åsberg Depression Rating Scale; HDRS-17, 17-item Hamilton Depression Rating Scale.

In SSD (Table 4), three studies reported lower α-diversity in the SSD groups compared to healthy controls (Ma et al., Reference Ma, Asif, Dai, He, Zheng, Wang and Chen2020; Xu et al., Reference Xu, Wu, Liang, He, Gu, Li and Wang2020; Zheng et al., Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019), four reported no differences (He et al., Reference He, Kosciolek, Tang, Zhou, Li, Ma and Chen2018; Li et al., Reference Li, Zhuo, Huang, Huang, Zhou, Xiong and Wu2020; Nguyen et al., Reference Nguyen, Kosciolek, Maldonado, Daly, Martin, McDonald and Jeste2018; Shen et al., Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018), two did not report on α-diversity (Schwarz et al., Reference Schwarz, Maukonen, Hyytiäinen, Kieseppä, Orešič, Sabunciyan and Suvisaari2018; Yuan et al., Reference Yuan, Zhang, Wang, Liu, Li, Kumar and Song2018) and one reported higher α-diversity in the SSD group (Zhu et al., Reference Zhu, Ju, Wang, Wang, Guo, Ma and Ma2020b). β-Diversity was different between groups in five studies (He et al., Reference He, Kosciolek, Tang, Zhou, Li, Ma and Chen2018; Nguyen et al., Reference Nguyen, Hathaway, Kosciolek, Knight and Jeste2021; Shen et al., Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018; Xu et al., Reference Xu, Wu, Liang, He, Gu, Li and Wang2020; Zheng et al., Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019), from those five, two studies reported tighter clustering in the healthy control group (Nguyen et al., Reference Nguyen, Kosciolek, Maldonado, Daly, Martin, McDonald and Jeste2018; Shen et al., Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018).

Table 4. Studies of the GM in schizophrenia-spectrum disorder

HC, healthy controls; FPE, first psychotic episode patients; SSD, schizophrenia-spectrum disorder patients; FSCZ, antipsychotic-free first episode patients; TSCZ, chronically antipsychotic-treated patients; OTU, operational taxonomic unit; PLS-DA, partial least-squares discriminant analysis; NMDS, non-metric multidimensional scaling; PCoA, principal coordinates analysis; BPRS, Brief Psychiatric Rating Scale; PANSS, Positive and Negative Syndrome Score.

In the present review more studies reported inconsistent findings (n = 7) or no differences (n = 8) than studies who reported lower α-diversity in the psychiatric disorders (n = 5). These results are in line with other studies (Sanada et al., Reference Sanada, Nakajima, Kurokawa, Barceló-Soler, Ikuse, Hirata and Kishimoto2020; Simpson et al., Reference Simpson, Diaz-Arteche, Eliby, Schwartz, Simmons and Cowan2021), suggesting that host–microbe interactions are more complex than can be modelled by α-diversity.

Findings at different taxonomic levels

A large number of bacterial taxa were significantly different in their relative abundance between control and psychiatric groups [MDD groups (Table 2), BD groups (Table 3) and SSD groups (Table 4)]. Interestingly, multiple bacterial taxa abundances were similar between psychiatric disorders (Fig. 2). All investigations reported taxonomic differences between the neuropsychiatric disorders and healthy control groups; in the next paragraphs the most important differences, similarities and findings are discussed.

Fig. 2. Taxonomic differences in neuropsychiatric disorders (at the phylum, family and genus levels), whereby ‘↑’ = higher relative abundance in the neuropsychiatric disorder group, ‘↓’ = lower relative abundance in the neuropsychiatric group and ‘I’ = no differences in abundance. The letters below the arrows refer to the studies the information was retrieved from and also can be connected to the letters in the tables. Studies: AZheng et al. (Reference Zheng, Zeng, Zhou, Liu, Fang, Xu and Fan2016); BJiang et al. (Reference Jiang, Ling, Zhang, Mao, Ma, Yin and Ruan2015); CKelly et al. (Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016); DChung et al. (Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019); EHuang et al. (Reference Huang, Shi, Li, Shen, Shi, Wang and Liang2018); FLai et al. (Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021); GLiu et al. (Reference Liu, Zhang, Wang, Wang, Zhang, Jiang and Duan2016); HZheng et al. (Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020); IRong et al. (Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019); JLiu et al. (Reference Liu, Rowan-Nash, Sheehan, Walsh, Sanzari, Korry and Belenky2020); KZheng et al. (Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019); LXu et al. (Reference Xu, Wu, Liang, He, Gu, Li and Wang2020); MNguyen et al. (Reference Nguyen, Kosciolek, Maldonado, Daly, Martin, McDonald and Jeste2018); NShen et al. (Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018); OHe et al. (Reference He, Kosciolek, Tang, Zhou, Li, Ma and Chen2018); PLi et al. (Reference Li, Zhuo, Huang, Huang, Zhou, Xiong and Wu2020); QRong et al. (Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019); RPainold et al. (Reference Painold, Mörkl, Kashofer, Halwachs, Dalkner, Bengesser and Reininghaus2018); SSchwarz et al. (Reference Schwarz, Maukonen, Hyytiäinen, Kieseppä, Orešič, Sabunciyan and Suvisaari2018); THu et al. (Reference Hu, Li, Huang, Lai, Li, Sublette and Xu2019); UZheng et al. (Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020); VPainold et al. (Reference Painold, Mörkl, Kashofer, Halwachs, Dalkner, Bengesser and Reininghaus2018); WRong et al. (Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019).

Firmicutes

The most abundant phylum in the human gut, Firmicutes, showed multiple inconsistencies at the phylum, family and genus levels (Fig. 2). At the genus level, divergent findings were reported for Ruminococcus, which was higher in one MDD study (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019), but lower in three other MDD studies (Jiang et al., Reference Jiang, Ling, Zhang, Mao, Ma, Yin and Ruan2015; Liu et al., Reference Liu, Rowan-Nash, Sheehan, Walsh, Sanzari, Korry and Belenky2020, Reference Liu, Zhang, Wang, Wang, Zhang, Jiang and Duan2016), one BD study (Hu et al., Reference Hu, Li, Huang, Lai, Li, Sublette and Xu2019) and one SSD study (Li et al., Reference Li, Zhuo, Huang, Huang, Zhou, Xiong and Wu2020). In line with these results, inconsistent findings were also reported in relation to clinical features. One study reported that decreased relative abundance of Ruminococcaceae was associated with an increase in negative symptoms (Nguyen et al., Reference Nguyen, Kosciolek, Maldonado, Daly, Martin, McDonald and Jeste2018). Another study reported Ruminococcaceae OTU725 to be negatively correlated with symptom severity in SSD (Zheng et al., Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019). In contrast, one study found a positive correlation of Ruminococcaceae with negative symptoms (Schwarz et al., Reference Schwarz, Maukonen, Hyytiäinen, Kieseppä, Orešič, Sabunciyan and Suvisaari2018). One MDD study found a positive correlation between Ruminococcus and symptom severity scores (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019). Another MDD study found a negative association of Ruminococcaceae with symptom severity (Liu et al., Reference Liu, Rowan-Nash, Sheehan, Walsh, Sanzari, Korry and Belenky2020).

Moreover, at the genus level Faecalibacterium were decreased in five MDD studies (Huang et al., Reference Huang, Shi, Li, Shen, Shi, Wang and Liang2018; Jiang et al., Reference Jiang, Ling, Zhang, Mao, Ma, Yin and Ruan2015; Kelly et al., Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016; Liu et al., Reference Liu, Rowan-Nash, Sheehan, Walsh, Sanzari, Korry and Belenky2020, Reference Liu, Zhang, Wang, Wang, Zhang, Jiang and Duan2016), one BD study (Hu et al., Reference Hu, Li, Huang, Lai, Li, Sublette and Xu2019) and one SSD study (Li et al., Reference Li, Zhuo, Huang, Huang, Zhou, Xiong and Wu2020). In line with these results Jiang et al. (Reference Jiang, Ling, Zhang, Mao, Ma, Yin and Ruan2015) found a negative correlation between Faecalibacterium and depressive symptom severity. Furthermore, Liu et al. (Reference Liu, Rowan-Nash, Sheehan, Walsh, Sanzari, Korry and Belenky2020) observed reduced Faecalibacterium in MDD patients with more severe symptoms. Moreover, the recently published systematic review and meta-analysis of Nikolova et al. (Reference Nikolova, Hall, Hall, Cleare, Stone and Young2021) observed depleted levels of Faecalibacterium in BD, MDD and schizophrenia as well. Faecalibacterium, especially species Faecalibacterium prausnitzii, is usually considered a ‘good’ gut bacterium and is associated with positive healthy outcomes, and its depletion with negative healthy outcomes (Ferreira-Halder, de Faria, & Andrade, Reference Ferreira-Halder, de Faria and Andrade2017; Gacesa et al., Reference Gacesa, Kurilshikov, Vich Vila, Sinha, Klaassen, Bolte and Weersma2022). Furthermore, F. prausnitzii was negatively associated with MDD (Gacesa et al., Reference Gacesa, Kurilshikov, Vich Vila, Sinha, Klaassen, Bolte and Weersma2022).

The genus Streptococcus was reported to be higher in three MDD studies (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019; Lai et al., Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019), one BD study (Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019) and one SSD study (Shen et al., Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018). In line with these results, Zheng et al. (Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019) reported a positive correlation between Streptococcaceae OTU834 and symptoms severity. At the family level, the relative abundance of Lactobacillaceae was reported to be higher in two SSD studies (Schwarz et al., Reference Schwarz, Maukonen, Hyytiäinen, Kieseppä, Orešič, Sabunciyan and Suvisaari2018; Shen et al., Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018). Consistently, at the genus level the relative abundance of Lactobacillus was significantly higher in two MDD studies (Lai et al., Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019), in one BD study (Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019) and in four SSD studies (He et al., Reference He, Kosciolek, Tang, Zhou, Li, Ma and Chen2018; Li et al., Reference Li, Zhuo, Huang, Huang, Zhou, Xiong and Wu2020; Schwarz et al., Reference Schwarz, Maukonen, Hyytiäinen, Kieseppä, Orešič, Sabunciyan and Suvisaari2018; Shen et al., Reference Shen, Xu, Li, Huang, Yuan, Wang and Liang2018). In line with these results, general symptom severity and positive symptom severity were positively correlated with Lactobacillus (Schwarz et al., Reference Schwarz, Maukonen, Hyytiäinen, Kieseppä, Orešič, Sabunciyan and Suvisaari2018). These are interesting results since specific strains of Lactobacillus are commonly used in probiotics (Simpson et al., Reference Simpson, Diaz-Arteche, Eliby, Schwartz, Simmons and Cowan2021). However, other studies have observed increased Lactobacillus as well in other disorders, like inflammatory bowel disease, indicating specific strains may have inflammatory potential (Wang et al., Reference Wang, Chen, Zhou, Wang, Song, Huang and Xia2014). Rocha-Ramírez et al. (Reference Rocha-Ramírez, Pérez-Solano, Castañón-Alonso, Moreno Guerrero, Ramírez Pacheco, García Garibay and Eslava2017) found that several Lactobacillus species increased proinflammatory cytokines such as interleukin-8 (IL-8), tumour necrosis factor-α, IL-12p70 and IL-6. Moreover, Zhu et al. (Reference Zhu, Ju, Wang, Wang, Guo, Ma and Ma2020b) reported increases of subspecies of Lactobacillus not typically present in the healthy gut in schizophrenia patients. It remains to be determined which species of Lactobacillus genus are increased in psychiatric disorders.

Actinobacteria

At the family level in the phylum Actinobacteria, Coriobacteriaceae were relatively more abundant in one BD study (Painold et al., Reference Painold, Mörkl, Kashofer, Halwachs, Dalkner, Bengesser and Reininghaus2018) and one SSD study (Zheng et al., Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019). From Coriobacteriaceae, the abundance of Eggerthella was relatively higher in four MDD studies (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019; Kelly et al., Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016; Lai et al., Reference Lai, Deng, Xu, Zhao, Xu, Liu and Rong2021; Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019), in one BD study (Rong et al., Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019) and one SSD study (Xu et al., Reference Xu, Wu, Liang, He, Gu, Li and Wang2020). In line with these results, Eggerthella correlated positively with depression, anxiety and stress scores (Chung et al., Reference Chung, Chen, Chou, Chen, Lee, Chuang and Kuo2019). Gacesa et al. (Reference Gacesa, Kurilshikov, Vich Vila, Sinha, Klaassen, Bolte and Weersma2022) found a positive association of the species Eggerthella lenta with BD. Moreover, Rekdal, Bess, Bisanz, Turnbaugh, and Balskus (Reference Rekdal, Bess, Bisanz, Turnbaugh and Balskus2019) suggest that E. lenta is capable of selectively removing the para-hydroxyl group of dopamine, thereby decreasing dopamine levels. This suggestion of Rekdal et al. (Reference Rekdal, Bess, Bisanz, Turnbaugh and Balskus2019) could explain the positive correlation found between Eggerthella and depression scores. Moreover, the BD patients in the study of Rong et al. (Reference Rong, Xie, Zhao, Lai, Wang, Xu and Liu2019) were at the time of the study in a major depressive episode. Eggerthella could be related to depression in multiple disorders and could therefore be a target for depression. Unfortunately, Eggerthella and E. lenta, previously known as Eubacterium lentum, have been underrecognized due to historical difficulties with phenotypic identification, therefore not much information is available about the bacterial species.

Only a few studies have investigated microbiome difference among subtypes of psychiatric disorders. Hu et al. (Reference Hu, Li, Huang, Lai, Li, Sublette and Xu2019) compared the GM of BD type 1 to BD type 2 patients with each other. They observed relatively higher relative abundance of the families Streptococcaceae and Erysipelotrichaceae, genera Streptococcus, Bacilli and Veillonella and lower relative abundance of the genus Ruminococcus in the BD type 1 group compared to the BD type 2 group. Zheng et al. (Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020) compared unipolar depression to bipolar depression and found Bacteroidaceae and Veilonellaceae to be higher and Enterobacteriaceae and Pseudomonadaceae lower in MDD v. BD. Studying the GM of subtypes of psychiatric disorders has the potential to be of great value in understanding differences between these subtypes.

Inconsistencies across studies may be attributable partly to the heterogeneity in sample characteristics across studies. Most studies used small sample sizes; only one study used a sample size of more than 100 patients (Zheng et al., Reference Zheng, Yang, Li, Wu, Liang, Yin and Wang2020). In addition, dietary habits may change among sites of study and socio-economic class of the participants. Then, psychiatric medication probably affects the microbiome. Methods used for obtaining the taxonomic profiles were not consistent. Moreover, statistical analysis used to compare GM composition between groups was quite heterogeneous across studies. Microbiota composition differences between groups were analysed by using several statistical methods, namely, analysis of composition of microbiomes, linear discriminant analysis effect size, Wilcoxon rank-sum test, Mann–Whitney U test, Kruskal–Wallis test and Welch's t test.

The most consistent findings across studies were higher relative abundances of the genera Streptococcus, Lactobacillus and Eggerthella and lower relative abundances of the genus Faecalibacterium in the neuropsychiatric disorders (Fig. 2), perhaps most interesting in the relationship between the psychiatric disorders and lower levels of Faecalibacterium. F. prausnitzii has been shown to have anti-inflammatory effects, to produce the SCFA butyrate and has been associated with improving the intestinal barrier by increasing levels of tight junction proteins occludin and E-cadherin (Carlsson et al., Reference Carlsson, Yakymenko, Olivier, Håkansson, Postma, Keita and Söderholm2013; He, Zhao, & Li, Reference He, Zhao and Li2021; Laval et al., Reference Laval, Martin, Natividad, Chain, Miquel, Desclée de Maredsous and Langella2015; Martín et al., Reference Martín, Miquel, Chain, Natividad, Jury, Lu and Bermúdez-Humarán2015). Next to that, F. prausnitzii has been associated with smoking, which is in line with the fact that the percentage of smokers is higher in psychiatric disorders (Gacesa et al., Reference Gacesa, Kurilshikov, Vich Vila, Sinha, Klaassen, Bolte and Weersma2022; Lê Cook et al., Reference Lê Cook, Wayne, Kafali, Liu, Shu and Flores2014). Multiple of the findings, like higher relative abundances of Actinobacteria and lower abundancy of Prevotella in MDD have been associated unhealthy dietary patterns. Higher relative abundances of Actinobacteria have been associated with high-fat and animal protein diet (Fritsch et al., Reference Fritsch, Garces, Quintero, Pignac-Kobinger, Santander, Fernández and Abreu2021). Low carbohydrate intake has been associated with reduced Prevotellaceae (Kang et al., Reference Kang, Park, Ilhan, Wallstrom, LaBaer, Adams and Krajmalnik-Brown2013). However, the majority of the studies did not control for diet, making it difficult to relate the findings to possible unhealthy diet patterns in the disorders.

Treatments targeting the GM

Understanding how our GM can be targeted could lead to the development of microbiota-based therapies.

Probiotics

Probiotics are defined as live organisms that exert a health benefit when ingested in an adequate amount. Probiotics contain living beneficial bacteria, traditionally from genera Lactobacilli and Bifidobacteria.

In MDD, Kazemi et al. (Reference Kazemi, Noorbala, Azam, Eskandari and Djafarian2019) conducted an 8 week randomized, double-blind, placebo-controlled study, in which 110 depressed patients were randomly assigned to receive probiotic (L. helveticus and B. longum), prebiotic (galactooligosaccharide) or placebo treatment. Probiotic supplementation resulted in a significant decrease in symptom severity compared to both the prebiotic and the placebo groups. Additionally, the serum kynurenine/tryptophan ratio was significantly decreased in the probiotic group compared to the placebo group. Another randomized, double-blind, placebo-controlled 8 weeks probiotics study by Akkasheh et al. (Reference Akkasheh, Kashani-Poor, Tajabadi-Ebrahimi, Jafari, Akbari, Taghizadeh and Esmaillzadeh2016) studied a probiotic (Lactobacillus acidophilus, Lactobacillus casei and Bifidobacterium bifidum) in 40 MDD patients. As a result symptom severity decreased significantly in the probiotic group. In addition, serum insulin levels, insulin resistance and serum high-sensitivity C-reactive protein concentrations were decreased.

In BD, Dickerson et al. (Reference Dickerson, Adamos, Katsafanas, Khushalani, Origoni, Savage and Yolken2018) found that the adjunctive probiotic treatment (Lactobacillus rhamnosus strain GG and Bifidobacterium animalis subsp. lactis strain Bb12) for 24 weeks prevented rehospitalization in patients with acute mania (n = 66). Probiotic treatment also resulted in fewer days of hospitalization. Another study (n = 38) found no effects of probiotics compared to placebo on symptom severity of both depression and mania (Shahrbabaki et al., Reference Shahrbabaki, Sabouri, Sabahi, Barfeh, Divsalar, Esmailzadeh and Ahmadi2020). In another longitudinal cohort study 20 euthymic individuals with BD received a probiotic called ‘OMNi-BiOTiC® Stress Repair’ (B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W22, L. casei W56, Lactobacillus paracasei W20, L. plantarum W62, L. salivarius W24, L. lactis W19) over a time period of 3 months (Reininghaus et al., Reference Reininghaus, Wetzlmair, Fellendorf, Platzer, Queissner, Birner and Dalkner2020). Cognition concerning attention and psychomotor processing speed was improved as well as executive functioning.

Three studies investigated the effect of probiotics in SSD patients. One studied the combination three bacteria (L. acidophilus, B. bifidum, L. reuteri and L. fermentum) and vitamin D in a randomized placebo-controlled trial (n = 60) (Ghaderi et al., Reference Ghaderi, Banafshe, Mirhosseini, Moradi, Karimi, Mehrzad and Asemi2019). The combination was associated with significant improvement in symptom severity and had beneficial effects on metabolic profiles in reducing fasting plasma glucose levels, insulin concentrations, triglycerides and total cholesterol levels. Another study investigated a probiotic (L. acidophilus, B. lactis, B. bifidum and B. longum) and selenium co-supplementation in a randomized placebo-controlled trial with 60 people with chronic schizophrenia (Jamilian & Ghaderi, Reference Jamilian and Ghaderi2021). The probiotic and selenium co-supplementation resulted in a significant improvement in symptom severity compared to the placebo group. However, in both studies it is uncertain which component of the intervention was responsible for the changes. Only one study studied the effect of a probiotic alone (Dickerson et al., Reference Dickerson, Stallings, Origoni, Katsafanas, Savage, Schweinfurth and Yolken2014). Their 65 participants were subjected to a double-blind adjunctive probiotic (L. rhamnosus strain GG and B. animalis subsp. lactis strain Bb12) or placebo therapy for 14 weeks. No significant differences were found in symptom severity between probiotic and placebo supplementation.

Prebiotics

Prebiotics are non-digestible fibres that are selectively metabolized by the intestinal microbiome. Prebiotics mostly include fructans and glucans. So far, only one study has studied the effects of prebiotics in SSD, namely, by using the purified prebiotic Bimuno™ galactooligosaccharides (B-GOS®) formulation. Thirty-nine non-hospitalized participants with non-affective psychosis received either B-GOS® or a placebo for 24 weeks. Participants performed the Brief Assessment of Cognition in Schizophrenia (BACS, Keefe et al., Reference Keefe, Goldberg, Harvey, Gold, Poe and Coughenour2004) and the B-GOS® group had significantly improved global cognitive performance, compared to the placebo group (Kao et al., Reference Kao, Safarikova, Marquardt, Mullins, Lennox and Burnet2019). While interesting, this study requires replication in larger studies.

Faecal microbiota transplantation

Faecal transplants from depressed patients to germ-free rats show interesting results. Kelly et al. (Reference Kelly, Borre, O’ Brien, Patterson, El Aidy, Deane and Dinan2016) showed that FMT from depressed patients to microbiota-depleted rats can induce behavioural and physiological features characteristic of MDD in the receiving animals, including anhedonia and anxiety-like behaviour, as well as an increased plasma kynurenine/tryptophan ratio. In two studies, FMT from schizophrenia patients to germ-free mice led to hyperactivity and reduced anxiety in the open-field test (Zheng et al., Reference Zheng, Zeng, Liu, Chen, Pan, Han and Xie2019; Zhu et al., Reference Zhu, Guo, Wang, Ju, Wang, Ma and Ma2020a). Zhu et al. (Reference Zhu, Guo, Wang, Ju, Wang, Ma and Ma2020a) also found that mice that received FMT from schizophrenia patients displayed impaired spatial learning and memory. Remarkable, however, was the heightened amount of social interaction these mice had compared to the control mice. In human, FMT has shown initial positive effects on autism spectrum disorder patients (Kang et al., Reference Kang, Adams, Coleman, Pollard, Maldonado, McDonough-Means and Krajmalnik-Brown2019, Reference Kang, Adams, Gregory, Borody, Chittick, Fasano and Krajmalnik-Brown2017).

The most traditional and widely used probiotics (consisting of Bifidobacterium spp. and Lactobacillus spp.) are safe, yet they are not disease-specific. Although different Lactobacillus and Bifidobacterium strains were used in the probiotic studies mentioned above, most of the GM studies in the psychiatric disorders already showed heightened Lactobacillus and Bifidobacterium levels compared to their control groups. Rather, F. prausnitzii could be a promising target for therapeutic purpose in psychiatric patients (Verhoog et al., Reference Verhoog, Taneri, Díaz, Marques-Vidal, Troup, Bally and Muka2019). Research in rats showed preventive and therapeutic effects of F. prausnitzii on chronic unpredictable mild stress-induced depression-like and anxiety-like behaviour (Hao, Wang, Guo, & Liu, Reference Hao, Wang, Guo and Liu2019). Supplementation of prebiotics consisting of inulin-oligofructose or inulin-type fructans and fructo-oligosaccharides have been demonstrated to increase F. prausnitzii levels (Dewulf et al., Reference Dewulf, Cani, Claus, Fuentes, Puylaert, Neyrinck and Delzenne2013; Hustoft et al., Reference Hustoft, Hausken, Ystad, Valeur, Brokstad, Hatlebakk and Lied2017; Ramirez-Fariaz et al., Reference Ramirez-Fariaz, Slezak, Fuller, Duncan, Holtrop and Louis2009), although this is not a consistent finding (Majid, Cole, Emery, & Whelan, Reference Majid, Cole, Emery and Whelan2014).

Conclusions

The GM revolution has opened new frontiers for examining the relation between the brain and the GM in the context of understanding and treating/preventing psychiatric disorders. This paper provides a detailed overview of current findings regarding alterations of the GM in MDD, BD and SSD patients. All the reviewed studies reported alterations of the GM in the psychiatric disorders. Alterations may partly be caused by medication use, and other lifestyle factors like smoking, diet and alcohol use. Diversity metrics and microbial relative abundance reported to be anomalous across articles varied. The most consistent findings across studies were higher relative abundances of the genera Streptococcus, Lactobacillus and Eggerthella and lower relative abundance of the anti-inflammatory butyrate-producing genus Faecalibacterium in the psychiatric disorders. All three increased genera were reported to be associated with higher symptom severity. The similarities found between the disorders in the relative abundances and the associations of certain genera and symptoms suggest overlap in MDD, BD and SSD. So far, the results of probiotics trials have been highly discrepant, though few have shown promising results. Findings on prebiotics and FMT are too limited to draw definitive conclusions. There is a need of expanding our knowledge on bacterial species in bigger populations with psychiatric disorders, including the influence of medication and dietary habits. Information about differences in specific bacterial strains could lead to the use of disease-specific pro/prebiotics. In the end, study of the GM could lead to a new strategy of treating psychiatric disorders.

Financial support

The research was supported by grants from the Stanley Medical Research Institute (grant number 18T-004) and ZonMW (Netherlands Organisation for Health Research and Development; grant number 636320010).

Conflict of interest

None.

Footnotes

*

These authors contributed equally to the manuscript.

References

Abouesh, A., Stone, C., & Hobbs, W. R. (2002). Antimicrobial-induced mania (antibiomania): A review of spontaneous reports. Journal of Clinical Pharmacology, 22(1), 7181. https://doi.org/10.1097/00004714-200202000-00012.Google ScholarPubMed
Akkasheh, G., Kashani-Poor, Z., Tajabadi-Ebrahimi, M., Jafari, P., Akbari, H., Taghizadeh, M., … Esmaillzadeh, A. (2016). Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition, 32(3), 315320. https://doi.org/10.1016/j.nut.2015.09.003.CrossRefGoogle ScholarPubMed
Alvarez-Mon, M. A., Gómez, A. M., Orozco, A., Lahera, G., Sosa, M. D., Diaz, D., … Alvarez-Mon, M. (2019). Abnormal distribution and function of circulating monocytes and enhanced bacterial translocation in major depressive disorder. Frontiers in Psychiatry, 10(812), 112. https://doi.org/10.3389/fpsyt.2019.00812.CrossRefGoogle ScholarPubMed
Anttila, V., Bulik-Sullivan, B., Finucane, H. K., Walters, R. K., Bras, J., Duncan, L., … Neale, B. M. (2018). Analysis of shared heritability in common disorders of the brain. Science, 360(6395), 140. https://doi.org/10.1126/science.aap8757.Google ScholarPubMed
Bäckhed, F., Roswall, J., Peng, Y., Feng, Q., Jia, H., Kovatcheva-Datchary, P., … Jun, W. (2015). Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host and Microbe, 17(5), 690703. https://doi.org/10.1016/j.chom.2015.04.004.CrossRefGoogle ScholarPubMed
Barber, G. S., Sturgeon, C., Fasano, A., Gascella, N. G., Eaton, W. W., McMahon, R. P., & Kelly, D. L. (2019). Elevated zonulin, a measure of tight-junction permeability, may be implicated in schizophrenia. Schizophrenia Research, 211, 111112. https://doi.org/https://doi.org/10.1016/j.schres.2019.07.006.CrossRefGoogle Scholar
Bastiaanssen, T. F. S., Cussotto, S., Claesson, M. J., Clarke, G., Dinan, T. G., & Cryan, J. F. (2020). Gutted! Unraveling the role of the microbiome in major depressive disorder. Harvard Review of Psychiatry, 28(1), 2639. https://doi.org/10.1097/HRP.0000000000000243.CrossRefGoogle ScholarPubMed
Beaver, M. H., & Wostmann, B. S. (1962). Histamine and 5-hydroxytryptamine in the intestinal tract of germ-free animals, animals harbouring one microbial species and conventional animals. British Journal of Pharmacology and Chemotherapy, 19(3), 385393. https://doi.org/10.1111/j.1476-5381.1962.tb01443.x.CrossRefGoogle ScholarPubMed
Bennett, E. J., Tennant, C. C., Piesse, C., Badcock, C. A., & Kellow, J. E. (1998). Level of chronic life stress predicts clinical outcome in irritable bowel syndrome. Gut, 43(2), 256261. https://doi.org/10.1136/gut.43.2.256.CrossRefGoogle ScholarPubMed
Bischoff, S. C., Barbara, G., Buurman, W., Ockhuizen, T., Schulzke, J.-D., Serino, M., … Wells, J. M. (2014). Intestinal permeability – A new target for disease prevention and therapy. BMC Gastroenterology, 14(1), 189. https://doi.org/10.1186/s12876-014-0189-7.CrossRefGoogle ScholarPubMed
Browning, J. S., & Houseworth, J. H. (1953). Development of new symptoms following medical and surgical treatment for duodenal ulcer. Psychosomatic Medicine, 15(4), 328336. https://doi.org/10.1097/00006842-195307000-00006.CrossRefGoogle ScholarPubMed
Cannon, M., Jones, P. B., & Murray, R. M. (2002). Obstetric complications and schizophrenia: Historical and meta-analytic review. American Journal of Psychiatry, 159(7), 10801092. https://doi.org/https://doi-org/10.1176/appi.ajp.159.7.1080.CrossRefGoogle ScholarPubMed
Carlessi, A. S., Borba, L. A., Zugno, A. I., Quevedo, J., & Réus, G. Z. (2021). Gut microbiota–brain axis in depression: The role of neuroinflammation. European Journal of Neuroscience, 53, 222235. https://doi.org/10.1111/ejn.14631.CrossRefGoogle ScholarPubMed
Carlsson, A. H., Yakymenko, O., Olivier, I., Håkansson, F., Postma, E., Keita, Å. V., & Söderholm, J. D. (2013). Faecalibacterium prausnitzii supernatant improves intestinal barrier function in mice DSS colitis. Scandinavian Journal of Gastroenterology, 48(10), 11361144. https://doi.org/10.3109/00365521.2013.828773.CrossRefGoogle ScholarPubMed
Chudal, R., Sourander, A., Polo-Kantola, P., Hinkka-Yli-Salomäki, S., Lehti, V., Sucksdorff, D., … Brown, A. S. (2014). Perinatal factors and the risk of bipolar disorder in Finland. Journal of Affective Disorders, 155(1), 7580. https://doi.org/10.1016/j.jad.2013.10.026.CrossRefGoogle ScholarPubMed
Chung, Y. C. E., Chen, H. C., Chou, H. C. L., Chen, I. M., Lee, M. S., Chuang, L. C., … Kuo, P. H. (2019). Exploration of microbiota targets for major depressive disorder and mood related traits. Journal of Psychiatric Research, 111(17), 7482. https://doi.org/10.1016/j.jpsychires.2019.01.016.CrossRefGoogle ScholarPubMed
Cryan, J. F., O'riordan, K. J., Cowan, C. S. M., Sandhu, K. V., Bastiaanssen, T. F. S., Boehme, M., … Dinan, T. G. (2019). The microbiota–gut–brain axis. Physiological Reviews, 99(4), 18772013. https://doi.org/10.1152/physrev.00018.2018.CrossRefGoogle ScholarPubMed
Desbonnet, L., Garrett, L., Clarke, G., Bienenstock, J., & Dinan, T. G. (2009). The probiotic Bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. Journal of Psychiatric Research, 43(2), 164174. https://doi.org/10.1016/j.jpsychires.2008.03.009.CrossRefGoogle Scholar
Dewulf, E. M., Cani, P. D., Claus, S. P., Fuentes, S., Puylaert, P. G. B., Neyrinck, A. M., … Delzenne, N. M. (2013). Insight into the prebiotic concept: Lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut, 62(8), 11121121. https://doi.org/10.1136/gutjnl-2012-303304.CrossRefGoogle ScholarPubMed
Dickerson, F., Adamos, M., Katsafanas, E., Khushalani, S., Origoni, A., Savage, C., … Yolken, R. H. (2018). Adjunctive probiotic microorganisms to prevent rehospitalization in patients with acute mania: A randomized controlled trial. Bipolar Disorders, 20(7), 614621. https://doi.org/10.1111/bdi.12652.CrossRefGoogle ScholarPubMed
Dickerson, F., Severance, E., & Yolken, R. (2017). The microbiome, immunity, and schizophrenia and bipolar disorder. Brain, Behavior, and Immunity, 62, 4652. https://doi.org/10.1016/j.bbi.2016.12.010.CrossRefGoogle ScholarPubMed
Dickerson, F. B., Stallings, C., Origoni, A., Katsafanas, E., Savage, C. L. G., Schweinfurth, L. A. B., … Yolken, R. H. (2014). Effect of probiotic supplementation on schizophrenia symptoms and association with gastrointestinal functioning. The Primary Care Companion for CNS Disorders, 16(1). https://doi.org/10.4088/PCC.13m01579.Google ScholarPubMed
Dzikowski, M., Juchnowicz, D., Dzikowska, I., Rog, J., Próchnicki, M., Kozioł, M., … Karakula-Juchnowicz, H. (2020). The differences between gluten sensitivity, intestinal biomarkers and immune biomarkers in patients with first-episode and chronic schizophrenia. Journal of Clinical Medicine, 9(11), 113. https://doi.org/10.3390/jcm9113707.CrossRefGoogle ScholarPubMed
El-Ansary, A. K., Bacha, A. B., & Kotb, M. (2012). Etiology of autistic features: The persisting neurotoxic effects of propionic acid. Journal of Neuroinflammation, 9, 114. https://doi.org/10.1186/1742-2094-9-74.CrossRefGoogle ScholarPubMed
Faresjö, Å., Grodzinsky, E., Johansson, S., Wallander, M. A., Timpka, T., & Åkerlind, I. (2007). Psychosocial factors at work and in every day life are associated with irritable bowel syndrome. European Journal of Epidemiology, 22(7), 473480. https://doi.org/10.1007/s10654-007-9133-2.CrossRefGoogle ScholarPubMed
Ferreira-Halder, C. V., de Faria, A. V. S., & Andrade, S. S. (2017). Action and function of Faecalibacterium prausnitzii in health and disease. Best Practice and Research: Clinical Gastroenterology, 31(6), 643648. https://doi.org/10.1016/j.bpg.2017.09.011.CrossRefGoogle ScholarPubMed
Foster, J. A., Baker, G. B., & Dursun, S. M. (2021). The relationship between the gut microbiome–immune system–brain axis and major depressive disorder. Frontiers in Neurology, 12, 19. https://doi.org/10.3389/fneur.2021.721126.CrossRefGoogle ScholarPubMed
Fritsch, J., Garces, L., Quintero, M. A., Pignac-Kobinger, J., Santander, A. M., Fernández, I., … Abreu, M. T. (2021). Low-fat, high-fiber diet reduces markers of inflammation and dysbiosis and improves quality of life in patients with ulcerative colitis. Clinical Gastroenterology and Hepatology, 19(6), 11891199.e30. https://doi.org/10.1016/j.cgh.2020.05.026.CrossRefGoogle Scholar
Gacesa, R., Kurilshikov, A., Vich Vila, A., Sinha, T., Klaassen, M. A. Y., Bolte, L. A., … Weersma, R. K. (2022). Environmental factors shaping the gut microbiome in a Dutch population. Nature, 604, 733739. https://doi.org/10.1101/2020.11.27.401125.Google Scholar
Genedi, M., Janmaat, I. E., Haarman, B. C. M., & Sommer, I. E. C. (2019). Dysregulation of the gut–brain axis in schizophrenia and bipolar disorder: Probiotic supplementation as a supportive treatment in psychiatric disorders. Current Opinion in Psychiatry, 32(3), 185195. https://doi.org/10.1097/YCO.0000000000000499.CrossRefGoogle ScholarPubMed
Ghaderi, A., Banafshe, H. R., Mirhosseini, N., Moradi, M., Karimi, M. A., Mehrzad, F., … Asemi, Z. (2019). Clinical and metabolic response to vitamin D plus probiotic in schizophrenia patients. BMC Psychiatry, 19(1), 110. https://doi.org/10.1186/s12888-019-2059-x.CrossRefGoogle ScholarPubMed
Golofast, B., & Vales, K. (2020). The connection between microbiome and schizophrenia. Neuroscience and Biobehavioral Reviews, 108(August 2019), 712731. https://doi.org/10.1016/j.neubiorev.2019.12.011.CrossRefGoogle Scholar
Haarman, B. C. (Benno), Riemersma-Van der Lek, R. F., Burger, H., Drexhage, H. A., & Nolen, W. A. (2016). The dysregulated brain: Consequences of spatial and temporal brain complexity for bipolar disorder pathophysiology and diagnosis. Bipolar Disorders, 18(8), 696701. https://doi.org/10.1111/bdi.12454.CrossRefGoogle ScholarPubMed
Hao, Z., Wang, W., Guo, R., & Liu, H. (2019). Faecalibacterium prausnitzii (ATCC 27766) has preventive and therapeutic effects on chronic unpredictable mild stress-induced depression-like and anxiety-like behavior in rats. Psychoneuroendocrinology, 104, 132142. https://doi.org/10.1016/j.psyneuen.2019.02.025.CrossRefGoogle ScholarPubMed
He, X., Zhao, S., & Li, Y. (2021). Faecalibacterium prausnitzii: A next-generation probiotic in gut disease improvement. Canadian Journal of Infectious Diseases and Medical Microbiology, 2021, 110. https://doi.org/10.1155/2021/6666114.Google Scholar
He, Y., Kosciolek, T., Tang, J., Zhou, Y., Li, Z., Ma, X., … Chen, X. (2018). Gut microbiome and magnetic resonance spectroscopy study of subjects at ultra-high risk for psychosis may support the membrane hypothesis. European Psychiatry, 53(2018), 3745. https://doi.org/10.1016/j.eurpsy.2018.05.011.CrossRefGoogle ScholarPubMed
Hu, S., Li, A., Huang, T., Lai, J., Li, J., Sublette, M. E., … Xu, Y. (2019). Gut microbiota changes in patients with bipolar depression. Advanced Science, 6(14), 111. https://doi.org/10.1002/advs.201900752.CrossRefGoogle ScholarPubMed
Huang, Y., Shi, X., Li, Z., Shen, Y., Shi, X., Wang, L., … Liang, Y. (2018). Possible association of firmicutes in the gut microbiota of patients with major depressive disorder. Neuropsychiatric Disease and Treatment, 14, 33293337. https://doi.org/10.2147/NDT.S188340.CrossRefGoogle ScholarPubMed
Hustoft, T. N., Hausken, T., Ystad, S. O., Valeur, J., Brokstad, K., Hatlebakk, J. G., & Lied, G. A. (2017). Effects of varying dietary content of fermentable short-chain carbohydrates on symptoms, fecal microenvironment, and cytokine profiles in patients with irritable bowel syndrome. Neurogastroenterology and Motility, 29(4), 19. https://doi.org/10.1111/nmo.12969.CrossRefGoogle ScholarPubMed
James, S. L., Abate, D., Abate, K. H., Abay, S. M., Abbafati, C., Abbasi, N., … Murray, C. J. L. (2018). Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: A systematic analysis for the global burden of disease study 2017. The Lancet, 392(10159), 17891858. https://doi.org/10.1016/S0140-6736(18)32279-7.CrossRefGoogle Scholar
Jamilian, H., & Ghaderi, A. (2021). The effects of probiotic and selenium co-supplementation on clinical and metabolic scales in chronic schizophrenia: A randomized, double-blind, placebo-controlled trial. Biological Trace Element Research, 199(12), 44304438. https://doi.org/10.1007/s12011-020-02572-3.CrossRefGoogle ScholarPubMed
Jiang, H., Ling, Z., Zhang, Y., Mao, H., Ma, Z., Yin, Y., … Ruan, B. (2015). Altered fecal microbiota composition in patients with major depressive disorder. Brain, Behavior, and Immunity, 48, 186194. https://doi.org/10.1016/j.bbi.2015.03.016.CrossRefGoogle ScholarPubMed
Kang, D. W., Adams, J. B., Coleman, D. M., Pollard, E. L., Maldonado, J., McDonough-Means, S., … Krajmalnik-Brown, R. (2019). Long-term benefit of microbiota transfer therapy on autism symptoms and gut microbiota. Scientific Reports, 9(1), 19. https://doi.org/10.1038/s41598-019-42183-0.Google ScholarPubMed
Kang, D. W., Adams, J. B., Gregory, A. C., Borody, T., Chittick, L., Fasano, A., … Krajmalnik-Brown, R. (2017). Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: An open-label study. Microbiome, 5(1), 116. https://doi.org/10.1186/s40168-016-0225-7.CrossRefGoogle Scholar
Kang, D. W., Park, J. G., Ilhan, Z. E., Wallstrom, G., LaBaer, J., Adams, J. B., … Krajmalnik-Brown, R. (2013). Reduced incidence of prevotella and other fermenters in intestinal microflora of autistic children. PLoS ONE, 8(7), 113. https://doi.org/10.1371/journal.pone.0068322.Google ScholarPubMed
Kao, A. C. C., Safarikova, J., Marquardt, T., Mullins, B., Lennox, B. R., & Burnet, P. W. J. (2019). Pro-cognitive effect of a prebiotic in psychosis: A double blind placebo controlled cross-over study. Schizophrenia Research, 208, 460461. https://doi.org/10.1016/j.schres.2019.03.003.CrossRefGoogle ScholarPubMed
Kazemi, A., Noorbala, A. A., Azam, K., Eskandari, M. H., & Djafarian, K. (2019). Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clinical Nutrition, 38(2), 522528. https://doi.org/10.1016/j.clnu.2018.04.010.CrossRefGoogle ScholarPubMed
Keefe, R. S. E., Goldberg, T. E., Harvey, P. D., Gold, J. M., Poe, M. P., & Coughenour, L. (2004). The brief assessment of cognition in schizophrenia: Reliability, sensitivity, and comparison with a standard neurocognitive battery. Schizophrenia Research, 68, 283297. https://doi.org/10.1016/j.schres.2003.09.011.CrossRefGoogle ScholarPubMed
Keita, Å. V., Söderholm, J. D., & Ericson, A. C. (2010). Stress-induced barrier disruption of rat follicle-associated epithelium involves corticotropin-releasing hormone, acetylcholine, substance P, and mast cells. Neurogastroenterology and Motility, 22(7), 770778. https://doi.org/10.1111/j.1365-2982.2010.01471.x.CrossRefGoogle ScholarPubMed
Kelly, J. R., Borre, Y., O’ Brien, C., Patterson, E., El Aidy, S., Deane, J., … Dinan, T. G. (2016). Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. Journal of Psychiatric Research, 82, 109118. https://doi.org/10.1016/j.jpsychires.2016.07.019.CrossRefGoogle ScholarPubMed
Kiecolt-Glaser, J. K., Wilson, S. J., Bailey, M. L., Andridge, R., Peng, J., Jaremka, L. M., … Belury, M. A. (2018). Marital distress, depression, and a leaky gut: Translocation of bacterial endotoxin as a pathway to inflammation. Psychoneuroendocrinology, 98, 5260. https://doi.org/10.1016/j.psyneuen.2018.08.007.CrossRefGoogle Scholar
Kiliaan, A. J., Saunders, P. R., Bijlsma, P. B., Cecilia Berin, M., Taminiau, J. A., Groot, J. A., & Perdue, M. H. (1998). Stress stimulates transepithelial macromolecular uptake in rat jejunum. American Journal of Physiology – Gastrointestinal and Liver Physiology, 275(5), 10371044. https://doi.org/10.1152/ajpgi.1998.275.5.g1037.CrossRefGoogle ScholarPubMed
Kitchens, R. L., & Thompson, P. A. (2005). Modulatory effects of sCD14 and LBP on LPS–host cell interactions. Journal of Endotoxin Research, 11(4), 225229. https://doi.org/10.1179/096805105X46565.CrossRefGoogle ScholarPubMed
Kılıç, F., Işık, Ü., Demirdaş, A., Doğuç, D. K., & Bozkurt, M. (2020). Serum zonulin and claudin-5 levels in patients with bipolar disorder. Journal of Affective Disorders, 266, 3742. https://doi.org/10.1016/j.jad.2020.01.117.CrossRefGoogle ScholarPubMed
Köhler, O., Petersen, L., Mors, O., Mortensen, P. B., Yolken, R. H., Gasse, C., & Benros, M. E. (2017). Infections and exposure to anti-infective agents and the risk of severe mental disorders: A nationwide study. Acta Psychiatrica Scandinavica, 135(2), 97105. https://doi.org/10.1111/acps.12671.CrossRefGoogle ScholarPubMed
Korpela, K., Helve, O., Kolho, K. L., Saisto, T., Skogberg, K., Dikareva, E., … de Vos, W. M. (2020). Maternal fecal microbiota transplantation in cesarean-born infants rapidly restores normal gut microbial development: A proof-of-concept study. Cell, 183(2), 324334. https://doi.org/10.1016/j.cell.2020.08.047.CrossRefGoogle ScholarPubMed
Lai, J., Gao, X., & Zhang, D. (2020). Gut microbial clues to bipolar disorder: State-of-the-art review of current findings and future directions. Clinical and Translational Medicine, 10, 114. https://doi.org/10.1002/ctm2.146.CrossRefGoogle ScholarPubMed
Lai, W. T., Deng, W. F., Xu, S. X., Zhao, J., Xu, D., Liu, Y. H., … Rong, H. (2021). Shotgun metagenomics reveals both taxonomic and tryptophan pathway differences of gut microbiota in major depressive disorder patients. Psychological Medicine, 51(1), 90101. https://doi.org/10.1017/S0033291719003027.CrossRefGoogle ScholarPubMed
Lambrichts, S., Van Oudenhove, L., & Sienaert, P. (2017). Antibiotics and mania: A systematic review. Journal of Affective Disorders, 219, 149156. https://doi.org/10.1016/j.jad.2017.05.029.CrossRefGoogle ScholarPubMed
Laval, L., Martin, R., Natividad, J. N., Chain, F, Miquel, F. C., Desclée de Maredsous, S., C., … Langella, P. (2015). Lactobacillus rhamnosus CNCM I-3690 and the commensal bacterium faecalibacterium prausnitzii A2-165 exhibit similar protective effects to induced barrier hyper-permeability in mice. Gut Microbes, 6(1), 19. https://doi.org/10.4161/19490976.2014.990784.CrossRefGoogle ScholarPubMed
Lê Cook, B., Wayne, G. F., Kafali, E. N., Liu, Z., Shu, C., & Flores, M. (2014). Trends in smoking among adults with mental illness and association between mental health treatment and smoking cessation. JAMA – Journal of the American Medical Association, 311(2), 172182. https://doi.org/10.1001/jama.2013.284985.CrossRefGoogle Scholar
Li, S., Zhuo, M., Huang, X., Huang, Y., Zhou, J., Xiong, D., … Wu, K. (2020). Altered gut microbiota associated with symptom severity in schizophrenia. PeerJ, 8, 122. https://doi.org/10.7717/peerj.9574.Google Scholar
Liang, X., Ye, J., Wen, Y., Li, P., Cheng, B., Cheng, S., … Zhang, F. (2021). Long-term antibiotic use during early life and risks to mental traits: An observational study and gene–environment-wide interaction study in UK Biobank cohort. Neuropsychopharmacology, 46(6), 10861092. https://doi.org/10.1038/s41386-020-00798-2.CrossRefGoogle ScholarPubMed
Lim, P. S., Chang, Y. K., & Wu, T. K. (2019). Serum lipopolysaccharide-binding protein is associated with chronic inflammation and metabolic syndrome in hemodialysis patients. Blood Purification, 47, 2836. https://doi.org/10.1159/000492778.CrossRefGoogle ScholarPubMed
Liu, R. T., Rowan-Nash, A. D., Sheehan, A. E., Walsh, R. F. L., Sanzari, C. M., Korry, B. J., & Belenky, P. (2020). Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults. Brain, Behavior, and Immunity, 88, 308324. https://doi.org/10.1016/j.bbi.2020.03.026.CrossRefGoogle Scholar
Liu, Y., Zhang, L., Wang, X., Wang, Z., Zhang, J., Jiang, R., … Duan, L. (2016). Similar fecal microbiota signatures in patients with diarrhea-predominant irritable bowel syndrome and patients with depression. Clinical Gastroenterology and Hepatology, 14(11), 16021611. https://doi.org/10.1016/j.cgh.2016.05.033.CrossRefGoogle ScholarPubMed
Lurie, I., Yang, Y.-X., Hayne, K., Mamtani, R., & Boursi, B. (2015). Antibiotic exposure and the risk for depression, anxiety, or psychosis: A neste case-control study. The Journal of Clinical Psychiatry, 76(11), 15221528. https://doi.org/10.4088/JCP.15m09961.CrossRefGoogle Scholar
Ma, X., Asif, H., Dai, L., He, Y., Zheng, W., Wang, D., … Chen, X. (2020). Alteration of the gut microbiome in first-episode drug-naïve and chronic medicated schizophrenia correlate with regional brain volumes. Journal of Psychiatric Research, 123, 136144. https://doi.org/10.1016/j.jpsychires.2020.02.005.CrossRefGoogle ScholarPubMed
Maes, M., Kanchanatawan, B., Sirivichayakul, S., & Carvalho, A. F. (2019a). In schizophrenia, increased plasma IgM/IgA responses to gut commensal bacteria are associated with negative symptoms, neurocognitive impairments, and the deficit phenotype. Neurotoxicity Research, 35(3), 684698. https://doi.org/10.1007/s12640-018-9987-y.CrossRefGoogle Scholar
Maes, M., Kubera, M., Leunis, J. C., Berk, M., Geffard, M., & Bosmans, E. (2013). In depression, bacterial translocation may drive inflammatory responses, oxidative and nitrosative stress (O&NS), and autoimmune responses directed against O&NS-damaged neoepitopes. Acta Psychiatrica Scandinavica, 127(5), 344354. https://doi.org/10.1111/j.1600-0447.2012.01908.x.CrossRefGoogle ScholarPubMed
Maes, M., Sirivichayakul, S., Kanchanatawan, B., & Vodjani, A. (2019b). Upregulation of the intestinal paracellular pathway with breakdown of tight and adherens junctions in deficit schizophrenia. Molecular Neurobiology, 56(10), 70567073. https://doi.org/10.1007/s12035-019-1578-2.CrossRefGoogle Scholar
Majid, H. A., Cole, J., Emery, P. W., & Whelan, K. (2014). Additional oligofructose/inulin does not increase faecal bifidobacteria in critically ill patients receiving enteral nutrition: A randomised controlled trial. Clinical Nutrition, 33(6), 966972. https://doi.org/10.1016/j.clnu.2013.11.008.CrossRefGoogle Scholar
Martín, R., Miquel, S., Chain, F., Natividad, J. M., Jury, J., Lu, J., … Bermúdez-Humarán, L. G. (2015). Faecalibacterium prausnitzii prevents physiological damages in a chronic low-grade inflammation murine model. BMC Microbiology, 15(1), 112. https://doi.org/10.1186/s12866-015-0400-1.CrossRefGoogle Scholar
Marx, W., McGuinness, A. J., Rocks, T., Ruusunen, A., Cleminson, J., Walker, A. J., … Fernandes, B. S. (2020). The kynurenine pathway in major depressive disorder, bipolar disorder, and schizophrenia: A meta-analysis of 101 studies. Molecular Psychiatry, 26(8), 41584178. https://doi.org/10.1038/s41380-020-00951-9.CrossRefGoogle ScholarPubMed
Massier, L., Chakaroun, R., Kovacs, P., & Heiker, J. T. (2021). Blurring the picture in leaky gut research: How shortcomings of zonulin as a biomarker mislead the field of intestinal permeability. Gut, 70(9), 18011802. https://doi.org/10.1136/gutjnl-2020-323026.CrossRefGoogle ScholarPubMed
Mitchell, C. M., Mazzoni, C., Hogstrom, L., Bryant, A., Bergerat, A., Cher, A., … Yassour, M. (2020). Delivery mode affects stability of early infant gut microbiota. Cell Reports Medicine, 1(9), 19. https://doi.org/10.1016/j.xcrm.2020.100156.CrossRefGoogle ScholarPubMed
Morch, R. H., Dieset, I., Færden, A., Reponen, E. J., Hope, S., Hoseth, E. Z., … Andreassen, O. A. (2019). Inflammatory markers are altered in severe mental disorders independent of comorbid cardiometabolic disease risk factors. Psychological Medicine, 49(10), 17494757. https://doi.org/10.1017/S0033291719000291.CrossRefGoogle ScholarPubMed
Moretti, M., Valvassori, S. S., Varela, R. B., Ferreira, C. L., Rochi, N., Benedet, J., … Quevedo, J. (2011). Behavioral and neurochemical effects of sodium butyrate in an animal model of mania. Behavioural Pharmacology, 22(8), 766772. https://doi.org/10.1097/FBP.0b013e32834d0f1b.CrossRefGoogle Scholar
Nankova, B. B., Agarwal, R., MacFabe, D. F., & La Gamma, E. F. (2014). Enteric bacterial metabolites propionic and butyric acid modulate gene expression, including CREB-dependent catecholaminergic neurotransmission, in PC12 cells – Possible relevance to autism spectrum disorders. PLoS ONE, 9(8), 116. https://doi.org/10.1371/journal.pone.0103740.CrossRefGoogle ScholarPubMed
Nguyen, T. T., Hathaway, H., Kosciolek, T., Knight, R., & Jeste, D. V. (2021). Gut microbiome in serious mental illnesses: A systematic review and critical evaluation. Schizophrenia Research, 234, 24–40. https://doi.org/10.1016/j.schres.2019.08.026.CrossRefGoogle ScholarPubMed
Nguyen, T. T., Kosciolek, T., Maldonado, Y., Daly, R. E., Martin, A. S., McDonald, D., … Jeste, D. V. (2018). Differences in gut microbiome composition between persons with chronic schizophrenia and healthy comparison subjects. Schizophrenia Research, 204, 2329. https://doi.org/10.1016/j.schres.2018.09.014.CrossRefGoogle ScholarPubMed
Nicholl, B. I., Halder, S. L., Macfarlane, G. J., Thompson, D. G., O'Brien, S., Musleh, M., & McBeth, J. (2008). Psychosocial risk markers for new onset irritable bowel syndrome – Results of a large prospective population-based study. Pain, 137(1), 147155. https://doi.org/10.1016/j.pain.2007.08.029.CrossRefGoogle ScholarPubMed
Nikolova, V. L., Hall, M. R. B., Hall, L. J., Cleare, A. J., Stone, J. M., & Young, A. H. (2021). Perturbations in gut microbiota composition in psychiatric disorders: A review and meta-analysis. JAMA Psychiatry, 78(12), 13431354. https://doi.org/10.1001/jamapsychiatry.2021.2573.CrossRefGoogle ScholarPubMed
Ohlsson, L., Gustafsson, A., Lavant, E., Suneson, K., Brundin, L., Westrin, , … Lindqvist, D. (2019). Leaky gut biomarkers in depression and suicidal behavior. Acta Psychiatrica Scandinavica, 139(2), 185193. https://doi.org/10.1111/acps.12978.CrossRefGoogle ScholarPubMed
O'Neill, S. M., Curran, E. A., Dalman, C., Kenny, L. C., Kearney, P. M., Clarke, G., … Khashan, A. S. (2016). Birth by caesarean section and the risk of adult psychosis: A population-based cohort study. Schizophrenia Bulletin, 42(3), 633641. https://doi.org/10.1093/schbul/sbv152.CrossRefGoogle ScholarPubMed
Painold, A., Mörkl, S., Kashofer, K., Halwachs, B., Dalkner, N., Bengesser, S., … Reininghaus, E. Z. (2018). A step ahead: Exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode. Bipolar Disorders, 21, 4049. https://doi.org/10.1111/bdi.12682.CrossRefGoogle ScholarPubMed
Purton, T., Staskova, L., Lane, M. M., Dawson, S. L., West, M., Firth, J., … Marx, W. (2021). Prebiotic and probiotic supplementation and the tryptophan–kynurenine pathway: A systematic review and meta analysis. Neuroscience and Biobehavioral Reviews, 123, 113. https://doi.org/10.1016/j.neubiorev.2020.12.026.CrossRefGoogle ScholarPubMed
Ramirez-Fariaz, C., Slezak, K., Fuller, Z., Duncan, A., Holtrop, G., & Louis, P. (2009). Effect of inulin on the human gut microbiota: Stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii. British Journal of Nutrition, 101(4), 533540. https://doi.org/10.1017/S0007114508019880.Google Scholar
Reininghaus, E. Z., Wetzlmair, L. C., Fellendorf, F. T., Platzer, M., Queissner, R., Birner, A., … Dalkner, N. (2020). The impact of probiotic supplements on cognitive parameters in euthymic individuals with bipolar disorder: A pilot study. Neuropsychobiology, 79(1), 6370. https://doi.org/10.1159/000492537.CrossRefGoogle Scholar
Rekdal, V. M., Bess, E. N., Bisanz, J. E., Turnbaugh, P. J., & Balskus, E. P. (2019). Discovery and inhibition of an interspecies gut bacterial pathway for levodopa metabolism. Science, 364(6445), 18. https://doi.org/10.1126/science.aau6323.Google Scholar
Resende, W. R., Valvassori, S. S., Réus, G. Z., Varela, R. B., Arent, C. O., Ribeiro, K. F., … Quevedoa, J. (2013). Effects of sodium butyrate in animal models of mania and depression: Implications as a new mood stabilizer. Behavioural Pharmacology, 24(7), 569579. https://doi.org/10.1097/FBP.0b013e32836546fc.CrossRefGoogle ScholarPubMed
Rocha-Ramírez, L. M., Pérez-Solano, R. A., Castañón-Alonso, S. L., Moreno Guerrero, S. S., Ramírez Pacheco, A., García Garibay, M., … Eslava, C. (2017). Probiotic lactobacillus strains stimulate the inflammatory response and activate human macrophages. Journal of Immunology Research, 2017, 114. https://doi.org/10.1155/2017/4607491.CrossRefGoogle ScholarPubMed
Rong, H., Xie, X. H., Zhao, J., Lai, W. T., Wang, M. B., Xu, D., … Liu, T. B. (2019). Similarly in depression, nuances of gut microbiota: Evidences from a shotgun metagenomics sequencing study on major depressive disorder versus bipolar disorder with current major depressive episode patients. Journal of Psychiatric Research, 113, 9099. https://doi.org/10.1016/j.jpsychires.2019.03.017.CrossRefGoogle Scholar
Sanada, K., Nakajima, S., Kurokawa, S., Barceló-Soler, A., Ikuse, D., Hirata, A., … Kishimoto, T. (2020). Gut microbiota and major depressive disorder: A systematic review and meta-analysis. Journal of Affective Disorders, 266, 113. https://doi.org/10.1016/j.jad.2020.01.102.CrossRefGoogle ScholarPubMed
Schwarz, E., Maukonen, J., Hyytiäinen, T., Kieseppä, T., Orešič, M., Sabunciyan, S., … Suvisaari, J. (2018). Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophrenia Research, 192, 398403. https://doi.org/10.1016/j.schres.2017.04.017.CrossRefGoogle ScholarPubMed
Severance, E. G., Dickerson, F., & Yolken, R. H. (2020). Complex gastrointestinal and endocrine sources of inflammation in schizophrenia. Frontiers in Psychiatry, 11, 19. https://doi.org/10.3389/fpsyt.2020.00549.CrossRefGoogle Scholar
Severance, E. G., Gressitt, K. L., Stallings, C. R., Origoni, A. E., Khushalani, S., Leweke, F. M., … Yolken, R. H. (2013). Discordant patterns of bacterial translocation markers and implications for innate immune imbalances in schizophrenia. Schizophrenia Research, 148(1–3), 130137. https://doi.org/10.1016/j.schres.2013.05.018.CrossRefGoogle Scholar
Sgritta, M., Dooling, S. W., Buffington, S. A., Momin, E. N., Francis, M. B., Britton, R. A., & Costa-Mattioli, M. (2019). Mechanisms underlying microbial-bediated changes in social behavior in mouse models of autism spectrum disorder. Neuron, 101(2), 246259. https://doi.org/10.1016/j.neuron.2018.11.018.CrossRefGoogle ScholarPubMed
Shahrbabaki, M. E., Sabouri, S., Sabahi, A., Barfeh, D., Divsalar, P., Esmailzadeh, M., & Ahmadi, A. (2020). The efficacy of probiotics for treatment of bipolar disorder-type 1: A randomized, double-blind, placebo controlled trial. Iranian Journal of Psychiatry, 15(1), 1016. https://doi.org/10.18502/ijps.v15i1.2435.Google Scholar
Shen, Y., Xu, J., Li, Z., Huang, Y., Yuan, Y., Wang, J., … Liang, Y. (2018). Analysis of gut microbiota diversity and auxiliary diagnosis as a biomarker in patients with schizophrenia: A cross-sectional study. Schizophrenia Research, 197, 470477. https://doi.org/10.1016/j.schres.2018.01.002.CrossRefGoogle ScholarPubMed
Sherwin, E., Sandhu, K. V., Dinan, T. G., & Cryan, J. F. (2016). May the force be with you: The light and dark sides of the microbiota–gut–brain axis in neuropsychiatry. CNS Drugs, 30(11), 10191041. https://doi.org/10.1007/s40263-016-0370-3.CrossRefGoogle ScholarPubMed
Simeonova, D., Stoyanov, D., Leunis, J. -C, Carvalho, A. F., Kubera, M., Murdjeva, M., & Maes, M. (2020). Increased serum immunoglobulin responses to gut commensal Gram-negative bacteria in unipolar major depression and bipolar disorder type 1, especially when melancholia is present. Neurotoxicity Research, 37(2), 338348. https://doi.org/10.1007/s12640-019-00126-7.CrossRefGoogle ScholarPubMed
Simpson, C. A., Diaz-Arteche, C., Eliby, D., Schwartz, O. S., Simmons, J. G., & Cowan, C. S. M. (2021). The gut microbiota in anxiety and depression – A systematic review. Clinical Psychology Review, 83(101943), 118. https://doi.org/10.1016/j.cpr.2020.101943.CrossRefGoogle ScholarPubMed
Skonieczna-żydecka, K., Grochans, E., Maciejewska, D., Szkup, M., Schneider-Matyka, D., Jurczak, A., … Stachowska, E. (2018). Faecal short chain fatty acids profile is changed in Polish depressive women. Nutrients, 10(12), 114. https://doi.org/10.3390/nu10121939.CrossRefGoogle ScholarPubMed
Söderholm, J. D., Yang, P. C., Ceponis, P., Vohra, A., Riddell, R., Sherman, P. M., & Perdue, M. H. (2002). Chronic stress induces mast cell-dependent bacterial adherence and initiates mucosal inflammation in rat intestine. Gastroenterology, 123(4), 10991108. https://doi.org/10.1053/gast.2002.36019.CrossRefGoogle ScholarPubMed
Stevens, B. R., Goel, R., Seungbum, K., Richards, E. M., Holbert, R. C., Pepine, C. J., … Genomics, F. (2018). Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut, 67(8), 15551557. https://doi.org/10.1136/gutjnl-2017-314759.Increased.CrossRefGoogle ScholarPubMed
Svensson, E., Horváth-Puhó, E., Thomsen, R. W., Djurhuus, J. C., Pedersen, L., Borghammer, P., & Sørensen, H. T. (2015). Vagotomy and subsequent risk of Parkinson's disease. Annals of Neurology, 78(4), 522529. https://doi.org/10.1002/ana.24448.CrossRefGoogle ScholarPubMed
Szczesniak, O., Hestad, K. A., Hanssen, J. F., & Rudi, K. (2016). Isovaleric acid in stool correlates with human depression. Nutritional Neuroscience, 19(7), 279283. https://doi.org/10.1179/1476830515Y.0000000007.CrossRefGoogle ScholarPubMed
Szeligowski, T., Yun, A. L., Lennox, B. R., & Burnet, P. W. J. (2020). The gut microbiome and schizophrenia: The current state of the field and clinical applications. Frontiers in Psychiatry, 11(156), 110. https://doi.org/10.3389/fpsyt.2020.00156.CrossRefGoogle ScholarPubMed
Tanaka, T., Matsuda, T., Hayes, L. N., Yang, S., Rodriguez, K., Severance, E. G., … Eaton, W. W. (2017). Infection and inflammation in schizophrenia and bipolar disorder. Neuroscience Research, 115, 5963. https://doi.org/10.1016/j.neures.2016.11.002.CrossRefGoogle ScholarPubMed
Thaiss, C. A., Zmora, N., Levy, M., & Elinav, E. (2016). The microbiome and innate immunity. Nature, 535(7610), 6574. https://doi.org/10.1038/nature18847.CrossRefGoogle ScholarPubMed
Valladares, R., Bojilova, L., Potts, A. H., Cameron, E., Gardner, C., Lorca, G., & Gonzalez, C. F. (2013). Lactobacillus johnsonii inhibits indoleamine 2,3-dioxygenase and alters tryptophan metabolite levels in BioBreeding rats. FASEB Journal, 27(4), 17111720. https://doi.org/10.1096/fj.12-223339.CrossRefGoogle ScholarPubMed
Valvassori, S., Resende, W., Budni, J., Dal-Pont, G., Bavaresco, D., Reus, G., … Quevedo, J. (2015). Sodium butyrate, a histone deacetylase inhibitor, reverses behavioral and mitochondrial alterations in animal models of depression induced by early- or late-life stress. Current Neurovascular Research, 12(4), 312320. https://doi.org/10.2174/1567202612666150728121121.CrossRefGoogle ScholarPubMed
Vanuytsel, T., Van Wanrooy, S., Vanheel, H., Vanormelingen, C., Verschueren, S., Houben, E., … Tack, J. (2014). Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut, 63(8), 12931299. https://doi.org/10.1136/gutjnl-2013-305690.CrossRefGoogle ScholarPubMed
Verhoog, S., Taneri, P. E., Díaz, Z. M. R., Marques-Vidal, P., Troup, J. P., Bally, L., … Muka, T. (2019). Dietary factors and modulation of bacteria strains of akkermansia muciniphila and faecalibacterium prausnitzii: A systematic review. Nutrients, 11(7), 120. https://doi.org/10.3390/nu11071565.CrossRefGoogle ScholarPubMed
Vicario, M., Guilarte, M., Alonso, C., Yang, P., Martínez, C., Ramos, L., … Santos, J. (2010). Chronological assessment of mast cell-mediated gut dysfunction and mucosal inflammation in a rat model of chronic psychosocial stress. Brain, Behavior, and Immunity, 24(7), 11661175. https://doi.org/10.1016/j.bbi.2010.06.002.CrossRefGoogle Scholar
Wang, W., Chen, L., Zhou, R., Wang, X., Song, L., Huang, S., … Xia, B. (2014). Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. Journal of Clinical Microbiology, 52(2), 398406. https://doi.org/10.1128/JCM.01500-13.CrossRefGoogle ScholarPubMed
Weber, N. S., Gressitt, K. L., Cowan, D. N., Niebuhr, D. W., Yolken, R. H., & Severance, E. G. (2018). Monocyte activation detected prior to a diagnosis of schizophrenia in the US Military New Onset Psychosis Project (MNOPP). Schizophrenia Research, 197, 465469. https://doi.org/10.1016/j.schres.2017.12.016.CrossRefGoogle Scholar
Whitlock, F. A. (1961). Some psychiatric consequences of gastrectomy. British Medical Journal, 1(5242), 15601564. https://doi.org/10.1136/bmj.1.5242.1828.CrossRefGoogle Scholar
Whittaker, R. H. (1972). Evolution and measurement of species diversity. Taxon, 21, 213251.CrossRefGoogle Scholar
World Health Organization (2006). The world health report : 2006 : working together for health. World Health Organization. https://apps.who.int/iris/handle/10665/43432.Google Scholar
Xu, R., Wu, B., Liang, J., He, F., Gu, W., Li, K., … Wang, M. (2020). Altered gut microbiota and mucosal immunity in patients with schizophrenia. Brain, Behavior, and Immunity, 85, 120127. https://doi.org/10.1016/j.bbi.2019.06.039.CrossRefGoogle ScholarPubMed
Yano, J. M., Yu, K., Donaldson, G. P., Shastri, G. G., Ann, P., Ma, L., … Hsiao, E. Y. (2015). Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell, 161(2), 264276. https://doi.org/10.1016/j.cell.2015.02.047.CrossRefGoogle ScholarPubMed
Yuan, X., Zhang, P., Wang, Y., Liu, Y., Li, X., Kumar, B. U., … Song, X. (2018). Changes in metabolism and microbiota after 24-week risperidone treatment in drug naïve, normal weight patients with first episode schizophrenia. Schizophrenia Research, 201, 299306. https://doi.org/10.1016/j.schres.2018.05.017.CrossRefGoogle ScholarPubMed
Zheng, P., Yang, J., Li, Y., Wu, J., Liang, W., Yin, B., … Wang, G. (2020). Gut microbial signatures can discriminate unipolar from bipolar depression. Advanced Science, 7(7), 111. https://doi.org/10.1002/advs.201902862.CrossRefGoogle ScholarPubMed
Zheng, P., Zeng, B., Liu, M., Chen, J., Pan, J., Han, Y., … Xie, P. (2019). The gut microbiome from patients with schizophrenia modulates the glutamate-glutamine-GABA cycle and schizophrenia-relevant behaviors in mice. Science Advances, 5(2), 111. https://doi.org/10.1126/sciadv.aau8317.CrossRefGoogle ScholarPubMed
Zheng, P., Zeng, B., Zhou, C., Liu, M., Fang, Z., Xu, X., … Fan, S. (2016). Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism. Molecular Psychiatry, 21, 786796. https://doi.org/10.1038/mp.2016.44.CrossRefGoogle ScholarPubMed
Zhu, F., Guo, R., Wang, W., Ju, Y., Wang, Q., Ma, Q., … Ma, X. (2020a). Transplantation of microbiota from drug-free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Molecular Psychiatry, 25(11), 29052918. https://doi.org/10.1038/s41380-019-0475-4.CrossRefGoogle Scholar
Zhu, F., Ju, Y., Wang, W., Wang, Q., Guo, R., Ma, Q., … Ma, X. (2020b). Metagenome-wide association of gut microbiome features for schizophrenia. Nature Communications, 11(1), 110. https://doi.org/10.1038/s41467-020-15457-9.Google Scholar
Figure 0

Fig. 1. (1) Environmental factors known to impinge on the human GM. (2) GM dysbiosis impairs intestinal permeability. (3) Increased intestinal permeability causes translocation of luminal components and reactivity of the intestinal immune system. (4) Bacterial translocation activates the gut–brain axis. (5) The gut and the brain communicate bidirectionally via several routes, including the vagal nerve, the HPA axis, immune mediators such as cytokines, and the production of bacterial metabolites, such as SCFAs. (6) The environmental factors, GM dysbiosis and increased permeability separately and in concert could contribute the development of psychiatric disorders. Created with BioRender.com.

Figure 1

Table 1. Main findings of studies assessing gut permeability in MDD, BD and SSD

Figure 2

Table 2. Studies of the GM in major depressive disorder

Figure 3

Table 3. Studies of the GM in individuals with BD

Figure 4

Table 4. Studies of the GM in schizophrenia-spectrum disorder

Figure 5

Fig. 2. Taxonomic differences in neuropsychiatric disorders (at the phylum, family and genus levels), whereby ‘↑’ = higher relative abundance in the neuropsychiatric disorder group, ‘↓’ = lower relative abundance in the neuropsychiatric group and ‘I’ = no differences in abundance. The letters below the arrows refer to the studies the information was retrieved from and also can be connected to the letters in the tables. Studies: AZheng et al. (2016); BJiang et al. (2015); CKelly et al. (2016); DChung et al. (2019); EHuang et al. (2018); FLai et al. (2021); GLiu et al. (2016); HZheng et al. (2020); IRong et al. (2019); JLiu et al. (2020); KZheng et al. (2019); LXu et al. (2020); MNguyen et al. (2018); NShen et al. (2018); OHe et al. (2018); PLi et al. (2020); QRong et al. (2019); RPainold et al. (2018); SSchwarz et al. (2018); THu et al. (2019); UZheng et al. (2020); VPainold et al. (2018); WRong et al. (2019).