The gut microbiome is a complex and dynamic community of commensal, symbiotic, and pathogenic microorganisms that exist in a bidirectional relationship with the host. Bacterial functions in the gut play a critical role in healthy host functioning, and its disruption can contribute to many medical conditions. The relationship between gut microbiota and the brain has gained attention in mental health due to the mounting evidence supporting the association of gut bacteria with mood and behavior. Patients with bipolar disorder exhibit an increased frequency of gastrointestinal illnesses such as inflammatory bowel disease, which mechanistically has been linked to microbial community function. While the heterogeneity in microbial communities between individuals might be associated with disease risk, it may also moderate the efficacy or adverse effects associated with the use of medication. The following review highlights published evidence linking the function of gut microbiota both to bipolar disorder risk and to the effect of medications that influence microbiota, inflammation, and mood symptoms.

Bipolar disorder (BD) is a severe psychological illness defined by shifts in mood and energy [1]. Without regard to nationality, ethnicity, or socioeconomic status, BD affects 1% of the world’s population, is associated with higher rates of substance abuse and cardiovascular disease, and represents a great source of disability [2-4]. The etiology of BD is not fully understood. Genome-wide association studies show that schizophrenia and BD genetically overlap and many identified risk genes are related to immune response and inflammation [5-7]. While genetic factors are recognized in the pathogenesis of BD, these identified risk genes account for only a small portion of disease risk.

Mounting evidence supports the presence of a bidirectional link between the brain and gut microbiota (termed the “gut-brain-axis”) that can affect the behavior and pathology of psychiatric illness. The human microbiome is the “community of commensal, symbiotic, and pathogenic microorganisms” that inhabit our bodies [8]. The last decade has brought about increased access to next-generation sequencing and bioinformatic techniques that has helped to show that gut microbiota play a critical role in human health but can also contribute to disease risk in specific populations [9]. Gut microbiome community structure is compositionally fluid between individuals and will also vary within the same individual depending on the space or time period sampled [10]. Current research aims to determine whether this nonuniformity in microbiome composition may contribute to disease risk or how individuals respond to medication leading to a suboptimal response to drug therapies. In this review, we aim to highlight published evidence that not only links gut microbiota function to BD but also describes the impact of gut microbes on pharmacotherapy.

Gastrointestinal (GI) pathologies are long recognized as common comorbidities in BD and other psychological illness, supporting the theory that GI pathology and psychological illness are interrelated. While, depending on the diagnostic criteria used, irritable bowel syndrome (IBS) is estimated to affect about 11% of the general population [11]. In contrast, rates of comorbidity with psychiatric disorders range from 54 to 94% in those seeking treatment for IBS [12, 13]. A meta-analysis comprised of 177,117 IBS patients and 192,092 healthy controls showed that the prevalence of BD specifically was significantly increased in the IBS population compared to healthy volunteers (OR = 2.48, p < 0.001) [14].

Patients with IBS also exhibit differences in brain morphology when compared to healthy volunteers [15, 16]. A recent study of IBS patients clustered the gut microbial communities into 2 different subtypes; either IBS participants exhibited a healthy control-like microbiome despite concomitant GI symptoms (HC-IBS) or participants exhibited a distinct microbial structure from healthy control subjects (IBS1) [17]. Participants exhibiting the distinct IBS1 microbial community subtype were not only enriched for individuals with early life trauma experiences (an environmental variable for psychiatric illness) but also showed correlated brain structural alterations that were associated with their microbiome community structure.

Gut-derived inflammation appears to be particularly relevant as a mode of bidirectional communication between the gut and the brain, an arrangement that is heavily influenced by the functioning of gut microbes [18-20]. Gut inflammation can cause a “leaky gut” phenotype by decreasing containment of gut contents, particularly gram-negative associated lipopolysaccharide, which can leak into the circulation and elicit both central and systemic inflammatory immune responses while selecting for the survival of specific bacterial species that can tolerate the host immune response [21-23]. Patients with BD often show low-grade peripheral inflammation with further increases in proinflammatory cytokine levels during mood episodes [24]. Patients with schizophrenia and BD have been shown to exhibit higher serum antibody levels to fungal organisms such as Saccharomyces cerevisiae and Candida albicans, as well as soluble CD14 (sCD14), a protein marker of bacterial translocation [25-29]. In addition to gut translocation of microbes, BD patients also show increased exposure to other gut-related markers such as food-derived proteins from the GI system [30].

Yet, significant variation in susceptibility to chronic inflammation exists in BD patients [30-32]. Specific environmental parameters, such as a history of childhood adversity, is associated with elevated proinflammatory plasma cytokines above that of diagnosis without early life trauma [31, 33]. Other data indicate that factors such as antipsychotic medications or dietary products can attenuate this leaky gut phenotype [25, 34]. However, this and other theories delineating inflammatory mechanisms remain grossly unspecific for psychological illness.

Altered immune-inflammatory activity evident in schizophrenia and BD can also manifest via changes in tryptophan metabolism pathways [35-37]. Tryptophan is a dietary amino acid and a precursor for both the kynurenic pathway and the serotonin (5-HT) synthesis pathway [37]. The predominant pathway for tryptophan metabolism is the kynurenic pathway, which transforms tryptophan into excitatory neurotransmitters, many of which are endogenous N-methyl-d-aspartate receptor (NMDAR) antagonists [38]. As part of this pathway, dietary tryptophan is metabolized hepatically, by tryptophan 2,3-dioxygenase (TDO2), or extra-hepatically, by indoleamine 2,3-dioxygenase 1 (IDO1), into kynurenine (Kyn) [35, 39]. Kynurenine is further metabolized to a number of biologically active metabolites such as kynurenic acid (KynA), an NMDAR antagonist [40], hydroxykynurenine (3-HK), which is metabolized further to quinolinic acid, a neurotoxin [41]. Data shows that excess kynurenine metabolites are seen in patients with a psychological illness such as BD [35, 36]. In a clinical study performed by Birner et al. [42], the peripheral blood tryptophan metabolite levels of 143 euthymic to mildly depressive BD patients were compared to those of 101 healthy controls. The authors found that levels of KynA were reduced in BD patients compared to controls. This decreased KynA measure was reflected in the increased 3-HK/Kyn and 3-HK/KynA ratios also detected in BD individuals.

Unlike TDO2, IDO1 is enriched in gut lymphoid tissue and inflammatory signals are known to increase its activity, effectively inducing the production of neurotoxic compounds which potentially contribute to cognitive dysfunction and psychosis [35, 43, 44]. Several independent studies demonstrate that microbial-derived H2O2 inhibits IDO1 resulting in decreased peripheral kyneurines and other tryptophan metabolites [20, 45, 46]. Interestingly, supplementation of a Lactobacillus-containing probiotic exhibited decreased IDO1 activity during chronic SIV infection in a macaque model of HIV/simian immunodeficiency virus [45]. However, the relationship between gut-derived IDO1 inhibition and BD symptom severity has not been examined in animals or humans.

There are already several recent reviews that elegantly summarize research linking gut microbiota to behavior or mental illness [47, 48]. However, mentioned below are some of the more pivotal studies that defined our current view of the “gut-brain axis.”

Sudo et al. [49] were the first to demonstrate that the presence of gut microbiota modulated the long-range hypothalamus-pituitary-adrenal reaction to stress [49]. These experiments showed that germ-free (GF) mice (mice raised in a sterile environment and devoid of gut bacteria) exhibited an elevated stress response as measured by an increased adrenocorticotrophic hormone and corticosterone release compared to control mice with gut microbiota. This exaggerated hypothalamus-pituitary-adrenal response was reversed by the introduction of Bifidobacterium infantis and was somewhat reversed with stool from conventionally raised mice. GF mice also exhibit reduced anxiety-like behavior in addition to altered levels of brain-derived neurotropic factors and other neurotransmitters [49, 50]. Importantly, this microbiota-behavior relationship has been demonstrated in several genetically distinct strains of GF mice, which strengthens the microbiota-behavior relationship [50-52]. Taken together, these preclinical studies demonstrate that the presence of gut microbiota, particularity in early development, is important to behavior in mice.

Evans et al. [53] analyzed the stool microbiome microbiome of clinical bipolar and control participants from the Prechter Longitudinal Study of Bipolar Disorder housed at the University of Michigan. The authors found significant differences in gut microbial communities between the bipolar and healthy control participants. Additionally, individuals with BD showed a decreased relative abundance of the gut microbe known as Faecalibacterium when compared to control participants. Interestingly, for participants with BD, the relative abundance of Faecalibacterium associated with better self-reported health measures based on the Short Form Health Survey (SF12), the Patient Health Questionnaire (PHQ9), the Pittsburg Sleep Quality Index (PSQI), the Generalized Anxiety Disorder scale (GAD7), and the Altman Mania Rating Scale (ASRM), and independently of covariates. Faecalibacterium is a prevalent gut gram-positive microorganism that has demonstrated anti-inflammatory properties [54] and a reported decreased representation in conditions such as inflammatory bowel disease [55], nonalcoholic steatohepatitis [56], and other psychiatric disorders like depression [57, 58].

Specific gut microbes have also been linked to symptoms of mood in a clinical cohort of major depressive disorder [59]. In this investigation, measures of species richness, or the total number of detected gut bacteria, were predictive of insomnia and depression while abundance of Enterobacteriaceae was predictive of anxiety. In the same investigation, Lactobacillus abundance and Enterococcus abundance were also positively related to psychomotor agitation. The consistency of investigations linking specific gut microbes to mood and behavior suggests that the lower gut microbes may be predictive of illness relevant to a depressed state. These data suggest that implementation of supplement or nutritional strategies that can therapeutically increase beneficial organisms, such as Faecalibacterium, in BD patients may be beneficial to reducing disease burden; however, this hypothesis needs to be tested with the appropriate study design.

Recently, Painold et al. [60] found decreased measures of species richness and diversity detected in fecal microbial samples of individuals with a BD diagnosis compared to healthy controls. Additionally, the authors identified a significant increase in the abundance of organisms classified in the Actinobacteria phylum and the class Coriobacteria from BD samples. Healthy controls were reported to exhibit a higher relative abundance of organisms such as Rumicococcaceae spp. and, like in the study of Evans et al. [53], Faecalibacterium spp. These data suggest that implementation of supplement or nutritional strategies that can therapeutically increase beneficial organisms, such as Faecalibacterium, in BD patients may improve mood symptoms, but this needs to be tested with prospective clinical trials.

Several pharmacotherapies including lithium, anticonvulsants, and atypical antipsychotics (AAP) are approved by the Federal Drug Administration for the acute and long-term management of BD. Although monotherapy is ideal, combination drug therapy is often required to achieve BD symptom remission [61]. Treatment regimens may include antidepressants which are added as acute adjunctive treatments for bipolar depressive symptoms. Treatment that results in improvement or remission of a BD episode is followed by worsening of symptoms or episode recurrence in 39–52% of patients, respectively [62]. The interaction between the microbiome and pharmacotherapy may explain the loss of drug effectiveness or the lack of remission of BD symptoms in some patients.

As more attention is paid to the microbiome-host relationship, it is becoming evident that gut microbes are important to the individualized response to pharmacotherapy. Over 60 drugs have been identified to have microbiome interactions according to the PharmacoMicrobiomics database (www.pharmacomicrobiomics.com). Microbiome-host interactions range from a direct influence on drug pharmacokinetics to indirect alteration of the host drug metabolism through modification of the hepatic enzyme activity [10, 63]. As gut microbes are the first point of contact between the body and oral medications, attention has recently been extended to considering the microbiome in precision medicine [64]. Nonuniformity in microbiome composition can contribute to how individuals respond to medication, leading to suboptimal treatment. Alternatively, we are learning that a large proportion of host-directed drugs (non-antimicrobials) exhibit direct activity against commensal microbes that can alter the normal functioning of gut microbes [65]. In this section, we review both clinical and preclinical studies that examine the relationship between drugs used in the treatment of BD and the gut microbiome.

Lithium

Lithium remains the gold standard pharmacotherapy for BD management [66]. While not much is known regarding the interaction between lithium and gut microbiota, a study by Cussotto et al. [67] investigated this relationship in vitro and in rats. In that study, lithium did not exhibit antimicrobial activity against the gram-negative organism Escherichia coli or the gram-positive organism Lactobacillus rhamnosus in vitro. The authors did, however, observe an increase in species richness and diversity in the gut microbiota in rats fed a lithium-supplemented chow, corresponding to approximately 150 mg/kg/day. Additionally, species belonging to the Clostridium, Peptoclostridium, Intestinibacter, and Christenellaceae genera were increased following lithium treatment.

Antipsychotics

AAP are commonly prescribed in BD for the treatment of acute mania and BD depressio, and also in BD maintenance treatment, with quetiapine being the one of the first-line treatments for bipolar depression and mania [61]. Population studies in mental health have noted the contribution of AAP to the burden of cardiac and metabolic disease in the mental health population [68]. A wealth of data has linked gut microbiota to obesity and metabolic disease and, therefore, the contribution of the microbiome to the AAP-associated metabolic risk is currently being investigated. A recent in vitro study showed that antipsychotics as a medication class show direct activity against commensal microbes, specifically Akkermansia muciniphila [65], an organism associated with metabolic health [69]. In GF mice [70], the presence of gut bacteria was determined to be a necessary component for olanzapine-mediated weight gain. Olanzapine treatment showed significant effects on a number of physiologic, inflammatory, and microbial parameters in a rat model [71]. Interestingly, many of these AAP-induced changes were more pronounced in female rats compared with males and were attenuated with coadministration of antibiotics [72].

Many of the observations of AAP in preclinical models have translated to human subjects. Use of the second-generation antipsychotic risperidone, and secondary weight gain has been associated with an altered gut microbiota in male adolescent children [73]. Additionally, in a BD human cohort, AAP treatment was associated with a decreased relative abundance of A. muciniphila and a decreased biodiversity in AAP-treated patients compared to non-AAP-treated BD patients [74, 75]. Due to this documented interaction of AAP and gut microbiota, considering the impact of prescribed medication on microbes will be an important variable when considering host-medication interactions in future studies.

Selective Serotonin Reuptake Inhibitors

Due to their potential to induce mania or rapid cycling, antidepressants are used conservatively in the treatment of BD [1]. Even so, about 34% of those with a BD diagnosis are treated with some sort of antidepressant [76]. Antidepressant drugs have long been known to exhibit a range of antimicrobial effects [77]. The common selective serotonin reuptake inhibitors (SSRI) sertraline, fluoxetine, and paroxetine show activity against gram-positive bacteria such as Staphylococcus and Enterococcus species [78, 79] and gram-negative bacteria such as Pseudomonas aeruginosa and Klebsiella pneumoniae [77, 80]. Specific SSRI, such as fluoxetine, have even been associated with an increased risk of developing a Clostridium difficile infection [81]. While the mechanism of action of SSRI for depression is not related with any antimicrobial effect of these drugs, potential changes in microbial communities may have an effect on other inflammatory or physiological parameters linked to mood.

Anticonvulsants

Anticonvulsant medications are often used as mood stabilizers in BD. The current approved mood stabilizers for BD are valproic acid, lamotrigine, and carbamazepine. While they have not been studied in the context of BD, some anticonvulsants have been found to alter gut microbiome communities in preclinical models. In a valproic acid (VPA)-induced rat model of autism [82], VPA administered to pregnant dams significantly decreased the fecal microbiome diversity in pups and changed the structure of the microbial composition to resemble those derived from patients with autism spectrum disorder. In another study investigating VPA treatment in the rat cecum microbiome [67], increases in the several species belonging to Clostridium, Peptoclostridium, Intestinibacter, and Christenellaceae genera were increased following treatment VPA, along with measurements of species diversity.

Lamotrigine has been shown to be an effective inhibitor of bacterial ribosomal biosynthesis in the model bacterium E. coli [83]. In vitro studies have also described that lamotrigine exhibits antimicrobial activity against gram-positive organisms such as Bacillus subtilis and Staphylococcus aureus [84]. It is imperative to gain insight into how treatment with drugs that exhibit antimicrobial activity in vitro affect the gut microbiome and treatment response in clinical BD cohorts.

Due to the rapid pace of microbial science discovery, many additional functions of the microbiome are likely to be discovered. Researchers are increasingly aware that the gut and the brain communicate and are looking to leverage actions of healthy gut microbiota to treat psychological conditions. Development of precision medicine therapies that exploit gut microbial processes may be beneficial in the near future. However, the microbiome is a complex and dynamic ecosystem and understanding its role in host illness and its potential for the treatment of BD will ultimately require more study.

The authors have no conflict of interests to declare.

Each author significantly contributed to the design, writing, and editing of this review article.

1.
Grande
I
,
Berk
M
,
Birmaher
B
,
Vieta
E
.
Bipolar disorder
.
Lancet
.
2016
Apr
;
387
(
10027
):
1561
72
.
[PubMed]
0140-6736
2.
Alonso
J
,
Petukhova
M
,
Vilagut
G
,
Chatterji
S
,
Heeringa
S
,
Üstün
TB
, et al
Days out of role due to common physical and mental conditions: results from the WHO World Mental Health surveys
.
Mol Psychiatry
.
2011
Dec
;
16
(
12
):
1234
46
.
[PubMed]
1359-4184
3.
Whiteford
HA
,
Degenhardt
L
,
Rehm
J
,
Baxter
AJ
,
Ferrari
AJ
,
Erskine
HE
, et al
Global burden of disease attributable to mental and substance use disorders: findings from the Global Burden of Disease Study 2010
.
Lancet
.
2013
Nov
;
382
(
9904
):
1575
86
.
[PubMed]
0140-6736
4.
Post
RM
.
The impact of bipolar depression
.
J Clin Psychiatry
.
2005
;
66
Suppl 5
:
5
10
.
[PubMed]
0160-6689
5.
Purcell
SM
,
Wray
NR
,
Stone
JL
,
Visscher
PM
,
O’Donovan
MC
,
Sullivan
PF
, et al;
International Schizophrenia Consortium
.
Common polygenic variation contributes to risk of schizophrenia and bipolar disorder
.
Nature
.
2009
Aug
;
460
(
7256
):
748
52
.
[PubMed]
0028-0836
6.
Lichtenstein
P
,
Yip
BH
,
Björk
C
,
Pawitan
Y
,
Cannon
TD
,
Sullivan
PF
, et al
Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study
.
Lancet
.
2009
Jan
;
373
(
9659
):
234
9
.
[PubMed]
0140-6736
7.
Van Snellenberg
JX
,
de Candia
T
.
Meta-analytic evidence for familial coaggregation of schizophrenia and bipolar disorder
.
Arch Gen Psychiatry
.
2009
Jul
;
66
(
7
):
748
55
.
[PubMed]
0003-990X
8.
Clarke
G
,
Stilling
RM
,
Kennedy
PJ
,
Stanton
C
,
Cryan
JF
,
Dinan
TG
.
Minireview: Gut microbiota: the neglected endocrine organ
.
Mol Endocrinol
.
2014
Aug
;
28
(
8
):
1221
38
.
[PubMed]
0888-8809
9.
McCarthy
JJ
,
McLeod
HL
,
Ginsburg
GS
.
Genomic medicine: a decade of successes, challenges, and opportunities
.
Sci Transl Med
.
2013
Jun
;
5
(
189
):
189sr4
.
[PubMed]
1946-6234
10.
ElRakaiby
M
,
Dutilh
BE
,
Rizkallah
MR
,
Boleij
A
,
Cole
JN
,
Aziz
RK
.
Pharmacomicrobiomics: the impact of human microbiome variations on systems pharmacology and personalized therapeutics
.
OMICS
.
2014
Jul
;
18
(
7
):
402
14
.
[PubMed]
1536-2310
11.
Lovell
RM
,
Ford
AC
.
Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis
.
Clin Gastroenterol Hepatol
.
2012
Jul
;
10
(
7
):
712
721.e4
.
[PubMed]
1542-3565
12.
Whitehead
WE
,
Palsson
O
,
Jones
KR
.
Systematic review of the comorbidity of irritable bowel syndrome with other disorders: what are the causes and implications?
Gastroenterology
.
2002
Apr
;
122
(
4
):
1140
56
.
[PubMed]
0016-5085
13.
Roy-Byrne
PP
,
Davidson
KW
,
Kessler
RC
,
Asmundson
GJ
,
Goodwin
RD
,
Kubzansky
L
, et al
Anxiety disorders and comorbid medical illness
.
Gen Hosp Psychiatry
.
2008
May-Jun
;
30
(
3
):
208
25
.
[PubMed]
0163-8343
14.
Tseng
PT
,
Zeng
BS
,
Chen
YW
,
Wu
MK
,
Wu
CK
,
Lin
PY
.
A meta-analysis and systematic review of the comorbidity between irritable bowel syndrome and bipolar disorder
.
Medicine (Baltimore)
.
2016
Aug
;
95
(
33
):
e4617
.
[PubMed]
0025-7974
15.
Piché
M
,
Chen
JI
,
Roy
M
,
Poitras
P
,
Bouin
M
,
Rainville
P
.
Thicker posterior insula is associated with disease duration in women with irritable bowel syndrome (IBS) whereas thicker orbitofrontal cortex predicts reduced pain inhibition in both IBS patients and controls
.
J Pain
.
2013
Oct
;
14
(
10
):
1217
26
.
[PubMed]
1526-5900
16.
Ellingson
BM
,
Mayer
E
,
Harris
RJ
,
Ashe-McNally
C
,
Naliboff
BD
,
Labus
JS
, et al
Diffusion tensor imaging detects microstructural reorganization in the brain associated with chronic irritable bowel syndrome
.
Pain
.
2013
Sep
;
154
(
9
):
1528
41
.
[PubMed]
0304-3959
17.
Labus
JS
,
Hollister
EB
,
Jacobs
J
,
Kirbach
K
,
Oezguen
N
,
Gupta
A
, et al
Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome
.
Microbiome
.
2017
May
;
5
(
1
):
49
.
[PubMed]
2049-2618
18.
Houser
MC
,
Tansey
MG
.
The gut-brain axis: is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis?
NPJ Parkinsons Dis
.
2017
Jan
;
3
(
1
):
3
.
[PubMed]
2373-8057
19.
Jobin
C
,
Sartor
RB
.
The I kappa B/NF-kappa B system: a key determinant of mucosalinflammation and protection
.
Am J Physiol Cell Physiol
.
2000
Mar
;
278
(
3
):
C451
62
.
[PubMed]
0363-6143
20.
Marcial
GE
,
Ford
AL
,
Haller
MJ
,
Gezan
SA
,
Harrison
NA
,
Cai
D
, et al
Lactobacillus johnsonii N6.2 Modulates the Host Immune Responses: A Double-Blind, Randomized Trial in Healthy Adults
.
Front Immunol
.
2017
Jun
;
8
:
655
.
[PubMed]
1664-3224
21.
Zhao
Y
,
Cong
L
,
Jaber
V
,
Lukiw
WJ
.
Microbiome-Derived Lipopolysaccharide Enriched in the Perinuclear Region of Alzheimer’s Disease Brain
.
Front Immunol
.
2017
Sep
;
8
:
1064
.
[PubMed]
1664-3224
22.
Abraham
C
,
Medzhitov
R
.
Interactions between the host innate immune system and microbes in inflammatory bowel disease
.
Gastroenterology
.
2011
May
;
140
(
6
):
1729
37
.
[PubMed]
0016-5085
23.
Schirmer
M
,
Smeekens
SP
,
Vlamakis
H
,
Jaeger
M
,
Oosting
M
,
Franzosa
EA
, et al
Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity
.
Cell
.
2016
Nov
;
167
(
4
):
1125
1136.e8
.
[PubMed]
0092-8674
24.
Bai
YM
,
Su
TP
,
Tsai
SJ
,
Wen-Fei
C
,
Li
CT
,
Pei-Chi
T
, et al
Comparison of inflammatory cytokine levels among type I/type II and manic/hypomanic/euthymic/depressive states of bipolar disorder
.
J Affect Disord
.
2014
Sep
;
166
:
187
92
.
[PubMed]
0165-0327
25.
Severance
EG
,
Alaedini
A
,
Yang
S
,
Halling
M
,
Gressitt
KL
,
Stallings
CR
, et al
Gastrointestinal inflammation and associated immune activation in schizophrenia
.
Schizophr Res
.
2012
Jun
;
138
(
1
):
48
53
.
[PubMed]
0920-9964
26.
Severance
EG
,
Gressitt
KL
,
Stallings
CR
,
Katsafanas
E
,
Schweinfurth
LA
,
Savage
CL
, et al
Candida albicans exposures, sex specificity and cognitive deficits in schizophrenia and bipolar disorder
.
NPJ Schizophr
.
2016
May
;
2
(
1
):
16018
.
[PubMed]
2334-265X
27.
Severance
EG
,
Gressitt
KL
,
Stallings
CR
,
Origoni
AE
,
Khushalani
S
,
Leweke
FM
, et al
Discordant patterns of bacterial translocation markers and implications for innate immune imbalances in schizophrenia
.
Schizophr Res
.
2013
Aug
;
148
(
1-3
):
130
7
.
[PubMed]
0920-9964
28.
Kim
YK
,
Jung
HG
,
Myint
AM
,
Kim
H
,
Park
SH
.
Imbalance between pro-inflammatory and anti-inflammatory cytokines in bipolar disorder
.
J Affect Disord
.
2007
Dec
;
104
(
1-3
):
91
5
.
[PubMed]
0165-0327
29.
Debnath
M
,
Berk
M
.
Th17 pathway-mediated immunopathogenesis of schizophrenia: mechanisms and implications
.
Schizophr Bull
.
2014
Nov
;
40
(
6
):
1412
21
.
[PubMed]
0586-7614
30.
Severance
EG
,
Gressitt
KL
,
Yang
S
,
Stallings
CR
,
Origoni
AE
,
Vaughan
C
, et al
Seroreactive marker for inflammatory bowel disease and associations with antibodies to dietary proteins in bipolar disorder
.
Bipolar Disord
.
2014
May
;
16
(
3
):
230
40
.
[PubMed]
1398-5647
31.
Dennison
U
,
McKernan
D
,
Cryan
J
,
Dinan
T
.
Schizophrenia patients with a history of childhood trauma have a pro-inflammatory phenotype
.
Psychol Med
.
2012
Sep
;
42
(
9
):
1865
71
.
[PubMed]
0033-2917
32.
Goldsmith
DR
,
Rapaport
MH
,
Miller
BJ
.
A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression
.
Mol Psychiatry
.
2016
Dec
;
21
(
12
):
1696
709
.
[PubMed]
1359-4184
33.
Meyer
U
,
Feldon
J
.
Prenatal exposure to infection: a primary mechanism for abnormal dopaminergic development in schizophrenia
.
Psychopharmacology (Berl)
.
2009
Nov
;
206
(
4
):
587
602
.
[PubMed]
0033-3158
34.
Severance
EG
,
Gressitt
KL
,
Stallings
CR
,
Katsafanas
E
,
Schweinfurth
LA
,
Savage
CL
, et al
Probiotic normalization of Candida albicans in schizophrenia: A randomized, placebo-controlled, longitudinal pilot study
.
Brain Behav Immun
.
2017
May
;
62
:
41
5
.
[PubMed]
0889-1591
35.
Anderson
G
,
Maes
M
.
Bipolar disorder: role of immune-inflammatory cytokines, oxidative and nitrosative stress and tryptophan catabolites
.
Curr Psychiatry Rep
.
2015
Feb
;
17
(
2
):
8
.
[PubMed]
1523-3812
36.
Myint
AM
,
Kim
YK
,
Verkerk
R
,
Park
SH
,
Scharpé
S
,
Steinbusch
HW
, et al
Tryptophan breakdown pathway in bipolar mania
.
J Affect Disord
.
2007
Sep
;
102
(
1-3
):
65
72
.
[PubMed]
0165-0327
37.
O’Mahony
SM
,
Clarke
G
,
Borre
YE
,
Dinan
TG
,
Cryan
JF
.
Serotonin, tryptophan metabolism and the brain-gut-microbiome axis
.
Behav Brain Res
.
2015
Jan
;
277
:
32
48
.
[PubMed]
0166-4328
38.
Anderson
G
,
Jacob
A
,
Bellivier
F
,
Geoffroy
PA
.
Bipolar Disorder: The Role of the Kynurenine and Melatonergic Pathways
.
Curr Pharm Des
.
2016
;
22
(
8
):
987
1012
.
[PubMed]
1381-6128
39.
Larkin
PB
,
Sathyasaikumar
KV
,
Notarangelo
FM
,
Funakoshi
H
,
Nakamura
T
,
Schwarcz
R
, et al
Tryptophan 2,3-dioxygenase and indoleamine 2,3-dioxygenase 1 make separate, tissue-specific contributions to basal and inflammation-induced kynurenine pathway metabolism in mice
.
Biochim Biophys Acta
.
2016
Nov
;
1860
(
11
11 Pt A
):
2345
54
.
[PubMed]
0006-3002
40.
Mok
MH
,
Fricker
AC
,
Weil
A
,
Kew
JN
.
Electrophysiological characterisation of the actions of kynurenic acid at ligand-gated ion channels
.
Neuropharmacology
.
2009
Sep
;
57
(
3
):
242
9
.
[PubMed]
0028-3908
41.
Lugo-Huitrón
R
,
Ugalde Muñiz
P
,
Pineda
B
,
Pedraza-Chaverrí
J
,
Ríos
C
,
Pérez-de la Cruz
V
. (
2013
) Quinolinic Acid: An Endogenous Neurotoxin with Multiple Targets. In: Oxidative Medicine and Cellular Longevity.
42.
Birner
A
,
Platzer
M
,
Bengesser
SA
,
Dalkner
N
,
Fellendorf
FT
,
Queissner
R
, et al
Increased breakdown of kynurenine towards its neurotoxic branch in bipolar disorder
.
PLoS One
.
2017
Feb
;
12
(
2
):
e0172699
.
[PubMed]
1932-6203
43.
Metz
R
,
Smith
C
,
DuHadaway
JB
,
Chandler
P
,
Baban
B
,
Merlo
LM
, et al
IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation
.
Int Immunol
.
2014
Jul
;
26
(
7
):
357
67
.
[PubMed]
0953-8178
44.
Penberthy
WT
;
W. T. Penberthy
.
Pharmacological targeting of IDO-mediated tolerance for treating autoimmune disease
.
Curr Drug Metab
.
2007
Apr
;
8
(
3
):
245
66
.
[PubMed]
1389-2002
45.
Vujkovic-Cvijin
I
,
Swainson
LA
,
Chu
SN
,
Ortiz
AM
,
Santee
CA
,
Petriello
A
, et al
Gut-Resident Lactobacillus Abundance Associates with IDO1 Inhibition and Th17 Dynamics in SIV-Infected Macaques
.
Cell Rep
.
2015
Nov
;
13
(
8
):
1589
97
.
[PubMed]
2639-1856
46.
Strasser
B
,
Geiger
D
,
Schauer
M
,
Gostner
JM
,
Gatterer
H
,
Burtscher
M
, et al
Probiotic Supplements Beneficially Affect Tryptophan-Kynurenine Metabolism and Reduce the Incidence of Upper Respiratory Tract Infections in Trained Athletes: A Randomized, Double-Blinded, Placebo-Controlled Trial
.
Nutrients
.
2016
Nov
;
8
(
11
):
8
.
[PubMed]
2072-6643
47.
Johnson
KV
,
Foster
KR
.
Why does the microbiome affect behaviour?
Nat Rev Microbiol
.
2018
Oct
;
16
(
10
):
647
55
.
[PubMed]
1740-1526
48.
Stefano
GB
,
Pilonis
N
,
Ptacek
R
,
Raboch
J
,
Vnukova
M
,
Kream
RM
.
Gut, Microbiome, and Brain Regulatory Axis: Relevance to Neurodegenerative and Psychiatric Disorders
.
Cell Mol Neurobiol
.
2018
Aug
;
38
(
6
):
1197
206
.
[PubMed]
0272-4340
49.
Sudo
N
,
Chida
Y
,
Aiba
Y
,
Sonoda
J
,
Oyama
N
,
Yu
XN
, et al
Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice
.
J Physiol
.
2004
Jul
;
558
(
Pt 1
):
263
75
.
[PubMed]
0022-3751
50.
Clarke
G
,
Grenham
S
,
Scully
P
,
Fitzgerald
P
,
Moloney
RD
,
Shanahan
F
, et al
The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner
.
Mol Psychiatry
.
2013
Jun
;
18
(
6
):
666
73
.
[PubMed]
1359-4184
51.
Diaz Heijtz
R
,
Wang
S
,
Anuar
F
,
Qian
Y
,
Björkholm
B
,
Samuelsson
A
, et al
Normal gut microbiota modulates brain development and behavior
.
Proc Natl Acad Sci USA
.
2011
Feb
;
108
(
7
):
3047
52
.
[PubMed]
0027-8424
52.
Neufeld
KM
,
Kang
N
,
Bienenstock
J
,
Foster
JA
(
2011
)
Reduced anxiety-like behavior and central neurochemical change in germ-free mice.
Neurogastroenterol Motil 23:255–264, e119 .
53.
Evans
SJ
,
Bassis
CM
,
Hein
R
,
Assari
S
,
Flowers
SA
,
Kelly
MB
, et al
The gut microbiome composition associates with bipolar disorder and illness severity
.
J Psychiatr Res
.
2017
Apr
;
87
:
23
9
.
[PubMed]
0022-3956
54.
Sokol
H
,
Pigneur
B
,
Watterlot
L
,
Lakhdari
O
,
Bermúdez-Humarán
LG
,
Gratadoux
JJ
, et al
Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients
.
Proc Natl Acad Sci USA
.
2008
Oct
;
105
(
43
):
16731
6
.
[PubMed]
0027-8424
55.
Sokol
H
,
Seksik
P
,
Furet
JP
,
Firmesse
O
,
Nion-Larmurier
I
,
Beaugerie
L
, et al
Low counts of Faecalibacterium prausnitzii in colitis microbiota
.
Inflamm Bowel Dis
.
2009
Aug
;
15
(
8
):
1183
9
.
[PubMed]
1078-0998
56.
Wong
VW
,
Tse
CH
,
Lam
TT
,
Wong
GL
,
Chim
AM
,
Chu
WC
, et al
Molecular characterization of the fecal microbiota in patients with nonalcoholic steatohepatitis—a longitudinal study
.
PLoS One
.
2013
Apr
;
8
(
4
):
e62885
.
[PubMed]
1932-6203
57.
Aizawa
E
,
Tsuji
H
,
Asahara
T
,
Takahashi
T
,
Teraishi
T
,
Yoshida
S
, et al
Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder
.
J Affect Disord
.
2016
Sep
;
202
:
254
7
.
[PubMed]
0165-0327
58.
Jiang
H
,
Ling
Z
,
Zhang
Y
,
Mao
H
,
Ma
Z
,
Yin
Y
, et al
Altered fecal microbiota composition in patients with major depressive disorder
.
Brain Behav Immun
.
2015
Aug
;
48
:
186
94
.
[PubMed]
0889-1591
59.
Mason
B
,
Koh
A
,
Enkevort
EV
,
Trivedi
M
.
676. Gut Microbiota Distributions Predict Mood Disorder Symptoms and Mediate Dietary Interactions
.
Biol Psychiatry
.
2017
;
81
(
10
):
S273
4
. 0006-3223
60.
Painold
A
,
Mörkl
S
,
Kashofer
K
,
Halwachs
B
,
Dalkner
N
,
Bengesser
S
, et al
A step ahead: exploring the gut microbiota in inpatients with bipolar disorder during a depressive episode
.
Bipolar Disord
.
2019
Feb
;
21
(
1
):
40
9
.
[PubMed]
1398-5647
61.
Yatham
LN
,
Kennedy
SH
,
Parikh
SV
,
Schaffer
A
,
Bond
DJ
,
Frey
BN
, et al
Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) 2018 guidelines for the management of patients with bipolar disorder
.
Bipolar Disord
.
2018
Mar
;
20
(
2
):
97
170
.
[PubMed]
1398-5647
62.
Tohen
M
,
Zarate
CA
 Jr
,
Hennen
J
,
Khalsa
HM
,
Strakowski
SM
,
Gebre-Medhin
P
, et al
The McLean-Harvard First-Episode Mania Study: prediction of recovery and first recurrence
.
Am J Psychiatry
.
2003
Dec
;
160
(
12
):
2099
107
.
[PubMed]
0002-953X
63.
Selwyn
FP
,
Cheng
SL
,
Klaassen
CD
,
Cui
JY
.
Regulation of Hepatic Drug-Metabolizing Enzymes in Germ-Free Mice by Conventionalization and Probiotics
.
Drug Metab Dispos
.
2016
Feb
;
44
(
2
):
262
74
.
[PubMed]
0090-9556
64.
Kuntz
TM
,
Gilbert
JA
.
Introducing the Microbiome into Precision Medicine
.
Trends Pharmacol Sci
.
2017
Jan
;
38
(
1
):
81
91
.
[PubMed]
0165-6147
65.
Maier
L
,
Pruteanu
M
,
Kuhn
M
,
Zeller
G
,
Telzerow
A
,
Anderson
EE
, et al
Extensive impact of non-antibiotic drugs on human gut bacteria
.
Nature
.
2018
Mar
;
555
(
7698
):
623
8
.
[PubMed]
0028-0836
66.
Lähteenvuo
M
,
Tanskanen
A
,
Taipale
H
,
Hoti
F
,
Vattulainen
P
,
Vieta
E
, et al
Real-world Effectiveness of Pharmacologic Treatments for the Prevention of Rehospitalization in a Finnish Nationwide Cohort of Patients With Bipolar Disorder
.
JAMA Psychiatry
.
2018
Apr
;
75
(
4
):
347
55
.
[PubMed]
2168-622X
67.
Cussotto
S
,
Strain
CR
,
Fouhy
F
,
Strain
RG
,
Peterson
VL
,
Clarke
G
, et al
Differential effects of psychotropic drugs on microbiome composition and gastrointestinal function
.
Psychopharmacology (Berl)
.
2019
May
;
236
(
5
):
1671
85
.
[PubMed]
0033-3158
68.
McEvoy
JP
,
Meyer
JM
,
Goff
DC
,
Nasrallah
HA
,
Davis
SM
,
Sullivan
L
, et al
Prevalence of the metabolic syndrome in patients with schizophrenia: baseline results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) schizophrenia trial and comparison with national estimates from NHANES III
.
Schizophr Res
.
2005
Dec
;
80
(
1
):
19
32
.
[PubMed]
0920-9964
69.
Schneeberger
M
,
Everard
A
,
Gómez-Valadés
AG
,
Matamoros
S
,
Ramírez
S
,
Delzenne
NM
, et al
Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice
.
Sci Rep
.
2015
Nov
;
5
(
1
):
16643
.
[PubMed]
2045-2322
70.
Morgan
AP
,
Crowley
JJ
,
Nonneman
RJ
,
Quackenbush
CR
,
Miller
CN
,
Ryan
AK
, et al
The antipsychotic olanzapine interacts with the gut microbiome to cause weight gain in mouse
.
PLoS One
.
2014
Dec
;
9
(
12
):
e115225
.
[PubMed]
1932-6203
71.
Davey
KJ
,
O’Mahony
SM
,
Schellekens
H
,
O’Sullivan
O
,
Bienenstock
J
,
Cotter
PD
, et al
Gender-dependent consequences of chronic olanzapine in the rat: effects on body weight, inflammatory, metabolic and microbiota parameters
.
Psychopharmacology (Berl)
.
2012
May
;
221
(
1
):
155
69
.
[PubMed]
0033-3158
72.
Davey
KJ
,
Cotter
PD
,
O’Sullivan
O
,
Crispie
F
,
Dinan
TG
,
Cryan
JF
, et al
Antipsychotics and the gut microbiome: olanzapine-induced metabolic dysfunction is attenuated by antibiotic administration in the rat
.
Transl Psychiatry
.
2013
Oct
;
3
(
10
):
e309
.
[PubMed]
2158-3188
73.
Bahr
SM
,
Tyler
BC
,
Wooldridge
N
,
Butcher
BD
,
Burns
TL
,
Teesch
LM
, et al
Use of the second-generation antipsychotic, risperidone, and secondary weight gain are associated with an altered gut microbiota in children
.
Transl Psychiatry
.
2015
Oct
;
5
(
10
):
e652
.
[PubMed]
2158-3188
74.
Flowers
SA
,
Evans
SJ
,
Ward
KM
,
McInnis
MG
,
Ellingrod
VL
.
Interaction Between Atypical Antipsychotics and the Gut Microbiome in a Bipolar Disease Cohort
.
Pharmacotherapy
.
2017
Mar
;
37
(
3
):
261
7
.
[PubMed]
0277-0008
75.
Flowers
SA
,
Baxter
NT
,
Ward
KM
,
Kraal
AZ
,
McInnis
MG
,
Schmidt
TM
, et al
Effects of Atypical Antipsychotic Treatment and Resistant Starch Supplementation on Gut Microbiome Composition in a Cohort of Patients with Bipolar Disorder or Schizophrenia
.
Pharmacotherapy
.
2019
Feb
;
39
(
2
):
161
70
.
[PubMed]
0277-0008
76.
Cascade
EF
,
Reites
J
,
Kalali
AH
,
Ghaemi
N
.
Antidepressants in bipolar disorder
.
Psychiatry (Edgmont)
.
2007
Mar
;
4
(
3
):
56
8
.
[PubMed]
1555-5194
77.
Macedo
D
,
Filho
AJ
,
Soares de Sousa
CN
,
Quevedo
J
,
Barichello
T
,
Júnior
HV
, et al
Antidepressants, antimicrobials or both? Gut microbiota dysbiosis in depression and possible implications of the antimicrobial effects of antidepressant drugs for antidepressant effectiveness
.
J Affect Disord
.
2017
Jan
;
208
:
22
32
.
[PubMed]
0165-0327
78.
Ayaz
M
,
Subhan
F
,
Ahmed
J
,
Khan
AU
,
Ullah
F
,
Ullah
I
, et al
Sertraline enhances the activity of antimicrobial agents against pathogens of clinical relevance
.
J Biol Res (Thessalon)
.
2015
Apr
;
22
(
1
):
4
.
[PubMed]
1790-045X
79.
Coban
AY
,
Tanriverdi Cayci
Y
,
Keleş Uludağ
S
,
Durupinar
B
.
[Investigation of antibacterial activity of sertralin]
.
Mikrobiyol Bul
.
2009
Oct
;
43
(
4
):
651
6
.
[PubMed]
0374-9096
80.
Kruszewska
H
,
Zareba
T
,
Tyski
S
.
Examination of antimicrobial activity of selected non-antibiotic medicinal preparations
.
Acta Pol Pharm
.
2012
Nov-Dec
;
69
(
6
):
1368
71
.
[PubMed]
0001-6837
81.
Rogers
MA
,
Greene
MT
,
Young
VB
,
Saint
S
,
Langa
KM
,
Kao
JY
, et al
Depression, antidepressant medications, and risk of Clostridium difficile infection
.
BMC Med
.
2013
May
;
11
(
1
):
121
.
[PubMed]
1741-7015
82.
Liu
F
,
Horton-Sparks
K
,
Hull
V
,
Li
RW
,
Martínez-Cerdeño
V
.
The valproic acid rat model of autism presents with gut bacterial dysbiosis similar to that in human autism
.
Mol Autism
.
2018
Dec
;
9
(
1
):
61
.
[PubMed]
2040-2392
83.
Stokes
JM
,
Davis
JH
,
Mangat
CS
,
Williamson
JR
,
Brown
ED
.
Discovery of a small molecule that inhibits bacterial ribosome biogenesis
.
eLife
.
2014
Sep
;
3
:
e03574
.
[PubMed]
2050-084X
84.
Qian
Y
,
Lv
PC
,
Fang
RQ
,
Song
ZC
,
Zhu
HL
.
Synthesis, antimicrobial activity of lamotrigine and its ammonium derivatives
.
J Chem Sci
.
2009
;
121
(
4
):
463
70
. 0253-4134

To be considered for publication in a special issue of Neuropsychobiology (“Effects of Nutrition, Exercise and the Gut-Brain-Axis on Psychiatric Disorders”).

Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.