Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

877954 ANP ANZJP ArticlesMcGovern et al.

Key Review

Australian & New Zealand Journal of Psychiatry

A review of the antimicrobial side 2019, Vol. 53(12) 1151­–1166


https://doi.org/10.1177/0004867419877954
DOI: 10.1177/0004867419877954

of antidepressants and its putative © The Royal Australian and


New Zealand College of Psychiatrists 2019

implications on the gut microbiome Article reuse guidelines:


sagepub.com/journals-permissions
journals.sagepub.com/home/anp

Abigail S McGovern , Adam S Hamlin and Gal Winter

Abstract
Objective: Serotonin reuptake inhibitors are the predominant treatment for major depressive disorder. In recent
years, the diversity of the gut microbiota has emerged to play a significant role in the occurrence of major depressive
disorder and other mood and anxiety disorders. Importantly, the role of the gut microbiota in the treatment of such
disorders remains to be elucidated. Here, we provide a review of the literature regarding the effects of physiologically
relevant concentrations of serotonin reuptake inhibitors on the gut microbiota and the implications this might have on
their efficacy in the treatment of mood disorders.
Methods: First, an estimation of gut serotonin reuptake inhibitor concentrations was computed based on pharmaco-
kinetic and gastrointestinal transit properties of serotonin reuptake inhibitors. Literature regarding the in vivo and in
vitro antimicrobial properties of serotonin reuptake inhibitors was gathered, and the estimated gut concentrations were
examined in the context of these data. Computer-based investigation revealed putative mechanisms for the antimicrobial
effects of serotonin reuptake inhibitors.
Results: In vivo evidence using animal models shows an antimicrobial effect of serotonin reuptake inhibitors on the gut
microbiota. Examination of the estimated physiological concentrations of serotonin reuptake inhibitors in the gastroin-
testinal tract collected from in vitro studies suggests that the microbial community of both the small intestine and the
colon are exposed to serotonin reuptake inhibitors for at least 4 hours per day at concentrations that are likely to exert
an antimicrobial effect. The potential mechanisms of the effect of serotonin reuptake inhibitors on the gut microbiota
were postulated to include inhibition of efflux pumps and/or amino acid transporters.
Conclusion: This review raises important issues regarding the role that gut microbiota play in the treatment of mood-
related behaviours, which holds substantial potential clinical outcomes for patients suffering from major depressive
disorder and other mood-related disorders.

Keywords
Depression, gut microbiota, serotonin reuptake inhibitor, antidepressants, gut microbiome, major depressive disorder,
selective serotonin reuptake inhibitors

Introduction
Antidepressants are used in the treatment of affective and First, while the synaptic effect of SSRIs is instantaneous,
anxiety disorders, most commonly for the treatment of reductions in MDD symptomology take several weeks
major depressive disorder (MDD; Abbing-Karahagopian
et al., 2014). They are in common use worldwide, with
annual usage estimated to be ~10% in Western population School of Science and Technology, University of New England,
(Abbing-Karahagopian et al., 2014; Brett et al., 2017; Pratt Armidale, NSW, Australia
et al., 2017). Despite selective serotonin reuptake inhibitors
Corresponding author:
(SSRIs) being the most commonly prescribed class of anti- Gal Winter, School of Science and Technology, University of New
depressants (Abbing-Karahagopian et al., 2014), there are England, Armidale, NSW 2351, Australia.
many uncertainties regarding SSRI treatment of MDD. Email: gwinterz@une.edu.au

Australian & New Zealand Journal of Psychiatry, 53(12)


1152 ANZJP Articles

(Jeon and Kim, 2016). Furthermore, only ~30–40% of the Specific examples include pharmaceuticals such as antidia-
MDD patients experience remission (Keller et al., 2007; betics (metformin; Forslund et al., 2015), proton pump
Rush et al., 2006) and of these patients, many (~20–40%) inhibitors (PPIs; Imhann et al., 2016; Jackson et al., 2016),
relapse even with continuing treatment (Rush et al., 2006; nonsteroidal anti-inflammatory drugs (NSAIDs; Rogers
Sim et al., 2015). As treatment-resistant MDD presents a and Aronoff, 2016) and atypical antipsychotics (AAPs;
substantive societal burden (Mrazek et al., 2014), a better Flowers et al., 2017). In a microbial community as diverse
understanding of the factors that may interact with SSRI as the gut ecosystem, it is logical to assume that some of the
mediation of antidepressant effect is imperative. microbes would be able to metabolise the consumed drugs
In recent years, the gut microbiota (GM) composition and that this will either have a growth-promoting effect or
has been associated with a number of illnesses, including a growth-inhibiting (antimicrobial) effect on microbes.
psychiatric disorders (Moya and Ferrer, 2016; Rojo et al., Indeed, microbial metabolism of over 240 pharmaceutical
2017). Recent studies point towards a strong link between drugs of all therapeutic classes has been shown in vitro
the diversity of the GM and depression (Winter et al., (Maier et al., 2018; Zimmermann et al., 2019). When con-
2018). MDD has been associated with reductions in micro- sidering the effect of drug metabolism on GM diversity, it
bial richness and diversity, as well as changes in relative is important to apprehend that metabolism of the drug by
abundances of specific microbial taxa (Winter et al., 2018). even one microbial strain will have a rolling effect on the
The gut microbial community is a complex ecological sys- rest of the microbial community and will lead to changes in
tem. It is composed mostly of bacteria from the Bacteroidetes the overall diversity.
and Firmicutes phyla (90%), with the Actinobacteria, As such, SSRIs have the capacity to exert a constant
Proteobacteria and Verrucomicrobia phyla comprising only antimicrobial pressure and affect community diversity,
a small part of the GM (Eckburg et al., 2005). Other symbi- especially considering that they are taken for long dura-
onts of the gut include fungi (particularly yeasts) and some tions. For instance, in the United States, around 25% of the
viruses (Lozupone et al., 2012). Gut microbes act as a func- individuals who used antidepressants between 2011 and
tional group in a manner dependent on metabolic activity, 2014 had done so for a period of greater than 10 years (Pratt
which is largely determined by the community composition et al., 2017). However, for SSRIs to affect the GM diver-
(Moya and Ferrer, 2016). The microbiota and associated sity, they would need to be present in the gut lumen at suf-
metabolic by-products may affect the host’s brain via a ficient concentrations, especially in the microbial-enriched
number of pathways, including (1) vagal signalling, (2) regions such as the ascending colon (Donaldson et al.,
production of neuroactive metabolites such as catechola- 2015).
mine neurotransmitters or short chain fatty acids or (3) In this review article, we present new evidence regard-
indirectly via the immune system (Bonaz et al., 2018; Fung ing in vivo antimicrobial activity of SSRIs. We then evalu-
et al., 2017; Kasubuchi et al., 2015; Rooks and Garrett, ate the extent by which GM is exposed to SSRIs by
2016; Stilling et al., 2016; Yano Jessica et al., 2015). evaluating the gastrointestinal (GI) concentrations of
SSRIs using pharmacokinetic (PK) properties and the tem-
poral nature of their GI transit. These concentrations are
Gut microbiome diversity and dynamics then compared with in vitro evidence assessing the effect
The dynamics of the gut microbial community are complex of SSRIs against bacteria and fungi. A computer-based
due to interactions between microbial species and interac- analysis is further performed to identify potential mecha-
tions of microbes with their environment. The literature nisms responsible for the antimicrobial effect of SSRIs.
suggests that the microbial community is drawn to a stable Finally, we discuss the clinical implications of these effects
equilibrium composition (Lozupone et al., 2012; Mehta and their relevance to SSRI treatment of mood-related
et al., 2018) whose stability is facilitated by competitive disorders.
inhibition, host regulation and adequate nutrients (Coyte
et al., 2015). However, a number of environmental and life- In vivo evidence for the
style factors, including drug consumption, may lead to dys-
biosis (Cryan and Dinan, 2019; David et al., 2014; Kundu antimicrobial activity of SSRIs
et al., 2017; Moloney et al., 2014; Rojo et al., 2017). The Recent studies using animal models provide in vivo evi-
most extreme example of this is antibiotics. Even a single dence for the antimicrobial activity of SSRIs. Cussotto
course of antibiotics substantially reduce richness and et al. (2019) demonstrated using rats that animals treated
diversity of GM (Eisen et al., 2008; Isaac et al., 2017); with fluoxetine (FLU) and escitalopram (ECIT) had an
however, over time the GM usually progresses towards its altered microbiota composition and that FLU-treated ani-
original equilibrium state (Rojo et al., 2017). Other phar- mals showed a decrease in Deferribacteres as determined
maceutical agents designed to target human cells rather by 16S rRNA sequencing analysis of faecal samples
than microbial cells have been shown to modulate the GM (Cussotto et al., 2019). Importantly, this study was con-
diversity (Maier et al., 2018; Zimmermann et al., 2019). ducted on healthy rat models and included administration

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1153

Figure 1. Box 1 shows the daily dosage range of several common SSRIs. These include fluoxetine (FLU), sertraline (SER),
paroxetine (PRX), fluvoxamine (FVX), escitalopram (ECIT) and citalopram (CIT). Box 2 shows the percentage of the drug that
is estimated to remain, unabsorbed, in the lumen and an estimate maximal concentration as the drug passes through the ileum.
All percentages are calculated based on the literature sourced mean pharmacokinetic parameters at times that represent the
GI transit as determined experimentally using radiolabelled tablets. All concentration estimates are calculated based on taking
the maximum dose of the drug and assume complete dissolution in an experimentally determined mean fluid volume for that
region of the digestive tract. The values in brackets give some idea of the possible variation between individuals given different
gastrointestinal transit speeds; they were computed using the mean time plus or minus one standard deviation. Box 3 shows the
percentage of the drug that is unabsorbed and an estimate maximal concentration at the time that half of the drug arrives at the
colon. Concentrations are therefore calculated using half of the remaining drug. Box 4 shows the percentage of the drug that is
unabsorbed and an estimate maximal concentration 4 hours later. Concentrations assume that the entire drug has reached the
colon. Box 5 shows the percentage of a radiolabelled dose of the given SSRI excreted in the faeces in mass balance studies.

of lower doses than the recommended daily dosages used in (MV), where rats are exposed to early life stress and devel-
Figure 1. Lyte et al. (2019) administered FLU at the recom- oped a depressive-like phenotype. The authors examined
mended daily dosage to healthy male mice and reported a the GM diversity and metabolic capacity of MV rats treated
significant, time-dependent alteration of microbial diver- with FLU during pregnancy and lactation, compared to a
sity, based on 16S rRNA analysis of faecal samples. The control group. Results from this study show that FLU
authors also report changes in body mass resulting from administration had a significant impact on GM diversity
FLU administration. Among the microbial genera decreased and metabolic activity of MV rats, for example, lowering
in abundance due to FLU administration was Lactobacillus, the abundance of genera Prevotella, Oscillospira and
which has been associated with body mass regulation (Lyte Ruminococcus. The latter genus was also identified as
and Brown, 2018). The authors suggest that depletion of decreased in abundance following administration of five
certain microbial genera due to FLU supplementation is different antidepressants by Lukic et al. (2019). The study
partially responsible for some of the known side effects used BALB/c mice as models as this strain is known for
such as weight loss (Lyte et al., 2019). An additional study having natural characteristics of exhibiting higher depres-
examined the effect of SSRIs on the microbiota of rats dur- sive-like behaviour and anxiety compared to other strains
ing pregnancy and lactation (Ramsteijn et al., 2018). The (Lukic et al., 2019). In follow-up experiments, BALB/c
study used female rat models of maternal vulnerability mice were treated with the antidepressant duloxetine with

Australian & New Zealand Journal of Psychiatry, 53(12)


1154 ANZJP Articles

or without a laboratory-prepared culture of Ruminococcus the absorption fraction is known (Supplementary File 1).
flavefaciens. It was found that supplementation of R. flave- Given that SSRIs are highly lipophilic and nearly com-
faciens reversed the behavioural symptoms observed fol- pletely absorbed (Hiemke and Härtter, 2000; Pawar et al.,
lowing the duloxetine administration, acting as an 2012), the absorption fraction was assumed to be 1.0.
anti-antidepressant. R. flavefaciens supplementation has Research findings of GI drug transit times for non-disinte-
also been shown to induce changes in cortical gene expres- grating radiolabelled tablets (Adkin et al., 1993; Davis
sion leading the authors to hypothesise a mechanism for et al., 1986) were used to estimate the proportion of drug
microbial regulation of antidepressant treatment efficiency that remains at different locations in the GIT. Estimate
(Lukic et al., 2019). concentrations have been calculated and are shown in
A limited number of observational studies in humans Figure 1, and the equations used are presented in
have examined SSRI use as a variable when comparing Supplementary File 1. These concentrations are the daily
subjects’ microbiome profiles. A large observational study theoretical maximum ([SSRI]max) and are based on the
(n > 2700, 113 taking SSRIs) examining the effects of 38 calculated amount remaining of the maximum daily dose
common diseases and 51 medications on the GM (Jackson (Figure 1, box 1; Hiemke and Härtter, 2000; van Harten,
et al., 2018) found negative associations between a number 1995; Wade et al., 2011) when completely dissolved in the
of taxa and SSRI use. In this study, depression did not sig- estimated average fluid volumes of different sections of
nificantly associate with the aforementioned taxa and SSRI the GIT. The calculations do not address potential cumu-
use had larger correlations with a greater number of relative lative effects of long-term drug exposure.
abundances. In a study of 76 elderly participants on polyp- The small intestine is home to a number of microbial
harmacy, antidepressant use was positively correlated with taxa (Donaldson et al., 2015). Figure 1, box 2 shows esti-
several taxa (Ticinesi et al., 2017). Investigation of the mates of remaining SSRI concentrations at a time that is
effects of NSAIDs on the microbiome found that citalo- estimated as being midway through small intestinal transit,
pram (CIT) was significantly associated with a 32% which combined gastric emptying and half of small intesti-
increase in abundance of a member of the Enterobacteriaceae nal transit (132 [±SD 60] minutes; Adkin et al., 1993;
family and found that SSRI use moderated the effects of Davis et al., 1986). Calculation of [SSRI]max used the calcu-
NSAIDs (Rogers and Aronoff, 2016). Studies (Vogt et al., lated mass of remaining drug and an average ileum fluid
2017; Zheng et al., 2016) found no correlation between volume of 31 mL (Sutton, 2009). Appreciable amounts of
microbiome structure and antidepressant use. However, as SSRIs are predicted as being present. However, greater pro-
these studies were not directly aimed at assessing the rela- portions of FLU, SER, PRX and FVX than ECIT and CIT
tionship between SSRI use and microbiome diversity, the are predicted as remaining (undissolved), which is consist-
conclusions drawn in this regard are limited by the number ent with these drugs taking less time to reach peak plasma
of participants and confounding factors. concentration (De Vane et al., 2002; Hiemke and Härtter,
2000; Jovanović et al., 2006; Qamar and Safila, 2015).
As the colon is home to the most abundant microbial pop-
Exposure of GM to SSRIs ulation, likely SSRI exposure was estimated for two points:
SSRIs are lipophilic and dissolve slowly (Filipski et al., arrival time and after 4 hours (Figure 1, boxes 3 and 4).
2013; Pawar et al., 2012; Seaman et al., 2005). In order to Concentrations were calculated based on a mean colonic
assess possible concentrations of SSRIs along the gastro- fluid volume of 11 mL (Schiller et al., 2005). Box 3 esti-
intestinal tract (GIT), evidence has been sourced from PK mates used the mean (±SD) time taken for 50% of a drug
and GI transit studies. We found PK research for a number to arrive at the colon of 210 (±111) minutes (Adkin et al.,
of SSRIs, including FLU, ECIT, sertraline (SER), parox- 1993) and 50% of the mass of the remaining drug. Box 4
etine (PRX), fluvoxamine (FVX) and CIT (Brown et al., shows estimates for 4 hours later and assumes that the
2005; van Gorp et al., 2012; Venkatakrishnan and Obach, entirety of the remaining drug is present in the colon.
2005). First-order rate constants for absorption, which Estimates indicate that substantial amounts of FLU, SER,
assume passive concentration-dependent absorption, PRX and FVX likely remain in the colon for at least 4 hours,
describe how rapidly a drug is absorbed as a function of ample time for these SSRIs to exert an antimicrobial effect.
the amount of drug yet to be absorbed (Mould and Upton, Finally, Figure 1, box 5 shows the proportion of excretion
2013). This is suitable given that SSRI uptake is a largely of SSRIs and their metabolites in the faeces, as identified
passive process (Pawar et al., 2012). Average absorption by mass balance studies (Hiemke and Härtter, 2000;
rate constants for the aforementioned SSRIs were those Lemberger et al., 1985; Qamar and Safila, 2015).
reported by Brown et al. (2005), van Gorp et al. (2012) The calculated [SSRI]max should be interpreted with cau-
and Venkatakrishnan and Obach (2005). These were then tion under the following limitations. (1) The calculations
used to estimate the amount of drug remaining after a assume total dissolution. However, the solubilities of
given amount of time. This can be estimated assuming hydrochloride salts of even the most lipophilic SSRIs SER
that the initial amount of drug at the absorption site and and FLU (4240 and 1100 mg/L, respectively; Childs et al.,

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1155

2004; Deak et al., 2006) would permit concentrations close The antibacterial activity of a drug can be described in
to estimates in Figure 1. In the small intestine, the dissolu- terms of its MIC against a given species and the concentra-
tion of SSRIs would be facilitated by bile salts (Mithani tions to which the species is likely exposed. According to the
et al., 1996) and colonic dissolution of SSRIs would likely Clinical Lab Standards Institute (CLSI), bacteria are classi-
be facilitated by colonic mechanical forces, phospholipids, fied as (1) susceptible to an agent if achievable concentra-
other fats and lectins (Shameem et al., 1995; Sutton, 2009). tions are above the MIC or (2) intermediate in susceptibility
(2) It should be noted that the PKs of SSRIs (De Vane et al., when only some isolates or strains are susceptible. When
2002; Hiemke and Härtter, 2000; Jovanović et al., 2006) attainable concentrations fall below the MIC, microbes are
and GI transit of medications (Adkin et al., 1993; Davis said to be (3) resistant (Patel, 2017). This terminology will
et al., 1986; Schiller et al., 2005) are highly variable. The be used when describing the efficacy of SSRIs as antimicro-
actual variability of gut concentrations of SSRIs in the pop- bials. The terms resistant, intermediate and susceptible are
ulation will far exceed the ranges reported in Figure 1. used in accordance with concentrations reported in Figure 1.
Overall, the main finding of this analysis is that the Many studies that determined MICs reported that standard
microbial community present in both the small intestine protocols were enforced (Ayaz et al., 2015b; Bohnert et al.,
and the colon is exposed to substantial concentrations of 2011) although some did not provide this information
SSRIs for a period of a minimum 4 hours on a daily basis. (Hendricks, 2007; Munoz-Bellido et al., 2000).
Given that the estimated average doubling time for microbes
in the mammalian gut is ~3 hours (Myhrvold et al., 2015),
Antibacterial activity
it is likely that this exposure will have a long-term effect on
the microbial population, in particular when considering The MICs for SSRIs sourced from the literature are shown
that SSRIs are taken daily and for a long period of time. in Table 1. For each SSRI, the table shows the range of
Furthermore, as demonstrated in Figure 1, box 5, metabo- MICs for tested species of Gram-positive, Gram-negative
lites of SSRIs are also passing through the GIT and may and acid-fast bacteria. The table indicates the number of
exert additional influence on the microbial population. strains upon which this was determined. The tested strains
Taken together, the information provided here offers cru- were either reference strains or clinical isolates.
cial context for the interpretation of in vitro assessments of Although these strains do not necessarily represent gut
the antimicrobial activity of SSRIs and their potential mod- microbes, a few general comments can be made with
ulation of the GM and implications for treatment efficacy. regards to the antimicrobial activity of SSRIs and the theo-
retical concentrations calculated in Figure 1. First and most
important, in the gut environment, many of the tested spe-
In vitro evidence for antimicrobial cies would be classified as susceptible or intermediate.
activity of SSRIs Importantly, this could remain the case for several hours.
Research analysed and methods The research therefore indicates that some SSRIs may exert
antimicrobial activity against gut microbes for several
The potential for the estimated concentrations of SSRIs in hours each day. The concentrations estimated in Figure 1
the gut to confer antimicrobial effect was estimated by indicate that the MICs of SER, FLU, PRX and FVX for a
reviewing the literature regarding antimicrobial effect of number of bacterial species would be possible in the ileum
SSRIs, as estimated by in vitro experiments. A number of in and ascending colon. The MICs listed in Table 1 indicate
vitro assays of the antimicrobial activity of SSRIs have that there is a substantial difference between different
been conducted over the last two decades. Much of this strains and species in susceptibility to SSRIs and show that
research involved determination of minimum inhibitory SER and FLU may have stronger inhibitory effects on
concentrations (MICs), which are considered to be the most Gram-positive bacteria than Gram-negative bacteria.
appropriate and comparable means of determining antimi- SSRIs differ in the degree to which they inhibit bacterial
crobial activity of a compound against a given strain, as growth. Examination of MICs for a variety of strains
determination involves adherence to a standardised proto- between different SSRIs indicates that SER and FLU have
col (Andrews, 2001). As such, these were targeted in litera- the strongest antimicrobial activity, followed by PRX and
ture searches. Searches were conducted on Google Scholar FVX, then ECIT and CIT (Bohnert et al., 2011; Hendricks,
and search terms included ‘Antimicrobial’, and the names 2007; Kalaycı et al., 2014; Munoz-Bellido et al., 2000). For
of the SSRIs ‘citalopram’, ‘escitalopram’, ‘fluoxetine’, SER, MIC ranges for all species fall below the mean
‘sertraline’, ‘paroxetine’ and ‘fluvoxamine’. The top 100 [SSRI]max estimate 4 hours after colonic arrival, indicating
results for each search were examined for MICs of SSRIs. that many microbes would be susceptible. This was further
The literature was also searched for research regarding supported by research involving disc diffusion assays. Disc
antifungal activity – particularly the determination of mini- diffusion assays involve placement of a drug-coated disc
mum fungicidal concentrations (MFCs), the fungal equiva- onto inoculated agar and incubation, thus allowing the size
lent to MICs (Lass-Flörl et al., 2001). of zone of microbial growth inhibition to be measured. The

Australian & New Zealand Journal of Psychiatry, 53(12)


1156 ANZJP Articles

Table 1. Minimum inhibitory concentrations (MICs) for different SSRIs against various bacteria.

SSRI Microbe MIC (mg/L) Reference


(no. strains or isolates tested) MIC

Sertraline
Gram-positive
Staphylococcus aureus (9) 10–60 Ayaz et al. (2015b)
(1) 4 Kalaycı et al. (2014)
(32) 4–32 Munoz-Bellido et al. (2000)
(1) 6.25 Kumar et al. (2015)
(10) 8–16 Hendricks (2007)
(1) 160 Kruszewska et al. (2004)
MRSA (1) 32 Kalaycı et al. (2014)
Staphylococcus epidermidis (4)16 Hendricks (2007)
(1) 4 Kalaycı et al. (2014)
(32) 4–16 Munoz-Bellido et al. (2000)
VRE (1) 8 Kalaycı et al. (2014)
Enterococcus faecalis (5) 16 Hendricks (2007)
(32) 2–16 Munoz-Bellido et al. (2000)
(1) 16 Kalaycı et al. (2014)
Streptococcus pyogenes (8)16 Hendricks (2007)
(32) 16–64 Munoz-Bellido et al. (2000)
Streptococcus pneumoniae (7) 16 Hendricks (2007)
(32) 16–64 Munoz-Bellido et al. (2000)
Bacillus subtilis (1) 8 Kalaycı et al. (2014)
(1) 6.25 Kumar et al. (2015)
Streptococcus agalactiae (32) 16–64 Munoz-Bellido et al. (2000)
Clostridium perfringens (32) 32–128 Munoz-Bellido et al. (2000)
Clostridium difficile (32) 32–128 Munoz-Bellido et al. (2000)
Corynebacterium group D2 2–8 Munoz-Bellido et al. (2000)
Gram-negative
Escherichia coli (6) 8.6–68.5 69
(16) 10–100 Ayaz et al. (2015b)
(1) 4 Kalaycı et al. (2014)
(32) 64–128 Munoz-Bellido et al. (2000)
(1) 50 Kumar et al. (2015)
Pseudomonas aeruginosa (1) 60 Ayaz et al. (2015b)
Proteus vulgaris (1) 16 Kalaycı et al. (2014)
(32) 128–512 Munoz-Bellido et al. (2000)
Klebsiella pneumoniae (1) 16 Kalaycı et al. (2014)
(32) 64–256 Munoz-Bellido et al. (2000)
Acinetobacter baumannii (1) 4 Kalaycı et al. (2014)
16
Yersinia enterocolitica (1) 16 Kalaycı et al. (2014)
Proteus mirabilis (32) 32–512 Munoz-Bellido et al. (2000)
Enterobacter cloacae (32) 64–128 Munoz-Bellido et al. (2000)

(continued)

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1157

Table 1. (Continued)

SSRI Microbe MIC (mg/L) Reference


(no. strains or isolates tested) MIC

Citrobacter freundii (32) 8–64 Munoz-Bellido et al. (2000)


Serratia marcescens (32) 256–(>512) Munoz-Bellido et al. (2000)
Morganella morganii (32) 256–512 Munoz-Bellido et al. (2000)
  Salmonella sp. (32) >512 Munoz-Bellido et al. (2000)
Haemophilus influenzae (32) 8–64 Munoz-Bellido et al. (2000)
  Moraxella catarrhalis (32) 4–16 Munoz-Bellido et al. (2000)
  Campylobacter jejuni (32) 2–32 Munoz-Bellido et al. (2000)
  Bacteroides fragilis (32) 16–32 Munoz-Bellido et al. (2000)
  Prevotella spp. (32) 16–64 Munoz-Bellido et al. (2000)
  Brucella spp. (32) 8–16 Munoz-Bellido et al. (2000)
Acid-fast
Mycobacterium tuberculosis (1) 2 Kalaycı et al. (2014)
(1) 1.6 Kumar et al. (2015)

Fluoxetine
Gram-positive
S. aureus (1) 4 Kalaycı et al. (2014)
(1) 12.5 Kumar et al. (2015)
MRSA (1) 128 Kalaycı et al. (2014)
S. epidermidis (1) 32 Kalaycı et al. (2014)
VRE (1) 16 Kalaycı et al. (2014)
E. faecalis (1) 32 Kalaycı et al. (2014)
B. subtilis (1) 4 Kalaycı et al. (2014)
(1) 12.5 Kumar et al. (2015)
Corynebacterium group D2 8–32 Munoz-Bellido et al. (2000)
Gram-negative
E. coli (6) 17.3–277 Bohnert et al. (2011)
(1) 16 Kalaycı et al. (2014)
(1) 100
P. vulgaris (1) 32 Kalaycı et al. (2014)
K. pneumonia (1) 32 Kalaycı et al. (2014)
A. baumannii (1) 32 Kalaycı et al. (2014)
Y. enterocolitica (1) 64 Kalaycı et al. (2014)
Acid-fast
M. tuberculosis (1) 8 Kalaycı et al. (2014)

Citalopram
Gram-positive
S. aureus (2) 4000–5000 Ayaz et al. (2015a)
(1) 16,000 Kruszewska et al. (2006)
(1) 13,000 Kruszewska et al. (2008)
(10) >256 Hendricks (2007)
(1) 256 Kalaycı et al. (2014)

(continued)

Australian & New Zealand Journal of Psychiatry, 53(12)


1158 ANZJP Articles

Table 1. (Continued)

SSRI Microbe MIC (mg/L) Reference


(no. strains or isolates tested) MIC

S. epidermidis (4) >256 Hendricks (2007)


E. faecalis (1) 7000 Ayaz et al. (2015a)
(5) >256
S. pyogenes (8) >256 Hendricks (2007)
S. pneumonia (7) >256 Hendricks (2007)
B. subtilis (1) 256 Kalaycı et al. (2014)
Gram-negative
E. coli (2) >800 µM Bohnert et al. (2011)
(2) 5000–7000 Ayaz et al. (2015a)
(1) 16,000 Kruszewska et al. (2006)
(1) 7000 Kruszewska et al. (2008)
(1) 256 Kalaycı et al. (2014)
P. aeruginosa (2) 4000–6000 Ayaz et al. (2015a)
Salmonella typhi (1) 6000 Ayaz et al. (2015a)
K. pneumonia (1) 6000 Ayaz et al. (2015a)
(1) 512 Kalaycı et al. (2014)
P. mirabilis (1) 6000 Ayaz et al. (2015a)
A. baumannii (1) 256 Kalaycı et al. (2014)

Fluvoxamine
Gram-positive
S. aureus (1) 4000 Kruszewska et al. (2010)
S. epidermidis (1) 64 Kalaycı et al. (2014)
B. subtilis (1) 64 Kalaycı et al. (2014)
Gram-negative
E. coli (1) 32 Kalaycı et al. (2014)
(1) 2000 Kruszewska et al. (2010)
P. aeruginosa (1) 10,000 Kruszewska et al. (2010)
P. vulgaris (1) 64 Kalaycı et al. (2014)
K. pneumoniae (1) 32 Kalaycı et al. (2014)
A. baumannii (1) 128 Kalaycı et al. (2014)
Y. enterocolitica (1) 256 Kalaycı et al. (2014)

Femoxetine
Gram-positive
S. aureus (1) 1000 Kumar et al. (2015)

Paroxetine
Gram-positive
S. aureus (1) 128 Kalaycı et al. (2014)
(1) 25 Kumar et al. (2015)
MRSA (1) 128 Kalaycı et al. (2014)
S. epidermidis (1) 32 Kalaycı et al. (2014)
VRE (1) 32 Kalaycı et al. (2014)

(continued)

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1159

Table 1. (Continued)

SSRI Microbe MIC (mg/L) Reference


(no. strains or isolates tested) MIC
E. faecalis (1) 64 Kalaycı et al. (2014)
B. subtilis (1) 256 Kalaycı et al. (2014)
(1) 25 Kumar et al. (2015)
Corynebacterium group D2 4–32 Munoz-Bellido et al. (2000)
Gram-negative
E. coli (6) 18.3–293 Bohnert et al. (2011)
(1) 128 Kalaycı et al. (2014)
(1) 100
P. vulgaris (1) 32 Kalaycı et al. (2014)
K. pneumoniae (1) 64 Kalaycı et al. (2014)
A. baumannii (1) 64 Kalaycı et al. (2014)
Y. enterocolitica (1) 64 Kalaycı et al. (2014)
Acid-fast
M. tuberculosis (1) 8 Kalaycı et al. (2014)

Escitalopram
Gram-positive
B. subtilis (1) 256 Kalaycı et al. (2014)
Gram-negative
P. vulgaris (1) 128 Kalaycı et al. (2014)
K. pneumoniae (1) 256 Kalaycı et al. (2014)
A. baumannii (1) 128 Kalaycı et al. (2014)
Y. enterocolitica (1) 512 Kalaycı et al. (2014)

SSRI: serotonin reuptake inhibitor; MIC: minimal inhibitory concentration; MRSA: methicillin-resistant Staphylococcus aureus; TCA: tricyclic
antidepressant; VRE: vancomycin-resistant Enterococcus.

authors demonstrated differences in the number of organ- have important roles in gut microbial ecology, antifungal
isms inhibited by different SSRIs. They found that SER, activity of SSRIs may also affect gut ecology. Particularly,
FLU and PRX affected the most species (12/13), followed Candida albicans interacts both with the host – through pro-
by FVX (7/13), and CIT and ECIT (5/13; Kalaycı et al., duction of immunomodulatory compounds – and bacterial
2014). symbionts (Huffnagle and Noverr, 2013).
The variability in MICs reported for different strains and
species may be an important factor in the gut, as intermicrobe
differences in inhibition would facilitate a change in commu- Antifungal activity
nity structure. Taxonomically, some assayed bacteria SSRIs exhibit some antifungal activity, which is generally
belonged to genera that are commonly associated with dys- weaker than their antibacterial activity. In assays of antifun-
biosis, including Enterococcus, Streptococcus, Lacto­bacillus, gal activity, fewer species and strains have been tested,
Clostridium, Bifidobacterium, Prevotella, Bac­teroides and from fewer genera. The MFCs found were tabulated and
Escherichia (Rojo et al., 2017). In regards to specific spe- can be found in Table 2. The inhibitory strength of different
cies, MICs indicate that Bacillus subtilis and Bacteroides SSRIs follows a similar trend in fungi as in bacteria. Studies
fragilis, which are commensals implicated in development of by Lass-Flörl et al. (2001) and Kalaycı et al. (2014) indicate
gut-associated lymphoid tissue (GALT; Rhee et al., 2004), that SER, FLU and PRX may have antifungal activity
could be inhibited by SER or FLU. Gut concentrations may against some strains at the estimated concentrations. The
also inhibit Escherichia coli but be less inhibitory to overall effect of SSRIs on fungal physiology is not fully
Clostridium species, both of which may be found in the gut understood; however, this could be an important direction
(Atarashi et al., 2011; Eckburg et al., 2005). As Candida spp. for future studies.

Australian & New Zealand Journal of Psychiatry, 53(12)


1160 ANZJP Articles

Table 2. Minimum fungicidal concentrations for different SSRIs against various species of fungi.

SSRI Microbe MFC(mg/L) Reference


(no. strains or isolates tested) MFC

Sertraline
Aspergillus fumigatus (10) 7–327 Lass-Flörl et al. (2001)a
(5) 80–100 Ayaz et al. (2015b)
Aspergillus niger (5) 20–80 Ayaz et al. (2015b)
(1) 64 Kalaycı et al. (2014)
Aspergillus flavus (5) 60–80 Ayaz et al. (2015b)
(9)14–475 Lass-Flörl et al. (2001)a
Aspergillus terreus (10) 29–237 Lass-Flörl et al. (2001)a
Candida parapsilosis (1) 59 Lass-Flörl et al. (2001)a
Candida albicans (1) 128 Kalaycı et al. (2014)
(1) 25 Kumar et al. (2015)
Fusarium solani (1) 80 Ayaz et al. (2015b)

Fluoxetine
A. fumigatus (10) 78–1250 Lass-Flörl et al. (2001)a
A. flavus (9) 156–2500 Lass-Flörl et al. (2001)a
A. terreus (10) 59–2500 Lass-Flörl et al. (2001)a
C. parapsilosis (1) 78–156 Lass-Flörl et al. (2001)a
C. albicans

Paroxetine
A. fumigatus (10) 62–2500 Lass-Flörl et al. (2001)a
A. flavus (9) 62–1000 Lass-Flörl et al. (2001)a
A. terreus (10) 500–1000 Lass-Flörl et al. (2001)a
C. parapsilosis (1) 500 Lass-Flörl et al. (2001)a
C. albicans (1) 25 Kumar et al. (2015)

Citalopram
A. fumigatus (10) 312–1250 Lass-Flörl et al. (2001)a
A. flavus (9) 312–2500 Lass-Flörl et al. (2001)a
A. terreus (10) 312–1250 Lass-Flörl et al. (2001)a
C. parapsilosis (1) 1250 Lass-Flörl et al. (2001)a
C. albicans

SSRI: serotonin reuptake inhibitor; MIC: minimal inhibitory concentration; MFC: minimum fungicidal concentrations.
a8-hour inhibition.

Antimicrobial mode of action are more hydrophobic (logP, 5.1, 4.1) than ECIT, CIT, PRX
and FVX (logP, 3.5, 3.5, 3.3, 3.6; Kim et al., 2016). SER
Intracellular effects and FLU may undergo passive diffusion across the phos-
In humans, SSRIs exert their effects on extracellular seroto- pholipid membrane with more ease, allowing interaction
nin levels by competitively inhibiting the serotonin trans- with cellular machinery (Hancock and Bell, 1989). This
porter (SERT or solute carrier family 6 member 4 hypothesis is consistent with research indicating that the
[SCL6A4]). However, the mechanism of action on microbes optimum lipophilicity for an antimicrobial is logP 5.5
is less understood. As discussed, FLU and SER may have (Wernert et al., 2004). One possible intracellular effect is
stronger antimicrobial action than other SSRIs. This could increased production of reactive oxygen species (ROS) and
be informative with respect to mechanism. As determined mutagenesis, as has been demonstrated with FLU and
by octanol–water partition coefficients (P), SER and FLU E. coli than FLU (Jin et al., 2018; Figure 2). While ROS

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1161

Figure 2. Potential antimicrobial mechanisms of SSRIs. SSRIs may gain access to the cytoplasm via passive diffusion. They may
inhibit microbial efflux systems and reduce excretion of antimicrobials, thereby reducing antimicrobial resistance. They may
also inhibit sodium-dependent symporters, including amino acid transporters and transporters for which substrates are not
yet known. This may affect nutrient intake and excretion of waste products and metabolites. SSRI, selective serotonin reuptake
inhibitors; NSS, neurotransmitter sodium symporter family; ROS, reactive oxygen species.

may mediate the effects of SSRIs on growth, the exact Sodium-dependent transport
mechanisms remain unclear.
The action of antidepressants on SERT may offer insights into
the potential mechanism of antimicrobial function. SSRIs
competitively inhibit SERT by binding at the serotonin bind-
Inhibition of efflux pumps
ing site and preventing conformational change (Grouleff
SSRIs have been shown to inhibit microbial efflux pumps, et al., 2016; Singh et al., 2007). Considering the large homol-
which contribute to antimicrobial resistance (Munoz- ogy between SERT and bacterial amino acid transporters
Bellido et al., 2000). Efflux systems extrude antimicrobials (Grouleff et al., 2016; Singh, 2014; Yamashita et al., 2005)
and other drugs from the cytoplasm. While the efflux sys- and the role these play in metabolism (Burkovski and Krämer,
tems are mainly associated with the expulsion of xenobiot- 2002), it is plausible that SSRIs exert similar effects in
ics, other physiological roles, such as in biofilm formation microbes and prevent the activity of amino acid transporters,
and virulence of some enteric pathogens, have been pro- leading to a deleterious effect on growth and survival.
posed (Opperman and Nguyen, 2015). Mechanisms of In order to ascertain the scope of potential targets for
efflux inhibition include competitive and non-competitive SSRIs in bacteria, a BLAST search (Altschul et al., 1990)
inhibition and clogging of the pump’s external pore (Askoura for the 100 most similar bacterial proteins in National
et al., 2011). Both SER and PRX were found to inhibit efflux Center for Biotechnology Information (NCBI) protein
pump activity in E. coli, Pseudomonas aeruginosa and database (Coordinators, 2018) was conducted for homo-
Staphylococcus aureus at concentrations below MIC logues of SERT. A total of 100 orthologues (E < 0.05) were
(Bohnert et al., 2011; Kaatz et al., 2003). Interestingly, the identified across a broad variety of bacterial species. All of
observed SSRI efflux inhibition was not associated with which were uncharacterized putative sodium-dependent
direct growth or survival inhibition (Bohnert et al., 2011). transporters. A multiple sequence alignment (Edgar, 2004;
Nonetheless, efflux inhibition may act in concert to promote Kumar et al., 2016) indicated that several important resi-
the activity of other antimicrobials (Bohnert et al., 2011). A dues in LeuT/SERT were conserved in all of these proteins.
number of studies have assessed the synergism between These included R30/104 and D404/E493, which are gating
SSRIs and various classes of antibiotics and have found residues, critical in the transport cycle (Grouleff et al.,
that, in some strains, the addition of SSRIs substantially 2016) and S355/438, a determinant of antidepressant bind-
reduced the antibiotic MICs by 2–8-fold (Ayaz et al., 2015b; ing affinity (Andersen et al., 2009). Therefore, there may be
Bohnert et al., 2011). This is suggestive of an indirect anti- a large number of functionally similar transporters to which
microbial mechanism of SSRIs (Figure 2). SSRIs could bind to and inhibit. As the substrates of these

Australian & New Zealand Journal of Psychiatry, 53(12)


1162 ANZJP Articles

transporters have not yet been determined, inhibition may (Schemann and Neunlist, 2004). Changes to motility and
have any number of metabolic consequences. Sodium- the mucosal environment can subsequently affect the GM
dependent transport needs to be investigated with respect to (Carabotti et al., 2015). SSRIs also affect the immune sys-
SSRI antimicrobial properties (Figure 2). tem. SERT expression is increased in activation of both
dendritic and B cells, and SSRIs have been shown to stimu-
Sub-inhibitory concentrations late natural killer (NK) cells (Ahern, 2011), inhibit B cells
apoptosis (Arreola et al., 2015) and promote the Th1, over
Given the wide variation in the efficacy of SSRIs between the Th2, response (Martino et al., 2012). This may have
species and strains, and that concentrations of SSRIs in the implications for gut microbes. Interaction with the immune
gut will fluctuate, the effect of sub-inhibitory concentra- system and gut modulation may therefore be another means
tions (<MIC) should be considered. Research has indicated by which SSRIs may manipulate the microbiome.
that most antimicrobials have a biphasic effect across a sub- Finally, SSRI treatment of anxiety and mood disorders
inhibitory concentration range (Davies et al., 2006). To this may indirectly impact the GM through brain to gut com-
effect, low concentrations are often stimulatory to growth munication. As research has indicated that stress and
and high concentrations become progressively more inhibi- depression have implications for microbial diversity
tory (Davies et al., 2006). The stimulatory, or hormetic, (Winter et al., 2018), it is probable that alleviation of symp-
effect on microbes is thought to be due to pro-survival toms would also impact the microbiota. Reduced stress
changes that at low concentrations outweigh negative may result in decreased sympathetic tone, which can affect
effects. This involves transcriptional alteration of around the GM, as GALT is directly sympathetically innervated
5–10% of transcripts including upregulation of pro-survival (Powell et al., 2017). Furthermore, some studies have found
and pro-growth factors (e.g. heat-shock proteins and pro- that SSRIs modulate hypothalamic pituitary adrenal (HPA)
teins involved in replication; Davies et al., 2006; Yim et al., axis activity. Research has identified both positive and neg-
2007). Transcriptional changes may occur at as little as 1% ative changes in plasma and salivary adrenocorticotropic
of the MIC (Yim et al., 2007). It is possible that at gut con- hormone (ACTH) and cortisol following treatment with
centrations of SSRIs some species would undergo hormetic different SSRIs (Glintborg et al., 2018; Nikisch et al., 2005)
changes while others would be inhibited. – although this is not always observed (Frase et al., 2018).
Cortisol modulation of the immune system may also facili-
tate a host-mediated effect of SSRIs on the GM (Powell
Potential frameworks for SSRIs
et al., 2017).
modulation of the gut microbial
community
In addition to their direct inhibitory activity, SSRIs may Implications for treatment efficacy
affect gut ecology indirectly by enhancing the activity of The effect of SSRIs on GM community composition may
other antimicrobial agents, for example, by inhibition of have implications for their treatment efficacy. While there
Efflux pump. Antibiotics are in common use, and evidence are no experimental data yet available to support this, there is
indicates that a baseline exposure to antimicrobials in house- evidence that antimicrobial-induced dysbiosis can have con-
hold products is sufficient to affect the GM (Yee and Gilbert, sequences for the brain and behaviour, in rodents. Most sig-
2016). Therefore, SSRIs may synergize with these antimi- nificantly, several studies identified increased depression-like
crobials to have a greater effect on microbial diversity. In behaviour in adult-treated rats and mice (Guida et al., 2018;
addition, SSRIs may also interact with gut microbial ecology Hoban et al., 2016) and decreased anxiety-like behaviour in
through their metabolic by-products, which are excreted into prenatally- and adolescent-treated mice (Desbonnet et al.,
the faeces (Hiemke and Härtter, 2000). Furthermore, gut 2015; Leclercq et al., 2017). It is important to note that
microbes may themselves metabolise SSRIs. For instance, some antimicrobials do have direct action on the brain
FLU has been found to be metabolised by a number of gut (Macedo et al., 2017). However, Guida et al. (2018) did
microbe strains in vitro (Zimmermann et al., 2019). While control for these direct effects by using an intraperitoneal
the effect of SSRI metabolites on gut microbes have not been injection control group. These studies show that inducing
characterised, they could impact the dynamic metabolic net- changes in GM composition can influence anxiety and
work in the gut and alter GM composition and function. depression-like behaviour and thus provide an empirical
Another avenue to consider as a potential effect of SSRIs basis for investigating the effect of SSRIs on the GM and
on the GM may involve a host-mediated component. SSRIs how this interacts with treatment efficacy.
may directly interact with gut cells and/or immune cells to There are a number of ways in which SSRI-mediated
modulate the gut environment (Mawe and Hoffman, 2013). dysbiosis could theoretically interfere with treatment effi-
One application might be altered availability of gut seroto- cacy. For instance, in some rodent studies antibiotic-
nin which could affect a large proportion of enteric neurons induced dysbiosis has been shown to reduce hippocampal
and alter peristalsis and reflex electrolyte secretion levels of brain-derived neurotrophic factor (BDNF;

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1163

Desbonnet et al., 2015; Frohlich et al., 2016; Guida et al., individuals, and in the magnitude and nature of the effects
2018), which is known to promote adult hippocampal neu- of antimicrobial effects of SSRIs, this may account for
rogenesis (Ortega-Martinez, 2015). Given the evidence that some of the interpersonal variabilities in treatment effi-
adult hippocampal neurogenesis may be necessary for the cacy with SSRIs.
action of a number of antidepressants (Sahay and Hen, Here, we have highlighted the need for research inves-
2007), GM-mediated changes in BDNF may be one means tigating the role of the gut–brain axis in the sporadic and
by which the antimicrobial action of SSRIs could affect unpredictable treatment efficacy of SSRIs. Future research
treatment. Gut-mediated changes in BDNF may be brought should attempt to establish the antimicrobial activity of
about by vagal nerve signalling (O’Leary et al., 2018) or SSRIs against gut microbes both in vitro and in vivo and
through changes in the production of metabolites such as determine the effect this activity has on treatment efficacy.
butyrate, which is known to increase BDNF and exert anti- Considering the significant role that gut microorganisms
depressant-like activity (Stilling et al., 2016). In addition, are discovered to play in human physiology, it is predicted
there could be gut-mediated effects on the serotonin sys- by the authors that this type of investigation will form a
tem. Antibiotic-induced dysbiosis has been shown to reduce routine assessment of drug efficacy in the future.
hippocampal serotonin (Hoban et al., 2016), and GM com-
position has implications for serotonin metabolism Acknowledgements
(O’Mahony et al., 2015). Given that SSRIs are milder anti- The authors thank Professor Natkunam Ketheesan for critically
microbials than antibiotics, SSRI-mediated GM changes reviewing the manuscript.
could, theoretically, interact positively or negatively with
drug efficacy, depending on which microbes were advan- Declaration of conflicting interests
taged or disadvantaged by the selection pressure. The author(s) declared no potential conflicts of interest with
respect to the research, authorship and/or publication of this
article.
Conclusion and future directions
In conclusion, there is substantial evidence that SSRIs may Funding
affect the community structure of the gut microbiome. The author(s) received no financial support for the research,
While there are no direct investigations of SSRI-induced gut authorship and/or publication of this article.
dysbiosis in humans, there is a considerable amount of
research investigating the antimicrobial properties of SSRIs. ORCID iDs
When this information is framed within the context of the
Abigail S McGovern https://orcid.org/0000-0002-9641-2768
amount of SSRIs likely remaining in the gut, assessed based
Gal Winter https://orcid.org/0000-0003-3789-395X
on findings of PK and GI transit studies, it is probable that
many microbes may be susceptible to SSRIs. Specifically,
SER, FLU and PRX may act as antimicrobials in the gut; Supplemental material
however, differences in absorption and GI transit could lead Supplemental material for this article is available online.
to interpersonal variability in degree of susceptibility to dif-
ferent drugs. Given the complex interactions that determine References
GM community structure, it is likely that these SSRIs would Abbing-Karahagopian V, Huerta C, Souverein PC, et al. (2014)
act as a persistent pressure, altering the stable state of the gut Antidepressant prescribing in five European countries: Application
microbiome. They may cause both increases and decreases of common definitions to assess the prevalence, clinical observa-
in the relative abundances of different gut taxa; given inter- tions, and methodological implications. European Journal of Clinical
Pharmacology 70: 849–857.
individual variability in gut microbiome structure, this effect Adkin D, Davis S, Sparrow R, et al. (1993) Colonic transit of different
would likely differ between individuals. sized tablets in healthy subjects. Journal of Controlled Release 23:
If SSRIs impact the GM, research suggests that they 147–156.
have the potential to interfere with treatment efficacy. Ahern GP (2011) 5-HT and the immune system. Current Opinion in
Studies of the behavioural and neurobiological effects of Pharmacology 11: 29–33.
Altschul SF, Gish W, Miller W, et al. (1990) Basic local alignment search
antibiotic-induced dysbiosis give insight into potential tool. Journal of Molecular Biology 215: 403–410.
means by which this may occur. Induced dysbiosis may Andersen J, Taboureau O, Hansen KB, et al. (2009) Location of the anti-
affect the serotoninergic system (Desbonnet et al., 2015), depressant binding site in the serotonin transporter: Importance of
which is the initial target of SSRIs, as well as adult hip- Ser-438 in recognition of citalopram and tricyclic antidepressants.
pocampal neurogenesis, which is thought to be an indi- Journal of Biological Chemistry 284: 10276–10284.
Andrews JM (2001) Determination of minimum inhibitory concentra-
rect target of SSRIs (Möhle et al., 2016). Given the tions. Journal of Antimicrobial Chemotherapy 48: 5–16.
complexity of the gut microbiome, there could be poten- Arreola R, Becerril-Villanueva E, Cruz-Fuentes C, et al. (2015)
tial for both positive and negative effects on treatment Immunomodulatory effects mediated by serotonin. Journal of
efficacy. Due to differences in the microbiomes of Immunology Research 2015: 354957.

Australian & New Zealand Journal of Psychiatry, 53(12)


1164 ANZJP Articles

Askoura M, Mattawa W, Abujamel T, et al. (2011) Efflux pump inhibitors Eisen JA, Dethlefsen L, Huse S, et al. (2008) The pervasive effects of
(EPIs) as new antimicrobial agents against Pseudomonas aeruginosa. an antibiotic on the human gut microbiota, as revealed by deep 16S
Libyan Journal of Medicine 6: 5870. rRNA Sequencing. PLoS Biology 6: e280.
Atarashi K, Tanoue T, Shima T, et al. (2011) Induction of colonic regu- Filipski KJ, Varma MV, El-Kattan AF, et al. (2013) Intestinal targeting of
latory T cells by indigenous Clostridium species. Science 331: drugs: Rational design approaches and challenges. Current Topics in
337–341. Medicinal Chemistry 13: 776–802.
Ayaz M, Subhan F, Ahmed J, et al. (2015a) Citalopram and venlafaxine Flowers SA, Evans SJ, Ward KM, et al. (2017) Interaction between atypi-
differentially augments antimicrobial properties of antibiotics. Acta cal antipsychotics and the gut microbiome in a bipolar disease cohort.
Poloniae Pharmaceutica: Drug Research 72: 1269–1278. Pharmacotherapy 37: 261–267.
Ayaz M, Subhan F, Ahmed J, et al. (2015b) Sertraline enhances the activ- Forslund K, Hildebrand F, Nielsen T, et al. (2015) Disentangling type 2
ity of antimicrobial agents against pathogens of clinical relevance. diabetes and metformin treatment signatures in the human gut micro-
Journal of Biological Research 22: 4. biota. Nature 528: 262–266.
Bohnert JA, Szymaniak-Vits M, Schuster S, et al. (2011) Efflux inhibition Frase L, Doerr JP, Feige B, et al. (2018) Different endocrine effects of an
by selective serotonin reuptake inhibitors in Escherichia coli. Journal evening dose of amitriptyline, escitalopram, and placebo in healthy par-
of Antimicrobial Chemotherapy 66: 2057–2060. ticipants. Clinical Psychopharmacology and Neuroscience 16: 253–261.
Bonaz B, Bazin T and Pellissier S (2018) The vagus nerve at the interface Frohlich EE, Farzi A, Mayerhofer R, et al. (2016) Cognitive impairment
of the microbiota-gut-brain axis. Frontiers in Neuroscience 12: 49. by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain
Brett J, Karanges EA, Daniels B, et al. (2017) Psychotropic medication communication. Brain, Behavior, and Immunity 56: 140–155.
use in Australia, 2007 to 2015: Changes in annual incidence, preva- Fung TC, Olson CA and Hsiao EY (2017) Interactions between the micro-
lence and treatment exposure. Australian and New Zealand Journal biota, immune and nervous systems in health and disease. Nature
of Psychiatry 51: 990–999. Neuroscience 20: 145–155.
Brown HS, Ito K, Galetin A, et al. (2005) Prediction of in vivo drug- Glintborg D, Altinok ML, Ravn P, et al. (2018) Adrenal activity and meta-
drug interactions from in vitro data: Impact of incorporating parallel bolic risk during randomized escitalopram or placebo treatment in
pathways of drug elimination and inhibitor absorption rate constant. PCOS. Endocrine Connections 7: 479–489.
British Journal of Clinical Pharmacology 60: 508–518. Grouleff J, Koldsø H, Miao Y, et al. (2016) Ligand binding in the extracel-
Burkovski A and Krämer R (2002) Bacterial amino acid transport proteins: lular vestibule of the neurotransmitter transporter homologue LeuT.
Occurrence, functions, and significance for biotechnological applica- ACS Chemical Neuroscience 8: 619–628.
tions. Applied Microbiology and Biotechnology 58: 265–274. Guida F, Turco F, Iannotta M, et al. (2018) Antibiotic-induced microbiota
Carabotti M, Scirocco A, Maselli MA, et al. (2015) The gut-brain axis: perturbation causes gut endocannabinoidome changes, hippocampal
Interactions between enteric microbiota, central and enteric nervous neuroglial reorganization and depression in mice. Brain, Behavior,
systems. Annals of Gastroenterology 28: 203. and Immunity 67: 230–245.
Childs SL, Chyall LJ, Dunlap JT, et al. (2004) Crystal engineering Hancock RE and Bell A (1989) Antibiotic Uptake into Gram-negative
approach to forming cocrystals of amine hydrochlorides with organic Bacteria: Perspectives in Antiinfective Therapy. New York: Springer,
acids. Molecular complexes of fluoxetine hydrochloride with ben- pp. 42–53.
zoic, succinic, and fumaric acids. Journal of the American Chemical Hendricks O (2007) Antimicrobial effects of selected non-antibiotics
Society 126: 13335–13342. on sensitivity and invasion of gram-positive bacteria. PhD Thesis,
Coordinators NR (2018) Database resources of the national center for bio- University of Southern Denmark, Odense.
technology information. Nucleic Acids Research 46: D8–D13. Hiemke C and Härtter S (2000) Pharmacokinetics of selective serotonin
Coyte KZ, Schluter J and Foster KR (2015) The ecology of the micro- reuptake inhibitors. Pharmacology & Therapeutics 85: 11–28.
biome: Networks, competition, and stability. Science 350: 663–666. Hoban AE, Moloney RD, Golubeva AV, et al. (2016) Behavioural and neu-
Cryan JF and Dinan TG (2019) Talking about a microbiome revolution. rochemical consequences of chronic gut microbiota depletion during
Nature Microbiology 4: 552–553. adulthood in the rat. Neuroscience 339: 463–477.
Cussotto S, Strain CR, Fouhy F, et al. (2019) Differential effects of psy- Huffnagle GB and Noverr MC (2013) The emerging world of the fungal
chotropic drugs on microbiome composition and gastrointestinal microbiome. Trends in Microbiology 21: 334–341.
function. Psychopharmacology 236: 1671–1685. Imhann F, Bonder MJ, Vila AV, et al. (2016) Proton pump inhibitors affect
David LA, Maurice CF, Carmody RN, et al. (2014) Diet rapidly and repro- the gut microbiome. Gut 65: 740–748.
ducibly alters the human gut microbiome. Nature 505: 559–563. Isaac S, Scher JU, Djukovic A, et al. (2017) Short- and long-term effects
Davies J, Spiegelman GB and Yim G (2006) The world of subinhibitory of oral vancomycin on the human intestinal microbiota. Journal of
antibiotic concentrations. Current Opinion in Microbiology 9: 445– Antimicrobial Chemotherapy 72: 128–136.
453. Jackson MA, Goodrich JK, Maxan M-E, et al. (2016) Proton pump
Davis S, Hardy J and Fara J (1986) Transit of pharmaceutical dosage inhibitors alter the composition of the gut microbiota. Gut 65:
forms through the small intestine. Gut 27: 886–892. 749–756.
De Vane CL, Liston HL and Markowitz JS (2002) Clinical pharmacokinet- Jackson MA, Verdi S, Maxan ME, et al. (2018) Gut microbiota associa-
ics of sertraline. Clinical Pharmacokinetics 41: 1247–1266. tions with common diseases and prescription medications in a popu-
Deak K, Takacs-Novak K, Tihanyi K, et al. (2006) Physico-chemical pro- lation-based cohort. Nature Communications 9: 2655.
filing of antidepressive sertraline: Solubility, ionisation, lipophilicity. Jeon SW and Kim YK (2016) Molecular neurobiology and promising new
Medicinal Chemistry 2: 385–389. treatment in depression. International Journal of Molecular Sciences
Desbonnet L, Clarke G, Traplin A, et al. (2015) Gut microbiota depletion 17: 381.
from early adolescence in mice: Implications for brain and behaviour. Jin M, Lu J, Chen Z, et al. (2018) Antidepressant fluoxetine induces
Brain, Behavior, and Immunity 48: 165–173. multiple antibiotics resistance in Escherichia coli via ROS-mediated
Donaldson GP, Lee SM and Mazmanian SK (2015) Gut biogeography of mutagenesis. Environment International 120: 421–430.
the bacterial microbiota. Nature Reviews Microbiology 14: 20–32. Jovanović D, Kilibarda V, Dordević S, et al. (2006) Bioequivalence testing
Eckburg PB, Bik EM, Bernstein CN, et al. (2005) Diversity of the human of a new tablet formulation of generic fluoxetine. European Journal
intestinal microbial flora. Science 308: 1635–1638. of Drug Metabolism and Pharmacokinetics 31: 35–40.
Edgar RC (2004) MUSCLE: Multiple sequence alignment with high accu- Kaatz GW, Moudgal VV, Seo SM, et al. (2003) Phenylpiperidine selec-
racy and high throughput. Nucleic Acids Research 32: 1792–1797. tive serotonin reuptake inhibitors interfere with multidrug efflux

Australian & New Zealand Journal of Psychiatry, 53(12)


McGovern et al. 1165

pump activity in Staphylococcus aureus. International Journal of tonin/norepinephrine balance and Th1/Th2 balance. Current
Antimicrobial Agents 22: 254–261. Neuropharmacology 10: 97–123.
Kalaycı S, Demirci S and Sahin F (2014) Antimicrobial properties of vari- Mawe GM and Hoffman JM (2013) Serotonin signalling in the gut:
ous psychotropic drugs against broad range microorganisms. Current Functions, dysfunctions and therapeutic targets. Nature Reviews
Psychopharmacology 3: 195–202. Gastroenterology & Hepatology 10: 473–486.
Kasubuchi M, Hasegawa S, Hiramatsu T, et al. (2015) Dietary gut micro- Mehta RS, Abu-Ali GS, Drew DA, et al. (2018) Stability of the human
bial metabolites, short-chain fatty acids, and host metabolic regula- faecal microbiome in a cohort of adult men. Nature Microbiology 3:
tion. Nutrients 7: 2839–2849. 347–355.
Keller MB, Trivedi MH, Thase ME, et al. (2007) The prevention of Mithani SD, Bakatselou V, TenHoor CN, et al. (1996) Estimation of the
recurrent episodes of depression with venlafaxine for two years increase in solubility of drugs as a function of bile salt concentration.
(PREVENT) study: Outcomes from the acute and continuation Pharmaceutical Research 13: 163–167.
phases. Biological Psychiatry 62: 1371–1379. Möhle L, Mattei D, Heimesaat Markus M, et al. (2016) Ly6Chi monocytes
Kim S, Thiessen PA, Bolton EE, et al. (2016) PubChem substance and provide a link between antibiotic-induced changes in gut microbiota
compound databases. Nucleic Acids Research 44: D1202–1213. and adult hippocampal neurogenesis. Cell Reports 15: 1945–1956.
Kruszewska H, Zareba T and Tyski S (2004) Examination of antimi- Moloney RD, Desbonnet L, Clarke G, et al. (2014) The microbiome:
crobial activity of selected non-antibiotic drugs. Acta Poloniae Stress, health and disease. Mammalian Genome 25: 49–74.
Pharmaceutica 61: 18–21. Mould DR and Upton RN (2013) Basic concepts in population modeling,
Kruszewska H, Zareba T and Tyski S (2006) Estimation of antimicro- simulation, and model-based drug development – part 2: Introduction
bial activity of selected non-antibiotic products. Acta Poloniae to pharmacokinetic modeling methods. CPT: Pharmacometrics &
Pharmaceutica 63: 457–460. Systems Pharmacology 2: e38.
Kruszewska H, Zareba T and Tyski S (2008) Examination of antibacte- Moya A and Ferrer M (2016) Functional redundancy-induced stability of
rial and antifungal activity of selected non-antibiotic products. Acta gut microbiota subjected to disturbance. Trends in Microbiology 24:
Poloniae Pharmaceutica: Drug Research 65: 779–782. 402–413.
Kruszewska H, Zareba T and Tyski S (2010) Examination of antimi- Mrazek DA, Hornberger JC, Altar CA, et al. (2014) A review of the clini-
crobial activity of selected non-antibiotic products. Acta Poloniae cal, economic, and societal burden of treatment-resistant depression:
Pharmaceutica 67: 733–736. 1996–2013. Psychiatric Services 65: 977–987.
Kumar MMK, Madhavi K, Varma PR, et al. (2015) Shape based virtual Munoz-Bellido J, Munoz-Criado S and Garcıa-Rodrıguez J (2000)
screening and discovery of potential antitubercular activity in ‘Non- Antimicrobial activity of psychotropic drugs: Selective serotonin
antibiotics’. International Journal of Drug Design and Discovery reuptake inhibitors. International Journal of Antimicrobial Agents 14:
6(1): 1430–1434. 177–180.
Kumar S, Stecher G and Tamura K (2016) MEGA7: Molecular evolu- Myhrvold C, Kotula JW, Hicks WM, et al. (2015) A distributed cell divi-
tionary genetics analysis version 7.0 for bigger datasets. Molecular sion counter reveals growth dynamics in the gut microbiota. Nature
Biology and Evolution 33: 1870–1874. Communications 6: 10039.
Kundu P, Blacher E, Elinav E, et al. (2017) Our gut microbiome: The Nikisch G, Mathe AA, Czernik A, et al. (2005) Long-term citalopram
evolving inner self. Cell 171: 1481–1493. administration reduces responsiveness of HPA axis in patients with
Lass-Flörl C, Dierich M, Fuchs D, et al. (2001) Antifungal properties of major depression: Relationship with S-citalopram concentrations
selective serotonin reuptake inhibitors against Aspergillus species in in plasma and cerebrospinal fluid (CSF) and clinical response.
vitro. Journal of Antimicrobial Chemotherapy 48: 775–779. Psychopharmacology 181: 751–760.
Leclercq S, Mian FM, Stanisz AM, et al. (2017) Low-dose penicillin in O’Leary OF, Ogbonnaya ES, Felice D, et al. (2018) The vagus nerve
early life induces long-term changes in murine gut microbiota, brain modulates BDNF expression and neurogenesis in the hippocampus.
cytokines and behavior. Nature Communications 8: 15062. European Neuropsychopharmacology 28: 307–316.
Lemberger L, Bergstrom R, Wolen R, et al. (1985) Fluoxetine: Clinical O’Mahony SM, Clarke G, Borre YE, et al. (2015) Serotonin, tryptophan
pharmacology and physiologic disposition. The Journal of Clinical metabolism and the brain-gut-microbiome axis. Behavioural Brain
Psychiatry 46: 14–19. Research 277: 32–48.
Lozupone CA, Stombaugh JI, Gordon JI, et al. (2012) Diversity, stability Opperman TJ and Nguyen ST (2015) Recent advances toward a molecular
and resilience of the human gut microbiota. Nature 489: 220–230. mechanism of efflux pump inhibition. Frontiers in Microbiology 6: 421.
Lukic I, Getselter D, Ziv O, et al. (2019) Antidepressants affect gut micro- Ortega-Martinez S (2015) A new perspective on the role of the CREB
biota and Ruminococcus flavefaciens is able to abolish their effects on family of transcription factors in memory consolidation via adult hip-
depressive-like behavior. Translational Psychiatry 9: 133. pocampal neurogenesis. Frontiers in Molecular Neuroscience 8: 46.
Lyte M and Brown DR (2018) Evidence for PMAT- and OCT-like biogenic Patel JB (2017) Performance Standards for Antimicrobial Susceptibility
amine transporters in a probiotic strain of Lactobacillus: Implications Testing. Wayne, PA: Clinical and Laboratory Standards Institute.
for interkingdom communication within the microbiota-gut-brain Pawar HA, Ayre AP and Lalitha K (2012) Development of validated ana-
axis. PloS ONE 13: e0191037. lytical method for in-vitro dissolution study of sertraline hydrochlo-
Lyte M, Daniels KM and Schmitz-Esser S (2019) Fluoxetine-induced ride capsules. Journal of Current Pharma Research 2: 560–565.
alteration of murine gut microbial community structure: Evidence for Powell N, Walker MM and Talley NJ (2017) The mucosal immune sys-
a microbial endocrinology-based mechanism of action responsible for tem: Master regulator of bidirectional gut-brain communications.
fluoxetine-induced side effects. PeerJ 7: e6199. Nature Reviews Gastroenterology & Hepatology 14: 143–159.
Macedo D, Filho A, Soares de Sousa CN, et al. (2017) Antidepressants, Pratt LA, Brody DJ and Gu Q (2017) Antidepressant Use among Persons
antimicrobials or both? Gut microbiota dysbiosis in depression and Aged 12 and Over: United States, 2011-2014. National Center for
possible implications of the antimicrobial effects of antidepressant Health Statistics Data Brief 283: 1–8.
drugs for antidepressant effectiveness. Journal of Affective Disorders Qamar F and Safila N (2015) Comparrative assay of citalopram in differ-
208: 22–32. ent media. Journal of Bioequivalence & Bioavailability 7: 4.
Maier L, Pruteanu M, Kuhn M, et al. (2018) Extensive impact of non- Ramsteijn AS, Jašarević E, Houwing DJ, et al. (2018) Antidepressant
antibiotic drugs on human gut bacteria. Nature 555: 623. treatment modulates the gut microbiome and metabolome during
Martino M, Rocchi G, Escelsior A, et al. (2012) Immunomodulation pregnancy and lactation in rats with a depressive-like phenotype.
mechanism of antidepressants: Interactions between sero- Epub ahead of print 19 December. DOI: 10.1101/501742.

Australian & New Zealand Journal of Psychiatry, 53(12)


1166 ANZJP Articles

Rhee K-J, Sethupathi P, Driks A, et al. (2004) Role of commensal bacteria Ticinesi A, Milani C, Lauretani F, et al. (2017) Gut microbiota composi-
in development of gut-associated lymphoid tissues and preimmune tion is associated with polypharmacy in elderly hospitalized patients.
antibody repertoire. The Journal of Immunology 172: 1118–1124. Scientific Reports 7: 11102.
Rogers MAM and Aronoff DM (2016) The influence of non-steroidal anti- van Gorp F, Duffull S, Hackett LP, et al. (2012) Population pharmacoki-
inflammatory drugs on the gut microbiome. Clinical Microbiology netics and pharmacodynamics of escitalopram in overdose and the
and Infection 22: 178. e171–e179. effect of activated charcoal. British Journal of Clinical Pharmacology
Rojo D, Méndez-García C, Raczkowska BA, et al. (2017) Exploring the 73: 402–410.
human microbiome from multiple perspectives: Factors altering its van Harten J (1995) Overview of the pharmacokinetics of fluvoxamine.
composition and function. FEMS Microbiology Reviews 41: 453–478. Clinical Pharmacokinetics 29: 1–9.
Rooks MG and Garrett WS (2016) Gut microbiota, metabolites and host Venkatakrishnan K and Obach RS (2005) In vitro-in vivo extrapolation
immunity. Nature Reviews Immunology 16: 341–352. of CYP2D6 inactivation by paroxetine: Prediction of nonstationary
Rush AJ, Trivedi MH, Wisniewski SR, et al. (2006) Acute and longer-term out- pharmacokinetics and drug interaction magnitude. Drug Metabolism
comes in depressed outpatients requiring one or several treatment steps: and Disposition 33: 845–852.
A STAR* D report. American Journal of Psychiatry 163: 1905–1917. Vogt NM, Kerby RL, Dill-McFarland KA, et al. (2017) Gut microbiome
Sahay A and Hen R (2007) Adult hippocampal neurogenesis in depression. alterations in Alzheimer’s disease. Scientific Reports 7: 13537.
Nature Neuroscience 10: 1110. Wade AG, Crawford GM and Yellowlees A (2011) Efficacy, safety and
Schemann M and Neunlist M (2004) The human enteric nervous system. tolerability of escitalopram in doses up to 50 mg in major depres-
Neurogastroenterology & Motility 16: 55–59. sive disorder (MDD): An open-label, pilot study. BMC Psychiatry
Schiller C, Frohlich CP, Giessmann T, et al. (2005) Intestinal fluid vol- 11: 42.
umes and transit of dosage forms as assessed by magnetic resonance Wernert GT, Winkler DA, Holan G, et al. (2004) Synthesis, biologi-
imaging. Alimentary Pharmacology & Therapeutics 22: 971–979. cal activity, and QSAR studies of antimicrobial agents containing
Seaman JS, Bowers SP, Dixon P, et al. (2005) Dissolution of common biguanide isosteres. Australian Journal of Chemistry 57: 77–85.
psychiatric medications in a Roux-en-Y gastric bypass model. Winter G, Hart RA, Charlesworth RPG, et al. (2018) Gut microbiome
Psychosomatics 46: 250–253. and depression: What we know and what we need to know. Nature
Shameem M, Katori N, Aoyagi N, et al. (1995) Oral solid controlled Reviews Neuroscience 29: 629–643.
release dosage forms: Role of GI-mechanical destructive forces and Yamashita A, Singh SK, Kawate T, et al. (2005) Crystal structure of a bac-
colonic release in drug absorption under fasted and fed conditions in terial homologue of Na+/Cl--dependent neurotransmitter transport-
humans. Pharmaceutical Research 12: 1049–1054. ers. Nature 437: 215–223.
Sim K, Lau WK, Sim J, et al. (2015) Prevention of relapse and recur- Yano Jessica M, Yu K, Donaldson Gregory P, et al. (2015) Indigenous
rence in adults with major depressive disorder: Systematic review bacteria from the gut microbiota regulate host serotonin biosynthesis.
and meta-analyses of controlled trials. The International Journal of Cell 161: 264–276.
Neuropsychopharmacology 19: pyv076. Yee AL and Gilbert JA (2016) Is triclosan harming your microbiome?
Singh SK (2014) LeuT: A prokaryotic stepping stone on the way to a eukar- Science 353: 348–349.
yotic neurotransmitter transporter structure. Channels 2: 380–389. Yim G, Wang HH and Davies J (2007) Antibiotics as signalling mole-
Singh SK, Yamashita A and Gouaux E (2007) Antidepressant binding site cules. Philosophical Transactions of the Royal Society of London B:
in a bacterial homologue of neurotransmitter transporters. Nature 448: Biological Sciences 362: 1195–1200.
952–956. Zheng P, Zeng B, Zhou C, et al. (2016) Gut microbiome remodeling
Stilling RM, van de Wouw M, Clarke G, et al. (2016) The neuropharma- induces depressive-like behaviors through a pathway mediated by the
cology of butyrate: The bread and butter of the microbiota-gut-brain host’s metabolism. Molecular Psychiatry 21: 786–796.
axis? Neurochemistry International 99: 110–132. Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, et al. (2019)
Sutton SC (2009) Role of physiological intestinal water in oral absorption. Mapping human microbiome drug metabolism by gut bacteria and
The AAPS Journal 11: 277–285. their genes. Nature 570: 462–467.

Australian & New Zealand Journal of Psychiatry, 53(12)

You might also like