You are on page 1of 18

JOURNAL OF NEUROCHEMISTRY | 2019 doi: 10.1111/jnc.

14661

University of Alberta, Edmonton, AB, Canada

Abstract compare antidepressants from three distinct classes (tricyclics,


Inflammatory insult to the central nervous system (CNS) can monoamine oxidase inhibitors, and SSRIs) on microglial
lead to development of depression, and subsequently depres- apoptosis. Primary microglia were treated with 1 lg/mL
sion is the most frequent psychiatric comorbidity following lipopolysaccharide and/or 10 lM antidepressants, and various
ischemic stroke, often limiting recovery and rehabilitation in apoptotic markers were assayed. Fluoxetine and its metabolite
patients. The initiators of inflammatory pathways in the CNS norfluoxetine decreased protein levels in cell lysates,
are microglia activated in response to acute ischemic stress, decreased cell viability of microglia, and increased the
and anti-depressants have been shown to have anti-inflam- expression of the apoptotic marker cleaved-caspase 3 in
matory effects in the CNS, promoting neuronal survival microglia. Live/dead nuclear staining also showed that fluox-
following ischemic insult. We have previously shown that the etine- or norfluoxetine-treated cultures contained greater
selective serotonin reuptake inhibitors (SSRIs) fluoxetine and numbers of dying microglial cells compared to vehicle-treated
citalopram promote neuronal survival after oxygen-glucose cultures. Cultures treated with citalopram, phenelzine, or
deprivation, an in vitro model of ischemia, by attenuating the imipramine showed no evidence of inducing microglial apop-
release of glutamate and D-serine from activated microglia. tosis. Our results demonstrate that fluoxetine and norfluoxetine
Interestingly, we found that fluoxetine-treated microglial cul- induce the apoptotic death of microglia, which may serve as a
tures contained fewer numbers of cells compared to other mechanism to attenuate the release of glutamate and D-serine
groups and hypothesized that fluoxetine and citalopram atten- from activated microglia.
uated the release of glutamate and D-serine by promoting the Keywords: antidepressant, fluoxetine, microglia, norfluoxetine.
apoptosis of microglia. The present study aimed to test and J. Neurochem. (2019) https://doi.org/10.1111/jnc.14661

Depression is a prevalent and highly debilitating mental the hypothalamic-pituitary-adrenal axis, and neurogenesis are
disorder affecting up to 15% of the population at least once in not sufficient to describe these mechanisms entirely. Consid-
their lifetime (Masi and Brovedani 2011). Although depressive erable work has demonstrated that inflammatory immune
symptoms are well defined, the mechanisms involved in the processes may contribute to the etiology of depression, and
onset and progression of depression are widely debated. Many interact with many of the neurobiological and neurochemical
hypotheses have been proposed to describe the neurobiology mechanisms described in the monoamine, hypothalamic-
of depression; however, hypotheses focused on monoamines, pituitary-adrenal axis, and neurogenesis hypotheses. The

Received September 25, 2018; revised manuscript received December FBS, fetal bovine serum; GC-MS, gas chromatography mass spec-
17, 2018; accepted December 20, 2018. troscopy; HPA, hypothalamic-pituitary-adrenal axis; HRP, horseradish
Address correspondence and reprint requests to Kathryn G. Todd, peroxidase; Iba1, ionized calcium-binding adapter protein-1; IETD, Z-
Neurochemical Research Unit, Department of Psychiatry, University of Ile-Glu(OMe)-Thr-Asp(OMe)-fluoromethylalketone; IL1 b, interleukin-
Alberta, 116th St and 85th Ave NW Edmonton, AB T6G2R3 Canada. 1b; LPS, lipopolysaccharide; MAOI, monoamine oxidase inhibitor;
E-mail: kgtodd@ualberta.ca MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide;
Abbreviations used: 5HT, 5-hydroxytryptamine; ANOVA, analysis of NEC, necrostatin-1; NED, naphthylethylenediamine dihydrochloride;
variance; BBB, blood brain barrier; CC3, cleaved caspase 3; CNS, NO, nitric oxide; OGD, oxygen-glucose deprivation; PBS, phosphate
central nervous system; DIV, days in vitro; DMEM/F12, Dulbecco’s buffered saline; RRID, research resource identifier; SDS, sodium dodecyl
modified Eagle medium/Ham’s F12 nutrient mix (1:1); EDTA, ethylene sulfate; SSRI, selective serotonin reuptake inhibitor; STS, staurosporine;
diamine tetraacetic acid; EGTA, ethylene glycol-bis(b-aminoethyl ether)- TCA, tricyclic antidepressant; TEEN, tris-EDTA-EGTA-NaCl buffer;
N,N,N0 ,N0 -tetraacetic acid; ELISA, enzyme linked immunosorbent assay; TNF, tumor necrosis factor; WB, western blotting.

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661 1


2 K. S. Dhami et al.

innate immune cells responsible for initiating inflammation in mediated by N-methyl-D-aspartic acid receptor signaling (Mat-
the central nervous system (CNS) are microglia. In the healthy sui et al. 1995; Wu et al. 2004; Takeuchi et al. 2005, 2006).
adult brain, microglia are found in a ramified state of active Surprisingly, we observed a reduction in cell number and
surveillance, characterized by a rod-shaped body and dynamic, protein levels of cultured microglia following fluoxetine
elongated processes. In addition to their surveillance roles, treatment, and hypothesized that fluoxetine and citalopram
microglia carry out maintenance and homeostatic roles and induced the cellular death of microglia, reducing the release of
contribute to ongoing plasticity of the CNS by regulating glutamate and D-serine from these inflammatory cells. In the
synaptic refinement, neurogenesis, and synaptic plasticity current exploratory study, we compare the effects of antide-
(Davalos et al. 2005 and Nimmerjahn et al. 2005). Microglial pressants from the TCA, MAOI, and SSRI classes on apoptotic
cells undergo morphological and functional changes in cellular death of cultured microglia. We present the novel
response to acute tissue damage, as in traumatic brain injury, finding that treatment with fluoxetine and its metabolite
infection, or stroke, and may be chronically activated in norfluoxetine induces the apoptotic cellular death of microglia.
various neurodegenerative diseases (Ling et al. 2001). Once This may serve as a mechanism to reduce the release of
activated, microglia acquire an amoeboid morphology, neurotoxic glutamate and D-serine from activated microglia.
actively phagocytose, and secrete various cytokines and pro-
inflammatory factors including interleukin 1-beta (IL-1b)
Methods and materials
(Kim et al. 2004 and Lai and Todd 2008), tumor necrosis
factor (TNF) (Jung et al. 2007 and Park et al. 2007), and nitric Primary mixed glial cultures
oxide (NO) (Bi et al. 2005 and Wu et al. 2007). All animal procedures were performed in accordance with the
Depression is a common psychiatric co-morbidity accom- University of Alberta Animal Care and Use Committee’s regulations
panying diseases such as Parkinson’s and Alzheimer’s under approved Animal Use Protocol AUP#0000343. Mixed glia
disease and insults such as stroke (Tanaka et al. 2006; Even were prepared from whole brains of postnatal day 1 Sprague-
Dawley rats (Charles River Laboratories, Montreal, QC, Canada,
and Weintraub 2010; Raskin and Durst 2010; Weintraub
CD 001, RRID: RGD_734476) acquired from the Science Animal
et al. 2010; Arbus et al. 2011 and El Husseini et al. 2012),
Support Services colony at the University of Alberta. Day 1 rat pups
wherein it is also a major factor limiting recovery and were killed by conscious decapitation and meninges were removed
rehabilitation in stroke patients (Li et al. 2012). Acute stroke and cells were dissociated by enzymatic digestion with 0.25%
causes irreversible damage to neurons within the ischemic trypsin-EDTA (Gibco, Rockville, MD, USA; 25200056) for
core and progressive, spreading injury to vulnerable neurons 25 min, followed by mechanical trituration in basal media [Dul-
in the surrounding penumbra, which is the pharmacological becco’s Modified Eagle Medium (DMEM/F-12, Gibco 11320033)
target for neuroprotective treatment in acute ischemic stroke supplemented with 10% fetal bovine serum (Gibco 12483) and 2%
(Liu et al. 2010). The primary objective in treatment of penicillin/streptomycin (P/S, Gibco 15140122)]. Mixed glia were
ischemic stroke is to salvage the penumbra as much and as plated on poly-L-lysine-coated (2 lg/mL, Sigma P0879) 12- or 24-
early as possible, and it has been reported that roughly half of well plates at a density of 1 9 105 cells viable per well in basal
media, determined via trypan blue exclusion method using a
all acute ischemic patients show potentially treatable penum-
hemocytometer. Mixed glia were maintained at 37°C in a 5% CO2
bra on magnetic resonance imaging (Rivers et al. 2006).
humidified incubator with complete media changes every 3–4 days
The primary treatment for depression before or after a stroke in vitro (DIV) with basal medium. Mixed glia were cultured for 21
is selective serotonin reuptake inhibitor (SSRI) antidepres- DIV to confluence prior to isolation of microglia.
sants; however, many investigations have shown that various
classes of antidepressants also attenuate the pro-inflammatory
Primary microglial culture
profile of microglia in the CNS (Obuchowicz et al. 2006; Microglia were isolated after 21 DIV from mixed glial cultures via
Hashioka et al. 2007; Tynan et al. 2012). Previously, we have trypsinization (Saura et al. 2003). Briefly, media was removed from
shown in an in vitro co-culture model of ischemia that cultured mixed cultures and the wells were washed with serum-free DMEM/F-
cortical neurons subjected to oxygen-glucose deprivation were 12, trypsin (diluted 1:3 in DMEM/F-12) was added and plates were
protected from inflammatory injury when microglia were pre- returned to incubator until the adherent cell layer detached (approx-
treated with the SSRI antidepressants fluoxetine and citalo- imately 30 min). Trypsin and debris were aspirated and fresh basal
pram (Dhami et al. 2013). Various monoamine oxidase media was added to the wells and incubated for 10 min to inactivate
inhibitor (MAOI) and tricyclic (TCA) class antidepressants, trypsin. Following inactivation, medium was aspirated and experi-
including phenelzine, imipramine, and clomipramine were mental media (serum-free DMEM/F12 supplemented with 2% P/S)
was added to the wells. Cultures were incubated overnight at 37°C
also investigated; however, these antidepressants failed to
with 5% CO2 and used for experiments the following day.
rescue the ischemic injured cortical neurons. Fluoxetine and
citalopram inhibited glutamate and D-serine release from
activated microglia, whereas the other antidepressants tested Microglial treatments
failed to inhibit this release. Glutamate and D-serine released Representative antidepressants from three drug classes were used in
from microglia can lead to excitatory neurotoxicity primarily pretreatment of microglia prior to co-culture: imipramine (5–40 lM as

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 3

indicated in figure legends, TCA, Sigma I7379), phenelzine (5–40 lM Nitrite assay
as indicated in figure legends, MAOI, Sigma P6777), and fluoxetine (5– Nitrite levels, an indirect measure of NO, were determined by the
40 lM as indicated in figure legends, SSRI, Eli Lilly F03072), Griess Reaction as described in previous reports (Green et al. 1982;
citalopram (5–40 lM as indicated in figure legends, SSRI, H- Lai and Todd 2008). Briefly, differentially treated media was
Lundbeck Lu10-171B), and norfluoxetine (5–40 lM as indicated in collected and nitrite measured in duplicate by incubation with equal
figure legends, SSRI, Eli Lilly 141296). Staurosporine (Sigma S6942) parts 1% sulfanilamide in 3N HCl and 0.1% naphthylethylenedi-
was used as a positive inducer of apoptosis at 100 ng/mL, while TNF (R amine dihydrochloride in distilled water. Absorbance (540 nm) was
& D systems DY510, 20 ng/mL) was used as a positive inducer of the read on a plate reader (SpectraMax M3, Molecular Devices, Palo
extrinsic apoptotic pathway (Boldin et al. 1996; Fernandes-Alnemri Alto, CA, USA). Values were calculated from measurement of
et al. 1996; Muzio et al. 1996). The caspase 8 inhibitor Z-Ile-Glu parallel nitrite standards (Sigma, St Louis, MO, USA). Extrapolated
(OMe)-Thr-Asp(OMe)-fluoromethylalketone (IETD, Enzo Life nitrite values were normalized to protein measured in the corre-
Sciences ALX-260-144, 50 lM) was used to inhibit extrinsic pathway sponding cell lysate.
activity and the RIP kinase inhibitor necrostatin-1 (30 lM, Sigma
N9037) was used to reverse this inhibition. All drugs were prepared as Enzyme-linked immunosorbant assay (ELISA)
100X stocks in aqueous solutions and diluted to the final concentrations Quantification of TNF was performed using ELISA kits (R & D
as indicated immediately prior to each experiment. Systems, Minneapolis, MN, USA; DY510) according to the
manufacturer’s protocol. Briefly, plates were coated with capture
Assessment of microglial viability and protein levels antibody (4 lg/mL anti-TNF in PBS) overnight, blocked with 1%
Microglial viability was assessed using the 3-(4,5-dimethylthiazol- bovine albumin (BSA) in PBS for 1 h, and samples and standards
2-yl)-2,5-diphenyltetrazolium bromide (MTT, Sigma M5655) col- loaded overnight at 4°C. After washing with PBS, signal was
orimetric assay. Briefly, 100 lL of 5 mg/mL MTT were added to detected with sequential incubation of detection antibody [225 ng/
microglial wells and incubated for 40 min at 37°C, 5% CO2. Media mL biotinylated anti-TNF in PBS + 1% BSA, 2 h at 21°C],
was aspirated and adherent cells were lysed with 210 lL of horseradish peroxidase-conjugated streptavidin [diluted 1 : 200 in
dimethyl sulfoxide. Absorbance was read at 570 nm on a microwell PBS + 1% BSA, 20 min at 21°C], and color developed with
plate reader (SpectraMax M3, Molecular Devices). tetramethylbenzidine (TMB) (270 mg of TMB in 40% methanol,
Protein was assayed by the bicinchoninic acid method (BCA assay, 100 mM sodium acetate, 18 mM citric acid, Sigma) with 0.03%
Pierce, Rockford, IL, USA; 23223) according to the manufacturer’s hydrogen peroxide. The reaction was stopped by adding 1.8 N
protocol. Media was collected and adherent cells were gently washed sulfuric acid to the wells and the absorbance was read at 450 nm
with phosphate buffered saline (PBS) (3x), and lysed with lysis buffer with background correction at 570 nm. Values were normalized to
(0.8% Triton X-100 and 0.2% sodium dodecyl sulfate (SDS) in tris- protein levels of the corresponding cell lysate.
EDTA-EGTA-NaCl buffer (50 mM Tris, 1 mM EDTA, 1 mM
EGTA, and 150 mM NaCl) and scraping. Lysates were measured in Live/dead staining of microglia
duplicate by addition of bicinchoninic acid reagent [copper (II) sulfate Live/dead assay was performed on cultured microglia using the
solution diluted in bicinchoninic acid (1 : 50; Pierce, as manufac- fluorescent nuclear stains SYTO 13 and SYTOX Orange (Life
turer’s protocol), after incubation at 37°C for 30 min and measure- Technologies, Grand Island, NY, USA; S7575 and S11368)
ment of absorbance at 562 nm. without fixation. Following treatments media was removed and
replaced with fresh DMEM/F-12 with SYTO 13 to a final
Immunocytochemistry concentration of 5 lM and incubated at 37°C at 5% CO2 for
Following experimental treatments, cells were washed (all subse- 110 min, followed by SYTOX orange (0.1 lM) for 10 min. The
quent wash steps were with PBS) and fixed with 10% phosphate- media was then removed, and wells were washed two times with
buffered formalin for 10 min. After three washes with PBS, the cells PBS and cover-slipped using aqueous mounting medium. Images
were incubated with a blocking solution [10% horse serum (Gibco were acquired on a Leica DM-6000 inverted fluorescence micro-
16050122) in PBS with 0.1% Triton X-100] for 1 h, washed, and scope (Leica Microsystems). Nine images were captured per well
incubated and co-labeled with Iba-1 (1 : 1000, Wako RRID: for each filter, and merged images were created by Image-J
AB_839504) or cleaved-caspase 3 (1 : 500, Cell Signaling Tech- software.
nology, Beverly, MA, USA; RRID:AB_2070042) primary antibod-
ies (diluted in PBS with 1% horse serum) overnight at 4°C. After Western blotting (WB)
subsequent washing, appropriate fluorescent-conjugated secondary WB was used to determine cleaved-caspase 3 (CC3), caspase 3
antibodies (Alexa Fluor 488 or 647, 1 : 200, Invitrogen RRID: (C3), caspase 8 (C8) and b-actin expression cell lysates from vehicle
AB_2536183, RRID:AB_162542, RRID:AB_2535792, and RRID: and antidepressant-treated microglia. Briefly, 9–12 microglial wells
AB_141607) were diluted in PBS for 1 h at 21°C. After a final (in 12-well plates) were pooled and lysed with tris-EDTA-EGTA-
wash, wells were mounted using aqueous mounting medium NaCl buffer (0.8% Triton X-100 and 0.2% SDS) after 24 h of
(Vectashield, Vector Laboratories, Burlingame, CA, USA). Images antidepressant treatment. 15% acrylamide gels (37.5 : 1 bis-
were acquired on a Leica DM-6000 inverted fluorescence micro- acrylamide) were loaded with 15–22 lg of protein per well. The
scope (Leica Microsystems, Wetzlar, Germany). Nine images were gels were run for 2 h under a voltage of 120 volts using BioRad
captured per well for each filter, image overlays were created using (Hercules, CA, USA) western blotting equipment in running buffer
Image-J. Image acquisition and analysis was carried out by a user (25 mM Tris, 250 mM glycine, and 3.5 mM SDS). Gels were then
blinded to the experimental conditions. placed in transfer buffer (40 mM glycine, 50 mM Tris, and 1 mM

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


4 K. S. Dhami et al.

SDS) and proteins were transferred to polyvinylidene fluoride


membrane (Millipore Corporation, Bedford, MA, USA) at 4°C
Results
overnight at 0.08 amps. The following day, the membranes were Fluoxetine decreases the viability of primary cultured
washed with wash buffer (20 mM Tris, 140 mM NaCl, and 1 : 50 microglia
Tween) and blocked for 1 h with 10% skim milk powder (diluted in Having previously demonstrated decreased microglial release
wash buffer) with gentle agitation at 21°C. After washing, the primary
of D-serine and glutamate after treatment with either
antibodies [rabbit anti-CC3 (1 : 1000, Cell Signaling RRID:
fluoxetine or citalopram (Dhami et al. 2013), we sought to
AB_2070042), rabbit anti-C3 (1 : 1000, Cell Signaling RRID:
AB_2069872), rabbit anti-C8 (1 : 1000, Cell Signaling RRID: investigate the possible mechanisms behind this result. Upon
AB_10545768), or mouse anti-rat b-actin (1 : 5000, Sigma RRID: close examination of data, we found that fluoxetine (10 lM)
AB_476744)] at the indicated dilutions (in PBS with 1% bovine significantly decreased the protein levels from microglial cell
serum albumin and 0.02% sodium azide in wash buffer) were lysates compared to vehicle-treated microglia and that this
incubated overnight on a shaker at 4°C. Subsequently, horseradish effect was dose-dependent, as higher doses of fluoxetine (30
peroxidase-conjugated 2° antibodies (diluted 1 : 10000 in PBS with and 40 lM) lowered these protein levels even further
5% skim milk solution, Santa Cruz RRID:AB_641181, RRID: (Fig. 1a). As protein levels in the cell lysate are generally
AB_641170) were incubated for 1 h at 21°C and detected using proportional to the number of cells in culture, we hypoth-
enhanced chemiluminescence (ECL, Amersham), exposed to Kodak esized that a possible mechanism behind the attenuation of
film for varying amounts of time, and developed using a Kodak
glutamate and D-serine release from activated microglia is by
processor.
the induction of microglial apoptosis after fluoxetine or
citalopram treatment. Measurement of microglial viability
Gas chromatography-mass spectrometry (GC-MS) after SSRI treatment showed that the addition of fluoxetine
Fluoxetine and norfluoxetine levels in medium were measured by
similarly decreased the viability of microglia compared to
GC-MS using a modification of an assay described previously in
vehicle-treated controls in a dose dependent manner
our laboratory (Rotzinger et al. 1997). Standards were prepared in
naive culture medium. Media collected from fluoxetine-, norfluox-
(Fig. 1b). When examining the morphology of fluoxetine-
etine-, or vehicle-treated microglia (450 lL) was used in the treated microglia under a phase-contrast microscope, we
analytical procedure. Fluvoxamine (100 ng) was added as internal observed that those cells treated with fluoxetine (10, 20, 30,
standard to the samples, which were basified (to pH 8.5) by the and 40 lM) showed an increased number of spherical-like
addition of 50 lL potassium bicarbonate. A solution of 2 lL cells compared to vehicle-treated controls, which showed
pentafluorobenzoyl chloride in 2 mL toluene was added to each more elongated cells and few spherical cells (data not
sample tube. The tubes were capped and shaken vigorously for shown). The morphological transformation of microglia from
15 min and centrifuged at 1800 g for 5 min in a benchtop elongated to spherical is one classic hallmark of apoptosis.
centrifuge. The organic phase was pipetted into 100 mm 9
13 mm screw cap culture tubes and taken to dryness in a Savant
Phenelzine, imipramine, and citalopram do not decrease the
evaporator (Speed Vac SC 110, Fisher Scientific). Toluene
viability of microglia
(100 lL) was added to each tube and the tubes were vortexed.
The toluene was transferred to a silanized glass insert and 2 lL was
As phenelzine, imipramine, or citalopram have been shown
injected onto an Agilent 6890 GC coupled to a 5973n mass to exert anti-inflammatory effects on microglia, and citalo-
spectrometer with an electron ionzation source. Data were collected pram attenuated the release of glutamate and D-serine from
in the single ion monitoring mode. The column used was HP-5MS lipopolysaccharide (LPS)-activated microglia comparable to
(5% phenyl methyl siloxane; 30 9 0.25 m, 0.25 lM), with a gas fluoxetine [self-cite], we sought to determine if comparable
flow rate of 1 mL/min helium. effects were seen on microglial viability. As expected,
phenelzine and imipramine did not decrease the level of
Statistical analysis protein in cell lysates (Fig. 1c) or the viability of microglia
Significance levels for data sets were calculated using a two-tailed t- (Fig. 1d) at any dose tested, though contrary to our
test for pairwise comparisons or a two-way ANOVA followed by expectations, citalopram did not affect the protein levels
Bonferroni post hoc test for multivariate comparisons as appropriate. (Fig. 1c) or viability of microglia (Fig. 1d) at any dose
Normality testing was not performed as the number of replicates tested. This disparity between the two SSRI antidepressants
(< 10) is insufficient for such testing, however parametric tests were fluoxetine and citalopram suggests that the effect of
used given the nature of sampling (biochemical sampling of cell fluoxetine on microglial viability may be unrelated to their
populations) assumes normality of the underlying biological analyte.
ability to inhibit serotonin reuptake.
No tests for outliers were performed on data. Differences were
Fluoxetine, but not citalopram, phenelzine, or imipramine,
considered significant at a p < 0.05, denoted by * and †. For all
molecular analyses each ‘n’ represents an independent culture
increases the number of compromised microglial cells and
preparation from a separate animal with three technical replicates reduces the percentage of live cells.
per condition, and appropriate numbers were determined based on In order to verify that the observed decrease in microglial
prior published examples using this model (Lai and Todd 2008; protein levels and viability following fluoxetine treatment
Dhami et al. 2013). was a result of cellular death we used a SYTO/SYTOX live/

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 5

(a) (b)

(c) (d)

Fig. 1 Fluoxetine decreases protein levels in microglial cell lysates 10 lM treatments n = 10. (c) Phenelzine, Imipramine, or citalopram
and decreases microglial viability in a concentration-dependent treatments of 5 or 10 lM showed no differences in the protein levels in
manner. (a) All fluoxetine-treated microglial cells showed a decrease cell lysates compared to vehicle-treated controls, n = 8. (d) Phenel-
in the protein levels in cell lysates compared to vehicle-treated zine at 5 lM increased microglial viability compared to vehicle-treated
controls. Microglia treated with 30 and 40 lM fluoxetine showed a controls; whereas all the other concentrations of phenelzine, imipra-
greater reduction in protein levels compared to 5 or 10 lM treatments, mine, or citalopram tested had no effect, n = 8. Each ‘n’ represents an
n = 10. (b) All fluoxetine-treated microglial cells, excluding 5 lM independent culture preparation from a separate animal. Statistical
treatments, showed a decrease in MTT absorbance (viability) com- significance, *p < 0.05 (relative to control), †p < 0.05 (relative to
pared to vehicle-treated controls. Microglia treated with 30 and 40 lM indicated condition).
fluoxetine showed a greater reduction in viability compared to 5 or

dead staining protocol to distinguish healthy intact cells from Fluoxetine, but not citalopram, phenelzine, or imipramine,
compromised damaged cells. SYTO is a membrane perme- increases cleavage of caspase 3 in microglia
able nuclear stain, while SYTOX is membrane impermeable, As cell death can occur through a number of distinct
thus cells with compromised plasma membranes will co-label mechanisms, we measured the expression of CC3 as a
while healthy cells will only retain the green SYTO dye. We marker of apoptotic activity in antidepressant-treated micro-
found that the number of co-stained cells (dead cells) in the glia. Apoptosis involves the cleavage of the protease caspase
imipramine (Fig. 2c), phenelzine (Fig. 2d), or citalopram 3 within the cytoplasm to produce CC3; therefore, apoptotic
(Fig. 2e) groups did not differ compared to vehicle-treated cells show decreases in caspase 3 and increases in CC3
controls (Fig. 2a). As a positive control for the initiation of levels. CC3 was first visualized in cultured microglia via
cell death, we treated microglia with 100 nM of stau- immunocytochemistry to measure the increase in apoptotic
rosporine. After the treatment with fluoxetine (Fig. 2b) or activity in microglia following fluoxetine treatment. Com-
staurosporine (Fig. 2f), we found a significant increase in the pared to vehicle-treated controls (Fig. 3a), microglia treated
percentage of co-stained cells compared to vehicle-treated with 10 lM fluoxetine displayed a significantly greater
controls (Fig. 2g). Therefore, we found that fluoxetine number of cells co-labeled with both ionized calcium-
induces cell death in microglia, whereas the other antide- binding adapter protein-1 (Iba1) and CC3 (Fig. 3b). The
pressants tested did not have this effect. treatment of microglia with 10 lM of imipramine (Fig. 3c),

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


6 K. S. Dhami et al.

(a) i iii

ii

(b)
i iii

ii

(c) (d)

Fig. 2 Live/dead assay of antidepressant-


treated non-activated microglia. (a) SYTO
green (i), SYTOX orange (ii), and the
merged image (iii) of STYO green and
SYTOX orange in vehicle-treated control
cells. (b) SYTO green (i), SYTOX orange
(e) (f) (ii), and the merged image (iii) of STYO
green and SYTOX orange of fluoxetine-
treated cells. (c–f) The merged images of
SYTO green and SYTOX orange of
imipramine (c), phenelzine (d), citalopram
(e), and staurosporine (f) treated cells. (g)
Quantification of the percentage of co-
stained microglia for the live and dead
stains across different treatment
conditions. Fluoxetine (10 lM) significantly
increased the number of microglial cells co-
(g) stained with the live and dead stain
compared to vehicle-treated controls;
whereas phenelzine, imipramine, and
citalopram did not have this effect at the
same concentration tested. Staurosporine,
a positive apoptosis-inducing control,
showed similar increases in the number of
microglial cells co-stained with the live and
dead stain compared to fluoxetine-treated
microglia. Scale bars = 80 lm. For each
condition n = 4 independent culture
preparations from a separate animal with
pictures taken from 9 fields per well within
each experiment. Statistical significance,
*p < 0.05 (relative to control), †p < 0.05
(relative to indicated condition).

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 7

(a) i iii Fig. 3 Immunofluorescence microscopy of Iba1 and cleaved-caspase


3 (CC3) expression in antidepressant-treated non-activated microglia.
(a) Iba1 labeling (i, red), CC3 labeling (ii, green), and the merged
image (iii) of Iba1 and CC3 labeling in vehicle-treated control cells. (b)
Iba1 labeling (i, red), CC3 labeling (ii, green), and the merged image
ii (iii) of Iba1 and CC3 in fluoxetine-treated cells. (c–f) The merged
images of Iba1 and CC3 labeling in imipramine (c), phenelzine (d),
citalopram (e), and staurosporine (f) treated cells. (g) Quantification of
the percentage of co-labeled microglia for both Iba1 and CC3 across
different treatment conditions. Fluoxetine significantly increased the
(b) number of cells co-expressing Iba1 (red) and CC3 (green) in microglia
i iii
compared to vehicle-treated controls; whereas imipramine, phenelzine,
or citalopram did not increase the number of cells co-expressing Iba1
and CC3 in microglia compared to vehicle-treated controls. Scale bar =
80 lm. For each condition n = 4 independent culture preparations
ii from a separate animal, with pictures taken from 9 fields per well.
Statistical significance, *p < 0.05 (relative to control), †p < 0.05
(relative to indicated condition). Examples of co-expressing microglia
are shown in merged images with arrows.

(c) (d)
phenelzine (Fig. 3d), or citalopram (Fig. 3e) showed no
differences in co-labeled cells compared to vehicle-treated
controls (Fig. 3a). As with the live/dead assay 100 nM of
staurosporine was used as a positive control for apoptosis
(Fig. 3f), and was found to significantly increase the number
of CC3 positive microglia (Fig. 3g). Western blot analysis
further showed that fluoxetine and staurosporine treatment
(e) (f) significantly increased CC3 levels as well as significantly
decreased caspase 3 levels within microglia compared to
vehicle-treated controls (Fig. 4a,d). Citalopram, imipramine,
or phenelzine treatment had no significant effect on CC3 or
caspase 3 expression levels (Fig. 4b and c). The cleavage of
caspase 3 to CC3 suggest that fluoxetine increased cell death
by inducing microglial apoptosis, whereas citalopram,
phenelzine, and imipramine do not.

Norfluoxetine attenuates the release of pro-inflammatory


(g)
factors from activated microglia and reduces microglial
viability
In a human subject, fluoxetine is primarily metabolized in the
liver to form norfluoxetine, a SSRI as potent as fluoxetine at
inhibiting 5-HT reuptake and with a much longer elimination
half-life than the parent compound (Sager et al. 2014). Since
norfluoxetine is highly prevalent in the depressed brain follow-
ing fluoxetine treatment, we investigated if norfluoxetine had
similar effects as fluoxetine in attenuating pro-inflammatory
factors released from microglia and reducing their cell viability.
First, we questioned whether microglia were able to convert
fluoxetine to norfluoxetine. To answer this question, we
measured the levels of fluoxetine and norfluoxetine in the
media of fluoxetine-treated microglia with GC-MS. If
microglia treated with fluoxetine converted the drug into
norfluoxetine, then we would measure a significant amount of
norfluoxetine within the medium compared to vehicle-treated
controls. GC-MS analysis 24 h after treatment with 10 lM

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


8 K. S. Dhami et al.

Fig. 4 Western Blot analysis for cleaved-caspase 3 and caspase 3 increased the expression of cleaved-caspase 3 (19 kDa) and
expression in microglial cell lysates following antidepressant treat- decreased the expression of caspase 3 (35 kDa) compared to
ments. (a) Fluoxetine increased the expression of cleaved-caspase 3 vehicle-treated controls. In all conditions, b-actin (43 kDa) expression
(19 kDa) and decreased caspase 3 (35 kDa) expression compared to was measured as a loading control. (e) Histograms displaying cleaved
vehicle-treated controls. (b) Citalopram did not alter the expression caspase 3 and (f) caspase 3 expression from western blotting (WB)
levels of cleaved-caspase 3 or caspase 3 compared to vehicle-treated analysis, n = 5. Each ‘n’ represents an independent culture prepara-
controls. (c) Imipramine and phenelzine did not alter the expression tion from a separate animal. Statistical significance, *p < 0.05 (relative
levels of cleaved-caspase 3 (19 kDa) or caspase 3 (35 kDa) compared to control), †p < 0.05 (relative to indicated condition).
to vehicle-treated controls. (d) Staurosporine (a positive control)

fluoxetine showed that there were insignificant levels of co-stained 10 lM norfluoxetine-treated microglia with
norfluoxetine present in the culture media, equivalent to only SYTO and SYTOX live/dead stains. Norfluoxetine (Fig. 6a)
0.09% conversion (Fig. 5a). As norfluoxetine is the major significantly increased the percentage of co-stained (dead)
metabolite present in the CNS during fluoxetine treatment we cells compared to vehicle-treated controls (Fig. 6b), In
measured the release of pro-inflammatory factors from non- addition, we measured the number of microglial cells co-
activated and LPS-activated microglial cells following norflu- labeled against Iba1 and CC3 following 10 lM norfluoxetine
oxetine treatment. Norfluoxetine significantly attenuated the treatment and found that norfluoxetine (Fig. 6c) significantly
release of NO and TNF from LPS-activated microglia (Fig. 5b increased the percentage of co-labeled cells compared to
and c), and decreased the viability of microglia in a dose- vehicle-treated controls (Fig. 6d). Western blot analysis
dependent manner comparable to fluoxetine treatment showed that 10 lM norfluoxetine treatment increased the
(Fig. 5e). Additionally, norfluoxetine decreased the protein expression of CC3 and decreased the expression of caspase 3
levels within microglial cell lysates (Fig. 5d). Thus, while in microglia compared to vehicle-treated controls comparable
microglia do not convert fluoxetine to norfluoxetine to any to the effects of fluoxetine (Fig. 6e–g).
significant extent, both of these SSRIs have similar effects in
decreasing microglial reactivity and viability. Fluoxetine and norfluoxetine affect microglial apoptosis via
the extrinsic pathway
Norfluoxetine increases the number of compromised As apoptosis can occur in response to external (extrinsic
microglia and CC3 expression in microglia pathway) or internal stimuli (intrinsic pathway), we sought to
To determine if norfluoxetine affects microglial viability by determine the site of effect of fluoxetine and norfluoxetine on
induction of apoptosis as seen with fluoxetine we first apoptosis. We measured the expression levels of caspase 8 in

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 9

Fig. 5 Norfluoxetine effects on the release


of pro-inflammatory factors from activated
microglia and on cell viability. (a) Gas
chromatography revealed high levels of
fluoxetine in the medium of 10 lM
fluoxetine-treated microglia, whereas
norfluoxetine levels were virtually
nonexistent. Therefore, microglia were not
converting fluoxetine to norfluoxetine to any
significant extent in culture, n = 3.
Norfluoxetine (10 lM) significantly
attenuated the release of nitric oxide (NO)
(B) and tumor necrosis factor (TNF) (C) from
lipopolysaccharide (LPS)-activated
microglia, n = 10. Norfluoxetine decreased
microglial cell viability (E) and protein levels
(D) within cell lysates at all doses tested,
n = 10. Each ‘n’ represents an independent
culture preparation from a separate animal.
Statistical significance, *p < 0.05 (relative
to control), †p < 0.05 (relative to indicated
condition).

fluoxetine- or norfluoxetine-treated microglia to determine if norfluoxetine-, and staurosporine-treated groups relative to


the extrinsic pathway in apoptosis was activated. Western control (Fig. 7a, Fig. 4f, Fig. 6e and g).
blots revealed that both 10 lM fluoxetine and 100 nM Interestingly, Fricker et al. (2013) found paradoxically
staurosporine decreased the level of caspase 8 in microglia that inhibition of caspase 8 with IETD resulted in the cell
compared to vehicle-treated controls (Fig. 7a and b). As a death of microglia and necrostatin-1, an inhibitor of a
positive control, microglia were treated with 20 ng/mL TNF downstream kinase involved in apoptotic death, prevented
to induce the extrinsic pathway of apoptosis. TNF similarly microglial caspase 8 inhibition-induced death. Therefore,
decreased the level of caspase 8, but 10 lM norfluoxetine caspase 8 activity may have a unique role microglial cell
failed to affect caspase 8 levels in treated microglia (Fig. 7a death compared to other cell types such as neurons. Given
and b). A decrease in caspase 8 levels is consistent with these findings, we hypothesized that fluoxetine may directly
cleavage of caspase 8 upstream to caspase 3 cleavage in the inhibit the activity and expression of caspase 8 to induce the
extrinsic pathway of apoptosis. In contrast, CC3 levels were death of microglia. We inhibited microglial activity of
significantly increased in fluoxetine-, norfluoxetine-, and caspase 8 with IETD and found a significant reduction in
staurosporine-treated groups compared to vehicle-treated MTT cell viability compared to vehicle-treated controls that
controls (Fig. 7a, Fig. 4e, Fig. 6e and f), while caspase 3 was reversed by treatment with necrostatin-1 (Fig. 7c).
levels were significantly decreased in TNF-, fluoxetine-, Microglial cultures treated with fluoxetine also showed a

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


10 K. S. Dhami et al.

(a) i iii (b)

ii

(c) (d)

i iii

ii

(f)

(e)

(g)

Fig. 6 Immunofluorescence microscopy and Western blot analysis of preparations from a separate animal, and each n represents pictures
cell death markers in norfluoxetine-treated microglial cultures. (a) taken from 9 fields per well. Scale bar = 80 lm. (e) Western Blot
SYTO green (i), SYTOX orange (ii), and the merged image (iii) of analysis for CC3 and caspase 3 expression in microglial cell lysates
STYO green and SYTOX orange of 10 lM norfluoxetine-treated cells. following 10 lM norfluoxetine or 100 nM staurosporine-treatment. (f–g)
(b) Quantification of the percentage of co-stained microglia for the live Histograms displaying CC3 and caspase 3 expression following 10 lM
and dead stains across different treatment conditions. (c) Iba1 labeling norfluoxetine treatment from western blotting (WB) analysis, n = 5.
(i, red), CC3 labeling (ii, green), and the merged image (iii) of Iba1 and Each ‘n’ represents an independent culture preparation from a
CC3 in 10 lM norfluoxetine-treated cells. (d) Quantification of the separate animal. Statistical significance, *p < 0.05 (relative to control),
percentage of co-labeled microglia for Iba1 and CC3 across different †p < 0.05 (relative to indicated condition).
treatment conditions. For each condition n = 4 independent culture

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 11

significant reduction in MTT cell viability compared to the number of microglia co-labeled with CC3 and Iba1 as
vehicle-treated controls, but necrostatin-1 failed to reverse measured using immunofluorescence microscopy, and
this effect (Fig. 7d); therefore, it is unlikely the mechanism increased the number of cells co-labeled with both the live
of inducing death by fluoxetine is through the inhibition of and dead stains. In addition, norfluoxetine decreased the
caspase 8 activity and expression in microglia. levels of protein within microglial cell lysates and reduced
MTT cell viability. These results raised an important issue in
that microglia may be converting fluoxetine into norfluox-
Discussion
etine, and that norfluoxetine may be the active component
Given prior observation that treatment with fluoxetine and responsible for inducing apoptosis. To test this, we treated
citalopram decreased glutamate and D-serine release from microglia with fluoxetine and measured the amounts of both
LPS-activated microglia, we hypothesized this effect was due norfluoxetine and fluoxetine in the cell media using GC-MS.
to induction of cell death SSRI antidepressants. Fluoxetine- We found relatively insignificant amounts of norfluoxetine in
treated microglial cultures contained significantly lower these lysates compared to the doses of norfluoxetine required
protein levels and reduced cell viability by MTT assay to induce cell death, demonstrating that the cultured
compared to vehicle-treated cultures after 24 h treatment. In microglia converted fluoxetine into norfluoxetine to only a
addition, using permeability-based live-dead fluorescent minute degree. We conclude that both fluoxetine and
staining, fluoxetine-treated microglial cells showed a greater norfluoxetine can independently induce the apoptotic cell
number of co-stained cells, comparable with cells treated death of microglia, without a requirement for conversion into
with staurosporine as a positive control for cell death. These norfluoxetine by microglia in vitro.
effects were not seen with cultures treated with citalopram or Fluoxetine and norfluoxetine may induce apoptosis by one
other antidepressants from the MAOI or TCA classes. The of two primary pathways, the extrinsic or intrinsic pathway
expression of the apoptotic marker CC3, measured with (Slee et al. 2001). To investigate if these antidepressants
immuno-fluorescence microscopy and Western blot, was initiated the extrinsic pathway, we measured the level of
significantly increased by fluoxetine but not citalopram caspase 8 in microglial cell lysates by western blotting.
(SSRI), imipramine (TCA) or phenelzine (MAOI). Fluox- Caspase 8 associates intracellularly with death receptors that
etine induced the apoptotic death of microglia, and this is a are members of the TNF receptor gene superfamily and is
possible mechanism by which this drug attenuates the release cleaved in response to receptor activation, going on to
of glutamate and D-serine from these cells. activate other downstream caspases such as caspase 3
Contrary to our initial hypothesis, citalopram treatment did (Kischkel et al. 1995; Locksley et al. 2001). We found that
not induce microglial apoptosis. As prior experiments have fluoxetine- or staurosporine-treated cells expressed lower
demonstrated that both fluoxetine and citalopram attenuate levels of caspase 8 compared to vehicle-treated controls.
the release of glutamate and D-serine from activated Norfluoxetine did not decrease caspase 8 levels but decreased
microglia, and as fluoxetine induces microglial apoptosis, caspase 3 levels while increasing cleaved-caspase 3 expres-
we hypothesized that citalopram would likewise induce sion. When we treated microglia with TNF, a positive
microglial apoptosis. Citalopram-treated microglial cultures inducer of extrinsic apoptosis, we found a decrease in
did not show any changes in the expression levels of cleaved- caspase 8 expression compared to vehicle-treated controls.
caspase 3 or caspase 3 with Western blot or immuno- One unexpected result in this experiment is that TNF
fluorescence and citalopram had no effect on live-dead treatment (positive control of extrinsic apoptosis) did not
staining compared to vehicle-treated controls. These unex- show increases in CC3 expression, but did decrease caspase
pected results led to questions about the mechanisms of 3 levels in microglia. One possible explanation is that the
citalopram’s effects on glutamate and D-serine release that concentration of TNF may not be sufficient to induce
still require investigation. apoptosis, though the observed decreased caspase 8 and
After oral administration in human subjects, fluoxetine is caspase 3 levels suggests activation of the extrinsic pathway.
largely metabolized through type I metabolism in the liver, Interestingly, there is evidence that caspase 8 activity may
with less than 10% excreted unchanged or as fluoxetine N- have a unique function in microglia in terms of inducing cell
glucuronide (Benfield et al. 1986). The remainder is metab- death compared to other cell types such as neurons.
olized to the active metabolite norfluoxetine and other Pharmacological inhibition of caspase 8 in microglia results
apparently non-active metabolites primarily by the enzymes in the cell death of microglia in a caspase 8 inhibition-
cytochrome P450 2D6 and 2C9 in the liver (Whirl-Carrillo induced form of apoptosis not seen in other cell types.
et al. 2012). Since the majority of fluoxetine is metabolized Necrostatin-1, an inhibitor of a downstream kinase involved
into norfluoxetine in the human, we investigated if norflu- in this death, prevents microglial caspase inhibition-induced
oxetine also induced apoptosis of microglia in culture. We death (Fricker et al. 2013). We investigated the possibility
found that norfluoxetine increased the expression of the that fluoxetine induced microglial death by acutely inhibiting
apoptotic marker CC3 while decreasing caspase 3, increased the activity of caspase 8 with IETD. Inhibition of microglial

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


12 K. S. Dhami et al.

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 13

Fig. 7 Western Blot analysis for CC3, caspase 3, and caspase 8 8 expression by WB analysis, n = 5. (c) Inhibition of caspase 8 with
expression in microglial cell lysates following fluoxetine and norfluox- IETD (50 lM) significantly reduced MTT cell viability compared to
etine-treatments and the effects of caspase-8 inhibition (by Z-Ile-Glu vehicle-treated controls and necrostatin-1 (30 lM) reversed this effect,
(OMe)-Thr-Asp(OMe)-fluoromethylalketone (IETD), fluoxetine, and n = 8. (d) Fluoxetine treatment (10 lM) significantly reduced MTT cell
necrostatin-1 on microglial cell viability. (a) Representative Western viability compared to vehicle-treated controls, similar to caspase 8-
blots showing levels of cleaved caspase 3 (19 kDa), caspase 3 (35 inhibited conditions, however, necrostatin-1 failed to reverse this effect,
kDa), b-actin (43 kDa, loading control), and caspase 8 (60 kDa) from n = 8. Each ‘n’ represents an independent culture preparation from a
staurosporine, tumor necrosis factor (TNF), norfluoxetine, and fluox- separate animal. Statistical significance, *p < 0.05 (relative to control),
etine treated microglia cell lysates. (b) Histograms displaying caspase †p < 0.05 (relative to indicated condition).

caspase 8 significantly reduced cell viability compared to 2013). In addition, the SSRI fluoxetine may induce
vehicle-treated controls and viability was rescued by necro- microglial apoptosis to help regulate the numbers of reactive
statin-1 treatment; however, necrostatin-1 failed to rescue and proliferative microglia in a region of ischemic insult.
microglial cells following fluoxetine treatment. It is thus The effects of these SSRIs may be beneficial following
unlikely that fluoxetine induces microglial death through the stroke, but there is evidence supporting a detrimental effect
inhibition of caspase 8, however, further experiments are of SSRI treatment prior to an ischemic event. In a 9-year
required to clarify the specific cell death pathway initiated by longitudinal study, Li et al. (2012) found that patients
fluoxetine. diagnosed with depression and prescribed SSRI antidepres-
In previous studies, we have shown that glutamate and D- sants had higher rates of stroke in later life, and this finding
serine release from LPS-activated microglia are factors that has been replicated by a number of studies (Bushnell 2011;
reduce the viability of oxygen-glucose deprivation-injured Andrews et al. 2012; Castro et al. 2012; Simmons et al.
cortical neurons. Fluoxetine is an antidepressant that 2012). Smoller (2011) reported that depressed post-
decreases the release of glutamate and D-serine from LPS- menopausal women had a 45% increased risk of stroke if
activated microglia and attenuates the loss in neuronal prescribed SSRI antidepressants and the risk of fatal stroke
viability. In this study, we have shown that fluoxetine was doubled. In support of this, Ried et al. (2011) reported
induces apoptosis in microglia and this could be a mecha- that depressed patients prescribed SSRI antidepressants were
nism behind the attenuation in glutamate and D-serine three times more likely to die following a stroke. The data in
release. The treatment with SSRI antidepressants such as our studies may help explain these findings. In the absence of
fluoxetine can be either beneficial or detrimental, depending microglia, tissue damage has been shown to be exacerbated
on context of co-morbid depression following stroke. In following a stroke, supporting a beneficial role of microglia
favor of beneficial effects of fluoxetine, microglia are in a (Lalancette-Hebert et al. 2007). We found that fluoxetine
highly reactive and proliferative state as they release a variety induces microglial apoptosis in vitro, and if fluoxetine had
of inflammatory and toxic factors following ischemia, and this same effect in vivo we would expect less microglia
anti-inflammatory drugs such as minocycline and fluoxetine available at times of insult and stress. This reduced
have been shown to protect against ischemic insults (Stoll availability in microglia may cause the increased risk of
et al. 1998; Schroeter et al. 1999; Yrjanheikki et al. 1999; fatal stroke in these depressed patients.
Lim et al. 2009). The frequency of depression following a In our study, we found that citalopram did not induce
stroke is highest after 1 month and the risk remains elevated microglial apoptosis, but others have found that citalopram
for several years. The treatment of stroke patients with SSRI does increase the susceptibility to fatal stroke in depressed
antidepressants has been shown to reduce the incidence of patients (Bushnell 2011; Mikami et al. 2011). One limitation
depression (Gothe et al. 2012; Loubinoux et al. 2012). For in our study was that we only investigated the effects of
instance, the SSRI fluvoxamine has been shown to alleviate citalopram treatment on microglial apoptosis for 24 h. It is
the diminished mood and sleep disturbances in patients with possible that longer or chronic treatments of citalopram may
post-stroke depression (Sunami et al. 2012). In addition, eventually induce microglial apoptosis. Similarly, as we did
chronic treatment with the SSRI citalopram has been shown not investigate the effects of the metabolites of citalopram on
to delay the degeneration of dopaminergic neurons in the the induction of microglial apoptosis, there remains a
midbrain, prevent striatal atrophy, and reduce the behavioral potential for effects of metabolites on apoptosis. Citalopram
symptoms in rodents following middle cerebral artery is metabolized into desmethylcitalopram by the enzymes
occlusion (Kronenberg et al. 2012). One interpretation of cytochrome P450 2C19 and 3A4 and desmethylcitalopram is
our findings is that the SSRIs fluoxetine and citalopram further metabolized into didesmethylcitalopram by cyto-
attenuate the reactive and proliferative state of microglia by chrome P450 2D6 in the liver (Sangkuhl et al. 2011). These
reducing their release of inflammatory and toxic factors, most metabolites are significantly less potent reuptake inhibitors of
importantly glutamate and D-serine, and ultimately reducing 5-HT, but we cannot rule out the possibility that these
neuronal toxicity after an ischemic insult (Dhami et al. metabolites induce microglial apoptosis. Although these

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


14 K. S. Dhami et al.

metabolites cross the blood brain barrier poorly compared to and D-serine to promote cortical neuronal viability following
their parent citalopram (Sangkuhl et al. 2011), studies should ischemic insult. Mol. Cell Neurosci. 56, 365–374.
El Husseini N., Goldstein L. B., Peterson E. D., Zhao X., Pan W., Olson
investigate the effects of these metabolites on microglial
D. M., Zimmer L. O., Williams J. W., Jr, Bushnell C. and
apoptosis. Laskowitz D. T. (2012) Depression and antidepressant use after
stroke and transient ischemic attack. Stroke 43, 1609–1616.
Even C. and Weintraub D. (2010) Case for and against specificity of
Acknowledgments and conflict of interest depression in Alzheimer’s disease. Psychiatry Clin. Neurosci. 64,
disclosure 358–366.
The authors gratefully acknowledge funding from Alberta Health Fernandes-Alnemri T., Armstrong R. C., Krebs J. et al. (1996) In vitro
activation of CPP32 and Mch3 by Mch4, a novel human apoptotic
Services and the Davey Endowment for Brain Research. The authors
cysteine protease containing two FADD-like domains. Proc. Natl
have no financial conflicts to declare. This manuscript is part of a Acad. Sci. USA 93, 7464–7469.
PhD thesis uploaded to https://era.library.ualberta.ca/items/8222be Fricker M1, Vilalta A, Tolkovsky AM and Brown GC. (2013). Caspase
4c-015f-43b2-bc11-ffaf3cebd55d. inhibitors protect neurons by enabling selective necroptosis of
All experiments were conducted in compliance with the ARRIVE inflamed microglia. J. Biol. Chem. 288(13):9145–52.
guidelines. Gothe F., Enache D., Wahlund L. O., Winblad B., Crisby M., Lokk J.
and Aarsland D. (2012) Cerebrovascular diseases and depression:
epidemiology, mechanisms and treatment. Panminerva Med. 54,
Open science badges 161–170.
Green L. C., Wagner D. A., Glogowski J., Skipper P. L., Wishnok J. S.
This article has received a badge for *Open Materials* and Tannenbaum S. R. (1982) Analysis of nitrate, nitrite, and
because it provided all relevant information to reproduce the [15N]nitrate in biological fluids. Anal. Biochem. 126, 131–138.
study in the manuscript. The complete Open Science Hashioka S., Klegeris A., Monji A., Kato T., Sawada M., McGeer P. L.
Disclosure form for this article can be found at the end of and Kanba S. (2007) Antidepressants inhibit interferon-gamma-
the article. More information about the Open Practices induced microglial production of IL-6 and nitric oxide. Exp.
Neurol. 206, 33–42.
badges can be found at https://cos.io/our-services/open-scie
Jung K. H., Kim M. J., Ha E., Kim H. K., Kim Y. O., Kang S. A., Chung
nce-badges/. J. H. and Yim S. V. (2007) The suppressive effect of beta-glucan
on the production of tumor necrosis factor-alpha in BV2 microglial
cells. Biosci. Biotechnol. Biochem. 71, 1360–1364.
References Kim W. K., Jang P. G., Woo M. S., Han I. O., Piao H. Z., Lee K., Lee
H., Joh T. H. and Kim H. S. (2004) A new anti-inflammatory agent
Andrews P. W., Thomson J. A., Jr, Amstadter A. and Neale M. C.
KL-1037 represses proinflammatory cytokine and inducible nitric
(2012) Primum non nocere: an evolutionary analysis of whether
oxide synthase (iNOS) gene expression in activated microglia.
antidepressants do more harm than good. Front Psychol. 3,
Neuropharmacology 47, 243–252.
117.
Kischkel F. C., Hellbardt S., Behrmann I., Germer M., Pawlita M.,
Arbus C., Gardette V., Cantet C.E., Andrieu S., Nourhashemi F., Schmitt
Krammer P. H. and Peter M. E. (1995) Cytotoxicity-dependent APO-
L., Vellas B. and Group R.F. (2011). Incidence and predictive
1 (Fas/CD95)-associated proteins form a death-inducing signaling
factors of depressive symptoms in Alzheimer’s disease: the
complex (DISC) with the receptor. EMBO J. 14, 5579–5588.
REAL.FR study. J Nutr Health Aging. 15, 609–617.
Kronenberg G., Balkaya M., Prinz V. et al. (2012) Exofocal
Benfield P., Heel R. C. and Lewis S. P. (1986) Fluoxetine. A review of dopaminergic degeneration as antidepressant target in mouse
its pharmacodynamic and pharmacokinetic properties, and model of poststroke depression. Biol. Psychiatry 72, 273–281.
therapeutic efficacy in depressive illness. Drugs 32, 481–508. Lai A. Y. and Todd K. G. (2008) Differential regulation of trophic and
Bi X. L., Yang J. Y., Dong Y. X., Wang J. M., Cui Y. H., Ikeshima T., proinflammatory microglial effectors is dependent on severity of
Zhao Y. Q. and Wu C. F. (2005) Resveratrol inhibits nitric oxide neuronal injury. Glia 56, 259–270.
and TNF-alpha production by lipopolysaccharide-activated Lalancette-Hebert M., Gowing G., Simard A., Weng Y. C. and Kriz J.
microglia. Int. Immunopharmacol. 5, 185–193. (2007) Selective ablation of proliferating microglial cells
Boldin M. P., Goncharov T. M., Goltsev Y. V. and Wallach D. (1996) exacerbates ischemic injury in the brain. J. Neurosci. 27, 2596–
Involvement of MACH, a novel MORT1/FADD-interacting 2605.
protease, in Fas/APO-1- and TNF receptor-induced cell death. Li C. T., Bai Y. M., Tu P. C., Lee Y. C., Huang Y. L., Chen T. J., Chang
Cell 85, 803–815. W. H. and Su T. P. (2012) Major depressive disorder and stroke
Bushnell C. (2011) Depression and the risk of stroke in women: an risks: a 9-year follow-up population-based, matched cohort study.
identification and treatment paradox. Stroke 42, 2718–2719. PLoS ONE 7, e46818.
Castro V. M., Gallagher P. J., Clements C. C. et al. (2012) Incident user Lim C. M., Kim S. W., Park J. Y., Kim C., Yoon S. H. and Lee J. K.
cohort study of risk for gastrointestinal bleed and stroke in (2009) Fluoxetine affords robust neuroprotection in the
individuals with major depressive disorder treated with postischemic brain via its anti-inflammatory effect. J. Neurosci.
antidepressants. BMJ Open 2, e000544. Res. 87, 1037–1045.
Davalos D., Grutzendler J., Yang G., Kim J. V., Zuo Y., Jung S., Ling E. A., Ng Y. K., Wu C. H. and Kaur C. (2001) Microglia: its
Littman D. R., Dustin M. L. and Gan W. B. (2005) ATP mediates development and role as a neuropathology sensor. Prog. Brain Res.
rapid microglial response to local brain injury in vivo. Nat. 132, 61–79.
Neurosci. 8, 752–758. Liu S., Levine S. R. and Winn H. R. (2010) Targeting ischemic
Dhami K. S., Churchward M. A., Baker G. B. and Todd K. G. (2013) penumbra: part I—from pathophysiology to therapeutic strategy. J.
Fluoxetine and citalopram decrease microglial release of glutamate Exp. Stroke Transl. Med. 15, 47–55.

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Fluoxetine induces microglial apoptosis 15

Locksley R. M., Killeen N. and Lenardo M. J. (2001) The TNF and TNF Saura J., Tusell J. M. and Serratosa J. (2003) High-yield isolation of
receptor superfamilies: integrating mammalian biology. Cell 104, murine microglia by mild trypsinization. Glia 44, 183–189.
487–501. Schroeter M., Jander S., Witte O. W. and Stoll G. (1999) Heterogeneity
Loubinoux I., Kronenberg G., Endres M., Schumann-Bard P., Freret T., of the microglial response in photochemically induced focal
Filipkowski R. K., Kaczmarek L. and Popa-Wagner A. (2012) ischemia of the rat cerebral cortex. Neuroscience 89, 1367–1377.
Post-stroke depression: mechanisms, translation and therapy. J. Simmons A., Pimentel R. and Lakkireddy D. (2012) Sudden cardiac
Cell Mol. Med. 16, 1961–1969. death in women. Rev Cardiovasc Med. 13, e37–e42.
Masi G. and Brovedani P. (2011) The hippocampus, neurotrophic factors Slee E. A., Adrain C. and Martin S. J. (2001) Executioner caspase-3, -6,
and depression: possible implications for the pharmacotherapy of and -7 perform distinct, non-redundant roles during the demolition
depression. CNS Drugs 25, 913–931. phase of apoptosis. J. Biol. Chem. 276, 7320–7326.
Matsui T., Sekiguchi M., Hashimoto A., Tomita U., Nishikawa T. and Smoller J. W. (2011) Do antidepressants raise the risk of stroke? Am. J.
Wada K. (1995) Functional comparison of D-serine and glycine in Psychiatry 168, 457–459.
rodents: the effect on cloned NMDA receptors and the extracellular Stoll G., Jander S. and Schroeter M. (1998) Inflammation and glial
concentration. J. Neurochem. 65, 454–458. responses in ischemic brain lesions. Prog. Neurobiol. 56, 149–171.
Mikami K., Jorge R. E., Moser D. J., Arndt S., Jang M., Solodkin A., Sunami E., Usuda K., Nishiyama Y., Otori T., Katsura K. and Katayama
Small S. L., Fonzetti P., Hegel M. T. and Robinson R. G. (2011) Y. (2012) A preliminary study of fluvoxamine maleate on
Increased frequency of first episode poststroke depression after depressive state and serum melatonin levels in patients after
discontinuation of escitalopram. Stroke 42, 3281–3283. cerebral infarction. Intern. Med. 51, 1187–1193.
Muzio M., Chinnaiyan A. M., Kischkel F. C. et al. (1996) FLICE, a Takeuchi H., Mizuno T., Zhang G., Wang J., Kawanokuchi J., Kuno R.
novel FADD-homologous ICE/CED-3-like protease, is recruited to and Suzumura A. (2005) Neuritic beading induced by activated
the CD95 (Fas/APO-1) death-inducing signaling complex. Cell 85, microglia is an early feature of neuronal dysfunction toward
817–827. neuronal death by inhibition of mitochondrial respiration and
Nimmerjahn A., Kirchhoff F. and Helmchen F. (2005) Resting axonal transport. J. Biol. Chem. 280, 10444–10454.
microglial cells are highly dynamic surveillants of brain Takeuchi H., Jin S., Wang J., Zhang G., Kawanokuchi J., Kuno R.,
parenchyma in vivo. Science 308, 1314–1318. Sonobe Y., Mizuno T. and Suzumura A. (2006) Tumor necrosis
Obuchowicz E., Kowalski J., Labuzek K., Krysiak R., Pendzich J. and factor-alpha induces neurotoxicity via glutamate release from
Herman Z. S. (2006) Amitriptyline and nortriptyline inhibit hemichannels of activated microglia in an autocrine manner. J.
interleukin-1 release by rat mixed glial and microglial cell Biol. Chem. 281, 21362–21368.
cultures. Int. J. Neuropsychopharmacol. 9, 27–35. Tanaka S., Ide M., Shibutani T., Ohtaki H., Numazawa S., Shioda S. and
Park Y. K., Chung Y. S., Kim Y. S., Kwon O. Y. and Joh T. H. (2007) Yoshida T. (2006) Lipopolysaccharide-induced microglial
Inhibition of gene expression and production of iNOS and TNF- activation induces learning and memory deficits without neuronal
alpha in LPS-stimulated microglia by methanol extract of cell death in rats. J. Neurosci. Res. 83, 557–566.
Phellodendri cortex. Int. Immunopharmacol. 7, 955–962. Tynan R. J., Weidenhofer J., Hinwood M., Cairns M. J., Day T. A. and
Raskin S. and Durst R. (2010) Bupropion as the treatment of choice in Walker F. R. (2012) A comparative examination of the anti-
depression associated with Parkinson’s disease and its various inflammatory effects of SSRI and SNRI antidepressants on LPS
treatments. Med. Hypotheses 75, 544–546. stimulated microglia. Brain Behav. Immun. 26, 469–479.
Ried L. D., Jia H., Feng H., Cameon R., Wang X., Tueth M. and Wu S. Weintraub D., Mavandadi S., Mamikonyan E. et al. (2010) Atomoxetine
S. (2011) Selective serotonin reuptake inhibitor treatment and for depression and other neuropsychiatric symptoms in Parkinson
depression are associated with poststroke mortality. Ann. disease. Neurology 75, 448–455.
Pharmacother. 45(7–8), 888–897. Whirl-Carrillo M., McDonagh E. M., Hebert J. M., Gong L., Sangkuhl
Rivers C. S., Wardlaw J. M., Armitage P. A., Bastin M. E., Carpenter T. K., Thorn C. F., Altman R. B. and Klein T. E. (2012)
K., Cvoro V., Hand P. J. and Dennis M. S. (2006) Do acute Pharmacogenomics knowledge for personalized medicine. Clin.
diffusion- and perfusion-weighted MRI lesions identify final infarct Pharmacol. Ther. 92, 414–417.
volume in ischemic stroke? Stroke 37, 98–104. Wu S. Z., Bodles A. M., Porter M. M., Griffin W. S., Basile A. S. and
Rotzinger S., Todd K. G., Bourin M., Coutts R. T. and Baker G. B. Barger S. W. (2004) Induction of serine racemase expression and
(1997) A rapid electron capture gas chromatographic method for D-serine release from microglia by amyloid beta-peptide. J.
the quantification of fluvoxamine in brain tissue. J. Pharmacol. Neuroinflammation 1, 2.
Toxicol. Methods 37, 129–133. Wu C. F., Bi X. L., Yang J. Y., Zhan J. Y., Dong Y. X., Wang J. H.,
Sager J. E., Lutz J. D., Foti R. S., Davis C., Kunze K. L. and Isoherranen Wang J. M., Zhang R. and Li X. (2007) Differential effects of
N. (2014) Fluoxetine- and norfluoxetine-mediated complex drug- ginsenosides on NO and TNF-alpha production by LPS-activated
drug interactions: in vitro to in vivo correlation of effects on N9 microglia. Int. Immunopharmacol. 7, 313–320.
CYP2D6, CYP2C19, and CYP3A4. Clin. Pharmacol. Ther. 95(6), Yrjanheikki J., Tikka T., Keinanen R., Goldsteins G., Chan P. H. and
653–662. Koistinaho J. (1999) A tetracycline derivative, minocycline, reduces
Sangkuhl K., Klein T. E. and Altman R. B. (2011) PharmGKB inflammation and protects against focal cerebral ischemia with a
summary: citalopram pharmacokinetics pathway. Pharmacogenet. wide therapeutic window. Proc. Natl Acad. Sci. USA 96, 13496–
Genomics 21, 769–772. 13500.

© 2019 International Society for Neurochemistry, J. Neurochem. (2019) 10.1111/jnc.14661


Open Practices Disclosure

Manuscript Title: Fluoxetine and its metabolite norfluoxetine induce microglial apoptosis
Corresponding Author: Kathryn G. Todd

Articles accepted to Journal of Neurochemistry after 01.2018 are eligible to earn badges that
recognize open scientific practices: publicly available data, material, or preregistered research
plans. Please read more about the badges in our author guidelines and Open Science Badges
page, and you can also find information on the Open Science Framework wiki.

[X] Please check this box if you are interested in participating.

To apply for one or more badges acknowledging open practices, please check the box(es)
corresponding to the desired badge(s) below and provide the information requested in the
relevant sections. To qualify for a badge, you must provide a URL, doi, or other permanent path
for accessing the specified information in a public, open-access repository. Qualifying public,
open-access repositories are committed to preserving data, materials, and/or registered
analysis plans and keeping them publicly accessible via the web in perpetuity. Examples
include the Open Science Framework (OSF) and the various Dataverse networks. Hundreds of
other qualifying data/materials repositories are listed at http://re3data.org/. Preregistration of an
analysis plan must take place via a publicly accessible registry system (e.g., OSF,
ClinicalTrials.gov or other trial registries in the WHO Registry Network, institutional registration
systems). Personal websites and most departmental websites do not qualify as
repositories.

Authors who wish to publicly post third-party material in their data, materials, or preregistration
plan must have the proper authority or permission agreement in order to do so.

There are circumstances in which it is not possible or advisable to share any or all data,
materials, or a research plan publicly. For example, there are cases in which sharing
participants’ data could violate confidentiality. If you would like your article to include an
explanation of such circumstances and/or provide links to any data or materials you have made
available—even if not under conditions eligible to earn a badge—you may write an alternative
note that will be published in a note in the article. Please check this box if you would like your
article to include an alternative note and provide the text of the note below:

[] Alternative note:
[] Open Data Badge
1. Provide the URL, doi, or other permanent path for accessing the data in a public, open-
access repository:

[] Confirm that there is sufficient information for an independent researcher to reproduce


all of the reported results, including codebook if relevant.

[] Confirm that you have registered the uploaded files so that they are time stamped
and cannot be age.

[X] Open Materials Badge


1. Provide the URL, doi, or other permanent path for accessing the materials in a
public, open-access repository: all relevant information is provided in the manuscript and
custom-made materials will be provided upon reasonable request.

[X] Confirm that there is sufficient information for an independent researcher to


reproduce all of the reported methodology.

[] Confirm that you have registered the uploaded files so that they are time stamped
and cannot be age.

[] Preregistered Badge
1. Provide the URL, doi, or other permanent path to the registration in a public,
open-access repository*:

2. Was the analysis plan registered prior to examination of the data or observing the outcomes?
If no, explain.**

3. Were there additional registrations for the study other than the one reported? If yes, provide
links and explain.*
*No badge will be awarded if (1) is not provided, or if (3) is answered “yes” without strong
justification

**If the answer to (2) is “no,” the notation DE (Data Exist) will be added to the badge, indicating
that registration postdates realization of the outcomes but predates analysis.

By signing below, authors affirm that the above information is accurate and complete, that any
third-party material has been reproduced or otherwise made available only with the permission
of the original author or copyright holder, and that publicly posted data do not contain
information that would allow individuals to be identified without consent.

Date: 2018-12-21____________

Name: Matthew Churchward (for Kathryn Todd)

Signature: _____________________________

You might also like