You are on page 1of 17

SCIENCE SIGNALING | RESEARCH ARTICLE

ALCOHOL ADDICTION Copyright © 2020


The Authors, some
Daily alcohol intake triggers aberrant synaptic pruning rights reserved;
exclusive licensee
leading to synapse loss and anxiety-like behavior American Association
for the Advancement
of Science. No claim
Renato Socodato1*†, Joana F. Henriques1*, Camila C. Portugal1*, Tiago O. Almeida1, to original U.S.
Joana Tedim-Moreira1, Renata L. Alves1, Teresa Canedo1,2, Cátia Silva1, Ana Magalhães1, Government Works
Teresa Summavielle1†, João B. Relvas1,2†

Alcohol abuse adversely affects the lives of millions of people worldwide. Deficits in synaptic transmission and in
microglial function are commonly found in human alcohol abusers and in animal models of alcohol intoxication.
Here, we found that a protocol simulating chronic binge drinking in male mice resulted in aberrant synaptic pruning
and substantial loss of excitatory synapses in the prefrontal cortex, which resulted in increased anxiety-like be-
havior. Mechanistically, alcohol intake increased the engulfment capacity of microglia in a manner dependent on
the kinase Src, the subsequent activation of the transcription factor NF-B, and the consequent production of the
proinflammatory cytokine TNF. Pharmacological blockade of Src activation or of TNF production in microglia,
genetic ablation of Tnf, or conditional ablation of microglia attenuated aberrant synaptic pruning, thereby pre-
venting the neuronal and behavioral effects of the alcohol. Our data suggest that aberrant pruning of excitatory
synapses by microglia may disrupt synaptic transmission in response to alcohol abuse.

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


INTRODUCTION adaptations in brain regions linked with escalating alcohol con-
Alcohol abuse is associated with pathophysiological changes in the sumption, tolerance, dependence, and relapse (13, 14). Consistent
brain and peripheral organs, many of which can result in life-threatening with a neuroimmune hypothesis for alcohol addiction (11), knock-
conditions. The effects of alcohol in the central nervous system ing out various genes involved in immune responses, which, in the
(CNS) often lead to behavioral deficits (including anxiety, cognitive brain, are exclusively expressed or highly enriched in microglia, de-
decline, and motor dysfunction), and impairment of synaptic function, creases voluntary alcohol intake in mice.
a major hallmark of alcohol abuse, likely underlies such behavioral In this study, we found that binge-level alcohol intake (about five
deficits. Alcohol detrimentally affects the pre- and postsynaptic drinks for an average person) over 10 consecutive days enhanced
compartments and the secretion/recycling of neurotransmitters, Src-to–tumor necrosis factor (TNF) signaling in prefrontal cortex
ultimately leading to the disruption of excitatory and inhibitory microglia, which boosted their engulfment capacity and led to aber-
neurotransmission (1, 2). This detrimental effect of alcohol on syn- rant synaptic pruning, culminating in synapse loss and anxiety-like
apses can be contributed by a well-established action of alcohol on behavior. Overall, our data suggest that aberrant synaptic pruning
neurons and, potentially, through an underappreciated action of by microglia might play an important role in the synaptic transmis-
alcohol on glial cells (3). sion deficits elicited by alcohol abuse.
Microglia, the major innate immune cell population in the brain
(4), maintain nervous tissue homeostasis, surveilling the CNS
parenchyma by continuously extending and retracting their cellular RESULTS
processes, monitoring for tissue damage or infections, and checking Alcohol intake produces a microglia-driven neuroimmune
the functional status of synapses (5). Upon CNS tissue damage or response in the prefrontal cortex
infection, microglia become activated, changing their morphology In mouse models of alcohol intoxication, alcohol intake leads to
(into a more amoeboid shape), phagocytic capacity, and transcrip- microglia activation and oxidative damage (15). We sought to study
tional profile to restore tissue homeostasis (5). In many neuro- the effect of alcohol intake on microglia reactivity and used a protocol
psychiatric disorders, however, microglia immune function becomes of ethanol (EtOH) exposure simulating repetitive binge-level drink-
impaired, often leading to overproduction of inflammatory mediators ing. For this, we administrated 1.5 g/kg of EtOH (a dosage equivalent
and exacerbated phagocytic activity, which can be detrimental to to five drinks for an adult person weighing 75 kg) or water for
synapses and negatively affect behavior (6, 7). 10 consecutive days to a microglia reporter [Cx3cr1EYFP-CreER/+; (16)]
In human alcohol abusers (8) and in animal models of alcohol mouse (Fig. 1A). Immunohistochemistry on prefrontal cortex tissue
intoxication (9), alteration of microglial function associates with sections from Cx3cr1EYFP-CreER/+ mice revealed that this EtOH
neuroimmune activation (10, 11) and may be directly involved in some exposure protocol induced a significant expansion of prefrontal
of the neurotoxic and adverse behavioral effects of alcohol intake cortex microglia [yellow fluorescent protein–positive (YFP+) cells;
(12). Activation of the innate immune system contributes to neuro- Fig. 1B]. Using flow cytometry, we confirmed this increased number
of microglia in Cx3cr1EYFP-CreER/+ mice gavaged with EtOH (Fig. 1C).
1
Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular EtOH also increased the expression of the reactivity markers CD45,
e Celular, Universidade do Porto, 4200-135 Porto, Portugal. 2Department of Bio- integrin alpha M (CD11b), and Iba1 in prefrontal cortex microglia
medicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal. (Fig. 1, C and D). Morphometry of prefrontal cortex microglia
*These authors contributed equally to this work.
†Corresponding authors. Email: renato.socodato@ibmc.up.pt (R.S.); tsummavi@ (identified by Iba1 labeling) using Sholl analysis revealed that EtOH
ibmc.up.pt (T.S.); jrelvas@ibmc.up.pt (J.B.R.). induced microglia hyperamification, meaning increased branching

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 1 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

200
Fig. 1. Alcohol intake elicits A Adult Cx3cr1EYFP-CreER/+ mice B HO *

YFP+ cells per mm2


*
EtOH
microglia activation in the 2
150 *
*
*

prefrontal cortex. (A) Schematic EtOH (1.5 g/kg) H2O (CT) *


*

100
of the regimen of repetitive *
*
*
* *
*
*

binge-level alcohol (EtOH) in-


*
*
50 *
*
* *
*

take to adult Cx3cr1EYFP-CreER/+


EtOH or H2O * * *
Anti-YFP (green) *
0
mice. Mice were given EtOH

H
O

O
(1.5 g/kg) or water (H2O; control)

Et
H
2
1 2 3 4 5 6 7 8 9 10 (Days)
by oral gavage for 10 consecu-
tive days. (B) Histological con-

H
O
focal analysis of microglia C D

O
H
2

Et
(immunostained against YFP)
on tissue sections from pre- Microglia
Iba1
frontal cortices of Cx3cr1EYFP-CreER/+
mice exposed to EtOH or H2O.

CD11b
Asterisks indicate the microglial
SSA

GAPDH
FSA

YFP
cell body. Scale bar, 50 m. Data FSH FSA CD11b CD45
are means ± SEM from six mice
200
per condition pooled across * * *

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


15,000 3500 1500
three independent experiments. * 150

Iba1/GAPDH
12,500
*P < 0.05 by Mann-Whitney test. 1250
CD11b MFI
# Microglia

CD45 MF
10,000 3000 100
(C) Flow cytometry analysis of 7500 1000
microglial numbers in neo- 5000 2500 50
750
cortices of Cx3cr1EYFP-CreER/+ mice 2500
0
exposed to EtOH or H2O. Micro­ 0 2000 500

H
O
H

H
glia were identified as being

O
O

O
O

Et
H
2
Et

Et

Et
H

H
2

2
CD11b+ and Cd45low/mid gated
Gated on microglia
from the double-positive CD11b+
+
YFP population. Data are means ±
SEM from eight (H2O) and nine 8
H2O
# Intersections

(EtOH) mice pooled across three *


E HO EtOH
6 * EtOH
independent experim e n t s .
2 *
4 *
*P < 0.05 by Mann-Whitney test. *
2 *
FSH, follicle-stimulating hor- * F 250
mone. TBK, TANK Binding Kinase, 0
mRNA levels (%)
200 * H2O
TICAM, Toll Like Receptor Adap-
0
6
12
18
24
30
36
42
48
54
60

Iba1 (green)
* EtOH
Distance from soma (µm) 150
tor Molecule. (D) Western blot
100
for Iba1 on lysates from prefrontal * * * * *
* * *
EYFP-CreER/+
cortices of Cx3cr1 mice 20
* 25 50 *
Number of processes

exposed to EtOH or H2O. GAPDH 20 0


Process length

15
(loading control). Data are
12

G .1
C 4
C r1
Tg C
C 1
M B
tk
Tr r4
C 2
A
15
3

em
1q

1q
1q

er
Pu

sf

fb

Tl
ry

pr

means ± SEM from six mice 10


P2

10
per condition pooled across 5 5
three independent experiments.
0 0
*P < 0.05 by Mann-Whitney test.
H

(E) Sholl analysis retrieved from


O

O
O

O
Et

Et
H

H
2

histological con­focal images of


Iba1 on tissue sections from pre-
frontal cortices of Cx3cr1EYFP-CreER/+ G Biological process (GO) Reactome pathways
mice exposed to EtOH or H2O
Positive regulation of NF-κB transcription factor activity Apoptosis
(n = 50 cells from five animals Positive regulation of immune response TNFR1-induced NF-κB signaling pathway
Innate immune response Caspase activation via death receptors in the presence of ligand
per group for each distance). Activation of immune response TRAF6-mediated induction of TAK1 complex within TLR4 complex

Scale bar, 20 m. Data are Regulation of immune response


Immune system process
TRIF-mediated programmed cell death
Activation of IRF3/IRF7 mediated by TBK1/IKK epsilon
means ± SEM from 50 cells for Positive regulation of immune system process
Positive regulation of response to stimulus
Immune system
IKK complex recruitment mediated by RIP1
each distance from five mice Positive regulation of IκB kinase/NF-κB signaling Innate immune system
Regulation of immune system process TRIF(TICAM1)-mediated TLR4 signaling
per condition pooled across 0 5 10 15 20 0 5 10 15 20
three independent experiments. –Log10 P value (FDR) –Log10 P value (FDR)

*P < 0.05 by two-way ANOVA


with Sidak’s multiple comparisons. (F) qRT-PCR from neocortices of Cx3cr1EYFP-CreER/+ mice exposed to EtOH or H2O. The clustered histogram shows the Z scores of mRNA
transcripts normalized to the H2O values. Data are means ± SEM from five mice per condition pooled across three independent experiments. *P < 0.05 by Mann-Whitney test.
(G) Pathway enrichment analysis inputting the transcripts shown in (F). Graphs show the top 10 most overrepresented ontology terms in the GO biological process and the
Reactome pathways. TRAF, TNF receptor–associated factor; TAK, transforming growth factor –activated kinase; IRF, interferon regulatory factor; IKK, IB kinase. TBK, TANK bind-
ing kinase; TICAM, toll like receptor adaptor molecule.

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 2 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

(or intersections) and process number without affecting process Preventing TNF production suppresses microglia activation
length (Fig. 1E). EtOH significantly altered the expression levels of after alcohol intake
the microglia-enriched mRNA transcripts P2ry12, Pu.1, Gpr34, Microglia are critical producers of TNF after acute binge alcohol
Csfr1, C1qC, Tgfr1, C1qB, Mertk, Tlr4, Trem2, and C1qA in the intake (23). To test whether microglia were also major producers of
neocortex of Cx3cr1EYFP-CreER/+ mice (Fig. 1F). Enrichment analysis TNF during our alcohol exposure protocol, we genetically ablated
based on Gene Ontology using those altered transcripts revealed microglia using a microglial iDTR (inducible diphtheria toxin receptor)/
that several immune-associated pathways were significantly modified Cre-lox system (16). We first validated the efficiency of this microglia
by EtOH intake, including pathways altering microglial activity, ablation system in the context of our experimental requirements by
innate immune response, and TNF–nuclear factor B (NF-B) sig- giving tamoxifen to Cx3cr1EYFP-CreER/+:R26iDTR/+ and Cx3cr1EYFP-CreER/+
naling (Fig. 1G). mice at P28 and P30 and diphtheria toxin 2 months later (Fig. 3A).
The effect of EtOH on microglia was also confirmed in wild-type Both immuno­histochemistry on prefrontal cortex tissue sections and
mice, displaying similar results regarding microglial cell number flow cytometry showed, as expected, that microglia were efficiently
(fig. S1A), microglia-enriched mRNA transcripts (fig. S1B), and eliminated in Cx3cr1EYFP-CreER/+:R26iDTR/+ mice (Fig. 3, B and C).
microglia morphology (fig. S1C) compared with Cx3cr1EYFP-CreER/+ Next, we evaluated the production of TNF in the prefrontal cortex of
mice that carry only one functional Cx3cr1 allele. We then evaluated Cx3cr1 EYFP-CreER/+ and microglia-depleted (Cx3cr1 EYFP-CreER/+:
the effect of alcohol on other immune cell populations. Compared R26iDTR/+) mice after EtOH exposure (Fig. 3D). Whereas EtOH sig-
with water-treated controls, EtOH did not alter the number of nificantly increased the mRNA and the protein amounts of TNF in
macrophages (EYFP+CD11b+CD45hi cells), B lymphocytes (CD19+ Cx3cr1EYFP-CreER/+ mice (Fig. 3, E and F), it did not increase the
CD45+CD11b−EYFP− cells), or T lymphocytes (CD3+CD45+CD11b− amounts of TNF in microglia-depleted mice (Fig. 3, E and F),

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


EYFP− cells) in the neocortex of Cx3cr1EYFP-CreER/+ mice (fig. S1, suggesting that microglia are the primary producers of TNF in the
D to F). Astrocytes also perform immune cell functions within the prefrontal cortex during EtOH exposure.
brain parenchyma, and alcohol exposure leads to astrocytic activa- We next used TNF-deficient [TNF knockout (KO)] mice or the
tion through several mechanisms (15, 17–22). Therefore, we tested immunomodulatory and brain-penetrant TNF blocker pomalido-
whether our EtOH exposure protocol would modulate astrocytic mide (PMD) (24–26) to test whether TNF production could lead to
reactivity. We found no significant difference in the number of pre- microgliosis. Using flow cytometry, we found that the EtOH-induced
frontal cortex astrocytes [glial fibrillary acidic protein–positive (GFAP+) microgliosis (Fig. 3G) was prevented entirely in TNF KO mice
cells] or in the expression of GFAP in EtOH-treated mice (fig. S1, G (Fig. 3G) or wild-type mice treated with PMD (Fig. 3G). The EtOH-­
and H), which might suggest that our experimental paradigm of induced alteration of the microglia-enriched genes P2ry12, Pu.1,
repetitive binge-level drinking did not produce a strong reaction in Gpr34, Csfr1, C1qC, Tgfr1, Mertk, Tlr4, Trem2, and C1qA was also
astrocytes. abrogated in TNF KO mice (Fig. 3H), indicating that microglial
Because our alcohol exposure protocol triggered microgliosis, TNF is involved in the microgliosis elicited by EtOH exposure.
we evaluated the transcript abundance of genes involved in classical
immune responses and antioxidant defenses in mice exposed to Alcohol intake modulates the Src–NF-B pathway
EtOH. We found that EtOH increased the transcript levels of Tlr2, to increase TNF production and trigger microglia activation
Cd14, Icam1, Slc23a2, Tspo, Tnf, Rip1, and Traf2 and decreased the Because the tyrosine kinase Src controls microglial activation via
transcript levels of Il-6, Ccl5, Tlr7, Mhc-II, Gclc, Traf1, Ccl2, Hmox1, increased production and secretion of TNF (27–29), we investigated
Cox2, Irak3, Tnxnrd1, and Gsr in the neocortex of Cx3cr1EYFP-CreER/+ whether EtOH could trigger the production of TNF in microglia via
mice (Fig. 2A). Moreover, other immune-related genes such as Il-18, Src. We found that the amount of active Src (phospho-Src Tyr416) was
Il-1b, Inf-b, Cd163, Socs3, Nrlp3, Nos2, Cx3cl1, Cxcl10, Arg1, Mrp8, significantly increased in the prefrontal cortex of Cx3cr1EYFP-CreER/+
Mrp14, Cxcl1, and Spp1 were not significantly altered by EtOH mice exposed to EtOH (Fig. 4A), but not in the prefrontal cortex of
(Fig. 2B). Pathway analysis of the differentially expressed genes microglia-depleted mice exposed to EtOH (Fig. 4A). In line with
after EtOH exposure revealed a substantial number of signifi- this, the effect of alcohol in increasing microglial Src activation was
cantly enriched pathways that were both directly and indirectly thoroughly reproduced in a cell-autonomous manner in primary
modulated by TNF (Fig. 2C). Most of those enriched pathways cortical microglia (analyzed by immunocytochemistry with an anti-
were not only related to immune responses but also included body against phospho-Src Tyr416; fig. S2A) and in the CHME3 micro­
cell migration, cell proliferation, cell adhesion, intracellular signal glial cell line [analyzed by live-cell imaging using a fluorescence
transduction, and cytokine biosynthesis and secretion (Fig. 2C). resonance energy transfer (FRET)–based Src biosensor; fig. S2B].
Network analysis of protein-protein interactions revealed the To confirm the relationship between Src activity and TNF ex-
prevalence of 10 major protein clusters to be modulated by EtOH pression, we used the clinically relevant Src blocker AZD0530 (30, 31)
intake (Fig. 2D). The TNF cluster was more intricately associated during EtOH exposure. We found that inhibiting Src with AZD0530
with Toll-like receptor 2 (TLR2) and interleukin-1 receptor– (fig. S2C) abrogated the increase in TNF elicited by EtOH (Fig. 4B). To
associated kinase 3 (IRAK3) clusters (Fig. 2D), suggesting that further confirm that alcohol modulates microglial Src to increase the
the alteration of neuroimmune pathways after EtOH exposure production of TNF, we inhibited Src, using the Src blocker SKI-1 (Src
might be coregulated via TNF, TLR2, and IRAK3. In addition, kinase inhibitor), in primary cortical microglia and found that SKI-1
several interacting proteins within the TNF cluster (Fig. 2D) are prevented the alcohol-­mediated increase of TNF production (fig. S2D).
known modulators/integrators of brain immune responses and To gain further mechanistic insight into how Src drives TNF
microglial activation, thereby indicating that the neuroimmune production in microglia during alcohol exposure, we focused on the
response triggered by EtOH intake might be strongly associated role of NF-B, a central regulator of inflammatory signaling in im-
with TNF. mune cells (32, 33). We measured the nuclear accumulation of the

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 3 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

A 300
* B 250
H2O
p65 subunit of NF-B as a functional
200 indicator of NF-B activation in CHME3

mRNA levels (%)


mRNA levels (%)
* EtOH
200
*
* * * 150
microglia using a green fluorescent pro-
* * *
100
* * *
* * *
100
tein (GFP)–tagged p65 construct. We
* * * * *
* 50
found that exposure to alcohol increased
0 0
nuclear GFP content in CHME3 micro­

3
b

M 8

2
p1
C b
Tl 6

g1

14
M 14

So 3
IL f

C l1
Tr sr
Tl 2
C r7

3
2
am 5

N 1
Tr k1

0
n 3

R f2

1
I 2

c2 lc
C II

C o
Ts -1

2
Sl Gc 1
Tn

H rd1

-1

16
-1

rp

os
-
-

rlp
r

cs
cl
I c Cc l glia (Fig. 4C) and that knocking down

ip

cl

l1
Tx rak

af
ox
-

3a

xc
ox

Ifn
G
a
hc

Sp
Ar

rp
D

IL

x3
IL

xc

N
D

N
C
m

C
Src (fig. S2E) abrogated the alcohol-­
C induced nuclear translocation of GFP-­p65

d
Regulation of programmed cell death

te
d

la
Regulation of anatomical structure morphogenesis

te

gu
Homeostatic process

la
Response to endogenous stimulus

subunit (Fig.  4C). Pharmacological

-re
gu
Regulation of transport

wn
-re
Regulation of apoptotic process

Up

Do
Regulation of cell proliferation Response to organic substance

blockade of Src using AZD0530 or SKI-1


Regulation of developmental process
Leukocyte migration

Significance
Regulation of homeostatic process Positive regulation of cellular metabolic process
Response to stress

Regulation of multicellular organismal process


Response to chemical stimulus

Regulation of biological quality


Neuron apoptotic process
also prevented the alcohol-induced ac-
Cytokine production
Positive regulation of metabolic process
Positive regulation of multicellular organismal process

Immune response MAPK cascade


tivation of the NF-B pathway, moni-
tored using the IB-miRFP703 reporter
Positive regulation of transport Cell migration
Immune system process Cell activation
Positive regulation of cell proliferation
Cytokine-mediated
signaling pathway
Positive regulation of immune system process
Regulation of immune system process
Multiorganism process
Regulation of protein metabolic process (34) (fig. S2F). Moreover, direct activa-
tion of Src in CHME3 microglia, using
Regulation of neuron apoptotic process
Regulation of immune effector process Immune effector process
Intracellular protein kinase cascade
Positive regulation of

rapamycin-based chemogenetics with


protein metabolic process Positive regulation of response to stimulus
Regulation of cell adhesion Inflammatory response Response to other organism
Vitamin metabolic process Regulation of response to stimulus

RapR-Src/FRB (FKBP–rapamycin binding)


Response to wounding Response to biotic stimulus
Response to bacterium Intracellular signal transduction Regulation of sequence_specific DNA binding transcription factor activity

(35), increased the nuclear translocation


Negative regulation of multicellular organismal process
Defense response Regulation of cell-cell adhesion
Regulation of molecular function Regulation of signal transduction

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


Positive regulation of sequence-specific DNA binding transcription factor activity

of the GFP-p65 subunit (fig. S2G). Paral-


Positive regulation of NF-κB transcription factor activity
Secretion by cell
Positive regulation of defense response
Regulation of defense response
Response to virus
Positive regulation of protein secretion
Regulation of IκB kinase/NF-κB cascade
Positive regulation of cytokine production
Regulation of immune response
leling the increase of NF-B activation
Protein import into nucleus Production of molecular mediator of immune response
IκB kinase/NF-κB cascade Positive regulation of immune response after Src activation, overexpressing the
constitutively active Src mutant SrcY527F
Regulation of cytokine secretion
Regulation of protein import into nucleus Regulation of cytokine biosynthetic process
Protein import Positive regulation of cytokine biosynthetic process
Cytokine metabolic process
Interleukin-8 production
Positive regulation of cytokine secretion Regulation of intracellular transport
Cytokine biosynthetic process
led to increased secretion of TNF from
Nuclear import

Interleukin-8 biosynthetic process


Regulation of nucleocytoplasmic transport
microglia (fig. S2H). Last, preventing NF-
B activation with the clinically relevant
inhibitor sulfasalazine (36, 37) completely
blocked the production of TNF elicited
D Tlr2 by SrcY527F in micro­glia (Fig. 4D). These
Tspo data suggest that alcohol drives TNF
k
or
er

production in microglia via Src and down-


tw
st
clu

ne
n
n

Pt
Pt

Degree of interaction

Degree of overlap

Tlr7
stream NF-B activation.
Tyk2
In line with such Src-dependent TNF
Casp1
Rhoa production during alcohol exposure,
in vivo blockade of Src with AZD0530
Rac1
Lyn Cav1
Ccl2
Hck
abrogated the EtOH-induced increase
Cd68
Fyn Jak2
Lck

Irak3
Fgr
Src
Tnfrsf1b
Notch1 of microglial numbers and prevented
Nfkbia
Nfkbie

Casp3
Ccl5 the EtOH-mediated alteration of the
Casp8
Adam10
Traf2 microglia-enriched genes P2ry12, Pu.1,
Csf1
Adam17 Gpr34, Csfr1, C1qC, Tgfr1, Mertk,
Ikbkg
Lamp2
Casp9
Tlr4, Trem2, and C1qA (Fig. 4, E and F).
Rela
Tnfrsf1a

Blocking microglial TNF signaling


Lamp1

Tnf
suppresses anxiety-like behavior
Hdac4

elicited by alcohol intake


Icam1 The neuroimmune hypothesis of alcohol
addiction (11) supports the notion that
Gclc Hmox1
alterations of neuroimmune signaling
might be responsible for some of the
Fig. 2. Alcohol intake triggers a TNF-associated neuroimmune response. (A and B) qRT-PCR from neocortices of behavioral deficits elicited by alcohol
Cx3cr1EYFP-CreER/+ mice exposed to EtOH or H2O. The clustered histogram shows the Z scores of mRNA transcripts abuse. Because EtOH induced the pro-
normalized to the H2O values. Transcripts differentially altered by EtOH are shown in (A); those not significantly al- duction of TNF by prefrontal cortex
tered by EtOH are shown in (B). Data are means ± SEM from five mice per condition pooled across three independent microglia, we tested whether EtOH could
experiments. *P < 0.05 by Mann-Whitney test. (C) Pathway enrichment analysis inputting the transcripts shown in (A).
drive changes in behavior. We found,
The topographic pathway map shows overrepresented (orange) and underrepresented (purple) terms (nodes) and
their degree of interaction (gray lines). Color ramps display the significance enrichment of each node. All nodes
using the elevated plus maze (EPM) test,
shown in the map are associated directly or indirectly with TNF signaling. MAPK, mitogen-activated protein kinase. that EtOH increased anxiety-like be-
(D) String-based protein-protein interaction (PPI) networks were constructed, inputting the transcripts shown in (A). havior in adult Cx3cr1EYFP-CreER/+ mice,
Protein clusters that were significantly modulated by EtOH are color-coded on the basis of their degree of association as revealed by the decreased time that
and significance, as indicated. EtOH-subjected mice spent on the open

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 4 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

Fig. 3. TNF production elic- A B Cx3cr1EYFP-CreER/+ Cx3cr1EYFP-CreER/+:R26iDTR/+ C 20,000 *


ited by alcohol intake drives Tamox DT
Cx3cr1EYFP-CreER/+
15,000
micro­glia activation in the

# Microglia
Cx3cr1EYFP-CreER/+:R26iDTR/+
prefrontal cortex. (A) Sche- 10,000
matic of tamoxifen-inducible P28 30 90 91 92 (Days)
5000
microglial ablation in the brain Iba-1 (green)
0
from postnatal day 28 (P28). DAPI (blue)

Tamox, tamoxifen; DT, diph-


theria toxin. (B) Histological E 500

TNF mRNA levels (%)


* Cx3cr1EYFP-CreER/+
confocal analysis for Iba1 on D DT 400
Tamox EtOH or H2O
tissue sections from prefrontal 300 Cx3cr1EYFP-CreER/+:R26iDTR/+

cortices of Cx3cr1EYFP-CreER/+ 200


and Cx3cr1EYFP-CreER/+:R26iDTR/+ P28 30 84 85 86 87 88 89 90 91 92 93 (Days)
100
after exposure to DT (n = 4
0
animals per genotype). Scale

2 O

H
2 O

H
bar, 50 m. (C) Flow cytometry

O
H

H
Et

Et
analysis of microglial numbers F Cx3cr1EYFP-CreER/+ Cx3cr1EYFP-CreER/+ Cx3cr1EYFP-CreER/+:R26iDTR/+ Cx3cr1EYFP-CreER/+:R26iDTR/+
in neocortices of Cx3cr1EYFP-CreER/+ 20 * Cx3cr1EYFP-CreER/+
H 2O EtOH H2O EtOH
EYFP-CreER/+ iDTR/+
and Cx3cr1 :R26 15 Cx3cr1EYFP-CreER/+:R26iDTR/+

% TNF area
after exposure to DT. Data are 10
means ± SEM from six mice

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


115 5
per condition pooled across
three independent experiments. 0

*P < 0.05 by Mann-Whitney test.


TNF

H
2 O

H
20

O
H

H
2

Et

Et
(D) Schematic of microglial
ablation during EtOH expo-
sure. (E) qRT-PCR from neo- G H 2O EtOH
cortices of Cx3cr1EYFP-CreER/+
105

105

*
and Cx3cr1EYFP-CreER/+:R26iDTR/+
15,000
DMSO
after microglia ablation during
104

104

# Microglia
DMSO

10,000
exposure to EtOH or H2O. Histo­ PMD
103

103

gram shows the Z scores of 5000 TNF KO

TNF transcripts normalized to


0

the Cx3cr1EYFP-CreER/+-H2O val- 0


O

H
O

O
ues. Data are means ± SEM from 0 10 10 10 0 10 10 10
3 4 5 3 4 5
O

O
H
2

Et

Et

Et
H

H
2

five mice per condition pooled 2

across three independent ex-


105

105

periments. *P < 0.05 by two-way H WT - H2O TNF KO - H2O


200
104

104

ANOVA with Sidak’s multiple WT - EtOH TNF KO - EtOH


PMD

*
mRNA levels (%)

comparisons. (F) Histological 150 *


103

103

confocal analysis for TNF on


100
0

tissue sections from prefrontal * * * * * *


EYFP-CreER/+ * *
cortices of Cx3cr1 and 0 10 10
3
104 5
0 10 10
3 4
10 5
50
Cx3cr1EYFP-CreER/+:R26iDTR/+ after
0
microglia ablation during ex-
12

34

r1

tk

r4

A
posure to EtOH or H2O. Scale
105

105

1q
1q

er
Pu

sf

fb

Tl
ry

pr

e
C

M
Tg

C
C
P2

Tr
G

bar, 100 m. Data are means ±


104

104
TNF KO

SEM from five mice per con-


dition pooled across three
103

103

independent experiments.
0

*P < 0.05 by two-way ANOVA


CD45

with Sidak’s multiple com- 0 10 10


3
104 5
0 10 10
3 4
10 5

parisons. (G) Flow cytometry CD11b

analysis of microglial numbers in neocortices of wild-type (WT) or TNF knockout (KO) mice exposed to EtOH or H2O. WT animals were also treated with DMSO or with
pomalidomide (PMD). Data are means ± SEM from at least eight mice per condition pooled across three independent experiments. *P < 0.05 by two-way ANOVA with
Sidak’s multiple comparisons. (H) qRT-PCR from neocortices of WT or TNF KO mice exposed to EtOH or H2O. Histogram shows the Z scores of mRNA transcripts normalized
to the WT-H2O values. Data are means ± SEM from five mice per condition pooled across three independent experiments. *P < 0.05 by two-way ANOVA with Sidak’s mul-
tiple comparisons.

arms (Fig. 5A). No differences were found in the distance traveled the open field (OF) test, as mice exposed to EtOH spent less time
in the EPM between EtOH-treated and water-treated mice (Fig. 5B). in the center of the OF arena than water-treated littermates
The anxiogenic effect of EtOH could also be observed in wild-type (fig. S3C).
mice in the EPM test (fig. S3, A and B). Moreover, the increase of EtOH did not produce significant changes in (i) general loco-
anxiety-like behavior elicited by EtOH exposure was reproduced in motor activity (fig. S3D) or repetitive behavior (fig. S3E) when mice

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 5 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

Fig. 4. Src activation elicited A 500 B 300 *

TNF mRNA levels (%)


DMSO

H
*

O
by alcohol intake leads to

% Src pTyr416/Src
H

H
2

2
Et

Et
400 Cx3cr1 EYFP-CreER/+ AZD0530
TNF production driving pre­ Src pTyr416 200
300
frontal cortex microglia acti- Cx3cr1EYFP-CreER/+:R26iDTR/+

vation. (A) Western blot analysis Src


200
100
for Src phospho-Tyr416 (active 100
Cx3cr1 Cx3cr1
EYFP-CreER/+ EYFP-CreER/+

form) on lysates from prefrontal : 0 0


cortices of Cx3cr1EYFP-CreER/+
R26 iDTR/+

2 O

H
2 O

2 O

H
2 O

H
O

O
H

H
and Cx3cr1EYFP-CreER/+:R26iDTR/+

Et

Et

Et

Et
after microglia ablation during
exposure to EtOH or H2O. Src
(loading control). Data are
C EV - CT EV - EtOH shSrc - CT shSrc - EtOH D EV SrcY527F SrcY527F + Sulfa
means ± SEM from five mice
100.0
per condition pooled across
three independent experiments.
*P < 0.05 by two-way ANOVA p65-GFP TNF 1.0
with Sidak’s multiple com-
parisons. (B) qRT-PCR from
neocortices of Cx3cr1EYFP-CreER/+
mice exposed to EtOH or H2O.
Mice were treated with either

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


DAPI CD11b
DMSO or the brain-penetrant
20,000 *
Src inhibitor AZD0530. Data *
*
GFP fluorescence

2.5
are means ± SEM from five mice 15,000

% TNF fluorescence
2.0
per condition pooled across
(nuclear)

10,000 1.5
three independent experi-
1.0
ments. *P < 0.05 by two-way 5000
0.5
ANOVA with Sidak’s multiple 0 0.0
comparisons. (C) CHME3 micro­

c Y5 S EV
+ 527
lfa
s Et T

c- CT

27 rc Y
H
sh hS OH
- -C

glial cultures expressing Src

Su
O
Sr rc-
EV EV

Et

or control shRNA were trans-

Sr
fected with a GFP-tagged p65
NF-B subunit (p65-GFP) con-
struct and were treated for
24 hours with EtOH (70 mM)
E H 2O EtOH 15,000
* DMSO
or H2O. Scale bar, 10 m. Data AZD0530
105
105

10,000
# Microglia

are means ± SEM from eight


104
104

independent cultures per


DMSO

5000
condition. *P < 0.05 by one-
103
103

0
way ANOVA with Sidak’s mul-
0
0

tiple comparisons. (D) N9


O

O
H

H
2

2
Et

Et

microglial cultures expressing 0 10 10 10 3 0 10 10 4 10 5 3 4 5

an empty pMSCV vector (EV) F 200 DMSO - H2O AZD0530 - H2O


*
Y527F
or the Src mutant were
105
105

DMSO - EtOH AZD0530 - EtOH


mRNA levels (%)

*
150
treated with 1 mM sulfasalazine
AZD0530

104
104

for 24 hours and immuno­ 100


*
stained for TNF and CD11b.
103
103

* * * * *
50 *
TNF fluorescence intensity per *
0
0

individual microglia in each


CD45

0
cell culture is coded according 0 10 10 10 3 0 10 104 10 5 3 4 5
12

.1

34

r1

tk

r4

A
br

1q
1q

er
Pu

sf

Tl
ry

pr

to the indicated pseudocolor CD11b


e
f
C

M
Tg

C
C
P2

Tr
G

scale. Scale bar, 20 m. Data


are means ± SEM from at least four independent cultures per condition. *P < 0.05 by one-way ANOVA with Sidak’s multiple comparisons. (E) Flow cytometry analysis of
microglial numbers in neocortices of Cx3cr1EYFP-CreER/+ mice exposed to EtOH or H2O. Mice were treated with either DMSO or AZD0530. Data are means ± SEM from eight
mice per condition pooled across three independent experiments. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons. (F) qRT-PCR from neocortices of
Cx3cr1EYFP-CreER/+ mice exposed to EtOH or H2O. Mice were also treated with DMSO or with AZD0530. The clustered histogram shows the Z scores of mRNA transcripts
normalized to the DMSO-H2O values. Data are means ± SEM from five mice per condition pooled across three independent experiments. *P < 0.05 by two-way ANOVA
with Sidak’s multiple comparisons.

were evaluated in the OF test and (ii) recognition memory when or directly (by inhibiting TNF expression with PMD) completely
mice were assessed on the novel object recognition test (fig. S3, F and G). suppressed the EtOH-induced increase in anxiety-like behavior
We then investigated whether the anxiogenic effect of EtOH in- (Fig. 5, A and B). To further confirm the role of microglia in the
take required the production of TNF by microglia. We found that anxiogenic effect of EtOH intake, we evaluated microglia-depleted
blocking TNF production indirectly (by inhibiting Src with AZD0530) mice in the EPM test. Whereas EtOH increased anxiety-like behavior

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 6 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

A 15 * B 2000 In pathological conditions, such as

Distance traveled (cm)


% Time in open arms

DMSO
1500
in Alzheimer’s disease, microglia can
AZD0530
10 phagocytose and prune healthy synapses,
1000 PMD a process termed synaptophagy (7). We
5
500 hypothesized that the EtOH-mediated
loss of excitatory synapses in the pre-
0 0
frontal cortex could be a direct conse-
O

2 O

2 O

H
quence of excessive engulfment of synaptic
O

O
H

H
2

2
Et

Et

Et

Et

Et

Et
structures by microglia. To investigate
whether microglia exposed to EtOH en-
gulfed more synapses in vivo, we evaluated
C 15 * D 2000 Distance traveled (cm) by immunofluorescence on prefrontal
% Time in open arms

Cx3cr1EYFP-CreER/+

1500 Cx3cr1EYFP-CreER/+
:R26iDTR/+ cortex tissue sections the amount of
10
PSD-95+ puncta within phagocytic struc-
1000 tures (labeled with CD68) in Iba1 +
5
500 microglia. Confocal imaging coupled
0
with 3D cell surface rendering revealed
0
that Iba1+ microglia from the prefrontal
O
O

H
H

O
O

H
H
O
O

cortex of mice exposed to EtOH con-


H
2

O
H
2

H
2
H
2
Et
Et

Et
Et

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


Fig. 5. Alcohol intake elicits anxiety-like behavior in a microglia-dependent manner. (A and B) Cx3cr1EYFP-CreER/+ tained significantly more PSD-95+ puncta
mice were injected with DMSO, AZD0530, or PMD; exposed to EtOH or H2O; and then evaluated in the elevated plus colocalizing with CD68 structures than
maze (EPM) test. Data are means ± SEM from at least nine mice per condition pooled across at least from three inde- prefrontal cortex microglia from mice
pendent experiments. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons. (C and D) Cx3cr1EYFP-CreER/+ gavaged with water (Fig. 7A). This
and Cx3cr1EYFP-CreER/+:R26iDTR/+ mice were given tamoxifen, treated with diphtheria toxin to ablate microglia during
microglial engulfment of prefrontal cor-
exposure to EtOH or H2O, and then evaluated in the EPM. Data are means ± SEM from six mice per condition pooled
tex postsynaptic elements required TNF
across three independent experiments. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons.
signaling because no significant differ-
ences in the engulfment of PSD-95 +
in control (Cx3cr1EYFP-CreER/+) mice, this effect was significantly pre- puncta were observed in TNF KO mice exposed to EtOH (Fig. 7A).
vented in microglia-depleted (Cx3cr1EYFP-CreER/+:R26iDTR/+) mice Conversely, EtOH neither increased PSD-95 engulfment by
(Fig. 5C). No differences were found in the distance traveled in the Iba1 + microglia in the CA1 region of the dorsal hippocampus
EPM between the genotypes or treatment groups (Fig. 5D). (fig. S5B) nor triggered the production of TNF in the dorsal hippo-
campus (fig. S5C).
Alcohol-induced TNF signaling increases microglia To further assess the engulfment of synapses by microglia, we
phagocytic activity, enabling microglia to prune prefrontal prepared synaptosomes (to isolate synaptic terminals) from the pre-
cortex synapses frontal cortex of adult mice and incubated microglial cultures with
Behavioral alterations, such as increased anxiety, can be caused by them. Immunofluorescence labeling of PSD-95 confirmed that
perturbations in the excitatory/inhibitory balance in prefrontal microglia efficiently engulfed synaptosomes prepared from the pre-
cortex circuits (38, 39), and exacerbation of microglia activation de- frontal cortex (Fig. 7B), further confirming that microglia ac-
creases the number of excitatory synapses in the neocortex (40). We tively phagocytose synapses in steady-state conditions. However,
found that the amounts of PSD-95 and vGlut1 (major proteins in- cultured microglia exposed to EtOH engulfed significantly more
volved in excitatory synaptic transmission) along with the number prefrontal cortex synaptosomes than control microglia (Fig. 7B).
of excitatory synapses (PSD-95+/vGlut1+ puncta) were significantly To further corroborate the excessive EtOH-mediated engulf-
decreased in prefrontal cortices of mice exposed to EtOH (Fig. 6A ment of synaptic structures, microglia cultures were incubated
and fig. S4A). The decrease of PSD-95 puncta, vGlut1 puncta, and with synaptosomes prepared from the prefrontal cortex of Thy1-
PSD-95+/vGlut1+ synapses elicited by EtOH intake was prevented YFP mice. Measuring microglia engulfment capacity by flow
in TNF KO mice (Fig. 6A and fig. S4A) and in microglia-depleted cytometry revealed that microglia exposed to EtOH displayed
mice (Fig.  6B and fig. S4B). Biochemical assessment further con- significantly increased engulfment of YFP+ synaptosomes com-
firmed that EtOH intake decreased, in a microglia-dependent pared with control microglia (Fig. 7C). Blocking TNF with the
manner, the amounts of PSD-95 and vGlut1 in the prefrontal cortex neutralizing antibody adalimumab (HUMIRA) completely pre-
(Fig. 6C), indicating that TNF production by microglia leads to loss vented the engulfment of YFP+ synaptosomes elicited by exposure
of excitatory synapses in the prefrontal cortex after EtOH intake. to EtOH (Fig. 7C).
Loss of synapses can be a consequence of increased neuronal cell In line with the view that EtOH intake increases microglia engulf-
death. However, we found that the number of neurons (stained with ment capacity, mice exposed to EtOH displayed a TNF-dependent
NeuN) in the prefrontal cortex was not significantly different between increase of the microglia engulfment module (represented by tran-
EtOH-subjected and water-treated mice (fig. S4C). The numbers of scripts coding for Adam12, Apoe, Axl, Ccr1, CD93, Cybb, Lyz2, Mrc1,
excitatory synapses in the CA1 region of the dorsal hippocampus of and Siglec1) (41) (Fig. 7D) and had a TNF-dependent increase of
EtOH-subjected mice were not significantly different from those prefrontal cortex microglia expressing the phagocytic marker CD68
of water-treated littermates (fig. S5A), which may help to explain (Fig. 7E). In this context, TNF signaling enabled microglial phago-
the specificity of the behavioral phenotype elicited by EtOH. cytosis because EtOH increased the phagocytic activity of cortical

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 7 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

A WT WT TNF KO TNF KO
H2O EtOH H2O EtOH

PSD-95 (red)
vGlut1 (green)

% PSD-95+/vGlut1+ puncta
WT
150 150 150
PSD-95 labeling area

vGlut1 labeling area


*** TNF KO
**** ****

(% of control)
(% of control)

100 (% of control) 100 100

50 50 50

0 0 0

H
O

O
O

H
2

2
H

H
2

H
2

Et

Et
Et

Et

Et

Et

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


B Cx3cr1EYFP-CreER/+ Cx3cr1EYFP-CreER/+ Cx3cr1EYFP-CreER/+:R26iDTR/+ Cx3cr1EYFP-CreER/+:R26iDTR/+
H2O EtOH H2O EtOH

PSD-95 (red)
vGlut1 (green)
% PSD-95+/vGlut1+ puncta

150 150 150 Cx3cr1EYFP-CreER/+


PSD-95 labeling area

vGlut1 labeling area

*** Cx3cr1EYFP-CreER/+:R26iDTR/+
**** ****
(% of control)
(% of control)

(% of control)

100 100 100

50 50 50

0 0 0
O

H
O

H
O

O
O

O
O

H
2

2
H

H
2

2
H

H
2

Et

Et
Et

Et
Et

Et
H

H
O

O
O

C
H

H
2

2
Et

Et

150 150 Cx3cr1EYFP-CreER/+


% PSD-95/GAPDH

% vGlut1/GAPDH

PSD-95 * * Cx3cr1EYFP-CreER/+:R26iDTR/+
100 100

vGlut1
50 50

GAPDH 0 0
O

2 O

H
O

O
H

H
2

Cx3cr1 EYFP-CreER/+
Cx3cr1EYFP-CreER/+
Et

Et

Et

Et

:
R26iDTR/+

Fig. 6. Alcohol intake elicits synapse loss in a microglia-dependent manner. (A) Confocal imaging analysis for PSD-95 (red) and vGlut1 (green) on tissue sections from
prefrontal cortices of WT and TNF KO mice after exposure to EtOH or H2O. Scale bar, 5 m. Data are means ± SEM from at least six mice per condition pooled across three
independent experiments. ****P < 0.0001 and ***P < 0.001 by two-way ANOVA with Sidak’s multiple comparisons. (B) Confocal imaging analysis for PSD-95 (red) and vGlut1
(green) on tissue sections from prefrontal cortices of Cx3cr1EYFP-CreER/+ and Cx3cr1EYFP-CreER/+:R26iDTR/+ mice after microglia ablation during exposure to EtOH or H2O. Scale
bar, 5 m. Data are means ± SEM from at least six mice per condition pooled across three independent experiments. ****P < 0.0001 and ***P < 0.001 by two-way ANOVA
with Sidak’s multiple comparisons. (C) Western blot analysis for PSD-95 and vGlut1 on lysates from prefrontal cortices of Cx3cr1EYFP-CreER/+ and Cx3cr1EYFP-CreER/+:
R26iDTR/+ mice after microglia ablation during exposure to EtOH or H2O. GAPDH (loading control). Data are means ± SEM from five mice per condition pooled across three
independent experiments. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons.

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 8 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

4000

CD68+/PSD-95+ puncta
Fig. 7. Alcohol intake enables A WT - H O WT
WT - EtOH TNF KO - H2O TNF KO - EtOH *

within Iba1+ cell


microglia to prune synapses. 2 3000 TNF KO
(A) Representative Imaris 3D sur- 2000
face rendering of confocal maxi- CD68 1000
mum projection images showing Iba1
PSD-95
0
volume reconstruction of PSD-

H
O

O
O

O
95 within CD68 structures in

Et

Et
H

H
2

2
microglia (Iba1+ cell) on tissue
sections from prefrontal cortices Microglia + H2O Microglia + EtOH Microglia + anti-TNF Microglia + anti-TNF + EtOH
of WT and TNF KO mice after ex- B C 100
posure to EtOH or H2O. Scale bar, 80
51.6% 66.8% 53.7% 50.9%

5 m. Data are means ± SEM from 60

Mode
12 microglia per condition derived 40

from four mice pooled across three 20

independent experiments. *P < 0.05


by two-way ANOVA with Sidak’s 0 10 10 0 10 10 0 10 10 0 10 10 3 4 3 4 3 4 3 4

multiple comparisons. (B) Immuno­ YFP+ synaptosomes

fluorescence images of PSD-95–


*
synaptosome within microglia
% Area occupied by PSD-95+

20 70 120
labeled prefrontal cortex synapto­

fluorescence in microglia
* 110
*

% Microglia engulfing
YFP+ synaptosomes
somes (red) inside cultured CHME3 60

Normalized YFP
15 100
microglia labeled with CellMask

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


50 90
10 80
dye (blue). Scale bar, 20 m. Data 40
60
30
are means ± SEM from 62 cells per 5 40
15
condition from three indepen- 20
0 0 0
dent experiments. *P < 0.05 by

O
O

H
F

H
H

F
O

TN

O
TN
O

O
O
O

unpaired t test. (C) Represent­

H
H

2
2
H
2

Et
Et

ET
ET
ET

ti-

+
+

ti-
+

+
+
an

ia
ia

an
+
+
ia

ative flow cytometry profiles for

F
F

gl
gl
gl

ia
ia
ia

TN
+

TN

+
ro
ro
ro

gl
gl
gl

ia

ia
ic
ic

ti-
ro
ro

ti-
ic

ro

gl

gl
YFP labeling in CHME3 microglia

M
M
M

an
an

ic
ic
ic

ro

ro
M
M
M

ic

ic

+
+
M

M
incubated for 24 hours with syn-

ia
ia

gl
gl

ro
ro

ic
aptosomes isolated from the pre­ ic

M
M
frontal cortex of Thy1-YFP mice. D 700
* WT - H2O TNF KO - H2O
In some conditions, microglial 600
WT - EtOH TNF KO - EtOH
cultures were pretreated for 500
mRNA levels (%)

24 hours with EtOH (70 mM) or


400
300
E 50
* WT
% CD68+ microglia

300 *
with the TNF blocking antibody 250 * * *
40
TNF KO
adalimumab (5 g/ml). Engulfment 200 * * *
* 30
ability of microglia was calculated 150 20
100
by comparing the percentage (left 50
10
graph) or the normalized MFIs 0 0
(right graph) of microglia express-
12

1
3

H
1
1
oe

2
l

O
Ax

ec
d9

rc
cr

yb

O
am

Ly
Ap

M
C

Et

Et
H

H
2

2
gl
C

ing a high YFP signal. Data are


Ad

Si

means ± SEM from six indepen-


dent experiments per condition.
*P < 0.05 by two-way ANOVA with 200
* WT
Beads per cell (%)

Sidak’s multiple comparisons. F WT - CT 150 TNF KO


(D) qRT-PCR from neocortices of WT - EtOH TNF KO - CT TNF KO - EtOH

WT and TNF KO mice exposed to 100


EtOH or H2O. The clustered histo­ 50
gram shows the Z scores of mRNA
transcripts normalized to the F-actin
Beads
0
DMSO-H 2 O values. Data are
T

H
C

C
O

O
Et

Et

means ± SEM from five mice per


condition pooled across three independent experiments. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons. (E) Flow cytometry analysis of CD68 expression
in microglia (gated as CD45midCD11b+ cell population) from prefrontal cortices of Cx3cr1EYFP-CreER/+ mice exposed to EtOH or H2O. Data are means ± SEM from five mice
per condition pooled across three independent experiments. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons. (F) Primary cortical microglial cultures were
treated with EtOH (70 mM) or H2O, incubated with fluorescent microbeads (green) for 90 min and stained with phalloidin (red). Scale bar, 20 m. Data are means ± SEM
from four independent experiments per condition. *P < 0.05 by two-way ANOVA with Sidak’s multiple comparisons.

microglia from wild-type mice, but not of cortical microglia from DISCUSSION
TNF KO mice (Fig. 7F). These data suggest that EtOH intake results Neuroinflammation is regarded as a major contributing factor in
in aberrant pruning of prefrontal cortex excitatory synapses through alcohol-induced brain damage (42–45). Various studies [including
microglial TNF signaling. chronic 20-week drinking—around 140 mg/dl—in 7-week-old

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 9 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

female mice (46) and chronic 5-week liquid diet feeding—around the increased anxiety-like behavior we observed likely reflects a more
215 mg/dl—in 6- to 8-week-old female mice (47, 48)] show that persistent anxious state driven by consecutive EtOH intake rather
heavy alcohol intake triggers a proinflammatory signature with than withdrawal-induced anxiety.
concomitant gliosis. However, despite inducing neuroimmune acti- A caveat of this study is that the observed phenotypes were only
vation and excitatory synapse loss, our model of repetitive binge-level reported for male mice. Sex differences in the emotional responses
alcohol intake in male mice did not produce a classical proinflam- of mice, such as those related to anxiety-like behavior, are well docu-
matory signature. Rather, our data are more in line with several mented and likely relate to variations in the amounts of sex hormones
transcriptomic studies (49–53) and specifically one RNA sequencing during the estrous cycle (66). The transcriptional program of female
study wherein microglia from the prefrontal cortex of mice chronically microglia and that of male microglia also vary substantially (67),
exposed to alcohol (using the every-other-day, two-bottle choice which, together with the fact that female mice are more prone to
paradigm) do not induce a classic proinflammatory signature (54). develop alcohol-related inflammation and neuronal damage (68, 69),
Likewise, transcript expression of TLR4 was rather decreased in our indicates that further studies using female mice are required to
EtOH exposure model, which is more in line with studies showing better understand the contribution of EtOH exposure to microglia-­
that acute alcohol exposure to human peripheral blood monocytes dependent synaptic pruning, synapse function, and anxiety-related
in culture suppresses TLR4 signaling (55–58). Thus, the dose and behavior.
duration of EtOH exposure appear to differentially affect neuro- Microglia participate in synaptic remodeling by phagocytic en-
immune activation and microglia proinflammatory signaling. gulfment of synaptic terminals (70). We found that EtOH exposure
In our study, Gene Ontology with pathway enrichment indicated increased the engulfment of synaptic structures by microglia likely
that TLR2 and the proinflammatory cytokine TNF were induced through enhancing their phagocytic capacity. In contrast, EtOH de-

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


upon EtOH intake and that these pathways may be coregulated in creases microglia phagocytosis of Escherichia coli (71) and amyloid
microglia. TLR2-dependent recruitment and activation of the (72) and also suppresses microglia phagocytosis stimulated by acti-
kinase Src stimulate several immune-related pathways, including vation of P2X4 receptors (73), thereby suggesting that EtOH might
NF-B activation (59), phospholipase C–to-integrin engagement (60), alter microglia phagocytic capacity in a context-specific manner. In
reactive oxygen species generation (61), interleukin-12 production addition, given that clearance activity by microglia varies substan-
(62), and, ultimately, production of proinflammatory mediators— tially across different brain regions (41), it is conceivable that pre-
including TNF—by microglia (29, 30). EtOH increased Src activa- frontal cortex microglia were more prone to engulf and prune synapses
tion, leading to the nuclear translocation of the p65 subunit of the than their hippocampal counterparts upon exposure to alcohol.
NF-B complex, which culminated in NF-B–dependent TNF pro- While further in-depth studies to identify the precise mechanisms
duction and secretion from microglia. Paralleling the effect of the underlying such regional differences and microglial heterogeneity
blockade of TNF production with PMD, inhibiting Src with AZD0530 are warranted, our finding that microglia can engulf postsynaptic
(a clinically relevant Src inhibitor) also prevented microgliosis. The elements in the prefrontal cortex provides important new mecha-
Src inhibitor also suppressed anxiety-like behavior elicited by EtOH nistic evidence into how the brain immune system may contribute
intake, suggesting that targeting the Src–NF-B–TNF pathway in to impaired synaptic transmission, a major detrimental consequence
microglia may prevent the anxiogenic effect of alcohol. The precise of alcohol abuse.
signaling mechanisms by which alcohol exposure elicits microglial
Src activation and NF-B-dependent transcription warrant further
investigation. Likewise, although our microglial ablation experi- MATERIALS AND METHODS
ments demonstrated that microglia were major producers of TNF Animals
in the prefrontal cortex during EtOH exposure, we cannot rule out All mouse experiments were reviewed by the i3S (Instituto de Investi-
that TNF produced in the periphery (the liver for instance) or even gação e Inovação em Saúde) animal ethical committee and were ap-
by astrocytes in response to EtOH may also have contributed to the proved by Direção-Geral de Alimentação e Veterinária. Animals
observed microglial phenotype. were maintained in standard laboratory conditions with an inverted
The prefrontal cortex contains glutamatergic excitatory neurons 12-hour light/12-hour dark cycle and were allowed free access to food
that synapse locally or project distally to cortical and subcortical and water. Mice were housed under specific pathogen–free conditions.
nuclei. Failure to set up and sustain proper excitatory connectivity Experiments were carried out following the 3Rs (Replacement,
leads to imbalanced neuronal activity across networks (63), which Reduction, and Refinement) ethics policy, and mice were kept on a
could explain at least some of the behavioral impairments found in C57BL/6 background. Because of the potential behavioral variability
various neurological disorders (26). Accordingly, the loss of pre- related to the estrous cycle in females (66), as well as to minimize
frontal cortex excitatory synapses elicited by EtOH could be suffi- intergroup variability and limit the number of animals, only male
cient to disrupt the excitatory/inhibitory balance of prefrontal mice (16 to 20 weeks old) were used in this study.
cortex neurons that project to anxiety-related centers in subcortical B6.129P2(Cg)-Cx3cr1tm2.1(cre/ERT2)Litt/WganJ mice (herein referred
regions, implying that EtOH-induced prefrontal cortex synapse loss to as Cx3cr1EYFP-CreER/+; the Jackson Laboratory stock no: 021160;
might directly affect alcohol-related anxiety. Although this is plau- RRID:IMSR_JAX:021160) were used to study brain microglia in
sible, our data do not formally exclude the possibility that microglia this work and were maintained as before (40). These mice express a
could be directly activated in other brain regions (such as the amyg- Cre-ERT2 fusion protein and an enhanced YFP (EYFP) from the
dala) to elicit the anxiogenic effect of EtOH. Anxiety has also been endogenous Cx3cr1 promoter. EYFP fluorescence is observed in
associated with alcohol withdrawal; however, because our behavior more than 95% of Iba1+ microglia in the brain (16, 40).
analyses were carried out at a time point at which the mice may be For microglial cell ablation experiments, Cx3cr1EYFP-CreER/+ mice were
expected to have a blood EtOH level of 100 to 120 mg/dl (64, 65), intercrossed with R26iDTR/+ [C57BL/6-Gt(ROSA)26Sortm1(HBEGF)Awai/J;

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 10 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

the Jackson Laboratory stock no: 007900; RRID:IMSR_JAX:007900] in addition to being correlated with a dosage inducing neuronal
mice. Genotypes of interest [Cx3cr1 EYFP-CreER/+ (control) and damage (77), it also simulates the amounts achieved in the blood
Cx3cr1EYFP-CreER/+:R26iDTR/+ (experimental)] were determined by during binge drinking.
polymerase chain reaction (PCR) using primers for R26-iDTR in-
sertion including ROSA26-forward: AAA GTC GCT CTG AGT TGT Pharmacological treatments
TAT, ROSA26-reverse: GCG AAG AGT TTG TCC TCA ACC, and Cx3cr1EYFP-CreER/+ and wild-type mice were subjected to a simulta-
wild-type reverse: GGA GCG GGA GAA ATG GAT ATG, as well as neous regimen of AZD0530 (Src inhibitor; 10 mg/kg) or dimethyl
primers for CreER-EYFP insertion including forward: AAG ACT CAC sulfoxide (DMSO; control) injections (pretreatment intraperitoneal
GTG GAC CTG CT, wild-type reverse: AGG ATG TTG ACT TCC injection followed by five intraperitoneal injections spaced by 24 hours
GAG TG, and mutant reverse: CGG TTA TTC AAC TTG CAC CA. during the 10-day EtOH exposure regimen). The same regimen was
TNF KO (referred to herein as TNF KO) mice were generously used for the immunomodulatory agent PMD (50 mg/kg). Tamoxifen
supplied by R. Applelberg (University of Porto). TNF KO mice were was given to adult Cx3cr1EYFP-CreER/+ and Cx3cr1EYFP-CreER/+:R26iDTR/+
genotyped by PCR using ATC CGC GAC GTG GAA CTG GCA mice as a solution in corn oil by oral gavage. Mice received two doses
GAA (forward) and CTG CCC GGA CTC CGC AAA GTC TAA of 10 mg of tamoxifen separated by 48 hours between doses. For micro­
(reverse) primer pair, as before (40). TNF KO mice display a single glia ablation, 8 weeks after the last tamoxifen pulse, Cx3cr1EYFP-CreER/+
band of 2 kb in the PCR gel. TNF-deficient mice were generated and Cx3cr1EYFP-CreER/+:R26iDTR/+ mice were given diphtheria toxin
from hemizygote progenitors, and wild-type littermates were used (1 g, intraperitoneally) for three consecutive days and sacri-
as controls. fice occurred 24 hours after the last diphtheria toxin injection.
Tg(Thy1-cre/ERT2,-EYFP)HGfng/PyngJ mice (also known as

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


SLICK-H [single-neuron labeling inducible Cre-mediated knockout Brain tissue preparation and immunohistochemistry
(SLICK)-line H] and herein termed Thy1-YFP; the Jackson Labora- After animal perfusion with ice-cold phosphate-buffered saline
tory stock no: 012708; RRID:IMSR_JAX:012708) were maintained (PBS; 15 ml) and fixation by perfusion with 4% paraformaldehyde
as before (74). These mice have constitutive and exclusive YFP label- (PFA), brains were postfixed by immersion in 4% PFA in PBS (pH 7.2)
ing of neurons driven by the endogenous Thy1 promoter (75). In overnight. After that, brains were washed with PBS and then cryo-
this work, Thy1-YFP mice were used for studying the engulfment protected using sucrose gradient in a row (15 and 30%). After 24 hours,
of synapses by microglia in vitro. brains were mounted in OCT (optimal cutting temperature)–­
embedding medium, frozen, and cryosectioned in the CM3050S
Drugs Cryostat (Leica Biosystems). Coronal sections from brains (30-m
Ethanol 99.9% (containing less than 0.0002% benzene) was obtained thickness) were collected nonsequentially on Superfrost ultra plus
from Merck. Src inhibitor 1 (SKI), sulfasalazine, tamoxifen, diphtheria slides. Tissue sections from controls and experimental mice en-
toxin, and rapamycin from Streptomyces hygroscopicus were from compassing identical stereological regions were collected on the
Sigma-Aldrich. AZD0530, PMD, and adalimumab were from same glass slide and stored at −20°C. Frozen sections were defrosted
Selleckchem. by at least 1 hour and hydrated with PBS for 15 min. Sections were
permeabilized with 0.25% Triton X-100 for 15 min, washed with
Alcohol intake protocol PBS for 10 min, and blocked [5% bovine serum albumin (BSA), 5%
Mice (genotypes are specified in Results and the figure legends) were fetal bovine serum (FBS), and 0.1% Triton X-100] for 1 hour. Pri-
habituated for 4 weeks in experimental rooms at the i3S animal facility. mary antibodies were incubated in a blocking solution in a hu-
Afterward, mice were randomly assigned to experimental groups. midified chamber overnight at 4°C. Secondary antibodies were
To emulate a pattern of repetitive binge-level alcohol intake, incubated for 2 hours in blocking solution. After the secondary
mice were given EtOH (1.5 g/kg; diluted to 25% in sterile tissue antibody, sections were washed three times for 10  min with
culture-grade water) by oral gavage daily for 10 consecutive days. PBS. Slides were coverslipped using glycergel or Immu-Mount and
Vehicle control mice were gavaged with water in equal amounts to visualized under a Leica TCS SP5 II confocal microscope.
those of EtOH-gavaged mice. For analyses, mice were euthanized
120 min after the last gavage. Animals were provided with food and Confocal imaging and morphometric analysis
water ad libitum throughout the experiments. Images from tissue sections of the prefrontal cortex and CA1 region
Measurements of blood alcohol concentration in adult C57BL/6 of the dorsal hippocampus were acquired using a Leica HC PL APO
mice show that 120 min after oral intake of 1.5 g/kg, around 100 to Lbl. Blue 20×/0.70 IMM/CORR (multi-immersion objective/
120 mg/dl of EtOH is detected in their blood (64, 65). On the basis correction collar) or a Leica HC PL APO CS 40×/1.10 CORR water
of a study by Pruett et al. (76), in which oral intake of alcohol is objective in 8-bit sequential mode using standard TCS (true confocal
compared in mice and humans to achieve similar peak blood alco- scanning) mode at 400 Hz, and the pinhole was kept at 1 airy in the
hol concentrations, the 1.5 g/kg daily dose we used would roughly Leica TCS SP5 II confocal microscope. Images were resolved at
correspond to 0.83 g/kg per day in humans or five drinks a day 1024 × 1024 pixels format illuminated with 2 to 5% DPSS561 561-nm
(at 12 g of alcohol per drink) for a person weighing 75 kg. Accord- wave laser using an hybrid HyD (hybrid) detector in the BrightR
ing to the National Institute of Alcohol Abuse and Alcoholism, mode, and entire Z-series were acquired from tissue sections.
binge drinking is defined as five or more drinks a day for men and Equivalent stereological regions were acquired for all tissue sections
four or more for women (www.niaaa.nih.gov/alcohol-health/ within a given slide.
overview-alcohol-consumption/moderate-binge-drinking). To quantify microglia, the number of YFP+ cells was manually
For in vitro studies, microglial cultures were exposed to 70 mM scored in stereological identical regions of the prefrontal cortex of
(320 mg/dl) EtOH. This concentration of EtOH was used because, stained sections (six images per section; five sections per animal for

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 11 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

each experimental group). To quantify GFAP, the number of GFAP+ lected, pelleted, washed extensively, and then counted in a Neubauer
cells was manually scored in stereological identical regions of the chamber using trypan blue exclusion to estimate the number of live
prefrontal cortex of stained sections (six images per section; eight cells. Single-cell suspensions (1 × 106 cells) were incubated with dif-
sections per animal for each experimental group). To quantify NeuN, ferent mixes of fluorescence-activated cell sorting (FACS) antibodies
the number of NeuN+ neurons was manually scored in stereological for 30 min at 4°C in the dark. Compensation settings were determined
identical regions of the prefrontal cortex-stained sections (six images using spleens from wild-type mice. Cell suspensions were evaluated
per section; eight sections per animal for each experimental group). on a FACS Canto II analyzer (BD Immunocytometry Systems).
To quantify TNF, briefly, stereological identical regions of TNF-­ Microglial cell numbers found on neocortices were estimated using
immunostained sections of the prefrontal cortex or the dorsal hippo­ Precision Count Beads (BioLegend 424902).
campal CA1 area (four images per section; six sections per animal
for each experimental group) were imaged, converted into 8-bit Preparation of lysates and Western blotting
grayscale, 3D volume-rendered, and thresholded. Using FIJI software, Cultures or mice tissues were lysed using radioimmunoprecipita-
the percentage of TNF-immunostained area was calculated for each tion assay–dithiothreitol (DTT) buffer [150 mM NaCl, 50 mM tris,
field and each section. 5 mM EGTA, 1% Triton X-100, 0.5% deoxycholate (DOC), and 0.1%
To quantify synapses, images from stereological identical pre- SDS] supplemented with complete-mini protease inhibitor cocktail
frontal cortex or dorsal hippocampal CA1 region from each experi- tablets, 1 mM DTT, and phosphatase inhibitor cocktail. Samples were
mental group (four images per section; six sections per animal for sonicated (six pulses of 1 s at 60 Hz) and centrifuged at 16,000g, 4°C
each experimental group) were acquired using a Leica HC PL APO for 10 min. The supernatants were collected, and the protein con-
CS 40×/1.10 CORR water objective at 1024 × 1024 pixels resolution centration was determined by the BCA (bicinchoninic acid assay)

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


with 8-bit bidirectional nonsequential scanner mode at 400 Hz and method. All samples were denatured with sample buffer [0.5 M
pinhole at 1 airy in the Leica TCS SP5 II confocal microscope. Z-stacks tris-HCl (pH 6.8), 30% glycerol, 10% SDS, 0.6 M DTT, and 0.02%
were converted to maximum projection images using the LAS AF bromophenol blue] at 95°C for 5 min and stored at −20°C until
(Leica Application Suite for Advanced Fluorescence) routine. Z-projections use. Samples were separated in SDS–polyacrylamide gel electro-
were background-subtracted using the rolling ball background sub- phoresis and transferred to polyvinylidene difluoride membranes,
traction built-in algorithm in FIJI and then images were upsampled which were incubated 12 hours (overnight) at 4°C with primary
using a bicubic interpolation routine. The number of double-positive antibodies. Membranes were washed in TBS-T buffer (pH 7.6),
PSD-95/vGlut1 puncta per square micrometer was manually scored incubated with peroxidase-conjugated secondary antibodies, and de-
for each image. veloped using an ECL chemiluminescence kit or an ECF fluorescence
To quantify PSD-95 engulfment by microglia, images from kit. Images were acquired in a Typhoon FLA 9000 system or ChemiDoc
stereological identical prefrontal cortex or dorsal hippocampal CA1 XRS System (Bio-Rad) and quantified by FIJI software.
region from each experimental group (six images per section; four
sections per animal for each experimental group) were acquired using Gene expression and bioinformatic analyses
a Leica HC PL APO CS 40×/1.10 CORR water objective at 1024 × RNA was extracted from neocortices using the Direct-zol RNA
1024 pixels resolution with 8-bit bidirectional scanner mode at 200 Hz MiniPrep Kit according to the manufacturer’s instructions. Com-
in the Leica TCS SP5 II confocal microscope. Using FIJI software, plementary DNA synthesis was performed using 500 ng of total
confocal Z stacks were background-subtracted and smoothened using RNA [deoxyribonuclease I (DNase I)–treated] with SuperScript III
a Sigma-Aldrich filter plus. CD68, PSD-95, and Iba1 volumes were First-Strand Synthesis SuperMix. Quantitative reverse transcription
reconstructed using 3D surface rendering of confocal Z stacks in (qRT)–PCR was carried out using iQ SYBR Green Supermix on an
Imaris. For quantification of PSD-95 engulfment by microglia, PSD-95 iQ5 multicolor real-time PCR detection system (Bio-Rad). The ex-
puncta embedded within volume-rendered CD68+ structures in pression of PCR transcripts was calculated using the 2−Ct with Yhwaz
Iba1+ cells were considered to be engulfed by microglia. Unbiased serving as the internal control gene. Statistical analyses on raw 2−Ct
measurements of PSD-95 puncta associated with CD68 were auto- values using unpaired t tests were performed for detecting differentially
matically performed using the Imaris colocalization package for expressed transcripts between sampled groups. For representation,
each microglia and used for statistical analyses. transcript expression values were converted into Z scores. ClueGo
To assess microglial morphology, the ramification of prefrontal (79) was used for unsupervised enrichment analyses and statistical
cortex Iba1+ microglia was evaluated as described before (78). Branches overrepresentation of immune signaling pathways. The NetworkAnalyst
were traced using the NeuronJ plugin, and data for each cell were 3.0 (80) was used for constructing topographic maps of over-
converted into SWC format using Bonfire. Individual segments were represented pathways from Gene Ontology (PANTHER/SLIM GO)
connected to NeuronStudio software and audited for any tracing and for generating protein-protein interaction networks (using
errors using Bonfire. Sholl analysis was performed by drawing con- the STRING database for the representation of interacting protein
centric circles around the cell body at defined radius increments, clusters).
separated by 6 m. The number of intersections of branches at each
defined circle was used to estimate the number of ramifications. Behavioral tests
Branching data were extracted using a custom MATLAB routine. All testing procedures were conducted in the dark phase of the
light/dark cycle. Before each session, mice were removed from their
Flow cytometry home cage in the colony room and brought into the adjacent testing
Mice were anesthetized and then perfused with ice-cold PBS. For rooms (illuminated with 100 lux and attenuated noise). Behavioral
single-cell suspensions, neocortices were quickly dissected on ice, placed tests were performed, as described previously (40), in the following
on ice-cold RPMI, and mechanically homogenized. Cells were col- order: (i) EPM, (ii) OF, and (iii) novel object recognition.

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 12 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

The EPM was made of opaque gray polyvinyl chloride consisting Immuno­labeling with CD11b showed a purity of 95 to 99% for
of four arms arranged in a plus-shaped format; two arms have sur- these cultures.
rounding walls (closed arms, 37 cm by 6 cm by 18 cm high), where-
as the two opposing arms have no walls (open arms, 37 cm by 6 cm). Microglial cell lines
The apparatus was elevated by 50 cm above the ground. Mice were The microglial cell line N9 was obtained by immortalization of pri-
placed on the central platform facing an open arm and were allowed mary cultures from the ventral mesencephalon and cerebral cortex
to explore the maze for 5 min. Frequency and time spent in the from embryonic day 12 ED12 (embryonic day 12) to ED13 CD1
open arms were obtained automatically (Smart 3.0 Panlab Harvard mouse embryos with the 3RV retrovirus carrying an activated
Apparatus) and used to assess anxiety-like behavior. v-myc oncogene (83). The microglial cell line CHME3 (human
For the OF test, mice were placed in the center of an OF apparatus microglial clone 3) was obtained from primary cultures of human
(40  cm by 40  cm by 40 cm) and then allowed to move freely for embryonic microglial cells by transfection with a plasmid encod-
10 min. The total distance traveled and locomotion in the peripheral ing for the large T antigen of SV40 (84). Cells were cultivated and
zone and center zone of the apparatus were obtained automatically maintained as before (27, 40).
using video tracking.
The novel object recognition test was performed as previously Synaptosomal preparations and microglia engulfment assay
described (81). Briefly, the same OF test apparatus was used, and To isolate prefrontal cortex synaptic terminals, synaptosomes were
the objects used were made of plastic, glass, or metal in three different freshly prepared using Syn-PER Synaptic Protein Extraction Reagent
shapes: cubes, pyramids, and cylinders. The test consists of three (catalog no. 87793, Thermo Fisher Scientific) precisely as recom-
phases. During the habituation phase, mice are allowed to explore mended by the manufacturer. Briefly, wild-type mice or Thy1-YFP

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


the apparatus for 10 min (period considered for the OF test). The were euthanized in a CO2 chamber. Their prefrontal cortex was col-
following day, the acquisition/sample phase starts by placing each lected, and homogenization was performed using a Dounce tissue
mouse in the apparatus with two identical objects (familiar) for grinder (~10 strokes). The homogenate was centrifuged at 1200g for
10 min. Then, the mouse goes back to its home cage. After 4 hours 10 min, and the pellet was discarded. The supernatant was centri-
(intertrial interval), the retention/choice session is performed. In fuged at 15,000g for 20 min. The resulting supernatant was discarded,
this phase, the apparatus contains a novel object and a copy of the and the synaptosomal pellet was resuspended in Syn-Per Synaptic
last familiar object; animals are allowed to explore these objects for Protein Extraction Reagent.
3 min. Exploration was defined as follows: Mouse touched the object CHME3 microglial cultures were seeded in 12-well culture plates
with its nose, or the mouse’s nose was directed toward the object at at a density of 2.5 × 104 cells per well in DMEM GlutaMAX-I sup-
a distance shorter than 2 cm (82). Circling or sitting on the object plemented with 10% FBS and 0.1% PenStrep and cultivated for
was not considered exploratory behavior. Behavioral data were col- 48 hours. Cultures were treated with an FcR (Fc-gamma receptors)–­
lected using the software Observer XT 7.0, Noldus. Increased time blocking solution (1:100) for 20 min and then refed with fresh medium
spent exploring the novel object serves as a measure of recognition containing vehicle (water), EtOH (70 mM), adalimumab (5 g/ml), or
memory for the familiar object. The discrimination index (DI) was adalimumab (5 g/ml) plus EtOH (70 mM). Cells were kept in those
calculated as an index of memory function, DI = (time exploring the conditions for 24 hours. Afterward, cells were refed with fresh me-
novel object − time exploring the familiar object)/(total time spent dium containing vehicle (water) plus synaptosomes (1:100), EtOH
exploring both objects). Positive values indicate increased time in- (70 mM) plus synaptosomes (1:100), adalimumab (5 g/ml) plus syn-
vestigating the novel object, which serves as an indication of object aptosomes (1:100), or adalimumab (5 g/ml) plus EtOH (70 mM)
discrimination. plus synaptosomes (1:100). Cells were cultured under these condi-
tions for an additional 24 hours. For analyses, microglial cultures
Primary cortical microglial cultures were washed extensively with PBS and either fixed with 4% PFA,
Primary cortical microglial cell cultures were performed as previously immunolabeled for PSD-95, and analyzed in a fluorescence micro-
described (27–29, 40). Briefly, 2-day-old mouse pups were eutha- scope or detached using Accutase solution (catalog no. A6964, Sigma-­
nized, and their cerebral cortices were dissected in Hanks’ balanced Aldrich), blocked in PBS–5% BSA, fixed with 2% PFA, resuspended
salt solution (pH 7.2) and digested with 0.07% trypsin plus 50 l in PBS–1% BSA, and analyzed in a flow cytometer.
(w/v) DNase for 15 min. The cells were gently dissociated using a
glass pipette in Dulbecco’s modified Eagle’s medium (DMEM) F12 Phagocytic assay in primary cortical microglia
GlutaMAX-I supplemented with 10% FBS and 0.1% gentamicin and Cell cultures were incubated with fluorescent latex microbeads
plated onto poly-d-lysine–coated T-flasks (75 cm2) at 1.5 × 106 cells/cm2. [0.001% (v/v) final concentration; Sigma-Aldrich] for 90 min.
Cultures were kept at 37°C and 95% air/5% CO2 in a humidified Cultures were washed 3× with PBS and fixed in 4% PFA (w/v) for
incubator, and culture media were changed every 3 days for up to 21 days. 12 min. Afterward, F-actin was stained using phalloidin, and cells
To obtain purified microglial cell cultures, culture flasks were sub- were visualized in a fluorescence microscope. The number of en-
jected to orbital shaking at 200 rpm for 2 hours. Culture supernatant gulfed microbeads per microglia was manually scored and plotted
was then collected into tubes, centrifuged at 453g for 5 min at room for statistical evaluation.
temperature. The supernatant was discarded, and the pellet contain-
ing microglia was resuspended in culture medium. Cells were then again Antibodies
seeded onto poly-d-lysine–coated 6- or 12-well culture plates at 2.5 × The following antibodies were used in this study: GFP (Abcam
105 cells/cm2 with DMEM F12 GlutaMAX-I supplemented with 10% FBS, catalog no. ab6673, RRID:AB_305643; 1:200), Iba1 (Wako catalog
0.1% gentamicin, and granulocyte-macrophage colony-stimulating no. 019-19741, RRID:AB_839504; 1:500), glyceraldehyde-3-phosphate
factor (1 ng/ml). Purified microglia were cultured for 5 to 8 days. dehydrogenase (Hytest catalog no. 5G4-9B3, RRID:AB_1616725;

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 13 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

1:20,000), CD11b clone M1/70.15 (BD Biosciences catalog no. 560456, Microsystems). A 440- to 520-nm dichroic mirror (CG1, Leica
RRID:AB_1645267; 1:100), phospho-Src family (Tyr416; Cell Signaling Microsystems) and a PlanApo 63× 1.3NA glycerol immersion ob-
Technology catalog no. 6943, RRID:AB_10013641; 1:100 to 1:200), jective were used for CFP and FRET images. Images were acquired
Src clone EPR5496 (Abcam catalog no. ab109381, RRID:AB_10865528; with 2 × 2 binning using a digital complementary metal-oxide semi-
1:1000), PSD95 clone 6G6-1C9 (Thermo Fisher Scientific catalog conductor camera (ORCA-Flash4.0 V2, Hamamatsu Photonics). At
no. MA1-045, RRID:AB_325399; 1:600), vGlut1 (Synaptic Systems each time point, CFP, FRET, and fared images were sequentially
catalog no. 135 303, RRID:AB_887875; 1:1000), CD68 clone FA-11 acquired using different filter combinations (27–29, 40). For quan-
(Bio-Rad catalog no. MCA1957T, RRID:AB_2074849; 1:400), Fc tifications, images were exported as 16-bit tiff files and processed in
Receptor Blocking Solution (BioLegend catalog no. 156603, FIJI software. The background was dynamically subtracted from all
RRID:AB_2783137; 1:50), GFAP (Abcam catalog no. ab7260, frames from both channels. Segmentation (on a pixel-by-pixel basis)
RRID:AB_305808; 1:100 to 1:500), NeuN (Millipore catalog no. and generation of 32-bit float-point ratiometric images were achieved
MAB377, RRID:AB_2298772; 1:400), allophycocyanin (APC) anti-­ using the precision FRET (PFRET) data processing software package
mouse/human CD11b antibody (BioLegend catalog no. 101212, for ImageJ (https://lvg.virginia.edu/digital-downloads/pfret-data-­
RRID:AB_312795; 0.25 g to 106 cells), Alexa Fluor 647 anti-mouse/ processing-software). The mean gray intensity values from ratio
human CD11b antibody (BioLegend catalog no. 101218, RRID: images were used for statistical calculations.
AB_389327; 0.25 g to 106 cells), phycoerythrin anti-mouse CD45
antibody (BioLegend catalog no. 103106, RRID:AB_312971; 0.25 g Cytokine release
to 106 cells), APC anti-mouse CD19 antibody (BioLegend catalog no. Culture medium was collected to tubes and centrifuged at 16,000g
115511, RRID:AB_313646; 0.25 g to 106 cells), APC/Cyanine7 at 4°C for 5 min. The supernatant was transferred to a new tube and

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


anti-mouse CD3 antibody (BioLegend catalog no. 100221, RRID:AB_ kept at −80°C. The concentration of TNF in cell culture supernatants
2057374; 0.25 g to 106 cells), and Brilliant Violet 421 anti-mouse was quantified by enzyme-linked immunosorbent assay following
CD68 antibody (BioLegend catalog no. 137017, RRID:AB_2562949; the instructions provided by the manufacturer (PeproTech, UK).
0.25 g to 106 cells). Absorbance at 405 nm, with wavelength correction at 650 nm, was
measured with a multimode microplate reader (Synergy HT, BioTek,
Primers USA). Values corresponding to nanograms per microliter were
The primers used in this study were purchased from Thermo Fisher obtained by extrapolating a standard concentration curve using re-
Scientific; their sequences are provided in table S1. combinant TNF.

Plasmids Statistics
Plasmids used in this study were purchased from Addgene: NF-B A 95% confidence interval was used for statistical evaluation, and
GFP–tagged p65 (RRID:Addgene_23255), IB-miRFP703 P < 0.05 was considered a statistically significant difference in all
(RRID:Addgene_80005), pLNCX chick src Y527F (RRID:Addgene_13660), sampled groups. Experimental units in individual replicates were previ-
pUSE-RapR-Src-myc (RRID:Addgene_25933), psPAX2 (RRID: ously evaluated for Gaussian distribution using the D’Agostino-­
Addgene_12260), pMD2.G (RRID:Addgene_12259), pUMVC Pearson omnibus normality test with GraphPad Prism. When
(RRID:Addgene_8449), pMSCV (RRID:Addgene_24828), and comparing only two experimental groups, a Mann-Whitney test for
pCherry-FRB (RRID:Addgene_25920). data with nonnormal distribution or an unpaired Student’s t test
with equal variance assumption for data with normal distribution
Retrovirus production was used. When comparing three to four independent conditions, a
Low-passage HEK293T cells were seeded in 100-mm culture dishes. one-way analysis of variance (ANOVA) followed by the Sidak’s
When cultures reached ~80% confluence, cells were cotransfected multiple comparisons test was used. When comparing treatment
overnight with virus-producing plasmids using the transfection re- effects within genotypes or mutants, a two-way ANOVA followed
agent jetPRIME. Transfection ratios were as follows: 6 g of short by the Sidak’s multiple comparisons test was used. Two-way ANOVA
hairpin RNA (shRNA) plasmids/3 g of psPAX2/3 g of VSVG was also used to compare values of microglia intersections retrieved
(vesicular stomatitis virus G) (2:1:1) for lentiviruses production or from Sholl analysis. To minimize bias, experimental groups were
8 g of SrcY527F construct/4 g of pUMVC/2 g of VSVG (4:2:1) for assigned through randomization. Specifically, sampled groups eval-
viral production. The next day, normal growth media replaced uated in Figs. 1 and 2 and all supplementary figures were assigned
transfection media, and cells were cultivated for an additional 48 hours. using “simple random sampling.” Sampled assessed groups in Figs. 3
Next, media with viral particles were collected and centrifuged at to 7 were assigned using “permuted block randomization.” All
906g for 15 min at 4°C, and the supernatant was collected into new quantifications were performed blinded.
tubes and kept at −80°C.
SUPPLEMENTARY MATERIALS
Live cell imaging and FRET stke.sciencemag.org/cgi/content/full/13/650/eaba5754/DC1
Microglial cells were plated on plastic-bottom culture dishes Fig. S1. Alcohol exposure does not activate other resident immune cells in the neocortex.
(-Dish 35 mm, iBidi). Imaging was performed using a Leica Fig. S2. Alcohol exposure modulates microglial TNF production through an Src–NF-B pathway.
Fig. S3. Alcohol exposure does not alter general locomotor activity or recognition memory.
DMI6000B inverted microscope. The excitation light source was a
Fig. S4. Alcohol induces synapse loss without affecting neuronal numbers.
mercury metal halide bulb integrated with an EL6000 light attenuator. Fig. S5. Alcohol exposure does not alter synapse number, postsynaptic engulfment, or TNF
High-speed, low-vibration external filter wheels [equipped with cyan production in the CA1 region of the dorsal hippocampus.
fluorescent protein (CFP)/YFP/farRed excitation and emission fil- Table S1. Primers.
ters] were mounted on the microscope (Fast Filter Wheels, Leica View/request a protocol for this paper from Bio-protocol.

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 14 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

REFERENCES AND NOTES transporter SVCT2 in microglia triggers an inflammatory phenotype. Sci. Signal. 10,
1. D. M. Lovinger, M. Roberto, Synaptic effects induced by alcohol. Curr. Top. Behav. eaal2005 (2017).
Neurosci. 13, 31–86 (2013). 28. R. Socodato, C. C. Portugal, T. Canedo, I. Domith, N. A. Oliveira, R. Paes-de-Carvalho,
2. K. P. Abrahao, A. G. Salinas, D. M. Lovinger, Alcohol and the brain: Neuronal molecular J. B. Relvas, M. Cossenza, c-Src deactivation by the polyphenol 3-O-caffeoylquinic acid
targets, synapses, and circuits. Neuron 96, 1223–1238 (2017). abrogates reactive oxygen species-mediated glutamate release from microglia
3. M. J. Lacagnina, P. D. Rivera, S. D. Bilbo, Glial and neuroimmune mechanisms as critical and neuronal excitotoxicity. Free Radic. Biol. Med. 79, 45–55 (2015).
modulators of drug use and abuse. Neuropsychopharmacology 42, 156–177 (2017). 29. R. Socodato, C. C. Portugal, I. Domith, N. A. Oliveira, V. S. Coreixas, E. C. Loiola, T. Martins,
4. M. Prinz, D. Erny, N. Hagemeyer, Ontogeny and homeostasis of CNS myeloid cells. A. R. Santiago, R. Paes-de-Carvalho, A. F. Ambrósio, J. B. Relvas, c-Src function is necessary
Nat. Immunol. 18, 385–392 (2017). and sufficient for triggering microglial cell activation. Glia 63, 497–511 (2015).
5. A. Crotti, R. M. Ransohoff, Microglial physiology and pathophysiology: Insights 30. S. Zhao, J. Yin, L. Zhou, F. Yan, Q. He, L. Huang, S. Peng, J. Jia, J. Cheng, H. Chen, W. Tao,
from genome-wide transcriptional profiling. Immunity 44, 505–515 (2016). X. Ji, Y. Xu, Z. Yuan, Hippo/MST1 signaling mediates microglial activation following acute
6. M. W. Salter, B. Stevens, Microglia emerge as central players in brain disease. Nat. Med. 23, cerebral ischemia–reperfusion injury. Brain Behav. Immun. 55, 236–248 (2016).
1018–1027 (2017). 31. A. C. Kaufman, S. V. Salazar, L. T. Haas, J. Yang, M. A. Kostylev, A. T. Jeng, S. A. Robinson,
7. A. Vilalta, G. C. Brown, Neurophagy, the phagocytosis of live neurons and synapses by E. C. Gunther, C. H. van Dyck, H. B. Nygaard, S. M. Strittmatter, Fyn inhibition rescues
glia, contributes to brain development and disease. FEBS J. 285, 3566–3575 (2018). established memory and synapse loss in Alzheimer mice. Ann. Neurol. 77, 953–971 (2015).
8. J. He, F. T. Crews, Increased MCP-1 and microglia in various regions of the human 32. T. Liu, L. Zhang, D. Joo, S.-C. Sun, NF-B signaling in inflammation. Signal Transduct.
alcoholic brain. Exp. Neurol. 210, 349–358 (2008). Target. Ther. 2, 17023 (2017).
9. J.-Y. Yang, X. Xue, H. Tian, X.-X. Wang, Y.-X. Dong, F. Wang, Y.-N. Zhao, X.-C. Yao, W. Cui, 33. G. Bonizzi, M. Karin, The two NF-B activation pathways and their role in innate
C.-F. Wu, Role of microglia in ethanol-induced neurodegenerative disease: Pathological and adaptive immunity. Trends Immunol. 25, 280–288 (2004).
and behavioral dysfunction at different developmental stages. Pharmacol. Ther. 144, 34. D. M. Shcherbakova, M. Baloban, A. V. Emelyanov, M. Brenowitz, P. Guo, V. V. Verkhusha,
321–337 (2014). Bright monomeric near-infrared fluorescent proteins as tags and biosensors
10. C. Cui, D. Shurtleff, R. A. Harris, Neuroimmune mechanisms of alcohol and drug addiction. for multiscale imaging. Nat. Commun. 7, 12405 (2016).
Int. Rev. Neurobiol. 118, 1–12 (2014). 35. A. V. Karginov, F. Ding, P. Kota, N. V. Dokholyan, K. M. Hahn, Engineered allosteric

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


11. J. Mayfield, L. Ferguson, R. A. Harris, Neuroimmune signaling: A key component of alcohol activation of kinases in living cells. Nat. Biotechnol. 28, 743–747 (2010).
abuse. Curr. Opin. Neurobiol. 23, 513–520 (2013). 36. H.-T. Gan, Y.-Q. Chen, Q. Ouyang, Sulfasalazine inhibits activation of nuclear factor-B
12. J. F. Henriques, C. C. Portugal, T. Canedo, J. B. Relvas, T. Summavielle, R. Socodato, in patients with ulcerative colitis. J. Gastroenterol. Hepatol. 20, 1016–1024 (2005).
Microglia and alcohol meet at the crossroads: Microglia as critical modulators of alcohol 37. C. K. Weber, S. Liptay, T. Wirth, G. Adler, R. M. Schmid, Suppression of NF-B activity by
neurotoxicity. Toxicol. Lett. 283, 21–31 (2018). sulfasalazine is mediated by direct inhibition of IB kinases  and . Gastroenterology
13. A. Warden, E. Erickson, G. Robinson, R. A. Harris, R. D. Mayfield, The neuroimmune 119, 1209–1218 (2000).
transcriptome and alcohol dependence: Potential for targeted therapies. 38. R. C. Froemke, Plasticity of cortical excitatory-inhibitory balance. Annu. Rev. Neurosci. 38,
Pharmacogenomics 17, 2081–2096 (2016). 195–219 (2015).
14. E. K. Erickson, E. K. Grantham, A. S. Warden, R. A. Harris, Neuroimmune signaling 39. R. Tatti, M. S. Haley, O. K. Swanson, T. Tselha, A. Maffei, Neurophysiology and regulation
in alcohol use disorder. Pharmacol. Biochem. Behav. 177, 34–60 (2019). of the balance between excitation and inhibition in neocortical circuits. Biol. Psychiatry
15. L. Qin, F. T. Crews, NADPH oxidase and reactive oxygen species contribute to alcohol- 81, 821–831 (2017).
induced microglial activation and neurodegeneration. J. Neuroinflammation 9, 5 (2012). 40. R. Socodato, C. C. Portugal, T. Canedo, A. Rodrigues, T. O. Almeida, J. F. Henriques,
16. C. N. Parkhurst, G. Yang, I. Ninan, J. N. Savas, J. R. Yates III, J. J. Lafaille, B. L. Hempstead, S. H. Vaz, J. Magalhães, C. M. Silva, F. I. Baptista, R. L. Alves, V. Coelho-Santos, A. P. Silva,
D. R. Littman, W.-B. Gan, Microglia promote learning-dependent synapse formation R. Paes-de-Carvalho, A. Magalhães, C. Brakebusch, A. M. Sebastião, T. Summavielle,
through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013). A. F. Ambrósio, J. B. Relvas, Microglia dysfunction caused by the loss of rhoa disrupts
17. S. Alfonso-Loeches, J. R. Ureña-Peralta, M. J. Morillo-Bargues, J. Oliver-De La Cruz, neuronal physiology and leads to neurodegeneration. Cell Rep. 31, 107796 (2020).
C. Guerri, Role of mitochondria ROS generation in ethanol-induced NLRP3 inflammasome 41. P. Ayata, A. Badimon, H. J. Strasburger, M. K. Duff, S. E. Montgomery, Y.-H. E. Loh, A. Ebert,
activation and cell death in astroglial cells. Front. Cell. Neurosci. 8, 216 (2014). A. A. Pimenova, B. R. Ramirez, A. T. Chan, J. M. Sullivan, I. Purushothaman, J. R. Scarpa,
18. M. Pascual-Lucas, S. Fernandez-Lizarbe, J. Montesinos, C. Guerri, LPS or ethanol triggers A. M. Goate, M. Busslinger, L. Shen, B. Losic, A. Schaefer, Epigenetic regulation of brain
clathrin- and rafts/caveolae-dependent endocytosis of TLR4 in cortical astrocytes. region-specific microglia clearance activity. Nat. Neurosci. 21, 1049–1060 (2018).
J. Neurochem. 129, 448–462 (2014). 42. C. J. Kane, P. D. Drew, Inflammatory responses to alcohol in the CNS: Nuclear receptors
19. R. Miñambres, R. M. Guasch, A. Perez-Aragó, C. Guerri, The RhoA/ROCK-I/MLC pathway is as potential therapeutics for alcohol-induced neuropathologies. J. Leukoc. Biol. 100,
involved in the ethanol-induced apoptosis by anoikis in astrocytes. J. Cell Sci. 119, 951–959 (2016).
271–282 (2006). 43. J. Montesinos, S. Alfonso-Loeches, C. Guerri, Impact of the innate immune response
20. S. L. Vallés, A. M. Blanco, M. Pascual, C. Guerri, Chronic ethanol treatment enhances inflammatory in the actions of ethanol on the central nervous system. Alcohol. Clin. Exp. Res. 40,
mediators and cell death in the brain and in astrocytes. Brain Pathol. 14, 365–371 (2004). 2260–2270 (2016).
44. F. T. Crews, R. Bechara, L. A. Brown, D. M. Guidot, P. Mandrekar, S. Oak, L. Qin, G. Szabo,
21. A. Pla, M. Pascual, C. Guerri, Autophagy constitutes a protective mechanism against
M. Wheeler, J. Zou, Cytokines and alcohol. Alcohol. Clin. Exp. Res. 30, 720–730 (2006).
ethanol toxicity in mouse astrocytes and neurons. PLOS ONE 11, e0153097 (2016).
45. N. M. Zahr, K. L. Kaufman, C. G. Harper, Clinical and pathological features of alcohol-
22. A. M. Blanco, S. L. Vallés, M. Pascual, C. Guerri, Involvement of TLR4/type I IL-1 receptor
related brain damage. Nat. Rev. Neurol. 7, 284–294 (2011).
signaling in the induction of inflammatory mediators and cell death induced by ethanol
46. S. Alfonso-Loeches, M. Pascual-Lucas, A. Blanco, I. Sanchez-Vera, C. Guerri, Pivotal role
in cultured astrocytes. J. Immunol. 175, 6893–6899 (2005).
of TLR4 receptors in alcohol-induced neuroinflammation and brain damage. J. Neurosci.
23. T. J. Walter, F. T. Crews, Microglial depletion alters the brain neuroimmune response
30, 8285–8295 (2010).
to acute binge ethanol withdrawal. J. Neuroinflammation 14, 86 (2017).
47. D. Lippai, S. Bala, T. Csak, E. A. Kurt-Jones, G. Szabo, Chronic alcohol-induced
24. G. W. Muller, R. Chen, S.-Y. Huang, L. G. Corral, L. M. Wong, R. T. Patterson, Y. Chen,
microRNA-155 contributes to neuroinflammation in a TLR4-dependent manner in mice.
G. Kaplan, D. I. Stirling, Amino-substituted thalidomide analogs: Potent inhibitors
PLOS ONE 8, e70945 (2013).
of TNF- production. Bioorg. Med. Chem. Lett. 9, 1625–1630 (1999).
48. D. Lippai, S. Bala, J. Petrasek, T. Csak, I. Levin, E. A. Kurt-Jones, G. Szabo, Alcohol-induced
25. C. Galustian, B. Meyer, M.-C. Labarthe, K. Dredge, D. Klaschka, J. Henry, S. Todryk, R. Chen,
IL-1 in the brain is mediated by NLRP3/ASC inflammasome activation that amplifies
G. Muller, D. Stirling, P. Schafer, J. B. Bartlett, A. G. Dalgleish, The anti-cancer agents neuroinflammation. J. Leukoc. Biol. 94, 171–182 (2013).
lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory 49. J. Liu, J. M. Lewohl, P. R. Dodd, P. K. Randall, R. A. Harris, R. D. Mayfield, Gene expression
cells. Cancer Immunol. Immunother. 58, 1033–1045 (2009). profiling of individual cases reveals consistent transcriptional changes in alcoholic
26. Z. Li, Y. Qiu, D. Personett, P. Huang, B. Edenfield, J. Katz, D. Babusis, Y. Tang, M. A. Shirely, human brain. J. Neurochem. 90, 1050–1058 (2004).
M. F. Moghaddam, J. A. Copland, H. W. Tun, Pomalidomide shows significant therapeutic 50. J. Liu, J. M. Lewohl, R. A. Harris, V. R. Iyer, P. R. Dodd, P. K. Randall, R. D. Mayfield, Patterns
activity against CNS lymphoma with a major impact on the tumor microenvironment of gene expression in the frontal cortex discriminate alcoholic from nonalcoholic
in murine models. PLOS ONE 8, e71754 (2013). individuals. Neuropsychopharmacology 31, 1574–1582 (2006).
27. C. C. Portugal, R. Socodato, T. Canedo, C. M. Silva, T. Martins, V. S. Coreixas, E. C. Loiola, 51. R. D. Mayfield, J. M. Lewohl, P. R. Dodd, A. Herlihy, J. Liu, R. A. Harris, Patterns of gene
B. Gess, D. Röhr, A. R. Santiago, P. Young, R. D. Minshall, R. Paes-de-Carvalho, expression are altered in the frontal and motor cortices of human alcoholics.
A. F. Ambrósio, J. B. Relvas, Caveolin-1–mediated internalization of the vitamin C J. Neurochem. 81, 802–813 (2002).

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 15 of 16


SCIENCE SIGNALING | RESEARCH ARTICLE

52. M. K. Mulligan, I. Ponomarev, R. J. Hitzemann, J. K. Belknap, B. Tabakoff, R. A. Harris, 75. P. Young, L. Qiu, D. Wang, S. Zhao, J. Gross, G. Feng, Single-neuron labeling
J. C. Crabbe, Y. A. Blednov, N. J. Grahame, T. J. Phillips, D. A. Finn, P. L. Hoffman, V. R. Iyer, with inducible Cre-mediated knockout in transgenic mice. Nat. Neurosci. 11, 721–728
G. F. Koob, S. E. Bergeson, Toward understanding the genetics of alcohol drinking (2008).
through transcriptome meta-analysis. Proc. Natl. Acad. Sci. U.S.A. 103, 6368–6373 (2006). 76. S. Pruett, W. Tan, G. E. Howell III, B. Nanduri, Dosage scaling of alcohol in binge exposure
53. E. Osterndorff-Kahanek, I. Ponomarev, Y. A. Blednov, R. A. Harris, Gene expression in brain models in mice: An empirical assessment of the relationship between dose, alcohol
and liver produced by three different regimens of alcohol consumption in mice: exposure, and peak blood concentrations in humans and mice. Alcohol 89, 9–17 (2020).
Comparison with immune activation. PLOS ONE 8, e59870 (2013). 77. C. Ikonomidou, P. Bittigau, M. J. Ishimaru, D. F. Wozniak, C. Koch, K. Genz, M. T. Price,
54. G. M. McCarthy, S. P. Farris, Y. A. Blednov, R. A. Harris, R. D. Mayfield, Microglial-specific V. Stefovska, F. Hörster, T. Tenkova, K. Dikranian, J. W. Olney, Ethanol-induced apoptotic
transcriptome changes following chronic alcohol consumption. Neuropharmacology neurodegeneration and fetal alcohol syndrome. Science 287, 1056–1060 (2000).
128, 416–424 (2018). 78. C. G. Langhammer, M. L. Previtera, E. S. Sweet, S. S. Sran, M. Chen, B. L. Firestein,
55. A. Dolganiuc, G. Bakis, K. Kodys, P. Mandrekar, G. Szabo, Acute ethanol treatment modulates Automated Sholl analysis of digitized neuronal morphology at multiple scales: Whole cell
Toll-like receptor-4 association with lipid rafts. Alcohol. Clin. Exp. Res. 30, 76–85 (2006). Sholl analysis versus Sholl analysis of arbor subregions. Cytometry A 77, 1160–1168
56. S. Oak, P. Mandrekar, D. Catalano, K. Kodys, G. Szabo, TLR2- and TLR4-mediated signals (2010).
determine attenuation or augmentation of inflammation by acute alcohol in monocytes. 79. G. Bindea, B. Mlecnik, H. Hackl, P. Charoentong, M. Tosolini, A. Kirilovsky, W.-H. Fridman,
J. Immunol. 176, 7628–7635 (2006). F. Pagès, Z. Trajanoski, J. Galon, ClueGO: A Cytoscape plug-in to decipher functionally
57. M. Pang, S. Bala, K. Kodys, D. Catalano, G. Szabo, Inhibition of TLR8- and TLR4-induced grouped gene ontology and pathway annotation networks. Bioinformatics 25,
Type I IFN induction by alcohol is different from its effects on inflammatory cytokine 1091–1093 (2009).
production in monocytes. BMC Immunol. 12, 55 (2011). 80. G. Zhou, O. Soufan, J. Ewald, R. E. W. Hancock, N. Basu, J. Xia, NetworkAnalyst 3.0: A visual
58. P. Mandrekar, S. Bala, D. Catalano, K. Kodys, G. Szabo, The opposite effects of acute analytics platform for comprehensive gene expression profiling and meta-analysis.
and chronic alcohol on lipopolysaccharide-induced inflammation are linked to IRAK-M Nucleic Acids Res. 47, W234–w241 (2019).
in human monocytes. J. Immunol. 183, 1320–1327 (2009). 81. M. Leger, A. Quiedeville, V. Bouet, B. Haelewyn, M. Boulouard, P. Schumann-Bard,
59. M. Manukyan, P. Nalbant, S. Luxen, K. M. Hahn, U. G. Knaus, RhoA GTPase activation T. Freret, Object recognition test in mice. Nat. Protoc. 8, 2531–2537 (2013).
by TLR2 and TLR3 ligands: Connecting via Src to NF-B. J. Immunol. 182, 3522–3529 82. A. Ennaceur, S. Michalikova, A. Bradford, S. Ahmed, Detailed analysis of the behavior

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


(2009). of Lister and Wistar rats in anxiety, object recognition and object location tasks. Behav.
60. S. Biswas, A. Zimman, D. Gao, T. V. Byzova, E. A. Podrez, TLR2 plays a key role in platelet Brain Res. 159, 247–266 (2005).
hyperreactivity and accelerated thrombosis associated with hyperlipidemia. Circ. Res. 83. M. Righi, L. Mori, G. De Libero, M. Sironi, A. Biondi, A. Mantovani, S. D. Donini,
121, 951–962 (2017). P. Ricciardi-Castagnoli, Monokine production by microglial cell clones. Eur. J. Immunol.
61. I.-T. Lee, S.-W. Wang, C.-W. Lee, C.-C. Chang, C.-C. Lin, S.-F. Luo, C.-M. Yang, Lipoteichoic 19, 1443–1448 (1989).
acid induces HO-1 expression via the TLR2/MyD88/c-Src/NADPH oxidase pathway 84. N. Janabi, S. Peudenier, B. Héron, K. H. Ng, M. Tardieu, Establishment of human microglial
and Nrf2 in human tracheal smooth muscle cells. J. Immunol. 181, 5098–5110 (2008). cell lines after transfection of primary cultures of embryonic microglial cells with the SV40
62. M. Wölfl, S. Schwinn, Y.-E. Yoo, M. L. Reß, M. Braun, M. Chopra, S. C. Schreiber, V. I. Ayala, large T antigen. Neurosci. Lett. 195, 105–108 (1995).
C. Ohlen, M. Eyrich, A. Beilhack, P. G. Schlegel, Src-kinase inhibitors sensitize human cells
of myeloid origin to Toll-like-receptor–induced interleukin 12 synthesis. Blood 122, Acknowledgments: We acknowledge the support of the following i3S Scientific Platforms:
1203–1213 (2013). Animal Facility, Cell Culture and Genotyping (CCGen), Translational Cytometry Unit (TraCy),
63. B. Haider, A. Duque, A. R. Hasenstaub, D. A. McCormick, Neocortical network activity and Advanced Light Microscopy (ALM), a member of the national infrastructure PPBI-
in vivo is generated through a dynamic balance of excitation and inhibition. J. Neurosci. Portuguese Platform of BioImaging (POCI-01–0145-FEDER-022122). We also acknowledge Rui
26, 4535–4545 (2006). Applelberg for generously supplying TNF KO mice. Funding: This work was financed by
64. N. Y. Golovenko, M. S. Zhuk, V. G. Zin’kovskii, O. V. Zhuk, M. V. Kopanitsa, FEDER - Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020 -
Pharmacokinetics of ethanol in mice with different alcohol motivation. Bull. Exp. Biol. Med. Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020,
132, 852–855 (2001). and by Portuguese funds through FCT - Fundação para a Ciência e a Tecnologia/Ministério da
65. J. C. Crabbe, C. J. Cotnam, A. J. Cameron, J. P. Schlumbohm, J. S. Rhodes, P. Metten, Ciência, Tecnologia e Ensino Superior in the framework of the project POCI-01-0145-
D. Wahlsten, Strain differences in three measures of ethanol intoxication in mice: FEDER-030647 (PTDC/SAU-TOX/30647/2017) in the T.S. laboratory. The projects FEDER
The screen, dowel and grip strength tests. Genes Brain Behav. 2, 201–213 (2003). Portugal [Norte-01-0145-FEDER-000008000008—Porto Neurosciences and Neurologic Disease
66. J. P. ter Horst, E. R. de Kloet, H. Schächinger, M. S. Oitzl, Relevance of stress and female sex Research Initiative at I3S, supported by Norte Portugal Regional Operational Programme
hormones for emotion and cognition. Cell. Mol. Neurobiol. 32, 725–735 (2012). (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European
67. A. Villa, P. Gelosa, L. Castiglioni, M. Cimino, N. Rizzi, G. Pepe, F. Lolli, E. Marcello, L. Sironi, Regional Development Fund (ERDF); FCOMP-01-0124-FEDER-021333] and FCT (PTDC/
E. Vegeto, A. Maggi, Sex-specific features of microglia from adult mice. Cell Rep. 23, MED-NEU/31318/2017) supported work in the J.B.R. laboratory. C.C.P. and R.S. hold
3501–3511 (2018). employment contracts financed by national funds through FCT—Fundação para a Ciência e a
68. M. Pascual, J. Montesinos, M. Marcos, J.-L. Torres, P. Costa-Alba, F. García-García, F.-J. Laso, Tecnologia, I.P., in the context of the program-contract described in paragraphs 4, 5, and 6 of
C. Guerri, Gender differences in the inflammatory cytokine and chemokine profiles art. 23 of Law no. 57/2016, of 29 August, as amended by Law no. 57/2017 of July 2019. T.C. was
induced by binge ethanol drinking in adolescence. Addict. Biol. 22, 1829–1841 (2017). supported by FCT (SFRH/BD/117148/2016). R.L.A. was supported by FCT (PD/
69. S. Alfonso-Loeches, M. Pascual, C. Guerri, Gender differences in alcohol-induced BD/114266/2016). A.M. was supported by FCT (IF/00753/2014). Author contributions: R.S.,
neurotoxicity and brain damage. Toxicology 311, 27–34 (2013). T.S., and J.B.R. designed the project. R.S., J.F.H., C.C.P., T.O.A., J.T.-M., R.L.A., T.C., C.S., and A.M.
70. L. Weinhard, G. di Bartolomei, G. Bolasco, P. Machado, N. L. Schieber, U. Neniskyte, performed experiments. R.S., T.S., and J.B.R. cosupervised the study. R.S. and J.B.R. wrote the
M. Exiga, A. Vadisiute, A. Raggioli, A. Schertel, Y. Schwab, C. T. Gross, Microglia remodel original draft. R.S., C.C.P., T.S., and J.B.R. reviewed and edited the manuscript. T.S. and J.B.R.
synapses by presynaptic trogocytosis and spine head filopodia induction. Nat. Commun. acquired funding. Competing interests: The authors declare that they have no competing
9, 1228 (2018). interests. Data and materials availability: All data needed to evaluate the conclusions in the
71. A. R. Aroor, R. C. Baker, Ethanol inhibition of phagocytosis and superoxide anion paper are present in the paper or the Supplementary Materials.
production by microglia. Alcohol 15, 277–280 (1998).
72. L. Gofman, J. M. Cenna, R. Potula, P2X4 receptor regulates alcohol-induced responses Submitted 15 December 2019
in microglia. J. Neuroimmune Pharmacol. 9, 668–678 (2014). Accepted 3 September 2020
73. S. Kalinin, M. González-Prieto, H. Scheiblich, L. Lisi, H. Kusumo, M. T. Heneka, Published 22 September 2020
J. L. M. Madrigal, S. C. Pandey, D. L. Feinstein, Transcriptome analysis of alcohol-treated 10.1126/scisignal.aba5754
microglia reveals downregulation of beta amyloid phagocytosis. J. Neuroinflammation
15, 141 (2018). Citation: R. Socodato, J. F. Henriques, C. C. Portugal, T. O. Almeida, J. Tedim-Moreira, R. L. Alves,
74. C. Karabiyik, R. Fernandes, F. R. Figueiredo, R. Socodato, C. Brakebusch, K. L. Lambertsen, T. Canedo, C. Silva, A. Magalhães, T. Summavielle, J. B. Relvas, Daily alcohol intake triggers
J. B. Relvas, S. D. Santos, Neuronal Rho GTPase Rac1 elimination confers neuroprotection aberrant synaptic pruning leading to synapse loss and anxiety-like behavior. Sci. Signal. 13,
in a mouse model of permanent ischemic stroke. Brain Pathol. 28, 569–580 (2018). eaba5754 (2020).

Socodato et al., Sci. Signal. 13, eaba5754 (2020) 22 September 2020 16 of 16


Daily alcohol intake triggers aberrant synaptic pruning leading to synapse loss and
anxiety-like behavior
Renato Socodato, Joana F. Henriques, Camila C. Portugal, Tiago O. Almeida, Joana Tedim-Moreira, Renata L. Alves, Teresa
Canedo, Cátia Silva, Ana Magalhães, Teresa Summavielle and João B. Relvas

Sci. Signal. 13 (650), eaba5754.


DOI: 10.1126/scisignal.aba5754

Microglia binge on synapses


Alcohol abuse has detrimental cognitive and behavioral consequences. Binge drinking activates resident
phagocytic immune cells in the brain called microglia in mice and is associated with anxiety in humans. Socodato et al.

Downloaded from http://stke.sciencemag.org/ on October 1, 2020


found that a binge drinking protocol in male mice induced microglia to selectively scavenge excitatory synapses between
neurons in the prefrontal cortex. The loss of these connections did not cause neuronal death during the study but instead
depressed neurotransmission and increased anxiety-like behaviors in the mice. These findings suggest that binge
drinking induces anxiety by activating microglia that destroy neuronal connections.

ARTICLE TOOLS http://stke.sciencemag.org/content/13/650/eaba5754

SUPPLEMENTARY http://stke.sciencemag.org/content/suppl/2020/09/18/13.650.eaba5754.DC1
MATERIALS

RELATED http://stke.sciencemag.org/content/sigtrans/12/569/eaar2124.full
CONTENT

REFERENCES This article cites 84 articles, 13 of which you can access for free
http://stke.sciencemag.org/content/13/650/eaba5754#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science Signaling (ISSN 1937-9145) is published by the American Association for the Advancement of Science, 1200 New
York Avenue NW, Washington, DC 20005. The title Science Signaling is a registered trademark of AAAS.
Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of
Science. No claim to original U.S. Government Works

You might also like