ArticlePDF Available

T1AM/TAAR1 System Reduces Inflammatory Response and β-Amyloid Toxicity in Human Microglial HMC3 Cell Line

Authors:

Abstract and Figures

Microglial dysfunction is one of the hallmarks and leading causes of common neurodegenerative diseases (NDDs), including Alzheimer’s disease (AD) and Parkinson’s disease (PD). All these pathologies are characterized by aberrant aggregation of disease-causing proteins in the brain, which can directly activate microglia, trigger microglia-mediated neuroinflammation, and increase oxidative stress. Inhibition of glial activation may represent a therapeutic target to alleviate neurodegeneration. Recently, 3-iodothyronamine (T1AM), an endogenous derivative of thyroid hormone (TH) able to interact directly with a specific GPCR known as trace amine-associated receptor 1 (TAAR1), gained interest for its ability to promote neuroprotection in several models. Nevertheless, T1AM’s effects on microglial disfunction remain still elusive. In the present work we investigated whether T1AM could inhibit the inflammatory response of human HMC3 microglial cells to LPS/TNFα or β-amyloid peptide 25–35 (Aβ25–35) stimuli. The results of ELISA and qPCR assays revealed that T1AM was able to reduce microglia-mediated inflammatory response by inhibiting the release of proinflammatory factors, including IL-6, TNFα, NF-kB, MCP1, and MIP1, while promoting the release of anti-inflammatory mediators, such as IL-10. Notably, T1AM anti-inflammatory action in HMC3 cells turned out to be a TAAR1-mediated response, further increasing the relevance of the T1AM/TAAR1 system in the management of NDDs.
Content may be subject to copyright.
Citation: Polini, B.; Ricardi, C.;
Bertolini, A.; Carnicelli, V.;
Rutigliano, G.; Saponaro, F.; Zucchi,
R.; Chiellini, G. T1AM/TAAR1
System Reduces Inflammatory
Response and β-Amyloid Toxicity in
Human Microglial HMC3 Cell Line.
Int. J. Mol. Sci. 2023,24, 11569.
https://doi.org/10.3390/
ijms241411569
Academic Editor: Masashi Tanaka
Received: 25 May 2023
Revised: 7 July 2023
Accepted: 13 July 2023
Published: 17 July 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
International Journal of
Molecular Sciences
Article
T1AM/TAAR1 System Reduces Inflammatory Response and
β-Amyloid Toxicity in Human Microglial HMC3 Cell Line
Beatrice Polini 1, *, Caterina Ricardi 1, Andrea Bertolini 1, Vittoria Carnicelli 1, Grazia Rutigliano 2,
Federica Saponaro 1, Riccardo Zucchi 1and Grazia Chiellini 1, *
1Department of Pathology, University of Pisa, 56100 Pisa, Italy; c.ricardi@student.unisi.it (C.R.);
a.bertolini@student.unisi.it (A.B.); vittoria.carnicelli@unipi.it (V.C.); federica.saponaro@unipi.it (F.S.);
riccardo.zucchi@unipi.it (R.Z.)
2Institute of Clinical Sciences, Imperial College London, London SW7 2AZ, UK;
grazia.rutigliano.gr@gmail.com
*Correspondence: beatrice.polini@farm.unipi.it (B.P.); grazia.chiellini@unipi.it (G.C.)
Abstract:
Microglial dysfunction is one of the hallmarks and leading causes of common neurodegen-
erative diseases (NDDs), including Alzheimer’s disease (AD) and Parkinson’s disease (PD). All these
pathologies are characterized by aberrant aggregation of disease-causing proteins in the brain, which
can directly activate microglia, trigger microglia-mediated neuroinflammation, and increase oxidative
stress. Inhibition of glial activation may represent a therapeutic target to alleviate neurodegeneration.
Recently, 3-iodothyronamine (T1AM), an endogenous derivative of thyroid hormone (TH) able to
interact directly with a specific GPCR known as trace amine-associated receptor 1 (TAAR1), gained
interest for its ability to promote neuroprotection in several models. Nevertheless, T1AM’s effects
on microglial disfunction remain still elusive. In the present work we investigated whether T1AM
could inhibit the inflammatory response of human HMC3 microglial cells to LPS/TNF
α
or
β
-amyloid
peptide 25–35 (A
β
25–35) stimuli. The results of ELISA and qPCR assays revealed that T1AM was
able to reduce microglia-mediated inflammatory response by inhibiting the release of proinflam-
matory factors, including IL-6, TNF
α
, NF-kB, MCP1, and MIP1, while promoting the release of
anti-inflammatory mediators, such as IL-10. Notably, T1AM anti-inflammatory action in HMC3 cells
turned out to be a TAAR1-mediated response, further increasing the relevance of the T1AM/TAAR1
system in the management of NDDs.
Keywords:
neurodegeneration; microglia; inflammation; neuroprotection; trace amine-associated
receptors type 1 (TAAR1); 3-iodothyronamine
1. Introduction
Microglial cells, the brain-resident immune cells, play a pivotal role in regulating the
cellular processes involved in the development of neuronal networks and the homeostasis
maintenance of the central nervous system (CNS) [
1
]. Notably, in response to abnormal
microenvironment factors, including, for example, stress, aging, injuries, infections, and
hypoxia–ischemia, microglia can assume different activation states, from neuroprotective
to neurodestructive, the equilibrium of which may contribute to the onset and outcome
of neuroinflammatory and neurodegenerative mechanisms [
2
]. Indeed, dysfunction of
microglia is one of the hallmarks and leading causes of common neurodegenerative diseases
(NDDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease
(HD), and amyotrophic lateral sclerosis (ALS) [
3
7
]. All these pathologies are characterized
by aberrant aggregation of disease-causing proteins in the brain [
8
], which can directly
activate microglia, trigger microglia-mediated neuroinflammation, and increase oxidative
stress [9].
Under physiological conditions microglial cells maintain a ramified cell shape, defined
as “resting state” (Figure 1), and vigilantly monitor and protect neuronal function. In
Int. J. Mol. Sci. 2023,24, 11569. https://doi.org/10.3390/ijms241411569 https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2023,24, 11569 2 of 15
the event of brain injury, infection, or neurodegeneration, microglia adopt a phagocytic
phenotype, defined as “M1 state”, in which the cell attains an amoeboid morphology, and
may secrete a wide variety of neurotoxic factors, including proinflammatory cytokines,
proteinases, and reactive oxygen species (ROS), leading to exacerbated damage and neu-
ronal death [
10
]. Accumulating evidence suggests that the initial classical activation of
microglia is followed by a secondary alternative activation, defined as “M2 state”, which is
important for neuronal cell repair, tissue remodeling, and suppression of inflammation [
11
]
(Figure 1). Since chronic inflammatory activation of microglia is correlated with common
NDDs, functional modulation of microglial phenotypes could represent a valid therapeutic
strategy [10,12,13].
Int.J.Mol.Sci.2023,24,115692of17
Underphysiologicalconditionsmicroglialcellsmaintainaramiedcellshape,de
nedas“restingstate”(Figure1),andvigilantlymonitorandprotectneuronalfunction.
Intheeventofbraininjury,infection,orneurodegeneration,microgliaadoptaphagocytic
phenotype,denedas“M1state”,inwhichthecellaainsanamoeboidmorphology,and
maysecreteawidevarietyofneurotoxicfactors,includingproinammatorycytokines,
proteinases,andreactiveoxygenspecies(ROS),leadingtoexacerbateddamageandneu
ronaldeath[10].Accumulatingevidencesuggeststhattheinitialclassicalactivationof
microgliaisfollowedbyasecondaryalternativeactivation,denedas“M2state”,which
isimportantforneuronalcellrepair,tissueremodeling,andsuppressionofinammation
[11](Figure1).Sincechronicinammatoryactivationofmicrogliaiscorrelatedwithcom
monNDDs,functionalmodulationofmicroglialphenotypescouldrepresentavalidther
apeuticstrategy[10,12,13].
Figure1.Diagramofmicrogliaactivation.Restingmicrogliamayturnintodistinctphenotypes,
namely,M1phenotypeandM2phenotype.M1classicalstatereleasesproinammatorycytokines
andcytotoxicsubstances,includingIL,IL6,TNFα,andROS,inducingneurologicaldamage.On
theotherhand,theM2alternativestateproducesantiinammatorycytokines,suchasIL10,able
toinhibittheproductionofproinammatorycytokinesbymicroglia,thusexertinganeuroprotec
tiveroleintheCNS.
Recently,3iodothyronamine(T1AM),anendogenousthyroidhormone(TH)deriv
ative,abletointeractdirectlywithaspecicGproteincoupledreceptorknownastrace
amineassociatedreceptor1(TAAR1)[14,15],hasgainedinterestforitsabilitytopromote
neuroprotectiveeectsinseveralmodels,includingseizurerelatedexcitotoxicdamage,
alteredautophagy,amyloidosis,andOGDinducedsynapticdysfunction[16–19],andto
ecientlycrosstheblood–brainbarrier(BBB)[20].TAAR1isanestablishedregulatory
proteinexpressedandbroadlydistributedinthebrainmonoaminergicsystems[21],
whichincludethedopaminergicsystem,thenoradrenergicsystem,andtheserotonergic
system[22].Collectively,allthesesystemsplayimportantrolesinmood,cognition,emo
tion,reward,learning,aention,andmotoractivity[23],andtheirdysregulationisasso
ciatedwithavarietyofneurologicalandneurodegenerativedisorders,severalofwhich
areamongtheleadingcausesofdeathanddisabilityworldwide[24].Recentstudiesre
vealedthatpharmacologicallytargetingTAAR1withintheCNShasresultedinsuccessful
clinicaltrialsforthetreatmentofschizophrenia,depression,addiction,andNDDs[25–
28].Morerecently,TAA R1expressionandfunctionalityinimmunesystemregulationand
Proinflammatory
phenotype
Microglia(resting)
Proinflammatory
stimuli
Antiinflammatory
stimuli
Antiinflammatory
phenotype
IL1β,IL6,TNF ,ROS
IL10
Inhibition
Figure 1.
Diagram of microglia activation. Resting microglia may turn into distinct phenotypes,
namely, M1 phenotype and M2 phenotype. M1 classical state releases proinflammatory cytokines
and cytotoxic substances, including IL-1ß, IL-6, TNF
α
, and ROS, inducing neurological damage. On
the other hand, the M2 alternative state produces anti-inflammatory cytokines, such as IL-10, able to
inhibit the production of proinflammatory cytokines by microglia, thus exerting a neuroprotective
role in the CNS.
Recently, 3-iodothyronamine (T1AM), an endogenous thyroid hormone (TH) deriva-
tive, able to interact directly with a specific G-protein coupled receptor known as trace
amine-associated receptor 1 (TAAR1) [
14
,
15
], has gained interest for its ability to promote
neuroprotective effects in several models, including seizure-related excitotoxic damage,
altered autophagy, amyloidosis, and OGD-induced synaptic dysfunction [
16
19
], and to effi-
ciently cross the blood–brain barrier (BBB) [
20
]. TAAR1 is an established regulatory protein
expressed and broadly distributed in the brain monoaminergic systems [
21
], which in-
clude the dopaminergic system, the noradrenergic system, and the serotonergic
system [22]
.
Collectively, all these systems play important roles in mood, cognition, emotion, reward,
learning, attention, and motor activity [
23
], and their dysregulation is associated with
a variety of neurological and neurodegenerative disorders, several of which are among
the leading causes of death and disability worldwide [
24
]. Recent studies revealed that
pharmacologically targeting TAAR1 within the CNS has resulted in successful clinical
trials for the treatment of schizophrenia, depression, addiction, and NDDs [
25
28
]. More
recently, TAAR1 expression and functionality in immune system regulation and immune
cell activation has become a topic of emerging interest [
29
]; nevertheless, so far, few studies
have examined the role of TAAR1 in CNS-resident neuroimmune cells [
30
,
31
]. Notably,
a recent report highlighted the expression of TAAR1 in macrophages/microglia bordering
multiple sclerosis (MS) lesions [
32
], supporting TAAR1 as a novel pharmacological target
in cells directly implicated in neuroinflammation.
Int. J. Mol. Sci. 2023,24, 11569 3 of 15
On these premises, we directed our attention to examining the potential of the
T1AM/TAAR1 signaling system in regulating brain neuroinflammatory responses. Using
HMC3 human microglia cells as an
in vitro
model [
33
], we demonstrated, for the first time,
that T1AM inhibits LPS/TNF
α
-induced inflammatory response through the activation of
TAAR1. Since it is well known that fibrillar amyloid-
β
(A
β
) peptides play an important
role in microglial activation in AD [
34
], we also showed that the T1AM-TAAR1 signaling
pathway was able to protect against A
β
-induced cytotoxic and inflammatory responses in
HMC3 cells, and that TAAR1 stimulation can inactivate microglial NF-κB signaling.
Even though still at a preliminary level, the results of our study highlight the ability of
T1AM to extensively modulate microglia-mediated neuroinflammation, providing further
insight into its possible therapeutic application for the prevention of neurodegeneration.
2. Results
2.1. T1AM Decreased the Inflammatory Phenotype of LPS/TNFα-Stimulated HMC3 Human
Microglial Cells
As previously reported [
35
37
], exposure of human microglial clone 3 (HMC3) cells
to LPS/TNF
α
stimulus for 24 h resulted in a significant increase in proinflammatory IL-6
release in cell culture media, while no effect on anti-inflammatory IL-10 levels was observed
as compared with control cells. In the present study, HMC3 microglial cell line was used as
an in vitro model to investigate the ability of T1AM to prevent neuroinflammation.
Dose–response experiments were carried out by exposing HMC3 cells to pretreat-
ment with increasing concentrations (0.1, 1, and 10
µ
M) of T1AM for 1 h, followed by
LPS/TNF
α
treatment for 24 h. ELISA assays on cell culture media revealed that T1AM
causes a significant (p< 0.05) dose-dependent reduction of IL-6 levels as compared to
LPS/TNF
α
-treated cells (Figure 2A). A significant (p< 0.05) dose-dependent increase in
IL-10 levels was also observed in the same set of experiments (Figure 2B), suggesting that
T1AM pretreatment may temper hyperinflammation. Moreover, gene expression analysis
revealed that pretreatment with 1 or 10
µ
M T1AM significantly decreases the expression of
inflammatory-response-related genes in LPS/TNF
α
-treated cells, including the monocyte
chemoattract protein-1 (MCP1), the macrophage inflammatory protein-1 (MIP1), and the
transcription factor NF-kB (Figure 3).
Int.J.Mol.Sci.2023,24,115694of17
Figure2.Releaseofpro‐andantiinammatoryinterleukins,IL–6(A)andIL–10(B),inducedby
dierentconcentrationsofT1AM.Datarepresentmeans±S.E.M.fromthreeindependentexperi
ments(n=3),performedinduplicate.Statisticalanalysiswasperformedbyordinaryoneway
ANOVAfollowedbyDunne’smultiplecomparisontest.Foreachexperimentalconditionase
lectedspecicpaernhasbeenused.
.
Figure3.T1AMdecreasestheexpressionofinammatoryresponserelatedgenesstimulatedby
LPS/TNFαtreatmentinHMC3cells.(A)monocytechemoaractprotein–1(MCP1),(B)macrophage
inammatoryprotein–1(MIP1),and(C)transcriptionfactorNFkB.Datarepresentmeans±S.E.M.
fromthreeindependentexperiments(n=3),performedinduplicate.Statisticalanalysiswasper
formedbyordinaryonewayANOVAfollowedbyDunne’smultiplecomparisontest.Foreach
experimentalconditionaselectedspecicpaernhasbeenused.
Inaddition,T1AMwasnontoxictomicroglialcellswhenusedat0.1,1,and10µM
concentrationsfor24h(Figure4).
0
100
200
300
400
500
IL-10 [pg/ml]
LPS/TNF
- + + + +
T1AM (µM)
- -
0.1 1 10
0.0031
0.0027
AB
0
250
500
750
1000
1250
1500
IL-6 [pg/ml]
LPS/TNF
- + + + +
T1AM (µM)
- -
0.1 1 10
0.0021
0.0048
0.0078
0.0037
0
2
4
6
8
10
MCP1 expression levels
(2
-

Ct
)
LPS/TNF
- + + +
T1AM (µM)
- -
1
10
0.0071
0.018
0.0029
0
2
4
6
8
MIP1 expression levels
(2
-

Ct
)
LPS/TNF
- + + +
T1AM (µM)
- -
1
10
0.0028 0.008
0.0015
0
1
2
3
4
NFKB expression levels
(2
-

Ct
)
LPS/TNF
- + + +
T1AM (µM)
- -
1
10
0.0078 0.032
0.0081
AB C
Figure 2.
Release of pro- and anti-inflammatory interleukins, IL–6 (
A
) and IL–10 (
B
), induced by dif-
ferent concentrations of T1AM. Data represent means
±
S.E.M. from three independent experiments
(n= 3), performed in duplicate. Statistical analysis was performed by ordinary one-way ANOVA
followed by Dunnett’s multiple comparison test. For each experimental condition a selected specific
pattern has been used.
Int. J. Mol. Sci. 2023,24, 11569 4 of 15
Int.J.Mol.Sci.2023,24,115694of17
Figure2.Releaseofpro‐andantiinammatoryinterleukins,IL–6(A)andIL–10(B),inducedby
dierentconcentrationsofT1AM.Datarepresentmeans±S.E.M.fromthreeindependentexperi
ments(n=3),performedinduplicate.Statisticalanalysiswasperformedbyordinaryoneway
ANOVAfollowedbyDunne’smultiplecomparisontest.Foreachexperimentalconditionase
lectedspecicpaernhasbeenused.
.
Figure3.T1AMdecreasestheexpressionofinammatoryresponserelatedgenesstimulatedby
LPS/TNFαtreatmentinHMC3cells.(A)monocytechemoaractprotein–1(MCP1),(B)macrophage
inammatoryprotein–1(MIP1),and(C)transcriptionfactorNFkB.Datarepresentmeans±S.E.M.
fromthreeindependentexperiments(n=3),performedinduplicate.Statisticalanalysiswasper
formedbyordinaryonewayANOVAfollowedbyDunne’smultiplecomparisontest.Foreach
experimentalconditionaselectedspecicpaernhasbeenused.
Inaddition,T1AMwasnontoxictomicroglialcellswhenusedat0.1,1,and10µM
concentrationsfor24h(Figure4).
0
100
200
300
400
500
IL-10 [pg/ml]
LPS/TNF
- + + + +
T1AM (µM)
- -
0.1 1 10
0.0031
0.0027
AB
0
250
500
750
1000
1250
1500
IL-6 [pg/ml]
LPS/TNF
- + + + +
T1AM (µM)
- -
0.1 1 10
0.0021
0.0048
0.0078
0.0037
0
2
4
6
8
10
MCP1 expression levels
(2
-

Ct
)
LPS/TNF
- + + +
T1AM (µM)
- -
1
10
0.0071
0.018
0.0029
0
2
4
6
8
MIP1 expression levels
(2
-

Ct
)
LPS/TNF
- + + +
T1AM (µM)
- -
1
10
0.0028 0.008
0.0015
0
1
2
3
4
NFKB expression levels
(2
-

Ct
)
LPS/TNF
- + + +
T1AM (µM)
- -
1
10
0.0078 0.032
0.0081
AB C
Figure 3.
T1AM decreases the expression of inflammatory-response-related genes stimulated by
LPS/TNF
α
treatment in HMC3 cells. (
A
) monocyte chemoattract protein–1 (MCP1), (
B
) macrophage
inflammatory protein–1 (MIP1), and (
C
) transcription factor NF-kB. Data represent means
±
S.E.M.
from three independent experiments (n= 3), performed in duplicate. Statistical analysis was per-
formed by ordinary one-way ANOVA followed by Dunnett’s multiple comparison test. For each
experimental condition a selected specific pattern has been used.
In addition, T1AM was nontoxic to microglial cells when used at 0.1, 1, and 10
µ
M
concentrations for 24 h (Figure 4).
Int.J.Mol.Sci.2023,24,115695of17
Figure4.MTTassayperformedwithdierentconcentrationsofT1AM.(A)EectofT1AMon
HMC3Cellviability.(B)EectofT1AMontheviabilityofLPS/TNFα–treatedcells.Datarepresent
means±S.E.M.fromthreeindependentexperiments,performedintriplicate(n=3).Statisticalanal
ysiswasperformedbyordinaryonewayANOVAfollowedbyTukey’smultiplecomparisontest.
Foreachexperimentalconditionaselectedspecicpaernhasbeenused.
2.2.T1AMUptakeandMetabolisminHMC3HumanMicroglialCells
T1AMisknowntobetransportedinsidecellsandrapidlytransformedinto3iodo
thyroaceticacid(TA1)[20,38,39].
Liquidchromatographywithtandemmassspectrometry(LCMS/MS)wasusedto
measureT1AMandTA1levelsinHMC3cells.AfterincubatingHMC3cellpreparations
with0.1µMT1AMfor5,15,30,and60minat37°C,thecellculturemediawerecollected,
andcelllysateswerepreparedaccordingtoapreviouslyreportedprocedure[40].
AsshowninTabl e1,T1AMwasrapidlytakenupbyHMC3cellsandcatabolizedto
TA1,indicatingthatinHMC3cells,amineoxidasesweremetabolicallyactive.Notably,
theproductofT1AMcatabolism,TA1,afterformingwasreleasedfromcells,anddetected
inthecollectedcellculturemedia,showingincreasingconcentrationsovertime.InHMC3
celllysates,theconcentrationofT1AMappearedtoremainconstantovertimeatvalues
correspondingtoapproximately25%oftheadministereddose,whereasonlyanegligible
amountofTA1 wasfound.
Tab le1.LCMS/MSmeasurementofT1AMandTA1levelsinHMC3cells
Time
(min)
MediumCellLysates
T1AM(nM)TA1(nM)T1AM(nM)TA1(nM)
0N.D.N.D.N.D.N.D.
564.1±2.700.31±0.02026.0±0.280.06±0.01
1557.2±5.400.66±0.2024.9±0.740.12±0.02
3056.7±8.101.33±0.1527.0±0.300.14±0.02
6053.4±3.603.81±0.8024.5±0.810.23±0.04
ConcentrationsofT1AMandTA1inmediumandcelllysatesafter0,5,15,30,and60minofinfusion.
Datarepresentmean±SEM,n=3pergroup,andareexpressedasnM.T1AMorTA1 contentswere
measuredinmediumandlysateHMC3cells,whichwereincubatedfor0,5,15,30,and60minwith
T1AM(0.1µM).N.D.,notdetectable.
0
20
40
60
80
100
120
Cell viability (% vs. CTRL)
T1AM (µM)
-
0.1 1 10
AB
0
20
40
60
80
100
120
Cell viability (% vs. CTRL)
LPS/TNF
- + + +
T1AM (µM)
- -
1 10
Figure 4.
MTT assay performed with different concentrations of T1AM. (
A
) Effect of T1AM
on HMC3 Cell viability. (
B
) Effect of T1AM on the viability of LPS/TNF
α
–treated cells. Data
represent
means ±S.E.M
. from three independent experiments, performed in triplicate (n= 3). Statis-
tical analysis was performed by ordinary one-way ANOVA followed by Tukey’s multiple comparison
test. For each experimental condition a selected specific pattern has been used.
2.2. T1AM Uptake and Metabolism in HMC3 Human Microglial Cells
T1AM is known to be transported inside cells and rapidly transformed into 3-iodothyroacetic
acid (TA1) [20,38,39].
Liquid chromatography with tandem mass spectrometry (LC–MS/MS) was used to
measure T1AM and TA1 levels in HMC3 cells. After incubating HMC3 cell preparations
with 0.1
µ
M T1AM for 5, 15, 30, and 60 min at 37
C, the cell culture media were collected,
and cell lysates were prepared according to a previously reported procedure [40].
As shown in Table 1, T1AM was rapidly taken up by HMC3 cells and catabolized to
TA1, indicating that in HMC3 cells, amine oxidases were metabolically active. Notably, the
Int. J. Mol. Sci. 2023,24, 11569 5 of 15
product of T1AM catabolism, TA1, after forming was released from cells, and detected in
the collected cell culture media, showing increasing concentrations over time. In HMC3
cell lysates, the concentration of T1AM appeared to remain constant over time at values
corresponding to approximately 25% of the administered dose, whereas only a negligible
amount of TA1 was found.
Table 1. LC–MS/MS measurement of T1AM and TA1 levels in HMC3 cells.
Time (min) Medium Cell Lysates
T1AM (nM) TA1 (nM) T1AM (nM) TA1 (nM)
0 N.D. N.D. N.D. N.D.
5 64.1 ±2.70 0.31 ±0.020 26.0 ±0.28 0.06 ±0.01
15 57.2 ±5.40 0.66 ±0.20 24.9 ±0.74 0.12 ±0.02
30 56.7 ±8.10 1.33 ±0.15 27.0 ±0.30 0.14 ±0.02
60 53.4 ±3.60 3.81 ±0.80 24.5 ±0.81 0.23 ±0.04
Concentrations of T1AM and TA1 in medium and cell lysates after 0, 5, 15, 30, and 60 min of infusion. Data
represent mean
±
SEM, n= 3 per group, and are expressed as nM. T1AM or TA1 contents were measured in
medium and lysate HMC3 cells, which were incubated for 0, 5, 15, 30, and 60 min with T1AM (0.1
µ
M). N.D.,
not detectable.
2.3. Trace Amine-Associated Receptor TAAR1 Is Involved in T1AM-Mediated Anti-Inflammatory
Response of Microglial Cells
The evidence that T1AM was able to suppress the response of microglia cells to
inflammatory stress prompted us to extend our investigation to its mechanism of action.
It is known from the literature that T1AM is a high-affinity ligand for TAAR1 [
14
], and
TAAR1 was demonstrated to mediate T1AM’s protective effect against synaptic plasticity
abnormalities in a mouse model of AD [
41
]. Therefore, we explored whether TAAR1 could
also be involved in T1AM-mediated anti-inflammatory response of HMC3 cells.
First, expression of TAAR1 in HMC3 microglial cells was assessed by qPCR analysis.
As shown in Figure 5, resting microglial cells showed TAAR1 expression, and no changes
were observed after 24 h treatment with LPS/TNFα.
Int.J.Mol.Sci.2023,24,115696of17
2.3.TraceAmineAssociatedReceptorTAAR1IsInvolvedinT1AMMediatedAntiInamma
toryResponseofMicroglialCells
TheevidencethatT1AMwasabletosuppresstheresponseofmicrogliacellstoin
ammatorystresspromptedustoextendourinvestigationtoitsmechanismofaction.
ItisknownfromtheliteraturethatT1AMisahighanityligandforTAAR1[14],
andTAAR1 wasdemonstratedtomediateT1AMsprotectiveeectagainstsynapticplas
ticityabnormalitiesinamousemodelofAD[41].Therefore,weexploredwhetherTAAR1
couldalsobeinvolvedinT1AMmediatedantiinammatoryresponseofHMC3cells.
First,expressionofTAAR1inHMC3microglialcellswasassessedbyqPCRanalysis.
AsshowninFigure5,restingmicroglialcellsshowedTAAR1expression,andnochanges
wereobservedafter24htreatmentwithLPS/TNFα.
Figure5.RealtimequanticationofTAAR1expressioninHMC3cellsbeforeandafterexposureto
LPS(10µg/mL)/TNFα (50ng/mL)treatmentfor24h.Datarepresentmeans±S.E.M.fromthree
independentexperiments,performedintriplicate(n=3).Statisticalanalysiswasperformedbyor
dinaryStudent’sttest.
Then,weinvestigatedwhetherTAAR1couldplayaroleinourmodelofinamed
microgliabyusingaTAAR1selectiveantagonist(EPPTB)andaselectiveagonist
(RO5166017)[42].
WeobservedthatT1AM’sprotectiveeectagainstmicrogliaactivationwasabolished
bycoadministrationofTAAR1selectiveantagonistEPPTB(5nM)(Figure6A,B).Con
versely,theadministrationoftheTAAR1agonistRO5166017(1µM)toLPS/TNFα‐stimu
latedHMC3cellsmimickedtheeectsonthereleaseofbothIL6andIL10previously
observedafteradministeringT1AM(10µM)(Figure6A,B).
HMC3
HMC3 + INF
0.0
0.1
0.2
0.3
TAAR1 mRNA expression
(2
-Ct
)
Figure 5.
Real-time quantification of TAAR1 expression in HMC3 cells before and after exposure
to LPS (10
µ
g/mL)/TNF
α
(50 ng/mL) treatment for 24 h. Data represent means
±
S.E.M. from
three independent experiments, performed in triplicate (n= 3). Statistical analysis was performed by
ordinary Student’s t-test.
Then, we investigated whether TAAR1 could play a role in our model of inflamed mi-
croglia by using a TAAR1 selective antagonist (EPPTB) and a selective agonist (RO5166017) [
42
].
We observed that T1AM’s protective effect against microglia activation was abolished
by coadministration of TAAR1 selective antagonist EPPTB (5 nM) (Figure 6A,B). Conversely,
Int. J. Mol. Sci. 2023,24, 11569 6 of 15
the administration of the TAAR1 agonist RO5166017 (1
µ
M) to LPS/TNF
α
-stimulated
HMC3 cells mimicked the effects on the release of both IL-6 and IL-10 previously observed
after administering T1AM (10 µM) (Figure 6A,B).
Int.J.Mol.Sci.2023,24,115697of17
Figure6.AbilityofT1AMtodecreasetheinammatoryphenotypeofLPS/TNFα‐stimulatedHMC3
cellsbythemodulationofTAAR1. Barsrepresenttherelease(pg/mL)ofIL–6(A)andIL–10(B)in
thepresenceofthedrugsattheindicatedconcentrations.Datarepresentmeans±S.E.M.frominde
pendentexperiments(n=3),performedinduplicate.Statisticalanalysiswasperformedbyordinary
onewayANOVAfollowedbyDunne’smultiplecomparisontest.Foreachexperimentalcondition
aselectedspecicpaernhasbeenused.
2.4.3IodothyroaceticAcid(TA1)WasNotAbletoDecreasetheInammatoryPhenotypeof
LPS/TNFα‐StimulatedHMC3HumanMicroglialCells
Since3iodothyroaceticacid(TA1),themajorcataboliteofT1AM[43–45],hasbeen
reportedtoberesponsibleforsomeeectselicitedaftertheadministrationofexogenous
T1AM,wecheckedwhetherTA1administrationcouldalsodecreasetheinammatory
phenotypeofLPS/TNFα‐stimulatedHMC3cells.WeobservedthatadministrationofTA1
(0.1,1,and10µM)toLPS/TNFα‐stimulatedHMC3cellswasnotabletoproduceanysig
nicanteectonbothIL6andIL10releasefromcells(Figure7),suggestingthatthede
creasedactivationofmicrogliaisexclusivelyduetotheactionofT1AMthroughtheinter
actionwiththeTAAR1receptor.
Figure7.3iodothyroaceticacidTA1doesnotdecreasetheinammatoryphenotypeofLPS/TNFα
stimulatedHMC3cells.AfterpretreatmentwithTA1 attheindicatedconcentrationsthereleaseof
0
250
500
750
1000
1250
1500
IL-6 [pg/ml]
LPS/TNF - + + + + + +
T
1
AM (µM) - -
10
-
-
10 -
RO5166017 (µM) - - -
0.25 1
-
1
EPPTB (µM)
- - -
-
-
1 1
0.0035 0.0041
0.0045
0.0031
0
200
400
600
IL-10 [pg/ml]
LPS/TNF - + + + + + +
T
1
AM (µM) - -
10
-
-
10 -
RO5166017 (µM) - - -
0.25 1
-
1
EPPTB (µM)
- - -
-
-
1 1
0.0023
0.0031
0.0019
AB
0
50
100
150
200
250
IL-10 [pg/ml]
LPS/TNF
- + + + +
TA
1
(µM) - +
0.1 1 10
AB
0
250
500
750
1000
1250
1500
IL-6 [pg/ml]
LPS/TNF
- + + + +
TA
1
(µM) - +
0.1 1 10
0.0023
Figure 6.
Ability of T1AM to decrease the inflammatory phenotype of LPS/TNF
α
-stimulated HMC3
cells by the modulation of TAAR1. Bars represent the release (pg/mL) of IL–6 (
A
) and IL–10 (
B
)
in the presence of the drugs at the indicated concentrations. Data represent means
±
S.E.M. from
independent experiments (n= 3), performed in duplicate. Statistical analysis was performed by
ordinary one-way ANOVA followed by Dunnett’s multiple comparison test. For each experimental
condition a selected specific pattern has been used.
2.4. 3-Iodothyroacetic Acid (TA1) Was Not Able to Decrease the Inflammatory Phenotype of
LPS/TNFα-Stimulated HMC3 Human Microglial Cells
Since 3-iodothyroacetic acid (TA1), the major catabolite of T1AM [
43
45
], has been
reported to be responsible for some effects elicited after the administration of exogenous
T1AM, we checked whether TA1 administration could also decrease the inflammatory
phenotype of LPS/TNF
α
-stimulated HMC3 cells. We observed that administration of TA1
(0.1, 1, and 10
µ
M) to LPS/TNF
α
-stimulated HMC3 cells was not able to produce any
significant effect on both IL-6 and IL-10 release from cells (Figure 7), suggesting that the
decreased activation of microglia is exclusively due to the action of T1AM through the
interaction with the TAAR1 receptor.
2.5. T1AM-TAAR1 System Protects against A
β25–35
-Mediated Release of Proinflammatory Factors
in HMC3 Cells
Studies have revealed that A
β
oligomers activate microglia to secrete proinflammatory
factors, including cytokines, chemokines, complements factors, and a large variety of free
radicals [
46
,
47
]. Suppressing the response of microglia cells to inflammatory stress may
attenuate AD pathology and lessen the disease progression [48,49].
We initially assessed the effects of
β
-amyloid peptide 25–35 (A
β
25–35) [
50
] on HMC3
cells’ viability. The cells were treated with two different concentrations (1 and 10
µ
M)
of A
β
25–35 for 24 or 48 h. A
β
25–35 was found to induce cytotoxicity in HMC3 cells in
a concentration- and time-dependent manner, with a 10
µ
M concentration promoting a 50%
reduction in cell viability after 24 h (Figure 8A). We thus used this condition to evaluate the
modulation of A
β
25–35 cytotoxicity in further experiments. HMC3 cells were treated with
10
µ
M A
β
25–35 in the presence of different concentrations (0.1, 1, and 10
µ
M) of T1AM
for 24 h to determine the effects of T1AM on
β
-amyloid-induced cytotoxicity. In line with
expectations, MTT assays indicated that T1AM was able to protect cells from A
β
-induced
Int. J. Mol. Sci. 2023,24, 11569 7 of 15
cytotoxicity in a concentration-dependent manner, with the identified best concentrations
of 1 and 10 µM being used in further experiments (Figure 8B).
Int.J.Mol.Sci.2023,24,115697of17
Figure6.AbilityofT1AMtodecreasetheinammatoryphenotypeofLPS/TNFα‐stimulatedHMC3
cellsbythemodulationofTAAR1. Barsrepresenttherelease(pg/mL)ofIL–6(A)andIL–10(B)in
thepresenceofthedrugsattheindicatedconcentrations.Datarepresentmeans±S.E.M.frominde
pendentexperiments(n=3),performedinduplicate.Statisticalanalysiswasperformedbyordinary
onewayANOVAfollowedbyDunne’smultiplecomparisontest.Foreachexperimentalcondition
aselectedspecicpaernhasbeenused.
2.4.3IodothyroaceticAcid(TA1)WasNotAbletoDecreasetheInammatoryPhenotypeof
LPS/TNFα‐StimulatedHMC3HumanMicroglialCells
Since3iodothyroaceticacid(TA1),themajorcataboliteofT1AM[43–45],hasbeen
reportedtoberesponsibleforsomeeectselicitedaftertheadministrationofexogenous
T1AM,wecheckedwhetherTA1administrationcouldalsodecreasetheinammatory
phenotypeofLPS/TNFα‐stimulatedHMC3cells.WeobservedthatadministrationofTA1
(0.1,1,and10µM)toLPS/TNFα‐stimulatedHMC3cellswasnotabletoproduceanysig
nicanteectonbothIL6andIL10releasefromcells(Figure7),suggestingthatthede
creasedactivationofmicrogliaisexclusivelyduetotheactionofT1AMthroughtheinter
actionwiththeTAAR1receptor.
Figure7.3iodothyroaceticacidTA1doesnotdecreasetheinammatoryphenotypeofLPS/TNFα
stimulatedHMC3cells.AfterpretreatmentwithTA1 attheindicatedconcentrationsthereleaseof
0
250
500
750
1000
1250
1500
IL-6 [pg/ml]
LPS/TNF - + + + + + +
T
1
AM (µM) - -
10
-
-
10 -
RO5166017 (µM) - - -
0.25 1
-
1
EPPTB (µM)
- - -
-
-
1 1
0.0035 0.0041
0.0045
0.0031
0
200
400
600
IL-10 [pg/ml]
LPS/TNF - + + + + + +
T
1
AM (µM) - -
10
-
-
10 -
RO5166017 (µM) - - -
0.25 1
-
1
EPPTB (µM)
- - -
-
-
1 1
0.0023
0.0031
0.0019
AB
0
50
100
150
200
250
IL-10 [pg/ml]
LPS/TNF
- + + + +
TA
1
(µM) - +
0.1 1 10
AB
0
250
500
750
1000
1250
1500
IL-6 [pg/ml]
LPS/TNF
- + + + +
TA
1
(µM) - +
0.1 1 10
0.0023
Figure 7.
3-iodothyroacetic acid TA1 does not decrease the inflammatory phenotype of LPS/TNF
α
stimulated HMC3 cells. After pretreatment with TA1 at the indicated concentrations the release
of IL–6 (
A
) and IL–10 (
B
) from LPS/TNF
α
–stimulated HMC3 cells were examined. Data represent
means
±
S.E.M. from three independent experiments (n= 3), performed in duplicate. Statistical
analysis was performed by ordinary one-way ANOVA followed by Dunnett’s multiple comparison
test. For each experimental condition a selected specific pattern has been used.
Figure 8.
MTT assays in HMC3 cells exposed to A
β
. (
A
) Effects on HMC3 cells’ viability of treatment
with
β
–amyloid peptide 25–35 (A
β
) at 1 and 10
µ
M for 24 or 48 h. (
B
) Effects on HMC3 cells’ viability
of pretreatment with T1AM at 0.1, 1, and 10
µ
M, followed by exposure to 10
µ
M A
β
for 24 h. Data
represent means
±
S.E.M. from three independent experiments, performed in triplicate (n= 3).
Statistical analysis was performed by ordinary one-way ANOVA followed by Dunnett’s multiple
comparison test. For each experimental condition a selected specific pattern has been used.
Having assessed the cytoprotective effects of T1AM, we moved on to examine the
ability of T1AM to suppress
β
-amyloid-induced upregulation of proinflammatory cytokines
production. Exposure of HMC3 cells to 10
µ
M A
β
25–35 for 24 h promoted a significant
increase in the secretion of common proinflammatory cytokines (TNF-
α
and IL-6), and no
effect on the release of anti-inflammatory cytokine IL-10 (Figure 9A–C). Then, we repeated
the experiment, exposing HMC3 cells to pretreatment with 1 and 10
µ
M T1AM. Compared
with the A
β
-treated group, the levels of TNF-
α
and IL-6 were found to show a significant
return to normal levels in the group receiving the combined A
β
25–35 and T1AM treatment
Int. J. Mol. Sci. 2023,24, 11569 8 of 15
(Figure 9A,B). In addition, pretreatment with T1AM significantly increased the secretion of
anti-inflammatory cytokine IL-10 (Figure 9C). Taken together, these findings indicated the
potential of T1AM to inhibit A
β
-induced microglia activation. Since TAAR1 has previously
been found to mediate the beneficial effects of T1AM on the inflammatory response of
LPS/TNF
α
-treated HMC3 cells, we subsequently proceeded to examine the role of TAAR1
in mediating the anti-inflammatory effects of T1AM in β-amyloid-induced HMC3 cells.
Int.J.Mol.Sci.2023,24,115699of17
repeatedtheexperiment,exposingHMC3cellstopretreatmentwith1and10µMT1AM.
ComparedwiththeAβ‐treatedgroup,thelevelsofTNF‐αandIL6werefoundtoshowa
signicantreturntonormallevelsinthegroupreceivingthecombinedAβ25–35and
T1AMtreatment(Figure9A,B).Inaddition,pretreatmentwithT1AMsignicantlyin
creasedthesecretionofantiinammatorycytokineIL10(Figure9C).Tak e ntogether,
thesendingsindicatedthepotentialofT1AMtoinhibitAβ‐inducedmicrogliaactivation.
SinceTAAR1 haspreviouslybeenfoundtomediatethebenecialeectsofT1AMonthe
inammatoryresponseofLPS/TNFα‐treatedHMC3cells,wesubsequentlyproceededto
examinetheroleofTAAR 1inmediatingtheantiinammatoryeectsofT1AMinβam
yloidinducedHMC3cells.
Figure9.T1AMpretreatmentmodulatedthereleaseofproinammatory(TNF‐α,IL6)andanti
inammatory(IL10)interleukinsinAβ‐inducedHMC3cells.Barsrepresenttherelease(pg/mL)of
TNFα(A),IL–6(B),andIL–10(C)inthepresenceofthedrugsattheindicatedconcentrations.Data
representmeans±S.E.M.fromthreeindependentexperiments(n=3),performedinduplicate.Sta
tisticalanalysiswasperformedbyordinaryonewayANOVAfollowedbyDunne’smultiplecom
parisontest.Foreachexperimentalconditionaselectedspecicpaernhasbeenused.
WeobservedthatT1AM’sprotectiveeectagainstAβ‐inducedmicrogliaactivation
wasabolishedbycoadministrationofTAAR1 selectiveantagonistEPPTB(1µM)(Figure
9A–C).
DysregulationofthetranscriptionfactorNF‐κBhasbeenwidelyassociatedwithAD,
leadingtoglialcellsactivationandneuroinammation[51].Moreimportantly,theinacti
vationofmicroglialNF‐κBhasbeenshowntorestorecognitivedecitsandhelpto
reestablishahomeostaticphenotypeinmicroglia[52].
Therefore,wedecidedtoevaluatetheinvolvementoftheNF‐κBsignalingpathway
inthebenecialeectsobservedwithT1AMinβamyloidinducedHMC3cells.Therefore,
werepeatedkeyAβ25–35andT1AMcotreatmentexperimentsandevaluatedNF‐κBac
tivity.Thecellswerethencollected,andphosphoNF‐κBP65(pP65)wasdetectedinthe
celllysates(Figure10).TheresultsshowedthatAβ25–35treatmentincreasedthephos
phorylationlevelofP65toactivatetheNF‐κBpathway,andT1AMsuppressedthisre
sponse(Figure10).Notably,inthepresenceofTA AR1selectiveantagonistEPPTB(1µM),
theeectofT1AMonNF‐κBactivationwascompletelyabolished(Figure10).

0
200
400
600
800
1000
TNF
[pg/ml]
A
(µM)
- 10 10 10 10
T1AM (µM)
- - 1 10 10
EPPTB (µM) - - - - 1
0.0037 0.0046
0.0041
0
200
400
600
800
IL-6 [pg/ml]
A
(µM)
- 10 10 10 10
T1AM (µM)
- - 1 10 10
EPPTB (µM) - - - - 1
0.0031 0.0042
0.0039
0
100
200
300
400
500
IL-10 [pg/ml]
A
(µM)
- 10 10 10 10
T1AM (µM)
- - 1 10 10
EPPTB (µM) - - - - 1
0.0022
0.0018
AB C
Figure 9.
T1AM pretreatment modulated the release of proinflammatory (TNF-
α
, IL-6) and anti-
inflammatory (IL-10) interleukins in A
β
-induced HMC3 cells. Bars represent the release (pg/mL)
of TNF–
α
(
A
), IL–6 (
B
), and IL–10 (
C
) in the presence of the drugs at the indicated concentrations.
Data represent means
±
S.E.M. from three independent experiments (n= 3), performed in duplicate.
Statistical analysis was performed by ordinary one-way ANOVA followed by Dunnett’s multiple
comparison test. For each experimental condition a selected specific pattern has been used.
We observed that T1AM’s protective effect against A
β
-induced microglia activa-
tion was abolished by co administration of TAAR1 selective antagonist EPPTB (1
µ
M)
(Figure 9A–C).
Dysregulation of the transcription factor NF-
κ
B has been widely associated with
AD, leading to glial cells activation and neuroinflammation [
51
]. More importantly, the
inactivation of microglial NF-
κ
B has been shown to restore cognitive deficits and help to
reestablish a homeostatic phenotype in microglia [52].
Therefore, we decided to evaluate the involvement of the NF-
κ
B signaling pathway in
the beneficial effects observed with T1AM in
β
-amyloid-induced HMC3 cells. Therefore,
we repeated key A
β
25–35 and T1AM cotreatment experiments and evaluated NF-
κ
B
activity. The cells were then collected, and phospho-NF-
κ
B P65 (p-P65) was detected
in the cell lysates (Figure 10). The results showed that A
β
25–35 treatment increased the
phosphorylation level of P65 to activate the NF-
κ
B pathway, and T1AM suppressed this
response (Figure 10). Notably, in the presence of TAAR1 selective antagonist EPPTB (1
µ
M),
the effect of T1AM on NF-κB activation was completely abolished (Figure 10).
Int. J. Mol. Sci. 2023,24, 11569 9 of 15
Int.J.Mol.Sci.2023,24,1156910of17
Figure10.TAAR 1activationmodulatedthephosphorylationofNF–kB(NF–kBp65)inAβ–induced
HMC3cells.Datarepresentmeans±S.E.M.fromthreeindependentexperiments(n=3),performed
induplicate.StatisticalanalysiswasperformedbyordinaryonewayANOVAfollowedbyDun
ne’smultiplecomparisontest.Foreachexperimentalconditionaselectedspecicpaernhasbeen
used.
3.Discussion
IncreasingevidencesupportstheconceptthatT1AM/TAAR1signalingispartofan
endogenoussystemthatcanbemodulatedtopreventneurodegeneration
[16,18,32,41,53,54].ADisthemostfrequentneurodegenerativedisorderintheelderly,
usuallycharacterizedbymemorydecitsandcognitivedecline.Cognitivefunctionisan
importantdeterminantofqualityoflife,especiallyintheelderly.Theprogressiveincrease
incirculatingproinammatorycytokinesininammaginghasbeenassociatedwithage
relatedcognitivedecline[55]aswellasenhancedneuroinammation,neurodegeneration,
andbrainreleaseofcytokinesbythemicroglia,whichactasresidentphagocyticinam
matorycellsinthebrain[56].Microglialcellsarevitalinrecruitingtheseinammatory
mediators,anddysregulatedmicroglialactivationrepresentsaneuropathologicalhall
markinADandmanyotherNDDs[57].MicroglialTA AR1signalinghasnotyetbeen
fullydened,butconsiderableoverlapexistsbetweentheimmuneactivatedstateand
TAAR1expression[29,58,59].PathologicalchangesinendogenousTAAR1agonists,asob
servedinpsychiatricdisorders,couldunderliealterationsinmicroglialfunctions[23,60].
Therefore,ascertainingtheabilityofT1AM,andendogenousagonistforTAAR 1willlead
toabeerunderstandingoftheroleofTAAR1asamodulatorofmicrogliadysregulation,
furtherexpandingthepotentialofdrugsthatinteractwithTAAR1 inthemanagementof
AD.
WeshowedherethatT1AMwasabletoreduceLPS/TNFα‐inducedIL6inHMC3
humanmicroglialcells,whileincreasingtheproductionofantiinammatoryinterleukin
IL10withoutaectingthecells’viability.Inaddition,inLPS/TNFα‐inducedHMC3cells,
treatmentwithT1AMwasobservedtodecreasetheexpressionofinammatoryresponse
relatedgenes,includingthemonocytechemoaractprotein1(MCP1),themacrophage
inammatoryprotein1(MIP1),andthetranscriptionfactorNFkB.
Regardingthemechanismofaction,T1AMdoesnotbindthenuclearthyroidhor
monereceptors(TRs)butitstimulateswithnanomolaranityTAAR1 ,aGproteincou
pledreceptorthatrecentlyemergedtohavearoleinimmunomodulation[53]andneu
roinammation[32].Nevertheless,thereiscurrentlynotmuchinformationregarding
TAAR1expressioninmicroglia[29].Inthepresentstudy,byperformingquantitativereal
0.0
0.5
1.0
1.5
2.0
phospho-NF-B p65
(fold increase vs control)
A (µM) - 10 10 10 10
T1AM (µM) - - 1 10 10
EPPTB (µM) - - - - 1
0.0029 0.017
0.0078
Figure 10.
TAAR1 activation modulated the phosphorylation of NF–kB (NF–kB p65) in A
β
–induced
HMC3 cells. Data represent means
±
S.E.M. from three independent experiments (n= 3), performed
in duplicate. Statistical analysis was performed by ordinary one-way ANOVA followed by Dunnett’s
multiple comparison test. For each experimental condition a selected specific pattern has been used.
3. Discussion
Increasing evidence supports the concept that T1AM/TAAR1 signaling is part of an en-
dogenous system that can be modulated to prevent neurodegeneration [
16
,
18
,
32
,
41
,
53
,
54
].
AD is the most frequent neurodegenerative disorder in the elderly, usually characterized
by memory deficits and cognitive decline. Cognitive function is an important determinant
of quality of life, especially in the elderly. The progressive increase in circulating proin-
flammatory cytokines in inflammaging has been associated with age-related cognitive
decline [
55
] as well as enhanced neuroinflammation, neurodegeneration, and brain release
of cytokines by the microglia, which act as resident phagocytic inflammatory cells in the
brain [
56
]. Microglial cells are vital in recruiting these inflammatory mediators, and dys-
regulated microglial activation represents a neuropathological hallmark in AD and many
other NDDs [
57
]. Microglial TAAR1 signaling has not yet been fully defined, but consider-
able overlap exists between the immune activated state and TAAR1 expression [
29
,
58
,
59
].
Pathological changes in endogenous TAAR1 agonists, as observed in psychiatric disorders,
could underlie alterations in microglial functions [
23
,
60
]. Therefore, ascertaining the ability
of T1AM, and endogenous agonist for TAAR1 will lead to a better understanding of the
role of TAAR1 as a modulator of microglia dysregulation, further expanding the potential
of drugs that interact with TAAR1 in the management of AD.
We showed here that T1AM was able to reduce LPS/TNF
α
-induced IL-6 in HMC3
human microglial cells, while increasing the production of anti-inflammatory interleukin
IL-10 without affecting the cells’ viability. In addition, in LPS/TNF
α
-induced HMC3 cells,
treatment with T1AM was observed to decrease the expression of inflammatory response-
related genes, including the monocyte chemoattract protein-1 (MCP1), the macrophage
inflammatory protein-1 (MIP1), and the transcription factor NF-kB.
Regarding the mechanism of action, T1AM does not bind the nuclear thyroid hormone
receptors (TRs) but it stimulates with nanomolar affinity TAAR1, a G protein-coupled
receptor that recently emerged to have a role in immunomodulation [
53
] and neuroin-
flammation [
32
]. Nevertheless, there is currently not much information regarding TAAR1
expression in microglia [
29
]. In the present study, by performing quantitative real-time
PCR analysis (qPCR), we demonstrated that TAAR1 is expressed in the human microglial
HMC3 cell line, and we found that after LPS/TNF
α
stimulation, no changing levels of
TAAR1 expression were observed in HMC3 cells.
Int. J. Mol. Sci. 2023,24, 11569 10 of 15
Using a pharmacological approach, we demonstrated that inhibition of TAAR1 was
sufficient to prevent T1AM’s anti-inflammatory effect on HMC3 cells, and, conversely, the
stimulation of TAAR1 in the absence of T1AM was able to attenuate the LPS/TNF
α
-induced
release of proinflammatory cytokines, as observed in T1AM-treated cells.
In this investigation, the pharmacokinetics results showed that T1AM was rapidly
taken up by HMC3 cells and catabolized by oxidative deamination to TA1, which was
largely released from cells.
Even though TA1 is not a ligand of TAAR1 [
61
], previous works have reported that
TA1 may contribute, at least in part, to some effects elicited after the administration of
exogenous T1AM [
41
,
43
45
,
62
]. Therefore, we checked whether TA1 administration could
also decrease the inflammatory phenotype of LPS/TNF
α
-stimulated HMC3 cells, but
obtained negative results, confirming the specific anti-inflammatory effect of T1AM.
A great number of studies have documented the ability of A
β
fibrils to directly stim-
ulate microglia
in vitro
to assume a neurotoxic phenotype characterized by secretion of
a plethora of proinflammatory molecules, ultimately leading to neuron loss [
63
,
64
]. We
found that aggregated A
β
25–35 peptide (10
µ
M, 24 h) significantly decreased viability
of HMC3 microglial cells. This was concentration-dependently attenuated by T1AM
(
0.1–10 µM
). Pretreatment with T1AM also suppressed the release of TNF
α
and proinflam-
matory IL-6 while increasing anti-inflammatory IL-10 levels. These effects were almost
completely abolished by treatment with the TAAR1 antagonist EPPTB. In addition, A
β
in-
duced activation of microglial NF-
κ
B by phosphorylation. Application of T1AM attenuated
this effect via TAAR1 activation, suggesting that the T1AM/TAAR1 system may protect
microglial cells against Aβ-induced cell injury by inhibition of inflammation.
In summary, we provided, for the first-time, preliminary evidence that the T1AM/TAAR1
signaling pathway has the potential to normalize factors that regulate microglia-mediated
neuroinflammation, further increasing the relevance of this system in the pathophysiology
of aging-related brain diseases like AD. In animal models, neuroinflammation has been
closely tied to impaired function of the hippocampus, associated with a loss of hippocampal
pyramidal neurons and entorhinal cells, leading to the disruption of long-term potentiation
(LTP) in hippocampal synapses [
65
,
66
]. Notably, the T1AM/TAAR1 system has been previ-
ously shown to restore LTP in the enthorinal cortex (EC) of mouse models of AD, suggesting
that T1AM and TAAR1 are part of an endogenous system that can be modulated to prevent
synaptic and behavioral deficits associated with A
β
-related toxicity [
41
]. The results of the
present study may help to elucidate the mechanisms by which the T1AM/TAAR1 system
exerts a neuroprotective effect, and the investigation of the relationship with neuroinflam-
mation markers could bring the story one step ahead. These preliminary data are strictly
necessary to provide the basis for the ongoing new
in vivo
study, which we are conducting
to clarify the contribution of neuroinflammation to altered hippocampal neuroplasticity, as
observed in mouse models of AD. Furthermore, the identification of cytokines specifically
associated with cognitive decline may provide novel biomarkers for the development of
interventions to slow or reverse AD.
4. Materials and Methods
4.1. Drugs
T1AM and the TAAR1 antagonist N-(3-ethoxyphenyl)-4-(pyrrolidin-1-yl)-3-trifluoromethyl-
benzamide (EPPTB) were purchased from Sigma-Aldrich (Milan, Italy); the TAAR1 agonist
(S)-4-[(ethylphenylamino) methyl]-4,5-dihydrooxazol-2-ylamine (RO5166017) was kindly
provided by Dr. Gainetdinov. T1AM metabolite 3-iodothyroacetic acid (TA1) was kindly
provided by Dr. Scanlan. Aliquots were stored at
20
C in DMSO as a 200 mM stock
solution and diluted to the desired final concentration in culture media.
4.2. Analysis of T1AM and TA1
T1AM and its metabolite 3-iodothyroacetic acid (TA1) were assayed in samples by
tandem mass spectrometry coupled to liquid chromatography (LC–MS/MS) by using
Int. J. Mol. Sci. 2023,24, 11569 11 of 15
a previously established procedure that allows the simultaneous detection of T1AM and
TA1 in each sample [
20
,
40
]. T1AM and TA1 intracellular concentrations were also assayed
by exposing cell lysate samples to the same LC–MS/MS procedure.
Briefly, aliquots (0.1 mL) from each sample collection were spiked with 10
µ
L of a suit-
able mixture of internal standards (deuterated T1AM and TA1). After adding methanol
(
0.4 mL
), the samples were shaken for 10 min and centrifuged at 22,780
×
gfor 10 min.
The supernatant was dried under a gentle stream of nitrogen, reconstituted with wa-
ter/methanol mixture (70/30 by volume), and injected into the LC–MS/MS system. The
latter included an Agilent 1290 UHPLC system (Santa Clara, CA, USA) coupled to an
AB-Sciex API 4000 triple quadrupole mass spectrometer (Concord, ON, Canada).
4.3. Cell Cultures and Reagents
The human microglial clone 3 cell line (HMC3) (ATCC
®
CRL-3304
,
Manassas, VA, USA
)
was cultured in high-glucose DMEM supplemented with 10% FBS, streptomycin (
100 g/mL
),
and penicillin (100 U/mL) (Sigma-Aldrich, Milan, Italy).
LPS (Escherichia coli 0111:B4), TNF
α
, and Amyloid
β
-Peptide 25–35 (A
β
25–35) were
purchased from Sigma-Aldrich (Milan, Italy).
4.4. MTT (Cell Viability Assay)
HMC3 cells were exposed to cell viability assays by using 3-(4,5-dimethylthiazol-2-
yl)-2,5-diphenyltetrazolium bromide (MTT, Sigma-Aldrich, Milan, Italy) reagent. Briefly,
HMC3 cells were exposed to increasing concentrations of T1AM (0.1–10
µ
M) and incubated
at 37
C for 24 h. Then, 0.5 mg/mL MTT reagent was added to each well, and the cells
were incubated for 3 h at 37
C. Next, 25
µ
L of the medium was removed from the wells,
and 50
µ
L of DMSO was added. After incubating for 10 min at 37
C, absorbance at 540 nm
was determined with an automated microplate reader (BIO-TEK, Winooski, VT, USA). The
percentage of cell viability was calculated as a percentage of vehicle-treated cells used as
control. The same procedure was also followed to detect the cytotoxic effect produced in
HMC3 cells after incubation with A
β
25–35 (1 and 10
µ
M, for 24 or 48 h) in the absence and
presence of increasing concentrations of T1AM (0.1, 1, and 10 µM).
4.5. Release of Inflammatory Molecules HMC3 Cells Treated with LPS/TNFα
Proinflammatory IL-6 and anti-inflammatory IL-10 levels were evaluated by specific
ELISA assays (RAB0306 (IL-6) and RAB0244 (IL-10), Sigma-Aldrich, Milan, Italy) on col-
lected culture media. Briefly, HMC3 cells were exposed to pretreatment with each of the
compounds under investigation (i.e., T1AM, TA1, or RO51660170) for 1 h followed by
treatment for 24 h with both LPS (10
µ
g/mL) and TNF
α
(50 ng/mL), generally indicated as
LPS/TNF
α
, and used as proinflammatory stimuli. Vehicle-treated cells were used as con-
trol. In competition experiments, the TAAR1 antagonist EPPTB (1
µ
M) was administered
15 min before proceeding with the administration of the selected TAAR1 agonist, namely,
T1AM or RO5166017.
4.6. Release of Inflammatory Molecules from Aβ25–35-Induced HMC3 Cells
The inflammatory response of HMC3 cells to A
β
25–35 (Sigma-Aldrich, Milan, Italy) ex-
posure (10
µ
M, 24 h) was evaluated by performing specific ELISA assays. In addition to IL-6
and IL-10 measurements, performed as described above, the levels of tumor necrosis factor
TNF
α
(RAB1089, Sigma-Aldrich, Milan, Italy) and nuclear factor kB (NF-kB) (
85-86082-11
,
ThermoFisher Scientific, Carlsbad, CA, USA) were also evaluated in collected culture media
and in cell lysates, respectively, following the corresponding manufacturer’s instructions.
Briefly, HMC3 cells were exposed to pretreatment with T1AM (1 or 10
µ
M) for 1 h followed
by A
β
25–35 (10
µ
M for 24 h), used as proinflammatory stimuli. Vehicle-treated cells were
used as control. In competition experiments, the TAAR1 antagonist EPPTB (
1µM
) was
administered 15 min before proceeding with the administration of T1AM (10 µM).
Int. J. Mol. Sci. 2023,24, 11569 12 of 15
4.7. Gene Expression Analysis
Total RNA was extracted from HMC3 cells using the RNeasy Mini kit (74104, Qiagen,
Hilden, Germany) following the manufacturer’s protocol. RNA concentration and purity
were determined by Nanodrop-1000 spectrophotometer and Qubit v.1 fluorometer plus
Qubit RNA HS Assay Kit (Thermo Fisher Scientific, Wilmington, DE, USA).
Total RNA (1
µ
g) was retrotranscribed into first-strand cDNA by using by iScriptTM
gDNA Clear cDNA Synthesis Kit (Bio-Rad, Milan, Italy) following manual protocol indications.
Relative quantity of gene transcripts was measured by real-time PCR on samples’
cDNA using an SYBRGreen chemistry and an CFX Connect Real-Time PCR Detection Sys-
tem (Bio-Rad, Milan, Italy). The PCR cycle program consisted of an initial 30 s denaturation
at 95
C followed by 40 cycles of 5 s denaturation at 95
C and 15 sec annealing/extension
at 60
C. A final melting protocol with ramping from 65
C to 95
C with 0.5
C increments
of 5 s was performed for verification of amplicon specificity and primer dimer formation.
Primers were designed with Beacon Designer Software v.8.0 (Premier Biosoft Inter-
national, Palo Alto, CA, USA) with a junction primer strategy whenever possible. In any
case, negative control of retrotranscription was performed to exclude any interference
from residual genomic DNA contamination. The primer sequences for real-time PCR are
reported in Table 2.
Table 2. Primer sequences of target genes.
Reference Sequence
(RefSeq) RNA Gene Symbol Primer Sequences
NM_002046 GAPDH
(F) 5
0
-CCCTTCATTGACCTCAACTACATG
(R) 50-TGGGATTTCCATTGATGACAAGC
NM_002982.4 MCP1 (F) 50-GAGAGGCTGAGACTAACC
(R) 50-TGATTGCATCTGGCTGAG
NM_002983 MIP1 (F) 50-ACTTTGAGACGAGCAGCCAGTG
(R) 50-TTTCTGGACCCACTCCTCACTG
NM_001404662 NFKB (F) 50-CCTTTCTCATCCCATCTTT
(R) 50-CCTCAATGTCCTCTTTCTG
NM_138327 TAAR1 (F) 50-GAGATCTGCTGAGCACTGTTGG
(R) 5
0
-CAGCATAGTAGCGGTCAATGGAG
All reactions were performed in triplicate and the amount of mRNA was calculated by
the comparative critical threshold (CT) method. To account for possible variations related
to cDNA input or the presence of PCR inhibitors, the endogenous reference gene GAPDH
was simultaneously quantified for each sample, and data were normalized accordingly.
4.8. Statistical Analysis
Statistical analyses were performed using GraphPad Prism version 9.0 for Mac (Graph-
Pad Software, San Diego, CA, USA). Data were subjected to t-tests or one-way ANOVA.
Significant differences among different treatments were calculated according to Dunnett’s
multiple comparisons test. Data are reported as mean
±
SEM. Differences at p< 0.05 were
considered statistically significant.
Author Contributions:
B.P. and G.C. designed the study and wrote the manuscript. C.R. and B.P.
performed ELISA tests and collected biochemical data. B.P. and V.C. conducted gene expression
studies. A.B. and C.R. carried out LC/MS-MS data collection. G.R., F.S. and R.Z. analyzed the data and
discussed the results. All authors have read and agreed to the published version of the manuscript.
Funding:
This work was supported by a grant Pfizer (Project ID: 67562227; G.C.) and a grant from
ETA (Project ID: 549901 to G.R.).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Data is contained within the article.
Int. J. Mol. Sci. 2023,24, 11569 13 of 15
Acknowledgments:
The authors would like to thank the CISUP Centre for Instrumentation Sharing,
University of Pisa for having placed the Sciex QTrap 6500+ mass spectrometer, which was used for
the T1AM assays, at the disposal of the authors.
Conflicts of Interest:
The authors declare no conflict of interest. The authors declare no competing
financial interest.
References
1.
Li, Q.; Barres, B.A. Microglia and Macrophages in Brain Homeostasis and Disease. Nat. Rev. Immunol.
2018
,18, 225–242.
[CrossRef]
2.
Mecha, M.; Feliú, A.; Carrillo-Salinas, F.J.; Rueda-Zubiaurre, A.; Ortega-Gutiérrez, S.; de Sola, R.G.; Guaza, C. Endocannabinoids
Drive the Acquisition of an Alternative Phenotype in Microglia. Brain Behav. Immun. 2015,49, 233–245. [CrossRef]
3. Reith, W. Neurodegenerative Erkrankungen. Radiologe 2018,58, 241–258. [CrossRef]
4.
Xu, L.; He, D.; Bai, Y. Microglia-Mediated Inflammation and Neurodegenerative Disease. Mol. Neurobiol.
2016
,53, 6709–6715.
[CrossRef]
5.
Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-Mediated Neuroinflammation in Neurodegenerative Diseases.
Semin. Cell Dev. Biol. 2019,94, 112–120. [CrossRef]
6.
Chen, W.-W.; Zhang, X.; Huang, W.-J. Role of Neuroinflammation in Neurodegenerative Diseases (Review). Mol. Med. Rep.
2016
,
13, 3391–3396. [CrossRef]
7.
Leng, F.; Edison, P. Neuroinflammation and Microglial Activation in Alzheimer Disease: Where Do We Go from Here? Nat. Rev.
Neurol. 2021,17, 157–172. [CrossRef]
8.
Jucker, M.; Walker, L.C. Self-Propagation of Pathogenic Protein Aggregates in Neurodegenerative Diseases. Nature
2013
,
501, 45–51. [CrossRef]
9. Jellinger, K.A. Basic Mechanisms of Neurodegeneration: A Critical Update. J. Cell. Mol. Med. 2010,14, 457–487. [CrossRef]
10.
Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms Underlying Inflammation in Neurodegeneration. Cell
2010,140, 918–934. [CrossRef]
11.
Mosser, D.M.; Edwards, J.P. Exploring the Full Spectrum of Macrophage Activation. Nat. Rev. Immunol.
2008
,8, 958–969.
[CrossRef]
12.
Thameem Dheen, S.; Kaur, C.; Ling, E.-A. Microglial Activation and Its Implications in the Brain Diseases. CMC
2007
,
14, 1189–1197. [CrossRef]
13. Lull, M.E.; Block, M.L. Microglial Activation and Chronic Neurodegeneration. Neurotherapeutics 2010,7, 354–365. [CrossRef]
14.
Scanlan, T.S.; Suchland, K.L.; Hart, M.E.; Chiellini, G.; Huang, Y.; Kruzich, P.J.; Frascarelli, S.; Crossley, D.A.; Bunzow, J.R.;
Ronca-Testoni, S.; et al. 3-Iodothyronamine Is an Endogenous and Rapid-Acting Derivative of Thyroid Hormone. Nat. Med.
2004
,
10, 638–642. [CrossRef]
15.
Barak, L.S.; Salahpour, A.; Zhang, X.; Masri, B.; Sotnikova, T.D.; Ramsey, A.J.; Violin, J.D.; Lefkowitz, R.J.; Caron, M.G.;
Gainetdinov, R.R. Pharmacological Characterization of Membrane-Expressed Human Trace Amine-Associated Receptor 1
(TAAR1) by a Bioluminescence Resonance Energy Transfer CAMP Biosensor. Mol. Pharmacol. 2008,74, 585–594. [CrossRef]
16.
Bellusci, L.; Laurino, A.; Sabatini, M.; Sestito, S.; Lenzi, P.; Raimondi, L.; Rapposelli, S.; Biagioni, F.; Fornai, F.; Salvetti, A.; et al.
New Insights into the Potential Roles of 3-Iodothyronamine (T1AM) and Newly Developed Thyronamine-Like TAAR1 Agonists
in Neuroprotection. Front. Pharmacol. 2017,8, 905. [CrossRef]
17.
Bellusci, L.; Runfola, M.; Carnicelli, V.; Sestito, S.; Fulceri, F.; Santucci, F.; Lenzi, P.; Fornai, F.; Rapposelli, S.; Origlia, N.; et al.
Endogenous 3-Iodothyronamine (T1AM) and Synthetic Thyronamine-Like Analog SG-2 Act as Novel Pleiotropic Neuroprotective
Agents through the Modulation of SIRT6. Molecules 2020,25, 1054. [CrossRef]
18.
Tozzi, F.; Rutigliano, G.; Borsò, M.; Falcicchia, C.; Zucchi, R.; Origlia, N. T1AM-TAAR1 Signalling Protects against OGD-Induced
Synaptic Dysfunction in the Entorhinal Cortex. Neurobiol. Dis. 2021,151, 105271. [CrossRef]
19.
Landucci, E.; Gencarelli, M.; Mazzantini, C.; Laurino, A.; Pellegrini-Giampietro, D.E.; Raimondi, L. N-(3-Ethoxy-Phenyl)-4-
Pyrrolidin-1-Yl-3-Trifluoromethyl-Benzamide (EPPTB) Prevents 3-Iodothyronamine (T1AM)-Induced Neuroprotection against
Kainic Acid Toxicity. Neurochem. Int. 2019,129, 104460. [CrossRef]
20.
Di Leo, N.; Moscato, S.; Borso’, M.; Sestito, S.; Polini, B.; Bandini, L.; Grillone, A.; Battaglini, M.; Saba, A.; Mattii, L.; et al. Delivery
of Thyronamines (TAMs) to the Brain: A Preliminary Study. Molecules 2021,26, 1616. [CrossRef]
21.
Xie, Z.; Miller, G.M. Trace Amine-Associated Receptor 1 as a Monoaminergic Modulator in Brain. Biochem. Pharmacol.
2009
,
78, 1095–1104. [CrossRef]
22.
Lindemann, L.; Meyer, C.A.; Jeanneau, K.; Bradaia, A.; Ozmen, L.; Bluethmann, H.; Bettler, B.; Wettstein, J.G.; Borroni, E.;
Moreau, J.-L.; et al.
Trace Amine-Associated Receptor 1 Modulates Dopaminergic Activity. J. Pharmacol. Exp. Ther.
2008
,324,
948–956. [CrossRef]
23.
Revel, F.G.; Moreau, J.-L.; Gainetdinov, R.R.; Bradaia, A.; Sotnikova, T.D.; Mory, R.; Durkin, S.; Zbinden, K.G.; Norcross, R.;
Meyer, C.A.; et al. TAAR1 Activation Modulates Monoaminergic Neurotransmission, Preventing Hyperdopaminergic and
Hypoglutamatergic Activity. Proc. Natl. Acad. Sci. USA 2011,108, 8485–8490. [CrossRef]
Int. J. Mol. Sci. 2023,24, 11569 14 of 15
24.
Al-Sabri, M.H.; Nikpour, M.; Clemensson, L.E.; Attwood, M.M.; Williams, M.J.; Rask-Anderson, M.; Mwinyi, J.; Schiöth, H.B. The
Regulatory Role of AP-2
β
in Monoaminergic Neurotransmitter Systems: Insights on Its Signalling Pathway, Linked Disorders
and Theragnostic Potential. Cell Biosci. 2022,12, 151. [CrossRef]
25.
Schwartz, M.D.; Canales, J.J.; Zucchi, R.; Espinoza, S.; Sukhanov, I.; Gainetdinov, R.R. Trace Amine-Associated Receptor 1:
A Multimodal Therapeutic Target for Neuropsychiatric Diseases. Expert Opin. Ther. Targets 2018,22, 513–526. [CrossRef]
26.
Koblan, K.S.; Kent, J.; Hopkins, S.C.; Krystal, J.H.; Cheng, H.; Goldman, R.; Loebel, A. A Non–D2-Receptor-Binding Drug for the
Treatment of Schizophrenia. N. Engl. J. Med. 2020,382, 1497–1506. [CrossRef]
27.
Nair, P.C.; Chalker, J.M.; McKinnon, R.A.; Langmead, C.J.; Gregory, K.J.; Bastiampillai, T. Trace Amine-Associated Receptor 1
(TAAR1): Molecular and Clinical Insights for the Treatment of Schizophrenia and Related Comorbidities. ACS Pharmacol. Transl.
Sci. 2022,5, 183–188. [CrossRef]
28.
Murtazina, R.Z.; Kuvarzin, S.R.; Gainetdinov, R.R. TAARs and Neurodegenerative and Psychiatric Disorders. In Handbook of Neu-
rotoxicity; Kostrzewa, R.M., Ed.; Springer International Publishing: Cham, Switzerland, 2021; pp. 1–18. ISBN 978-3-030-71519-9.
29.
Fleischer, L.M.; Somaiya, R.D.; Miller, G.M. Review and Meta-Analyses of TAAR1 Expression in the Immune System and Cancers.
Front. Pharmacol. 2018,9, 683. [CrossRef]
30.
Cisneros, I.E.; Ghorpade, A. Methamphetamine and HIV-1-Induced Neurotoxicity: Role of Trace Amine Associated Receptor 1
CAMP Signaling in Astrocytes. Neuropharmacology 2014,85, 499–507. [CrossRef]
31.
Dave, S.; Chen, L.; Yu, C.; Seaton, M.; Khodr, C.E.; Al-Harthi, L.; Hu, X.-T. Methamphetamine Decreases K
+
Channel Function in
Human Fetal Astrocytes by Activating the Trace Amine-Associated Receptor Type-1. J. Neurochem. 2019,148, 29–45. [CrossRef]
32.
Barnes, D.A.; Galloway, D.A.; Hoener, M.C.; Berry, M.D.; Moore, C.S. TAAR1 Expression in Human Macrophages and Brain
Tissue: A Potential Novel Facet of MS Neuroinflammation. Int. J. Mol. Sci. 2021,22, 11576. [CrossRef]
33.
Dello Russo, C.; Cappoli, N.; Coletta, I.; Mezzogori, D.; Paciello, F.; Pozzoli, G.; Navarra, P.; Battaglia, A. The Human Microglial
HMC3 Cell Line: Where Do We Stand? A Systematic Literature Review. J. Neuroinflamm. 2018,15, 259. [CrossRef]
34. Merighi, S.; Nigro, M.; Travagli, A.; Gessi, S. Microglia and Alzheimer ’s Disease. Int. J. Mol. Sci. 2022,23, 12990. [CrossRef]
35.
Gado, F.; Ferrisi, R.; Polini, B.; Mohamed, K.A.; Ricardi, C.; Lucarini, E.; Carpi, S.; Domenichini, F.; Stevenson, L.A.;
Rapposelli, S.; et al.
Design, Synthesis, and Biological Activity of New CB2 Receptor Ligands: From Orthosteric and Allosteric
Modulators to Dualsteric/Bitopic Ligands. J. Med. Chem. 2022,65, 9918–9938. [CrossRef]
36.
Ferrisi, R.; Gado, F.; Polini, B.; Ricardi, C.; Mohamed, K.A.; Stevenson, L.A.; Ortore, G.; Rapposelli, S.; Saccomanni, G.;
Pertwee, R.G.; et al.
Design, Synthesis and Biological Evaluation of Novel Orthosteric-Allosteric Ligands of the Cannabinoid
Receptor Type 2 (CB2R). Front. Chem. 2022,10, 984069. [CrossRef]
37.
Ferrisi, R.; Polini, B.; Ricardi, C.; Gado, F.; Mohamed, K.A.; Baron, G.; Faiella, S.; Poli, G.; Rapposelli, S.; Saccomanni, G.; et al. New
Insights into Bitopic Orthosteric/Allosteric Ligands of Cannabinoid Receptor Type 2. Int. J. Mol. Sci. 2023,24, 2135. [CrossRef]
38.
Ghelardoni, S.; Chiellini, G.; Frascarelli, S.; Saba, A.; Zucchi, R. Uptake and Metabolic Effects of 3-Iodothyronamine in Hepatocytes.
J. Endocrinol. 2014,221, 101–110. [CrossRef]
39.
Assadi-Porter, F.; Reiland, H.; Sabatini, M.; Lorenzini, L.; Carnicelli, V.; Rogowski, M.; Selen Alpergin, E.; Tonelli, M.; Ghelardoni,
S.; Saba, A.; et al. Metabolic Reprogramming by 3-Iodothyronamine (T1AM): A New Perspective to Reverse Obesity through
Co-Regulation of Sirtuin 4 and 6 Expression. Int. J. Mol. Sci. 2018,19, 1535. [CrossRef]
40.
Saba, A.; Chiellini, G.; Frascarelli, S.; Marchini, M.; Ghelardoni, S.; Raffaelli, A.; Tonacchera, M.; Vitti, P.; Scanlan, T.S.; Zucchi, R.
Tissue Distribution and Cardiac Metabolism of 3-Iodothyronamine. Endocrinology 2010,151, 5063–5073. [CrossRef]
41.
Accorroni, A.; Rutigliano, G.; Sabatini, M.; Frascarelli, S.; Borsò, M.; Novelli, E.; Bandini, L.; Ghelardoni, S.; Saba, A.;
Zucchi, R.; et al
. Exogenous 3-Iodothyronamine Rescues the Entorhinal Cortex from
β
-Amyloid Toxicity. Thyroid
2020
,30,
147–160. [CrossRef]
42.
Siemian, J.N.; Zhang, Y.; Li, J.-X. Trace Amine-Associated Receptor 1 Agonists RO5263397 and RO5166017 Attenuate Quinpirole-
Induced Yawning but Not Hypothermia in Rats. Behav. Pharmacol. 2017,28, 590–593. [CrossRef]
43.
Laurino, A.; Landucci, E.; Raimondi, L. Central Effects of 3-Iodothyronamine Reveal a Novel Role for Mitochondrial Monoamine
Oxidases. Front. Endocrinol. 2018,9, 290. [CrossRef]
44.
Laurino, A.; De Siena, G.; Saba, A.; Chiellini, G.; Landucci, E.; Zucchi, R.; Raimondi, L. In the Brain of Mice, 3-Iodothyronamine
(T1AM) Is Converted into 3-Iodothyroacetic Acid (TA1) and It Is Included within the Signaling Network Connecting Thyroid
Hormone Metabolites with Histamine. Eur. J. Pharmacol. 2015,761, 130–134. [CrossRef]
45.
Musilli, C.; De Siena, G.; Manni, M.E.; Logli, A.; Landucci, E.; Zucchi, R.; Saba, A.; Donzelli, R.; Passani, M.B.;
Provensi, G.; et al.
Histamine Mediates Behavioural and Metabolic Effects of 3-Iodothyroacetic Acid, an Endogenous End Product of Thyroid
Hormone Metabolism: A Novel Link between Thyroid and Histamine. Br. J. Pharmacol. 2014,171, 3476–3484. [CrossRef]
46.
Shi, S.; Liang, D.; Chen, Y.; Xie, Y.; Wang, Y.; Wang, L.; Wang, Z.; Qiao, Z. Gx-50 Reduces
β
-Amyloid-Induced TNF-
α
, IL-1
β
, NO,
and PGE
2
Expression and Inhibits NF-KB Signaling in a Mouse Model of Alzheimer’s Disease. Eur. J. Immunol.
2016
,46, 665–676.
[CrossRef]
47.
Cisbani, G.; Rivest, S. Targeting Innate Immunity to Protect and Cure Alzheimer’s Disease: Opportunities and Pitfalls. Mol.
Psychiatry 2021,26, 5504–5515. [CrossRef]
48.
Liu, Y.-Y.; Bian, J.-S. Hydrogen Sulfide Protects Amyloid-
β
Induced Cell Toxicity in Microglia. JAD
2011
,22, 1189–1200. [CrossRef]
Int. J. Mol. Sci. 2023,24, 11569 15 of 15
49.
Ranaivo, H.R.; Craft, J.M.; Hu, W.; Guo, L.; Wing, L.K.; Van Eldik, L.J.; Watterson, D.M. Glia as a Therapeutic Target: Se-
lective Suppression of Human Amyloid-
β
-Induced Upregulation of Brain Proinflammatory Cytokine Production Attenuates
Neurodegeneration. J. Neurosci. 2006,26, 662–670. [CrossRef]
50.
Naldi, M.; Fiori, J.; Pistolozzi, M.; Drake, A.F.; Bertucci, C.; Wu, R.; Mlynarczyk, K.; Filipek, S.; De Simone, A.; Andrisano, V.
Amyloid
β
-Peptide 25–35 Self-Assembly and Its Inhibition: A Model Undecapeptide System to Gain Atomistic and Secondary
Structure Details of the Alzheimer’s Disease Process and Treatment. ACS Chem. Neurosci. 2012,3, 952–962. [CrossRef]
51.
Mattson, M.P.; Camandola, S. NF-KB in Neuronal Plasticity and Neurodegenerative Disorders. J. Clin. Investig.
2001
,107, 247–254.
[CrossRef]
52.
Wang, C.; Fan, L.; Khawaja, R.R.; Liu, B.; Zhan, L.; Kodama, L.; Chin, M.; Li, Y.; Le, D.; Zhou, Y.; et al. Microglial NF-KB Drives
Tau Spreading and Toxicity in a Mouse Model of Tauopathy. Nat. Commun. 2022,13, 1969. [CrossRef]
53.
Christian, S.L.; Berry, M.D. Trace Amine-Associated Receptors as Novel Therapeutic Targets for Immunomodulatory Disorders.
Front. Pharmacol. 2018,9, 680. [CrossRef]
54.
Rutigliano, G.; Accorroni, A.; Zucchi, R. The Case for TAAR1 as a Modulator of Central Nervous System Function. Front.
Pharmacol. 2018,8, 987. [CrossRef]
55.
Atienza, M.; Ziontz, J.; Cantero, J.L. Low-Grade Inflammation in the Relationship between Sleep Disruption, Dysfunctional
Adiposity, and Cognitive Decline in Aging. Sleep Med. Rev. 2018,42, 171–183. [CrossRef]
56.
Sama, D.M.; Norris, C.M. Calcium Dysregulation and Neuroinflammation: Discrete and Integrated Mechanisms for Age-Related
Synaptic Dysfunction. Ageing Res. Rev. 2013,12, 982–995. [CrossRef]
57. Akiyama, H. Inflammation and Alzheimer’s Disease. Neurobiol. Aging 2000,21, 383–421. [CrossRef]
58.
Liang, J.; Takeuchi, H.; Doi, Y.; Kawanokuchi, J.; Sonobe, Y.; Jin, S.; Yawata, I.; Li, H.; Yasuoka, S.; Mizuno, T.; et al. Excita-
tory Amino Acid Transporter Expression by Astrocytes Is Neuroprotective against Microglial Excitotoxicity. Brain Res.
2008
,
1210, 11–19. [CrossRef]
59.
Pubill, D.; Canudas, A.M.; Pallàs, M.; Camins, A.; Camarasa, J.; Escubedo, E. Different Glial Response to Methamphetamine- and
Methylenedioxymethamphetamine-Induced Neurotoxicity. Naunyn Schmiedebergs Arch. Pharmacol.
2003
,367, 490–499. [CrossRef]
60.
Tremblay, M.-È. Microglial Functional Alteration and Increased Diversity in the Challenged Brain: Insights into Novel Targets for
Intervention. Brain Behav. Immun. Health 2021,16, 100301. [CrossRef]
61.
Chiellini, G.; Nesi, G.; Digiacomo, M.; Malvasi, R.; Espinoza, S.; Sabatini, M.; Frascarelli, S.; Laurino, A.; Cichero, E.;
Macchia, M.; et al.
Design, Synthesis, and Evaluation of Thyronamine Analogues as Novel Potent Mouse Trace Amine As-
sociated Receptor 1 (mTAAR1) Agonists. J. Med. Chem. 2015,58, 5096–5107. [CrossRef]
62.
Laurino, A.; Gencarelli, M.; Raimondi, L. The 3-Iodothyronamine (T1AM) and the 3-Iodothyroacetic Acid (TA1) Indicate a Novel
Connection with the Histamine System for Neuroprotection. Eur. J. Pharmacol. 2021,912, 174606. [CrossRef] [PubMed]
63.
Akiyama, H.; Arai, T.; Kondo, H.; Tanno, E.; Haga, C.; Ikeda, K. Cell Mediators of Inflammation in the Alzheimer Disease Brain.
Alzheimer Dis. Assoc. Disord. 2000,14, S47–S53. [CrossRef] [PubMed]
64.
Gabrielli, M.; Prada, I.; Joshi, P.; Falcicchia, C.; D’Arrigo, G.; Rutigliano, G.; Battocchio, E.; Zenatelli, R.; Tozzi, F.;
Radeghieri, A.; et al.
Microglial Large Extracellular Vesicles Propagate Early Synaptic Dysfunction in Alzheimer’s Disease. Brain 2022,145, 2849–2868.
[CrossRef]
65.
Hauss-Wegrzyniak, B.; Lynch, M.A.; Vraniak, P.D.; Wenk, G.L. Chronic Brain Inflammation Results in Cell Loss in the Entorhinal
Cortex and Impaired LTP in Perforant Path-Granule Cell Synapses. Exp. Neurol. 2002,176, 336–341. [CrossRef] [PubMed]
66.
Kelly, Á.; Vereker, E.; Nolan, Y.; Brady, M.; Barry, C.; Loscher, C.E.; Mills, K.H.G.; Lynch, M.A. Activation of P38 Plays a Pivotal
Role in the Inhibitory Effect of Lipopolysaccharide and Interleukin-1
β
on Long Term Potentiation in Rat Dentate Gyrus. J. Biol.
Chem. 2003,278, 19453–19462. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... To explore in more detail the potential of newly developed CB2R bitopic ligand FD22a to target neurodegeneration, in the present study we investigated the ability of FD22a to counteract the detrimental effects produced by the neurotoxic Aβ fragment 25-35 (Aβ [25][26][27][28][29][30][31][32][33][34][35] [30] in human cellular models of neurodegeneration, such as human microglial (HMC3) and human glioblastoma-astrocytoma (U87-MG) cell lines. ...
... In these models FD22a prevented the cytotoxic and proinflammatory effects of Aβ 25-35 and efficiently counteracted the depression of autophagy caused by Aβ [25][26][27][28][29][30][31][32][33][34][35] . ...
... Aβ [25][26][27][28][29][30][31][32][33][34][35] peptide (NH 2 -Gly-Ser-Asn-Lys-Gly-Ala-Ile-Ile-Gly-Leu-Met-COOH) (A4559, Sigma-Aldrich, Milan, Italy) was initially dissolved in double-distilled water to obtain 1 mM concentration and stored at −20 • C. To form aggregated diffusible oligomers, the solution was incubated at 37 • C for 5 days [31], then diluted in medium to the indicated concentration, just prior to cell treatments. FD22a was dissolved in DMSO to obtain a 50 mM stock solution which was kept at 4 • C. Before the experiments FD22a stock solution was diluted into the cell culture medium to the desired experimental concentration, and the final DMSO concentration was maintained no higher than 0.1%. ...
Article
Full-text available
Neurodegenerative diseases (NDDs) are progressive multifactorial disorders of the nervous system sharing common pathogenic features, including intracellular misfolded protein aggregation, mitochondrial deficit, and inflammation. Taking into consideration the multifaceted nature of NDDs, development of multitarget-directed ligands (MTDLs) has evolved as an attractive therapeutic strategy. Compounds that target the cannabinoid receptor type II (CB2R) are rapidly emerging as novel effective MTDLs against common NDDs, such as Alzheimer’s disease (AD). We recently developed the first CB2R bitopic/dualsteric ligand, namely FD22a, which revealed the ability to induce neuroprotection with fewer side effects. To explore the potential of FD22a as a multitarget drug for the treatment of NDDs, we investigated here its ability to prevent the toxic effect of β-amyloid (Aβ25–35 peptide) on human cellular models of neurodegeneration, such as microglia (HMC3) and glioblastoma (U87-MG) cell lines. Our results displayed that FD22a efficiently prevented Aβ25–35 cytotoxic and proinflammatory effects in both cell lines and counteracted β-amyloid-induced depression of autophagy in U87-MG cells. Notably, a quantitative proteomic analysis of U87-MG cells revealed that FD22a was able to potently stimulate the autophagy–lysosomal pathway (ALP) by activating its master transcriptional regulator TFEB, ultimately increasing the potential of this novel CB2R bitopic/dualsteric ligand as a multitarget drug for the treatment of NDDs.
... Since cell death and inflammation along the olfactory pathway were detected, we examined the mRNA levels of several molecules, including the olfactory receptor 558 (Olfr558), Taar1 [40,42], and formyl peptide receptor 2 (Fpr2) [47], which are either involved in the above-mentioned cellular events or olfaction. The results indicated that, among these molecules, the levels of Taar1 were significantly diminished in several olfaction-related regions, including the OB, the cortex, and the hypothalamus in PD mice ( Figure 4A-C). ...
... (2) Regulating the activity of astrocytes: Activation of TAAR1 in astrocytes significantly downregulates the level of the glutamate transporter EAAT-2, leading to an accumulation of glutamate and subsequent cytotoxicity [39,64] due to the corresponding influx of Ca 2+ into the cells to trigger apoptotic signals and the release of IL-1β, resulting in apoptosis and neuroinflammation [48]. The following two possible mechanisms are speculated according to previous reports and require further investigation: (3) Regulating the activity of microglia: Activation of TAAR1 in microglia can suppress the release of proinflammatory factors such as IL-6, TNF-α, NF-κB, MCP1, and MIP1, while simultaneously promoting the release of anti-inflammatory mediators like IL-10, thus fulfilling an anti-inflammatory role [40]. (4) Regulating dopaminergic neurons: As previously reported, dopaminergic neuronal damage in the OB could result in olfactory dysfunction [46], which indicates that dopaminergic neurons might also participate in olfaction. ...
Article
Full-text available
Parkinson’s disease (PD) is characterized not only by motor symptoms but also by non-motor dysfunctions, such as olfactory impairment; the cause is not fully understood. Our study suggests that neuronal loss and inflammation in brain regions along the olfactory pathway, such as the olfactory bulb (OB) and the piriform cortex (PC), may contribute to olfactory dysfunction in PD mice, which might be related to the downregulation of the trace amine-associated receptor 1 (TAAR1) in these areas. In the striatum, although only a decrease in mRNA level, but not in protein level, of TAAR1 was detected, bioinformatic analyses substantiated its correlation with PD. Moreover, we discovered that neuronal death and inflammation in the OB and the PC in PD mice might be regulated by TAAR through the Bcl-2/caspase3 pathway. This manifested as a decrease of anti-apoptotic protein Bcl-2 and an increase of the pro-apoptotic protein cleaved caspase3, or through regulating astrocytes activity, manifested as the increase of TAAR1 in astrocytes, which might lead to the decreased clearance of glutamate and consequent neurotoxicity. In summary, we have identified a possible mechanism to elucidate the olfactory dysfunction in PD, positing neuronal damage and inflammation due to apoptosis and astrocyte activity along the olfactory pathway in conjunction with the downregulation of TAAR1.
... It has been observed to modulate sleep [36], exploratory activity, memory acquisition and retention [24]. T 1 AM showed neuroprotective actions in models of seizure-related excitotoxic damage, altered autophagy, amyloidosis, ischemia-reperfusion injury, and neuroinflammation [25][26][27][28]37]. In these studies, T 1 AM was administered either through perfusion on ex vivo brain acute/organotypic slices or through intracerebral microinjection, while we used a systemic administration. ...
... However, we hypothesise a role for TAAR1 due to growing evidence of its involvement in neuropsychiatric disorders [43,44]. Furthermore, we have previously demonstrated that T1AM neuroprotective actions in models of amyloidosis, ischemia-reperfusion injury, and neuroinflammation are dependent on TAAR1 [27,28,37]. TAAR1 was reported to activate distinct signalling pathways depending on its localization in different cellular compartments. ...
Article
Full-text available
Mood alterations, anxiety, and cognitive impairments associated with adult-onset hypothyroidism often persist despite replacement treatment. In rodent models of hypothyroidism, replacement does not bring 3-iodothyronamine (T1AM) brain levels back to normal. T1AM is a thyroid hormone derivative with cognitive effects. Using a pharmacological hypothyroid mouse model, we investigated whether augmenting levothyroxine (L-T4) with T1AM improves behavioural correlates of depression, anxiety, and memory and has an effect on hippocampal neurogenesis. Hypothyroid mice showed impaired performance in the novel object recognition test as compared to euthyroid mice (discrimination index (DI): 0.02 ± 0.09 vs. 0.29 ± 0.06; t = 2.515, p = 0.02). L-T4 and L-T4+T1AM rescued memory (DI: 0.27 ± 0.08 and 0.34 ± 0.08, respectively), while T1AM had no effect (DI: −0.01 ± 0.10). Hypothyroidism reduced the number of neuroprogenitors in hippocampal neurogenic niches by 20%. L-T4 rescued the number of neuroprogenitors (mean diff = 106.9 ± 21.40, t = 4.99, pcorr = 0.003), while L-T4+T1AM produced a 30.61% rebound relative to euthyroid state (mean diff = 141.6 ± 31.91, t = 4.44, pcorr = 0.004). We performed qPCR analysis of 88 genes involved in neurotrophic signalling pathways and found an effect of treatment on the expression of Ngf, Kdr, Kit, L1cam, Ntf3, Mapk3, and Neurog2. Our data confirm that L-T4 is necessary and sufficient for recovering memory and hippocampal neurogenesis deficits associated with hypothyroidism, while we found no evidence to support the role of non-canonical TH signalling.
... In microglia cells, TAAR1 agonist T1AM is capable of reducing the inflammatory response stimulated by Aβ, a factor in tumor necrosis (TNFα), and by lipopolysaccharide (LPS). The inflammatory response is on the part of the microglia via the inhibition of pro-inflammatory factors' release (IL-6, TNFα, NF-kB, MCP1 and MIP1), stimulating the release of anti-inflammatory mediators (IL-10) [39]. Interestingly, the effect of ethanol causes an increase in TAAR1 expression in human microglia cell strains HMO6, which may indicate the influence of alcohol consumption on the functioning of the immune blood-brain barrier [12,56]. ...
Article
Full-text available
Trace amines are a separate, independent group of biogenic amines, close in structure to classical monoamine neurotransmitters such as dopamine, serotonin, and norepinephrine that include many products of the endogenous or bacteria-mediated decarboxylation of amino acids. A family of G protein-coupled trace amine-associated receptors (in humans, TAAR1, TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) that senses trace amines was discovered relatively recently. They are mostly investigated for their involvement in the olfaction of volatile amines encoding innate behaviors and their potential contribution to the pathogenesis of neuropsychiatric disorders, but the expression of the TAAR family of receptors is also observed in various populations of cells in the immune system. This review is focused on the basic information of the interaction of trace amines and their receptors with cells of the general immune systems of humans and other mammals. We also overview the available data on TAARs’ role in the function of individual populations of myeloid and lymphoid cells. With further research on the regulatory role of the trace amine system in immune functions and on uncovering the contribution of these processes to the pathogenesis of the immune response, a significant advance in the field could be expected. Furthermore, the determination of the molecular mechanisms of TAARs’ involvement in immune system regulation and the further investigation of their potential chemotactic role could bring about the development of new approaches for the treatment of disorders related to immune system dysfunctions.
... In the brain, TAAR1 proved to be an important modulator of the major monoamine (dopamine and serotonin) and glutamate signaling pathways, directing the attention of researchers on the therapeutic implications of TAAR1 ligands in neuropsychiatric disorders [3,12,13]. The pharmaceutical company Hoffmann-La Roche was an early leader in TAAR1 drug discovery and registered several patents [14] as well as published preclinical studies [15][16][17][18][19] with selective TAAR1 agonists. TAAR1 agonists now give promise to be a new generation of antipsychotic medications, as evidenced by two compounds that have entered clinical trials, SEP-363856 (ulotaront-Sunovion) and RO6889450 (ralmitaront-La Roche) [13,20,21]. ...
Article
Full-text available
Trace amine-associated receptor 1 (TAAR1) is an attractive target for the design of innovative drugs to be applied in diverse pharmacological settings. Due to a non-negligible structural similarity with endogenous ligands, most of the agonists developed so far resulted in being affected by a low selectivity for TAAR1 with respect to other monoaminergic G protein-coupled receptors, like the adrenoreceptors. This study utilized comparative molecular docking studies and quantitative-structure activity relationship (QSAR) analyses to unveil key structural differences between TAAR1 and alpha2-adrenoreceptor (α 2-ADR), with the aim to design novel TAAR1 agonists characterized by a higher selectivity profile and reduced off-target effects. While the presence of hydrophobic motives is encouraged towards both the two receptors, the introduction of polar/positively charged groups and the ligand conformation deeply affect the TAAR1 or α 2-ADR putative selectivity. These computational methods allowed the identification of the α 2 A-ADR agonist guanfacine as an attractive TAAR1-targeting lead compound, demonstrating nanomolar activity in vitro. In vivo exploration of the efficacy of guanfacine showed that it is able to decrease the locomotor activity of dopamine transporter knockout (DAT-KO) rats. Therefore, guanfacine can be considered as an interesting template molecule worthy of structural optimization. The dual activity of guanfacine on both α 2-ADR and TAAR1 signaling and the related crosstalk between the two pathways will deserve more in-depth investigation.
Article
Full-text available
The search for novel effective TAAR1 ligands continues to draw great attention due to the wide range of pharmacological applications related to TAAR1 targeting. Herein, molecular docking studies of known TAAR1 ligands, characterized by an oxazoline core, have been performed in order to identify novel promising chemo-types for the discovery of more active TAAR1 agonists. In particular, the oxazoline-based compound S18616 has been taken as a reference compound for the computational study, leading to the development of quite flat and conformationally locked ligands. The choice of a “Y-shape” conformation was suggested for the design of TAAR1 ligands, interacting with the protein cavity delimited by ASP103 and aromatic residues such as PHE186, PHE195, PHE268, and PHE267. The obtained results allowed us to preliminary in silico screen an in-house series of pyrimidinone-benzimidazoles (1a–10a) as a novel scaffold to target TAAR1. Combined ligand-based (LBCM) and structure based (SBCM) computational methods suggested the biological evaluation of compounds 1a–10a, leading to the identification of derivatives 1a–3a (hTAAR1 EC50 = 526.3–657.4 nM) as promising novel TAAR1 agonists.
Article
Full-text available
Very recently, we have developed a new generation of ligands targeting the cannabinoid receptor type 2 (CB2R), namely JR compounds, which combine the pharmacophoric portion of the CB2R positive allosteric modulator (PAM), EC21a, with that of the CB2R selective orthosteric agonist LV62, both synthesized in our laboratories. The functional examination enabled us to identify JR14a, JR22a, and JR64a as the most promising compounds of the series. In the current study, we focused on the assessment of the bitopic (dualsteric) nature of these three compounds. Experiments in cAMP assays highlighted that only JR22a behaves as a CB2R bitopic (dualsteric) ligand. In parallel, computational studies helped us to clarify the binding mode of these three compounds at CB2R, confirming the bitopic (dualsteric) nature of JR22a. Finally, the potential of JR22a to prevent neuroinflammation was investigated on a human microglial cell inflammatory model.
Article
Full-text available
There is a huge need for novel therapeutic and preventative approaches to Alzheimer’s disease (AD) and neuroinflammation seems to be one of the most fascinating solutions. The primary cell type that performs immunosurveillance and helps clear out unwanted chemicals from the brain is the microglia. Microglia work to reestablish efficiency and stop further degeneration in the early stages of AD but mainly fail in the illness’s later phases. This may be caused by a number of reasons, e.g., a protracted exposure to cytokines that induce inflammation and an inappropriate accumulation of amyloid beta (Aβ) peptide. Extracellular amyloid and/or intraneuronal phosphorylated tau in AD can both activate microglia. The activation of TLRs and scavenger receptors, inducing the activation of numerous inflammatory pathways, including the NF-kB, JAK-STAT, and NLRP3 inflammasome, facilitates microglial phagocytosis and activation in response to these mediators. Aβ/tau are taken up by microglia, and their removal from the extracellular space can also have protective effects, but if the illness worsens, an environment that is constantly inflamed and overexposed to an oxidative environment might encourage continuous microglial activation, which can lead to neuroinflammation, oxidative stress, iron overload, and neurotoxicity. The complexity and diversity of the roles that microglia play in health and disease necessitate the urgent development of new biomarkers that identify the activity of different microglia. It is imperative to comprehend the intricate mechanisms that result in microglial impairment to develop new immunomodulating therapies that primarily attempt to recover the physiological role of microglia, allowing them to carry out their core function of brain protection.
Article
Full-text available
It is well known that G protein–coupled receptors (GPCRs) assume multiple active states. Orthosteric ligands and/or allosteric modulators can preferentially stabilize specific conformations, giving rise to pathway-biased signaling. One of the most promising strategies to expand the repertoire of signaling-selective GPCR activators consists of dualsteric agents, which are hybrid compounds consisting of orthosteric and allosteric pharmacophoric units. This approach proved to be very promising showing several advantages over monovalent targeting strategies, including an increased affinity or selectivity, a bias in signaling pathway activation, reduced off-target activity and therapeutic resistance. Our study focused on the cannabinoid receptor type 2 (CB2R), considered a clinically promising target for the control of brain damage in neurodegenerative disorders. Indeed, CB2R was found highly expressed in microglial cells, astrocytes, and even in some neuron subpopulations. Here, we describe the design, synthesis, and biological evaluation of two new classes of potential dualsteric (bitopic) CB2R ligands. The new compounds were obtained by connecting, through different linkers, the pharmacophoric portion of the CB2R positive allosteric modulator (PAM), EC21a, with that of the CB2R selective orthosteric agonist LV62, both developed in our laboratories. A preliminary screening enabled us to identify compound JR64a as the most promising of the series. Indeed, functional examination highlighted a signaling ‘bias’ in favor of G protein activation over βarrestin2 recruitment, combined with high affinity for CB2R and the ability to efficiently prevent inflammation in human microglial cells (HMC3) exposed to LPS/TNFα stimulation, thus demonstrating great promise for the treatment of neurodegenerative diseases.
Article
Full-text available
Monoaminergic neurotransmitter systems play a central role in neuronal function and behaviour. Dysregulation of these systems gives rise to neuropsychiatric and neurodegenerative disorders with high prevalence and societal burden, collectively termed monoamine neurotransmitter disorders (MNDs). Despite extensive research, the transcriptional regulation of monoaminergic neurotransmitter systems is not fully explored. Interestingly, certain drugs that act on these systems have been shown to modulate central levels of the transcription factor AP-2 beta (AP-2β, gene: TFAP2Β). AP-2β regulates multiple key genes within these systems and thereby its levels correlate with monoamine neurotransmitters measures; yet, its signalling pathways are not well understood. Moreover, although dysregulation of TFAP2Β has been associated with MNDs, the underlying mechanisms for these associations remain elusive. In this context, this review addresses AP-2β, considering its basic structural aspects, regulation and signalling pathways in the controlling of monoaminergic neurotransmitter systems, and possible mechanisms underpinning associated MNDS. It also underscores the significance of AP-2β as a potential diagnostic biomarker and its potential and limitations as a therapeutic target for specific MNDs as well as possible pharmaceutical interventions for targeting it. In essence, this review emphasizes the role of AP-2β as a key regulator of the monoaminergic neurotransmitter systems and its importance for understanding the pathogenesis and improving the management of MNDs.
Article
Full-text available
Activation of microglia is a prominent pathological feature in tauopathies, including Alzheimer’s disease. How microglia activation contributes to tau toxicity remains largely unknown. Here we show that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, activated by tau, drives microglial-mediated tau propagation and toxicity. Constitutive activation of microglial NF-κB exacerbated, while inactivation diminished, tau seeding and spreading in young PS19 mice. Inhibition of NF-κB activation enhanced the retention while reduced the release of internalized pathogenic tau fibrils from primary microglia and rescued microglial autophagy deficits. Inhibition of microglial NF-κB in aged PS19 mice rescued tau-mediated learning and memory deficits, restored overall transcriptomic changes while increasing neuronal tau inclusions. Single cell RNA-seq revealed that tau-associated disease states in microglia were diminished by NF-κB inactivation and further transformed by constitutive NF-κB activation. Our study establishes a role for microglial NF-κB signaling in mediating tau spreading and toxicity in tauopathy. Wang et al show that microglial NF-κB activation is essential for tau spreading and tau-mediated spatial learning and memory deficits in tauopathy mice. Inactivation of NF-κB reversed tau associated microglial states and rescued autophagy deficits.
Article
Full-text available
Synaptic dysfunction is an early mechanism in Alzheimer’s disease that involves progressively larger areas of the brain over time. However, how it starts and propagates is unknown. Here we show that Aβ released by microglia in association with large extracellular vesicles (Aβ-EVs) alters dendritic spine morphology in vitro, at the site of neuron interaction, and impairs synaptic plasticity both in vitro and in vivo in the entorhinal cortex-dentate gyrus circuitry. 1 h after Aβ-EV injection into the mouse entorhinal cortex, long-term potentiation (LTP) was impaired in the entorhinal cortex but not in the dentate gyrus, its main target region, while 24 h later it was impaired also in the dentate gyrus, revealing a spreading of LTP deficit between the two regions. Similar results were obtained upon injection of EVs carrying Aβ naturally secreted by CHO7PA2 cells, while neither Aβ42alone nor inflammatory EVs devoid of Aβ were able to propagate LTP impairment. Using optical tweezers combined to time-lapse imaging to study Aβ-EV-neuron interaction, we show that Aβ-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aβ-EV motility was inhibited, no propagation of LTP deficit occurred along the entorhinal-hippocampal circuit, implicating large EV motion at the neuron surface in the spreading of LTP impairment. Our data indicate the involvement of large microglial EVs in the rise and propagation of early synaptic dysfunction in Alzheimer’s disease, and suggest a new mechanism controlling the diffusion of large EVs and their pathogenic signals in the brain parenchyma, paving the way for novel therapeutic strategies to delay the disease.
Article
Full-text available
TAAR1 is a neuroregulator with emerging evidence suggesting a role in immunomodulation. Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system. Here, we investigate TAAR1 expression in human primary monocytes, peripherally-derived macrophages, and MS brain tissue. RT-qPCR was used to assess TAAR1 levels in MS monocytes. Using a previously validated anti-human TAAR1 antibody and fluorescence microscopy, TAAR1 protein was visualized in lipopolysaccharide-stimulated or basal human macrophages, as well as macrophage/microglia populations surrounding, bordering, and within a mixed active/inactive MS lesion. In vivo, TAAR1 mRNA expression was significantly lower in MS monocytes compared to age- and sex-matched healthy controls. In vitro, TAAR1 protein showed a predominant nuclear localization in quiescent/control macrophages with a shift to a diffuse intracellular distribution following lipopolysaccharide-induced activation. In brain tissue, TAAR1 protein was predominantly expressed in macrophages/microglia within the border region of mixed active/inactive MS lesions. Considering that TAAR1-mediated anti-inflammatory effects have been previously reported, decreased mRNA in MS patients suggests possible pathophysiologic relevance. A shift in TAAR1 localization following pro-inflammatory activation suggests its function is altered in pro-inflammatory states, while TAAR1-expressing macrophages/microglia bordering an MS lesion supports TAAR1 as a novel pharmacological target in cells directly implicated in MS neuroinflammation.
Article
The design of dualsteric/bitopic agents as single chemical entities able to simultaneously interact with both the orthosteric and an allosteric binding site represents a novel approach in medicinal chemistry. Biased dualsteric/bitopic agents could enhance certain signaling pathways while diminishing the others that cause unwanted side effects. We have designed, synthesized, and functionally characterized the first CB2R heterobivalent bitopic ligands. In contrast to the parent orthosteric compound, our bitopic ligands selectively target CB2R versus CB1R and show a functional selectivity for the cAMP signaling pathway versus βarrestin2 recruitment. Moreover, the most promising bitopic ligand FD-22a displayed anti-inflammatory activity in a human microglial cell inflammatory model and antinociceptive activity in vivo in an experimental mouse model of neuropathic pain. Finally, computational studies clarified the binding mode of these compounds inside the CB2R, further confirming their bitopic nature.
Article
Schizophrenia is a complex and severe mental illness. Current treatments for schizophrenia typically modulate dopaminergic neurotransmission by D2-receptor blockade. While reducing positive symptoms of schizophrenia, current antipsychotic drugs have little clinical effect on negative symptoms and cognitive impairments. For the last few decades, discovery efforts have sought nondopaminergic compounds with the aim to effectively treat the broad symptoms of schizophrenia. In this viewpoint, we provide an overview on trace-amine associated receptor-1 (TAAR1), which presents a clinically validated nondopaminergic target for treating schizophrenia and related disorders, with significantly less overall side-effect burden. TAAR1 agonists may also be specifically beneficial for the substance abuse comorbidity and metabolic syndrome that is often present in patients with schizophrenia.
Article
The 3-iodothyronamine (T1AM) and 3-iodothryoacetic acid (TA1), are endogenous occurring compounds structurally related with thyroid hormones (THs, the pro-hormone T4 and the active hormone T3) initially proposed as possible mediators of the rapid effects of T3. However, after years from their identification, the physio-pathological meaning of T1AM and TA1 tissue levels remains an unsolved issue while pharmacological evidence indicates both compounds promote in rodents central and peripheral effects with mechanisms which remain mostly elusive. Pharmacodynamics of T1AM includes the recognition of G-coupled receptors, ion channels but also biotransformation into an active metabolite, i.e. the TA1. Furthermore, long term T1AM treatment associates with post-translational modifications of cell proteins. Such array of signaling may represent an added value, rather than a limit, equipping T1AM to play different functions depending on local expression of targets and enzymes involved in its biotransformation. Up to date, no information regarding TA1 mechanistic is available. We here review some of the main findings describing effects of T1AM (and TA1) which suggest these compounds interplay with the histaminergic system. These data reveal T1AM and TA1 are part of a network of signals involved in neuronal plasticity including neuroprotection and suggest T1AM and TA1 as lead compounds for a novel class of atypical psychoactive drugs.