Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

European Journal of Pharmacology 946 (2023) 175650

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Losartan attenuates acetic acid enema-induced visceral hypersensitivity by


inhibiting the ACE1/Ang II/AT1 receptor axis in enteric glial cells
Yating Sun a, Xiaohui Liu b, Lianli Wang a, Laifu Li a, Xiaojing Quan a, Haitao Shi a, Ting Wang a,
Lin Mei a, Yindi Chen c, Yue Zhang a, Jingyao Li a, Ruiting Meng a, Jinhai Wang a, Fei Dai a, *
a
Department of Gastroenterology, Second Affifiliated Hospital of Xi’an Jiaotong University, Xi’an, China
b
Department of Bone and Joint Surgery, Second Affifiliated Hospital of Xi’an Jiaotong University, Xi’an, China
c
Department of Gastroenterology, Xi’an People’s Hospital, Xi’an, China

A R T I C L E I N F O A B S T R A C T

Keywords: Enteric glial cells (EGCs) play an important role in visceral hypersensitivity associated with irritable bowel syn­
Visceral hypersensitivity drome (IBS). Losartan (Los) is known to reduce pain; however, its function in IBS is unclear. The present study
Enteric glial cell aimed to investigate Los’s therapeutic effect on visceral hypersensitivity in IBS rats. Thirty rats were randomly
Irritable bowel syndrome
divided into control, acetic acid enema (AA), AA + Los low, medium and high dose groups in vivo. EGCs were
Losartan
treated with lipopolysaccharide (LPS) and Los in vitro. The molecular mechanisms were explored by assessing the
AT1 receptor
Inflammation expression of EGC activation markers, pain mediators, inflammatory factors and angiotensin-converting enzyme
1(ACE1)/angiotensin II (Ang II)/Ang II type 1 (AT1) receptor axis molecules in colon tissue and EGCs. The results
showed that the rats in the AA group showed significantly higher visceral hypersensitivity than the control rats,
which was alleviated by different doses of Los. The expression of GFAP, S100β, substance P (SP), calcitonin gene-
related peptide (CGRP), transient receptor potential vanilloid 1 (TRPV1), tumor necrosis factor (TNF), inter­
leukin-1β (IL-1β) and interleukin-6 (IL-6) was considerably increased in colonic tissues of AA group rats and LPS-
treated EGCs compared with control rats and EGCs, and reduced by Los. In addition, Los reversed ACE1/Ang II/
AT1 receptor axis upregulation in AA colon tissues and LPS-treated EGCs. These results show that Los inhibits
ACE1/Ang II/AT1 receptor axis upregulation by suppressing EGC activation, resulting in reduced expression of
pain mediators and inflammatory factors, thereby alleviating visceral hypersensitivity.

et al., 2016). This state of EGCs is referred to as the reactive EGC


phenotype. In addition, calcitonin gene-related peptide (CGRP) and
1. Introduction
transient receptor potential vanilloid 1 (TRPV1) are also closely asso­
ciated with visceral hypersensitivity in relation to IBS (Akbar et al.,
Visceral hypersensitivity is the main driver of abdominal pain in
2008; Shi et al., 2015).
relation to irritable bowel syndrome (IBS), which may be associated
Considerable evidence supports the concept of a local tissue or
with various factors (Pusceddu and Gareau, 2018; Sengupta, 2009).
cellular renin–angiotensin–aldosterone system (RAAS) (Karnik et al.,
Recently, considerable attention has been given to the role of enteric
2015; Obukhov et al., 2020). Some studies have shown that the local
glial cells (EGCs) and the secretory mediators of the enteric nervous
RAAS can be involved in inflammation and pain (Bali et al., 2014; Karnik
system (ENS) in the visceral hypersensitivity response to IBS.
et al., 2015). Ang II mainly binds with high affinity to Ang II type 1
EGCs are a prominent component of the ENS. Upon alteration of the
(AT1) receptor to generate inflammatory responses and reactive oxygen
gastrointestinal environment, EGCs are activated, calcium-binding
species (Xu et al., 2017) and participates in nociception (Hegazy et al.,
protein S100β and glial fibrillary acidic protein (GFAP) expression is
2020a). Ang II has been shown to co-localize with AT1 receptor, SP,
upregulated, and substance P (SP) is released and expressed to partici­
CGRP, and TRPV1 in the dorsal root ganglion (DRG) (Anand et al., 2015;
pate in visceral hypersensitivity (Grubišić and Gulbransen, 2017; Wang

* Corresponding author.
E-mail addresses: 18204311643@163.com (Y. Sun), liuxiaohuixjtuer@163.com (X. Liu), 15737939767@163.com (L. Wang), 1114502232@qq.com (L. Li),
quanxiaojing77@126.com (X. Quan), shihaitao7@xjtu.edu.cn (H. Shi), wttingzai@163.com (T. Wang), milly_meilin@outlook.com (L. Mei), 593825787@qq.com
(Y. Chen), zhangyue_zyzy@163.com (Y. Zhang), 893139621@qq.com (J. Li), 952463828@qq.com (R. Meng), Jinhaiwang@hotmail.com (J. Wang), daifei68@
xjtu.edu.cn (F. Dai).

https://doi.org/10.1016/j.ejphar.2023.175650
Received 31 October 2022; Received in revised form 2 March 2023; Accepted 7 March 2023
Available online 10 March 2023
0014-2999/© 2023 Elsevier B.V. All rights reserved.
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Abbreviations: GFAP Glial fibrillary acidic protein


SP Substance P
EGC Enteric glial cells CGRP Calcitonin gene-related peptide
IBS Irritable bowel syndrome TRPV1 Transient receptor potential vanilloid 1
Ang Angiotensin RAAS Renin-angiotensin-aldosterone system
AT1 Ang II type 1 ACE1 Angiotensin-converting enzyme 1
Los Losartan MAPK Mitogen-activated protein kinase
VMR Visceral motor response NF- κB Nuclear factor-κB
CRD Colorectal balloon dilation EMG Electromyography
LPS Lipopolysaccharide AUC Area under the curve
ENS Enteric nervous system IL-1β Interleukin-1β
IHC Immunohistochemistry IL-6 Interleukin-6
IF Immunofluorescence TNF Tumor necrosis factor
qRT-PCR Quantitative real-time PCR SD Standard deviation

Patil et al., 2010), which provides strong evidence supporting the groups were administered with 0.2–0.3 ml of 0.5% acetic acid (0.2 ml
involvement of Ang II binding to AT1 receptor in nociception. AT1 re­ during the 1st week and 0.3 ml during the 2 nd week after the start of
ceptor activation in spinal dorsal horn neurons and astrocytes causes modelling) on the 8th-21st day of life by daily insertion of an epidural
activation of the mitogen-activated protein kinase (MAPK) pathway, catheter (1 mm in diameter) through the anus for 2–3 cm after lubri­
which is involved in spinal pain transmission (Nemoto, 2018; Ogata cation with paraffin oil to form a visceral hypersensitivity model. The
et al., 2016). AT1 receptor blockers or angiotensin-converting enzyme 1 control group was perfused with equal amounts of saline. From day 22
(ACE1) inhibitors, on the other hand, can prevent and relieve pain (Bali until week 7, rats in all groups were left untreated, with a normal diet
et al., 2014; Marques-Lopes et al., 2009). and water intake, and at week 8, the Los intraperitoneal intervention
Losartan (Los) is a selective AT1 receptor blocker with significant was started for the AA + Los-L group (10 mg/kg/d), the AA + Los-M
anti-inflammatory effects (Kalynovska et al., 2020; Yamamoto et al., group (25 mg/kg/d) and the AA + Los-H group (50 mg/kg/d) with
2015). Whereas inflammation may contribute to the induction and reference to several common dosages of Los in previous studies (Aka­
maintenance of pain, studies have shown that Los may attenuate colonic zawa et al., 2021; Collister et al., 1996; Xiu et al., 2002). The remaining
injury by inhibiting pro-inflammatory cytokine production in the two groups were given an equal volume of saline for 10 days of treat­
colonic mucosa (Shi et al., 2016). Several studies have demonstrated the ment. During the experiment, the body weight of the rats was recorded
presence of Ang II and AT1 receptor in the ENS, that Ang II excites every 7 days.
neurons in the myenteric and submucosal plexuses (Wang et al., 2005).
However, the role of the ACE1/Ang II/AT1 receptor axis in EGC acti­
vation and visceral hypersensitivity in IBS is unclear. 2.3. Assessment of visceral sensation
Therefore, we hypothesized that the ACE1/Ang II/AT1 receptor axis
may interact with EGCs to participate in IBS visceral hypersensitivity Previous research has confirmed the visceral hypersensitivity of IBS
and that Los may improve IBS visceral hypersensitivity by modulating patients with increased sensitivity to CRD (Keszthelyi et al., 2012). In
the ACE1/Ang II/AT1 receptor axis to inhibit EGC activation. We this study, visceral sensation was assessed by extra-abdominal oblique
explored the mechanism involving Los intervention in an acetic acid muscle electromyography (EMG) measurements after CRD (Nozu et al.,
enema-induced IBS rat model and lipopolysaccharide (LPS)-induced 2017). At the end of the Los intervention, pentobarbital (30 mg/kg,
EGCs in vitro experiments. intraperitoneal injection) was used to anaesthetize fasted Sprague‒
Dawley rats intraperitoneally for extra-abdominal oblique muscle elec­
trode implantation, and the free end of the electrode was fixed behind
2. Materials and methods
the neck through a subcutaneous tunnel for EMG signal measurement.
EMG signals were recorded by a CRD instrument (BL420 Data Acquisi­
2.1. Animals
tion and Analysis System, Chengdu TME Technology Co., Ltd., China) 3
days after surgery, and the visceromotor response (VMR) was measured
Thirty male experimental clean-grade neonatal Sprague‒Dawley
in each group of rats. The volumes of balloon injection were 0 ml, 0.4 ml,
rats were purchased from the Animal Experiment Center of Xi’an Jiao­
0.8 ml and 1.2 ml, and the recordings were repeated three times. The
tong University and were housed in the same cage as their mothers until
VMR value was calculated using the area under the curve (AUC) at 20 s
day 21, at which point they were separated from their mothers. All
during dilation minus the area at 20 s during the baseline period before
animals were housed in a specific-pathogen-free environment with a 12-
dilation.
h light and 12-h dark cycle, and the animals had free access to food and
water. All animal experimental procedures were performed in accor­
dance with National Institutes of Health guidelines and approved by the 2.4. Colonic tissue specimen collection
Ethics Committee of Xi’an Jiaotong University (No. 2017-127, approved
February 28, 2017). At the end of the visceral hypersensitivity test, the rats were anaes­
thetized with pentobarbital, and each group was given 250 ml of saline
2.2. Visceral hypersensitivity rat model and drug treatments for intracardiac perfusion. 5 cm colonic samples were collected from 8
cm proximal to the anus and divided into three parts. One section of the
Thirty neonatal Sprague‒Dawley rats were randomly divided into colon tissue was fixed in 4% paraformaldehyde solution, dehydrated,
five groups: the control group (n = 6), acetic acid enema group (AA, n = embedded in paraffin and sectioned for immunofluorescence (IF) and
6), AA + Los (HY-17512A, MCE) low-dose group (AA + Los-L, n = 6), immunohistochemistry (IHC) staining, and the other two sections were
AA + Los medium-dose group (AA + Los-M, n = 6), and AA + Los high- rapidly frozen in liquid nitrogen and then transferred to a − 80 ◦ C freezer
dose treatment group (AA + Los-H, n = 6). The rats in the four treatment for quantitative real-time PCR (qRT‒PCR) and Western blotting.

2
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

2.5. Cell culture Table 1


QRT-PCR primer pair sequences for Sprague‒Dawley rats.
Rat EGCs (CRL-2621, ATCC) were grown in medium supplemented Gene Primers, 5′ -3′
with 10% foetal bovine serum (FBS, Gibco), 100 U/mL penicillin and
GFAP Sense AAACAGAAGAGTGGTATCGG
100 μg/mL streptomycin (Gibco) and incubated at 5% CO2 and 37 ◦ C. Antisense TAGTCATTAGCCTCGTGCTT
The cells were treated with LPS (L2630, Sigma–Aldrich) or Los in vitro S100β Sense TTCAGGGAGAGAGGGTGACAA
experimental studies. Antisense CTTCCTGCTCTTTGATTTCCTCC
CGRP Sense CTTTCCTGGTTGTCAGCATCTT
Antisense AAGTTGTCCTTCACCACACCTC
2.6. Cell proliferation and viability assay SP Sense ATGAAAATCCTCGTGGCGGT
Antisense CAGCATCCCGTTTGCCCATT
Cell proliferation and viability were assessed using cell counting kit- TRPV1 Sense AAGGATGGAACAACGGGCTAG
Antisense TCCTGGTAGTGAAGATGTGGG
8 (CCK-8) (Dojindo, Japan). EGCs (CRL-2621, ATCC) were inoculated IL-1β Sense TGTGATGTTCCCATTAGAC
into 96-well plates at a density of 3*103 cells/well and treated with Antisense AATACCACTTGTTGGCTTA
different concentrations of Los (0, 10− 9, 10− 8, 10− 7, 10− 6, 10− 5, 10− 4, IL-6 Sense GGAAGTTGGGGTAGGAAGGA
and 10− 3 M), while a negative control was set up to observe the effects of Antisense TGTGATGTTCCCATTAGAC
TNF Sense CCCAATCTGTGTCCTTCTAACT
the drug on cell proliferation and viability. Based on the above results,
Sense CAGCGTCTCGTGTGTTTCT
the cellular proliferation and viability tests were performed on the cells Ang II Sense GAAGACCCTGCGAGATAAGC
after LPS (10 μg/ml) intervention with the appropriate range of Los Antisense ACTGGCTACACCTCTTGCCT
concentrations. AT1 R Sense CTCAAGCCTGTCTACGAAAATGAG
Antisense GTGAATGGTCCTTTGGTCGT
ACE1 Sense AGACTTGCCTGTGACCTTTC
2.7. Cell treatment Antisense CTGTGTAGATGCTTGGGTGTAG
β-actin Sense CCCGCGAGTACAACCTTCTTG
EGCs were inoculated uniformly into well plates and divided into the Antisense GTCATCCATGGCGAACTGGTG

control, LPS, Los, and LPS + Los groups. LPS and Los were dissolved with Abbreviation: GFAP, glial fibrillary acidic protein; CGRP, calcitonin gene-related
phosphate buffered saline (PBS). Cells were starved for 12 h at 50% cell peptide; SP, substance P; TRPV1, transient receptor potential vanilloid 1; IL-1β,
density. The LPS group was given 10 μg/ml LPS-treated cells (LPS− 5), interleukin-1β; IL-6, interleukin-6; TNF, tumor necrosis factor; Ang II, angio­
the Los group was given 10− 6 M Los-treated cells (Los− 6), the LPS + Los tensin II; AT1R, ang II type 1 receptor; ACE1, angiotensin-converting enzyme 1.
group was given cells pre-treated with 10− 6 M Los for 2 h followed by
LPS (10 μg/ml)-stimulated cells (LPS− 5+Los− 6), and the control group 2.10. ELISA
was given equal amounts of PBS. Afterwards, the cells were incubated at
37 ◦ C (5% CO2) for 24 h, RNA was extracted for qRT‒PCR, and protein To assess the levels of pain mediators and inflammatory factors
was extracted after 48 h of incubation and the cell supernatant was released by EGC after its activation, we collected cell supernatants after
collected for Western blotting and Enzyme-linked immunosorbent assay 48 h of cell treatment and measured SP and IL-6 levels in the cell su­
(ELISA) assays, respectively. pernatants using a rat substance P ELISA assay kit (MLBio, China) and a
rat IL-6 ELISA assay kit (Quanzhou Ruixin Biotechnology, China),
2.8. QRT-PCR respectively, according to the manufacturer’s instructions.

Total RNA was extracted from colonic tissue or cultured EGCs using 2.11. IHC staining
standard methods with RNAiso-Plus (9109, Takara Biotechnology) and
reverse transcribed using a cDNA kit (Genstar, cat# A223-10). The ob­ Paraffin-embedded tissue was cut into 4 μm slices for IHC staining.
tained cDNA was analysed by qRT‒PCR using a Thermo Fisher Scientific All sections were routinely dewaxed, rehydrated, and treated with goat
Applied Biosystems 7500/7500 Fast qRT‒PCR (QuantStudio™ Design & serum to seal non-specific binding sites. Sections were incubated over­
Analysis SE Software) detection system and 2X Universal SYBR Green night at 4 ◦ C with primary antibodies against each antigen: anti-GFAP
Fast qPCR Mix (ABclonal, RK21203) as follows: denaturation (95 ◦ C, 3 (1:2000, Proteintech) and anti-SP (1:100, Affinity). They were then
min) for 1 cycle and annealing (95 ◦ C, 5 s) and extension (60 ◦ C, 32 s) for incubated with horseradish peroxidase-coupled secondary antibodies
45 cycles. All experiments were repeated 3 times, and the Ct values were (1:250) for 1 h at room temperature. Immunohistochemical staining of
normalized according to the expression of β-actin. The primer sequences colon tissue sections was performed according to the instructions for the
used in this study are listed in Table 1. diaminobenzidine kit (ZLI-9018, ZSGB-BIO), and five random fields of
view were examined for each sample at 400× magnification. The results
2.9. Western blot were finally analysed using ImageJ software.

Protein was extracted from rat colon tissue or EGCs by lysis in pre- 2.12. IF staining
chilled RIPA lysis buffer containing 1 mM PMSF and phosphatase in­
hibitor. After electrophoresis and membrane transfer, the membranes 2.12.1. Tissue double IF staining
were closed at room temperature in 0.5% skim milk powder (P0216, All sections were routinely dewaxed, rehydrated, permeabilized with
Beyotime) for 2 h and incubated overnight at 4 ◦ C with anti-GFAP 0.2% Triton X-100 for 10 min and then closed in PBS containing 5% goat
(1:5000, 4B2E10, Proteintech), anti-SP (1:1000, DF7522, Affinity), serum for 30 min. The expression of AT1 receptor in colon tissue EGCs
anti-ACE1 (1:500, 24743-1-AP, Proteintech) and anti-GAPDH (1:10000, was detected by double IF staining. EGC cells were labelled with anti-
60004-1-Ig, Proteintech), followed by 1 h of incubation at room tem­ GFAP, and sections were incubated overnight at 4 ◦ C with anti-GFAP
perature with a 1:5000 dilution of horseradish peroxidase-coupled sec­ (1:50, sc-33673, Santa Cruz Biotechnology) and anti-AT1 receptor
ondary antibody, and the bands were visualized using (1:50, Proteintech). Then, the cells were incubated with fluor488-
electrochemiluminescent solution (Millipore) exposure on a BioRad coupled secondary antibody (Immunoway, 1:250) and fluro568-
imaging system. The relative expression of each protein was determined coupled secondary antibody (Immunoway, 1:250) for 2 h at room
using ImageJ software for quantitative analysis and GAPDH as an in­ temperature protected from light. Finally, the slices were stained with
ternal reference. DAPI for 7 min and sealed with an anti-fluorescence quencher.

3
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

2.12.2. Cellular IF staining start of the enema (week 0), with no significant differences. The body
Cells were inoculated into 24-well plates and treated for 24 h ac­ weight of the control rats was slightly higher than that of the rats in the
cording to the protocol described above. Then, the cells were fixed with other 4 groups at the beginning of the first week of the acetic acid enema
4% paraformaldehyde, permeabilized and blocked as described previ­ treatment (p < 0.05), and the body weight of the control rats was still
ously, followed by incubation with anti-AT1 receptor (1:50, Pro­ slightly higher than that of the rats in the other 4 groups at 2–8 weeks,
teintech) at 4 ◦ C overnight. The cells were then incubated with fluro568- but there was no statistically significant difference, suggesting that the
coupled secondary antibody (Immunoway, 1:250) protected from light growth of rats was slightly and transiently affected during the initial
and finally stained with DAPI and sealed with anti-fluorescence acetic acid enema, but the effect was not evident after the rats reached
quencher. adulthood. Additionally, Los treatment had no significant effect on the
Tissue sections and cell crawls were observed and photographed growth of rats (Fig. 1A).
under a fluorescence microscope (Nikon, Japan) and analysed for fluo­ Assessment of the VMR revealed that with an increasing balloon
rescence intensity using ImageJ software. injection volume, the EMG wave amplitude and density increased
significantly in the AA group compared to the control group, with sta­
tistically significant differences between the two groups at 0.8 ml and
2.13. Statistical analysis
1.2 ml CRD (p < 0.001), suggesting successful modelling and that acetic
acid treatment in neonatal rats produces persistent visceral hypersen­
Data are presented as the mean ± standard deviation (SD). One-way
sitivity (Fig. 1B and C). After 10 days of intervention at each dosage of
or two-way analysis of variance (ANOVA) was used to compare data
Los, compared with those of the AA group, the EMG wave amplitude and
between groups. GraphPad Prism 8.0 was used for statistical analysis. P
density of the AA + Los-L, AA + Los-M and AA + Los-H groups at 0.8 ml
values < 0.05 were considered statistically significant.
(p AA+Los-L < 0.05, p AA+Los-M < 0.001, p AA+Los-H < 0.01) and 1.2 ml (p
AA+Los-L < 0.05, p AA+Los-M / AA+Los-H < 0.001) CRD were significantly
3. Results reduced, and the VMR reduction in the AA + Los-M group was signifi­
cantly larger than that in the AA + Los-L group at 1.2 ml CRD (p < 0.05)
3.1. Los ameliorated the VMR of viscerally hypersensitive rats (Fig. 1B and C).

The body weights of the rats were similar among groups before the

Fig. 1. Effects of Los on the body weight and VMR of


viscerally hypersensitive rats. (A) Weekly changes in
the body weight of rats in each group from week 0 of
enema treatment. (B) Quantitative analysis of the ef­
fect of Los on the VMR of rats with different volumes
(0 ml, 0.4 ml, 0.8 ml, and 1.2 ml) of CRD in viscerally
hypersensitive rats. (C) Typical representative EMG
images of the effect of Los on the VMR under different
volumes (0 ml, 0.4 ml, 0.8 ml, and 1.2 ml) of CRD in
viscerally hypersensitive rats. Data are expressed as
the mean ± SD, n = 6. ns, p > 0.05; *p < 0.05, **p <
0.01 and ***p < 0.001 for Control vs. AA; #p < 0.05,
##
p < 0.01 and ###p < 0.001 for AA vs. AA + Los-L
or AA + Los-M or AA + Los –H; Δp < 0.05 AA + Los-L
vs. AA + Los-M. AA, acetic acid enema; Los-L, los­
artan low-dose; Los-M, losartan medium-dose; Los-H,
losartan high-dose; CRD, colorectal balloon dilation;
VMR, visceral motor response.

4
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

3.2. Los reduced the mRNA expression of colonic inflammatory factors in was significantly higher in the AA group than in the control group
viscerally hypersensitive rats (Fig. 3B), and IHC (p < 0.001) (Fig. 3C and D) and Western blotting (p <
0.001) (Fig. 3E and F) showed that GFAP protein expression was also
To determine whether the visceral hypersensitivity in adult rats was significantly increased. GFAP mRNA (p AA+Los-L < 0.05, p AA+Los-M <
due to colonic inflammation after neonatal AA enema, we performed 0.001, p AA+Los-H < 0.01) and S100β mRNA (p AA+Los-L / AA+Los-M / AA+Los-
qRT‒PCR to assess the mRNA expression levels of the colonic tissue H < 0.001) expression was decreased in colonic tissues after different
inflammatory factors IL-1β, IL-6 and TNF. The mRNA expression levels doses of Los treatment compared with the AA group (Fig. 3B), and IHC (p
of TNF (p < 0.01) and IL-1β (p < 0.001) were significantly higher in the AA+Los-L < 0.05, p AA+Los-M / AA+Los-H < 0.001) (Fig. 3C and D) and
colonic tissue of rats with visceral hypersensitivity than in the control Western blot (p AA+Los-L < 0.05, p AA+Los-M / AA+Los-H < 0.001) (Fig. 3E
group rats, and IL-6 (p > 0.05) mRNA expression was slightly higher, but and F) showed that GFAP protein expression was also significantly
there was no statistically significant difference. Different doses of Los reduced, suggesting that the involvement of Los in regulating visceral
treatment all reduced TNF (p AA+Los-L / AA+Los-M < 0.05, p AA+Los-H < hypersensitivity is associated with inhibition of EGC activation.
0.01), IL-1β (p AA+Los-L / AA+Los-M / AA+Los-H < 0.001), and IL-6 (p AA+Los-L
< 0.05, p AA+Los-M / AA+Los-H > 0.05) mRNA expression (Fig. 2). This 3.4. AT1 receptor was co-localized and up-regulated with GFAP in the
result suggested an inflammatory response in the colonic tissue of colonic tissues of viscerally hypersensitive rats
viscerally hypersensitive rats and that Los could alleviate visceral hy­
persensitivity by attenuating the inflammatory response in colonic To clarify the expression of RAAS components in colonic tissues, we
tissue. showed by IF double-labelling that AT1 receptor was expressed in all
layers of the colon but mainly in the mucosal layer, submucosal plexus
3.3. Los decreased the expression of SP, CGRP, TRPV1, GFAP, and and intermuscular plexus and co-localized with GFAP, and both were
S100β in viscerally hypersensitive rats increased in the AA group compared with the control group (Fig. 4A).
Western blot analysis showed that the expression of ACE1 was increased
To investigate whether Los ameliorates the VMR in viscerally hy­ in the colon tissue of the AA group compared with the control group (p
persensitive rats by reducing the expression of pain mediators in colonic < 0.05), and the expression of ACE1 in the colon tissue was decreased
tissues, we detected and evaluated the relevant molecules using qRT‒ after different doses of Los intervention compared with the AA group
PCR, Western blotting and IHC. The relative expression of SP mRNA, (Fig. 4B and C) (p AA+Los-M / AA+Los-H < 0.05), but there was no statis­
CGRP mRNA and TRPV1 mRNA was significantly higher in the colon tically significant difference between the AA + Los-L and AA groups (p
tissue of the AA group than in the control group (p SP / CGRP / > 0.05), suggesting that Los improves visceral hypersensitivity by
modulating colonic tissue RAAS components.
TRPV1<0.001) (Fig. 3A), and the IHC (p < 0.001) (Fig. 3C and D) and
Western blot (p < 0.001) (Fig. 3E and F) results showed that SP protein
expression was also significantly increased. SP mRNA (p AA+Los-L < 0.01, 3.5. Los protected EGC proliferation and viability against LPS treatment
p AA+Los-M / AA+Los-H < 0.05), CGRP mRNA (p AA+Los-L / AA+Los-M / AA+Los- in vitro
H < 0.001) and TRPV1 mRNA (p AA+Los-M < 0.01, p AA+Los-L / AA+Los-H <
0.001) expression was significantly decreased in colonic tissues First, we compared the effects of different concentrations of Los
following different doses of Los treatment compared with the AA group (10− 9 M to 10− 3 M) on EGC proliferation and viability (Fig. 5A). Los
(Fig. 3A), while IHC (p AA+Los-L / AA+Los-M / AA+Los-H < 0.001) (Fig. 3C and treatment of EGCs for 24 h had no significant effect on EGC proliferation
D) and Western blot (p AA+Los-M < 0.05, p AA+Los-H < 0.01) (Fig. 3E and F) and viability at 10− 9 M to 10− 4 M (p > 0.05), while it significantly
showed that SP protein expression was also significantly reduced. inhibited EGC proliferation at 10− 3 M (p < 0.001). At 48 h, Los had no
To investigate whether the above effects were associated with EGC significant effect on EGC proliferation and viability at low concentra­
activation, we evaluated the changes in GFAP mRNA and S100β mRNA tions (10− 9 M to 10− 6 M) (p > 0.05), while it significantly inhibited
expression in the colonic tissues of rats in each group. The relative these variables at 10− 5 M to 10− 3 M (p −105 < 0.01, p −104 −/ 310 < 0.001).
expression of GFAP mRNA (p < 0.001) and S100β mRNA (p < 0.001) Next, we examined the effects of different concentrations of Los

Fig. 2. Los reduced the mRNA expression of colonic inflammatory factors in viscerally hypersensitive rats. Analysis of the mRNA expression of the inflammatory
factors TNF, IL-1β, and IL-6 in the colonic tissues of rats in each group. Data are expressed as the mean ± SD, n = 6. ns, p > 0.05; **p < 0.01 and ***p < 0.001 for
Control vs. AA; #p < 0.05, ##p < 0.01 and ###p < 0.001 for AA vs. AA + Los-L or AA + Los-M or AA + Los-H. AA, acetic acid enema; Los-L, losartan low-dose; Los-M,
losartan medium-dose; Los-H, losartan high-dose; TNF, tumor necrosis factor; IL-1β, interleukin-1β; IL-6, interleukin-6.

5
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Fig. 3. Los reduced pain mediators and EGC activation markers in the colons of viscerally hypersensitive rats. (A) Analysis of the mRNA expression of the pain
mediators SP, CGRP, and TRPV1 in each group of rats. (B) Analysis of the mRNA expression of the EGC activation markers GFAP and S100β in each group. (C)
Representative images (400× magnification, scale bar = 50 μm) of IHC for SP (indicated in brown) and GFAP (indicated in brown) in each group of rats. (D) The
mean optical density of SP and GFAP (mean density: IOD/area) was analysed using ImageJ. (E) Representative images of SP and GFAP protein expression detected by
Western blot. (F) Quantitative analysis of Western blot results for SP and GFAP. The results are presented as the mean ± SD, n = 6. ***p < 0.001 for Control vs. AA;
#
p < 0.05, ##p < 0.01 and ###p < 0.001 for AA vs. AA + Los-L or AA + Los-M or AA + Los-H. AA, acetic acid enema; Los-L, losartan low-dose; Los-M, losartan
medium-dose; Los-H, losartan high-dose; SP, substance P; CGRP, calcitonin gene-related peptide; TRPV1, transient receptor potential vanilloid 1; GFAP, glial
fibrillary acidic protein; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

(10− 7 M to 10− 5 M) on the proliferation and viability of LPS (10 ug/ml)- 3.6. Los inhibited the activation of EGCs induced by LPS
treated cells (Fig. 5B). The proliferation and viability of EGCs were
partially inhibited by LPS treatment at 24 h (p < 0.05) and significantly As mentioned previously, EGC activation plays an important role in
inhibited at 48 h and 72 h (p < 0.001). In contrast, Los inhibited this IBS visceral hypersensitivity. To investigate whether Los can reduce EGC
adverse effect, and both 10− 7 M and 10− 5 M concentrations had a pro­ activation, we performed qRT‒PCR, revealing that EGCs were signifi­
tective effect on EGC proliferation and viability at 24 h (p < 0.05), while cantly activated by LPS treatment, and the mRNA expression of its
10− 6 M also showed partial protection, but the difference was not sta­ activation markers GFAP and S100β was significantly increased (p <
tistically significant (p = 0.105). At 48 h, all three concentrations from 0.001), while the expression of both was significantly decreased after
10− 7 M to 10− 5 M had a significant protective effect on EGC prolifera­ pre-treatment with Los (p < 0.001) (Fig. 6A). Subsequently, we further
tion and viability (p < 0.001), and there were no significant differences verified by Western blot that Los significantly reduced the expression
between the three concentrations (p > 0.05). At 72 h, all three con­ level of GFAP protein induced by LPS activation of EGCs (p < 0.01)
centrations from 10− 7 M to 10− 5 M had a significant protective effect on (Fig. 6B and C).
EGC proliferation and viability, and both 10− 7 M and 10− 6 M were
significantly better than 10− 5 M (p −vs.
7
5 − 5
6
10 10< 0.001, p vs.
10 10< 0.001). Based 3.7. Los reduced LPS-induced pain mediators production and release
− 6
on this, a concentration of 10 M was used for the subsequent experi­ following EGC activation
mental study in this study.
Pain mediators play an important role in visceral hypersensitivity in
relation to IBS, and to investigate whether Los could reduce the pro­
duction of pain mediators in EGCs, we performed relevant validation

6
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Fig. 4. Localization and expression of AT1 receptor


and ACE1 in the colonic tissues of viscerally hyper­
sensitive rats. (A) Representative images of GFAP and
AT1 receptor IF co-localization in colon tissues of the
control and AA groups (green fluorescence indicates
GFAP-positive cells, red fluorescence indicates AT1
receptor-positive cells; 200 × magnification; scale
bar = 100 μm). (B) Representative image of Western
blot to detect ACE1 protein expression. (C) Quanti­
tative analysis of Western blot results for ACE1. The
results are presented as the mean ± SD, n = 6. *p <
0.05 for Control vs. AA; #p < 0.05 for AA vs. AA +
Los-M or AA + Los-H. AA, acetic acid enema; Los-L,
losartan low-dose; Los-M, losartan medium-dose;
Los-H, losartan high-dose; SP, substance P; GFAP,
glial fibrillary acidic protein; AT1R, ang II type 1 re­
ceptor; DAPI, 4′ ,6-diamidino-2′ -phenylindole; ACE1,
angiotensin-converting enzyme 1; GAPDH,
glyceraldehyde-3-phosphate dehydrogenase.

Fig. 5. Los protected cell proliferation and viability


against LPS treatment in vitro. (A) CCK-8 analysis was
used to detect the effect of different concentrations
(10− 9 M to 10− 3 M) of Los on EGC proliferation and
viability at 24 h and 48 h. (B) Proliferation and
viability analysis of EGCs pre-treated with 10− 7, 10− 6
and 10− 5 M Los for 2 h, followed by LPS (10 μg/ml)
treatment of EGCs for 24 h, 48 h and 72 h, respec­
tively. Data are presented as the mean ± SD, n = 3.
ns, p > 0.05; **p < 0.01 and ***p < 0.001 for Control
vs. Los; &&p < 0.01 and &&&p < 0.001 for Control vs.
LPS− 5; ∇p < 0.05 and ∇∇∇p < 0.001 for LPS− 5 vs.
LPS− 5+Los− 7; ###p < 0.001 for LPS− 5 vs.
LPS− 5+Los− 6; up < 0.05 and uuup <0.001 for LPS− 5
vs. LPS− 5+Los− 5. LPS, lipopolysaccharide; Los,
losartan.

Fig. 6. Los (10− 6 M) inhibited LPS-induced (10 μg/ml) EGC activation. (A) Analysis of the mRNA expression of GFAP and S100β in each group of EGCs. (B)
Representative image of GFAP protein expression detected by Western blot. (C) Quantitative analysis of the Western blot results for GFAP. Data are presented as the
mean ± SD, n = 3. **p < 0.01 and ***p < 0.001 for Control vs. LPS− 5; ##p < 0.01 and ###p < 0.001 for LPS− 5 vs. Los− 6 or LPS− 5+Los− 6. LPS, lipopolysaccharide;
Los, losartan; GFAP, glial fibrillary acidic protein; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

using qRT‒PCR, Western blotting and ELISA. The results suggested that the protein level, the results showed that LPS treatment significantly
at the mRNA level, both CGRP mRNA and TRPV1 mRNA were signifi­ increased SP protein expression (p < 0.001) and release (p < 0.001), and
cantly increased in EGCs post-LPS treatment (p < 0.001), and Los Los inhibited its expression (p < 0.01) (Fig. 7B and C) and release (p <
significantly decreased the expression of both (p < 0.001) (Fig. 7A). At 0.01) (Fig. 7D).

7
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Fig. 7. Los (10− 6 M) reduced the expression of pain mediators following LPS (10 μg/ml)-induced EGC activation. (A) Analysis of the mRNA expression of CGRP and
TRPV1 in each group of EGCs. (B) Representative image of SP protein expression detected by Western blot. (C) Quantitative analysis of SP by Western blot. (D) The
concentration of SP in each group of cell supernatant by ELISA. Data are presented as the mean ± SD, n = 3. ***p < 0.001 for Control vs. LPS− 5; ##p < 0.01 and ###p
< 0.001 for LPS− 5 vs. Los− 6 or LPS− 5+Los− 6. LPS, lipopolysaccharide; Los, losartan; GFAP, glial fibrillary acidic protein; TRPV1, transient receptor potential vanilloid
1; CGRP, calcitonin gene-related peptide; SP, substance P; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

3.8. Los inhibited LPS-induced upregulation of the ACE1/Ang II/AT1 Some researchers have proposed that infusion of dilute acetic acid in
receptor axis post-EGC activation rats during 8–21 days after birth produces sustained visceral hypersen­
sitivity with little apparent injury or inflammation (Qian et al., 2009).
To investigate whether Los regulates IBS visceral hypersensitivity by The same modelling method was used in this study. Since there are sex
affecting the changes in RAAS components in EGCs, we verified the differences in visceral hypersensitivity in IBS (Mayer et al., 2004), we
results by qRT‒PCR, IF and Western blotting. The results suggested that used only male rats to avoid such differences. After adulthood, rats with
at the mRNA level, the expression levels of ACE1 mRNA, Ang II mRNA acetic acid enema exhibited significantly increased sensitivity to CRD
and AT1 receptor mRNA in EGCs post-LPS treatment were significantly compared to that of the control group, which is consistent with previous
increased (p < 0.001), while Los treatment significantly decreased the findings (Qian et al., 2009), suggesting that the visceral hypersensitivity
expression of the above molecules (p Ang II < 0.05, p ACE1 / AT1 receptor < model of IBS rats was successfully constructed. While different doses of
0.001) (Fig. 8A). At the protein level, by IF, we also found that LPS Los could reduce visceral hypersensitivity in IBS rats, we further
significantly increased the expression level of AT1 receptor in EGCs (p < explored the potential mechanism.
0.001), while Los treatment reversed this effect (p < 0.001) (Fig. 8B and Abnormal activation of EGCs is associated with visceral sensitivity
C). By Western blotting, we further confirmed that LPS treatment and intestinal barrier function (Long et al., 2018; Meira De-Faria et al.,
significantly increased ACE1 protein expression (p < 0.01), while this 2021). One study found increased expression of pro-inflammatory cy­
effect could be inhibited by Los (p < 0.05) (Fig. 8D and E). tokines such as IL-1β and TNF when treating intestinal epithelial cells
with reactive EGC-conditioned medium, which may lead to leaky gut
3.9. Los reduced LPS-induced inflammatory factors expression and microenvironment. Furthermore, the reactive EGC phenotype may also
release post-EGC activation increase DNA breaks through increased inflammation and reactive ox­
ygen species production (Kimono et al., 2019), leading to programmed
Inflammation is closely related to IBS-related visceral hypersensi­ cell death of EGC (Macchioni et al., 2017), which ultimately leads to
tivity; therefore, we finally explored whether Los could reduce the abnormal sensory function from malfunctioning intestinal neurons
production and release of inflammatory factors in EGCs. We found that (Bassotti et al., 2018). In the present study, we found that Los reduced
both TNF mRNA and IL-6 mRNA were significantly increased in EGCs the expression of GFAP and S100β as well as SP, TRPV1 and CGRP in
post-LPS treatment (p TNF < 0.001, p IL-6 < 0.01) by qRT‒PCR assay, and colonic tissues of viscerally hypersensitive rats, suggesting that Los may
Los significantly reduced the expression of both (p < 0.001) (Fig. 9A). alleviate visceral hypersensitivity in IBS rats by inhibiting the activation
Further by ELISA of the cell supernatant, we found that IL-6 release was of EGCs and thus reducing the secretion of pain mediators. This phe­
also significantly increased after EGCs activation (p < 0.001), which was nomenon was further confirmed in in vitro experiments. In addition, we
inhibited by Los (p < 0.001) (Fig. 9B). These results suggest that Los found that cell proliferation viability was significantly inhibited after
improves IBS-related visceral hypersensitivity by reducing the produc­ LPS treatment of EGC, especially at 48h and 72h, while Los significantly
tion and release of inflammatory factors after the activation of EGCs. suppressed this adverse effect. Some previous studies have shown that
LPS stimulates glial cells to proliferate responsively (de Almeida et al.,
4. Discussion 2020), while others have shown that LPS inhibits their proliferation and
induces apoptosis (Letournel-Boulland et al., 1994; Wang et al., 2018;
The acetic acid enema-induced colonic hypo-inflammation model is Xu et al., 2008). It is speculated that the differences between the results
often used to explore the visceral hypersensitivity associated with IBS. of different studies may be due to differences in the time and dose of
The present study shows for the first time that Los administration re­ intervention.
duces visceral hypersensitivity in IBS rats, which may be caused by in­ There is growing evidence that the binding of Ang II to AT1 receptor
hibition of EGC activation in colonic tissues. In combination with is involved in the regulation of pain (Marques-Lopes et al., 2009). Ang II
cellular experiments, it was hypothesized that acetic acid enema has been found to act selectively on non-peptidergic C-fibre neurons,
induced visceral hypersensitivity in relation to IBS, EGC activation and CGRP + nerve endings and myelinated fibres (Benitez et al., 2020). It
RAAS activation and increased Ang II secretion in EGCs and binding to was found that CGRP + neurons in the DRG express AT1 receptors in an
AT1 receptor, resulting in the release of pro-inflammatory cytokines inflammatory environment and that the number of CGRP + neurons was
such as TNF, IL-1β and IL-6 from EGCs and thus aggravating EGC acti­ significantly reduced after Los administration (Benitez et al., 2020). In
vation. At the same time, EGC activation leads to overexpression of the the present experiments, we found that AT1 receptors were co-localized
pain mediators SP, TRPV1, and CGRP, causing visceral hypersensitivity and positively correlated with GFAP in rat colonic tissues, while ACE1
in association with IBS. expression levels were significantly increased in colonic tissues of

8
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Fig. 8. Los (10− 6 M) reduced the expression of RAAS-related molecules following LPS-induced (10 μg/ml) EGC activation. (A) Analysis of the mRNA expression of
Ang II, AT1 receptor, and ACE1 in each group of EGCs. (B) Representative IF images of AT1 receptor expression in each group of EGCs (red fluorescence indicates
AT1 receptor-positive cells; 200 × magnification; scale bar = 100 μm). (C) The mean fluorescence density of AT1 receptor-positive staining was analysed using
ImageJ software. (D) Representative image of Western blot used to detect ACE1 protein expression. (E) Quantitative analysis of Western blot results for ACE1. Data
are presented as the mean ± SD, n = 3. **p < 0.01 and ***p < 0.001 for Control vs. LPS− 5; #p < 0.05, ##p < 0.01 and ###p < 0.001 for LPS− 5 vs. Los− 6 or
LPS− 5+Los− 6. LPS, lipopolysaccharide; Los, losartan; ACE1, angiotensin-converting enzyme 1; Ang II, angiotensin II; AT1R, ang II type 1 receptor; DAPI, 4′ ,6-dia­
midino-2′ -phenylindole; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

9
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Fig. 9. Los (10− 6 M) reduced the level of inflamma­


tory factors after LPS-induced (10 μg/ml) EGC acti­
vation. (A) Analysis of mRNA expression of the
inflammatory factors TNF and IL-6 in each group of
EGCs. (B) The concentration of IL-6 in each group of
cell supernatant by ELISA. Data are presented as the
mean ± SD, n = 3. **p < 0.01 and ***p < 0.001 for
Control vs. LPS− 5; ##p < 0.01 and ###p < 0.001 for
LPS− 5 vs. Los− 6 or LPS− 5+Los− 6. LPS, lipopolysac­
charide; Los, losartan; TNF, tumor necrosis factor; IL-
6, interleukin-6.

visceral hypersensitive rats compared to control rats, which combined hypersensitivity in relation to IBS. In addition, it was found that in­
with increased expression of ACE1, AngII and AT1R in activated EGCs in flammatory cytokines activate p38 MAPK, leading to increased expres­
in vitro experiments, suggesting the presence of AT1 receptor expression sion of injurious ion channels and transport to peripheral nerves, leading
in colonic tissue EGCs of visceral hypersensitive rats and upregulation of to long-term afferent nerve sensitization and inflammatory pain (Ji
the ACE1/Ang II/AT1 receptor axis. One study revealing that the et al., 2002; Linley et al., 2010). In contrast, Los attenuates oxidative
ACE1/Ang II/AT1 receptor axis is highly expressed in inflammatory stress and reduces neuroinflammation and cell death (Abdul-Muneer
bowel disease colon tissue (Jacobs et al., 2019), and that low inflam­ et al., 2018). In this study, Los inhibited the elevation of IL-1β, IL-6 and
mation is also present in IBS colon tissue (Grabauskas et al., 2022) seems TNF in colonic tissues of viscerally hypersensitive rats and EGCs.
to provide evidence supporting the possible involvement of colonic local Therefore, we speculate that Los attenuates the inflammatory response
RAAS in the development of IBS. Pain mediators and inflammatory and inhibits EGC activation by blocking the ACE1/Ang II/AT1 receptor
factor signals in the gut wall are sensed by extrinsic, primary afferent axis and consequently reduces visceral hypersensitivity in IBS rats,
neurons with cell bodies in the DRG. The signals are transduced via which may involve inhibition of MAPK and NF-κBp65 pathways. Su­
secondary neurons in the spinal cord to different somatosensory areas in pernatants from mucosal biopsies of IBS patients were found to sensitize
the brain and are processed by the central nervous system to produce the colon-injurious DRG neurons (Valdez-Morales et al., 2013; Wouters
corresponding sensations (Vermeulen et al., 2014). Sensitization of ion et al., 2016), and blocking AT1 receptor attenuates the inflammatory
channels such as TRPV1 in submucosal neurons of the colon that response to DRG (Kalynovska et al., 2020; Kim et al., 2019). Therefore,
perceive toxic stimuli by bioactive substances released within the gut losartan may alleviate visceral hypersensitivity by attenuating the in­
wall and the release of pain mediators such as CGRP and SP can lead to flammatory response in the central nervous system.
visceral hypersensitivity (Anand et al., 2007; Tang et al., 2022, 2022van We verified the protective effect of Los on visceral hypersensitivity in
Wanrooij et al., 2014; Wouters et al., 2016). In the present study, we IBS rats by in vivo treatment with different doses of Los and in vitro ex­
found that Los decreased the expression of ACE1 as well as pain medi­ periments. However, there are still shortcomings in this study. In the
ators TRPV1, CGRP and SP in colonic tissue of visceral hypersensitive present experiment, rats were treated with 10, 25, and 50 mg/kg/d, the
rats and EGC, and it is reasonable to speculate that Los may attenuate usual doses of Los in previous studies, and although all three doses
visceral afferent nerve sensitivity by inhibiting AT1 receptors to reduce showed protective effects, they did not exhibit a significant dose-
visceral hypersensitivity. dependence, presumably related to the complex visceral hypersensitiv­
Inflammation can induce and sustain pain development, and the ity mechanism of IBS and insufficient sample size. Since the mechanism
release of inflammatory factors and chemokines may sensitize periph­ of visceral hypersensitivity associated with IBS is complex, including
eral nerves and participate in pain development, with these substances neuroendocrine mediators, intestinal flora and mucosal hypo-
being further released by the glial cells themselves (Ebersberger, 2018). inflammation, the results of this study are limited to the rat model of
It has been shown that in a rat model of nerve crush and transection, AT1 acetic acid enema and need to be further validated in other IBS models
receptor blockers can improve nerve healing and thus relieve neuro­ and different species, including clinical specimens.
pathic pain by inhibiting the production of IL-1β and caspase-3 (Yuksel
et al., 2015). The MAPK and NF-κBp65 pathways are two classical in­ 5. Conclusion
flammatory pathways. ACE1/Ang II/AT1 receptor axis activation is
usually accompanied by activation of the MAPK pathway (Wenzel et al., The present study shows for the first time that the ACE1/Ang II/AT1
2001), and MAPK pathway activation induces and exacerbates inflam­ receptor axis is upregulated in the colon tissue EGCs of rats with IBS-
matory responses and pain development (Ji et al., 2009; PARK et al., related visceral hypersensitivity and that Los significantly ameliorates
2009). Los has been found to reduce tissue inflammatory damage by the visceral hypersensitivity induced by acetic acid enema, possibly by
decreasing MAPK and NF-κBp65 activation in a variety of inflammatory mediating AT1 receptor on colon tissue EGCs. Therefore, inhibition of
injury-related models (Abdel-Latif et al., 2020; Wang et al., 2019). Los the ACE1/Ang II/AT1 receptor axis may be an effective therapeutic
can also have an analgesic effect on neuropathic pain by inhibiting target for alleviating visceral hypersensitivity in relation to IBS, which
MAPK and NF-κBp65 activation and the production of inflammatory provides a new therapeutic strategy for the clinical treatment of IBS.
factors such as TNF and IL-6 (Hegazy et al., 2020b; Kalynovska et al.,
2020). Many studies have suggested that inflammatory factors such as Ethics statement
TNF, IL-1β, and IL-6 are significantly increased in colorectal tissues of
IBS rats (Gwee et al., 2003; Lee et al., 2017), and the hyperinflammatory The animal study was reviewed and approved by the Ethics Com­
state can activate EGCs to further secrete inflammatory factors (Stoffels mittee of Xi’an Jiaotong University.
et al., 2014), sensitizing the ENS and ultimately promoting visceral

10
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Funding de Almeida, M., Souza, C., Dourado, N.S., Da, S.A., Ferreira, R.S., David, J.M., David, J.
P., Costa, M., Da, S.V., Butt, A.M., Costa, S.L., 2020. Phytoestrogen agathisflavone
ameliorates neuroinflammation-induced by LPS and IL-1β and protects neurons in
The research was funded by National Natural Science Foundation of cocultures of glia/neurons. Biomolecules 10. https://doi.org/10.3390/
China (NSFC: 81770540). biom10040562.
Ebersberger, A., 2018. The analgesic potential of cytokine neutralization with
biologicals. Eur. J. Pharmacol. 835, 19–30. https://doi.org/10.1016/j.
CRediT authorship contribution statement ejphar.2018.07.040.
Grabauskas, G., Gao, J., Wu, X., Zhou, S.Y., Turgeon, D.K., Owyang, C., 2022. Gut
microbiota alter visceral pain sensation and inflammation via modulation of
Yating Sun: Conceptualization, Data curation, Formal analysis, synthesis of resolvin D1 in colonic tuft cells. Gastroenterology. https://doi.org/
Investigation, Writing – original draft. Xiaohui Liu: Investigation, 10.1053/j.gastro.2022.07.053.
Methodology, Validation, Writing – original draft. Lianli Wang: Inves­ Grubišić, V., Gulbransen, B.D., 2017. Enteric glia: the most alimentary of all glia.
J. Physiol. 595, 557–570. https://doi.org/10.1113/JP271021.
tigation, Validation, Software. Laifu Li: Investigation, Data curation.
Gwee, K., Collins, S.M., Read, N.W., Rajnakova, A., Deng, Y., Graham, J.C.,
Xiaojing Quan: Formal analysis, Visualization. Haitao Shi: Conceptu­ McKendrick, M.W., Moochhala, S.M., 2003. Increased rectal mucosal expression of
alization, Supervision. Ting Wang: Conceptualization, Visualization. interleukin 1β in recently acquired post-infectious irritable bowel syndrome. Gut 52,
Lin Mei: Methodology, Software. Yindi Chen: Supervision. Yue Zhang: 523. https://doi.org/10.1136/gut.52.4.523.
Hegazy, N., Rezq, S., Fahmy, A., 2020a. Renin-angiotensin system blockade modulates
Methodology. Jingyao Li: Visualization. Ruiting Meng: Validation. both the peripheral and central components of neuropathic pain in rats: role of
Jinhai Wang: Writing – review & editing. Fei Dai: Conceptualization, calcitonin gene–related peptide, substance P and nitric oxide. Basic Clin Pharmacol
Supervision, Writing – review & editing, Funding acquisition. 127, 451–460. https://doi.org/10.1111/bcpt.13453.
Hegazy, N., Rezq, S., Fahmy, A., 2020b. Mechanisms involved in superiority of
angiotensin receptor blockade over ACE inhibition in attenuating neuropathic pain
induced in rats. Neurotherapeutics 17, 1031–1047. https://doi.org/10.1007/
Declaration of competing interest s13311-020-00912-8.
Jacobs, J.D., Wagner, T., Gulotta, G., Liao, C., Li, Y.C., Bissonnette, M., Pekow, J., 2019.
Impact of angiotensin II signaling blockade on clinical outcomes in patients with
The authors declare that the research was conducted in the absence
inflammatory bowel disease. Digest. Dis. Sci. 64, 1938–1944. https://doi.org/
of any commercial or financial relationships that could be construed as a 10.1007/s10620-019-5474-4.
potential conflict of interest. Ji, R., Gereau, R.W., Malcangio, M., Strichartz, G.R., 2009. MAP kinase and pain. Brain
Res. Rev. 60, 135–148. https://doi.org/10.1016/j.brainresrev.2008.12.011.
Ji, R., Samad, T.A., Jin, S., Schmoll, R., Woolf, C.J., 2002. p38 MAPK activation by NGF
Data availability in primary sensory neurons after inflammation increases TRPV1 levels and maintains
heat hyperalgesia. Neuron 36, 57–68. https://doi.org/10.1016/S0896-6273(02)
Data will be made available on request. 00908-X.
Kalynovska, N., Diallo, M., Sotakova-Kasparova, D., Palecek, J., 2020. Losartan
attenuates neuroinflammation and neuropathic pain in paclitaxel-induced peripheral
Acknowledgements neuropathy. J. Cell Mol. Med. 24, 7949–7958. https://doi.org/10.1111/
jcmm.15427.
Karnik, S.S., Unal, H., Kemp, J.R., Tirupula, K.C., Eguchi, S., Vanderheyden, P.M.L.,
Special thanks to the Brain Science Research Center of the First Thomas, W.G., Ohlstein, E.H., 2015. International union of basic and clinical
Affiliated Hospital of Xi’an Jiaotong University for providing us with pharmacology. XCIX. Angiotensin receptors: interpreters of pathophysiological
EGC (ATCC). angiotensinergic stimuli. Pharmacol. Rev. 67, 754. https://doi.org/10.1124/
pr.114.010454.
Keszthelyi, D., Troost, F.J., Masclee, A.A., 2012. Irritable Bowel Syndrome: methods,
References Mechanisms, and Pathophysiology. Methods to assess visceral hypersensitivity in
irritable bowel syndrome. Am J PhysioL-Gastr l 303, G141–G154. https://doi.org/
10.1152/ajpgi.00060.2012.
Abdel-Latif, G.A., Elwahab, A.H.A., Hasan, R.A., ElMongy, N.F., Ramzy, M.M., Louka, M.
Kim, E., Hwang, S., Kim, H., Abdi, S., Kim, H.K., 2019. Losartan, an angiotensin II type 1
L., Schaalan, M.F., 2020. A novel protective role of sacubitril/valsartan in
receptor antagonist, alleviates mechanical hyperalgesia in a rat model of
cyclophosphamide induced lung injury in rats: impact of miRNA-150-3p on NF-κB/
chemotherapy-induced neuropathic pain by inhibiting inflammatory cytokines in the
MAPK signaling trajectories. SCI REP-UK 10, 13045. https://doi.org/10.1038/
dorsal root ganglia. Mol. Neurobiol. 56, 7408–7419. https://doi.org/10.1007/
s41598-020-69810-5.
s12035-019-1616-0.
Abdul-Muneer, P.M., Bhowmick, S., Briski, N., 2018. Angiotensin II causes neuronal
Kimono, D., Sarkar, S., Albadrani, M., Seth, R., Bose, D., Mondal, A., Li, Y., Kar, A.N.,
damage in stretch-injured neurons: protective effects of losartan, an angiotensin T1
Nagarkatti, M., Nagarkatti, P., Sullivan, K., Janulewicz, P., Lasley, S., Horner, R.,
receptor blocker. Mol. Neurobiol. 55, 5901–5912. https://doi.org/10.1007/s12035-
Klimas, N., Chatterjee, S., 2019. Dysbiosis-associated enteric glial cell immune-
017-0812-z.
activation and redox imbalance modulate tight junction protein expression in gulf
Akazawa, Y., Fujioka, T., Ide, H., Yazaki, K., Honjo, O., Sun, M., Friedberg, M.K., 2021.
war illness pathology. Front. Physiol. 10, 1229. https://doi.org/10.3389/
Impaired right and left ventricular function and relaxation induced by pulmonary
fphys.2019.01229. eCollection 2019.
regurgitation are not reversed by tardive antifibrosis treatment. Am J Physiol-Heart
Lee, J.Y., Kim, N., Nam, R.H., Sohn, S.H., Lee, S.M., Choi, D., Yoon, H., Kim, Y.S., Lee, H.
C 321, H38–H51. https://doi.org/10.1152/ajpheart.00467.2020.
S., Lee, D.H., 2017. Probiotics reduce repeated water avoidance stress-induced
Akbar, A., Yiangou, Y., Facer, P., Walters, J.R.F., Anand, P., Ghosh, S., 2008. Increased
colonic microinflammation in Wistar rats in a sex-specific manner. PLoS One 12,
capsaicin receptor TRPV1-expressing sensory fibres in irritable bowel syndrome and
e188992. https://doi.org/10.1371/journal.pone.0188992.
their correlation with abdominal pain. Gut 57, 923. https://doi.org/10.1136/
Letournel-Boulland, M.L., Fages, C., Rolland, B., Tardy, M., 1994. Lipopolysaccharides
gut.2007.138982.
(LPS), up-regulate the IL-1-mRNA and down-regulate the glial fibrillary acidic
Anand, P., Aziz, Q., Willert, R., Van Oudenhove, L., 2007. Peripheral and central
protein (GFAP) and glutamine synthetase (GS)-mRNAs in astroglial primary cultures.
mechanisms of visceral sensitization in man. Neuro Gastroenterol. Motil. 19, 29–46.
Eur. Cytokine Netw. 5, 51–56.
https://doi.org/10.1111/j.1365-2982.2006.00873.x.
Linley, J.E., Rose, K., Ooi, L., Gamper, N., 2010. Understanding inflammatory pain: ion
Anand, U., Yiangou, Y., Sinisi, M., Fox, M., MacQuillan, A., Quick, T., Korchev, Y.E.,
channels contributing to acute and chronic nociception. Pflueg. Arch. Eur. J. Physiol.
Bountra, C., McCarthy, T., Anand, P., 2015. Mechanisms underlying clinical efficacy
459, 657–669. https://doi.org/10.1007/s00424-010-0784-6.
of angiotensin II type 2 receptor (AT2R) antagonist EMA401 in neuropathic pain:
Long, X., Li, M., Li, L.X., Sun, Y.Y., Zhang, W.X., Zhao, D.Y., Li, Y.Q., 2018. Butyrate
clinical tissue and in vitro studies. Mol. Pain 11, s12915–s12990. https://doi.org/
promotes visceral hypersensitivity in an IBS-like model via enteric glial cell-derived
10.1186/s12990-015-0038-x.
nerve growth factor. Neuro Gastroenterol. Motil. 30, e13227 https://doi.org/
Bali, A., Singh, N., Jaggi, A.S., 2014. Renin–angiotensin system in pain: existing in a
10.1111/nmo.13227.
double life? J RENIN-ANGIO-ALDO S 15, 329–340. https://doi.org/10.1177/
Macchioni, L., Davidescu, M., Fettucciari, K., Petricciuolo, M., Gatticchi, L., Gioè, D.,
1470320313503694.
Villanacci, V., Bellini, M., Marconi, P., Roberti, R., Bassotti, G., Corazzi, L., 2017.
Bassotti, G., Macchioni, L., Corazzi, L., Marconi, P., Fettucciari, K., 2018. Clostridium
Enteric glial cells counteract Clostridium difficile Toxin B through a NADPH
difficile-related postinfectious IBS: a case of enteroglial microbiological stalking
oxidase/ROS/JNK/caspase-3 axis, without involving mitochondrial pathways. SCI
and/or the solution of a conundrum? Cell. Mol. Life Sci. 75, 1145–1149. https://doi.
REP-UK 7, 45569. https://doi.org/10.1038/srep45569.
org/10.1007/s00018-017-2736-1.
Marques-Lopes, J., Pinto, M., Pinho, D., Morato, M., Patinha, D., Albino-Teixeira, A.,
Benitez, S.G., Seltzer, A.M., Messina, D.N., Foscolo, M.R., Patterson, S.I., Acosta, C.G.,
Tavares, I., 2009. Microinjection of angiotensin II in the caudal ventrolateral
2020. Cutaneous inflammation differentially regulates the expression and function
medulla induces hyperalgesia. Neuroscience 158, 1301–1310. https://doi.org/
of Angiotensin-II types 1 and 2 receptors in rat primary sensory neurons.
10.1016/j.neuroscience.2008.11.044.
J. Neurochem. 152, 675–696. https://doi.org/10.1111/jnc.14848.
Collister, J.P., Hornfeldt, B.J., Osborn, J.W., 1996. Hypotensive response to losartan in
normal rats. Hypertension 27, 598–606. https://doi.org/10.1161/01.HYP.27.3.598.

11
Y. Sun et al. European Journal of Pharmacology 946 (2023) 175650

Mayer, E.A., Berman, S., Chang, L., Naliboff, B.D., 2004. Sex-based differences in predominant irritable bowel syndrome patients: a role for PAR2. Am. J.
gastrointestinal pain. Eur. J. Pain 8, 451–463. https://doi.org/10.1016/j. Gastroenterol. 108, 1634–1643. https://doi.org/10.1038/ajg.2013.241.
ejpain.2004.01.006. van Wanrooij, S.J., Wouters, M.M., Van Oudenhove, L., Vanbrabant, W., Mondelaers, S.,
Meira De-Faria, F., Casado-Bedmar, M., Mårten Lindqvist, C., Jones, M.P., Walter, S.A., Kollmann, P., Kreutz, F., Schemann, M., Boeckxstaens, G.E., 2014. Sensitivity testing
Keita, Å.V., 2021. Altered interaction between enteric glial cells and mast cells in the in irritable bowel syndrome with rectal capsaicin stimulations: role of TRPV1
colon of women with irritable bowel syndrome. Neuro Gastroenterol. Motil. 33, upregulation and sensitization in visceral hypersensitivity? Am. J. Gastroenterol.
e14130 https://doi.org/10.1111/nmo.14130. 109, 99–109. https://doi.org/10.1038/ajg.2013.371.
Nemoto, W., 2018. Behavioral and molecular pharmacological study of the role of Vermeulen, W., De Man, J.G., Pelckmans, P.A., De Winter, B.Y., 2014. Neuroanatomy of
angiotensin II in spinal pain transmission. Yakugaku Zasshi 138, 1235–1240. lower gastrointestinal pain disorders. World J. Gastroenterol. 20, 1005–1020.
https://doi.org/10.1248/yakushi.18-00124. https://doi.org/10.3748/wjg.v20.i4.1005.
Nozu, T., Miyagishi, S., Nozu, R., Takakusaki, K., Okumura, T., 2017. Repeated water Wang, G., Wang, X., Hu, H., Fang, X., Liu, S., Gao, N., Xia, Y., Wood, J.D., 2005.
avoidance stress induces visceral hypersensitivity: role of interleukin-1, interleukin- Angiotensin receptors and actions in Guinea pig enteric nervous system. Am J
6, and peripheral corticotropin-releasing factor. J Gastroen Hepatol. 32, 1958–1965. PhysioL-Gastr l 289, G614–G626. https://doi.org/10.1152/ajpgi.00119.2005.
https://doi.org/10.1111/jgh.13787. Wang, P., Du, C., Chen, F., Li, C., Yu, Y., Han, T., Akhtar, S., Zuo, X., Tan, X., Li, Y., 2016.
Obukhov, A.G., Stevens, B.R., Prasad, R., Li Calzi, S., Boulton, M.E., Raizada, M.K., BDNF contributes to IBS-like colonic hypersensitivity via activating the enteroglia-
Oudit, G.Y., Grant, M.B., 2020. SARS-CoV-2 infections and ACE2: clinical outcomes nerve unit. SCI REP-UK 6, 20320. https://doi.org/10.1038/srep20320.
linked with increased morbidity and mortality in individuals with diabetes. Diabetes Wang, S., Zhang, H., Geng, B., Xie, Q., Li, W., Deng, Y., Shi, W., Pan, Y., Kang, X.,
69, 1875–1886. https://doi.org/10.2337/dbi20-0019. Wang, J., 2018. 2-arachidonyl glycerol modulates astrocytic glutamine synthetase
Ogata, Y., Nemoto, W., Nakagawasai, O., Yamagata, R., Tadano, T., Tan-No, K., 2016. via p38 and ERK1/2 pathways. J. Neuroinflamm. 15, 220. https://doi.org/10.1186/
Involvement of spinal angiotensin II system in streptozotocin-induced diabetic s12974-018-1254-x.
neuropathic pain in mice. Mol. Pharmacol. 90, 205. https://doi.org/10.1124/ Wang, X., Chen, X., Huang, W., Zhang, P., Guo, Y., Körner, H., Wu, H., Wei, W., 2019.
mol.116.104133. Losartan suppresses the inflammatory response in collagen-induced arthritis by
Park, H.S., Kim, S.R., Lee, Y.C., 2009. Impact of oxidative stress on lung diseases. inhibiting the MAPK and NF-κB pathways in B and T cells. Inflammopharmacology
Respirology 14, 27–38. https://doi.org/10.1111/j.1440-1843.2008.01447.x. 27, 487–502. https://doi.org/10.1007/s10787-018-0545-2.
Patil, J., Schwab, A., Nussberger, J., Schaffner, T., Saavedra, J.M., Imboden, H., 2010. Wenzel, S., Taimor, G., Piper, H.M., Schlüeter, K., 2001. Redox-sensitve intermediates
Intraneuronal angiotensinergic system in rat and human dorsal root ganglia. Regul. mediate angiotensin II-induced p38 MAP kinase activation, AP-1 binding activity,
Pept. 162, 90–98. https://doi.org/10.1016/j.regpep.2010.03.004. and TGF-β expression in adult ventricular cardiomyocytes. Faseb. J. 15, 1–23.
Pusceddu, M.M., Gareau, M.G., 2018. Visceral pain: gut microbiota, a new hope? https://doi.org/10.1096/fj.00-0827fje.
J. Biomed. Sci. 25, 73. https://doi.org/10.1186/s12929-018-0476-7. Wouters, M.M., Balemans, D., Van Wanrooy, S., Dooley, J., Cibert-Goton, V., Alpizar, Y.
Qian, A., Liu, X., Yao, W., Wang, H., Sun, J., Zhou, L., Yuan, Y., 2009. Voltage-gated A., Valdez-Morales, E.E., Nasser, Y., Van Veldhoven, P.P., Vanbrabant, W., Van der
potassium channels in IB4-positive colonic sensory neurons mediate visceral Merwe, S., Mols, R., Ghesquière, B., Cirillo, C., Kortekaas, I., Carmeliet, P.,
hypersensitivity in the rat. Off. J. Am. College Gastroenterol. ¦ ACG 104, 2014–2027. Peetermans, W.E., Vermeire, S., Rutgeerts, P., Augustijns, P., Hellings, P.W.,
https://doi.org/10.1038/ajg.2009.227. Belmans, A., Vanner, S., Bulmer, D.C., Talavera, K., Vanden Berghe, P., Liston, A.,
Sengupta, J.N., 2009. Visceral pain: the neurophysiological mechanism. In: Canning, B. Boeckxstaens, G.E., 2016. Histamine receptor H1–mediated sensitization of TRPV1
J., Spina, D. (Eds.), Sensory Nerves. Springer Berlin Heidelberg, Berlin, Heidelberg, mediates visceral hypersensitivity and symptoms in patients with irritable bowel
pp. 31–74. https://doi.org/10.1007/978-3-540-79090-7_2. syndrome. Gastroenterology 150, 875–887. https://doi.org/10.1053/j.
Shi, H., Liu, C., Ding, L., Zheng, Y., Fei, X., Lu, L., Zhou, X., Yuan, J., Xie, J., 2015. gastro.2015.12.034.
Alterations in serotonin, transient receptor potential channels and protease-activated Xiu, J.C., Wu, P., Xu, J.P., Guo, Z., Lai, W., Zhang, Y., Li, S., Li, J., Liu, Y., 2002. Effects of
receptors in rats with irritable bowel syndrome attenuated by Shugan decoction. long-term enalapril and losartan therapy of heart failure on cardiovascular
World J. Gastroenterol.: WJG 21, 4852. https://doi.org/10.3748/wjg.v21.i16.4852. aldosterone. J. Endocrinol. Invest. 25, 463–468. https://doi.org/10.1007/
Shi, Y., Liu, T., He, L., Dougherty, U., Chen, L., Adhikari, S., Alpert, L., Zhou, G., Liu, W., BF03344039.
Wang, J., Deb, D.K., Hart, J., Liu, S.Q., Kwon, J., Pekow, J., Rubin, D.T., Zhao, Q., Xu, X., Kim, J.A., Zuo, Z., 2008. Isoflurane preconditioning reduces mouse microglial
Bissonnette, M., Li, Y.C., 2016. Activation of the renin-angiotensin system promotes activation and injury induced by lipopolysaccharide and interferon-γ. Neuroscience
colitis development. SCI REP-UK 6, 27552. https://doi.org/10.1038/srep27552. 154, 1002–1008. https://doi.org/10.1016/j.neuroscience.2008.04.013.
Stoffels, B., Hupa, K.J., Snoek, S.A., van Bree, S., Stein, K., Schwandt, T., Vilz, T.O., Xu, X., Wang, H., Xing, C., Gu, Y., Liu, S., Xu, H., Hu, M., Song, L., 2017. IRE1α/XBP1s
Lysson, M., Veer, C.V., Kummer, M.P., Hornung, V., Kalff, J.C., de Jonge, W.J., branch of UPR links HIF1α activation to mediate ANGII-dependent endothelial
Wehner, S., 2014. Postoperative ileus involves interleukin-1 receptor signaling in dysfunction under particulate matter (PM) 2.5 exposure. SCI REP-UK 7, 1–16.
enteric glia. Gastroenterology 146, 176–187. https://doi.org/10.1053/j. https://doi.org/10.1038/s41598-017-13156-y.
gastro.2013.09.030. Yamamoto, S., Zhong, J., Yancey, P.G., Zuo, Y., Linton, M.F., Fazio, S., Yang, H.,
Tang, H.Y., Chen, X.Q., Wang, H., Chu, H.R., Zhu, C.F., Huang, S., Zhang, M.T., Shen, G. Narita, I., Kon, V., 2015. Atherosclerosis following renal injury is ameliorated by
M., 2022. Acupuncture relieves the visceral pain of diarrhea-predominant irritable pioglitazone and losartan via macrophage phenotype. Atherosclerosis 242, 56–64.
bowel syndrome rats by regulating P2X4 expression. Am. J. Transl. Res. 14, https://doi.org/10.1016/j.atherosclerosis.2015.06.055.
5563–5573. Yuksel, T.N., Halici, Z., Demir, R., Cakir, M., Calikoglu, C., Ozdemir, G., Unal, D., 2015.
Valdez-Morales, E.E., Overington, J., Guerrero-Alba, R., Ochoa-Cortes, F., Ibeakanma, C. Investigation of the effect of telmisartan on experimentally induced peripheral nerve
O., Spreadbury, I., Bunnett, N.W., Beyak, M., Vanner, S.J., 2013. Sensitization of injury in rats. Int. J. Neurosci. 125, 464–473. https://doi.org/10.3109/
peripheral sensory nerves by mediators from colonic biopsies of diarrhea- 00207454.2014.948115.

12

You might also like