ArticlePDF Available

Visceral hyperalgesia caused by peptide YY deletion and Y2 receptor antagonism OPEN

Authors:

Abstract and Figures

Altered levels of colonic peptide YY (PYY) have been reported in patients suffering from functional and inflammatory bowel disorders. While the involvement of neuropeptide Y (NPY) and Y receptors in the regulation of nociception is well established, the physiological role of PYY in somatic and visceral pain is poorly understood. In this work, the role of PYY in pain sensitivity was evaluated using PYY knockout (PYY(−/−)) mice and Y2 receptor ligands. PYY(−/−) mice were more sensitive to somatic thermal pain compared to wild type (WT) mice. Visceral pain was assessed by evaluating pain-related behaviors, mouse grimace scale (MGS) and referred hyperalgesia after intrarectal administration of allyl isothiocyanate (AITC, 1 or 2%) or its vehicle, peanut oil. The pain-related behaviors induced by AITC were significantly exaggerated by PYY deletion, whereas the MGS readout and the referred hyperalgesia were not significantly affected. The Y2 receptor antagonist, BII0246, increased pain-related behaviors in response to intrarectal AITC compared to vehicle treatment while the Y2 receptor agonist, PYY(3–36), did not have a significant effect. These results indicate that endogenous PYY has a hypoalgesic effect on somatic thermal and visceral chemical pain. The effect on visceral pain seems to be mediated by peripheral Y2 receptors.
Content may be subject to copyright.
1
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
www.nature.com/scientificreports
Visceral hyperalgesia caused by
peptide YY deletion and Y2 receptor
antagonism
Ahmed M. Hassan1,*, Piyush Jain1,*, Raphaela Mayerhofer1, Esther E. Fröhlich1, Aitak Farzi1,
Florian Reichmann1, Herbert Herzog2 & Peter Holzer1
Altered levels of colonic peptide YY (PYY) have been reported in patients suering from functional
and inammatory bowel disorders. While the involvement of neuropeptide Y (NPY) and Y receptors in
the regulation of nociception is well established, the physiological role of PYY in somatic and visceral
pain is poorly understood. In this work, the role of PYY in pain sensitivity was evaluated using PYY
knockout (PYY(/)) mice and Y2 receptor ligands. PYY(/) mice were more sensitive to somatic
thermal pain compared to wild type (WT) mice. Visceral pain was assessed by evaluating pain-related
behaviors, mouse grimace scale (MGS) and referred hyperalgesia after intrarectal administration of
allyl isothiocyanate (AITC, 1 or 2%) or its vehicle, peanut oil. The pain-related behaviors induced by
AITC were signicantly exaggerated by PYY deletion, whereas the MGS readout and the referred
hyperalgesia were not signicantly aected. The Y2 receptor antagonist, BII0246, increased pain-
related behaviors in response to intrarectal AITC compared to vehicle treatment while the Y2 receptor
agonist, PYY(3–36), did not have a signicant eect. These results indicate that endogenous PYY has a
hypoalgesic eect on somatic thermal and visceral chemical pain. The eect on visceral pain seems to be
mediated by peripheral Y2 receptors.
e gut hormone peptide YY (PYY) is a member of the neuropeptide Y (NPY) family which also includes pan-
creatic polypeptide. PYY release is regulated by food intake, the parasympathetic nervous system, inammatory
mediators, and other gut hormones including cholecystokinin, vasoactive intestinal polypeptide, gastrin, and
glucagon-like peptide-11–4. PYY is secreted by L-cells of the gastrointestinal tract (GIT) which release it in the
form of PYY(1–36). e amino acids tyrosine and proline are cleaved from the N-terminus of the polypeptide by
the dipeptidyl peptidase IV (DDP-IV) enzyme to produce PYY(3–36). is changes the pharmacological proper-
ties of the peptide. While PYY(1–36) binds to all Y receptor subtypes, PYY(3–36) has a higher anity to the Y2
subtype3. e two peptides have endocrine actions in the GIT where they act as brakes to inhibit gastric emptying,
intestinal motility, mouth to anus transit time, and electrolyte secretion in the intestine1,2.
In addition to regulation of GIT functions and food intake, the NPY system is involved in the regulation of
several physiological functions including stress coping, emotional-aective behavior, cognition, neurogenesis,
immunity, and nociception5. Knockout of Y1 receptors is associated with thermal, chemical, and mechanical
somatic hyperalgesia and exaggerated acetic acid- and MgSO4 -induced visceral pain5,6. Electrophysiological
studies conrm a role of spinal Y1 and Y2 receptors in controlling postsynaptic currents in pain signaling path-
ways7,8. NPY acting through Y1 receptors controls the production of several neuropeptides including substance P
and calcitonin gene-related peptide (CGRP), which subserve pronociceptive functions in the CNS9. In addition to
their eect on spinal pain pathways, Y receptor ligands aect visceral pain mediated by vagal aerent pathways, as
c-Fos expression in the nucleus tractus solitarii (NTS) following intragastric acid challenge is enhanced in Y2 and
Y4 receptor knockout mice10. Furthermore, Y1 receptors are involved in the descending control of pain signaling.
For example, NPY injection into the arcuate nucleus reduces somatic pain in rats, an eect that is blocked by Y1
but not Y2 receptor antagonists11. In spite of the well-established role of NPY in the regulation of pain sensitivity,
the role of the gut hormone PYY in nociception is poorly understood.
1Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology,
Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria. 2Neurobiology Research Program, Garvan
Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia. *These authors
contributed equally to this work. Correspondence and requests for materials should be addressed to P.H. (email:
peter.holzer@medunigraz.at)
Received: 13 September 2016
Accepted: 13 December 2016
Published: 20 January 2017
OPEN
www.nature.com/scientificreports/
2
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
Expression of colonic PYY is altered in patients suering from functional and inammatory bowel disorders.
e density of PYY-containing cells in the colon is lowered in patients with irritable bowel syndrome (IBS)12,13
and Crohn’s disease (CD)14,15. Similarly, patients suering from ulcerative colitis (UC) have lower rectal PYY
levels compared to healthy controls16. Given that IBS and inammatory bowel disease (IBD) are associated with
pain, we hypothesized that a deciency in PYY contributes to the hyperalgesia associated with these pathologies.
To examine a possible role of PYY in nociception, we rst investigated somatic and visceral pain percep-
tion in PYY knockout (PYY(/)) mice. Somatic sensitivity to thermal pain was tested with the plantar test17
while visceral sensitivity to chemical pain was quantied by the pain reactions to intrarectal allyl isothiocyanate
(AITC)18,19. e visceral pain readouts included spontaneous pain-related behaviors, locomotor activity, facial
pain expressions, and referred hyperalgesia18–20. Concomitantly, the expression of NPY, Y1, and Y2 receptor
mRNA in the spinal cord of wild type (WT) and PYY(/) mice was assessed. Since PYY(3–36), the main circu-
latory form of PYY, has a preferential anity to Y2 receptors3, the eects of the Y2 receptor agonist, PYY(3–36),
and the Y2 receptor antagonist, BIIE0246, on visceral pain sensitivity were also evaluated.
Results
PYY(/) mice are hypersensitive to thermal pain. Compared to WT mice, PYY(/) mice were more
sensitive to thermal pain as indicated by a signicantly shorter withdrawal latency in the plantar test (t19.5 = 3.3;
p = 0.004) (Fig.1).
PYY knockout exaggerates pain-related behaviors induced by intrarectal AITC. Following
intrarectal instillation of PO or AITC (2%), one-way ANOVA revealed statistically signicant dierences in
pain-related behaviors (F(3,24) = 4.6; p = 0.011) and time spent freezing (F(3,24) = 4.6; p = 0.003) while the dif-
ferences in the latency to rst pain-related behavior and time spent grooming were not statistically signicant
(Fig.2). Post-hoc testing revealed that pain-related behaviors (stretching, squashing, arching and licking of the
abdomen) were enhanced in PYY(/) mice that received AITC (PYY(/) + AITC group) compared to WT mice
that received PO (WT + PO group) and WT mice that received AITC (WT + AITC group) (Fig.2A). e time
spent freezing was prolonged in both the WT + AITC and the PYY(/) + AITC groups relative to the WT + PO
group (Fig.2B). None of the parameters recorded in the LabMaster system (horizontal activity, vertical activity,
traveling distance) during the rst 15 min aer intrarectal instillation of PO or AITC were signicantly dierent
between the experimental groups (Supplement;FigureS1).
PYY knockout does not alter AITC-induced facial pain expressions. Following intrarectal instil-
lation of PO or AITC (2%), one-way ANOVA revealed statistically signicant dierences in Δ MGS among the
experimental groups ((F(3,23) = 53.1; p < 0.001) (Fig.3). Post hoc testing revealed that the Δ MGS in the groups
which received AITC was signicantly higher than in the groups that received PO while other dierences between
the groups were statistically not signicant (Fig.3).
PYY knockout does not alter AITC-induced referred hyperalgesia. e mechanical pain threshold
(MPT) on the plantar surface of the hind paws was measured with von Frey laments in WT and PYY(/) mice
before and aer intrarectal treatment with PO or AITC (1%). e baseline value reects somatic sensitivity to
mechanical pain while the dierence between the pre- and post-treatment measurements was taken as index
of referred hyperalgesia which accompanies visceral pain. One-way ANOVA failed to disclose any statistically
signicant dierences in MPT among the experimental groups both under baseline conditions and aer intra-
rectal administration of PO and AITC (Fig.4A,B). A comparison of baseline and post-treatment recordings
Figure 1. Eect of PYY knockout (PYY(/)) on withdrawal latency in the plantar test. e data shown are
means + SEM, n = 11 per group; **p < 0.01.
www.nature.com/scientificreports/
3
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
with the paired t test revealed, however, that in the WT + PO group the MPT aer instillation of PO was signif-
icantly higher than at baseline. In contrast, Δ MPT was signicantly dierent among the experimental groups
(F(3,20) = 3.3; p = 0.041). Post-hoc testing showed that the Δ MPT was signicantly lower in the PYY(/) + AITC
compared to the WT + PO group but did not signicantly dier compared to the WT + AITC group (Fig.4C).
MPT measurements were also taken on the abdomen. e quantitative MPT and Δ MPT readouts over the
abdomen showed a similar but highly variable pattern, and no signicant dierences could be detected among the
experimental groups (Supplement,FigureS2).
AITC increases spinal NPY and Y1 receptor mRNA expression in PYY(/) but not WT mice. e
eect of intrarectal administration of PO and AITC (1%, 0.05 ml) on the expression of NPY as well as Y1 and
Y2 receptor mRNA in the lumbosacral spinal cord was examined in WT and PYY(/) mice. One-way ANOVA
revealed significant differences among the experimental groups with regard to NPY mRNA (F(3,18) = 3.3;
p = 0.045) (Fig.5A) and Y1 receptor mRNA (F(3,18) = 3.7; p = 0.032) expression (Fig.5B) while Y2 receptor mRNA
expression did not dier to a statistically signicant extent (Fig.5C). Post-hoc testing revealed that NPY mRNA
Figure 2. Eect of intrarectally administered AITC (2%, 0.1 ml) and PYY knockout (PYY(/)) on pain-related
behaviors (A), time spent freezing (B), latency to rst pain-related behavior (C), and time spent grooming (D).
e behavioral parameters were evaluated blindly from 15 min videos recorded immediately aer intrarectal
treatment. One-way ANOVA revealed statistically signicant dierences among the groups with regard to pain-
related behaviors (p < 0.05) and the time spent freezing (p < 0.01). e data shown are means + SEM, n = 6–8
per group; *p < 0.05, **p < 0.01 compared to WT + PO; #p < 0.05 compared to WT + AITC.
www.nature.com/scientificreports/
4
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
and Y1 mRNA expression in the PYY(/) + AITC group was signicantly higher than in the WT + PO group
(Fig.5A,B).
Y2 receptor antagonism increases pain-related behaviors induced by intrarectal AITC. In this
experiment, the eects of the Y2 receptor agonist PYY(3–36) (0.2 mg/kg) and the Y2 receptor antagonist BII0246
(0.03 mmol/kg) on visceral pain sensitivity were evaluated in conjunction with intrarectal administration of AITC
(2%) to C57BL/6 N mice. e results revealed that the Y2 receptor antagonist increased pain-related behaviors
induced by intrarectal AITC while the Y2 receptor agonist had no eect.
As depicted in Fig.6, one-way ANOVA with Welch’s correction disclosed statistically signicant dierences
among the groups with regard to pain-related behaviors (Welch’s F(6,21.5) = 9.4; p < 0.001), time spent freezing
(Welch’s F(6,19.1) = 26.1; p < 0.001), latency to rst pain-related behavior (Welch’s F(6,18.2) = 15.6; p < 0.001), and
time spent grooming (Welch’s F(6,18.7) = 10.9; p < 0.001).
Post-hoc testing failed to reveal any signicant dierences in pain-related behavior counts between the vehi-
cle + PO group and vehicle + AITC group. Signicantly higher pain-related behavior counts were recorded in the
BIIE0246 + AITC and BIIE0246 + PYY(3–36) + AITC groups compared to the vehicle + PO and BIIE0246 + PO
groups. e BIIE0246 + AITC group also exhibited signicantly higher pain-related behavior counts than the
vehicle + AITC group (Fig.6A). e time spent freezing was signicantly higher in all groups receiving AITC
compared to PO, but no signicant dierences were observed within the groups receiving PO or within the groups
receiving AITC (Fig.6B). Similarly, the time spent grooming and the latency to the rst pain-related behavior
were shorter in all groups receiving AITC than in the PO group, while no signicant dierences were observed
within groups receiving PO or within groups receiving AITC (Fig.6C,D). The parameters recorded by the
LabMaster system (horizontal activity, vertical activity, traveling distance) measured for 15 min aer intrarectal
injection of PO or AITC were not signicantly dierent among the experimental groups (Supplement,FigureS3).
Discussion
Although the role of Y receptors in nociception is well established9, relatively little is known about the specic
implication of PYY in pain regulation. In this work, we examined the eects of genetic PYY deletion and Y2
receptor ligands on somatic sensitivity to thermal and mechanical pain and visceral sensitivity to chemical pain.
To probe chemonociception in the rectum we used AITC which stimulates transient receptor potential ankyrin 1
(TRPA1) and transient receptor potential vanilloid 1 (TRPV1) ion channels expressed by nociceptive aerent
nerve bers21,22. On the somatic level, PYY(/) mice were found to be hypersensitive to thermal somatic pain. On
the visceral level, PYY knockout exaggerated pain-related behaviors in response to intrarectal AITC. In conrma-
tion of these results, the Y2 receptor antagonist BII0246 increased pain-related behaviors in response to intrarec-
tal AITC. ese ndings attest to an involvement of PYY in the regulation of somatic and visceral pain sensitivity.
To study visceral pain and hyperalgesia by a minimally invasive method that does not require surgery, we
took advantage of the models of Laird et al.18 and Langford et al.20 and the recording of locomotor activity19 to
gauge pain by a multitude of behavioral readouts considered to reect several dimensions of pain experience. By
recording several behavioral manifestations of visceral pain (stretching, squashing, arching, licking of the abdo-
men)18 we found that intrarectal AITC evoked pain-related behaviors only in PYY(/) mice but not in WT mice.
It appears as if PYY knockout enhances the susceptibility of the mice to respond to the stress and mechanical
Figure 3. Eect of intrarectally administered AITC (1%, 0.05 ml) and PYY knockout (PYY(/)) on the mouse
grimace scale (Δ MGS) expressed as action units. One-way ANOVA revealed statistically signicant dierences
among the groups (p < 0.001). e data shown are means + SEM, n = 6–7 per group; ***p < 0.001 compared to
WT + PO; 𝛿𝛿𝛿p < 0.001 compared to PYY(/) + PO.
www.nature.com/scientificreports/
5
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
irritation associated with the instillation procedure with pain-related behaviors. is phenotype is consistent with
impaired stress coping and enhanced depression-like behavior seen in PYY(/) mice23. e moderate behavioral
response of the WT mice to intrarectal AITC was untypical when compared with the more pronounced response
observed in C57BL/6 mice19,24. Although the reason for this discrepancy is not understood, we think of a strain
eect, given that the genetic background of the WT animals is dierent from that of C57BL/6 mice.
e MGS described by Langford et al.20 is a novel method to assess the pain experience in response to sev-
eral noxious stimuli. In addition, recording of the MGS does not interfere with the evaluation of pain-related
behaviors or referred hyperalgesia. On the basis of our study we contend that a combination of several behavioral
methods to assess visceral pain, including facial pain expression, a variety of pain-related behaviors and referred
hyperalgesia, addresses several dimensions of nociception and has the potential to overcome strain-related dif-
ferences in the nociceptive responses to noxious stimuli. Although, for instance, the WT mice used here did not
show the typical pain-related behaviors described by Laird et al.18 in response to intrarectal AITC, they exhibited
a very prominent MGS response: the Δ MGS was more than 20 fold larger in response to AITC than aer admin-
istration of PO. On the other hand, the eect of PYY knockout on MGS was not statistically signicant.
A particular dimension of the visceral pain experience manifests itself in the phenomenon of referred
pain. We used a decrease in the MPT of the hindpaws as an index of referred hyperalgesia and found that the
Figure 4. Mechanical pain threshold (MPT) of the plantar surface of the hindpaws in WT and PYY knockout
(PYY(/)) mice before (baseline, A) and aer (post-treatment, B) intrarectal administration of PO or AITC
(1%, 0.05 ml) and the dierence between the two measurements (Δ MPT, C). MPT was assessed with von Frey
hairs, and the values represent the average of measurements in both hind paws. With regard to Δ MPT, one-
way ANOVA revealed statistically signicant dierences among the groups (p < 0.05). e data shown are
means + or SEM, n = 5–7 per group; *p < 0.05 compared to WT + PO.
Figure 5. Eect of intrarectal AITC (1%) on the relative expression of NPY (A), Y1 receptor (B), and Y2
receptor (C) mRNA in the lumbosacral spinal cord. Tissue was collected 1 h aer intrarectal administration
of PO or AITC. Relative mRNA expression was assessed with real time PCR. One-way ANOVA revealed
statistically signicant dierences among the groups with regard to NPY and Y1 receptor but not Y2 receptor
mRNA expression. e data shown are means + SEM, n = 5–6 per group; *p < 0.05 compared to WT + PO.
www.nature.com/scientificreports/
6
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
AITC-induced reduction of the MPT was most pronounced in PYY(/) mice. It must not be overlooked, how-
ever, that the AITC-induced Δ MPT in PYY(/) mice was only nominally, but not signicantly, dierent from the
AITC-induced Δ MPT in WT mice. e absence of a signicant AITC-evoked referred hyperalgesia in WT mice
is consistent with the relative resistance of this genotype towards pain-related behaviors induced by AITC. It is
worth noting, however, that in WT but not PYY(/) mice administration of the vehicle (PO) increased the MPT
of the hindpaws. is rise of the MPT may be interpreted as stress-induced analgesia which is known to be strain
dependent25,26. If so, the MPT changes in the dierent experimental groups may represent the composite result of
stress-induced analgesia and referred hyperalgesia evoked by intrarectal irritation.
Since PYY(3–36), the circulatory form of PYY, has a higher anity for Y2 receptors than PYY itself5, we exam-
ined the eects of this Y2 receptor agonist (0.2 mg/kg) and the Y2 receptor antagonist BIIE0246 (0.03 mmol/kg)
Figure 6. Eects of subcutaneously injected BIIE0246 (0.03 mmol/kg) and intraperitoneally injected PYY
(3–36) (0.2 mg/kg) on pain evoked by intrarectally administered PO or AITC (2%, 0.05 ml) in C57BL/6 N
mice. Shown are pain-related behaviors (A), time spent freezing (B), latency to rst pain-related behavior
(C), and time spent grooming (D). e behavioral parameters were evaluated blindly from 15-min videos
recorded immediately aer intrarectal treatment. One-way ANOVA with Welch’s correction revealed signicant
dierences among the groups. e data shown are means + SEM, n = 6 for PO groups and 9–12 for AITC
groups; *p < 0.05, **p < 0.01, ***p < 0.001 compared to vehicle + PO, #p < 0.05 compared to vehicle + AITC.
www.nature.com/scientificreports/
7
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
on pain-related behaviors induced by AITC (2%) in C57BL/6 N mice. e experiments revealed that Y2 recep-
tor antagonism led to visceral hyperalgesia in response to intrarectal AITC, while Y2 receptor agonism had no
eect. Specically, PYY(3–36) failed to attenuate pain-related behaviors and to attenuate the eect of BII0246
to exaggerate AITC-induced pain-related behaviors. e inecacy of PYY(3–36) could be due to pharmacody-
namic reasons, pharmacokinetic factors and/or particular experimental conditions. First, the simplest explana-
tion would be to assume that dosing of PYY(3–36) was inadequate. It should be noted, however, that the dose
used here (0.2 mg/kg) is considered to be a high dose, given that doses as low as 0.01 mg/kg are sucient to
enhance c-Fos expression in the brain and to induce several behavioral changes in mice27,28. Second, the failure of
PYY(3–36) to antagonize the hyperalgesic eect of BII0246 may be due to dierent pharmacokinetic proles of
the two compounds, PYY(3–36) being unable to achieve eective concentrations at the site where BII0246 exerts
its hyperalgesic action. ird, it should not go unnoticed that a failure of PYY(3–36) to induce expected pharma-
cological eects is a common observation in the literature29. e inconsistent ecacy of PYY(3–36) in vivo has
been attributed to the stress in handling the animals, its poor pharmacokinetic prole in rodents, or concomitant
activation of Y1 and Y5 receptors, as PYY(3–36) is not totally receptor-selective30,31.
e ndings of this study that PYY knockout and Y2 receptor antagonism enhance particular aspects of
AITC-evoked visceral pain behavior raise the question as to the site in the pain pathway where PYY and Y2
receptors operate. Y1 and Y2 receptors are widely expressed in the primary somatic and visceral pain pathways,
including the skin and colon, the inferior ganglion of the vagus nerve, the dorsal root ganglia, and the dorsal horn
of the spinal cord9,32–35. An implication of the NPY system in AITC-evoked rectal pain can be deduced from the
increased expression of NPY and Y1 receptor mRNA in the spinal cord of PYY(/) mice. e functional impli-
cation of these ndings is not clear. However, given that NPY and Y1 receptor signaling in the spinal cord have
an analgesic function8, it could be contended that the increase in spinal NPY and Y1 receptor mRNA expression
in PYY(/) mice reects a counterregulatory mechanism to balance the hyperalgesic state in this genotype. Due
to the widespread distribution of multiple Y receptors in the peripheral and central pain pathways it is dicult
to hypothesize on the sites where PYY could inuence visceral and somatic pain, given that PYY can cross the
blood brain barrier36. e observation that both somatic and visceral pain sensitivity was increased in PYY(/)
mice is compatible with both a peripheral and central site of action. In contrast, BII0246 acts primarily by block-
ing peripheral Y2 receptors37, which suggests that peripheral Y2 receptors play a role in controlling visceral pain
signaling. is contention is in keeping with the expression of Y2 receptors by vagal and spinal primary sensory
neurons in rodents and rabbits9,32,38.
In conclusion, PYY knockout and Y2 receptor antagonism have a hyperalgesic eect in mice, exaggerating
particular behavioral aspects of both somatic and visceral pain. Circumstantial evidence indicates that the eect
on visceral pain is mediated by peripheral Y2 receptors. Our experimental observations in mice may have a trans-
lational value in view of the deciency of PYY reported to occur in the colon of IBS and IBD patients12–15. e
results of the current work suggest that the decreased PYY content in the lower GIT of IBS and IBD patients may
contribute to the visceral pain associated with these pathologies.
Methods
Experimental animals. PYY(/) mice were generated by removing the entire coding sequence including
the initiation start as reported previously39. e genetic background of the knockout as well as WT mice was a 1:1
mixture of C57Bl/6 and129/SvJ. e PYY(/) mice and the age-matched WT mice were 2–4 months old when
used in the plantar test and in the assessment of spontaneous pain-related behaviors and 3–9 months old when
MGS and referred pain were assessed in parallel. ree dierent lots of mice were used for the plantar test, the
assessment of spontaneous pain-related behaviors, and the combined assessment of MGS and referred pain. e
animals subjected to the MGS and referred pain measurements were also used to assess the expression of NPY, Y1
receptor and Y2 receptor expression in the spinal cord. e total number of WT and PYY(/) mice employed in
this study amounted to 77 male mice.
e eects of BII0246 and PYY(3–36) on pain behavior were evaluated in a total of 59 male C57BL/6 N mice
obtained from Charles River (Sulzfeld, Germany). e C57BL/6 N mice arrived in the institutional animal house
at 8 weeks of age and were housed 2–3 per cage. ey were habituated for 2 weeks before they were subjected to
the experiments.
Ethical statement. All experiments were approved by an ethical committee at the Federal Ministry of
Science, Research, and Economy of the Republic of Austria (BMWFW-66.010/0054-WF/II/3b/2014) and con-
ducted according to the Directive of the European Communities Council of 24 November 1986 (86/609/EEC)
and the Directive of the European Parliament and of the Council of 22 September 2010 (2010/63/EU).
Intrarectal administration. Chemically induced nociception in the colon was induced by intrarectal AITC
administered through a Teon feeding cannula (length 38.1 mm, gauge 20) (Scanbur, Karlslunde, Denmark).
Vaseline (Rösch & Handel, Vienna, Austria) was applied to the perianal region and the tip of the cannula to
reduce any discomfort induced by the insertion of the cannula. AITC (1% (v/v)) in a volume of 0.05 ml in the
MGS and referred pain experiment, 2% in a volume of 0.1 ml in assessment of pain related behavior in WT and
PYY(/) mice and 2% in a volume of 0.05 ml in assessment of pain related behaviors in C57BL/6 N mice or its
vehicle, PO, was injected intrarectally through the feeding cannula18,19,21. e concentration (1%) and volume
(0.05 ml) of AITC used in the MGS and referred pain experiment was chosen to avoid any ceiling eect as noted
with higher dosing in pilot trials.
Drug treatment. PYY(3–36) lyophilized powder (Sigma-Aldrich, Vienna, Austria) was dissolved in saline
(0.9% NaCl) containing 1% bovine serum albumin. Aliquots were stored at 70 °C in polypropylene tubes and
www.nature.com/scientificreports/
8
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
thawed on the day of experiment. e peptide was injected intraperitoneally at the dose of 0.2 mg/kg (volume:
0.05 ml/10 g body weight). is dose of PYY(3–36) is sucient to induce several behavioral changes in mice27.
BIIE0246 (Tocris, Bristol, UK) was dissolved in 30% polyethylene glycol 200 in distilled water, prepared freshly
on the day of the experiment and injected subcutaneously (SC) at the dose of 0.03 mmol/kg (volume: 0.05 ml/10 g
body weight). is dose of BIIE0246 was used previously to evaluate the role of Y2 receptors in vagal aerent
signaling10. To assess the eects of the Y2 receptor agonist PYY(3–36) and the Y2 receptor antagonist BIIE0246,
7 groups of mice were used. e time line of the experiments was such that, rst, the mice received a SC injection
of BIIE0246 or its vehicle, 5 min later IP PYY(3–36) or its vehicle, and aer a further 5 min PO or 2% AITC intra-
rectally. e 7 experimental groups were as follows:
(1) SC vehicle, IP vehicle, PO intrarectally (vehicle + PO)
(2) SC vehicle, IP vehicle, 2% AITC intrarectally (vehicle + AITC)
(3) SC vehicle, IP PYY(3–36), PO intrarectally (PYY(3–36) + PO)
(4) SC vehicle, IP PYY(3–36), 2% AITC intrarectally (PYY(3–36) + AITC)
(5) SC BIIE0246, IP vehicle, PO intrarectally (BIIE0246 + PO)
(6) SC BIIE0246, IP vehicle, 2% AITC intrarectally (BIIE0246 + AITC)
(7) SC BIIE0246, IP PYY(3–36), 2% AITC intrarectally (BIIE0246 + PYY(3–36) + AITC)
After intrarectal administration of 2% AITC, pain-related behaviors and locomotion were assessed as
described below.
Assessment of pain-related behaviors. Aer intrarectal administration of AITC or vehicle, the behav-
ior of mice was video-recorded in the LabMaster system (TSE Systems, Bad Homburg, Germany) for 15 min. A
blinded trained investigator used the event marker module of the Videomot 2 tracking soware (TSE Systems) to
evaluate the video-recordings with regard to the following pain-related behaviors18,19,24: (a) stretching of the trunk
and squashing (pressing the abdomen towards the oor of the cage and stretching of the body), (b) licking of the
lower part of the abdomen, and (c) arching of the trunk.
e time spent freezing and grooming was also calculated from the video-recordings. Horizontal and vertical
locomotor activity was measured simultaneously with the LabMaster system19. Details about the LabMaster sys-
tem are available in the supplement.
The mouse grimace scale (MGS). Facial pain expression was evaluated with the MGS as described
previously20,40 with a slight modification that allowed for the simultaneous assessment of referred hyper-
algesia. Mice were kept on a wire mesh (Ugo Basile) in a homemade plexiglass box (9 cm × 5 cm × 5 cm;
length × height × width). All sides of the box except one (9 cm × 5 cm) were covered with white paper. Mice
were le to habituate in the box for 30 min. ereaer a 20-min video recording under baseline conditions was
taken with a Canon Legira HF R406 video camera. Aer taking the video, MPT was measured as described
later. Aerwards the mice received 1% AITC or the vehicle (PO) intrarectally as described above, followed by
another 20-min video recording for the assessment of the MGS and a second measurement of MPT. e MGS was
assessed as described in the supplement. e Δ MGS value was calculated by subtracting the baseline MGS from
post-treatment MGS and used as an index of visceral pain. One hour aer intrarectal treatment the mice were
sacriced by decapitation aer they had been deeply anesthetized with pentobarbital (150 mg/kg IP). e lum-
bosacral spinal cords were collected, shock frozen in liquid nitrogen and stored at 70 °C until RNA extraction.
Assessment of mechanical pain threshold (MPT) and referred pain. Referred pain18,24 was evalu-
ated in parallel with the MGS assessment and determined by two measurements of MPT over the plantar surface
of the hind paws and abdomen. e rst measurement of MPT was made under baseline conditions aer the
baseline MGS video had been taken. e second measurement of MPT was taken 20 min aer intrarectal admin-
istration of PO or 1% AITC when the video-recording of the post-treatment MGS had been completed. e MPT
was evaluated with von Frey laments (Bioseb, Vitrolles, France) using the simplied up-down method (SUDO
method)41 as described in the supplement. e Δ MPT value was calculated by subtracting the baseline MPT from
the post-treatment MPT. A reduction of Δ MPT was considered as an index of referred pain.
Real time PCR. NPY, Y1 receptor and Y2 receptor mRNA expression in the lumbosacral spinal cord
was assessed by real-time PCR. RNA was extracted with the RNeasy Lipid Tissue Mini Kit (Qiagen, Hilden,
Germany). Reverse-transcription was performed with the High Capacity cDNA Reverse Transcription kit as
described by the manufacturer (Applied Biosystems, Foster City, CA, USA). e PCR conditions and the primer
sequences are described in the supplement. GAPDH and PGK were used as reference genes. Quantitative values
of mRNA relative to control were calculated with the 2−ΔΔCT method42.
Statistical analysis. SPSS 22 and SigmaPlot 13 were used for statistical analysis and graphic presentation of
the results. e data were analyzed with two-sample t-test, paired t-test, or one-way ANOVA, as appropriate. Post
hoc pairwise comparisons with Tukey’s test were made when one-way ANOVA revealed signicant dierences
among the groups. Log transformation was considered whenever needed to meet one-way ANOVA assumptions.
When log transformation was not sucient to meet the equal variability assumption of one-way ANOVA as
tested by Levenes test, the Welch correction and post-hoc Games-Howell test were used. Dierences between
groups in t test, one-way ANOVA, and post-hoc tests were considered signicant if p 0.05. All data are pre-
sented as means + SEM, n referring to the number of mice in each group.
www.nature.com/scientificreports/
9
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
References
1. Ballantyne, G. H. Peptide YY(1–36) and peptide YY(3–36): Part I. Distribution, release and actions. Obes. Surg. 16, 651–658 (2006).
2. Cox, H. M. Peptide YY: a neuroendocrine neighbor of note. Peptides 28, 345–351 (2007).
3. Neary, M. T. & Batterham, . L. Peptide YY: food for thought. Physiol. Behav. 97, 616–619 (2009).
4. Cosun, T. et al. Activation of prostaglandin E receptor 4 triggers secretion of gut hormone peptides GLP-1, GLP-2, and PYY.
Endocrinology 154, 45–53 (2013).
5. Holzer, P., eichmann, F. & Farzi, A. Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut–brain axis. Neuropeptides
(2012).
6. Naveilhan, P. et al. educed antinociception and plasma extravasation in mice lacing a neuropeptide Y receptor. Nature 409,
513–517 (2001).
7. Moran, T. D., Colmers, W. F. & Smith, P. A. Opioid-lie actions of neuropeptide Y in rat substantia gelatinosa: Y1 suppression of
inhibition and Y2 suppression of excitation. J. Neurophysiol. 92, 3266–3275 (2004).
8. Smith, P. A., Moran, T. D., Abdulla, F., Tumber, . . & Taylor, B. . Spinal mechanisms of NPY analgesia. Peptides 28, 464–474
(2007).
9. Brumovsy, P., Shi, T. S., Landry, M., Villar, M. J. & Hofelt, T. Neuropeptide tyrosine and pain. Trends Pharmacol. Sci. 28, 93–102
(2007).
10. Wultsch, T. et al. Endogenous neuropeptide Y depresses the aerent signaling of gastric acid challenge to the mouse brainstem via
neuropeptide Y type Y2 and Y4 receptors. Neuroscience 136, 1097–1107 (2005).
11. Li, J. J., Zhou, X. & Yu, L. C. Involvement of neuropeptide Y and Y1 receptor in antinociception in the arcuate nucleus of
hypothalamus, an immunohistochemical and pharmacological study in intact rats and rats with inammation. Pain 118, 232–242
(2005).
12. El-Salhy, M., Gundersen, D., Hatleba, J. G., Gilja, O. H. & Hausen, T. Abnormal rectal endocrine cells in patients with irritable
bowel syndrome. egul. Pept. 188, 60–65 (2014).
13. El-Salhy, M., Hatleba, J. G., Gilja, O. H. & Hausen, T. Densities of rectal peptide YY and somatostatin cells as biomarers for the
diagnosis of irritable bowel syndrome. Peptides 67, 12–19 (2015).
14. El-Salhy, M., Danielsson, A., Stenling, . & Grimelius, L. Colonic endocrine cells in inammatory bowel disease. J. Intern. Med. 242,
413–419 (1997).
15. El-Salhy, M., Suhr, O. & Danielsson, A. Peptide YY in gastrointestinal disorders. Peptides 23, 397–402 (2002).
16. Tari, A. et al. Peptide YY abnormalities in patients with ulcerative colitis. Jpn. J. Med. 27, 49–55 (1988).
17. Allen, J. W. & Yash, T. L. Assessment of acute thermal nociception in laboratory animals. Methods Mol. Med. 99, 11–24 (2004).
18. Laird, J. M., Martinez-Caro, L., Garcia-Nicas, E. & Cervero, F. A new model of visceral pain and referred hyperalgesia in the mouse.
Pain 92, 335–342 (2001).
19. Jain, P. et al. Behavioral and molecular processing of visceral pain in the brain of mice: impact of colitis and psychological stress.
Front. Behav. Neurosci. 9, 177 (2015).
20. Langford, D. J. et al. Coding of facial expressions of pain in the laboratory mouse. Nat. Methods 7, 447–449 (2010).
21. Mitrovic, M., Shahbazian, A., Boc, E., Pabst, M. A. & Holzer, P. Chemonociceptive signalling from the colon is enhanced by mild
colitis and bloced by inhibition of transient receptor potential anyrin 1 channels. Br. J. Pharmacol. 160, 1430–1442 (2010).
22. Everaerts, W. et al. e capsaicin receptor TPV1 is a crucial mediator of the noxious eects of mustard oil. Curr. Biol. 21, 316–321
(2011).
23. Painsipp, E. et al. Neuropeptide Y and peptide YY protect from weight loss caused by Bacille Calmette-Guerin in mice. Br. J.
Pharmacol. 170, 1014–1026 (2013).
24. Eijelamp, N. et al. Increased visceral sensitivity to capsaicin aer DSS-induced colitis in mice: spinal cord c-Fos expression and
behavior. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G749–57 (2007).
25. Mosowitz, A. S., Terman, G. W. & Liebesind, J. C. Stress-induced analgesia in the mouse: strain comparisons. Pain 23, 67–72
(1985).
26. Przewloca, B. et al. e dierence in stress-induced analgesia in C57BL/6 and DBA/2 mice: a search for biochemical correlates. Pol.
J. Pharmacol. Pharm. 40, 497–506 (1988).
27. Stadlbauer, U., Langhans, W. & Meyer, U. Administration of the Y2 receptor agonist PYY3-36 in mice induces multiple behavioral
changes relevant to schizophrenia. Neuropsychopharmacology 38, 2446–2455 (2013).
28. Stadlbauer, U., Weber, E., Langhans, W. & Meyer, U. e Y2 receptor agonist PYY(3-36) increases the behavioural response to
novelty and acute dopaminergic drug challenge in mice. Int. J. Neuropsychopharmacol. 17, 407–419 (2014).
29. Tschop, M. et al. Physiology: does gut hormone PYY3-36 decrease food intae in rodents? Nature 430, 1 p following 165; discussion
2 p following 165 (2004).
30. Batterham, . et al. Physiology: does gut hormone PYY3-36 decrease food intae in rodents? (reply). Nature 430 (2004).
31. DeCarr, L. B. et al. A long-acting selective neuropeptide Y2 receptor PEGylated peptide agonist reduces food intae in mice. Bioorg.
Med. Chem. Lett. 17, 1916–1919 (2007).
32. Ghilardi, J. ., Allen, C. J., Vigna, S. ., McVey, D. C. & Mantyh, P. W. Cholecystoinin and neuropeptide Y receptors on single rabbit
vagal aerent ganglion neurons: site of prejunctional modulation of visceral sensory neurons. Brain es. 633, 33–40 (1994).
33. Zhang, X. et al. Expression and regulation of the neuropeptide Y Y2 receptor in sensory and autonomic ganglia. Proc. Natl. Acad. Sci.
USA 94, 729–734 (1997).
34. Goumain, M., Voisin, T., Lorinet, A. M. & Laburthe, M. Identication and distribution of mNA encoding the Y1, Y2, Y4, and Y5
receptors for peptides of the PP-fold family in the rat intestine and colon. Biochem. Biophys. es. Commun. 247, 52–56 (1998).
35. Landry, M., Holmberg, ., Zhang, X. & Hofelt, T. Eect of axotomy on expression of NPY, galanin, and NPY Y1 and Y2 receptors
in dorsal root ganglia and the superior cervical ganglion studied with double-labeling in situ hybridization and
immunohistochemistry. Exp. Neurol. 162, 361–384 (2000).
36. Nonaa, N., Shioda, S., Nieho, M. L. & Bans, W. A. Characterization of blood-brain barrier permeability to PYY3-36 in the
mouse. J. Pharmacol. Exp. er. 306, 948–953 (2003).
37. Brothers, S. P. et al. Selective and brain penetrant neuropeptide y y2 receptor antagonists discovered by whole-cell high-throughput
screening. Mol. Pharmacol. 77, 46–57 (2010).
38. Brumovsy, P. et al. Neuropeptide Y2 receptor protein is present in peptidergic and nonpeptidergic primary sensory neurons of the
mouse. J. Comp. Neurol. 489, 328–348 (2005).
39. Boey, D. et al. Peptide YY ablation in mice leads to the development of hyperinsulinaemia and obesity. Diabetologia 49, 1360–1370
(2006).
40. Leach, M. C. et al. e assessment of post-vasectomy pain in mice using behaviour and the Mouse Grimace Scale. PLoS One 7,
e35656 (2012).
41. Bonin, . P., Bories, C. & De oninc, Y. A simplied up-down method (SUDO) for measuring mechanical nociception in rodents
using von Frey laments. Mol. Pain 10, 26-8069-10-26 (2014).
42. Schmittgen, T. D. & Liva, . J. Analyzing real-time PC data by the comparative CT method. Nature protocols 3, 1101–1108 (2008).
www.nature.com/scientificreports/
10
Scientific RepoRts | 7:40968 | DOI: 10.1038/srep40968
Acknowledgements
is work was supported by the Austrian Science Fund (FWF grants P23097-B18, P25912-B23 and W1241-B18).
e authors thank Margit Eichholzer for running the PCR assays, and Jerey Mogil and Susana Sotocinal, McGill
University, Montreal, Canada, for their guidance and help in setting up the mouse grimace scale recordings.
Author Contributions
A.M.H., P.J. and P.H. designed the experiments and wrote the manuscript. A.M.H. and P.J. acquired the behavioral
data and performed the statistical analysis. H.H. generated the PYY(/) mice. R.M. and E.E.F. collected the spinal
cords for PCR analysis. F.R. contributed to the validation and analysis of the PCR assay and A.F. contributed to
the analysis of the LabMaster data. All authors revised the manuscript.
Additional Information
Supplementary information accompanies this paper at http://www.nature.com/srep
Competing nancial interests: e authors declare no competing nancial interests.
How to cite this article: Hassan, A. M. et al. Visceral hyperalgesia caused by peptide YY deletion and Y2
receptor antagonism. Sci. Rep. 7, 40968; doi: 10.1038/srep40968 (2017).
Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
is work is licensed under a Creative Commons Attribution 4.0 International License. e images
or other third party material in this article are included in the article’s Creative Commons license,
unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license,
users will need to obtain permission from the license holder to reproduce the material. To view a copy of this
license, visit http://creativecommons.org/licenses/by/4.0/
© e Author(s) 2017
... Data were extracted from the included studies by three independent reviewers (A.L.W., Y.L., T.H.B.) using an electronic form developed by the authors. All reviewers initially performed independent reviews of the same 3 studies [26][27][28] to pilot the extraction tool and check for data consistency. Following this, the remaining studies were allocated between the 3 reviewers, with each study being extracted by one reviewer. ...
... In 10 studies, summation of the individual action units scores was performed to arrive at the final score (maximum score of 10 for five FAUs). The method of achieving the final score was unclear in the remaining two studies [28,58]. A number of studies accounted for individual responses to pain by using mean difference scores in data presentation and analysis to correct for baseline grimace scores. ...
... Based on this discussion, perhaps the most reliable corroborating measure against which to assess the MGS is another readout of spontaneous pain, with composite pain behaviour scoring being the only measure to completely fulfil this description. In the studies that compared these two readouts, the direction of effect was aligned, but the studies are few in number (5) [28,47,49,53,56]. ...
Article
Full-text available
The Mouse Grimace Scale (MGS) was developed 10 years ago as a method for assessing pain through the characterisation of changes in five facial features or action units. The strength of the technique is that it is proposed to be a measure of spontaneous or non-evoked pain. The time is opportune to map all of the research into the MGS, with a particular focus on the methods used and the technique’s utility across a range of mouse models. A comprehensive scoping review of the academic literature was performed. A total of 48 articles met our inclusion criteria and were included in this review. The MGS has been employed mainly in the evaluation of acute pain, particularly in the pain and neuroscience research fields. There has, however, been use of the technique in a wide range of fields, and based on limited study it does appear to have utility for pain assessment across a spectrum of animal models. Use of the method allows the detection of pain of a longer duration, up to a month post initial insult. There has been less use of the technique using real-time methods and this is an area in need of further research.
... Data were extracted from the included studies by three independent reviewers (AW, YL, THB) using an electronic form developed by the authors (Appendix 2). All reviewers initially performed independent review of the same 3 studies Dwivedi et al., 2016;Hassan et al., 2017) to pilot the extraction tool and check for data consistency. Following this the remaining studies were allocated between the 3 reviewers; each study being extracted by one reviewer. ...
... The method of achieving the final score was unclear in the remaining 2 studies. (Hassan et al., 2017;Mitchell et al., 2020) A number of studies accounted for individual responses to pain by using mean difference scores in data presentation and analysis to correct for baseline grimace scores. For studies where the whisker position FAU was excluded, 50% of the studies used mice (6/12) that were black coloured, 33% (4/12) white, and 17% (2/12) brown coloured (X 2 (2, N = 12) = 3, p=0.22). ...
... In the studies that compared these two readouts, the direction of effect was aligned but the studies are few in number (5). (Hassan et al., 2017;Jurik et al., 2014;Leach et al., 2012;Miller et al., 2015; Another finding of this review that questions the construct validity of the MGS is the change in grimace scores in response to techniques that would not be expected to elicit pain. Out of the eleven studies that examined the MGS over the 24 hour period after an intervention, that were expected to elicit none or momentary pain, six found grimace score elevations. ...
Preprint
Full-text available
The Mouse Grimace Scale (MGS) was developed 10 years ago to assess pain through characterisation of changes in five facial features or action units. The strength of the technique is that it is proposed to be a measure of spontaneous or non-evoked pain. A comprehensive scoping review of the academic literature was performed. The MGS has been employed mainly in evaluation of acute pain, particularly in the pain and neuroscience research fields. There has however been use of the technique in a wide range of fields, and based on limited study it does appear to have utility for pain assessment across a spectrum of animal models. Use of the method does allow detection of pain of a longer duration, up to a month post-initial insult. There has been less use of the technique using real-time methods and this is an area in need of further research.
... Allyl isothiocyanate (AITC)-induced hypersensitivity was assessed using the methods described previously (57). Mice were habituated in the 15 cm×10 cm×8 cm observation cage 20 minutes. ...
Preprint
It is widely said that healthy intestinal environment takes essential role for better mental condition. One of the known dietary nutrients which maintains intestinal environment is the dietary fiber. Recent study showed that maintaining intestinal environment by dietary fiber succeeded to alleviate the psychiatric disorder symptoms in animals. However, such effects have only been reported with soluble fiber, which is highly fermentable and promotes short-chain fatty acid (SCFA) production, and not with insoluble fiber. Therefore, we aimed to verify whether insoluble fiber, such as cellulose, can alter emotion via changes in the gut. We divided mice into two groups and fed either standard diet (SD, contains both insoluble and soluble dietary fibers) or cellulose rich diet (CRD, contains cellulose alone as the dietary fibers). The CRD-fed mice displayed 1) the increased the anxiety-like behavior accompanied with 2) the modified amygdalar dopamine signaling. We further found the decreased intestinal SCFA levels along with intestinal permeability, dysmotility and hypersensitivity in CRD-fed mice. These behavioral and physiological effect of CRD has been completely abolished in vagotomized mice, indicating the direct link between intestinal environment exacerbation to the emotion through gut-brain axis. Additionally, the opioid antagonist abolished the CRD-induced anxiety, suggesting the involvement of opioidergic system to the anxiety which may evoked by increased amygdalar dopamine levels. Altogether, our findings suggest that consumption of cellulose alone as the dietary fiber may evoke intestinal abnormalities which fires the vagus nerve then opiodergic system and amygdalar dopamine upregulation, resulting in the enhancement of anxiety.
... 23 Peptide YY (PYY) is a gut hormone that suppresses food intake 24 and reportedly has a hypoalgesic effect on somatic thermal and visceral chemical pain as a pain inhibitor. 25 Ion channels (ICs) include members of multiple gene families, such as transient receptor potential (TRP) channels, acid sensing ion channels (ASICs) involved in the detection of nociceptive signals, [26][27][28] and voltage gated Na + channels (VDSC) and voltage gated Ca 2+ channels (VDCC) for the propagation and transmission of the pain signal, which are critical for detecting noxious stimuli in primary sensory neurons. 29 TRP channels are involved in pain sensations generated by chemical or thermal stimulation. ...
Article
Full-text available
Nociception and pain sensation are important neural processes in humans to avoid injury. Many proteins are involved in nociception and pain sensation in humans; however, the evolution of these proteins in animals is unknown. Here, we chose nociception- and pain-related proteins, including G protein-coupled receptors (GPCRs), ion channels (ICs), and neuropeptides (NPs), which are reportedly associated with nociception and pain in humans, and identified their homologs in various animals by BLAST, phylogenetic analysis and protein architecture comparison to reveal their evolution from protozoans to humans. We found that the homologs of transient receptor potential channel A 1 (TRPA1), TRAPM, acid-sensing IC (ASIC), and voltage-dependent calcium channel (VDCC) first appear in Porifera. Substance-P receptor 1 (TACR1) emerged from Coelenterata. Somatostatin receptor type 2 (SSTR2), TRPV1 and voltage-dependent sodium channels (VDSC) appear in Platyhelminthes. Calcitonin gene-related peptide receptor (CGRPR) was first identified in Nematoda. However, opioid receptors (OPRs) and most NPs were discovered only in vertebrates and exist from agnatha to humans. The results demonstrated that homologs of nociception and pain-related ICs exist from lower animal phyla to high animal phyla, and that most of the GPCRs originate from low to high phyla sequentially, whereas OPRs and NPs are newly evolved in vertebrates, which provides hints of the evolution of nociception and pain-related proteins in animals and humans.
... This simple test can help to identify pain located in the visceral area and could be a great tool for the identification of spontaneous pain caused by endometrial lesions in rodents [56]. Spontaneous nociceptive behavior can be evaluated by measuring either the time spent grooming (washing or licking the abdominal area) or by the number of grooming repetitions over a certain period of time, as selected according to the investigator's discretion [54,63,66]. ...
Article
Full-text available
Chronic pain induced by endometriosis is a maladaptive pain experienced by half of women with this disease. The lack of pharmacological treatments suitable for the long-term relief of endometriosis-associated pain, without an impact on fertility, remains an urgent unmet need. Progress has been slowed by the absence of a reproducible rodent endometriosis model that fully replicates human physiopathological characteristics, including pain symptoms. Although pain assessment in rodents is a complicated task requiring qualified researchers, the choice of the behavioral test is no less important, since selecting inappropriate tests can cause erroneous data. Pain is usually measured with reflex tests in which hypersensitivity is evaluated by applying a noxious stimulus, yet this ignores the associated emotional component that could be evaluated via non-reflex tests. We conducted a systematic review of endometriosis models used in rodents and the number of them that studied pain. The type of behavioral test used was also analyzed and classified according to reflex and non-reflex tests. Finally, we determined the most used reflex tests for the study of endometriosis-induced pain and the main non-reflex behavioral tests utilized in visceral pain that can be extrapolated to the study of endometriosis and complement traditional reflex tests.
... In a recent study, Hassan et al. used PYY knockout mice to investigate the relationship between pain, PYY, and the Y2 receptor. Their data suggested that the Y2 receptor antagonist and knockout of PYY increased visceral pain [117]. However, future studies are needed to confirm the effect of PYY on visceral pain and to investigate the details of the mechanism. ...
Article
Full-text available
Neurological and psychiatric patients have increased dramatically in number in the past few decades. However, effective treatments for these diseases and disorders are limited due to heterogeneous and unclear pathogenic mechanisms. Therefore, further exploration of the biological aspects of the disease, and the identification of novel targets to develop alternative treatment strategies, is urgently required. Systems-level investigations have indicated the potential involvement of the brain–gut axis and intestinal microbiota in the pathogenesis and regulation of neurological and psychiatric disorders. While intestinal microbiota is crucial for maintaining host physiology, some important sensory and regulatory cells in the host should not be overlooked. Intestinal epithelial enteroendocrine cells (EECs) residing in the epithelium throughout intestine are the key regulators orchestrating the communication along the brain-gut-microbiota axis. On one hand, EECs sense changes in luminal microorganisms via microbial metabolites; on the other hand, they communicate with host body systems via neuroendocrine molecules. Therefore, EECs are believed to play important roles in neurological and psychiatric disorders. This review highlights the involvement of EECs and subtype cells, via secretion of endocrine molecules, in the development and regulation of neurological and psychiatric disorders, including Parkinson’s disease (PD), schizophrenia, visceral pain, neuropathic pain, and depression. Moreover, the current paper summarizes the potential mechanism of EECs in contributing to disease pathogenesis. Examination of these mechanisms may inspire and lead to the development of new aspects of treatment strategies for neurological and psychiatric disorders in the future.
... The MGS was successfully used in models expected to produce pain of a neuropathic nature, for example in headache and migraine [55,87] and craniotomy [65]. There is also suggestion that pain of a visceral nature elevates scores based on studies evaluating colonic nociception [88], pelvic pain [89], colitis [61], and alimentary mucositis [62,63]. Hereditary sickle cell disease frequently leads to painful episodes in human patients. ...
Article
Full-text available
Animals’ facial expressions are widely used as a readout for emotion. Scientific interest in the facial expressions of laboratory animals has centered primarily on negative experiences, such as pain, experienced as a result of scientific research procedures. Recent attempts to standardize evaluation of facial expressions associated with pain in laboratory animals has culminated in the development of “grimace scales”. The prevention or relief of pain in laboratory animals is a fundamental requirement for in vivo research to satisfy community expectations. However, to date it appears that the grimace scales have not seen widespread implementation as clinical pain assessment techniques in biomedical research. In this review, we discuss some of the barriers to implementation of the scales in clinical laboratory animal medicine, progress made in automation of collection, and suggest avenues for future research.
Article
The intestinal homeostasis maintained by the gut microbiome and relevant metabolites is essential for health, and its disturbance leads to various intestinal or extraintestinal diseases. Recent studies suggest that gut microbiome-derived metabolites short-chain fatty acids (SCFAs) are involved in different neurological disorders (such as chronic pain). SCFAs are produced by bacterial fermentation of dietary fibers in the gut and contribute to multiple host processes, including gastrointestinal regulation, cardiovascular modulation, and neuroendocrine-immune homeostasis. Although SCFAs have been implicated in the modulation of chronic pain, the detailed mechanisms that underlie such roles of SCFAs remain to be further investigated. In this review, we summarize currently available research data regarding SCFAs as a potential therapeutic target for chronic pain treatment and discuss several possible mechanisms by which SCFAs modulate chronic pain.
Chapter
Although currently available therapeutics provide moderate relief from acute pain, they are typically limited by adverse side effects, including tolerance, addiction, and constipation when used for treatment of chronic pain. Thus, there is a significant medical need for novel therapies devoid of serious adverse effects for the treatment of chronic pain. Epigenetic drugs, such as histone deacetylase inhibitors and histone acetyltransferase inhibitors, have been shown in experimental models to have a potential role in managing pain perception. This chapter will review spinal and central pain pathways mediating nociception and the basic molecular epigenetic mechanisms of pain processing. We will discuss the animal models, which have enhanced our understanding of how epigenetic processes may play a role in the pathophysiological mechanisms of chronic pain. Throughout the chapter, the authors have attempted to critically evaluate the latest research from both experimental models and human studies to highlight the future role of epigenetic drugs in managing both acute and chronic pain. However, it is important to note that the role of epigenetics in pain perception is a developing field. Therefore, the exact epigenetic mechanisms underlying chronic pain perception remain to be established.
Article
Abdominal migraine is a type of functional abdominal pain disorder that affects 0.2% to 4.1% of children. It consists of paroxysmal, recurrent, and acute abdominal pain attacks with associated symptoms, including pallor, nausea, vomiting, anorexia, headache, and photophobia. In between episodes, patients return to their baseline health. Abdominal migraine is a clinical diagnosis. Its diagnostic criteria are outlined under the Rome IV criteria and the International Classification of Headache Disorders III criteria. Hypothesized contributors to its pathophysiology include a combination of visceral hypersensitivity, gut-brain enteric nervous system alterations, and psychological factors. Treatment is focused on preventive measures and mostly includes nonpharmacologic approaches. Possible pharmacologic treatments include abortive medications used for migraine headaches such as analgesics and antiemetics. Abdominal migraine is likely underdiagnosed and is poorly understood. Individuals who have abdominal migraine report a lower quality of life, rendering it an important diagnosis. The aim of this article is to review the epidemiology, clinical presentation, pathophysiology, diagnosis, and treatment of abdominal migraine in children.
Article
Full-text available
Gastrointestinal disorders with abdominal pain are associated with central sensitization and psychopathologies that are often exacerbated by stress. Here we investigated the impact of colitis induced by dextran sulfate sodium (DSS) and repeated water avoidance stress (WAS) on spontaneous and nociception-related behavior and molecular signaling in the mouse brain. DSS increased the mechanical pain sensitivity of the abdominal skin while both WAS and DSS enhanced the mechanical and thermal pain sensitivity of the plantar skin. These manifestations of central sensitization were associated with augmented c-Fos expression in spinal cord, thalamus, hypothalamus, amygdala and prefrontal cortex. While WAS stimulated phosphorylation of mitogen-activated protein kinase (MAPK) p42/44, DSS activated another signaling pathway, both of which converged on c-Fos. The DSS- and WAS-induced hyperalgesia in the abdominal and plantar skin and c-Fos expression in the brain disappeared when the mice were subjected to WAS+DSS treatment. Intrarectal allyl isothiocyanate (AITC) evoked aversive behavior (freezing, reduction of locomotion and exploration) in association with p42/44 MAPK and c-Fos activation in spinal cord and brain. These effects were inhibited by morphine, which attests to their relationship with nociception. DSS and WAS exerted opposite effects on AITC-evoked p42/44 MAPK and c-Fos activation, which indicates that these transduction pathways subserve different aspects of visceral pain processing in the brain. In summary, behavioral perturbations caused by colitis and psychological stress are associated with distinct alterations in cerebral signaling. These findings provide novel perspectives on central sensitization and the sensory and emotional processing of visceral pain stimuli in the brain.
Article
Full-text available
Irritable bowel syndrome (IBS) is a common chronic disorder. IBS diagnosis is a diagnosis of exclusion since there are no blood tests, radiological or endoscopic examinations for this disorder. Although several attempts have been made to develop a symptoms-based diagnosis, such systems are not widely used in clinics. Several tests and examinations measuring pathological findings in IBS have been considered for the diagnosis of IBS, but none of them has proved useful as a biomarker. Abnormalities in the cell densities of rectal peptide YY (PYY) and somatostatin cells have been reported in IBS patients. The aim of the present study was to determine the utility of these abnormalities as biomarkers for the diagnosis of IBS. Patients with IBS established according to Rome III criteria (n=101) were included in this study (71 females and 30 males with a mean age of 35 years; range 18-61 years), and 62 healthy subjects (38 females and 24 males with a mean age of 41 years; range 18-65 years) were recruited as controls. Both the patients and controls underwent colonoscopy during which rectal biopsy samples were taken. The tissue samples were immunostained for PYY and somatostatin, and the number of stained cells was quantified relative to both the area of epithelial cells and per microscopic field. The density of PYY cells was significantly lower in IBS patients than in the healthy controls (P<0.0001); receiver operator characteristic (ROC) analysis revealed an area under the ROC curve (AUC) of 0.99. The somatostatin cell density in IBS patients was higher than in the controls (P<0.0001); ROC analysis revealed an AUC of 0.86. The densities of the rectal PYY and somatostatin cells appear to be clinically effective biomarkers for IBS. Furthermore, measurement of these parameters is inexpensive, rapid and does not require considerable experience or sophisticated equipment. Copyright © 2015. Published by Elsevier Inc.
Article
Full-text available
The results of Tschöp et al.¹ on the lack of effect of peripheral administration of PYY3–36 on food intake in rodents are at odds with both the published literature2, 3, 4, 5, 6, 7, ⁸ (our Table 1) and with earlier data generated by the Tschöp laboratory.
Article
Full-text available
The measurement of mechanosensitivity is a key method for the study of pain in animal models. This is often accomplished with the use of von Frey filaments in an up-down testing paradigm. The up-down method described by Chaplan et al. (J Neurosci Methods 53:55-63, 1994) for mechanosensitivity testing in rodents remains one of the most widely used methods for measuring pain in animals. However, this method results in animals receiving a varying number of stimuli, which may lead to animals in different groups receiving different testing experiences that influences their later responses. To standardize the measurement of mechanosensitivity we developed a simplified up-down method (SUDO) for estimating paw withdrawal threshold (PWT) with von Frey filaments that uses a constant number of five stimuli per test. We further refined the PWT calculation to allow the estimation of PWT directly from the behavioral response to the fifth stimulus, omitting the need for look-up tables. The PWT estimates derived using SUDO strongly correlated (r > 0.96) with the PWT estimates determined with the conventional up-down method of Chaplan et al., and this correlation remained very strong across different levels of tester experience, different experimental conditions, and in tests from both mice and rats. The two testing methods also produced similar PWT estimates in prospective behavioral tests of mice at baseline and after induction of hyperalgesia by intraplantar capsaicin or complete Freund's adjuvant. SUDO thus offers an accurate, fast and user-friendly replacement for the widely used up-down method of Chaplan et al.
Article
Full-text available
The gastrointestinal hormone PYY3-36 is a preferential Y2 neuropeptide Y (NPY) receptor agonist. Recent evidence indicates that PYY3-36 acts on central dopaminergic pathways, but its influence on dopamine-dependent behaviours remains largely unknown. We therefore explored the effects of peripheral PYY3-36 treatment on the behavioural responses to novelty and to dopamine-activating drugs in mice. In addition, we examined whether PYY3-36 administration may activate distinct dopamine and γ-aminobutyric acid (GABA) cell populations in the mesoaccumbal and nigrostriatal pathways. We found that i.p. PYY3-36 injection led to a dose-dependent increase in novel object exploration. The effective dose of PYY3-36 (1 μg/100 g body weight) also potentiated the locomotor reaction to the indirect dopamine receptor agonist amphetamine and increased stereotyped climbing/leaning responses following administration of the direct dopamine receptor agonist apomorphine. PYY3-36 administration did not affect activity of midbrain dopaminergic cells as evaluated by double immuno-enzyme staining of the neuronal early gene product c-Fos with tyrosine hydroxylase. PYY3-36 did, however, lead to a marked increase in the number of cells co-expressing c-Fos with glutamic acid decarboxylase in the nucleus accumbens and caudate putamen, indicating activation of GABAergic cells in dorsal and ventral striatal areas. Our results support the hypothesis that acute administration of the preferential Y2 receptor agonist PYY3-36 modulates dopamine-dependent behaviours. These effects do not seem to involve direct activation of midbrain dopamine cells but instead are associated with neuronal activation in the major input areas of the mesoaccumbal and nigrostriatal pathways.
Article
Full-text available
Immune challenge of mice with Bacille Calmette-Guérin (BCG) has been reported to cause transient weight loss and a behavioural sickness response. While BCG-induced depression involves the kynurenine pathway, weight loss occurs independently of this factor. Since neuropeptide Y (NPY) and peptide YY (PYY) are involved in the regulation of food intake, we hypothesized that they play a role in the BCG-induced weight loss. Male wild-type (WT), PYY knockout (PYY-/-), NPY knockout (NPY-/-) and NPY-/-;PYY-/- double knockout mice were injected with vehicle or BCG (approximately 10(8) CFU per mouse), and their weight, locomotion, exploration and ingestion were recorded for 2 weeks post-treatment. Deletion of PYY and NPY aggravated the BCG-induced loss of body weight, which was most pronounced in NPY-/-;PYY-/- mice (maximum loss: 15 %). The weight loss in NPY-/-;PYY-/- mice did not normalize during the 2 week observation period. BCG suppressed the circadian pattern of locomotion, exploration and food intake. However, these changes took a different time course than the prolonged weight loss caused by BCG in NPY-/-;PYY-/- mice. The effect of BCG to increase circulating interleukin-6 (measured 16 days post-treatment) remained unaltered by knockout of PYY, NPY or NPY plus PYY. These data show that NPY and PYY are both required to protect from the action of BCG-evoked immune challenge to cause prolonged weight loss and disturb energy balance. The findings attest to an important role of NPY and PYY in orchestrating homeostatic reactions to infection and immune stimulation.
Article
Full-text available
Functional changes in neuropeptide Y (NPY) signaling at the Y2 receptor subtype have been widely implicated in stress-related neuropsychiatric illnesses such as depression and anxiety disorders. Altered Y2 receptor signaling may also play a role in the precipitation of behavioral and cognitive symptoms associated with schizophrenia. To seek preclinical evidence for this possibility, we explored the functional consequences of treatment with the selective Y2 receptor agonist PYY3-36 using translational tests for the assessment of schizophrenia-relevant behavioral and cognitive deficits in mice. We found that acute systemic administration of PYY3-36 at a low dose (1 μg/100 g body weight) or high dose (20 μg/100 g body weight) profoundly impaired social interaction without affecting innate anxiety. PYY3-36 treatment at the high dose further led to a disruption of sensorimotor gating in the form of prepulse inhibition deficiency. This effect was fully antagonized by acute treatment with the preferential dopamine D2 receptor antagonist haloperidol, but not with clozapine. In addition, both doses of PYY3-36 impaired selective associative learning in the latent inhibition paradigm and spatial working memory in a matching-to-position water maze test. The wide range of abnormalities induced by PYY3-36 suggests that signaling at the Y2 subtype of NPY receptors is critical for a number of behavioral and cognitive functions, some of which are highly relevant to schizophrenia and related psychotic disorders. At least some of the behavioral deficits induced by augmentation of Y2 receptor signaling may involve increased dopaminergic activity.Neuropsychopharmacology accepted article preview online, 10 June 2013; doi:10.1038/npp.2013.146.
Article
Two different methods of presenting quantitative gene expression exist: absolute and relative quantification. Absolute quantification calculates the copy number of the gene usually by relating the PCR signal to a standard curve. Relative gene expression presents the data of the gene of interest relative to some calibrator or internal control gene. A widely used method to present relative gene expression is the comparative CT method also referred to as the 2−ΔΔCT method. This protocol provides an overview of the comparative CT method for quantitative gene expression studies. Also presented here are various examples to present quantitative gene expression data using this method.
Article
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder. In a previous study the total number of endocrine cells in the rectum of IBS patients, as detected by chromogranin A, did not differ from that of healthy controls. While the total endocrine cell content of the rectum appears to be unchanged in IBS patients, changes in particular endocrine cells cannot be excluded. This study was undertaken, therefor, to investigate the cell density of different rectal endocrine cell types in (IBS) patients. Fifty patients with IBS (41 females and 9 males) were included in the study. Thirty patients had diarrhoea (IBS-D) and 20 had constipation (IBS-C) as the predominant symptom. Twenty-seven subjects were included as controls (19 females and 8 males). Rectal biopsy specimens were immunostained using the avidin-biotin-complex method for serotonin, peptide YY (PYY), pancreatic polypeptide (PP), and oxyntomodulin and somatostatin cells. The cell densities were quantified by computerised image analysis. The serotonin cell density did not differ significantly, although a type II statistical error cannot be excluded, due to the small size of the sample. The densities of PYY and Oxyntomodulin cells were significantly lower and that of somatostatin were significantly higher in IBS patients than controls. These abnormalities were observed in both IBS-D and IBS-C patients. The abnormalities in the endocrine cells observed in this study in the rectum differed considerably from those seen in the colon of IBS patients. This indicates that caution in using the rectum to represent the large intestine in these patients. These abnormalities could be primary (genetic) or secondary to changes in the gut hormones found in other segments of the gut and/or other pathological processes. Although the-cause-and effect relationship of the abnormalities found in rectal endocrine cells is difficult to elucidate, they might contribute to the symptoms associated with IBS. The densities of PYY and somatostatin cells are potential biomarkers with good sensitivity and specificity for the diagnosis of IBS.