ArticlePDF Available

Distribution of Secretin Receptors in the Rat Central Nervous System: an in situ Hybridization Study

Authors:

Abstract and Figures

Secretin shows a wide distribution in the brain. Functional significance of central secretin is stressed since it has been associated with autism and schizophrenia. The presence of the secretin receptor was previously demonstrated in the brain by different methods. Neurons in the cerebellum, hypothalamic paraventricular and supraoptic nuclei, and in the vascular organ of lamina terminalis were shown to express secretin receptor mRNA by using in situ hybridization with digoxigenin-labeled probe. In this work, we used a very sensitive radioactive in situ hybridization technique and systematically mapped the expression of secretin receptor mRNA in the brain. The densest labeling was observed in the nucleus of solitary tract and in the laterodorsal thalamic nucleus, where decreasing number of receptors was seen in the vascular organ of lamina terminalis, and the lateral habenular complex, and then in the supraoptic nucleus. Only a few scattered labeled cells were observed in the median frontal gyrus, entorhinal cortex, hypothalamic paraventricular nucleus, perifornical region, lateral hypothalamic area, head of the caudate nucleus, spinal trigeminal nucleus, and cerebellum. Secretin receptor mRNA showed a far wider distribution than was known before, suggesting a more significant functional relevance than thought earlier.
Content may be subject to copyright.
Distribution of Secretin Receptors in the Rat Central Nervous
System: an in situ Hybridization Study
Zsuzsanna E. Tóth &Andrea Heinzlmann &Hitoshi Hashimoto &Katalin Köves
Received: 2 July 2012 /Accepted: 24 September 2012 / Published online: 11 October 2012
#Springer Science+Business Media New York 2012
Abstract Secretin shows a wide distribution in the brain.
Functional significance of central secretin is stressed since it
has been associated with autism and schizophrenia. The
presence of the secretin receptor was previously demonstrat-
ed in the brain by different methods. Neurons in the cere-
bellum, hypothalamic paraventricular and supraoptic nuclei,
and in the vascular organ of lamina terminalis were shown
to express secretin receptor mRNA by using in situ hybrid-
ization with digoxigenin-labeled probe. In this work, we
used a very sensitive radioactive in situ hybridization tech-
nique and systematically mapped the expression of secretin
receptor mRNA in the brain. The densest labeling was
observed in the nucleus of solitary tract and in the latero-
dorsal thalamic nucleus, where decreasing number of recep-
tors was seen in the vascular organ of lamina terminalis, and
the lateral habenular complex, and then in the supraoptic
nucleus. Only a few scattered labeled cells were observed in
the median frontal gyrus, entorhinal cortex, hypothalamic
paraventricular nucleus, perifornical region, lateral hypotha-
lamic area, head of the caudate nucleus, spinal trigeminal
nucleus, and cerebellum. Secretin receptor mRNA showed a
far wider distribution than was known before, suggesting a
more significant functional relevance than thought earlier.
Keywords Brain stem .Forebrain .In vitro transcription .
Radioactive labeling .Mapping
Abbreviations
BNST Bed nucleus of stria terminalis
Bp Base pair
cAMP Cyclic adenosine monophosphate
CRH Corticotropin-releasing hormone
GABA Gamma-aminobutyric acid
ICV Intracerebroventricular
IN Intranasal
IV Intravenous
PBS Phosphate-buffered saline
PCR Polymerase chain reaction
NST Nucleus of solitary tract
PV Paraventricular nucleus
SO Supraoptic nucleus
SSC Sodium chloridesodium citrate buffer
UTP Uracil triphosphate
VOLT Vascular organ of lamina terminalis
Introduction
Secretin, one of the so-calledgastrointestinal peptides,
was first described in enteroendocrine S cells of the duode-
num and in Langerhans islets (Sundler and Hakanson 1988).
With the use of radioimmunoassay, secretin was also found
in several regions of the central nervous system including
the thalamus, hypothalamus, olfactory bulb, cerebral cortex,
septum, striatum, hippocampus, midbrain, pons, and medul-
la (Charlton et al. 1981). Secretin and its mRNA were also
J Mol Neurosci (2013) 50:172178
DOI 10.1007/s12031-012-9895-1
Z. E. Tóth
Neuromorphological and Neuroendocrine Research Laboratory,
Department of Anatomy, Histology and Embryology,
Semmelweis University and the Hungarian Academy of Sciences,
1094 Budapest, Hungary
A. Heinzlmann :K. Köves (*)
Department of Human Morphology and Developmental Biology,
Semmelweis University,
Tűzoltó u. 58,
1094 Budapest, Hungary
e-mail: koves.katalin@med.semmelweis-univ.hu
H. Hashimoto
Laboratory of Medicinal Pharmacology,
Graduate School of Pharmaceutical Sciences, Osaka University,
Yamada-oka,
Suita, Osaka, Japan
shown in the Purkinje cells of the cerebellar cortex (Yung
et al. 2001).
In our laboratory, using immunohistochemistry, we ob-
served secretin immunopositive elements in various parts of
human, cat, and rat central nervous systems including the
Purkinje cells of the cerebellar cortex, central cerebellar nu-
clei, pyramidal cells of the motor cortex, hippocampus, supe-
rior olivary nucleus, trapezoid body in the pons, and in the
sensory ganglia (Koves et al. 2002,2004; Heinzlmann et al.
2011). It has also become evident that secretin is synthetized
in the hypothalamic supraoptic (SO) and paraventricular (PV)
nuclei (Welch et al. 2004; Chu et al. 2009), and it regulates
water homeostasis (Chu et al. 2009). It was also shown that
intravenous (IV), intracerebroventricular (ICV), or intranasal
(IN) administration of secretin influenced the behavior of rats,
mice, and humans. In rats, its ICV administration decreased
the open field locomotor activity and the novel object ap-
proach (Charlton et al. 1983). In a special mouse model
(Japanese waltzing mice), ICV and IN secretin attenuated the
hyperactive repetitive movement (Koves et al. 2011;
Heinzlmann et al. 2012). In autistic children, a single IV
secretin injection improved eye contact, alertness, and expan-
sion of expressive language (Horvath et al. 1998).
Question arises concerning the site of action of secretin.
It is likely that secretin exerts its effect in places where (1) it
induces neural activity (appearance of c-fos) and (2) where
its receptors occur.
1. IV infusion of secretin into rats induced c-fos gene ex-
pression in the central amygdala, area postrema, the bed
nucleus of the stria terminalis (BNST), external lateral
parabrachial nucleus, and SO (Goulet et al. 2003). After
ICV administration of secretin, many regions, such as
several brain stem, hypothalamic, and limbic structures
(septum and amygdala) and the medial bank of the ante-
rior prefrontal, orbitofrontal, and piriform cortex, showed
activation of Fos protein in awake, freely moving rats
(Welch et al. 2003). It is clear from the above-mentioned
data that secretin influences c-fos expression in the struc-
tures that are involved in behavior, stress adaptation, and
visceral responses.
2. Secretin receptors in the central nervous system were first
demonstrated by Fremeau and his coworkers (1983).
They applied radiolabeled secretin binding assay using
tissue homogenate. The highest level of the secretin re-
ceptor was observed in the cerebellum, and a gradually
decreasing amount was detected in the frontal cortex,
striatum, hippocampus, thalamus, hypothalamus, medul-
la/pons, and midbrain. The above-mentioned data sug-
gested a relationship between secretin receptors and
gamma-aminobutyric acid (GABA) receptors. Indeed,
two decades later, it was demonstrated that, in the cere-
bellum, secretin facilitated the GABA-ergic inhibitory
input onto Purkinje cells via a postsynaptic and cAMP-
dependent mechanism as a retrograde messenger (Yung et
al. 2001,2006). An in vitro autoradiographic investiga-
tion suggested a different distribution (Nozaki et al.
2002). The radiolabeled secretin was bound in higher
concentration to the nucleus of solitary tract (NST) than
in the laterodorsal thalamic and accumbens nuclei. Mod-
erate binding was found in the orbital, cingulate, piriform,
frontal, parietal, and entorhinal cortices, caudate/putamen,
hippocampus, lateral septal nucleus, olfactory bulb,
amygdala, hypothalamus, pineal body, pituitary gland,
dorsal raphe nucleus, locus coeruleus, and cerebellum.
Weak binding was seen in the corpus callosum. A re-
search group (Tay et al. 2004) studied expression levels
of secretin and secretin receptor mRNA in several brain
regions of rat ranging in age from postnatal days 7 to 60
by quantitative real-time PCR. Both secretin and its re-
ceptor showed highest expression levels at postnatal days
7 and 14 compared to later time points. Interestingly,
secretin receptor mRNA was most abundant in the cere-
bellum, while secretin mRNA expression was strongest in
the NST. In situ hybridization using digoxigenin-labeled
probe was used to identify secretin receptor mRNA-
expressing neurons in the Purkinje and basket cells of
the cerebellum and in the deep cerebellar nuclei (Yung
et al. 2006), as well as in the magno- and parvocellular
PV, SO, and in the vascular organ of the lamina terminalis
(VOLT) (Chu et al. 2009; Lee et al. 2010). In our recent
study, using radioactive labeling of the secretin receptor
probe, we described secretin receptor mRNA in all layers
of the cerebellar cortex of male rats (molecular, Purkinje
cell, and granule cell layers in both vermis and hemi-
spheres) (Heinzlmann et al. 2012).
The aim of our present work was to precisely map the
secretin receptor mRNA-expressing neurons in the forebrain
and brain stem by in situ hybridization histochemistry, giving
a high morphological resolution. We applied radioactive la-
beling of the secretin receptor probe by [
35
S]-uracil triphos-
phate (UTP) incorporated by in vitro transcription to achieve a
maximal sensitivity.
Experimental Procedures
Animals
Adult male Wistar rats (Semmelweis University, Budapest,
Hungary) weighing approximately 400 g were used for our
experiments. The rats were kept in a temperature-controlled
vivarium (22 ± 2 °C). The lights were on at 7:00 a.m. and off
at 7:00 p.m. The animals were fed with standard lab chow and
tap water ad libitum. Five animals were decapitated; the brains
J Mol Neurosci (2013) 50:172178 173
were immediately removed, frozen on dry ice, and stored
at 70 °C until use. The animals were treated according to
the rules of the European Communities Council Directive
(86/609/EEC), permission no. 22.1/1158/2010.
Tissue Sectioning
The tissue blocks were warmed up to 20 °C, and 12-μm-
thick serial coronal sections were cut on cryostat (Cryotome,
Thermo Shandon, Pittsburg, PA). The sections were mounted
on Superfrost Plus slides (Thermo Scientific, Budapest,
Hungary), dried on a hot plate (37 °C), and then stored
at 70 °C again until use.
In situ Hybridization
Probe Preparation
The preparation of the probe was a critical step for the suc-
cessful assay. A vector (CDM8) containing the complete rat
secretin receptor cDNA (1,796 bp) was kindly provided by Dr.
Hashimoto and Dr. Baba (Ishihara et al. 1991). A 453-bp-long
fragment according to 7831,235 bp of the original cDNA
was subcloned into a pBluescript II SK (+) vector and verified
by sequencing. Antisense and sense cRNA probes, labeled by
[
35
S]-UTP and used for in situ hybridization, were produced
by in vitro transcription using the MAXIscript in vitro tran-
scription kit (Invitrogen, Budapest, Hungary).
Hybridization and Visualization
Before hybridization, sections were fixed in 4 % paraformal-
dehyde dissolved in phosphate-buffered saline (PBS) (pH
7.4), washed in PBS, and then treated with 0.25 % acetic
anhydride in 0.1 M triethanolamine HCl (pH 8.0) for
10 min. The sections were then rinsed in ×2 sodium chlo-
ridesodium citrate buffer (SSC, pH 7.0), dehydrated and
delipidated in a subsequent series of 70-85-95-100-95 % eth-
anol, and finally air dried. Hybridization was performed over-
night at 55 °C with 10
6
cpm/slide radioactively labeled
secretin receptor riboprobe in a humid chamber. On the fol-
lowing day, the sections were washed in ×4 SSC buffer for 4 ×
5 min at room temperature; then, they were treated in RnaseA
(20 μg/ml; Sigma, Budapest, Hungary) buffer (pH 8.0) con-
taining 500 mM NaCl, 10 mM TrisHCl, and 0.25 mM ethi-
lenediaminetetraacitic acid for 30 min at 37 °C and then for
5 min in each of the following buffer solutions: ×2, ×1, and
×0.5 SSC at room temperature and finally in ×0.1 SSC at
65 °C for 2 × 30 min. Then, the slides were let to cool down
and were washed in PBS. The slides were dipped into NTB
nuclear track emulsion (Carestream Health Inc., Rochester,
NY). After 8 weeks of exposition time at 4 °C in dark, the
reaction was developed using Kodak Dektol developer and
fixer (Sigma) at 18 °C. The slides were counterstained with
Giemsa solution, dried, and coverslipped with DePeX (all
from Sigma).
Results
Secretin Receptors in the Forebrain
With the use of antisense probe, secretin receptors were found
in many regions of the forebrain; however, there was no signal
using the sense probe. We observed a considerable number of
positive cells in the VOLT (Fig. 1a, b). Secretin receptors were
also found in the hypothalamic magnocellular nuclei, SO
(Fig. 1c, d), and PV (Fig. 1e, f), but not in the anterior
commissural nucleus. The number of silver grain containing
cells was greater in the SO than in the PV.
Scattered labeled cells were seen in the median frontal
gyrus, head of the caudate nucleus, perifornical region, ento-
rhinal cortex, and lateral hypothalamic area (not shown).
There was no labeling in the BNST, septum, amygdala, and
Fig. 1 Microphotographs demonstrating secretin receptor mRNA ex-
pression in hypothalamic frontal sections. a,bVascular organ of the
lamina terminalis; c,dsupraoptic nucleus; e,fparaventricular nucleus.
b,d, and fHigh power details of a,c, and e, respectively. Arrows
indicate secretin receptor mRNA-expressing cells. Asterisks indicate
same structures in aand b,c, and das well as in eand f, respectively.
3Vthird ventricle, Ffornix, OX optic chiasm, PRE preoptic recess, PV
hypothalamic paraventricular nucleus, SO supraoptic nucleus, VOLT
vascular organ of the lamina terminalis. Scale 0150 μmina,c, and e
and 30 μminb,d, and f
174 J Mol Neurosci (2013) 50:172178
hippocampus. In the forebrain, the densest labeling was seen
in the laterodorsal thalamic nucleus (Fig. 2a, b). Significant
number of labeled cells was seen in the lateral habenula
(Fig. 2c, d).
Secretin Receptors in the Brain Stem
The density of secretin receptors was very high in NST
(Fig. 3a, b). A few weakly labeled cells were also seen in
the second-order sensory neurons of the spinal trigeminal
nucleus (not shown).
Discussion
On the basis of previous results (Yung et al. 2001,2006;
Koves et al. 2002,2004), it is appropriate to accept secretin
as a neuropeptide synthetized in both central and peripheral
nervous systems. As it was mentioned in the introduction,
secretin receptors in the central nervous system were demon-
strated by the following several methods: radiolabeled secretin
binding assay using tissue homogenate, in vitro autoradio-
graphic investigation, quantitative real-time PCR, and in situ
hybridization using digoxigenin-labeled probe (Fremeau et al.
1983; Nozaki et al. 2002; Tay et al. 2004). Our paper is the
first in which secretin receptors were demonstrated in the
forebrain and brain stem using radioactive in situ hybridiza-
tion. This method is very sensitive and gives high morpho-
logical resolution, enabling identification of the cell groups
where secretin receptor mRNA is present.
The question arises: what may be the role of secretin recep-
tors in the various regions of the forebrain and brain stem. One
of the regions, where we observed strong in situ hybridization
signal, was the laterodorsal thalamic nucleus. This is in agree-
ment with the earlier findings (Nozaki et al. 2002). The later-
odorsal thalamic nucleus receives information from the visual
cortex and projects to the limbic system. It plays a role in spatial
learning and memory (van Groen et al. 2002).
A very dense secretin receptor mRNA labeling was also
seen in the NST, confirming previous data (Nozaki et al.
2002; Tay et al. 2004). The NST relays gustatory, olfactory,
and visceral sensory information toward upper brain centers.
Afferent fibers from cranial nerves VII, IX, and X convey
taste to its rostral portion and general visceral sense to its
caudal part. This nucleus is also an important autonomic
regulatory center (Garcia-Diaz et al. 1988). Secretin there-
fore may influence all of the above-mentioned functions
through its receptors in the NST.
VOLT is another region where secretin and its receptors
are present. As it was shown, secretin through the magno-
cellular neurons in the PV and SO, which also express
secretin receptor mRNA, has a crucial role in the water
homeostasis (Chu et al. 2009). The strong secretin receptor
mRNA expression in the VOLT supports this hypothesis.
VOLT possesses osmoreceptors and is involved in osmoti-
cally stimulated vasopressin release, as well as in central
hyperosmolality-induced increases of sympathetic nerve ac-
tivity and arterial blood pressure (McKinley et al. 2004; Shi
et al. 2007). Secretin increased vasopressin release from
hypothalamic explants, and dehydration evoked secretin
release from the posterior pituitary to the systemic circula-
tion (Chu et al. 2009). This research group also showed
secretin and secretin receptor transcripts in the VOLT using
real-time PCR and digoxigenin-labeled riboprobe in in situ
hybridization assay (Lee et al. 2010). They also demon-
strated that angiotensin II exerts its effect through secretin and
its receptors on water homeostasis using secretin and secretin
knockout mice. Our morphological results support this
hypothesis.
Fig. 2 Microphotographs demonstrating secretin receptor mRNA ex-
pression in the laterodorsal thalamic nucleus in the forebrain. a,bLater-
odorsal thalamic nucleus; c,dlateral habenular nucleus. b,dHigh power
details of aand c, respectively. Arrows show secretin receptor mRNA-
expressing cells. Asterisks indicate same structures in aand bas well as in
cand d,respectively.3Vthird ventricle, Hipp hippocampus, LHN lateral
habenular nucleus, LD laterodorsal thalamic nucleus. Scale 0150 μmina
and cand 30 μminband d
Fig. 3 Microphotographs demonstrating secretin receptor mRNA ex-
pression in the medulla oblongata (a,b). bHigh power detail of a.Arrows
show secretin receptor mRNA-expressing cells. Asterisks indicate same
structures in aand b.4Vfourth ventricle, GCL granule cell layer, MOL
molecular cell layer, NST nucleus of the solitary tract. Scale 0150 μmina
and cand 30 μminband d
J Mol Neurosci (2013) 50:172178 175
In this study, we not only confirmed previous data but also
identified so far unrecognized regions in the brain, where
secretin receptor mRNA is expressed. These are the lateral
habenular nucleus, perifornical and lateral hypothalamic
areas, and spinal trigeminal nucleus. The central axons of
the secretin immunoreactive primary sensory neurons of the
trigeminal ganglion terminate in the spinal trigeminal nucleus
(Heinzlmann et al. 2011). This observation well correlates
with our new observation that this nucleus expresses secretin
receptors. Our morphological data support the physiological
observation that secretin counteracts the analgesic effect of
morphine (Babarczy et al. 1995).
In our recently published paper (Heinzlmann et al. 2012),
we described many strongly labeled cells in the granule cell
layer, a few scatteredcells in the molecular cell layer, and only
very rare labeling in the Purkinje cell layer of the cerebellar
cortex in male rats. The labeling in the granule cell layer
suggests that the receptors may also be present in those
GABA-ergic cells which participate in forming the cerebellar
glomeruli. Yung and his coworkers (2001, 2006) described
strong secretin receptor mRNA expression in the Purkinje
cells, a weaker in the basket cells in the molecular cell layer,
and no labeling in the granule cell layer. However, they used
nonradioactive in situ hybridization method and worked on
SpragueDawley rats, while we used radioactive in situ hy-
bridization and Wistar rats. Additionally, their probe was
specific to the region according to 213639 bp of rat secretin
receptor cDNA, (GenBank accession no. NM031115), while
ours contained a nonoverlapping region (7831,235 bp).
Therefore, the different results can also be explained by the
existence of putative splice variants of the secretin receptor in
the brain. Although splice variants were not yet identified in
VOLT
P
CC
OX
P
AC
CC
OX
PV
LHN
Cer
NST
CPu LD
AC
Cer
Lateral 0.10 mm
Lateral 0.40 mm
Lateral 2.62 mm
Lateral 1.40 mm
POT
AC
SO
CC
LH
FNST
Cer
LD
Sp5
CC
CPu
Fig. 4 Schematic illustration of the occurrence and density of secretin
receptor mRNA expression in sagittal sections of the brain, according to
stereotaxic coordinates of Paxinos and Watson (2007). Distance from the
midline is labeled in millimeters. The number of asterisks indicates the
density of the signal. AC anterior commissure, CC corpus callosum, Cer
cerebellar hemisphere. Single and quadruple asterisks indicate molecular
and granule cell layers, respectively; CPu caudate putamen, Fperifornical
area, LH lateral hypothalamus, LHN lateral habenular nucleus, LD later-
odorsal thalamic nucleus, OT optic tract, OX optic chiasm, Ppituitary, PV
hypothalamic paraventricular nucleus, Sp5 spinal trigeminal nucleus,
NST nucleus of the solitary tract, SO supraoptic nucleus, VOLT vascular
organ of the lamina terminalis. The data concerning the cerebellum
derives from the paper of Heinzlmann et al. (2012)
Table 1 Summary of
the distribution of
secretin receptor
mRNA-expressing
neurons in the forebrain,
brain stem, and
cerebellum
Semiquantitative analy-
sis of the in situ
hybridization signal:
+ occasional, ++ week,
+++ moderate,
++++ strong
labeling, no labeling.
The data concerning the
cerebellum were
published in our previ-
ous paper (Heinzlmann
et al. 2012)
Forebrain structures
median frontal gyrus +
entorhinal cortex +
VOLT +++
SO ++
PV +
perifornical area +
lateral hypothalamus +
BNST
septum
laterodorsal thalamus ++++
lateral habenular nucleus +++
amygdala
hippocampus
head of caudate nucleus +
Brain stem structures
nucleus of the solitary tract ++++
spinal trigeminal nucleus +
Cerebellum
vermis +
molecular cell layer +
granule cell layer ++++
176 J Mol Neurosci (2013) 50:172178
healthy tissues, they were demonstrated in bronchopulmonary
carcinoid tumors by autoradiography and real-time PCR
(Korner et al. 2008). Since there were other areas in the brain,
where secretin binding was found by others like in the septum,
amygdala, and hippocampus (Fremeau et al. 1983;Nozakiet
al. 2002), we did not see any labeling. Therefore, we suppose
that the expression of the secretin receptor in these areas was
below the detection level in rats (400 g bw) we used. Nozaki
and Fremeau examined much younger animals, with a body
weight of 160180 and 200250 g, respectively (Fremeau et
al. 1983;Nozakietal.2002). As it has been demonstrated that
secretin receptor mRNA expression decreases with age, it is
especially low, for example, in the amygdala and hippocam-
pus, by the age of 60 days (Tay et al. 2004).
The lateral habenular nucleus projects to the midbrain
dopaminergic systems and may be involved in reward-
seeking behaviors (Maia 2009). The lateral hypothalamic
and perifornical areas participate in several functions like the
sleepwake and food intake regulation and regulation of the
body fluid homeostasis as well as in depression, anxiety, and
reward (Johnson and Thunhorst 1997; Valassi et al. 2008;
Chung et al. 2011; Kitka et al. 2011). Thus, secretin may be
associated with all of these roles.
Finally, it was concluded that the secretin receptors show a
more wide distribution in the brain than it was known before.
Our present and previous data (Heinzlmann et al. 2012)
obtained by radioactive in situ hybridization are semiquanti-
tatively illustrated and summarized in Fig. 4and Table 1,
respectively. The newly identified secretin receptor mRNA-
expressing areas are partly related to functions of secretin
previously accepted but also raise the possibility that secretin
may be related with new functions.
Acknowledgments We are grateful to Mrs. Anna Takács and Judit
Kerti for their excellent technical assistance. This work was supported by
ETT grant 495/09 to ZE Tóth and by the Department of Human
Morphology and Developmental Biology, Semmelweis University.
References
Babarczy E, Szabo G, Telegdy G (1995) Effects of secretin on acute
and chronic effects of morphine. Pharmacol Biochem Behav
51:469472
Charlton CG, ODonohue TL, Miller RL, Jacobowitz DM (1981)
Secretin immunoreactivity in rat and pig brain. Peptides 2
(Suppl 1):4549
Charlton CG, Miller RL, Crawley JN, Handelmann GE, ODonohue
TL (1983) Secretin modulation of behavioral and physiological
functions in the rat. Peptides 4:739742
Chu JY, Lee LT, Lai CH, Vaudry H, Chan YS, Yung WH, Chow BK
(2009) Secretin as a neurohypophysial factor regulating body
water homeostasis. Proc Natl Acad Sci U S A 106:1596115966
Chung S, Parks GS, Lee C, Civelli O (2011) Recent updates on the
melanin-concentrating hormone (MCH) and its receptor system:
lessons from MCH1R antagonists. J Mol Neurosci 43:115121
Fremeau RT Jr, Jensen RT, Charlton CG, Miller RL, ODonohue TL,
Moody TW (1983) Secretin: specific binding to rat brain mem-
branes. J Neurosci 3:16201625
Garcia-Diaz DE, Jimenez-Montufar LL, Guevara-Aguilar R, Wayner
MJ, Armstrong DL (1988) Olfactory and visceral projections to
the nucleus of the solitary tract. Physiol Behav 44:619624
Goulet M, Shiromani PJ, Ware CM, Strong RA, Boismenu R, Rusche
JR (2003) A secretin i.v. infusion activates gene expression in the
central amygdala of rats. Neuroscience 118:881888
Heinzlmann A, Toth ZE, Koves K (2011) Secretin mRNA in the
subdivision of primary sensory neurons in the trigeminal ganglion
of rats. J Mol Neurosci 43:101108
Heinzlmann A, Kiss G, Tóth ZE et al (2012) Intranasal application of
secretin, similarly to intracerebroventricular administration, influ-
ences the motor behavior of mice probably through specific
receptors. J Mol Neurosci 48(3):558564
Horvath K, Stefanatos G, Sokolski KN, Wachtel R, Nabors L, Tildon JT
(1998) Improved social and language skills after secretin adminis-
tration in patients with autistic spectrum disorders. J Assoc Acad
Minor Phys 9:915
Ishihara T, Nakamura S, Kaziro Y, Takahashi T, Takahashi K, Nagata S
(1991) Molecular cloning and expression of a cDNA encoding the
secretin receptor. EMBO J 10:16351641
Johnson AK, Thunhorst RL (1997) The neuroendocrinology of thirst
and salt appetite: visceral sensory signals and mechanisms of
central integration. Front Neuroendocrinol 18:292353
Kitka T, Adori C, Katai Z et al (2011) Association between the
activation of MCH and orexin immunorective neurons and REM
sleep architecture during REM rebound after a three day long
REM deprivation. Neurochem Int 59:686694
Korner MU, Hayes GM, Carrigan PE, Rehmann R, Miller LJ, Reubi
JC (2008) Wild-type and splice-variant secretin receptors in lung
cancer: overexpression in carcinoid tumors and peritumoral lung
tissue. Mod Pathol 21:387395
Koves K, Kausz M, Reser D, Horvath K (2002) What may be the
anatomical basis that secretin can improve the mental functions in
autism? Regul Pept 109:167172
Koves K, Kausz M, Reser D et al (2004) Secretin and autism: a basic
morphological study about the distribution of secretin in the nervous
system. Regul Pept 123:209216
Koves K, Kiss G, Heinzlmann A et al (2011) Secretin attenuates the
hereditary repetitive hyperactive movements in a mouse model. J
Mol Neurosci 43:109114
Lee VH, Lee LT, Chu JY, Lam IP, Siu FK, Vaudry H, Chow BK (2010)
An indispensable role of secretin in mediating the osmoregulatory
functions of angiotensin II. FASEB J 24:50245032
Maia TV (2009) Reinforcement learning, conditioning, and the brain:
successes and challenges. Cogn Affect Behav Neurosci 9:343364
McKinley MJ, Mathai ML, McAllen RM et al (2004) Vasopressin secre-
tion: osmotic and hormonal regulation by the lamina terminalis. J
Neuroendocrinol 16:340347
Nozaki S, Nakata R, Mizuma H, Nishimura N, Watanabe Y, Kohashi
R, Watanabe Y (2002) In vitro autoradiographic localization of
(125)i-secretin receptor binding sites in rat brain. Biochem
Biophys Res Commun 292:133137
Paxinos G, Watson C (eds) (2007) The rat brain in stereotaxic coor-
dinates. Academic Press, San Diego
Shi P, Stocker SD, Toney GM (2007) Organum vasculosum laminae
terminalis contributes to increased sympathetic nerve activity
induced by central hyperosmolality. Am J Physiol Regul Integr
Comp Physiol 293:R2279R2289
Sundler F, Hakanson R (1988) Peptide hormone-producing endocrine/
paracrine cells in the gastro-entero-pancreatic region. In: Bjorklund
A, Hokfelt T, Owman C (eds) The peripheral nervous system.
Handbook of chemical neuroanatomy. Elsevier, Amsterdam, pp
219295
J Mol Neurosci (2013) 50:172178 177
Tay J, Goulet M, Rusche J, Boismenu R (2004) Age-related and
regional differences in secretin and secretin receptor mRNA levels
in the rat brain. Neurosci Lett 366:176181
Valassi E, Scacchi M, Cavagnini F (2008) Neuroendocrine control of
food intake. Nutr Metab Cardiovasc Dis 18:158168
van Groen T, Kadish I, Wyss JM (2002) The role of the laterodorsal
nucleus of the thalamus in spatial learning and memory in the rat.
Behav Brain Res 136:329337
Welch MG, Keune JD, Welch-Horan TB, Anwar N, Anwar M,
Ruggiero DA (2003) Secretin activates visceral brain regions in
the rat including areas abnormal in autism. Cell Mol Neurobiol
23:817837
Welch MG, Keune JD, Welch-Horan TB, Anwar N, Anwar M, Ludwig RJ,
Ruggiero DA (2004) Secretin: hypothalamic distribution and hypoth-
esized neuroregulatory role in autism. Cell Mol Neurobiol 24:219241
Yung WH, Leung PS, Ng SS, Zhang J, Chan SC, Chow BK (2001)
Secretin facilitates GABA transmission in the cerebellum. J
Neurosci 21:70637068
Yung WH, Chan YS, Chow BK, Wang JJ (2006) The role of secretin in
the cerebellum. Cerebellum 5:4348
178 J Mol Neurosci (2013) 50:172178
... SCT elicits its regulatory functions following it is binding to the signaling form of its receptor, SCTR, which was previously identified in the hypothalamus using in vitro autoradiography 46 , in situ hybridization 47 , and RNA sequencing 48 . Here, we further confirmed the presence of SCTR in VMH by reanalyzing the published single-cell RNA sequencing dataset from a mouse model 49 (Supplementary Fig. 12) and Arrows indicate lipids in bone marrow. ...
Article
Full-text available
Secretin, though originally discovered as a gut-derived hormone, is recently found to be abundantly expressed in the ventromedial hypothalamus, from which the central neural system controls satiety, energy metabolism, and bone homeostasis. However, the functional significance of secretin in the ventromedial hypothalamus remains unclear. Here we show that the loss of ventromedial hypothalamus-derived secretin leads to osteopenia in male and female mice, which is primarily induced by diminished cAMP response element-binding protein phosphorylation and upregulation in peripheral sympathetic activity. Moreover, the ventromedial hypothalamus-secretin inhibition also contributes to hyperphagia, dysregulated lipogenesis, and impaired thermogenesis, resulting in obesity in male and female mice. Conversely, overexpression of secretin in the ventromedial hypothalamus promotes bone mass accrual in mice of both sexes. Collectively, our findings identify an unappreciated secretin signaling in the central neural system for the regulation of energy and bone metabolism, which may serve as a new target for the clinical management of obesity and osteoporosis.
... Our current knowledge about secretin signaling in the brain (Zhang and Chow, 2014) is limited. In situ hybridization histochemistry explored the distribution of secretin receptor mRNA-expressing cells in the brain, including the OVLT region (Toth et al., 2013). The release of secretin from the hypothalamus has been reported earlier (Chu et al., 2006). ...
Article
Full-text available
Rising serum estradiol triggers the surge release of gonadotropin-releasing hormone (GnRH) at late proestrus leading to ovulation. We hypothesized that proestrus evokes alterations in peptidergic signaling onto GnRH neurons inducing a differential expression of neuropeptide-, growth factor-, and orphan G-protein-coupled receptor (GPCR) genes. Thus, we analyzed the transcriptome of GnRH neurons collected from intact, proestrous and metestrous GnRH-green fluorescent protein (GnRH-GFP) transgenic mice using Affymetrix microarray technique. Proestrus resulted in a differential expression of genes coding for peptide/neuropeptide receptors including Adipor1, Prokr1, Ednrb, Rtn4r, Nmbr, Acvr2b, Sctr, Npr3, Nmur1, Mc3r, Cckbr , and Amhr2 . In this gene cluster, Adipor1 mRNA expression was upregulated and the others were downregulated. Expression of growth factor receptors and their related proteins was also altered showing upregulation of Fgfr1, Igf1r, Grb2, Grb10 , and Ngfrap1 and downregulation of Egfr and Tgfbr2 genes. Gpr107 , an orphan GPCR, was upregulated during proestrus, while others were significantly downregulated ( Gpr1, Gpr87, Gpr18, Gpr62, Gpr125, Gpr183, Gpr4 , and Gpr88 ). Further affected receptors included vomeronasal receptors ( Vmn1r172, Vmn2r-ps54 , and Vmn1r148 ) and platelet-activating factor receptor ( Ptafr ), all with marked downregulation. Patch-clamp recordings from mouse GnRH-GFP neurons carried out at metestrus confirmed that the differentially expressed IGF-1, secretin, and GPR107 receptors were operational, as their activation by specific ligands evoked an increase in the frequency of miniature postsynaptic currents (mPSCs). These findings show the contribution of certain novel peptides, growth factors, and ligands of orphan GPCRs to regulation of GnRH neurons and their preparation for the surge release.
... Sctr is a member of the type II G-protein-coupled receptor family, which is mainly expressed in the pancreas [7], stomach [8], and kidneys [9]. It was also reported that it is expressed in several regions of the brain, including the cerebellum, hippocampus, central amygdala, median frontal gyrus, entorhinal cortex, hypothalamic paraventricular nucleus, perifornical region, lateral hypothalamic area, head of the caudate nucleus, and spinal trigeminal nucleus [6,10,20]. However, it has not been ascertained whether secretin receptors are expressed in the SCN. ...
Article
In mammals, the timing of behavior and physiological activity is controlled by the suprachiasmatic nucleus (SCN) in the hypothalamus. Incidentally, secretin is a peptide hormone that promotes digestive activities and regulates water reabsorption. In recent studies, exogenous administration of secretin has been reported to induce secretion of oxytocin in the supraoptic nucleus of the hypothalamus and modulate social behavior. These results indicate that secretin is involved in the neural network that controls social behavior and plays important roles in the central nervous system. In the present study, we investigated the effects of secretin on circadian rhythms, by assessing circadian rhythms during wheel-running behavior in secretin receptor-deficient (Sctr-/-) mice. Male adult wild-type (WT) and Sctr-/- mice were housed in separate cages containing a wheel. Every minute of the wheel-running activity was monitored during the normal light-dark (LD) cycle (12:12 h) and in constant darkness (DD). Significant differences were observed in the free-running period between the WT and Sctr-/- mice. However, no significant differences were observed in the daily wheel-running revolutions between WT and Sctr-/- mice, in the LD and DD conditions. Moreover, the ratio of the daily activity phase to the rest phase (α/ρ) was significantly smaller in Sctr-/- than that in WT mice in the DD condition. Secretin receptors were expressed in the SCN cells. These findings suggest that secretin receptors are involved in the central circadian clock in the SCN and the circadian system in general.
... Dense labeling of secretin receptor was observed in the NTS and in the laterodorsal nucleus of the thalamus. Expression of secretin receptor was also found in the hypothalamus (Toth et al., 2013). Intracerebroventricular injection of secretin increased the expression of c-Fos in several brain regions including the area postrema, medial region of the NTS, paraventricular nucleus, and various cortical areas indicating a central action of the hormone in rats. ...
Article
Full-text available
In mammals, reproduction is regulated by a wide range of metabolic hormones that maintain the proper energy balance. In addition to regulating feeding and energy expenditure, these metabolic messengers also modulate the functional performance of the hypothalamic-pituitary-gonadal (HPG) axis. Secretin, a member of the secretin-glucagon-vasoactive intestinal peptide hormone family, has been shown to alter reproduction centrally, although the underlying mechanisms have not been explored yet. In order to elucidate its central action in the neuroendocrine regulation of reproduction, in vitro electrophysiological slice experiments were carried out on GnRH-GFP neurons in male mice. Bath application of secretin (100 nM) significantly increased the frequency of the spontaneous postsynaptic currents (sPSCs) to 118.0 ± 2.64% compared to the control, and that of the GABAergic miniature postsynaptic currents (mPSCs) to 147.6 ± 19.19%. Resting membrane potential became depolarized by 12.74 ± 4.539 mV after secretin treatment. Frequency of evoked action potentials (APs) also increased to 144.3 ± 10.8%. The secretin-triggered elevation of the frequency of mPSCs was prevented by using either a secretin receptor antagonist (3 μM) or intracellularly applied G-protein-coupled receptor blocker (GDP-β-S; 2 mM) supporting the involvement of secretin receptor in the process. Regarding the actions downstream to secretin receptor, intracellular blockade of protein kinase A (PKA) with KT-5720 (2 μM) or intracellular inhibition of the neuronal nitric oxide synthase (nNOS) by NPLA (1 μM) abolished the stimulatory effect of secretin on mPSCs. These data suggest that secretin acts on GnRH neurons via secretin receptors whose activation triggers the cAMP/PKA/nNOS signaling pathway resulting in nitric oxide release and in the presynaptic terminals this retrograde NO machinery regulates the GABAergic input to GnRH neurons.
... After hybridizations, sections were apposed to a BAS-MS imaging plate (Fuji Photo Film Co., Ltd., Kanagawa, Japan, NJ) for 2 (DRs) and 7 (TH) days, and then data were read out by a Fujifilm FLA-8000 Image Analyzer. Sections with D1R and D2R labeling were dipped in Kodak NTB nuclear emulsion (Carestream Health Inc., Rochester, NY) for 5 days according to the manufacturer's instructions and developed using Kodak developer and fixer (Sigma) [29,30].The sections were stained with Giemsa (Sigma) except those with prior Fos immunohistochemistry. ...
Article
Full-text available
Background/objectives: Dysfunction in reward-related aspects of feeding, and consequent overeating in humans, is a major contributor to obesity. Intrauterine undernutrition and overnutrition are among the predisposing factors, but the exact mechanism of how overeating develops is still unclear. Consummatory behavior is regulated by the medial shell (mSh) of the accumbens nucleus (Nac) through direct connections with the rostral part of the lateral hypothalamic area (LHA). Our aim was to investigate whether an altered Nac-LHA circuit may underlie hyperphagic behavior. Subjects/methods: Intrauterine protein-restricted (PR) male Wistar rats were used as models for hyperphagia. The experiments were performed using young adult control (normally nourished) and PR animals. Sweet condensed milk (SCM) served as a reward to test consumption and subsequent activation (Fos+) of Nac and LHA neurons. Expression levels of type 1 and 2 dopamine receptors (D1R, D2R) in the Nac, as well as tyrosine hydroxylase (TH) levels in the ventral tegmental area, were determined. The D1R agonist SKF82958 was injected into the mSh-Nac of control rats to test the effect of D1R signaling on SCM intake and neuronal cell activation in the LHA. Results: A group of food reward-representing D1R+ neurons was identified in the mSh-Nac. Activation (Fos+) of these neurons was highly proportional to the consumed palatable food. D1R agonist treatment attenuated SCM intake and diminished the number of SCM-activated cells in the LHA. Hyperphagic PR rats showed increased intake of SCM, reduced D1R expression, and an impaired response to SCM-evoked neuronal activation in the mSh-Nac, accompanied by an elevated number of Fos+ neurons in the LHA compared to controls. Conclusions: Sensitivity of food reward-representing neurons in the mSh-Nac determines the level of satisfaction that governs cessation of consumption, probably through connections with the LHA. D1R signaling is a key element in this function, and is impaired in obesity-prone rats.
... Using the more sensitive auto-radiographic binding approach, secretinbinding sites have been found in brain stem, hippocampus, caudate, cerebellum, cingulate nuclei, and orbital cortex (Nozaki et al., 2002). Consistently, the region-specific RT-PCR study has identified secretin receptor within cerebellum, hippocampus, NTS, lateral dorsal thalamic nucleus, lamina terminalis, lateral habenular complex, supraoptic nucleus (SON) and paraventricular nucleus (PVN) of hypothalamus, and CeA (Tay et al., 2004;Toth et al., 2013). Generally speaking, secretin receptor presents a much wider spatial distribution than secretin ligand (Ng et al., 2002), indicating pleiotropic roles of secretin inside the brain via specific receptor binding within certain regions. ...
Article
Full-text available
Secretin is a polypeptide hormone initially identified for its gastrointestinal functions. However, emerging evidences show wide distribution of secretin and secretin receptor across various brain regions from cerebral cortex, hippocampus, hypothalamus to cerebellum. In this mini review, we will firstly describe the region-specific expression pattern of secretin and secretin receptor in the brain, followed by a summary of central physiological and neurological functions mediated by secretin. Using genetic manipulation and pharmaceutical approaches, one can elucidate the role of secretin in mediating various neurological functions from simple behaviors, such as water and food intake, to more complex functions including emotion, motor, and learning or memory. At last, current weakness and future perspectives of secretin in the central nervous system will be discussed, aiming to provide the potency of using secretin or its analog for treating various neurological disorders.
... The rat secretin receptor was found widely distributed in the brain when examined by in situ hybridization. 17 Secretin receptors were found in the Purkinje cells of the cerebellum and other areas of the human brain. 18 Secretin receptors are also present in gastrinoma cells, 19 which is the basis of its use in the diagnosis of Zollinger-Ellison syndrome. ...
Article
Objective: The aim of this study was to compare the hemodynamic parameters from the anesthesia records of children who underwent upper gastrointestinal endoscopy (esophagogastroduodenoscopy [EGD]) with and without secretin pancreatic function tests (sPFTs). Methods: The hemodynamic parameters were retrieved from an electronic anesthesia database. The secretin group consisted of 186 children, and the age- and sex-matched control group included 136 patients who did not have sPFTs. Results: There was no difference in the demographic parameters (age and sex) between the 2 groups. The secretin group had a lower height and body mass index. The sPFT resulted in an average 3-minute extension of the endoscopic procedure. The heart rate increased during the EGD in both groups and was higher (averaged 7 beats per minute) in the secretin group than the EGD-only group. There were mild elevations on the systolic and diastolic blood pressures. None of these changes were clinically significant. There were no complications reported during the anesthesia and procedures in the 2 groups. Conclusions: Secretin PFT is a safe procedure. It only slightly prolongs the total procedure and anesthesia time. There were no clinically significant changes in the vital parameters in the secretin group, and there were no adverse effects recorded.
... Secretin is synthesized in various brain regions including the hypothalamus, hippocampus, cerebellum, cerebral cortex and brainstem (18)(19)(20)(21). The secretin receptor is also distributed widely in the brain including the brainstem, cerebellum, cerebral cortex, hypothalamus and hippocampus (21)(22)(23)(24). Secretin receptor-deficient mice have been suggested to have deficits in social behavior (25). ...
Article
Full-text available
Background: Social recognition underlies social behavior in animals, and patients with psychiatric disorders associated with social deficits show abnormalities in social recognition. Oxytocin is implicated in social behavior and has received attention as an effective treatment for sociobehavioral deficits. Secretin receptor-deficient mice show deficits in social behavior. The relationship between oxytocin and secretin concerning social behavior remains to be determined. Methods: Expression of c-Fos in oxytocin neurons and release of oxytocin from their dendrites after secretin application were investigated. Social recognition was examined after intracerebroventricular or local injection of secretin, oxytocin, or an oxytocin receptor antagonist in rats, oxytocin receptor-deficient mice, and secretin receptor-deficient mice. Electron and light microscopic immunohistochemical analysis was also performed to determine whether oxytocin neurons extend their dendrites into the medial amygdala. Results: Supraoptic oxytocin neurons expressed the secretin receptor. Secretin activated supraoptic oxytocin neurons and facilitated oxytocin release from dendrites. Secretin increased acquisition of social recognition in an oxytocin receptor-dependent manner. Local application of secretin into the supraoptic nucleus facilitated social recognition, and this facilitation was blocked by an oxytocin receptor antagonist injected into, but not outside of, the medial amygdala. In the medial amygdala, dendrite-like thick oxytocin processes were found to extend from the supraoptic nucleus. Furthermore, oxytocin treatment restored deficits of social recognition in secretin receptor-deficient mice. Conclusions: The results of our study demonstrate that secretin-induced dendritic oxytocin release from supraoptic neurons enhances social recognition. The newly defined secretin-oxytocin system may lead to a possible treatment for social deficits.
... www.frontiersin.org Purkinje neuron, basket cell SCTR gene (17,19) Medulla oblongata NTS SCTR gene (10) a SCT peptide was found in pyramidal cells in cerebral cortex from humans, and colchicine-treated rats (7) but with less (11) or no forebrain expression (18) In concurrent histological and electrophysiological examinations, higher apoptosis of neural progenitor cells in DG during postnatal development occurred, along with impaired LTP (32). In summary, SCT was involved in hippocampal neurogenesis and neural transmission including synaptic plasticity, all of which contributed to social behavioral and spatial learning as illustrated in Figure 1B. ...
Article
Full-text available
Secretin (SCT) was firstly discovered as a gut peptide hormone in stimulating pancreatic secretion, while its novel neuropeptide role has drawn substantial research interests in recent years. SCT and its receptor (SCTR) are widely expressed in different brain regions, where they exert multiple cellular functions including neurotransmission, gene expression regulation, neurogenesis, and neural protection. As all these neural functions ultimately can affect behaviors, it is hypothesized that SCT controls multiple behavioral paradigms. Current findings support this hypothesis as SCT–SCTR axis participates in modulating social interaction, spatial learning, water and food intake, motor coordination, and motor learning behaviors. This mini-review focuses on various aspects of SCT and SCTR in hippocampus, hypothalamus, and cerebellum including distribution profiles, cellular functions, and behavioral phenotypes to elucidate the link between cellular mechanisms and behavioral control.
Article
Full-text available
Secretin and its receptors show wide distribution in the central nervous system. It was demonstrated previously that intravenous (i.v.) and intracerebroventricular (i.c.v.) application of secretin influenced the behavior of rat, mouse, and human. In our previous experiment, we used a special animal model, Japanese waltzing mice (JWM). These animals run around without stopping (the ambulation distance is very limited) and they do not bother with their environment. The i.c.v. secretin attenuated this hyperactive repetitive movement. In the present work, the effect of i.c.v. and intranasal (i.n.) application of secretin was compared. We have also looked for the presence of secretin receptors in the brain structures related to motor functions. Two micrograms of i.c.v. secretin improved the horizontal movement of JWM, enhancing the ambulation distance. It was nearly threefold higher in treated than in control animals. The i.n. application of secretin to the left nostril once or twice a day or once for 3 days more effectively enhanced the ambulation distance than i.c.v. administration. When secretin was given twice a day for 3 days it had no effect. Secretin did not improve the explorative behavior (the rearing), of JWM. With the use of in situ hybridization, we have found very dense secretin receptor labeling in the cerebellum. In the primary motor cortex and in the striatum, only a few labeled cells were seen. It was supposed that secretin exerted its effect through specific receptors, mainly present in the cerebellum.
Article
Full-text available
Fluid balance is critical to life and hence is tightly controlled in the body. Angiotensin II (ANGII), one of the most important components of this regulatory system, is recognized as a dipsogenic hormone that stimulates vasopressin (VP) expression and release. However, detailed mechanisms regarding how ANGII brings about these changes are not fully understood. In the present study, we show initially that the osmoregulatory functions of secretin (SCT) in the brain are similar to those of ANGII in mice and, more important, we discovered the role of SCT as the link between ANGII and its downstream effects. This was substantiated by the use of two knockout mice, SCTR(-/-) and SCT(-/-), in which we show the absence of an intact SCT/secretin receptor (SCTR) axis resulted in an abolishment or much reduced ANGII osmoregulatory functions. By immunohistochemical staining and in situ hybridization, the proteins and transcripts of SCT and its receptor are found in the paraventricular nucleus (PVN) and lamina terminalis. We propose that SCT produced in the circumventricular organs is transported and released in the PVN to stimulate vasopressin expression and release. In summary, our findings identify SCT and SCTR as novel elements of the ANGII osmoregulatory pathway in maintaining fluid balance in the body.
Article
Full-text available
It was previously demonstrated that secretin influenced the behavior of rats investigated by open-field test. In the present experiment, we have compared the effect of intracerebroventricular administration of 2 μg of secretin on the behavior of CFLP white and Japanese waltzing mice. These latter animals exhibit stereotypic circular movements. The effect of secretin on the horizontal (ambulation) and vertical movements (rearing and jumping) was investigated in open-field test. The ambulation time and distance were shorter, and the number of rearing and jumping were much lower in Japanese waltzing mice than in CFLP white mice during 30 min-experimental period. In white mice, 2 μg of secretin had no effect on the above-mentioned parameters; however, in Japanese waltzing mice, secretin enhanced the ambulation time and distance to the level of CFLP white mice, but did not influence the rearing and jumping. On the basis of the results, it was concluded that intracerebroventricularly administered secretin attenuated the stereotypic (circulating) movement and improved the horizontal movement indicated by the normalization of the ambulation time and distance; however, it did not influence the explorative behavior (rearing and jumping) in our special animal model.
Article
Full-text available
Melanin-concentrating hormone (MCH) is a 19-amino-acid cyclic peptide which was originally found to lighten skin color in fish that is highly conserved among many species. MCH interacts with two G-protein-coupled receptors, MCH1R and MCH2R, but only MCH1R is expressed in rodents. MCH is mainly synthesized in the lateral hypothalamus and zona incerta, while MCH1R is widely expressed throughout the brain. Thus, MCH signaling is implicated in the regulation of many physiological functions. The identification of MCH1R has led to the development of small-molecule MCH1R antagonists that can block MCH signaling. MCH1R antagonists are useful not only for their potential therapeutic value, but also for understanding the physiological functions of the endogenous MCH system. Here, we review the physiological functions of the MCH system which have been investigated using MCH1R antagonists such as food intake, anxiety, depression, reward, and sleep. This will help us understand the physiological functions of the MCH system and suggest some of the potential applications of MCH1R antagonists in human disorders.
Article
The effects of intracerebroventricularly (ICV) administered secretin on the analgesic, tolerance-inducing, and dependence-inducing actions of morphine were investigated, in adult, male CFLP mice. Secretin administered doses ICV did not itself affect pain sensitivity in a heat-radiant tail flick test. However, it depressed the acute nociceptive effect of a single subcutaneous (SC) dose of morphine (4 mg/kg) after ICV (1 or 10 ng/ animal) secretin administration. A dose of 10 ng secretin facilitated the development of acute morphine tolerance. On the other hand, none of the doses applied had any influence on chronic morphine tolerance, where animals were implanted SC with a morphine- containing pellet and the pain sensitivity was measured 3 days later. Morphine withdrawal signs were also evaluated by injecting naloxone. In a 100-ng dose, secretin increased the latency of the withdrawal jumping response; the peptide did not modify the other abstinence signs. These data suggest that central secretin administration can modify the analgesic effect of morphine.
Article
The primary sensory neurons use glutamate as a major neurotransmitter. Several neuropeptides are also found in these neurons. In our laboratory we demonstrated secretin-like immunoreactivity in primary sensory neurons of several species including human, rat and cat. In the present experiment utilizing in situ hybridization, we have demonstrated for the first time that secretin is not only immunostained but is also expressed in the primary sensory neurons of the trigeminal ganglion of male rats. In intact rats, secretin mRNA was not observed; we had to use intracerebroventricular colchicine administration to induce the expression of secretin. Secretin was expressed in about 5% of the cells in all the three subdivisions of the trigeminal ganglion. The secretin-synthetizing cells were large and medium sized, and their mean diameter was about 50 μm. When we compared the percentage and the size of secretin to that of calcitonin gene-related peptide (CGRP), substance-P (SP) and vasoactive intestinal polypeptide (VIP) cells, it was found that CGRP, SP and VIP are present in about 15-20% of the cells and their mean diameter is about 20-25 μm. The morphometric data indicate that secretin is present in a subdivision of neurons that is different from the subdivision of the CGRP, SP and VIP cells. It is suggested that secretin may modulate the function of the primary neurotransmitter.