ArticlePDF Available

Evidence for mediation of nociception by injection of the NK-3 receptor agonist, senktide, into the dorsal periaqueductal gray of rats

Authors:

Abstract and Figures

Ultrasound vocalizations (USVs) at approximately 22 kHz are usual components of the defensive response of rats. However, depending on the neural substrate that is activated, such as the dorsal periaqueductal gray (dPAG), USV emissions may be reduced. Activation of neurokinin-1 (NK-1)-mediated mechanisms of the dPAG causes analgesia, reduced 22 kHz USVs, and anxiogenic-like effects in rats exposed to the elevated plus maze (EPM). Involvement of other types of neurokinin receptors in this activation has not yet been evaluated. The present study examined whether local injections of the selective NK-3 agonist senktide (1-100 pmol/0.2 microL) into the dPAG can (1) cause anxiogenic effects in the EPM, (2) influence novelty-induced 22 kHz USVs, or (3) change nociceptive reactivity in the tail-flick test. Senktide elicited a significant increase in exploratory behavior, an effect accompanied by hyperalgesia and an increase in the number of 22 kHz USVs. The nociceptive effects, increased locomotor activity, and USV emissions elicited by local injections of senktide (50 pmol/0.2 microL) were reduced by prior injections of the selective NK-3 receptor antagonist SB222200 (50 pmol/0.2 microL) into the dPAG. These findings show that NK-3 receptors in the dPAG mediate nociceptive responses in this area, contrasting with the known fear-related processes mediated by NK-1 receptors in the dPAG. Both hyperalgesia and fear-related processes are accompanied by emissions of 22 kHz USVs.
Content may be subject to copyright.
ORIGINAL INVESTIGATION
Evidence for mediation of nociception by injection
of the NK-3 receptor agonist, senktide, into the dorsal
periaqueductal gray of rats
Gabriel S. Bassi &Ana C. Broiz &Margarete Z. Gomes &
Marcus L. Brandão
Received: 17 July 2008 / Accepted: 1 December 2008 / Published online: 18 December 2008
#Springer-Verlag 2008
Abstract
Rationale Ultrasound vocalizations (USVs) at approxi-
mately 22 kHz are usual components of the defensive
response of rats. However, depending on the neural
substrate that is activated, such as the dorsal periaqueductal
gray (dPAG), USV emissions may be reduced. Activation
of neurokinin-1 (NK-1)-mediated mechanisms of the dPAG
causes analgesia, reduced 22 kHz USVs, and anxiogenic-
like effects in rats exposed to the elevated plus maze
(EPM). Involvement of other types of neurokinin receptors
in this activation has not yet been evaluated.
Objectives The present study examined whether local injec-
tions of the selective NK-3 agonist senktide (1-100 pmol/
0.2 μL) into the dPAG can (1) cause anxiogenic effects in the
EPM, (2) influence novelty-induced 22 kHz USVs, or (3)
change nociceptive reactivity in the tail-flick test.
Results Senktide elicited a significant increase in exploratory
behavior, an effect accompanied by hyperalgesia and an
increase in the number of 22 kHz USVs. The nociceptive
effects, increased locomotor activity, and USV emissions
elicited by local injections of senktide (50 pmol/0.2 μL) were
reduced by prior injections of the selective NK-3 receptor
antagonist SB222200 (50 pmol/0.2 μL) into the dPAG.
Conclusions These findings show that NK-3 receptors in
the dPAG mediate nociceptive responses in this area,
contrasting with the known fear-related processes mediated
by NK-1 receptors in the dPAG. Both hyperalgesia and
fear-related processes are accompanied by emissions of 22
kHz USVs.
Keywords Elevated plus maze .Ultrasonic vocalizations .
NK-3 receptors .Dorsal periaqueductal gray .Senktide
Introduction
The dorsal periaqueductal gray (dPAG) is a central structure
of the so-called brain aversion system responsible for
several behavioral and somatic responses characteristic of
high states of fear in rats (Adams 1979; Bandler and
DePaulis 1988; Graeff et al. 1990; Brandão et al. 2005) cats
(Hunsperger 1956), monkeys (Jurgens and Pratt 1979), and
mice (Miczek et al. 1985). This reaction is similar to those
observed in animals confronted by predators or dangerous
environmental cues and has been associated with panic
attacks (Graeff 2004; Vianna et al. 2001; Brandão et al.
2005). Stimulation of the dPAG also produces antinocicep-
tion considered part of the unconditioned defense reaction
(Fanselow and Helmstetter 1988; Brandão et al. 1990;
Castilho et al. 2002).
The association of pain and fear generated in the dPAG
has led many laboratories to explore the involvement of
tachykinin substance P-mediated mechanisms in these
processes. The three main tachykinins, substance P, neuro-
kinin A, and neurokinin B, constitute a family of neuro-
peptides interacting with three distinct neurokinin (NK)
receptors. Substance P binds preferentially to the NK-1
receptor, NKA binds to the NK-2 receptor, and neurokinin
B binds to the NK-3 receptor (Pennefather et al. 2004;
Regoli et al. 1994). In mammals, substance P is the most
abundant tachykinin in the central nervous system, where it
Psychopharmacology (2009) 204:1324
DOI 10.1007/s00213-008-1434-y
G. S. Bassi :A. C. Broiz :M. Z. Gomes :M. L. Brandão
Instituto de Neurociências & Comportamento-INeC,
Campus USP,
14040-901 Ribeirão Preto, SP, Brasil
G. S. Bassi :A. C. Broiz :M. Z. Gomes :M. L. Brandão (*)
Laboratório de Psicobiologia, FFCLRP,
Campus USP, Avenida Bandeirantes 3900,
14049-901 Ribeirão Preto, SP, Brasil
e-mail: mbrandao@usp.br
is widely distributed in brain regions involved in the
regulation of affective behavior and the mediation of stress
responses, such as the amygdala, septum, hippocampus,
hypothalamus, and periaqueductal gray (PAG; Barbaresi
1998; Commons and Valentino 2002; Hietala et al. 2005;
Maeno et al. 1993; Nagano et al. 2006; Rigby et al. 2005).
Previous animal studies have shown that exposure to a
variety of aversive and stressful situations alter substance
P transmission in various brain regions (Bannon et al. 1986;
Brodin et al. 1994; Ebner et al. 2004;Krameretal.1998;
Rosen et al. 1992; Siegel et al. 1987). Substantial concen-
trations of substance P have been found in the dPAG
(Ljungdahl et al. 1978;Lietal.1990; Barbaresi 1998). NK-1
and NK-2 receptors are mainly involved in the mediation of
depressant- and anxiety-like behaviors in different animal
species (Ebner and Singewald 2006;Dablehetal.2005;
Kramer et al. 1998; Varty et al. 2002,Duarteetal.2004).
Local injections of substance P agonists into the dPAG
elicit a variety of anxiety-like behaviors in animals,
including conditioned place aversion and less time spent in
the open arms of the elevated plus maze (Aguiar and
Brandão 1996; De Araújo et al. 1998,2001a). Injections
of a C-terminal fragment of substance P also induce
behavioral activation with defensive characteristics (De
Araújo et al. 1999,2001b). The anxiogenic-like effects of
substance P in the dPAG appear to be mediated by NK-1
receptors (Mongeau et al. 1998; De Araújo et al. 2001b;
Duarte et al. 2004; Bassi et al. 2007b). The observation that
NK-1 receptor antagonists may be effective in the treatment
of depression in patients with symptoms of appreciable
anxiety has led to research efforts aimed at the development
of therapies for both depression and anxiety (Kramer et al.
1998; Santarelli et al. 2001; Ranga and Krishnan 2002;
Quartara and Altamura 2006).
An important component of the defensive behavior
repertoire of rats and a reliable measure of anxiety-like
behavior in myomorph rodents is the phenomenon of
ultrasonic vocalizations (USVs), more specifically 22 kHz
vocalizations (van der Poel et al. 1989; Blanchard et al. 1991;
Commissaris et al. 2000; Brudzynski and Chiu 1995; Wohr
et al. 2005; Tomazini et al. 2006). Rats emit vocalizations of
approximately 22 kHz only under experimental conditions
of potential or distal threat, not immediate or proximal threat,
suggesting that these vocalizations are associated with
anxiety rather than fear (Jelen et al. 2003). Such vocaliza-
tions may therefore be relevant to intraspecies signaling or
warning in situations of potential danger (Brudzynski and
Holland 2005).
Depending on the neural substrate of fear/anxiety that is
activated, such as the dPAG and inferior colliculus, USVs
may decrease (Nobre et al. 2003; Bassi et al. 2007a).
Interestingly, activation of NK-1-mediated mechanisms of
the dPAG causes analgesia, a reduction in 22 kHz USVs,
and anxiogenic-like effects in rats exposed to the elevated
plus maze (Bassi et al. 2007b). However, the involvement
of NK-3 receptors in the defensive repertoire induced by
activation of the dPAG has not yet been thoroughly
analyzed. To examine this issue in greater detail, we studied
whether injection of the NK-3 agonist senktide (10
100 pmol/0.2 μL) into the dPAG induces anxiogenic effects
in rats submitted to the elevated plus maze, changes their
nociceptive reactivity, or influences novelty-induced
22 kHz USVs recorded within the 22-kHz frequency range.
Senktide was chosen because of its preferential affinity
for NK-3 receptors (Regoli et al. 1994; Jenkinson et al.
2000; Yip and Chahl 1999,2001; Sculptoreanu and
de Groat 2007).
Materials and methods
General methodology
Animals
One hundred thirty-one male Wistar rats from the Univer-
sity of São Paulo vivarium were used. The animals were
transported to a local vivarium in the laboratory when they
were 45 days old and weighed an average of 230 g. The
room was maintained under constant temperature (23 ± 1°C)
with a 12:12 h light/dark cycle (lights on between 0700 and
1900 hours). They had free access to food and water
throughout the experiment, and care was taken to ensure
minimal handling and stress (Nunes Mamede Rosa et al.
2005). The rats were housed in groups of five per cage
(40×30×25 cm). The experiments were conducted between
900 and 1400 hours and complied with the recommen-
dations of the Brazilian Society for Neuroscience and
Behavior, which are based on the US National Institutes of
Health Guide for the Care and Use of Laboratory Animals.
Surgery
The animals were anesthetized with tribromoethanol
(250 mg/kg, i.p.) and placed in a stereotaxic frame (David
Kopf, Tujunga, CA, USA). The upper incisor bar was set
3.3 mm below the interaural line such that the skull was
horizontal between bregma and lambda. A unilateral
stainless steel guide cannula (12 mm, 24 gauge) aimed at
the dPAG was implanted in each animal. The cannula was
introduced at an angle of 10° with lambda serving as the
reference for each plane: anterior/posterior 0.3 mm,
medial/lateral 1.2 mm, dorsal/ventral 2.2 mm (Paxinos
and Watson 1997). For all groups, the cannulae were fixed
to the skull by means of acrylic resin and two stainless steel
screws. At the end of surgery, each guide cannula was
14 Psychopharmacology (2009) 204:1324
sealed with a stainless steel wire to protect it from
blockage. Afterward, the animals were transported to the
vivarium and housed in pairs. Seven days after surgery, the
animals were submitted to the testing sessions. Each animal
was subjected to only one of the tests described below.
Microinjection procedure
The animal was gently wrapped in a cloth and held by the
experimenter for 2 min, and a thin dental needle (0.3 mm,
outside diameter) was introduced through the guide cannula
until its lower end was 1 mm below the guide cannula. The
injection needle was connected to a 5 μL Hamilton syringe
by means of a polyethylene tube. A total volume of 0.2 μL
was used for injections into the dPAG. The solutions were
injected into the dPAG (0.2 μL/min), driven by an infusion
pump (Harvard Apparatus, South Natick, MA, USA). The
displacement of an air bubble inside the polyethylene
catheter (PE-10; Becton-Dickinson, Franklin Lakes, NJ,
USA) connecting the syringe needle to the intracerebral
needle was used to monitor the microinjection. The needle
was held in place for an additional 1 min to maximize
diffusion away from the tip of the needle.
Drugs
The NK-3 receptor agonist senktide (succinyl-[Asp
6
, Me-
Phe
8
]SP
6-11
) was obtained from Sigma (Brazil). The drug
was dissolved and diluted to the desired concentration with
phosphate-buffered saline (pH 7.4) shortly before use.
Senktide was injected at doses of 1, 10, 50, and 100 pmol/
0.2 μL (depending on the group of animals) immediately
before the tests. The doses of senktide were selected based on
pilot experiments and on a previous study (Ribeiro et al. 1999).
Histology
Upon completion of the experiments, the animals were
deeply anesthetized with urethane and perfused intra-
cardially with 0.9% saline followed by formalin solution
(10%). Three hours later, the brains were immersed in 30%
sucrose. Seven days later, the brains were frozen. Serial
60 μm brain sections were cut using a microtome to
localize the positions of the cannulae tips according to the
rat brain atlas (Paxinos and Watson 1997).
Experiment I: involvement of NK-3 receptors of the dPAG
in the modulation of anxiety-related behavior
Apparatus
The elevated plus maze was made of wood and had two
open arms (50×10 cm) perpendicular to two enclosed arms
of the same size with 50-cm-high walls, with the exception
of the central part (10×10 cm) where the arms crossed. The
apparatus was elevated 50 cm above the floor (Pellow et al.
1985; Anseloni and Brandão 1997). The behavior of the
animals was recorded with a video camera (Everfocus, Sao
Paulo, Brazil) positioned above the maze. The signal was
relayed to a monitor in another room via a closed-circuit
television camera to discriminate all forms of behavior.
Luminosity at the level of the open arms of the elevated
plus maze was 20 lx. The maze was cleaned thoroughly
after each test using damp and dry cloths.
Procedure
The effects of senktide injected into the dPAG of rats were
assessed with five groups of animals (n= 8 in all groups):
(1) saline, (2) 1 pmol senktide, (3) 10 pmol senktide,
(4) 50 pmol senktide, (5) 100 pmol senktide. Five minutes
after the dPAG injections, the animals were placed in the
maze for 5-min sessions. Experimental sessions were
conducted between 1200 and 1800 hours. Rats were placed
individually in the center of the maze facing an enclosed
arm and allowed 5 min of free exploration of the maze. An
observer trained to measure ethological elevated plus
maze parameters subsequently scored the videotapes. The
behavioral categories were scored using a software (Noldus,
Amsterdam, The Netherlands) which allowed measurement
of the number of entries in both the open and closed arms
and the time spent in different parts of the maze. An arm
entry or exit was defined as all four paws entering or exiting
an arm, respectively. These data were used to calculate the
percentage of open arm entries and percentage of time spent
in the open arms. A thorough description of the use of the
elevated plus maze test in this laboratory can be found
elsewhere (Anseloni and Brandão 1997).
Statistical analysis
The data obtained in the elevated plus maze test were
analyzed using one-way analysis of variance (ANOVA) for
each variable in the study, followed by the Student
NewmanKeuls post hoc test. Values of p<0.05 were
considered statistically significant.
Experiment II: involvement of NK-3 receptors in the dPAG
in the generation of 22 kHz ultrasonic vocalizations
Recording of ultrasonic vocalizations
The apparatus used for recording and analyzing USVs
consisted of a testing box (25×15×12 cm) made of steel
bars spaced approximately 12 mm apart. This experimental
chamber was situated inside of a larger, padded, echo-free
Psychopharmacology (2009) 204:1324 15
(sound-attenuated), ventilated box (60 × 40 × 45 cm) with a
28-W red light bulb located at the top of the chamber. For
recording and analyzing USVs, an Electret ultrasound
microphone (Emkay FG-3629; Avisoft Bioacoustics,
Berlin, Germany) that is sensitive to frequencies of 1
100 kHz with a flat frequency response was used. The
microphone was connected via an Avisoft UltraSoundGate
116 USB audio device (Avisoft Bioacoustics) to a com-
puter, where acoustic data were displayed in real time by
Avisoft Recorder (version 2.7; Avisoft Bioacoustics) and
were recorded with a sampling rate of 214,285 Hz in 16-bit
format. For acoustical analysis, recordings were transferred
to SASLab Pro (version 4.38; Avisoft Bioacoustics), and a
fast Fourier transformation was performed (512 FFT-length,
100% frame, Hamming window, 75% time window
overlap). Spectrograms were produced at a frequency
resolution of 488 Hz and a time resolution of 0.512 ms.
Call detection was provided by an automatic threshold-
based algorithm (threshold, 10 dB; start/end threshold,
20 dB) and a hold-time mechanism (hold time, 20 ms).
A lower cut-off frequency of 1 kHz was used for the
analysis of the USV parameters. Various parameters derived
from the average spectrum of the entire element were
determined automatically. The number of calls emitted at
each frequency served as the statistical unit in each
subject. Vocalizations recorded at frequencies below
20 kHz are operationally defined in this study as audible
vocalizations, although this term refers to the human
capacities.
The sessions consisted of placing the animals individu-
ally inside the experimental chamber for 15 min. During
each testing session, the microphone was placed through a
hole in the middle of the roof of the chamber, 40 cm above
the floor, to record the entire spectrum of USVs. A video
camera linked to a television was used to monitor all
behavior for the 15-min recording period. The animals were
allocated to four groups: (1) saline (n= 8), (2) 1 pmol
senktide (n= 6), (3) 10 pmol senktide (n=9), and (4)
50 pmol senktide (n=10).
The USV emissions obtained from this experiment were
stored on a hard disk and subsequently transferred to tables
in the Microsoft Excel spreadsheet program (Redmond,
WA, USA) for off-line analysis.
Statistical analysis
Data are expressed as mean ± SEM. Vocalizations were
analyzed by two-way repeated-measures ANOVA. The
group factor refers to treatments, and the condition factor
refers to the recorded frequencies. StudentNewmanKeuls
post hoc comparisons were performed whenever significant
overall Fvalues were obtained. Values of p<0.05 were
considered statistically significant.
Experiment III: influence of NK-3 receptors of the dPAG
on the tail-flick latency test
Tail-flick test
This test was based on DAmour and Smiths(1941)
method. Rats were placed in a restraining tube, from which
their tails protruded. Radiant heat was focused on the lower
third of the tail of the animal (Ugo Basile, Varese, Italy).
Movement of the tail activates a photocell, turning off both
the light and a reaction timer. The light intensity was
adjusted to achieve baseline latencies of 2.5 and 3.5 s. A
maximum latency of 6 s (i.e., the cutoff) was established to
minimize tail damage. The baseline trial consisted in
determining the average of three individual tests separated
by a 5-min interval before injection of drug or saline into
the dPAG. After the determination of baseline tail-flick
latency, saline or senktide at 1, 10, and 50 pmol (n=8 in all
groups, with the exception of the saline group that had ten
animals) was microinjected into the dPAG according to
each group assignment. Six other tail-flick latencies were
recorded across the experiments at 5-min intervals. This
procedure has been used successfully in this laboratory
(De Luca-Vinhas et al. 2006; Bassi et al. 2007b).
Statistical analysis
Data are expressed as mean ± SEM. Tail-flick latencies
were analyzed by two-way repeated-measures ANOVA.
The group factor refers to treatments, and the condition
factor refers to the time before and after tail-flick latency
recordings. StudentNewmanKeuls post hoc comparisons
were performed whenever significant overall Fvalues were
obtained. Values of p<0.05 were considered statistically
significant.
Results
Histological examination of the midbrain slices indicated
that all cannulae tips were located within the dorsal portion
of the PAG. The upper panels of Fig. 1show outlines of the
injection sites, and a representative site of microinjection
into the dPAG is shown in the lower panel.
Experiment I
The first experiment was designed to evaluate the influence
of senktide on anxiety-like behaviors of rats tested in the
elevated plus maze. Senktide injection led to increased
exploration of the open and closed arms in this animal
model of anxiety. Figure 2shows a significant increase in
the frequency of entries into both arms (F
4,35
=7.23 and
16 Psychopharmacology (2009) 204:1324
3.00 for the open and closed arms, respectively; p< 0.05 in
both cases). For both open- and closed-arm entries, post
hoc analyses showed that these effects were caused by
doses of 50 and 100 pmol/0.2 μL. As a consequence, the
statistical analysis of the proportion (percentage) of time
spent and entries into the open arms compared with total
time of the test and entries into both open and closed arms
of the maze did not reveal any significant effect.
Experiment II
Figure 3illustrates the effects of senktide injections on
audible(Fig. 3a) and ultrasonic (Fig. 3b) vocalizations.
Whereas all saline-injected rats emitted some vocalizations at
the frequencies of 412 kHz during the test sessions, only
12% of the animals injected with saline emitted USVs. Two-
way ANOVA revealed a significant main effect of treatment
on USVs (F
3,116
=8.19, p<0.05) and frequencies (F
4,116
=
7.18, p<0.05). A significant interaction was observed
between treatment and frequencies of USVs (F
12,116
=2.66,
p<0.05). Significant changes were observed in the number
of audible vocalizations caused by treatments (F
3,116
=10.61,
p<0.05) and in the frequency of audiblevocalizations
(F
4,116
=6.71, p<0.05). A significant interaction was observed
between treatment and frequency in the number of calls
(F
12,116
=2.77, p<0.05). Post hoc comparisons revealed that
senktide injection into the dPAG caused an increase in the
number of USVs recorded at 2024 kHz and a reduction in
this parameter of audiblevocalizations at the range of 8
10 kHz compared with saline-injected control animals.
Audiblevocalizations (0.04 s/call) were shorter than
ultrasonic vocalizations (0.4 s/call).
Experiment III
Figure 4shows the tail-flick latencies (mean ± SEM) mea-
sured before drug administration (10, 5, and 0 min) and
over six 5-min intervals after injection into the dPAG. No
differences were detected between groups during the three
baseline tail-flick latencies. Repeated-measures ANOVA
revealed significant effects of treatment (F
3,240
=9.09; p<
0.05) and time (F
8,240
=8.08; p<0.05) and a significant
interaction between treatment and time (F
24,240
=2.90; p<
0.05). Post hoc pairwise comparisons indicated that these
differences were attributable to the groups senktide 10 and
50 pmol 520 min after dPAG injections compared with the
saline group. Thermal hyperalgesia peaked 20 min after
injection of drug and returned to normal within 30 min.
Fig. 1 a Outlines of all injec-
tion sites in the dPAG (gray
areas) on cross-sections from
the atlas of Paxinos and Watson
(1997). The numbers below the
brain diagrams represent the
atlas frontal coordinates in
millimeters posterior to bregma.
bRepresentative photomicro-
graph of an injection site in the
dorsal periaqueductal gray
matter (dPAG). Scale bar
represents 400 μm. DR dorsal
raphe nucleus, dPAG dorsal
periaqueductal gray, vPAG
ventrolateral periaqueductal
gray, SC superior colliculus
Psychopharmacology (2009) 204:1324 17
Experiment IV
After the completion of Experiments IIII, we examined
whether the observed effects of senktide were selectively
mediated by NK-3 receptors. In this additional study, the
effects of senktide were challenged with intra-dPAG
injections of the selective NK-3 receptor antagonist
SB222200 (Massi et al. 2000, Sarau et al. 2000). Using
the same procedures described above, rats were tested in
the elevated plus maze and USVs and tail-flick tests. The
elevated plus maze was first used to preliminarily evaluate
the dose of SB222200 to be used. Intra-dPAG injections of
SB222200 diluted in dimethyl sulfoxide (DMSO 7.5%
vehicle) at a dose of 50 pmol did not change the
exploratory behavior of animals in the elevated plus maze;
however, a dose of 100 pmol caused motor deficits assessed
by a reduction in closed arm entries. For this study, rats
were also randomly allocated to one of four treatment
groups (n=6 for each group): (1) vehicle + saline, (2)
vehicle + senktide 50 pmol; (3) SB222200 50 pmol +
senktide 50 pmol, (4) SB222200 50 pmol + saline. A 5-min
interval separated the two injections. Immediately after the
second injection, the animals were submitted to one of the
three tests separated by an interval of 24 h.
Similar to the previous experiments, only rats with
cannulae tips located within the dorsal portion of the
PAG were used in experiment IV. The data obtained in
this experiment are shown in Fig. 5. The treatments
increased exploration of the open and closed arms of the
elevated plus maze. Figure 5a shows that these animals
exhibited a significant increase in the frequency of entries
into both open and closed arms (F
3,23
=4.73 and 8.91
for open and closed arms, respectively; p<0.05inboth
cases). For both open- and closed-arm entries, post hoc
analyses revealed that these effects were attributable to
the intra-dPAG senktide injections. These effects were
inhibited by SB222200 treatment. The effects of injections
of this antagonist alone did not produce any significant
effects.
Figure 5b illustrates the effects of combined treatments
on the number of USVs. Two-way ANOVA revealed a
main effect of treatment on USVs (F
3,80
=20.78; p<0.05)
and frequencies (F
4,80
=16.11; p<0.05). A significant
interaction was observed between treatment and frequencies
for number of USVs (F
12,80
=4.98; p<0.05). Post hoc
comparisons revealed that senktide injection into the dPAG
caused an increase in the number of USVs recorded at
22 kHz, which was significantly reduced in animals
challenged with prior injections of SB222200. The effects
of intra-dPAG injections of this antagonist alone did not
produce any significant effects.
Figure 5c shows the tail-flick latencies measured
before drug administration and over six 5-min intervals
after combined injections into the dPAG. Repeated-
measures ANOVA revealed significant effects of treat-
ment (F
3,168
=3.73; p<0.05) and time (F
8,168
=4.46;
p<0.05) and a significant interaction between treatment
and time (F
24,168
=2.30; p<0.05). Post hoc pairwise com-
parisons indicated that these differences were attributable
to the senktide group 510 min after dPAG injections
compared with controls. This hyperalgesia was signifi-
cantly reduced in the group of animals that received prior
injections of SB222200. The effects of intra-dPAG
injections of this antagonist alone did not produce any
significant effects.
Fig. 2 Mean (±SEM) number of entries into the open and closed arms
of the elevated plus maze and percentage of entries in the open arms
compared with total entries in both arms in controls and in rats under
the effects of senktide injections into the dPAG. *p<0.05, compared
with the control group (Saline). n=8 animals for each group
18 Psychopharmacology (2009) 204:1324
Discussion
Substance P has been implicated in the mediation of fear in
certain regions of the brain aversion system, such as the
amygdala, medial hypothalamus, and dPAG (Shaikh et al.
1993; De Araújo et al. 1999,2001a). Local injections of
substance P agonists into the dPAG elicit a variety of
anxiety-like behaviors in animals, including conditioned
place aversion and less time spent in the open arms of the
elevated plus maze, a widely used animal model of anxiety
Fig. 3 Effects of senktide
injections into the dorsal
periaqueductal gray on the
number of vocalizations emitted
at frequencies of 1826 kHz
during the 15-min test of
novelty exposure. Data are
expressed as mean±SEM.
*p<0.05, compared with 4 kHz
emissions in the control group
(Saline). Pound sign p<0.05,
compared with the same
frequency in the control group.
Two-way ANOVA followed by
NewmanKeuls post hoc test.
Saline (n=8), senktide 1 pmol
(n=6), senktide 10 pmol (n= 9),
senktide 50 pmol (n=10)
Fig. 4 Time-course of tail-flick
latencies during each 5-min
period across the baseline period
(10, 5, and 0 min) and up to
30 min after injection of saline
or senktide into the dPAG of rats
submitted to the tail-flick test.
Data are expressed as mean±
SEM. *p<0.05 different from
the control group (Saline). Two-
way repeated-measures ANOVA
followed by StudentNewman
Keuls post hoc test. n=8 in each
group, with the exception of
n=10 for the 50 pmol group.
The arrow indicates the time of
injection into the dPAG
Psychopharmacology (2009) 204:1324 19
(Aguiar and Brandão 1994; De Araújo et al. 1998,2001b).
NK-1 receptors appear to be involved in the aversive effects
of substance P in the dPAG. However, the role of NK-2 and
NK-3 receptors in the dPAG in the modulation of fear is
still unclear. Despite the differences in the localization of
NK-3 receptors in rodents, a significant density of NK-3
receptors is present in the PAG of rats (Bergstrom et al.
1987; Shughrue et al. 1996, Langlois et al. 2001). The
present study, therefore, sought further evidence of the
involvement of neurokinin mechanisms of the dPAG in the
defense reaction by exploring the effects of injection of the
NK-3 receptor-selective agonist senktide into the dPAG
(Regoli et al. 1994; Jenkinson et al. 2000; Yip and Chahl
1999,2001; Sculptoreanu and de Groat 2007). Hyper-
algesia and 22 kHz USV emissions were found to be the
main effects produced by injection of senktide at doses of
10 and 50 pmol/0.2 μL into the dPAG, without any
detectable anxiogenic-like effects assessed by the elevated
Fig. 5 a Effects of injections of
senktide (50 pmol/0.2 μL) and
SB222200 (50 pmol/0.2 μL)
into the dPAG on the number of
entries into the open and closed
arms of the elevated plus maze.
Data are expressed as mean±
SEM. bEffects of injections of
senktide (50 pmol/0.2 μL) and
SB220200 (50 pmol/0.2 μL)
into the dPAG on the number of
vocalizations emitted by rats at
frequencies of 1826 kHz
during the 15-min test of
novelty exposure. Data are
expressed as mean±SEM.
cTime-course of tail-flick
latencies during each 5-min
period across the baseline period
(10, 5, and 0 min) and up to
30 min after combined treat-
ments in rats submitted to the
tail-flick test. Data are expressed
as mean± SEM. *p< 0.05, com-
pared with the control group
(VSvehiclesaline).
#
p<0.05,
compared with the Vehicle-
senktide (V-Senktide) group.
ANOVA followed by the
NewmanKeuls post hoc test.
n=6 for all groups. Injections
were separated by a 5-min
interval. Rats were placed in the
test apparatus soon after the
second injection
20 Psychopharmacology (2009) 204:1324
plus maze. These animals did not show any other signs of
distress, such as urination, defecation, struggle, or abnormal
breathing patterns, but senktide produced enhanced
exploratory behavior when injected into the dPAG of rats
tested in the elevated plus maze. These animals entered the
open and closed arms of the maze more often than controls,
which is characteristic of heightened motor activity in this
test. These effects cannot be attributed to anxiolytic-like
effects because the proportion of time spent in the open
arms was not statistically different than control animals.
The present findings contrast with recent data obtained
in a similar study from this laboratory, in which injections
of the NK-1 receptor agonist SAR-Met-SP into the dPAG
caused anxiogenic-like effects in the elevated plus maze,
analgesia in the tail-flick test, and reduced USV emissions
(Bassi et al. 2007b). NK-1-mediated mechanisms have been
associated with anxiety-like behavior in several animal
species (Ebner and Singewald 2006; Dableh et al. 2005;
Kramer et al. 1998; Varty et al. 2002; Duarte et al. 2004).
The anxiogenic-like effects of substance P in the dPAG
may be mediated by NK-1 receptors (Mongeau et al. 1998;
De Araújo et al. 2001b; Duarte et al. 2004; Bassi et al.
2007a,b).
USVs in the present study lasted longer than audible
calls in a pattern similar to those of previous studies,
suggesting that these types of calls reflect distinct affective
states (Wohr et al. 2005). Rats emit USVs at frequencies of
2024 kHz when exposed to moderately stressful condi-
tions, such as when they are exposed to potentially
dangerous situations (Blanchard et al. 1991; Vivian et al.
1994; Brudzynski and Chiu 1995; Commissaris et al. 2000;
Nobre and Brandão 2004). However, a nonmonotonic
function may ensue when rats with past stressful experience
are exposed to threatening conditions of an intense nature
(Nunes Mamede Rosa et al. 2005; Tomazini et al. 2006).
Thus, an intense condition of fear or proximal danger, such
as when the animals are confronted by predators, produces
opposite effects (i.e., a reduction in USVs at 2224 kHz)
that are not sensitive to the anxiolytic effects of midazolam
(Tomazini et al. 2006). Consistent with this, rats have been
claimed to only emit 22 kHz vocalizations under experi-
mental conditions associated with anxiety and not fear
(Jelen et al. 2003). In fact, uncontrollable stressors, such as
imminent attack by predators or electrical stimulation of the
dorsal midbrain, led to freezing and a reduction in USVs at
22 kHz (Nobre and Brandão 2004; Nunes Mamede Rosa et
al. 2005). In this respect, previous studies in this laboratory
have shown that injection of substance P or NK-1 agonists
into the dPAG leads to a reduction in the number and
duration of 22 kHz USVs (De Araújo et al. 1998,1999,
2001a,b). Moreover, activation of NK-1 mechanisms in the
dPAG also produces anxiogenic-like effects assessed by
the elevated plus maze and antinociception evaluated by the
tail-flick test (Bassi et al. 2007b). Antinociception has been
considered to be an important sensory component of fear
(Fanselow and Helmstetter 1988). Such a secondary
response to the stimulation of the dPAG may activate a
descending inhibitory mechanism of pain associated with
the ventrolateral region of the PAG. Indeed, substance P
administration into this region had antinociceptive effects in
several animal models, including the tail-flick test (Xin
et al. 1997; Rosen et al. 2004). Notably, these effects appear
to be mediated selectively by NK-1 receptors because they
were blocked by selective NK-1 receptor antagonists.
Ultrasonic vocalizations in the 22 kHz range have been
used to address the emotional-affective responses to painful
stimuli under various experimental conditions, including
arthritic pain (Ardid et al. 1993; Vivian and Miczek 1998;
Jourdan et al. 1995,1998; Calvino et al. 1996; Dinh et al.
1999; Han et al. 2005). The hyperalgesia caused by
injections of 10 or 50 pmol/0.2 μL of senktide into the
dPAG was accompanied by a concomitant increase in the
emission of 22 kHz USVs. Interestingly, these effects were
also accompanied by a decrease in audibleUSVs in the
810 kHz frequency range. These latter effects may be
reflexive emotional reactivity to the nociceptive effects of
senktide (i.e., aversion to these injections has the effect of
producing higher USVs frequencies). Considering that
when rats are in pain they emit frequencies denoting
suffering (22 kHz), they should reduce the vocalizations
they normally emit (810 kHz) in conditions without
distress. In fact, NK-3 receptors are known to be involved
in the processing of information leading to central sensiti-
zation and nociception. This biological function could be
subserved by excitation of a subpopulation of neurons of
the PAG that result from NK-3 receptor activation in this
region, consistent with a recent study that administered
senktide into the PAG (Drew et al. 2005). Another study
showed that intracerebroventricular injections of senktide
induced strong neuronal activation in the PAG measured by
the expression of the protooncogene c-fos (Smith and Flynn
2000). Also supporting the present findings, activation of
NK-3 receptors has been found to increase motor activity
and facilitate the electrically evoked nociceptive flexor
reflex in adult rats (Linden and Seybold 1999; Gaudreau
and Plourde 2003; De Souza Silva et al. 2006). Pretreat-
ment with N(G)-nitro-L-arginine methyl ester (30 nmol), a
nitric oxide synthase inhibitor, blocked the hyperalgesic
effect of senktide, suggesting that senktide-induced thermal
hyperalgesia is also mediated by the production of nitric
oxide (Linden et al. 1999). Unfortunately, these studies did
not clearly distinguish between ventral and dorsal regions
of the PAG, so direct comparisons with the present results
found after intra-dPAG administration cannot be made,
especially when the drug effects on NK receptors depend
on a particular subregion of the PAG (Chahl 2006). Indeed,
Psychopharmacology (2009) 204:1324 21
some studies have shown an antinociceptive effect of NK-3
receptors after intrathecal injections of NK-3 receptor
agonists (Papir-Kircheli et al. 1987; Laneuville et al. 1988;
Couture et al. 1993; Chahl 2006). The results of experiment
IV confirm that the nociceptive effects, increased locomotor
activity, and USV emissions elicited by local injections of
senktide into the dPAG were attributable to activation of
NK-3 receptors in this region. These effects were all
reduced by prior injections of the selective NK-3 receptor
antagonist SB222200 into the dPAG. It is also important to
mention that the fact that the use of young adult rats in this
study might have had an impact on the reported results
especially because it is known that the spontaneous
behavior of rats at this age is highly influenced by their
emotional reactivity (Adriani and Laviola 2004; Tomazini
et al. 2006). Nonetheless, more studies are needed to firmly
establish the role of NK-3 receptors and its endogenous
agonist neurokinin B.
In summary, the present results show that activation of
NK-3-mediated mechanisms of the dPAG produces a
pattern of effects distinct from those mediated by NK-1
mechanisms found in the dPAG. The vocal and behavioral
profiles are consistent with activation of acute pain
mechanisms that lead to increased psychomotor activation
accompanied by 22 kHz USVs that may reflect the
emotional-affective state of the animal. These data associate
NK-3 mechanisms of the dPAG with the processing of
sensory stimulation in the dPAG. These effects contrast
with those attributed to the NK-1 receptor subtype in the
dPAG in the control of pathological anxiety states.
Acknowledgments This work wassupported by FAPESP (06/06354-5
and 06/03930-5) and CNPq (06/472030-0).
References
Adams DB (1979) Brain mechanisms for offense, defense, and
submission. Behav Brain Sci 2:200241
Adriani W, Laviola G (2004) Windows of vulnerability to psycho-
pathology and therapeutic strategy in the adolescent rodent
model. Behav Pharmacol 15:34152
Aguiar MS, Brandão ML (1994) Conditioned place aversion produced
by microinjections of substance P into the periaqueductal gray of
rats. Behav Pharmacol 5:369373
Aguiar MS, Brandão ML (1996) Effects of microinjections of the
neuropeptide substance P in the dorsal periaqueductal gray on the
behaviour of rats in the plus-maze test. Physiol Behav 60:11831186
Anseloni VZ, Brandão ML (1997) Ethopharmacological analysis of
behaviour of rats using variations of the elevated plus-maze.
Behav Pharmacol 8:533540
Ardid D, Jourdan D, Eschalier A, Arabia C, Le Bars D (1993)
Vocalization elicited by activation of A delta- and C-fibres in the
rat. NeuroReport 5:105108
Bandler R, Depaulis A (1988) Elicitation of intraspecific defence
reactions in the rat from midbrain periaqueductal grey by
microinjection of kainic acid, without neurotoxic effects. Neurosci
Lett 88:291296
Bannon MJ, Deutch AY, Tam SY, Zamir N, Eskay RL, Lee JM,
Maggio JE, Roth RH (1986) Mild footshock stress dissociates
substance P from substance K and dynorphin from Met- and
Leu-enkephalin. Brain Res 381:393396
Barbaresi P (1998) Immunocytochemical localization of substance
P receptor in rat periaqueductal gray matter: a light and electron
microscopic study. J Comp Neurol 398:473490
Bassi GS, Nobre MJ, Carvalho MC, Brandão ML (2007a) Substance
P injected into the dorsal periaqueductal gray causes anxiogenic
effects similar to the long-term isolation as assessed by ultra-
sound vocalizations measurements. Behav Brain Res 182:301
307
Bassi GS, Nobre MJ, de Araújo JE, Brandão ML (2007b) Anxiogenic
effects of activation of NK-1 receptors of the dorsal periaqueductal
gray as assessed by the elevated plus-maze, ultrasound vocaliza-
tions and tail-flick tests. Neuropeptides 41:365374
Bergström L, Torrens Y, Saffroy M, Beaujouan JC, Lavielle S,
Chassaing G, Morgat JL, Glowinski J, Marquet A (1987) [
3
H]
neurokinin B and
125
I-Bolton Hunter eledoisin label identical
tachykinin binding sites in the rat brain. J Neurochem 48:125
133
Blanchard RJ, Blanchard DC, Agullana R, Weiss SM (1991) Twenty-
two kHz alarm cries to presentation of a predator, by laboratory
rats living in visible burrow systems. Physiol Behav 50:967972
Brandão ML, Coimbra NC, Borges PC (1990) Effects of morphine
and midazolam on reactivity to peripheral noxious and central
aversive stimuli. Neurosci Biobehav Rev 14:495499
Brandão ML, Borelli KG, Nobre MJ, Santos JM, Albrechet-Souza L,
Oliveira AR, Martinez RC (2005) Gabaergic regulation of the
neural organization of fear in the midbrain tectum. Neurosci
Biobehav Rev 29:12991311
Brodin E, Rosen A, Schott E, Brodin K (1994) Effects of sequential
removal of rats from a group cage, and of individual housing of
rats, on substance P, cholecystokinin and somatostatin levels in
the periaqueductal grey and limbic regions. Neuropeptides
26:253260
Brudzynski SM, Chiu EM (1995) Behavioural responses of laboratory
rats to playback of 22 kHz ultrasonic calls. Physiol Behav
57:10391044
Brudzynski SM, Holland G (2005) Acoustic characteristics of air puff-
induced 22-kHz alarm calls in direct recordings. Neurosci
Biobehav Rev 29:11691180
Calvino B, Besson JM, Boehrer A, Depaulis A (1996) Ultrasonic
vocalization (2228 kHz) in a model of chronic pain, the arthritic
rat: effects of analgesic drugs. NeuroReport 7:581584
Castilho VM, Macedo CE, Brandão ML (2002) Role of benzo-
diazepine and serotonergic mechanisms in conditioned freezing
and antinociception using electrical stimulation of the dorsal
periaqueductal gray as unconditioned stimulus in rats. Psycho-
pharmacology 165:7785
Chahl LA (2006) Tachykinins and neuropsychiatric disorders. Curr
Drug Targets 7:9931003
Commissaris RL, Palmer A, Neophytou S, Graham M, Beckett S,
Marsden CA (2000) Acoustically elicited behaviours in Lister
hooded and Wistar rats. Physiol Behav 68:521531
Commons KG, Valentino RJ (2002) Cellular basis for the effects of
substance P in the periaqueductal gray and dorsal raphe nucleus.
J Comp Neurol 447:8297
Couture R, Boucher S, Picard P, Regoli D (1993) Receptor charac-
terization of the spinal action of neurokinins on nociception: a
three receptor hypothesis. Regul Pept 46:426429
DAmour FE, Smith DL (1941) A method for determining loss of pain
sensation. J Pharmacol Exp Ther 72:9
22 Psychopharmacology (2009) 204:1324
Dableh LJ, Yashpal K, Rochford J, Henry JL (2005) Antidepressant-
like effects of neurokinin receptor antagonists in the forced swim
test in the rat. Eur J Pharmacol 507:99105
De Araújo JE, Huston JP, Brandão ML (1998) Aversive effects of the
C-fragment of substance P in the dorsal periaqueductal gray
matter. Exp Brain Res 123:8489
De Araújo JE, Silva RC, Huston JP, Brandão ML (1999) Anxiogenic
effects of substance P and its 7-11 C-terminal, but not the 1-7 N-
terminal, injected into the dorsal periaqueductal gray. Peptides
20:14371443
De Araújo JE, Huston JP, Brandão ML (2001a) Opposite effects of
substance P fragments C (anxiogenic) and N (anxiolytic) injected
into dorsal periaqueductal gray. Eur J Pharmacol 432:4351
De Araújo JE, Huston JP, Brandão ML (2001b) Place aversion
induced by microinjections of C-fragment of substance P into the
dorsal periaqueductal gray of rats is mediated by tachykinin NK1
receptors. Peptides 22:14471452
De Luca-Vinhas MC, Macedo CE, Brandão ML (2006) Pharmaco-
logical assessment of the freezing, antinociception, and exploratory
behavior organized in the ventrolateral periaqueductal gray. Pain
121:94104
De Souza Silva MA, Mello EL Jr, Müller CP, Jocham G, Maior RS,
Huston JP, Tomaz C, Barros M (2006) The tachykinin NK3
receptor antagonist SR142801 blocks the behavioral effects of
cocaine in marmoset monkeys. Eur J Pharmacol 536:269278
Dinh HK, Larkin A, Gatlin L, Piepmeier E Jr (1999) Rat ultrasound
model for measuring pain resulting from intramuscularly injected
antimicrobials. J Pharm Sci Technol 53:4043
Drew GM, Mitchell VA, Vaughan CW (2005) Postsynaptic actions of
substance P on rat periaqueductal grey neurons in vitro.
Neuropharmacology 49:587595
Duarte FS, Testolin R, De Lima TC (2004) Further evidence on the
anxiogenic-like effect of substance P evaluated in the elevated
plus-maze in rats. Behav Brain Res 154:501510
Ebner K, Rupniak NM, Saria A, Singewald N (2004) Substance P in
the medial amygdala: emotional stress-sensitive release and
modulation of anxiety-related behavior in rats. Proc Natl Acad
Sci U S A 101:42804285
Ebner K, Singewald N (2006) The role of substance P in stress and
anxiety responses. Amino Acids 31:251272
Fanselow MS, Helmstetter FJ (1988) Conditional analgesia, defensive
freezing, and benzodiazepines. Behav Neurosci 102:233243
Gaudreau GA, Plourde V (2003) Role of tachykinin NK1, NK2 and
NK3 receptors in the modulation of visceral hypersensitivity in
the rat. Neurosci Lett 351:5962
Graeff FG (2004) Serotonin, the periaqueductal gray and panic.
Neurosci Biobehav Rev 28:239259
Graeff FG, Audi EA, Almeida SS, Graeff EO, Hunziker MH (1990)
Behavioral effects of 5-HT receptor ligands in the aversive brain
stimulation, elevated plus-maze and learned helplessness tests.
Neurosci Biobehav Rev 14:501506
Han JS, Bird GC, Li W, Jones J, Neugebauer V (2005) Computerized
analysis of audible and ultrasonic vocalizations of rats as a
astandardized measure of pain-related behavior. J Neurosci
Methods 141:261269
Hietal J, Nyman MJ, Eskola O, Laakso A, Gronroos T, Oikonen V,
Bergman J, Haaparanta M, Forsback S, Marjamaki P, Lehikoinen
P, Goldberg M, Burns D, Hamill T, Eng WS, Coimbra A,
Hargreaves R, Solin O (2005) Visualization and quantification of
neurokinin-1 (NK1) receptors in the human brain. Mol Imaging
Biol 7:262272
Hunsperger RW (1956) Affective reaction from electric stimulation of
brain stem in cats. Helv Physiol Pharmacol Acta 14:7092
Jelen P, Soltysik S, Zagrodzka J (2003) 22-kHz ultrasonic vocalization
in rats as an index of anxiety but not fear: behavioral and
pharmacological modulation of affective state. Behav Brain Res
141:6372
Jenkinson KM, Mann PT, Southwell BR, Furness JB (2000)
Independent endocytosis of the NK
1
and NK
3
tachykinin
receptors in neurons of the rat myenteric plexus. Neuroscience
100:191199
Jourdan D, Ardid D, Chapuy E, Eschalier A, Le Bars D (1995)
Audible and ultrasonic vocalization elicited by single electrical
nociceptive stimuli to the tail in the rat. Pain 63:237249
Jourdan D, Ardid D, Chapuy E, Le Bars D, Eschalier A (1998) Effect
of analgesics on audible and ultrasonic pain-induced vocalization
in the rat. Life Sci 63:17611768
Jürgens U, Pratt R (1979) Role of the periaqueductal grey in vocal
expression of emotion. Brain Res 167:367378
Kramer MS, Cutler N, Feighner J, Shrivastava R, Carman J, Sramek
JJ, Reines SA, Liu G, Snavely D, Wyatt-Knowles E, Hale JJ,
Mills SG, MacCoss M, Swain CJ, Harrison T, Hill RG, Hefti F,
Scolnick EM, Cascieri MA, Chicchi GG, Sadowski S, Williams
AR, Hewson L, Smith D, Carlson EJ, Hargreaves RJ, Rupniak
NM (1998) Distinct mechanism for antidepressant activity by
blockade of central substance P receptors. Science 281:1640
1645
Laneuville O, Dorais J, Couture R (1988) Characterization of the
effects produced by neurokinins and three agonists selective for
neurokinin receptor subtypes in a spinal nociceptive reflex of the
rat. Life Sci 42:12951305
Langlois X, Wintmolders C, te Riele P, Leysen JE, Jurzak M (2001)
Detailed distribution of neurokinin 3 receptors in the rat, guinea
pig and gerbil brain: a comparative autoradiographic study.
Neuropharmacology 40:242253
Li YQ, Jia HG, Rao ZR, Shi JW (1990) Serotonin-, substance P- or
leucine-enkephalin-containing neurons in the midbrain periaque-
ductal gray and nucleus raphe dorsalis send projection fibers to
the central amygdaloid nucleus in the rat. Neurosci Lett 120:124
127
Linden DR, Seybold VS (1999) Spinal neurokinin3 receptors mediate
thermal but not mechanical hyperalgesia via nitric oxide. Pain
80:309317
Linden DR, Jia YP, Seybold VS (1999) Spinal neurokin3 receptors
facilitate the nociceptive flexor reflex via a pathway involving
nitric oxide. Pain 80:301308
Ljungdahl A, Hökfelt T, Nilsson G (1978) Distribution of substance
P-like immunoreactivity in the central nervous system of the rat:
I. Cell bodies and nerve terminals. Neuroscience 3:861943
Maeno H, Kiyama H, Tohyama M (1993) Distribution of the
substance P receptor (NK-1 receptor) in the central nervous
system. Brain Res Mol Brain Res 18:4358
Massi M, Panocka I, De Caro G (2000) The psychopharmacology of
tachykin NK-3 receptors in laboratory animals. Peptides 21:
15971609
Miczek KA, Thompson ML, Shuster L (1985) Naloxone injections
into the periaqueductal grey area and arcuate nucleus block
analgesia in defeated mice. Psychopharmacology 87:3942
Mongeau R, De Oca BM, Fanselow MS, Marsden CA (1998)
Differential effects of neurokinin-1 receptor activation in sub-
regions of the periaqueductal gray matter on conditional and
unconditional fear behaviors in rats. Behav Neurosci 112:1125
1135
Nagano M, Saitow F, Haneda E, Konishi S, Hayashi M, Suzuki H (2006)
Distribution and pharmacological characterization of primate NK-1
and NK-3 tachykinin receptors in the central nervous system of the
rhesus monkey. Br J Pharmacol 147:316323
Nobre MJ, Brandão ML (2004) Analysis of freezing behavior and
ultrasonic vocalization in response to foot-shocks, ultrasound
signals and GABAergic inhibition in the inferior colliculus:
Psychopharmacology (2009) 204:1324 23
effects of muscimol and midazolam. Eur Neuropsychopharmacol
14:4552
Nobre MJ, Sandner G, Brandão ML (2003) Enhancement of acoustic
evoked potentials and impairment of startle reflex induced by
reduction of GABAergic control of the neural substrates of
aversion in the inferior colliculus. Hear Res 84:8290
Nunes Mamede Rosa ML, Nobre MJ, Ribeiro Oliveira A, Brandão
ML (2005) Isolation-induced changes in ultrasonic vocalization,
fear-potentiated startle and prepulse inhibition in rats. Neuro-
psychobiology 51:248255
Papir-Kricheli D, Frey J, Laufer R, Gilon C, Chorev M, Selinger Z,
Devor M (1987) Behavioural effects of receptor-specific sub-
stance P agonists. Pain 31:263276
Paxinos G, Watson C (1997) The rat brain in stereotaxic coordinates.
Academic, Sydney
Pellow S, Chopin P, File SE, Briley M (1985) Validation of open:
closed arm entries in the elevated plus-maze as a measure of
anxiety in the rat. J Neurosci Methods 14:149167
Pennefather JN, Lecci A, Candenas ML, Patak E, Pinto FM, Maggi
CA (2004) Tachykinins and tachykinin receptors: a growing
family. Life Sci 74:14451463
Quartara L, Altamura M (2006) Tachykinin receptors antagonists:
from research to clinic. Curr Drug Targets 7:975992
Ranga K, Krishnan R (2002) Clinical experience with substance P
receptor (NK1) antagonists in depression. J Clin Psychiatry 63
(Suppl 11):2529
Regoli D, Boudon A, Fauchere JL (1994) Receptors and antagonists
for substance P and related peptides. Pharmacol Rev 46:551599
Ribeiro SJ, Teixeira RM, Calixto JB, De Lima TC (1999) Tachykinin
NK
3
receptor involvement in anxiety. Neuropeptides 33:181188
Rigby M, O'Donnell R, Rupniak NM (2005) Species differences in
tachykinin receptor distribution: further evidence that the
substance P (NK1) receptor predominates in human brain. J
Comp Neurol 490:335353
Rosen A, Brodin K, Eneroth P, Brodin E (1992) Short-term restraint
stress and s.c. saline injection alter the tissue levels of substance
P and cholecystokinin in the peri-aqueductal grey and limbic
regions of rat brain. Acta Physiol Scand 146:341348
Rosen A, Zhang YX, Lund I, Lundeberg T, Yu LC (2004) Substance P
microinjected into the periaqueductal gray matter induces
antinociception and is released following morphine administra-
tion. Brain Res 1001:8794
Santarelli L, Gobbi G, Debs PC, Sibille ET, Blier P, Hen R, Heath MJ
(2001) Genetic and pharmacological disruption of neurokinin 1
receptor function decreases anxiety-related behaviors and increases
serotonergic function. Proc Natl Acad Sci U S A 98:19121917
Sarau HM, Griswold DE, Bush B, Potts W, Sandhu P, Lundberg D,
Foley JJ, Schmidt DB, Webb EF, Martin LD, Legos JJ, Whitmore
RG, Barone FC, Medhurst AD, Luttmann MA, Giardina GA,
Hay DW (2000) Non peptide tachykinin receptor antagonists: II.
Pharmacological and pharmacokinetic profile of SB-222200, a
central nervous system penetrant, potent and selective NK-3
receptor antagonist. J Pharmacol Exp Ther 295:373381
Sculptoreanu A, de Groat WC (2007) Neurokinins enhance excitability
in capsaicin-responsive DRG neurons. Exp Neurol 205:92100
Shaikh MB, Steinberg A, Siegel A (1993) Evidence that substance P
is utilized in medial amygdaloid facilitation of defensive rage
behavior in the cat. Brain Res 625:283294
Shughrue PJ, Lane MV, Merchenthaler I (1996) In situ hybridization
analysis of the distribution of neurokinin-3 mRNA in the rat
central nervous system. J Comp Neurol 372:395414
Siegel RA, Duker EM, Pahnke U, Wuttke W (1987) Stress-induced
changes in cholecystokinin and substance P concentrations in
discrete regions of the rat hypothalamus. Neuroendocrinology
46:7581
Smith ME, Flynn FW (2000) Distribution of Fos-like immunoreac-
tivity within the rat brain following intraventricular injection of
the selective NK
3
receptor agonist senktide. J Comp Neurol
426:413428
Tomazini FM, Reimer A, Albrechet-Souza L, Brandão ML (2006)
Opposite effects of short- and long-duration isolation on
ultrasonic vocalization, startle and prepulse inhibition in rats. J
Neurosci Methods 153:114120
Varty GB, Cohen-Williams ME, Morgan CA, Pylak U, Duffy RA,
Lachowicz JE, Carey GJ, Coffin VL (2002) The gerbil ele-
vated plus-maze II: anxiolytic-like effects of selective neuro-
kinin NK1 receptor antagonists. Neuropsychopharmacology
27:371379
Vianna DM, Landeira-Fernandez J, Brandão ML (2001) Dorsolateral
and ventral regions of the periaqueductal gray matter are
involved in distinct types of fear. Neurosci Biobehav Rev 25:
711719
van der Poel AM, Noach EJ, Miczek KA (1989) Temporal patterning
of ultrasonic distress calls in the adult rat: effects of morphine
and benzodiazepines. Psychopharmacology 97:147148
Vivian JA, Miczek KA (1998) Effects of μand δopioid agonists and
antagonists on affective vocal and reflexive pain responses
during social stress in rats. Psychopharmacology 139:364375
Vivian JA, Farrell WJ, Sapperstein SB, Miczek KA (1994) Diazepam
withdrawal: effects of diazepam and gepirone on acoustic startle-
induced 22 kHz ultrasonic vocalizations. Psychopharmacology
114:101108
Wohr M, Borta A, Schwarting RK (2005) Overt behavior and
ultrasonic vocalization in a fear conditioning paradigm: a dose
response study in the rat. Neurobiol Learn Mem 84:228240
Xin L, Geller EB, Liu-Chew LY, Chew C, Adler MW (1997)
Substance P release in the rat periaqueductal gray and preoptic
anterior hypothalamus after noxious cold stimulation: effect of
selective mu and kappa opioid agonists. J Pharmacol Exp Ther
282:10551063
Yip J, Chahl LA (1999) Distribution of Fos-like immunoreactivity in
guinea-pig brain following administration of the neurokinin-1
receptor agonist, [SAR
9
,MET(O
2
)
11
]substance P. Neuroscience
94:663673
Yip J, Chahl LA (2001) Localization of NK1 and NK3 receptors in
guinea-pig brain. Regul Pept 98:5562
24 Psychopharmacology (2009) 204:1324
... The present study compared the behavioral effects of administering the higher effective dose of SP (100 pmol/ 0.2 μl) in the MeA, CeA, and BLA and challenged these effects by administering the NK-1 antagonist spantide II. Spantide was used because it has been used in several studies in our laboratory over the past decade (Bassi et al., 2007b(Bassi et al., , 2009Brenes et al., 2012;Carvalho et al., 2013Carvalho et al., , 2015. Strong immunoreactivity of NK-1 receptors is found in the MeA in rats, with sparse immunoreactivity in the BLA and low levels in the CeA (Smith et al., 1999;Ribeiro-da-Silva and Hökfelt, 2000), justifying the use of spantide II as a suitable tool for assessing NK-1 activity because it possesses much stronger antagonist activity for NK-1 receptors than for NK-2 and NK-3 receptors (Janecka et al., 2005;Maggi et al., 1991). ...
... Five minutes after the last microinjection, six additional tail-flick latencies were recorded at 5-min intervals. This procedure has been used successfully in our laboratory (Bassi et al., 2007a(Bassi et al., , 2009). ...
... First, bilateral inactivation of the amygdala decreases shock-conditioned vocalizations (Borszcz and Leaton, 2003;Lee et al., 2001), and USV emissions via stimulation of the amygdala can only be produced after the conditioning of stimuli (e.g., light and shock). Second, unconditioned USVs can only be produced in brain structures that are located in the lower levels of the neuroaxis because stimulation of the dPAG and anterior hypothalamus produces USVs (Bassi et al., 2007b(Bassi et al., , 2009Brudzynski and Barnabi, 1996). In our opinion, the amygdala serves as a feedback structure that regulates acoustic components of the modulation of the actual emotion, such as intensity and frequency. ...
Article
Rationale: The production of unconditioned defensive behaviors has been related to the amygdala, a key component of the encephalic aversion system. Microinjection of the neuropeptide substance P (SP) in the amygdala elicits defensive behaviors via the activation of type 1 neurokinin (NK-1) receptors. However, no studies have investigated whether intra-amygdala SP/NK-1 mechanisms can elicit other types of defensive responses, such as antinociception and ultrasonic vocalizations (USVs). Methods: The present study investigated the effects of SP-induced activation of the neurokininergic system in three main nuclei of the amygdala-basolateral (BLA), central (CeA), and medial (MeA) nuclei-in rats that were subjected to the elevated plus maze (EPM), tail-flick test, and USV recording. The effects of SP in these amygdaloid nuclei were challenged with combined injections of the NK-1 receptor antagonist spantide. Results: The present study showed that SP injections in the CeA and MeA but not BLA exerted anxiogenic-like effects. In contrast to the CeA, the anxiogenic-like effects of SP in the MeA were not dependent on NK-1 mechanisms. In the tail-flick test, SP microinjections produced antinociceptive effects only in the MeA through NK-1 receptor activation. No USV emissions were detected after the SP microinjections. Conclusions: The present study showed that NK-1 receptors in the CeA and MeA but not BLA are involved in defensive reactions to conditions of fear. The present results may provide a better understanding of the neurochemical mediation of fear states.
... Spantide was injected at the dose of 100 pmol/0.2 μL, which was chosen on the basis of previous studies (7,22). ...
... Ten minutes later, aversive thresholds and the time spent freezing after dPAG or IC stimulation were determined again. The drug dose and waiting time after the injections were selected from previous studies in this laboratory (7,22). ...
... NK-1 receptors appear to be involved in the aversive effects of substance P in the dPAG, but the role of NK-2 and NK-3 receptors in this region in the modulation of fear is still unclear. Previous studies from this laboratory showed that NK-1-mediated mechanisms, but not NK-3, are involved in the mediation of defensive behavior elicited by stimulation of the dPAG (22). The anti-aversive effects of spantide in the present study support the hypothesis that NK-1 receptors in the dPAG play a pro-aversive role and corroborate and extend the proposed involvement of the NK system in the modulation of defensive behavior in this region. ...
Article
Full-text available
Electrical stimulation of midbrain tectum structures, particularly the dorsal periaqueductal gray (dPAG) and inferior colliculus (IC), produces defensive responses, such as freezing and escape behavior. Freezing also ensues after termination of dPAG stimulation (post-stimulation freezing). These defensive reaction responses are critically mediated by γ-aminobutyric acid and 5-hydroxytryptamine mechanisms in the midbrain tectum. Neurokinins (NKs) also play a role in the mediation of dPAG stimulation-evoked fear, but how NK receptors are involved in the global processing and expression of fear at the level of the midbrain tectum is yet unclear. The present study investigated the role of NK-1 receptors in unconditioned defensive behavior induced by electrical stimulation of the dPAG and IC of male Wistar rats. Spantide (100 pmol/0.2 μL), a selective NK-1 antagonist, injected into these midbrain structures had anti-aversive effects on defensive responses and distress ultrasonic vocalizations induced by stimulation of the dPAG but not of the IC. Moreover, intra-dPAG injections of spantide did not influence post-stimulation freezing or alter exploratory behavior in rats subjected to the elevated plus maze. These results suggest that NK-1 receptors are mainly involved in the mediation of defensive behavior organized in the dPAG. Dorsal periaqueductal gray-evoked post-stimulation freezing was not affected by intra-dPAG injections of spantide, suggesting that NK-1-mediated mechanisms are only involved in the output mechanisms of defensive behavior and not involved in the processing of ascending aversive information from the dPAG.
... The present results give support to the notion that NK-1-and NK-3-mediated mechanisms have distinct roles in the dPAG. Contrasting with the known fear-related processes mediated by NK-1 receptors, NK-3 receptors in the dPAG mediate nociceptive responses in this area (Bassi et al., 2009). In this latter study, senktide-a selective NK-3 agonist-elicited a significant increase in exploratory behavior in the elevated plus-maze test, an effect accompanied by hyperalgesia and an increase in the number of 22 kHz ultrasonic vocalizations (USVs). ...
... Also, in a previous study from this laboratory, SB 222200 did not show any effect in the plus-maze test at doses of 50 and 100 mol, with the latter dose causing a reduction in the closed arm entries (Bassi et al., 2009, p 18). In this study, similar injections of the NK-3 agonist senktide into the dPAG increased the locomotor activity without causing any significant effect on the emotional variables of the elevated plus-maze test (Bassi et al., 2009). In line with this report, the present data show that SB 222200 selectively acted on the motor activity by decreasing the number of crossings and rearing. ...
... * PϽ0.05, different from the corresponding groups tested in the different context (two-way ANOVA followed by Bonferroni test). than with fear processing or expression of defensive behaviors, as already shown (Bassi et al., 2009). However, when applied systemically, the NK-3 agonist, senktide, was also shown to be pro-amnestic in rats and mice and to increase ACh levels in the frontal cortex, amygdala, and hippocampus of aged rats (Schäble et al., 2011;Zlomuzica et al., 2008). ...
Article
The dorsal periaqueductal gray (dPAG) is the main output structure for the defensive response to proximal aversive stimulation. Panic-like responses, such as freezing and escape behaviors, often result when this structure is electrically stimulated. Freezing also ensues after termination of the dPAG stimulation (post-stimulation freezing (PSF)). GABA and 5-HT have been proposed as the main neuromediators of these defense reactions. Neurokinins (NKs) also play a role in the defense reaction; however, it is unclear how the distinct types of NK receptors are involved in the expression of these fear responses. This study investigated the role of NK-1 and NK-3 receptors in the unconditioned defensive behaviors induced by electrical stimulation of the dPAG of rats, with and without previous experience with contextual fear conditioning (CFC). Spantide (100 ρmol/0.2 μl) and SB 222200 (50 and 100 ρmol/0.2 μl), selective antagonists of NK-1 and NK-3 receptors, respectively, were injected into the dPAG. Injection of spantide had antiaversive effects as determined by stimulation of the dPAG in naive animals and in animals subjected previously to CFC. SB 222200 also increased these aversive thresholds but only at doses that caused a motor deficit. Moreover, neither spantide nor SB 222200 influenced the PSF. The results suggest that NK-1 receptors are mainly involved in the mediation of the defensive behaviors organized in the dPAG. Because dPAG-evoked PSF was not affected by intra-dPAG injections of either spantide or SB 222200, it is suggested that neurokinin-mediated mechanisms are not involved in the processing of ascending aversive information from the dPAG.
... Recently, Zelikowsky et al. found that chronic isolation stress causes a brain-wide upregulation of Tac2 expression, and this change is necessary and sufficient for the behavioural influence of chronic isolation stress (Zelikowsky et al. 2018). In the rat dPAG, injection of an NK3R (neurokinin receptor which preferentially binds NKB) agonist causes an increase in exploratory behaviour in an elevated-plus maze, hyperalgesia and an increase in 22kHz USV (Bassi et al. 2009). ...
Thesis
The midbrain periaqueductal grey (PAG) is commonly recognised as the exit relay for the coordination and execution of a wide range of instinctive behaviours, such as defense, reproduction and predation. In line with its functional diversity, are the range of inputs it receives from higher cortical and subcortical areas as well as ascending spinal pathways, and the various neurotransmitter and neuromodulatory mechanisms active in its different subregions. However, the lack of a comprehensive cell-type classification of the PAG hinders systematic investigations into the intricacies of its many behavioural roles. Here, we applied high-throughput single neuronal nucleus RNA-sequencing to profile transcriptomes of adult mouse PAG neurons. Our data revealed at least 9 distinct PAG neuronal subpopulations, marked by differential expressions of neurotransmitter, neuromodulator and ion channel genes. In addition, using a combination of optogenetic manipulations and a carefully designed defense test battery, we identified separate functions of dPAG vGlut2+, PACAP+ and Tac2+ neurons in triggering and modulating defensive behaviour. We showed that dlPAG vGlut2+ neurons project to the Cuneiform nucleus, and this projection is an output pathway for PAG elicited escape behaviour. Our work supports the existence of molecularly distinct, functionally divergent pathways in the PAG underlying defensive behaviour, and demonstrates a framework towards a systematic dissection of cell-type specific functions of complex brain regions.
... NK3 receptors are widely distributed in the brain (often together with NK1 receptors), e.g. in the prefrontal cortex, hippocampus, amygdala and in the hypothalamus, where they are involved in the regulation of the release of gonadotropinreleasing hormones [54]. NK3 also functions in the pain transmission and locomotor activity [55,56]. ...
Article
Over the last few decades, depression has become one of the major public health problems in our society. This problem is connected not only with morbidity, but also with treatment, specifically with the effectiveness of the therapy as well as the concomitant side effects of available antidepressants. Major depressive disorder is a complex clinical entity, including different molecular mechanisms and neurological processes. This complexity is a challenge for scientists seeking to discover an innovatory antidepressant drug with multiple and complementary mechanisms of action. In this review, we discuss the role of melatonin, neurokinin, neurotrophic tyrosine kinase and glucocorticoid receptors in depression and antidepressant-like effects.
... Nevertheless, since C-terminal fragments may have a greater affinity for NK 2 and NK 3 receptors than SP1-11 [13], which preferentially binds to NK 1 receptors [4], a possible involvement of the NK 3 subtype in the biological effects induced by SP6-11(C-terminal) cannot be excluded, as previously observed by our group. NK 3 receptors have been associated with locomotor activity, pain processing and promnestic effects [14], but the exact role of NK 3 receptors in anxiety has not been determined yet. The distribution of these receptors shows dense populations in the brain cortex, amygdala, medial septal nucleus, hypothalamus, and locus coeruleus [15,16], cerebral areas implicated in the underlying mechanisms of fear and anxiety (for reviews, see Refs. ...
Article
Introduction: Substance P (SP) is a neuropeptide widely expressed throughout the fear-processing pathways of the brain. SP is cleaved by several proteolytic enzymes in amino (N-) and carboxy (C-) terminal sequences, which can have biological activities per se. We have previously shown that the anxiogenic-like effects elicited by SP6-11(C-terminal), a specific metabolite of SP, are mediated via NK1 and NK2 receptors. Nevertheless, there are evidences that C-terminal fragments may have a greater affinity for NK3 receptors. Objectives: The aim of the present study was to further investigate the possible involvement of NK3 receptors in the anxiogenic-like effects induced by SP6-11(C-terminal). Methods: Adult male Wistar rats were intracerebroventricularly (i.c.v.) treated with SR142801 (NK3 receptors antagonist) or vehicle one minute to prior SP6-11(C-terminal) or vehicle. Other experimental groups received SP6-11(C-terminal) or vehicle i.c.v. one minute prior to senktide (NK3 receptors agonist) or vehicle. After five minutes, the animals were behaviorally evaluated in the elevated plus-maze test (EPM). Results: SR142801 (100 pmol) or SP6-11(C-terminal) (10 pmol) reduced all the parameters of open-arms exploration and increased the number of protected stretch-attend postures in the EPM, indicating an anxiogenic-like effect. Senktide (10 pmol) promoted an opposite effect on these behavioral parameters, characterizing an anxiolytic-like profile. Pretreatment with SR142801, in an ineffective dose, potentiated the SP6-11-induced anxiety, especially in the unprotected head-dipping and protected stretch-attend postures behaviors. Moreover, the anxiolytic-like effect induced by senktide (1 pmol) was prevented by SP6-11. Conclusions: Our results give support to the involvement of NK3 receptors in the anxiogenic-like actions of SP6-11(C-terminal), where this metabolite seems to behave as an antagonist, in a way similar to SR142801.
Article
Full-text available
The coexistence of chronic pain and anxiety is a common clinical phenomenon. Here, the role of tachykinin receptor 3 (NK3R) in the lateral habenula (LHb) in trigeminal neuralgia and in pain-associated anxiety was systematically investigated. First, electrophysiological recording showed that bilateral LHb neurons are hyperactive in a mouse model of trigeminal neuralgia made by partial transection of the infraorbital nerve (pT-ION). Chemicogenetic activation of bilateral LHb glutamatergic neurons in naive mice induced orofacial allodynia and anxiety-like behaviors, and pharmacological activation of NK3R in the LHb attenuated allodynia and anxiety-like behaviors induced by pT-ION. Electrophysiological recording showed that pharmacological activation of NK3R suppressed the abnormal excitation of LHb neurons. In parallel, pharmacological inhibition of NK3R induced orofacial allodynia and anxiety-like behavior in naive mice. The electrophysiological recording showed that pharmacological inhibition of NK3R activates LHb neurons. Neurokinin B (NKB) is an endogenous high-affinity ligand of NK3R, which binds NK3R and activates it to perform physiological functions, and further neuron projection tracing showed that the front section of the periaqueductal gray (fPAG) projects NKB-positive nerve fibers to the LHb. Optogenetics combined with electrophysiology recordings characterize the functional connections in this fPAG NKB → LHb pathway. In addition, electrophysiological recording showed that NKB-positive neurons in the fPAG were more active than NKB-negative neurons in pT-ION mice. Finally, inhibition of NKB release from the fPAG reversed the analgesic and anxiolytic effects of LHb Tacr3 overexpression in pT-ION mice, indicating that fPAG NKB → LHb regulates orofacial allodynia and pain-induced anxious behaviors. These findings for NK3R suggest the cellular mechanism behind pT-ION in the LHb and suggest that the fPAG NKB → LHb circuit is involved in pain and anxiety comorbidity. This previously unrecognized pathway might provide a potential approach for relieving the pain and anxiety associated with trigeminal neuralgia by targeting NK3R.
Article
Full-text available
Studies have previously demonstrated a relationship between social status and anxiety disorders such as panic disorder. Repeated episodes of panic attacks do not occur in combination with an actual fear stimulus or stressor. However, social ranking modulates the perception of the social signals of a threat or stressor. The hypothalamic nuclei are well‑known for their role in the elaboration of fear‑induced reactions. The dorsomedial hypothalamus (DMH) and the ventromedial hypothalamic (VMH) nuclei are hypothalamic subnuclei involved in the processing of threatening stimuli‑evoked aversive response and innate fear development. These structures are also located in the medial amygdala‑hypothalamus‑brainstem circuit that modulates innate fear‑induced defensive behaviors. This work aimed to investigate the relationship between social hierarchy and innate fear‑induced panic‑like responses in male rats. In our study, the dominance tube test was used to determine the social hierarchy. Then, DMH/VMH nuclei were unilaterally implanted with a guide cannula. After intra‑DMH/VMH injection of bicuculline (GABAA receptor antagonist), both innate fear induction and differences in dominant/subordinate rats were evaluated by the open field test. Intra‑DMH/VMH bicuculline increased the frequency of defensive immobility, forward escape movements, and crossing behaviors, as well as the duration of defensive immobility and forward escape movements in dominant rats. Subordinate rats showed a higher frequency of defensive attention, defensive immobility, and crossing than dominant rats. Additionally, dominant rats demonstrated a lower duration of defensive attention and defensive immobility than subordinate rats. Dominant rats seemed to adopt a form of innate‑fear characterized by increased proactivity with the environment. In contrast, subordinate rats exhibited a reactive form of innate‑fear characterized by passivity and freezing.
Article
The Tacr3 gene encodes tachykinin receptor 3 (NK3R), which belongs to the tachykinin receptor family. This family of proteins includes typical G protein-coupled receptors and belongs to the rhodopsin subfamily, and NK3R functions by binding to its high-affinity ligand, neurokinin B(NKB). The role of Tacr3/NK3R in growth and reproduction has been extensively studied, but Tacr3/NK3R is also widely expressed in the nervous system from the spinal cord to the brain and is involved in both physiological and pathological processes in the nervous system, including mood disorders, chronic pain, learning and memory deficiencies, Alzheimer's disease, Parkinson's disease, addiction-related processes, hypoxic-ischemic encephalopathy, body fluid management, neural development, and schizophrenia. Here, we summarize the structure of NK3R/NKB and its cellular signaling as well as the expression of Tacr3/NK3R in the nervous system, and we provide a comprehensive summary of the role of Tacr3/NK3R in neurological diseases, including reproduction-related disorders and other neurological diseases. At the end of this review, we propose the hypothesis that Tacr3/NK3R mediates a variety of brain functions by affecting the excitability of different neurons with specific functions. Based on this "excited or not" hypothesis, more studies related to Tacr3 should be carried out in other nervous system diseases in order to better understand the biological roles of Tacr3.
Article
Neurokinin B (NKB) is one member of an evolutionarily conserved family of neuropeptides, the tachykinins. Preferential binding of NKB to endogenous NK3 receptors affects a variety of biological and physiological processes, including endocrine secretions, sensory transmission, and fluid and electrolyte homeostasis. In light of its widespread biological actions, immunohistochemical detection of the c-Fos protein product was used to study the distribution of neuronal activation in the rat brain caused by intraventricular (icv) injections of the selective NK3 receptor agonist (succinyl-[Asp(6), N-Me-Phe(8)] substance P [6-11]), senktide. Quantitative analysis revealed that treatment with isotonic saline or 200 ng senktide resulted in the differential expression of Fos-like immunoreactivity (FLI) throughout the brain. Senktide induced the highest number of FLI neurons in the lateral septum, bed nucleus of the stria terminalis, amygdala, paraventricular nucleus of the hypothalamus, median preoptic nucleus, organum vasculosum of the lamina terminalis, supraoptic nucleus, periaqueductal gray, and medial nucleus of the solitary tract compared to isotonic saline controls. Additional regions that contained elevated FLI following icy injection of senktide, relative to saline injection, included the cerebral cortex, lateral hypothalamic nucleus, suprachiasmatic nucleus, ventral tegmental area, substantia nigra, inferior colliculus, locus coeruleus, zona incerta, and arcuate nucleus. Our data indicate that activation of NK3 receptors induces the expression of FLI within circumscribed regions of the rat brain. This pattern of neuronal activation overlaps with nuclei known to regulate homeostatic processes, such as endocrine secretion, cardiovascular function, salt intake, and nociception.(C) 2000 Wiley-Liss, Inc.
Article
The dorsal periaqueductal gray (DPAG) is one of the main structures involved in the integration of defensive behavior in the brain. In order to investigate the participation of neuropeptides in the generation of aversive slates, semicarbazide, a glutamic acid decarboxylase inhibitor, and substance P, an active neuropeptide, were injected into the DPAG and their effects evaluated in the open field and the place conditioning tests. While semicarbazide and substance P both increased locomotor activity only substance P increased grooming in the open field. In the place conditioning procedure similar aversion conditioning was produced by both drugs. These results confirm previous data showing that semicarbazide in the DPAG causes place aversion through reduction of tonic inhibitory mechanisms on neural substrates of aversion. Such mechanisms may include substance P neurons as substance P microinjection into the DPAG also functioned as an unconditioned stimulus in the place aversion test. (C) Lippincott-Raven Publishers.