ArticlePDF Available

Corticotropin-Releasing Factor Receptors Couple to Multiple G-Proteins to Activate Diverse Intracellular Signaling Pathways in Mouse Hippocampus: Role in Neuronal Excitability and Associative Learning

Authors:

Abstract and Figures

Corticotropin-releasing factor (CRF) exerts a key neuroregulatory control on stress responses in various regions of the mammalian brain, including the hippocampus. Using hippocampal slices, extracts, and whole animals, we investigated the effects of human/rat CRF (h/rCRF) on hippocampal neuronal excitability and hippocampus-dependent learning in two mouse inbred strains, BALB/c and C57BL/6N. Intracellular recordings from slices revealed that application of h/rCRF increased the neuronal activity in both mouse inbred strains. Inhibition of protein kinase C (PKC) by bisindolylmaleimide I (BIS-I) prevented the h/rCRF effect only in hippocampal slices from BALB/c mice but not in slices from C57BL/6N mice. Inhibition of cAMP-dependent protein kinase (PKA) by H-89 abolished the h/rCRF effect in slices from C57BL/6N mice, with no effect in slices from BALB/c mice. Accordingly, h/rCRF elevated PKA activity in hippocampal slices from C57BL/6N mice but increased only PKC activity in the hippocampus of BALB/c mice. These differences in h/rCRF signal transduction were also observed in hippocampal membrane suspensions from both mouse strains. In BALB/c mice, hippocampal CRF receptors coupled to G(q/11) during stimulation by h/rCRF, whereas they coupled to G(s), G(q/11), and G(i) in C57BL/6N mice. As expected on the basis of the slice experiments, h/rCRF improved context-dependent fear conditioning of BALB/c mice in behavioral experiments, and BIS-I prevented this effect. However, although h/rCRF increased neuronal spiking in slices from C57BL/6N mice, it did not enhance conditioned fear. These results indicate that the CRF system activates different intracellular signaling pathways in mouse hippocampus and may have distinct effects on associative learning depending on the mouse strain investigated.
Content may be subject to copyright.
Corticotropin-Releasing Factor Receptors Couple to Multiple
G-Proteins to Activate Diverse Intracellular Signaling
Pathways in Mouse Hippocampus: Role in Neuronal
Excitability and Associative Learning
Thomas Blank,
1
Ingrid Nijholt,
1
Dimitris K. Grammatopoulos,
2
Harpal S. Randeva,
2
Edward W. Hillhouse,
2
and
Joachim Spiess
1
1
Department of Molecular Neuroendocrinology, Max Planck Institute for Experimental Medicine, D-37075 Goettingen, Germany, and
2
Sir Quinton Hazell
Molecular Medicine Research Centre, Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
Corticotropin-releasing factor (CRF) exerts a key neuroregulatory control on stress responses in various regions of the mammalian brain,
including the hippocampus. Using hippocampal slices, extracts, and whole animals, we investigated the effects of human/rat CRF
(h/rCRF) on hippocampal neuronal excitability and hippocampus-dependent learning in two mouse inbred strains, BALB/c and C57BL/
6N. Intracellular recordings from slices revealed that application of h/rCRF increased the neuronal activity in both mouse inbred strains.
Inhibition of protein kinase C (PKC) by bisindolylmaleimide I (BIS-I) prevented the h/rCRF effect only in hippocampal slices from
BALB/c mice but not in slices from C57BL/6N mice. Inhibition of cAMP-dependent protein kinase (PKA) by H-89 abolished the h/rCRF
effect in slices from C57BL/6N mice, withno effect in slices from BALB/c mice. Accordingly, h/rCRF elevated PKA activity in hippocampal
slices from C57BL/6N mice but increased only PKC activity in the hippocampus of BALB/c mice. These differences in h/rCRF signal
transduction were also observed in hippocampal membrane suspensions from both mouse strains. In BALB/c mice, hippocampal CRF
receptors coupled to G
q/11
during stimulation by h/rCRF, whereas they coupled to G
s
,G
q/11
, and G
i
in C57BL/6N mice. As expected on the
basis of the slice experiments, h/rCRF improved context-dependent fear conditioning of BALB/c mice in behavioral experiments, and
BIS-I prevented this effect. However, although h/rCRF increased neuronal spiking in slices from C57BL/6N mice, it did not enhance
conditioned fear. These results indicate that the CRF system activates different intracellular signaling pathways in mouse hippocampus
and may have distinct effects on associative learning depending on the mouse strain investigated.
Key words: neuronal excitability; h/rCRF; PKC; PKA; classical fear conditioning; G-protein; mouse; hippocampus
Introduction
Corticotropin-releasing factor (CRF) is a 41 amino acid neu-
ropeptide that has been implicated in both physiological and
behavioral responses to stress (Spiess et al., 1981; Vale et al.,
1981). During exposure to stress, CRF can be secreted directly
from nerve terminals located in the hippocampus. Specifically,
numerous, large CRF-immunoreactive neurons have been found
in the hippocampal CA1 and CA3 region (Swanson et al., 1983;
Merchenthaler, 1984). Previous studies have shown the modula-
tion of hippocampus-dependent learning and memory by CRF.
Human/rat CRF (h/rCRF) injected directly into the dentate gyrus
consistently enhanced memory retention in rats in a one-way
passive avoidance task (Lee et al., 1993). Injection of h/rCRF into
the dorsal hippocampus shortly before the training enhanced
context- and tone-dependent fear conditioning in BALB/c mice
through CRF receptor 1 (CRFR1) (Radulovic et al., 1999). In
addition to the effects on hippocampal learning tasks, CRF exerts
a profound action on hippocampal neuronal activity. Recent
studies have demonstrated that h/rCRF produces a long-lasting
enhancement of synaptic efficacy in the rat hippocampus in vivo
(Wang et al., 1998, 2000). h/rCRF reversibly increases the spiking
of rat hippocampal pyramidal cells (Aldenhoff et al., 1983) and
enhances the amplitude of CA1 population spikes evoked by
stimulation of the Schaffer collateral pathway (Hollrigel et al.,
1998). We showed recently that application of h/rCRF facilitates
the induction and stability of long-term potentiation (LTP) un-
der defined stimulation conditions in area CA1 of mouse hip-
pocampal slices (Blank et al., 2002).
To examine the signal transduction pathways of h/rCRF in
mouse hippocampus, we studied the G-protein and second-
messenger activation after CRF receptor stimulation in hip-
pocampi of two mouse inbred strains, C57BL/6N and BALB/c.
We chose these two inbred strains because C57BL/6 and BALB/c
mice have repeatedly been found to differ strongly in several
behavioral responses (Oliverio et al., 1973; Peeler and Nowa-
kowski, 1987; Beuzen and Belzung, 1995) and in neurodevelop-
mental and neurochemical parameters (Nowakowski, 1984). For
example, BALB/c mice exhibit stronger anxiety-like responses in
the light– dark choice test (Beuzen and Belzung, 1995), in the
Received May 31, 2002; revised Oct. 25, 2002; accepted Oct. 29, 2002.
This work was supported by the Max Planck Society. D.K.G. is a Wellcome Trust Career Development Fellow. We
thank Dr. Klaus Eckart for the peptide synthesis of [Glu
11,16
] astressin and h/rCRF.
Correspondence should be addressed to Thomas Blank, Department of Molecular Neuroendocrinology, Max
Planck Institute for Experimental Medicine, Hermann-Rein-Straße 3, D-37075 Goettingen, Germany. E-mail:
blank@em.mpg.de.
Copyright © 2003 Society for Neuroscience 0270-6474/03/230700-08$15.00/0
700 The Journal of Neuroscience, January 15, 2003 23(2):700 –707
open-field paradigm (Oliverio et al., 1973), and in a runway tra-
versal locomotor activity test (Peeler and Nowakowski, 1987).
The impact of h/rCRF on neuronal excitability of CA1 pyramidal
cells was investigated in hippocampal slices from both mouse
inbred strains. Finally, we investigated the effect of h/rCRF on
hippocampus-dependent learning in C57BL/6N and BALB/c
mice.
Materials and Methods
Animals. Experiments were performed with male BALB/c and C57BL/6N
mice (Charles River, Sultzfeld, Germany) 912 weeks old. The mice were
housed individually and maintained on a 12 hr light/dark cycle (lights on
at 7:00 A.M.) with access to food and water ad libitum. All experimental
procedures were in accordance with the European Council Directive
(86/609/EEC) and the Animal Section Law under the supervision of the
District Government of Braunschweig (Lower Saxony, Germany).
Electrophysiology. Mice were briefly anesthetized with isoflurane and
then decapitated. In 1 min, the skull was opened, and the brain was
removed and transferred to ice-cold artificial CSF (aCSF) solution of the
following composition (in m
M): 130 NaCl, 3.5 KCl, 1.25 NaH
2
PO
4
, 1.5
MgSO
4
, 2 CaCl
2
, 24 NaHCO
3
, and 10 glucose, pH 7.4 (equilibrated with
95% O
2
5% CO
2
). Hippocampi were dissected from the chilled brain
hemispheres on ice. Transverse hippocampal slices (400
m) were ob-
tained on a McIlwain tissue chopper (Mickle Laboratory Engineering,
Surrey, UK) and kept submerged (minimum of 1 hr at room temperature
before recordings) in aCSF.
Conventional intracellular recording techniques were used, with glass
microelectrodes filled with 3
M potassium acetate. Microelectrodes were
pulled from borosilicate glass capillaries (World Precision Instruments,
Sarasota, FL) on a horizontal electrode puller (Zeitz-Instrumente, Augs-
burg, Germany). The microelectrode tip resistances ranged from 60 to
100 M for recordings from mouse hippocampal neurons. Intracellular
signals were recorded with a single-electrode voltage-clamp amplifier
(SEC-05L; NPI Electronics, Tamm, Germany), which performed
current-clamp measurements at high switching frequencies in the range
of 2530 kHz. Bridge balance was monitored throughout the experiment
and adjusted as required. Traces were stored on a computer using Pulse
7.4 software (Heka, Lambrecht, Germany) for offline analysis. For intra-
cellular recordings, only neurons were included that exhibited over-
shooting action potentials, stable membrane potentials of at least 60
mV, and input resistances of 35 M. Input resistance was determined
by measuring the voltage deflection at the end of a 100 msec hyperpolar-
ization current step (0.2 nA). Depolarizing current pulses of 35 msec
duration were injected through the recording electrode to elicit single
action potentials. Spike frequency adaptation was investigated by inject-
ing each cell with a series of 600 msec depolarizing current pulses (0.21
nA; increment, 100 pA). To compare neuronal responses, the membrane
potential of each cell was manually clamped to 65 mV by discontinuous
current injection. In all electrophysiological experiments, n values repre-
sent the number of slices.
Drugs. h/rCRF (Ru¨hmann et al., 1996) and [Glu
11,16
] astressin (Eckart
et al., 2001) were synthesized in our laboratory as described. H-89 and
bisindolylmaleimide I (BIS-I) were obtained from Calbiochem (San Di-
ego, CA). Phorbol 12,13-dibutyrate (PDBu) and 4
-phorbol were both
purchased from Sigma (St. Louis, MO).
Drug treatment. [Glu
11,16
] astressin was dissolved in aCSF to a final
concentration of 280
M. h/rCRF stock solutions were prepared in 10 mM
acetic acid. For cannula injections, dilutions in aCSF to a final concen-
tration of 400 ng/
l were prepared immediately before the experiments.
The final pH of the peptide solution was 7.4. BIS-I was stored as 1 m
M
stock solution in dimethylsulfoxide (DMSO). For injection, the solution
was diluted with aCSF to a final concentration of 0.4 nmol/
l. PDBu and
4
-phorbol were both dissolved in DMSO to 5
g/
l. For injection, the
solutions were diluted with aCSF to a final concentration of 10 ng/
l.
Cannulation. Double guide cannulas (C235; Plastics One, Roanoke,
VA) were implanted using a stereotactic holder during anesthesia with
1.2% avertin (0.02 ml/g, i.p.) under aseptic conditions as described pre-
viously (Stiedl et al., 2000; Blank et al., 2002). Each double guide cannula
with inserted dummy cannula and dust cap was fixed to the skull of the
mouse with dental cement. The cannulas were placed into both lateral
brain ventricles, with anteroposterior (AP) coordinates zeroed at bregma
AP 0 mm, lateral 1 mm, and depth 3 mm or directed toward both dorsal
hippocampi, AP 1.5 mm, lateral 1 mm, and depth 2 mm (Franklin and
Paxinos, 1997). The animals were allowed to recover for 4 5 d before the
experiments started. On the day of the experiment, bilateral injections
were performed using an infusion pump (CMA/100; CMA Microdialy-
sis, Solna, Sweden) at a constant rate of 0.33
l/min (final volume, 0.25
l per side). Cannula placement was verified post hoc in all mice by
injection of methylene blue. For electrophysiological experiments, dou-
ble guide cannula placement was verified by unilateral methylene blue
injection.
Fear conditioning. The fear conditioning experiments were performed
as described previously (Stiedl et al., 2000; Blank et al., 2002) using a
computer-controlled fear conditioning system (TSE, Bad Homburg,
Germany). Fear conditioning was performed in a Plexiglas cage (36
21 20 cm) within a fear conditioning box constantly illuminated (12 V,
10 W halogen lamp, 100 500 lux). In the conditioning box, a high-
frequency loudspeaker (KT-25-DT; Conrad, Hirschau, Germany) pro-
vided constant background noise [white noise, 68 dB sound pressure
level (SPL)]. The training (conditioning) consisted of a single trial. The
mouse was exposed to the conditioning context (180 sec) followed by a
tone (30 sec, 10 kHz, 75 dB SPL, pulsed 5 Hz). After termination of the
tone, a foot shock (0.7 mA, 2 sec, constant current) was delivered through
a stainless steel grid floor. The mouse was removed from the fear condi-
tioning box 30 sec after shock termination to avoid an aversive associa-
tion with the handling procedure. Under these conditions, the context
served as background stimulus. Background contextual fear condition-
ing but not foreground contextual fear conditioning, in which the tone is
omitted during training, has been shown to involve the hippocampus
(Phillips and LeDoux, 1994). Memory tests were performed 24 hr after
fear conditioning. Contextual memory was tested in the fear condition-
ing box for 180 sec without tone or shock presentation (with background
noise). Freezing, defined as lack of movement except for respiration and
heart beat, was assessed as the behavioral parameter of the defensive
reaction of mice (Blanchard and Blanchard, 1969; Bolles and Riley, 1973;
Fanselow and Bolles, 1979) by a time-sampling procedure every 10 sec
throughout the memory test. In addition, activity-derived measures (in-
activity, mean activity, and exploratory area) were recorded by a photo-
beam system (10 Hz detection rate).
Protein kinase A and protein kinase C assays. cAMP-dependent protein
kinase (PKA) and protein kinase C (PKC) activities were assayed using
the PepTag Assay for nonradioactive detection of PKC or PKA (Promega,
Madison, WI) on the basis of the phosphorylation of fluorescent-tagged
PKC- or PKA-specific peptides. After incubation in either aCSF or 250
n
M h/rCRF for 30 min, hippocampal slices were placed in ice-cold ho-
mogenization buffer [20 m
M Tris-HCl, pH 7.4, 2 mM EDTA, 2 mM EGTA,
48 m
M mercaptoethanol, 0.32 M sucrose, and freshly added protease
inhibitor cocktail tablet (Boehringer Mannheim, Mannheim, Germa-
ny)]. The tissue was homogenized with a Teflon-plastic homogenizer
and centrifuged at 100,000 g for 30 min in a Beckman Instruments
(Fullerton, CA) XL-80 ultracentrifuge. The resulting supernatant con-
tained the PKA preparation. The pellet was rehomogenized in homoge-
nization buffer and sonicated (four times for 15 sec), incubated for 30
min with Triton X-100 (0.2%), and centrifuged at 100,000 g for 30
min. The supernatant contained the membrane-bound PKC prepara-
tion, which was used for the PKC assay. Protein concentrations were
determined with the Bradford assay (Bio-Rad, Munich, Germany). The
assay was performed as described by the manufacturer. An aliquot of the
PKA preparation was incubated for 30 min at 30°C in PepTag PKA 5
reaction buffer (in m
M: 100 Tris-HCl, pH 7.4, 50 MgCl
2
, and 5 ATP) and
0.4
g/
l of the PKA-specific peptide substrate PepTag A1 (L-R-R-A-S-
L-G; Kemptide). The same procedure was used for the PKC preparations
that were incubated in PepTag PKC reaction buffer (in m
M: 100 HEPES,
pH 7.4, 6.5 CaCl
2
, 5 DTT, 50 MgCl
2
, and 5 ATP) containing 0.4
g/
lof
the PKC-specific peptide substrate PepTag C1 (P-L-S-R-T-L-S-V-A-A-
K). The reaction was stopped by heating to 95°C for 10 min. Phosphor-
ylation of the PKA- and PKC-specific substrates was used to measure
Blank et al. CRF-Mediated Excitability in Mouse Hippocampus J. Neurosci., January 15, 2003 23(2):700 –707 701
kinase activity. Phosphorylated and unphosphorylated PepTag peptides
were separated on a 0.8% agarose gel by electrophoresis. The gel was
photographed with a transilluminator, and bands indicating substrate
phosphorylated by PKA or PKC were quantified by densitometry (Win-
Cam 2.2; Cybertech, Berlin, Germany). For the PKA and PKC assays, 4.5
and 6.5
g of protein, respectively, were applied.
Western blotting. Hippocampi of C57BL/6N or BALB/c mice were dis-
sected out and homogenized in TBS (10 m
M Tris, pH 7.6, and 150 mM
NaCl), 10% sucrose, and a protease inhibitor cocktail tablet (Boehringer
Mannheim). The homogenate was centrifuged at 20,000 g for 30 min
at 4°C. The supernatant was removed, and the membrane pellet was
resuspended in a second identical wash step and centrifuged again at
20,000 g for 30 min at 4°C. The supernatant was removed, and the
membrane pellet was resuspended in TBS, 1 m
M EDTA, and 1% sodium
cholate and incubated for 60 min with constant mixing at 4°C. By cen-
trifugation at 155,000 g for 60 min (4°C), the supernatant containing
soluble membrane proteins was obtained. Protein concentrations were
determined with a Bradford assay (Bio-Rad). Equal amounts of protein
for each group were separated on a 10% SDS gel and transferred to an
Immobilon-P membrane (Millipore, Bedford, MA) using a semidry
transfer apparatus. The blot was probed using an anti-G
q/11
subunit
antibody (1:4000; Calbiochem), an anti-G
s
subunit antibody (1:1000;
NEN, Boston, MA), or an antibody directed against G
i-1,2,3
-protein
(1:200; Calbiochem). These antibodies were detected by secondary anti-
bodies conjugated to alkaline phosphatase. CDP-Star (Tropix, Bedford,
MA) was used as a chemiluminescence substrate. During dephosphory-
lation, the substrate decomposed, producing a prolonged emission of
light that was imaged on photographic film (Fuji Super RX; Fujifilm,
Tokyo, Japan). The relative density of the bands was measured by densi-
tometry using the software WinCam 2.2 for Windows.
Preparation of hippocampal membranes. Membranes were prepared as
described previously (Grammatopoulos et al., 2001). Hippocampi of
C57BL/6N or BALB/c mice were homogenized in Dulbeccos PBS (ex-
traction buffer) containing 10 m
M MgCl
2
,2mM EGTA, 1.5 gm/l bovine
serum albumin (BSA) (w/v), 0.15 m
M bacitracin, and 1 mM phenylmeth-
ylsulfonylfluoride (PMSF), pH 7.2, at 22°C. The homogenate was centri-
fuged at 1500 g for 30 min at 4°C. The pellet was discarded, and the
supernatant was spun at 45,000 g for 60 min at 4°C. Using the homog-
enizer, the final pellet was resuspended in 10 ml of the described extrac-
tion buffer. The protein concentration of the membrane suspension was
determined using the bicinchoninic acid method (Smith et al., 1985)
with BSA as a standard.
Synthesis of
32
P-GTP-
-azidoanilide and photolabeling of G
subunits.
32
P-GTP-
-azidoanilide (
32
P-GTP-AA) was synthesized as described
previously (Schwindinger et al., 1998). Mouse hippocampal membranes
were incubated in a darkroom with or without h/rCRF (100 n
M) for 5
min at 30°C before the addition of 5
Ci of
32
P-GTP-AA in 120
lof50
m
M HEPES buffer, pH 7.4, containing 30 mM KCl, 10 mM MgCl
2
,1mM
benzamidine, 5
M GDP, and 0.1 mM EDTA. After incubation for 3 min
at 30°C, membranes were collected by centrifugation and resuspended in
100
l of the above buffer containing 2 mM glutathione, placed on ice,
and exposed to UV light (254 nm) at a distance of 5 cm for 5 min.
G-protein immunoprecipitation.
32
P-GTP-AA-labeled G-proteins
were precipitated by centrifugation and solubilized in 120
l of 2% SDS.
Then, 360
lof10mM Tris-HCl buffer, pH 7.4, containing 1% (v/v)
Triton X-100, 1% (v/v) deoxycholate, 0.5% (w/v) SDS, 150 m
M NaCl, 1
m
M DTT, 1 mM EDTA, 0.2 mM PMSF, and 10
g/ml aprotinin was added,
and insoluble material was removed by centrifugation. Solubilized mem-
branes were divided into 100
l aliquots, and each aliquot was incubated
with 10
l of undiluted G-protein antiserum at 4°C. Subsequently, 50
l
of protein A-Sepharose beads (10% w/v in the above buffer) was added,
and the incubation was continued at 4°C overnight. The beads were
collected by centrifugation, washed twice, and dried under vacuum. The
immune complexes were dissociated from protein A by reconstitution in
Laemmlis buffer (100
l) and boiling in a water bath for 5 min. Samples
were subjected to gel electrophoresis. The gels were stained with
Coomassie blue, dried, and exposed to Fuji x-ray film at 70°Cfor
25 d. The relative density of the bands was measured by optical
density scanning using the software Scion Image-
3b for Windows
(Scion, Frederick, MD).
Statistics. Statistical comparisons were made by using Students t test
and ANOVA. Data were expressed as mean SEM. Significance was
determined at the level of p 0.05.
Results
In hippocampal slices from both C57BL/6N and BALB/c mice,
stable intracellular recordings were obtained from CA1 pyrami-
dal neurons. The resting membrane potentials of pyramidal neu-
rons from C57BL/6N mice (68.4 0.9 mV; n 38) and
BALB/c mice (69.6 1.2 mV; n 33) did not differ signifi-
cantly, nor did the membrane input resistance of CA1 cells of
C57BL/6N mice (56.5 3.5 M; n 38) and BALB/c mice
(58.7 3M; n 33) differ significantly from each other.
Likewise, there were no significant differences between the spike
amplitudes, with values of 63.4 1.2 mV (n 19) found for
C57BL/6N mice and 62.2 1.3 mV (n 13) for BALB/c mice.
When mouse CA1 pyramidal cells of either strain were excited
by prolonged depolarizing current pulses, they responded with
prolonged spiking (Fig. 1A). The discharge rate was highest at the
beginning of the current pulse (1 nA) and declined to a steady rate
during the course of the depolarizing pulse (Fig. 1B). Increasing
stimulus intensities elicited enhanced neuronal spiking. In re-
sponse to strong depolarizing current pulses (1 nA, 600 msec),
C57BL/6N and BALB/c mouse pyramidal cells fired 18.7 2.5
(n 12) and 18.6 7.3 (n 7) spikes, respectively (Fig. 1C).
h/rCRF was applied to mouse hippocampal slices to investi-
gate the effects on the neuronal spiking behavior. The number of
spikes elicited by a 600 msec depolarizing current pulse was in-
creased by 88 24% (n 7; p 0.05) in C57BL/6N (data not
shown) mice and by 87 39% (n 8; p 0.05) in BALB/c mice
after addition of 250 n
M h/rCRF (Fig. 2A). After 30 min of wash-
ing in aCSF, spiking was still elevated by 85 21% (n 7; p
0.05) in C57BL/6N (data not shown) mice and by 86 35% (n
8; p 0.05) in pyramidal cells from BALB/c mice (Fig. 2A).
Within 90 min, the firing rate returned to control values and was
no longer significantly different from the firing rate before
h/rCRF application in C57BL/6N mice (2 6%; n 7) and
Figure 1. A, Representative intracellular recordings from CA1 pyramidal neurons in hip-
pocampal slices from C57BL/6N mice and BALB/c mice showing responses to 600 msec depo-
larizing current pulses. B, Number of spikes elicited in 100 msec fragments during a single
depolarizing (depol.) current pulse (600 msec, 1 nA). C, Plot of the number of spikes elicited by
a 600 msec depolarizing pulse versus stimulus (stim.) intensity.
702 J. Neurosci., January 15, 2003 23(2):700 –707 Blank et al. CRF-Mediated Excitability in Mouse Hippocampus
BALB/c mice (4 7%; n 8) (Fig. 2A). In CA1 hippocampal
neurons from both mouse strains, the h/rCRF effect was an-
tagonized by the CRF receptor antagonist [Glu
11,16
] astressin
(Fig. 2B).
In subsequent experiments, we investigated the underlying
second-messenger pathways activated by h/rCRF to increase the
neuronal excitability in mouse hippocampus. When slices were
preincubated with the selective and cell-permeable PKA inhibitor
H-89, the firing rate of hippocampal neurons from C57BL/6N
mice was not significantly enhanced by h/rCRF (7 5%; n 6;
p NS) (Fig. 3A). In contrast, after the H-89 treatment, h/rCRF
still enhanced the neuronal activity of hippocampal neurons
from BALB/c mice by 55 10% (n 5; p 0.05) (Fig. 3B).
When hippocampal slices from BALB/c mice were preincubated
with BIS-I, a highly selective cell-permeable PKC inhibitor, sub-
sequent h/rCRF application did not significantly increase the
neuronal firing rate (4 2%; n 5; p NS) (Fig. 3D). In
contrast, after BIS-I treatment, h/rCRF application still enhanced
neuronal spiking in CA1 cells from C57BL/6N mice by 52 9%
(n 6; p 0.05) (Fig. 3C). In these mice, bath application of the
potent PKC activator PDBu increased the spiking behavior of
hippocampal neurons by 79 24% (n 5; p 0.05) (Fig. 3E).
Under basal conditions, PKA activity, as measured by the
phosphorylated state of a PKA-specific target peptide, was lower
in hippocampal brain slices from C57BL/6N mice than in hip-
pocampal brain slices from BALB/c mice. After h/rCRF treat-
ment, PKA activity in hippocampal slices from C57BL/6N mice
was increased, whereas it was decreased in hippocampal slices
from BALB/c mice compared with the corresponding PKA activ-
ities in control slices (Fig. 4A). Because membrane translocation
of PKC is considered to be an indicator of PKC activation (Kraft
and Anderson, 1983), we assayed PKC activity in the membrane-
bound fraction of hippocampal slice homogenates. After h/rCRF
incubation of slices, PKC activity was apparent only in hip-
pocampal slices of BALB/c mice (Fig. 4B), with no detectable
PKC activity in hippocampal slices of C57BL/6N mice. The ob-
served differences in the activation of second-messenger path-
ways after h/rCRF application can be attributed to variations in
the abundance of G-proteins. However, using immunoblots, we
Figure 2. Effect of h/rCRF on neuronal spiking of BALB/c mouse CA1 pyramidal cells elicited
by 600msec depolarizing currentpulses. A, Traces were sampled before, during, and30 and 90
minafter h/rCRF (250n
M,10 min) application.B,Recordings were madebeforeand 20 minafter
coapplicationof h/rCRF(250n
M,10 min)and[Glu
11,16
]astressin (1
M)over aperiodof10 min.
Pulse intensity was kept constant during each experiment; holding potential, 65 mV.
Figure 3. Effectof thePKC inhibitor BIS-I and ofthe PKAinhibitor H-89 on h/rCRF-mediated
modulation ofexcitability. Representative recordingsin CA1 pyramidal cells from C57BL/6N (A,
C ) and BALB/c (B, D) mice showing the effect of 250 n
M h/rCRF applied over a period of 20 min
after preincubation with BIS-I (1.2
M, 1 hr) or H-89 (10
M, 3 hr). E, Spiking behavior of CA1
pyramidal cells from C57BL/6N before and during bath application of PDBu (100 n
M). Pulse
intensity was kept constant during each experiment.
Figure 4. PKA and PKC activity in hippocampal slices of C57BL/6N and BALB/c mice. Hip-
pocampal slices were incubated in either 250 n
M h/rCRF (30 min) or aCSF (30 min, as control).
Partially purified homogenates of these slices (n 11) from six animals were tested for the
ability tophosphorylate a PKA-specific(L-R-R-A-S-L-G; Kemptide) (A) ora PKC-specific (P-L-S-
R-T-L-S-V-A-A-K) ( B)peptidic substrate ina nonradioactive assay.Identical amountsofprotein
were used for each sample.
Blank et al. CRF-Mediated Excitability in Mouse Hippocampus J. Neurosci., January 15, 2003 23(2):700 –707 703
did not observe any significant differences in the abundance
of G
s
-, G
I
-, and G
q/11
-proteins (Fig. 5
A). In subsequent ex-
periments, we analyzed CRF receptor-mediated activation of
G-proteins in hippocampal membrane suspensions. After
h/rCRF application, the nonhydrolyzable GTP analog
32
P-
GTP-AA binds to the GTP-binding site of activated G-protein
-chains and forms a stable complex, which can be identified
with specific G
antibodies (Offermanns et al., 1991). Thus, spe
-
cific activation of individual G-proteins can be demonstrated. In
hippocampal membranes of C57BL/6N mice, h/rCRF induced
activation of G
s
,G
i
, and G
q/11
with an order of potency G
s
G
q/11
G
i
, whereas in hippocampal membranes of BALB/c mice,
only stimulation of G
q/11
was detectable after h/rCRF treatment
(Fig. 5B,C).
To further delineate the impact of the observed different
h/rCRF-mediated signaling pathways on learning and memory,
mice were subjected to contextual fear conditioning, a
hippocampus-dependent associative learning paradigm (Kim
and Fanselow, 1992; Phillips and LeDoux, 1992, 1994). When
BALB/c mice received a bilateral h/rCRF injection intracerebrov-
entricularly (n 7) (Fig. 6A) and were trained 2 hr after the
injection, contextual fear was significantly enhanced compared
with naive ( p 0.05; n 9) (Fig. 6A) and vehicle-treated ( p
0.01; n 30) animals (Fig. 6 A). This h/rCRF effect was prevented
by either [Glu
11,16
] astressin (n 7) or BIS-I (n 7). Both
compounds had no effect when applied alone (Fig. 6 A). To ex-
clude the possibility that h/rCRF was acting via a brain structure
that has projections to the hippocampus, h/rCRF and BIS-I were
administered locally into the dorsal hippocampus. Contextual
fear was also significantly elevated when h/rCRF was injected
intrahippocampally ( p 0.05; n 6) (Fig. 6A). BIS-I had no
effect when administered intrahippocampally alone (n 5) but
abolished the h/rCRF-mediated enhancement of conditioned
fear (n 6; p NS) (Fig. 6 A). In C57BL/6N mice, freezing was
not significantly changed when h/rCRF was injected 2 hr (n 15;
p NS) (Fig. 6B) before the training session. However, injection
of PDBu 2 hr before the training (n 9) significantly enhanced
contextual fear compared with the contextual fear of naive ( p
0.05; n 9) (Fig. 6B) and vehicle-treated ( p 0.05; n 27)
animals (Fig. 6B). There was no significant change of contextual
fear after injection of the inactive isomer 4
-phorbol ( p NS;
n 4) (Fig. 6B).
Discussion
In this study, we provide evidence that signal processing of
h/rCRF in mouse hippocampus was mediated through two dif-
ferent signal transduction pathways. Slice experiments revealed
that h/rCRF increased CA1 hippocampal neuronal activity via
PKC in the hippocampus of BALB/c mice and via PKA in the
hippocampus of C57BL/6N mice. Hippocampus-dependent
learning evaluated by context-dependent fear conditioning was
improved only in BALB/c mice after h/rCRF injection but not in
C57BL/6N mice. Western blots from mouse hippocampal mem-
brane proteins showed identical amounts of the relevant
G-protein subunits in both mouse strains. However, application
of h/rCRF induced activation of G
q/11
in the hippocampus of
BALB/c mice and G
s
,G
q/11
, and G
i
in the hippocampus of
C57BL/6N mice. h/rCRF increased neuronal excitability in the
hippocampus of both mouse strains but improved fear condi-
tioning only in BALB/c and not in C57BL/6N mice. Thus, it
might be concluded that the h/rCRF-induced increase in neuro-
nal activity is not sufficient to enhance fear conditioning but that
the stimulation of specific intracellular signaling cascades is also
required. In support of this hypothesis, we observed recently that
inhibition of hippocampal Ca
2
/calmodulin-dependent kinase
II (CaMKII) prevents stress-mediated facilitation of fear condi-
tioning with no effect on primed hippocampal LTP (Blank et al.,
2002). This observation implies that facilitation of neuronal ac-
tivity was necessary along with activation of CaMKII to enhance
fear conditioning.
In mouse hippocampus, CRFR1 was reported to be the pre-
dominant CRF receptor subtype (Van Pett et al., 2000). However,
we cannot conclude whether the differences in G-protein activa-
tion result from the different coupling of a single receptor sub-
Figure 5. h/rCRF-induced activation of G
s
-, G
I
-, and G
q/11
-proteins. A, Basal levels of G
s
,G
i
,
and G
q/11
in hippocampal membrane fractions from C57BL/6Nand BALB/c mice. The bar graph
summarizesWestern blot data(mean SEM)of three independentexperimentseachwithfive
animals per mouse strain. B, Autoradiograph of h/rCRF-induced photolabeling of G
subunit
subtypes from hippocampal membranes of C57BL/6N (n 30) and BALB/c (n 30) mice.
Membranes were incubated with
32
P-GTP-AA in the presence and absence of h/rCRF (100 nM),
followed by UV cross-linking and immunoprecipitation of the G
subunit subtypes using spe-
cific antibodies. Proteins were resolved by SDS-PAGE, followed by autoradiographic visualiza-
tion. C, Bar graph summarizing autoradiograph data. *p 0.05 indicates statistically signifi-
cant differences.
704 J. Neurosci., January 15, 2003 23(2):700 –707 Blank et al. CRF-Mediated Excitability in Mouse Hippocampus
type or the different coupling in combination with differences in
the distribution profile of CRF receptor subtypes in the hip-
pocampus of both mouse strains.
All of the known effects of CRF in the rat hippocampus in-
volve receptor-coupled activation of G
s
and adenylate cyclase and
an increase in cellular levels of cAMP (Chen et al., 1986; Battaglia
et al., 1987; Pihoker et al., 1992; Haug and Storm, 2000). This is in
agreement with the activation of G
s
in hippocampi of C57BL/6N
mice. However, it was reported that h/rCRF also activates the
phospholipase C (PLC)PKC-pathway in rat Leydig cells (Ulisse
et al., 1990), in cultured rat astrocytes (Takuma et al., 1994), in rat
cerebellum (Miyata et al., 1999), and in rat cerebral cortex
(Grammatopoulos et al., 2001). In addition, Malenka et al.
(1986) reported that activation of PKC markedly reduces accom-
modation of neuronal spiking in rat hippocampal pyramidal
cells. Both aspects together are in agreement with our conclusion
that, in BALB/c mice, G
q/11
-dependent PKC activation mediated
the h/rCRF-induced increase of neuronal activity. Surprisingly,
PKA activity was reduced in hippocampal slices from BALB/c
mice during application of h/rCRF. This effect might be initiated
by G
q/11
stimulation, which has been shown to be associated with
an increase of the abundance of G-protein
␤␥
subunits. These
subunits inhibit type I adenylyl cyclase and thereby decrease PKA
activity (Taussig et al., 1993; Chen et al., 1997). Activation of
G
q/11
,G
s
, and G
i
, as observed in hippocampi of C57BL/6N mice,
synergistically stimulates adenylyl cyclase type 2 (Lustig et al.,
1993), thus also increasing the cAMP formation. In the
membrane-bound fraction of hippocampal slice homogenates
prepared from C57BL/6N mice, no PKC activity was detected
after h/rCRF application. We did not detect any significant con-
tribution of PKC to the h/rCRF-induced increase in neuronal
spiking behavior of CA1 pyramidal cells in C57BL/6N mice.
However, the treatment of hippocampal slices from C57BL/6N
mice with BIS-I and the H-89 treatment of slices from BALB/c
mice showed the tendency to reduce the spiking rate compared
with controls. This observation suggests that, in both mouse
strains, neuronal activity is sensitive to changes in PKA and PKC
activity.
Our observation that only PKA was ac-
tivated in hippocampal slices of
C57BL/6N mice during application of
h/rCRF might be because receptors with
dual signaling properties often stimulate
different pathways with different effica-
cies. A
3
adenosine receptors, for example,
interact with G
i
-proteins and, to a lesser
extent, with G
q/11
-proteins in CHO cells
(Palmer et al., 1995). These receptors were
shown to inhibit adenylyl cyclase in all cell
types tested, whereas stimulation of PLC
was cell type dependent. Although acti-
vated CRF receptors coupled to G
q/11
in
hippocampal membranes of C57BL/6N
mice, h/rCRF neither activated PKC in
hippocampal slices nor enhanced the con-
ditioned fear response. This result is sur-
prising because, in experiments using
PDBu, we demonstrated that hippocam-
pal neuronal excitability and conditioned
fear of C57BL/6N mice was enhanced by
activation of PKC. In contrast, h/rCRF
also stimulated G
q/11
in hippocampal
membranes of BALB/c mice and im-
proved hippocampus-dependent learning via activation of PKC
in this mouse strain. Similar results were reported by Fordyce et
al. (1985), who found that stimulation of hippocampal PKC ac-
tivity enhances contextual learning, as determined by the fear
conditioning task in DBA mice. In the hippocampus of C57BL/6J
mice, a PKA-dependent period for contextual memory consoli-
dation develops between 1 and 3 hr after training (Bourtchou-
ladze et al., 1998). Considering the activation of the PKA system
in the hippocampus of C57BL/6N mice during h/rCRF applica-
tion, it is surprising that h/rCRF did not facilitate contextual fear
conditioning in C57BL/6N mice. In a recent study, the crucial
temporal relationship between PKA inhibition and training nec-
essary to produce impairment of the consolidation of fear mem-
ory was demonstrated (Bourtchouladze et al., 1998). A narrow
time window exists for PKA inhibition before the training. When
mice are treated with PKA inhibitor 20 30 min before contextual
conditioning, they show dramatic amnesia. However, inhibition
of PKA 3 hr before training does not affect retention 24 hr after
training. Thus, in the present study, h/rCRF might have had no
effect on long-term contextual memory in C57BL/6N mice be-
cause PKA was not activated within the decisive time window.
To summarize, we demonstrated that h/rCRF activated at
least two different signaling cascades in mouse hippocampus, the
PLCPKC pathway (via interaction with G
q/11
) and the cAMP
PKA pathway (via interaction with G
s
,G
q/11
, and G
i
). Future
experiments will have to determine whether hippocampal CRF
receptors can switch their coupling between different G-protein
subunits triggered by the activation of specific signaling events
such as protein phosphorylation (Lawler et al., 2001). Alterna-
tively, the observed multisignaling activity of h/rCRF might be
caused by the activation of different types of CRF receptors cou-
pling to G
s
and to G
q/11
to initiate independent activation of
adenylyl cyclase and PLC. These findings suggest a possible inter-
mediary role for differential CRF receptor coupling in determin-
ing distinct endocrine and behavioral stress responses. In support
of this hypothesis, both mouse strains are differentially respon-
sive to neurogenic, psychogenic, and systemic stress, with a
greater stress reactivity and adrenal glucocorticoid release in
Figure 6. Effect of h/rCRF on context-dependent fear conditioning of BALB/c ( A) and C57BL/6N ( B) mice injected with aCSF,
h/rCRF, [Glu
11,16
] astressin,PDBu, or 4
-phorbol 2hr before the training asindicated. For combinedtreatment, [Glu
11,16
] astres
-
sin and BIS-I were given 15 min before h/rCRF application. Freezing was measured in the retention test performed 24 hr after
training. Injectionswere performedintracerebroventricularly (i.c.v.)or intrahippocampally(i.h.) asindicated. *p 0.05 indicates
statistically significant differences versus vehicle-injected animals and naive animals.
Blank et al. CRF-Mediated Excitability in Mouse Hippocampus J. Neurosci., January 15, 2003 23(2):700 –707 705
BALB/cByJ mice than in C57BL/6ByJ mice (Anisman et al.,
2001). Our results add to the existing data showing that the ge-
netic background can affect the behavioral phenotypes of genet-
ically modified mice generated for elucidating the molecular basis
of learning and memory (McNamara et al., 1998; Dobkin et al.,
2000; Dockstader and van der Kooy, 2001). In view of the contri-
bution of the hippocampus to numerous forms of learning (for
review, see Kesner et al., 2000; Kim and Baxter, 2001; Maren,
2001) and the fact that h/rCRF represents an early signal in the
neuroendocrine response to stress (Koob and Bloom, 1985), our
present findings may represent an important step toward under-
standing the cellular and molecular processes underlying inter-
strain variability concerning the impact of stress on learning and
memory (Brush et al., 1988; Francis et al., 1995; Palmer and
Prinz, 1999).
References
Aldenhoff JB, Gruol DL, Rivier J, Vale W, Siggins GR (1983) Corticotropin
releasing factor decreases postburst hyperpolarizations and excites hip-
pocampal neurons. Science 221:875877.
Anisman H, Hayley S, Kelly O, Borowski T, Merali Z (2001) Psychogenic,
neurogenic, and systemic stressor effects on plasma corticosterone and be-
havior: mouse strain-dependent outcomes. Behav Neurosci 115:443454.
Battaglia G, Webster EL, De Souza EB (1987) Characterization of cortico-
tropin-releasing factor receptor-mediated adenylate cyclase activity in the
rat central nervous system. Synapse 1:572581.
Beuzen A, Belzung C (1995) Link between emotional memory and anxiety
states: a study by principal component analysis. Physiol Behav 58:111118.
Blanchard RJ, Blanchard DC (1969) Passive and active reactions to fear-
eliciting stimuli. J Comp Physiol Psychol 68:129135.
Blank T, Nijholt I, Eckart K, Spiess J (2002) Priming of long-term potenti-
ation in mouse hippocampus by corticotropin-releasing factor and acute
stress: implications for hippocampus-dependent learning. J Neurosci
22:3788 3794.
Bolles RC, Riley AL (1973) Freezing as an avoidance response: another look
at the operant-respondent distinction. Learn Motiv 4:268275.
Bourtchouladze R, Abel T, Berman N, Gordon R, Lapidus K, Kandel ER
(1998) Different training procedures recruit either one or two critical
periods for contextual memory consolidation, each of which requires
protein synthesis and PKA. Learn Mem 5:365374.
Brush FR, Del Paine SN, Pellegrino LJ, Rykaszewski IM, Dess NK, Collins PY
(1988) CER suppression, passive-avoidance learning, and stress-induced
suppression of drinking in the Syracuse high- and low-avoidance strains
of rats (Rattus norvegicus). J Comp Psychol 102:337349.
Chen FM, Bilezikjian LM, Perrin MH, Rivier J, Vale W (1986) Cortico-
tropin releasing factor receptor-mediated stimulation of adenylate cyclase
activity in the rat brain. Brain Res 381:4957.
Chen Y, Weng G, Li J, Harry A, Pieroni J, Dingus J, Hildebrandt JD, Guarnieri
F, Weinstein H, Iyengar R (1997) A surface on the G protein beta-
subunit involved in interactions with adenylyl cyclases. Proc Natl Acad Sci
USA 94:27112714.
Dobkin C, Rabe A, Dumas R, El Idrissi A, Haubenstock H, Brown WT (2000)
Fmr1 knockout mouse has a distinctive strain-specific learning impair-
ment. Neuroscience 100:423 429.
Dockstader CL, van der Kooy D (2001) Mouse strain differences in opiate
reward learning are explained by differences in anxiety, not reward or
learning. J Neurosci 21:90779081.
Eckart K, Jahn O, Radulovic J, Tezval H, van Werven L, Spiess J (2001) A
single amino acid serves as an affinity switch between the receptor and the
binding protein of corticotropin-releasing factor: implications for the de-
sign of agonists and antagonists. Proc Natl Acad Sci USA 98:1114211147.
Fanselow MS, Bolles RC (1979) Naloxone and shock-elicited freezing in the
rat. J Comp Physiol Psychol 93:736744.
Fordyce DE, Clark VJ, Paylor R, Wehner JM (1985) Enhancement of
hippocampally-mediated learning and protein kinase C activity by ox-
iracetam in learning-impaired DBA/2 mice. Brain Res 672:170176.
Francis DD, Zaharia MD, Shanks N, Anisman H (1995) Stress-induced dis-
turbances in Morris water-maze performance: interstrain variability.
Physiol Behav 58:57 65.
Franklin KBJ, Paxinos G (1997) The mouse brain in stereotaxic coordinates.
San Diego: Academic.
Grammatopoulos DK, Randeva HS, Levine MA, Kanellopoulou KA, Hill-
house EW (2001) Rat cerebral cortex corticotropin-releasing hormone
receptors: evidence for receptor coupling to multiple G-proteins. J Neu-
rochem 76:509 519.
Haug T, Storm JF (2000) Protein kinase A mediates the modulation of the slow
Ca
2
-dependent K
current, I
sAHP
, by the neuropeptides CRF, VIP, and
CGRP in hippocampal pyramidal neurons. J Neurophysiol 83:20712079.
Hollrigel GS, Chen K, Baram TZ, Soltesz I (1998) The pro-convulsant ac-
tions of corticotropin-releasing hormone in the hippocampus of infant
rats. Neuroscience 84:7179.
Kesner RP, Gilbert PE, Wallenstein GV (2000) Testing neural network
models of memory with behavioral experiments. Curr Opin Neurobiol
10:260 265.
Kim JJ, Baxter MG (2001) Multiple brain-memory systems: the whole does
not equal the sum of its parts. Trends Neurosci 24:324330.
Kim JJ, Fanselow MS (1992) Modality-specific retrograde amnesia of fear.
Science 256:675 677.
Koob GF, Bloom FE (1985) Corticotropin-releasing factor and behavior.
Fed Proc 44:259 263.
Kraft AS, Anderson WB (1983) Phorbol esters increase the amount of Ca
2
,
phospholipid-dependent protein kinase associated with plasma mem-
brane. Nature 301:621 623.
Lawler OA, Miggin SM, Kinsella BT (2001) Protein kinase A-mediated
phosphorylation of serine 357 of the mouse prostacyclin receptor regu-
lates its coupling to G
s
-, to G
i
-, and to G
q-
coupled effector signaling. J Biol
Chem 276:33596 33607.
Lee EH, Lee CP, Wang HI, Lin WR (1993) Hippocampal CRF, NE, and
NMDA system interactions in memory processing in the rat. Synapse
14:144 153.
Lustig KD, Conklin BR, Herzmark P, Taussig R, Bourne HR (1993) Type II
adenylylcyclase integrates coincident signals from G
s
,G
i
, and G
q
. J Biol
Chem 268:13900 13905.
Malenka RC, Madison DV, Andrade R, Nicoll RA (1986) Phorbol esters
mimic some cholinergic actions in hippocampal pyramidal neurons.
J Neurosci 6:475 480.
Maren S (2001) Neurobiology of pavlovian fear conditioning. Annu Rev
Neurosci 24:897931.
McNamara RK, Stumpo DJ, Morel LM, Lewis MH, Wakeland EK, Blackshear
PJ, Lenox RH (1998) Effect of reduced myristoylated alanine-rich C ki-
nase substrate expression on hippocampal mossy fiber development and
spatial learning in mutant mice: transgenic rescue and interactions with
gene background. Proc Natl Acad Sci USA 95:1451714522.
Merchenthaler I (1984) Corticotropin releasing factor (CRF)-like immuno-
reactivity in the rat central nervous system: extrahypothalamic distribu-
tion. Peptides 5:53 69.
Miyata M, Okada D, Hashimoto K, Kano M, Ito M (1999) Corticotropin-
releasing factor plays a permissive role in cerebellar long-term depression.
Neuron 22:763775.
Nowakowski RS (1984) The mode of inheritance of a defect in lamination in
the hippocampus of BALB/c mice. J Neurogenet 1:249258.
Offermanns S, Schultz G, Rosenthal W (1991) Identification of receptor-
activated G proteins with photoreactive GTP analog, [alpha-
32
P]GTP
azidoanilide. Methods Enzymol 195:286301.
Oliverio A, Eleftheriou RS, Bailey DW (1973) A gene influencing active
avoidance performance in mice. Physiol Behav 11:497501.
Palmer AA, Printz MP (1999) Strain differences in Fos expression following
airpuff startle in spontaneously hypertensive and Wistar Kyoto rats. Neu-
roscience 89:965978.
Palmer TM, Gettys TW, Stiles GL (1995) Differential inter-action with and
regulation of multiple G-proteins by the rat A3 adenosine receptor. J Biol
Chem 270:1689516902.
Peeler DF, Nowakowski RS (1987) Genetic factors and the measurement of
exploratory activity. Behav Neural Biol 48:90103.
Phillips RG, LeDoux JE (1992) Differential contribution of amygdala and
hippocampus to contextual and cued fear conditioning. Behav Neurosci
106:274 285.
Phillips RG, LeDoux JE (1994) Lesions of the dorsal hippocampal forma-
tion interfere with background but not foreground contextual fear con-
ditioning. Learn Mem 1:34 44.
Pihoker C, Cain ST, Nemeroff CB (1992) Postnatal development of regional
706
J. Neurosci., January 15, 2003 23(2):700 –707 Blank et al. CRF-Mediated Excitability in Mouse Hippocampus
binding of corticotropin-releasing factor and adenylate cyclase activity in
the rat brain. Prog Neuropsychopharmacol Biol Psychiatry 16:581586.
Radulovic J, Ru¨hmann A, Liepold T, Spiess J (1999) Modulation of learning
and anxiety by corticotropin-releasing factor (CRF) and stress: differen-
tial roles of CRF receptors 1 and 2. J Neurosci 19:50165025.
Ru¨hmann A,Ko¨pkeAKE, Dautzenberg FM, Spiess J (1996) Synthesis and char-
acterization of a photoactivatable analog of corticotropin-releasing factor for
specific receptor labeling. Proc Natl Acad Sci USA 93:1060910613.
Schwindinger WF, Fredericks J, Watkins L, Robinson H, Bathon JM, Pines M,
Suva LJ, Levine MA (1998) Coupling of the PTH/PTHrP receptor to
multiple G-proteins: direct demonstration of receptor activation of G
s
,
G
q/11
, and G
i(1)
by [alpha-
32
P]GTP-gamma-azidoanilide photoaffinity
labeling. Endocrine 8:201209.
Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, Provenzano
MD, Fujimoto EK, Goeke NM, Olson BJ, Klenk DC (1985) Measure-
ment of protein using bicinchoninic acid. Anal Biochem 150:76 85.
Spiess J, Rivier J, Rivier C, Vale W (1981) Primary structure of corticotropin-
releasing factor from ovine hypothalamus. Proc Natl Acad Sci USA
78:65176521.
Stiedl O, Birkenfeld K, Palve M, Spiess J (2000) Impairment of conditioned
contextual fear of C57BL/6J mice by intracerebral injections of the NMDA
receptor antagonist APV. Behav Brain Res 116:157168.
Swanson LW, Sawchenko PE, Rivier J, Vale WW (1983) Organization of ovine
corticotropin-releasing factor immunoreactive cells and fibers in the rat
brain: an immunohistochemical study. Neuroendocrinology 36:165186.
Takuma K, Matsuda T, Yoshikawa T, Kitanaka J, Gotoh M, Hayata K, Baba A
(1994) Corticotropin-releasing factor stimulates Ca
2
influx in cultured
rat astrocytes. Biochem Biophys Res Commun 199:11031107.
Taussig R, Quarmby LM, Gilman AG (1993) Regulation of purified type I
and type II adenylylcyclases by G protein beta gamma subunits. J Biol
Chem 268:9 12.
Ulisse S, Fabbri A, Tinajero JC, Dufau ML (1990) A novel mechanism of
action of corticotropin releasing factor in rat Leydig cells. J Biol Chem
265:1964 1971.
Vale W, Spiess J, Rivier C, Rivier J (1981) Characterization of a 41-residue
ovine hypothalamic peptide that stimulates secretion of corticotropin and
beta-endorphin. Science 213:1394 1397.
Van Pett K, Viau V, Bittencourt JC, Chan RK, Li HY, Arias C, Prins GS, Perrin M,
Vale W, Sawchenko PE (2000) Distribution of mRNAs encoding CRF re-
ceptors in brain and pituitary of rat and mouse. J Comp Neurol 428:191212.
Wang HL, Wayner MJ, Chai CY, Lee EH (1998) Corticotropin-releasing
factor produces a long-lasting enhancement of synaptic efficacy in the
hippocampus. Eur J Neurosci 10:34283437.
Wang HL, Tsai LY, Lee EH (2000) Corticotropin-releasing factor produces
a protein synthesis-dependent long-lasting potentiation in dentate gyrus
neurons. J Neurophysiol 83:343349.
Blank et al. CRF-Mediated Excitability in Mouse Hippocampus J. Neurosci., January 15, 2003
23(2):700 –707 707
... Depending on which CRF-R is activated by CRF, different molecular signaling pathways can be activated by different G-proteins [38][39][40].Nevertheless, the molecular mechanisms by which CRF peptides regulate spine density in acute stress and synaptic structural changes in response to the stress are still unclear. We are assuming that the major function of CRF is to regulate the distribution of transmembrane proteins in the postsynaptic membrane, most probably via cytoskeletal changes. ...
Article
Full-text available
The neuropeptide corticotropin-releasing factor (CRF) exerts a pivotal role in modulating neuronal activity in the mammalian brain. The effects of CRF exhibit notable variations, depending on factors such as duration of exposure, concentration, and anatomical location. In the CA1 region of the hippocampus, the impact of CRF is dichotomous: chronic exposure to CRF impairs synapse formation and dendritic integrity, whereas brief exposure enhances synapse formation and plasticity. In the current study, we demonstrate long-term effects of acute CRF on the density and stability of mature mushroom spines ex vivo. We establish that both CRF receptors are present in this hippocampal region, and we pinpoint their precise subcellular localization within synapses by electron microscopy. Furthermore, both in vivo and ex vivo data collectively demonstrate that a transient surge of CRF in the CA1 activates the cyclin-dependent kinase 5 (Cdk5)-pathway. This activation leads to a notable augmentation in CRF-dependent spine formation. Overall, these data suggest that upon acute release of CRF in the CA1-SR synapse, both CRF-Rs can be activated and promote synaptic plasticity via activating different downstream signaling pathways, such as the Cdk5-pathway.
... The specific signaling systems altered following deafening in this study provide a set of candidate mechanisms that may influence song. For example, corticotropinreleasing hormone binding protein (CRHBP) , one of the most strongly downregulated genes in RA following deafening, modulates activity in the CRH signaling pathway (Kemp et al., 1998), which has diverse effects on long-term potentiation, neuronal excitability, and spine dynamics in central circuits (Aldenhoff et al., 1983;Blank et al., 2003;Chen et al., 2008;Fox and Gruol, 1993;Kratzer et al., 2013;Li et al., 2016). Such evidence suggests that the dynamic modulation of neuropeptides could play a prominent role in regulating birdsong stability and plasticity and may similarly influence the control of other stable sensorimotor skills such as human speech. ...
Article
Full-text available
Sensory feedback is required for the stable execution of learned motor skills, and its loss can severely disrupt motor performance. The neural mechanisms that mediate sensorimotor stability have been extensively studied at systems and physiological levels, yet relatively little is known about how disruptions to sensory input alter the molecular properties of associated motor systems. Songbird courtship song, a model for skilled behavior, is a learned and highly structured vocalization that is destabilized following deafening. Here, we sought to determine how the loss of auditory feedback modifies gene expression and its coordination across the birdsong sensorimotor circuit. To facilitate this system-wide analysis of transcriptional responses, we developed a gene expression profiling approach that enables the construction of hundreds of spatially-defined RNA-sequencing libraries. Using this method, we found that deafening preferentially alters gene expression across birdsong neural circuitry relative to surrounding areas, particularly in premotor and striatal regions. Genes with altered expression are associated with synaptic transmission, neuronal spines, and neuromodulation and show a bias toward expression in glutamatergic neurons and Pvalb/Sst-class GABAergic interneurons. We also found that connected song regions exhibit correlations in gene expression that were reduced in deafened birds relative to hearing birds, suggesting that song destabilization alters the inter-region coordination of transcriptional states. Finally, lesioning LMAN, a forebrain afferent of RA required for deafening-induced song plasticity, had the largest effect on groups of genes that were also most affected by deafening. Combined, this integrated transcriptomics analysis demonstrates that the loss of peripheral sensory input drives a distributed gene expression response throughout associated sensorimotor neural circuitry and identifies specific candidate molecular and cellular mechanisms that support the stability and plasticity of learned motor skills.
... The specific signaling systems altered following deafening in this study provide a set of candidate mechanisms that may influence song. For example, corticotropinreleasing hormone binding protein (CRHBP) , one of the most strongly downregulated genes in RA following deafening, modulates activity in the CRH signaling pathway (Kemp et al., 1998), which has diverse effects on long-term potentiation, neuronal excitability, and spine dynamics in central circuits (Aldenhoff et al., 1983;Blank et al., 2003;Chen et al., 2008;Fox and Gruol, 1993;Kratzer et al., 2013;Li et al., 2016). Such evidence suggests that the dynamic modulation of neuropeptides could play a prominent role in regulating birdsong stability and plasticity and may similarly influence the control of other stable sensorimotor skills such as human speech. ...
Article
Full-text available
Sensory feedback is required for the stable execution of learned motor skills, and its loss can severely disrupt motor performance. The neural mechanisms that mediate sensorimotor stability have been extensively studied at systems and physiological levels, yet relatively little is known about how disruptions to sensory input alter the molecular properties of associated motor systems. Songbird courtship song, a model for skilled behavior, is a learned and highly structured vocalization that is destabilized following deafening. Here, we sought to determine how the loss of auditory feedback modifies gene expression and its coordination across the birdsong sensorimotor circuit. To facilitate this system-wide analysis of transcriptional responses, we developed a gene expression profiling approach that enables the construction of hundreds of spatially-defined RNA-sequencing libraries. Using this method, we found that deafening preferentially alters gene expression across birdsong neural circuitry relative to surrounding areas, particularly in premotor and striatal regions. Genes with altered expression are associated with synaptic transmission, neuronal spines, and neuromodulation and show a bias toward expression in glutamatergic neurons and Pvalb/Sst-class GABAergic interneurons. We also found that connected song regions exhibit correlations in gene expression that were reduced in deafened birds relative to hearing birds, suggesting that song destabilization alters the inter-region coordination of transcriptional states. Finally, lesioning LMAN, a forebrain afferent of RA required for deafening-induced song plasticity, had the largest effect on groups of genes that were also most affected by deafening. Combined, this integrated transcriptomics analysis demonstrates that the loss of peripheral sensory input drives a distributed gene expression response throughout associated sensorimotor neural circuitry and identifies specific candidate molecular and cellular mechanisms that support the stability and plasticity of learned motor skills.
... The specific signaling systems altered following deafening in this study provide a set of candidate mechanisms that may influence song. For example, corticotropinreleasing hormone binding protein (CRHBP) , one of the most strongly downregulated genes in RA following deafening, modulates activity in the CRH signaling pathway (Kemp et al., 1998), which has diverse effects on long-term potentiation, neuronal excitability, and spine dynamics in central circuits (Aldenhoff et al., 1983;Blank et al., 2003;Chen et al., 2008;Fox and Gruol, 1993;Kratzer et al., 2013;Li et al., 2016). Such evidence suggests that the dynamic modulation of neuropeptides could play a prominent role in regulating birdsong stability and plasticity and may similarly influence the control of other stable sensorimotor skills such as human speech. ...
Article
Full-text available
Sensory feedback is required for the stable execution of learned motor skills, and its loss can severely disrupt motor performance. The neural mechanisms that mediate sensorimotor stability have been extensively studied at systems and physiological levels, yet relatively little is known about how disruptions to sensory input alter the molecular properties of associated motor systems. Songbird courtship song, a model for skilled behavior, is a learned and highly structured vocalization that is destabilized following deafening. Here, we sought to determine how the loss of auditory feedback modifies gene expression and its coordination across the birdsong sensorimotor circuit. To facilitate this system-wide analysis of transcriptional responses, we developed a gene expression profiling approach that enables the construction of hundreds of spatially-defined RNA-sequencing libraries. Using this method, we found that deafening preferentially alters gene expression across birdsong neural circuitry relative to surrounding areas, particularly in premotor and striatal regions. Genes with altered expression are associated with synaptic transmission, neuronal spines, and neuromodulation and show a bias toward expression in glutamatergic neurons and Pvalb/Sst-class GABAergic interneurons. We also found that connected song regions exhibit correlations in gene expression that were reduced in deafened birds relative to hearing birds, suggesting that song destabilization alters the inter-region coordination of transcriptional states. Finally, lesioning LMAN, a forebrain afferent of RA required for deafening-induced song plasticity, had the largest effect on groups of genes that were also most affected by deafening. Combined, this integrated transcriptomics analysis demonstrates that the loss of peripheral sensory input drives a distributed gene expression response throughout associated sensorimotor neural circuitry and identifies specific candidate molecular and cellular mechanisms that support the stability and plasticity of learned motor skills.
... The specific signaling systems altered following deafening in this study provide a set of candidate mechanisms that may influence song. For example, corticotropinreleasing hormone binding protein (CRHBP) , one of the most strongly downregulated genes in RA following deafening, modulates activity in the CRH signaling pathway (Kemp et al., 1998), which has diverse effects on long-term potentiation, neuronal excitability, and spine dynamics in central circuits (Aldenhoff et al., 1983;Blank et al., 2003;Chen et al., 2008;Fox and Gruol, 1993;Kratzer et al., 2013;Li et al., 2016). Such evidence suggests that the dynamic modulation of neuropeptides could play a prominent role in regulating birdsong stability and plasticity and may similarly influence the control of other stable sensorimotor skills such as human speech. ...
Article
Full-text available
Sensory feedback is required for the stable execution of learned motor skills, and its loss can severely disrupt motor performance. The neural mechanisms that mediate sensorimotor stability have been extensively studied at systems and physiological levels, yet relatively little is known about how disruptions to sensory input alter the molecular properties of associated motor systems. Songbird courtship song, a model for skilled behavior, is a learned and highly structured vocalization that is destabilized following deafening. Here, we sought to determine how the loss of auditory feedback modifies gene expression and its coordination across the birdsong sensorimotor circuit. To facilitate this system-wide analysis of transcriptional responses, we developed a gene expression profiling approach that enables the construction of hundreds of spatially-defined RNA-sequencing libraries. Using this method, we found that deafening preferentially alters gene expression across birdsong neural circuitry relative to surrounding areas, particularly in premotor and striatal regions. Genes with altered expression are associated with synaptic transmission, neuronal spines, and neuromodulation and show a bias toward expression in glutamatergic neurons and Pvalb/Sst-class GABAergic interneurons. We also found that connected song regions exhibit correlations in gene expression that were reduced in deafened birds relative to hearing birds, suggesting that song destabilization alters the inter-region coordination of transcriptional states. Finally, lesioning LMAN, a forebrain afferent of RA required for deafening-induced song plasticity, had the largest effect on groups of genes that were also most affected by deafening. Combined, this integrated transcriptomics analysis demonstrates that the loss of peripheral sensory input drives a distributed gene expression response throughout associated sensorimotor neural circuitry and identifies specific candidate molecular and cellular mechanisms that support the stability and plasticity of learned motor skills.
Article
The role of the TAAR1 receptor, one of the trace amine-associated receptors (TAARs) family, in the formation of the behavioral component of the stress response was studied. The behavior of female TAAR1 knockout mice and wild-type (WT) mice was investigated in tests of elevated plus maze and elevated zero maze (EPM and EZM) and forced swimming test (FST) under normal conditions and after uncontrolled restraint stress exposure for 30 min. In the EPM test, the initial level of locomotor and exploratory activity, as well as the anxiety, was identical in both groups of mice. In the EZM test, the initial indicators of anxiety in female TAAR1 KO mice compared to female WT mice were higher, and locomotor activity was lower. When testing mice in the EZM 30 minutes after the end of stress exposure, it was found that the anxiety in female WT mice sharply increased, and the indicators of locomotor activity and exploratory behavior significantly decreased. The behavioral indicators in the EZM test in TAAR1 KO mice before and after stress were identical. A pronounced behavioral component of the stress response was observed in both TAAR1 KO and WT mice during testing in EPM. There were no significant differences between TAAR1 KO and WT mice during testing in EPM four hours after stress exposure. In the FST test the latency to the first immobility was initially longer in TAAR1 KO mice compared to the WT mice, but 24 h after the stress this indicator has significantly decreased. As a result, TAAR1 KO and WT mice no longer differed in all behavioral indicators in the FST. Three weeks after acute restraint stress, both TAAR1 KO and WT groups showed a significant increase in immobility duration and a decrease in latency to the first immobility, however no difference between the both groups of animals were found. Thereby, we found the complete absence of behavioral change immediately after stressor exposure in TAAR1 KO compared to the WT mice.
Article
Background and Purpose Pancreatic islets are modulated by cross‐talk among different cell types and paracrine signalling plays important roles in maintaining glucose homeostasis. Urocortin 3 (UCN3) secreted by pancreatic β cells activates the CRF 2 receptor (CRF 2 R) and downstream pathways mediated by different G protein or arrestin subtypes in δ cells to cause somatostatin (SST) secretion, and constitutes an important feedback circuit for glucose homeostasis. Experimental Approach Here, we used Arrb1 −/− , Arrb2 −/− , Gs fl/fl and Gq fl/fl knockout mice, the G 11 ‐shRNA‐GFP fl/fl lentivirus, as well as functional assays and pharmacological characterization to study how the coupling of G s , G 11 and β ‐arrestin1 to CRF 2 R contributed to UCN3‐induced SST secretion in pancreatic δ cells. Key Results Our study showed that CRF 2 R coupled to a panel of G protein and arrestin subtypes in response to UCN3 engagement. While RyR3 phosphorylation by PKA at the S156, S2706 and S4697 sites may underlie the Gs‐mediated UCN3‐ CRF 2 R axis for SST secretion, the interaction of SYT1 with β ‐arrestin1 is also essential for efficient SST secretion downstream of CRF 2 R. The specific expression of the transcription factor Stat6 may contribute to G 11 expression in pancreatic δ cells. Furthermore, we found that different UCN3 concentrations may have distinct effects on glucose homeostasis, and these effects may depend on different CRF 2 R downstream effectors. Conclusions and Implications Collectively, our results provide a landscape view of signalling mediated by different G protein or arrestin subtypes downstream of paracrine UCN3‐ CRF 2 R signalling in pancreatic β ‐ δ ‐cell circuits, which may facilitate the understanding of fine‐tuned glucose homeostasis networks.
Article
Full-text available
The role of the TAAR1 receptor, one of the trace amine-associated receptors (TAARs) family, in the formation of the behavioral component of the stress response was studied. The behavior of female TAAR1 knockout (TAAR1 KO) mice and wild-type (WT) mice was investigated in tests of elevated plus maze and elevated zero maze (EPM and EZM) and forced swimming test (FST) under normal conditions and after uncontrolled restraint stress exposure for 30 min. In the EPM test, the initial level of locomotor and exploratory activity, as well as the anxiety, was identical in both groups of mice. In the EZM test, the initial indicators of anxiety in female TAAR1 KO mice compared to female WT mice were higher, and locomotor activity was lower. When testing mice in the EZM 30 minutes after the end of stress exposure, it was found that the anxiety in female WT mice sharply increased, and the indicators of locomotor activity and exploratory behavior significantly decreased. The behavioral indicators in the EZM test in TAAR1 KO mice before and after stress were identical. A pronounced behavioral component of the stress response was observed in both TAAR1 KO and WT mice during testing in EPM. There were no significant differences between TAAR1 KO and WT mice during testing in EPM four hours after stress exposure. In the FST test the latency to the first immobility was initially longer in TAAR1 KO mice compared to the WT mice, but 24 hours after the stress this indicator has significantly decreased. As a result, TAAR1 KO and WT mice no longer differed in all behavioral indicators in the FST. Three weeks after acute restraint stress, both TAAR1 KO and WT groups showed a significant increase in immobility duration and a decrease in latency to the first immobility, however no difference between the both groups of animals were found. Thereby, we found the complete absence of behavioral change immediately after stressor exposure in TAAR1 KO compared to the WT mice.
Article
Ethanol (EtOH) has effects on numerous cellular molecular targets, and alterations in synaptic function are prominent among these effects. Acute exposure to EtOH activates or inhibits the function of proteins involved in synaptic transmission, while chronic exposure often produces opposing and/or compensatory/homeostatic effects on the expression, localization, and function of these proteins. Interactions between different neurotransmitters (e.g., neuropeptide effects on release of small molecule transmitters) can also influence both acute and chronic EtOH actions. Studies in intact animals indicate that the proteins affected by EtOH also play roles in the neural actions of the drug, including acute intoxication, tolerance, dependence, and the seeking and drinking of EtOH. The present chapter is an update of our previous Lovinger and Roberto (Curr Top Behav Neurosci 13:31-86, 2013) chapter and reviews the literature describing these acute and chronic synaptic effects of EtOH with a focus on adult animals and their relevance for synaptic transmission, plasticity, and behavior.
Article
Full-text available
The RIIβ subunit of cAMP‐dependent protein kinase A (PKA) is expressed in the brain and adipose tissue. RIIβ‐knockout mice show leanness and increased UCP1 in brown adipose tissue. The authors have previously reported that RIIβ reexpression in hypothalamic GABAergic neurons rescues the leanness. However, whether white adipose tissue (WAT) browning contributes to the leanness and whether RIIβ‐PKA in these neurons governs WAT browning are unknown. Here, this work reports that RIIβ‐KO mice exhibit a robust WAT browning. RIIβ reexpression in dorsal median hypothalamic GABAergic neurons (DMH GABAergic neurons) abrogates WAT browning. Single‐cell sequencing, transcriptome sequencing, and electrophysiological studies show increased GABAergic activity in DMH GABAergic neurons of RIIβ‐KO mice. Activation of DMH GABAergic neurons or inhibition of PKA in these neurons elicits WAT browning and thus lowers body weight. These findings reveal that RIIβ‐PKA in DMH GABAergic neurons regulates WAT browning. Targeting RIIβ‐PKA in DMH GABAergic neurons may offer a clinically useful way to promote WAT browning for treating obesity and other metabolic disorders.
Article
Full-text available
Type I and type II adenylylcyclases have been purified after expression in Sf9 cells, each by application of a two-step purification protocol. The specific activities of the essentially homogeneous enzymes are approximately 7 and 2 mumol.min-1.mg-1, respectively. Each purified enzyme preparation is activated by G(salpha), but they are regulated in an opposite fashion by G protein betagamma subunits. Purified betagamma inhibits G(salpha)-stimulated type I adenylylcyclase directly, while betagamma activates type II adenylylcyclase and potentiates the G(salpha)-mediated stimulation of the enzyme. This is the first demonstration of the activation of a purified effector molecule by G protein betagamma subunits.
Article
Full-text available
Two G protein-coupled receptors have been identified that bind corticotropin-releasing factor (CRF) and urocortin (UCN) with high affinity. Hybridization histochemical methods were used to shed light on controversies concerning their localization in rat brain, and to provide normative distributional data in mouse, the standard model for genetic manipulation in mammals. The distribution of CRF-R1 mRNA in mouse was found to be fundamentally similar to that in rat, with expression predominating in the cerebral cortex, sensory relay nuclei, and in the cerebellum and its major afferents. Pronounced species differences in distribution were few, although more subtle variations in the relative strength of R1 expression were seen in several forebrain regions. CRF-R2 mRNA displayed comparable expression in rat and mouse brain, distinct from, and more restricted than that of CRF-R1. Major neuronal sites of CRF-R2 expression included aspects of the olfactory bulb, lateral septal nucleus, bed nucleus of the stria terminalis, ventromedial hypothalamic nucleus, medial and posterior cortical nuclei of the amygdala, ventral hippocampus, mesencephalic raphe nuclei, and novel localizations in the nucleus of the solitary tract and area postrema. Several sites of expression in the limbic forebrain were found to overlap partially with ones of androgen receptor expression. In pituitary, rat and mouse displayed CRF-R1 mRNA signal continuously over the intermediate lobe and over a subset of cells in the anterior lobe, whereas CRF-R2 transcripts were expressed mainly in the posterior lobe. The distinctive expression pattern of CRF-R2 mRNA identifies additional putative central sites of action for CRF and/or UCN. Constitutive expression of CRF-R2 mRNA in the nucleus of the solitary tract, and stress-inducible expression of CRF-R1 transcripts in the paraventricular nucleus may provide a basis for understanding documented effects of CRF-related peptides at a loci shown previously to lack a capacity for CRF-R expression or CRF binding. Other such “mismatches” remain to be reconciled. J. Comp. Neurol. 428:191–212, 2000. © 2000 Wiley-Liss, Inc.
Article
The effects of several stressors were assessed in inbred strains of mice, BALB/cByJ and C57BL/6ByJ, thought to be differentially reactive to stressors. Behavioral reactivity was greater in BALB/cByJ mice with respect to open-field emergence, step-down responding, response to a predator (rat) or to fox urine odor. Neurogenic insults (e.g., footshock, forced swim, restraint) and a systematic stressor (intraperitoneal interlukin-1β treatment) likewise provoked a greater rise of plasma corticisterone in the BALB/cByJ mice. Psychogenic stressors (e.g., novel open-field exposure, acoustic startle stimuli) also enhanced plasma corticosterone to a greater extent in BALB/cByJ mice, but such an outcome was not apparent following predator-related cues. In appears that whereas stressor reactivity and adrenal glucorticoid release may be exaggerated in BALB/cByJ mice, such effects may be dependent on the specific characteristic of the stressor situation. (PsycINFO Database Record (c) 2012 APA, all rights reserved)
Article
Bicinchoninic acid, sodium salt, is a stable, water-soluble compound capable of forming an intense purple complex with cuprous ion (Cu1+) in an alkaline environment. This reagent forms the basis of an analytical method capable of monitoring cuprous ion produced in the reaction of protein with alkaline Cu2+ (biuret reaction). The color produced from this reaction is stable and increases in a proportional fashion over a broad range of increasing protein concentrations. When compared to the method of Lowry et al., the results reported here demonstrate a greater tolerance of the bicinchoninate reagent toward such commonly encountered interferences as nonionic detergents and simple buffer salts. The stability of the reagent and resulting chromophore also allows for a simplified, one-step analysis and an enhanced flexibility in protocol selection. This new method maintains the high sensitivity and low protein-to-protein variation associated with the Lowry technique.
Article
Marked differences were observed across strains of mice (i.e., DBA/2J, C57BL/6J, BALB/cByJ and CD-1 mice) in acquisition, performance and reversal of a place learning response in a Morris water-maze. While DBA/2J, C57BL/6J and CD-1 mice typically learned the response readily, only 20% of BALB/cByJ mice acquired the response. Commensurate with the effects of hippocampal disturbances, the performance deficits in BALB/cByJ mice were not evident when the position of the platform in the water-maze was cued. Exposure to uncontrollable foot shock did not affect the acquisition or performance of this response in the former three strains, but provoked a modest disruption of reversal performance in DBA/2J mice and markedly impaired reversal performance in BALB/cByJ mice. It seemed, however, that the response strategies adopted in these strains could be distinguished from one another. In the reversal paradigm BALB/cByJ mice initially persisted in returning to the original training quadrant rather than to the new goal quadrant. Following 2 days of training the perseveration was no longer apparent and animals seemed to adopt a random search strategy. In contrast, DBA/2J mice, which exhibited a smaller stress-induced disturbance, did not display a perseverative response style. These data suggest that inescapable shock does not disturb response-outcome associations, but may result from the induction of a perseverative response style. However, it appears that the mechanisms responsible for an interference of performance may not be uniform across strains. The mechanisms responsible for the poor performance of BALB/cByJ mice have not been determined, but the behavioral profile demonstrated is consistent with that associated with hippocampal disturbances and excessive glucocorticoid release in response to environmental stressors.
Article
Groups of rats were trained with shock either contingent on freezing (punishment procedure) or contingent on not freezing (avoidance procedure). Although the different contingencies produced different levels of freezing behavior, these levels were attained immediately rather than over a number of trials. This result, together with the results of control rats, suggest that while freezing can be controlled by both punishment and avoidance procedures, in both cases the effects on freezing are due to elicitation rather than learning.
Article
The wide distribution of corticotrophin-releasing hormone (CRH) receptors in brain and periphery appear to be important in integrating the responses of the brain, endocrine and immune systems to physiological, psychological and immunological stimuli. The type 1 receptors are highly expressed throughout the cerebral cortex, a region involved in cognitive function and modulation of stress responses, where they are coupled to the adenylyl cyclase system. Using techniques that analyse receptor-mediated guanine-nucleotide binding protein (G-proteins) activation, we recently demonstrated that expressed type 1alpha CRH receptors are capable of activating multiple G-proteins, which suggests that CRH can regulate multiple signalling pathways. In an effort to characterize the intracellular signals generated by CRH in the rat cerebral cortex we sought to identify G-proteins activated by CRH in a physiological membrane environment. Rat cerebral cortical membrane suspensions were analysed for the ability of CRH to
Article
Most contemporary theories of memory are based on the assumption that memory can be divided into multiple psychological systems that are subserved by different neural substrates and that contribute to performance in a relatively independent manner. Although the study of individual memory systems has proved to be enormously useful, recent data increasingly point towards complex interactions between memory systems during performance of any given memory task. Three basic classes of interactions between different memory systems (competition, synergism and independence) are presented that appear to be congruent with the findings of many behavioral studies. Consideration of interactions among multiple memory systems will enhance our current understanding of memory by encouraging the view that memory systems are dynamic interactive units, rather than independent modules that act in isolation.