Next Article in Journal
A Case of Double Standard: Sex Differences in Multiple Sclerosis Risk Factors
Next Article in Special Issue
Excitatory Effects of Calcitonin Gene-Related Peptide (CGRP) on Superficial Sp5C Neurons in Mouse Medullary Slices
Previous Article in Journal
Effects of Lifestyle Intervention in Tissue-Specific Lipidomic Profile of Formerly Obese Mice
Previous Article in Special Issue
Leucokinin and Associated Neuropeptides Regulate Multiple Aspects of Physiology and Behavior in Drosophila
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Brain Region-Dependent Effects of Neuropeptide Y on Conditioned Social Fear and Anxiety-Like Behavior in Male Mice

Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(7), 3695; https://doi.org/10.3390/ijms22073695
Submission received: 19 March 2021 / Revised: 29 March 2021 / Accepted: 30 March 2021 / Published: 2 April 2021
(This article belongs to the Special Issue Neuropeptides, Receptors, and Behavior)

Abstract

:
Neuropeptide Y (NPY) has anxiolytic-like effects and facilitates the extinction of cued and contextual fear in rodents. We have previously shown that the intracerebroventricular administration of NPY reduces the expression of social fear in a mouse model of social fear conditioning (SFC). In the present study, we aimed to identify the brain regions that mediate these effects of NPY. We show that NPY (0.1 nmol/0.2 µL/side) reduces the expression of SFC-induced social fear in a brain-region-dependent manner. In more detail, NPY reduced the expression of social fear when administered into the dorsolateral septum (DLS) and central amygdala (CeA), but not when administered into the dorsal hippocampus (DH), medial amygdala (MeA) and basolateral amygdala (BLA). We also investigated whether the reduced expression of social fear might partly be due to a reduced anxiety-like behavior, and showed that NPY exerted anxiolytic-like effects when administered into the DH, DLS, CeA and BLA, but not when administered into the MeA. This study identifies the DLS and the CeA as brain regions mediating the effects of NPY on the expression of social fear and suggests that partly distinct neural circuitries mediate the effects of NPY on the expression of social fear and on anxiety-like behavior.

1. Introduction

Neuropeptide Y (NPY) is the most abundant and widely distributed neuropeptide in the mammalian brain. It regulates important biological and pathophysiological functions, such as blood pressure, food intake, neuroendocrine secretions, seizures, neuronal excitability and neuroplasticity [1,2,3,4,5,6]. These effects are mediated by at least five different G-protein-coupled receptors, among which the Y1, Y2, Y4 and Y5 subtypes are localized in the brain [7,8,9]. NPY is expressed at high levels in brain regions involved in emotional behavior and fear-related behavior, suggesting a regulatory role of NPY in these behaviors. The brain regions expressing NPY include, among others, the amygdala, hippocampus, septum, cerebral cortex, locus coeruleus, periaqueductal gray, basal ganglia, hypothalamus and thalamus [10,11,12]. Indeed, a considerable amount of the literature supports an anxiolytic function of NPY [13,14,15]. As such, intracerebroventricular (i.c.v.) administration of NPY has anxiolytic-like effects [16], which can also be seen after NPY administration directly into the central amygdala (CeA) [17], locus coeruleus [18], lateral septum [19], dentate gyrus and the CA1 region of the hippocampus [20], indicating that these brain regions mediate the anxiolytic-like effects of NPY. The anxiolytic-like effects can also be seen in rats with a higher innate number of NPY-positive cells in the CeA [21] and after localized overexpression of NPY within the amygdala [22]. Similar brain region-specific effects of NPY were described on social interaction. As such, NPY promotes social interaction when administered into the dorsolateral septum (DLS) [23] and basolateral amygdala (BLA) [24], but does not affect social interaction when administered into the intramedial septum [23] and CeA [24]. These anxiolytic and prosocial effects of NPY suggest its potential benefit in disorders associated with social anxiety and fear. This might also be suggested by the fact that NPY affects different aspects of fear-related behavior. In fear conditioning paradigms, for example, i.c.v.-administered NPY impairs the acquisition of cued and contextual fear [25,26,27], impairs the retention and retrieval of cued fear [28,29,30] and facilitates the extinction of cued and contextual fear [27,28,31,32,33]. The effects of NPY on the expression of cued fear also seem to be brain region-dependent, since NPY inhibits the expression of cued fear when administered into the BLA, but not when administered into the medial amygdala (MeA) [28,29]. These studies suggest that NPY acts as a resilience factor against exaggerated fear responses after stressful and adverse events and that these effects are brain-region-dependent. However, little is known about the involvement of NPY in the modulation of fear responses after adverse social events or about the neural circuitries mediating these effects.
Recently, in a model of social fear conditioning (SFC), we could show that i.c.v.-administered NPY reduces the expression of social fear, but does not alter the acquisition of social fear [34]. This suggests that although NPY does not prevent the formation of aversive social memories, it might improve the recovery from a traumatic social experience. Moreover, i.c.v.-administered NPY also reduces the expression of antidepressant-resistant social fear [35], further supporting its potential benefit in disorders associated with social anxiety and fear. In the present study, we aimed to identify the brain regions mediating the effects of NPY on the expression of social fear. Therefore, we administered NPY into the dorsal hippocampus (DH), DLS, CeA, MeA and BLA before social fear extinction. These brain regions were selected because the amygdala and the septo-hippocampal circuits have strong NPY-ergic innervation [10,11,36], and are important components of the neural circuitry controlling anxiety-related behaviors, fear-related behaviors, social behaviors and stress responses [36,37,38,39]. We also investigated whether the effects of NPY on the expression of social fear might be due to altered anxiety-like behavior.

2. Results

To investigate whether NPY alters the expression of social fear when administered into the DH, DLS, CeA, MeA and/or BLA, separate groups of conditioned (SFC+) and unconditioned (SFC) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) into these brain regions 10 min before social fear extinction on day 2. On day 1, during SFC, SFC+ mice received mild electric foot shocks each time they investigated an unknown conspecific, whereas SFC mice investigated an unknown conspecific without receiving foot shocks. On day 2, during social fear extinction, we assessed the time that the SFC+ and SFC mice spent investigating three empty cages (i.e., non-social investigation) and six unknown conspecifics enclosed in wire mesh cages (i.e., social investigation) as a read-out of non-social and social fear, respectively.

2.1. NPY Does Not Affect the Expression of Social Fear When Administered into the DH

During SFC on day 1, SFC+ and SFC mice showed similar investigation of the non-social stimulus (empty cage), reflecting a similar pre-conditioning non-social anxiety (Figure 1a; F(3,28) = 0.684; p = 0.570). All SFC+ mice received a similar number of foot shocks during SFC, reflecting similar levels of distress during conditioning and similar social fear learning (T(14) = −0.798; p = 0.438). During social fear extinction on day 2, all SFC+ and SFC mice showed a similar investigation of the non-social stimuli (three empty cages; ns1–ns3), indicating that SFC did not induce an unspecific non-social fear (Figure 1b). However, all SFC+ mice showed a reduced investigation of the social stimuli (six unknown conspecifics; s1–s6) compared with respective SFC mice, independent of treatment, reflecting an increased social fear (conditioning effect F(1,28) = 53.556; p < 0.001; conditioning x treatment effect F(1,28) = 0.024; p = 0.879). This indicates that NPY does not reduce the expression of social fear when administered into the DH.

2.2. NPY Reduces the Expression of Social Fear When Administered into the DLS

During SFC on day 1, SFC+ and SFC mice showed similar investigation of the non-social stimulus, reflecting a similar pre-conditioning non-social anxiety (Figure 2a; F(3,28) = 0.661; p = 0.583). All SFC+ mice received a similar number of foot shocks during SFC, reflecting similar levels of distress during conditioning and similar social fear learning (T(14) = 0.532; p = 0.603). During social fear extinction on day 2 (Figure 2b), all SFC+ and SFC mice showed a similar non-social investigation, indicating that SFC did not induce an unspecific non-social fear. While Veh-treated SFC+ mice showed a reduced social investigation compared with all other groups, reflecting social fear, NPY increased social investigation starting from the first social stimulus, reflecting a reduced expression of social fear (conditioning x treatment effect F(1,28) = 47.150; p < 0.001; stimulus x conditioning x treatment effect F(8,224) = 11.611; p < 0.001). This indicates that NPY reduces the expression of social fear when administered into the DLS.

2.3. NPY Reduces the Expression of Social Fear When Administered into the CeA, But Not When Administered into the MeA or BLA

During SFC on day 1, SFC+ and SFC mice showed similar investigation of the non-social stimulus, reflecting a similar pre-conditioning non-social anxiety (CeA: Figure 3a; F(3,28) = 0.236; p = 0.871; MeA: Figure 3c; F(3,28) = 0.429; p = 0.734; BLA: Figure 3e; F(3,28) = 1.040; p = 0.390). All SFC+ mice received a similar number of foot shocks during SFC, reflecting similar levels of distress during conditioning and similar social fear learning (CeA: T(14) = −0.323; p = 0.751; MeA: T(14) = −0.851; p = 0.409; BLA: T(14) = 1.111; p = 0.285). During social fear extinction on day 2 (Figure 3b,d,f), all mice showed a similar non-social investigation, indicating that SFC did not induce an unspecific non-social fear. While all Veh-treated SFC+ mice showed social fear, NPY reduced the expression of social fear when administered into the CeA, but not when administered into the MeA or BLA (CeA: Figure 3b; conditioning x treatment effect F(1,28) = 17.210; p < 0.001; stimulus x conditioning x treatment effect F(8,224) = 2.769; p = 0.006; MeA: Figure 3d; conditioning effect F(1,28) = 50.287; p < 0.001; conditioning x treatment effect F(1,28) = 0.237; p = 0.630; BLA: Figure 3f; conditioning effect F(1,28) = 43.611; p < 0.001; conditioning x treatment effect F(1,28) = 0.113; p = 0.739). This indicates that NPY reduces the expression of social fear when administered into the CeA, but not when administered into the MeA or BLA.

2.4. NPY Exerts Anxiolytic-Like Effects When Administered into the DH, DLS, CeA and BLA, But Not When Administered into the MeA

We also investigated whether the reduced expression of social fear in NPY-treated SFC+ mice might be due to a reduced anxiety-like behavior. By decreasing anxiety-like behavior, NPY might enable SFC+ mice to approach the social stimuli faster and, thereby, to express less social fear. To investigate whether NPY alters anxiety-like behavior when administered into the DH, DLS, CeA, MeA and/or BLA, mice were administered either Veh (0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) into these brain regions 10 min before they were tested on the elevated plus-maze (EPM). NPY exerted anxiolytic-like effects when administered into the DH (Figure 4a; T(14) = −2.699; p = 0.017), DLS (Figure 4b; T(14) = −2.845; p = 0.013), CeA (Figure 4c; T(14) = −2.202; p = 0.045) and BLA (Figure 4e; T(14) = −2.325; p = 0.036), as indicated by the increased percentage of time spent on the open arms of the EPM in NPY-treated mice compared with Veh-treated mice. When administered into the MeA, however, NPY did not alter anxiety-like behavior (Figure 4d; T(14) = −0.527; p = 0.606). This indicates that NPY exerts anxiolytic-like effects when administered into the DH, DLS, CeA and BLA, but not when administered into the MeA, and suggests that partly distinct neural circuitries mediate the effects of NPY on the expression of social fear and on anxiety-like behavior.
NPY did not alter locomotor activity when administered into any of these brain regions, as indicated by the similar number of entries into the closed arms of the EPM between NPY-treated and Veh-treated mice (DH: T(14) = −0.396; p = 0.698; DLS: T(14) = −0.284; p = 0.780; CeA: T(14) = −0.715; p = 0.486; MeA: T(14) = −0.444; p = 0.664; BLA: T(14) = −0.402; p = 0.693). This suggests that the anxiolytic-like effects (i.e., the increased time spent on the open arms of the EPM) observed after NPY administration are not due to a general increase in locomotor activity.

3. Discussion

The present study demonstrates, for the first time, that NPY reduces the expression of SFC-induced social fear in a brain-region-dependent manner in male mice. In more detail, we could show that when administered into the DLS and CeA, NPY reduced the expression of social fear. In contrast, when administered into the DH, MeA and BLA, NPY did not affect the expression of social fear. We could also show that NPY exerted anxiolytic-like effects when administered into the DH, DLS, CeA and BLA, but not when administered into the MeA. These results suggest that distinct brain regions are recruited to mediate the effects of NPY on the expression of social fear and on anxiety-like behavior.
In previous studies, we have shown that i.c.v.-administered NPY reduces the expression of SFC-induced social fear [34,35]. Here, we extended these findings and localized these effects of NPY in the DLS and CeA. The amygdala is the central component of the fear circuitry and is involved in the perception, learning and expression of fear. Within the amygdala, the CeA constitutes the output relay for the functional consequences of amygdala activation by fearful stimuli, and together with the BLA, coordinates the behavioral and physiological correlates of fear expression [40,41]. Although Fendt et al. [29] have shown that NPY decreased the expression of conditioned fear when administered into the amygdala, a direct involvement of the CeA in the effects of NPY on conditioned fear was not reported previously. The BLA seems to mediate the fear-reducing effects of NPY in a stimulus-specific manner. As such, NPY inhibited the expression of cued fear when administered into the BLA [28], but did not reduce the expression of social fear when administered into the BLA (Figure 3f), suggesting that the BLA mediates the effects of NPY on cued (non-social) fear, but not on social fear. Alternatively, differences in the form of conditioning (e.g., classical conditioning for cued fear versus operant conditioning for social fear) might contribute to the differential role of BLA NPY on fear expression. The MeA was implicated to a lesser extent in fear processes and does not seem to mediate the effects of NPY on conditioned fear, as NPY did not affect the expression of cued fear [28] or the expression of social fear (Figure 3d) when administered into the MeA. The septum and the hippocampus are key components of the behavioral inhibition system regulating emotional behavior and cognitive functions, especially learning and memory, and the septo-hippocampal circuits are important for fear-related behaviors [42,43]. The hippocampus also processes information about the context, i.e., the environment of a fearful situation [43]. The DLS was previously shown to mediate the effects of another neuropeptide, oxytocin, on the expression of social fear [44]. This suggests that multiple neuropeptide systems, including oxytocin and NPY, particularly at the level of the DLS, are regulating the expression of social fear. The DH does not seem to be involved in the effects of NPY on the expression of social fear, which indicates that the processing of context-dependent information related to social fear might not be regulated by NPY.
Similar to its i.c.v. effects [34,35], NPY increased social investigation only in SFC+ mice, but not in SFC mice when administered into the DLS and CeA, suggesting that NPY increases social investigation only in individuals with low or impaired sociability. This resembles the effects of other neuropeptides, such as oxytocin or neuropeptide S, which were also shown to reduce social fear in SFC+ mice without further increasing social investigation in SFC mice [44,45].
We also investigated whether the reduced expression of social fear in NPY-treated SFC+ mice might be due to a reduced anxiety-like behavior. By decreasing anxiety-like behavior, NPY might enable SFC+ mice to approach the social stimuli faster and, thereby, to express less social fear. Even though we observed anxiolytic-like effects after the administration of NPY both in the DLS and in the CeA, confirming previous findings [17,19], these anxiolytic-like effects are unlikely to completely explain the effects of NPY on the expression of social fear, especially because similar anxiolytic-like effects occurred after administration of NPY into the DH [20] (Figure 4a) and BLA (Figure 4e), regions in which NPY did not reduce the expression of social fear. Interestingly, Sajdyk et al. [24,46] reported anxiolytic-like effects of NPY after administration into the DLS and BLA, but not into the CeA in the social interaction test, a validated test for assessing both anxiety-like behavior [47] and social behavior [48] in rodents. These partly different results might suggest that the neural circuitries mediating the effects of NPY on social behavior and on anxiety-like behavior are different to some extent.
Taken together, we have shown that NPY reduces the expression of SFC-induced social fear when administered into the DLS and CeA, but not when administered into the DH, MeA and BLA. We also showed that NPY exerted anxiolytic-like effects when administered into the DH, DLS, CeA and BLA, but not when administered into the MeA. These results suggest that partly distinct neural circuitries mediate the effects of NPY on the expression of social fear and on anxiety-like behavior.

4. Materials and Methods

4.1. Animals

CD1 mice (Charles River, Sulzfeld, Germany, 10 weeks of age) were individually housed for 1 week before experiments started and remained single-housed throughout the experiments. Mice were kept under standard laboratory conditions (12:12 light/dark cycle, lights on at 07:00 h, 22 °C, 60% humidity, food and water ad libitum). Experiments were performed during the light phase, between 09:00 and 14:00 h. All efforts were made to minimize animal suffering and to reduce the number of animals used.

4.2. Stereotaxic Cannula Implantation

Implantation of the guide cannula (23 G, 8 mm length; Injecta GmbH, Klingenthal, Germany) for bilateral infusions was performed under ketamine-xylazine anesthesia (intraperitoneal injection of 120 mg/kg Ketavet® and 16 mg/kg Rompun®, respectively) as previously described [44,49], 1 mm above the DH (from Bregma: AP − 2.0 mm, L ± 1.5 mm, D + 1.4 mm), DLS (AP + 0.3 mm, L ± 0.5 mm, D + 1.6 mm), CeA (AP − 1.2 mm, L ± 2.8 mm, D + 3.8 mm), MeA (AP − 1.1 mm, L ± 2.5 mm, D + 4.5 mm) or BLA (AP − 1.2 mm, L ± 3.1 mm, D + 3.8 mm). After surgery, mice were handled for 5 days before experiments started.

4.3. Intracerebral Infusions

Mice received bilateral DH, DLS, CeA, MeA or BLA infusions of either vehicle (Veh; distilled H2O; 0.2 µL/side) or porcine NPY (0.1 nmol/0.2 µL/side; PeptaNova, Sandhausen, Germany) via an infusion cannula (25 G, 9 mm length) inserted into the guide cannula and connected via polyethylene tubing to a Hamilton syringe. The infusion system was left in place for 30 s following the infusion to allow diffusion of the solution.
The correct infusion site was verified (Figure S1); accordingly, 1 DH, 1 DLS, 3 CeA, 2 MeA and 2 BLA cannulas were not implanted correctly, and these mice were excluded from the study. NPY dose and timing of administration were selected based on previous studies [26,49].

4.4. Social Fear Conditioning (SFC) Paradigm

To induce social fear, mice were conditioned during SFC and social investigation was assessed during social fear extinction as a read-out of social fear.
SFC. SFC was performed with a computerized fear conditioning system (TSE System GmbH, Bad Homburg, Germany) as previously described [34,35,44,45,50,51,52]; see [48] for a schematic representation of the SFC paradigm. Mice were placed in the conditioning chamber (45 × 22 × 40 cm) and, after a 30-s habituation period, an empty wire mesh cage (7 × 7 × 6 cm) was placed as a non-social stimulus near one of the short walls. After 3 min, the non-social stimulus was replaced by an identical cage containing an unfamiliar mouse. Unconditioned (SFC) mice were allowed to investigate this social stimulus for 3 min, whereas conditioned (SFC+) mice were given a 1-s mild electric foot shock (0.7 mA) each time they investigated, i.e., made direct contact with the social stimulus. Mice received between 1 and 4 foot shocks, with a variable inter-shock interval, depending on when direct social contact was made. The number of foot shocks was assessed as a measure of distress and social fear learning. Mice were returned to their home cage when no further social contact was made for 2 min (average duration of SFC approximately 10 min). All SFC+ mice investigated the social stimulus and could be conditioned. The time mice spent investigating the non-social stimulus, as a pre-conditioning measure of non-social anxiety, was analyzed.
Social fear extinction. One day after SFC, mice were exposed in their home cage to three non-social stimuli, i.e., empty cages identical to the cage used during SFC, to assess non-social investigation as a parameter of non-social fear. Mice were then exposed to six unfamiliar social stimuli, i.e., mice enclosed in wire mesh cages, to assess social investigation as a parameter of social fear. Each stimulus was placed near a short wall of the home cage and presented for 3 min, with a 3-min inter-exposure interval. The test was recorded and analyzed using JWatcher (V 1.0, Macquarie University, Sydney, Australia and UCLA, Los Angeles, CA, USA). The non-social investigation was defined as a direct sniffing of the empty cage, whereas social investigation was defined as a direct sniffing of the cage and/or of the social stimulus inside the cage.

4.5. Elevated Plus-Maze (EPM) Test

To investigate whether NPY alters anxiety-like behavior, mice were tested on the EPM as previously described [46,51]. An increased percentage of time spent on the open arms (150 lx) indicated reduced anxiety-like behavior. The number of entries into the closed arms (30 lx) during the 5-min testing period indicated locomotor activity.

4.6. Statistical Analysis

For statistical analysis, SPSS (Version 24, SPSS Inc., Chicago, IL, USA) was used. Data were analyzed by Student’s t-test, one-way ANOVA or three-way ANOVA for repeated measures, followed by a Bonferroni’s post-hoc analysis whenever appropriate. Statistical significance was set at p < 0.05.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijms22073695/s1.

Author Contributions

Conceptualization, I.Z.; methodology, I.Z.; validation, I.Z.; formal analysis, I.Z.; investigation, I.Z.; resources, J.K.; writing—original draft preparation, I.Z.; writing—review and editing, J.K. and I.Z.; visualization, I.Z.; project administration, I.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Experiments were conducted in accordance with the Guide for the Care and Use of Laboratory Animals of the Government of Unterfranken (approval code 55.2 DMS-2532-2-314, approval date 13.12.16) and the guidelines of the NIH.

Acknowledgments

We thank Gunther Moll for permission to use the SFC System.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stanley, B.G.; Leibowitz, S.F. Neuropeptide Y: Stimulation of feeding and drinking by injection into the paraventricular nucleus. Life Sci. 1984, 35, 2635–2642. [Google Scholar] [CrossRef]
  2. Colmers, W.F.; Bleakman, D. Effects of neuropeptide Y on the electrical properties of neurons. Trends Neurosci. 1994, 17, 373–379. [Google Scholar] [CrossRef]
  3. Vezzani, A.; Sperk, G.; Colmers, W.F. Neuropeptide Y: Emerging evidence for a functional role in seizure modulation. Trends Neurosci. 1999, 22, 25–30. [Google Scholar] [CrossRef]
  4. Michalkiewicz, M.; Michalkiewicz, T.; Kreulen, D.L.; McDougall, S.J. Increased blood pressure responses in neuropeptide Y transgenic rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2001, 281, 417–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Magni, P. Hormonal control of the neuropeptide Y system. Curr. Protein Pept. Sci. 2003, 4, 45–57. [Google Scholar] [CrossRef]
  6. Hökfelt, T.; Stanic, D.; Sanford, S.D.; Gatlin, J.C.; Nilsson, I.; Paratcha, G.; Ledda, F.; Fetissov, S.; Lindfors, C.; Herzog, H.; et al. NPY and its involvement in axon guidance, neurogenesis, and feeding. Nutrition 2008, 24, 860–868. [Google Scholar] [CrossRef] [PubMed]
  7. Dumont, Y.; Fournier, A.; St-Pierre, S.; Quirion, R. Comparative characterization and autoradiographic distribution of neuropeptide Y receptor subtypes in the rat brain. J. Neurosci. 1993, 13, 73–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Dumont, Y.; Fournier, A.; St-Pierre, S.; Quirion, R. Autoradiographic distribution of [125I]Leu31,Pro34]PYY and [125I]PYY3-36 binding sites in the rat brain evaluated with two newly developed Y1 and Y2 receptor radioligands. Synapse 1996, 22, 139–158. [Google Scholar] [CrossRef]
  9. Parker, R.M.; Herzog, H. Regional distribution of Y-receptor subtype mRNAs in rat brain. Eur. J. Neurosci. 1999, 11, 1431–1448. [Google Scholar] [CrossRef]
  10. Chang, R.S.; Lotti, V.J.; Chen, T.B.; Cerino, D.J.; Kling, P.J. Neuropeptide Y (NPY) binding sites in rat brain labeled with 125I-Bolton-Hunter NPY: Comparative potencies of various polypeptides on brain NPY binding and biological responses in the rat vas deferens. Life Sci. 1985, 37, 2111–2122. [Google Scholar] [CrossRef]
  11. De Quidt, M.E.; Emson, P.C. Distribution of neuropeptide Y-like immunoreactivity in the rat central nervous system-II. Immunohistochemical analysis. Neuroscience 1986, 18, 545–618. [Google Scholar] [CrossRef]
  12. Lynch, D.R.; Walker, M.W.; Miller, R.J.; Snyder, S.H. Neuropeptide Y receptor binding sites in rat brain: Differential autoradiographic localizations with 125I-peptide YY and 125I-neuropeptide Y imply receptor heterogeneity. J. Neurosci. 1989, 9, 2607–2619. [Google Scholar] [CrossRef] [Green Version]
  13. Bowers, M.E.; Choi, D.C.; Ressler, K.J. Neuropeptide regulation of fear and anxiety: Implications of cholecystokinin, endogenous opioids, and neuropeptide Y. Physiol. Behav. 2012, 107, 699–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Heilig, M. The NPY system in stress, anxiety and depression. Neuropeptides 2004, 38, 213–224. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, G.; Feder, A.; Wegener, G.; Bailey, C.; Saxena, S.; Charney, D.; Mathé, A.A. Central functions of neuropeptide Y in mood and anxiety disorders. Expert Opin. Ther. Targets 2011, 15, 1317–1331. [Google Scholar] [CrossRef] [PubMed]
  16. Heilig, M.; Soderpalm, B.; Engel, J.A.; Widerlov, E. Centrally administered neuropeptide Y (NPY) produces anxiolytic-like effects in animal anxiety models. Psychopharmacology 1989, 98, 524–529. [Google Scholar] [CrossRef] [PubMed]
  17. Heilig, M.; McLeod, S.; Brot, M.; Heinrichs, S.C.; Menzaghi, F.; Koob, G.F.; Britton, K.T. Anxiolytic-like action of neuropeptide Y: Mediation by Y1 receptors in amygdala, and dissociation from food intake effects. Neuropsychopharmacology 1993, 8, 357–363. [Google Scholar] [CrossRef]
  18. Kask, A.; Rägo, L.; Harro, J. Anxiolytic-like effect of neuropeptide Y (NPY) and NPY13-36 microinjected into vicinity of locus coeruleus in rats. Brain Res. 1998, 788, 345–348. [Google Scholar] [CrossRef]
  19. Trent, N.L.; Menard, J.L. Infusions of neuropeptide Y into the lateral septum reduce anxiety-related behaviors in the rat. Pharmacol. Biochem. Behav. 2011, 99, 580–590. [Google Scholar] [CrossRef]
  20. Smiałowska, M.; Wierońska, J.M.; Domin, H.; Zieba, B. The effect of intrahippocampal injection of group II and III metobotropic glutamate receptor agonists on anxiety; the role of neuropeptide Y. Neuropsychopharmacology 2007, 32, 1242–1250. [Google Scholar] [CrossRef]
  21. Sharko, A.C.; Kaigler, K.F.; Fadel, J.R.; Wilson, M.A. Ethanol-induced anxiolysis and neuronal activation in the amygdala and bed nucleus of the stria terminalis. Alcohol 2016, 50, 19–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Thorsell, A.; Repunte-Canonigo, V.; O’Dell, L.E.; Chen, S.A.; King, A.R.; Lekic, D.; Koob, G.F.; Sanna, P.P. Viral vector-induced amygdala NPY overexpression reverses increased alcohol intake caused by repeated deprivations in Wistar rats. Brain 2007, 130, 1330–1337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kask, A.; Nguyen, H.P.; Pabst, R.; Von Hörsten, S. Neuropeptide Y Y1 receptor-mediated anxiolysis in the dorsocaudal lateral septum: Functional antagonism of corticotropin-releasing hormone-induced anxiety. Neuroscience 2001, 104, 799–806. [Google Scholar]
  24. Sajdyk, T.J.; Vandergriff, M.G.; Gehlert, D.R. Amygdalar neuropeptide Y Y1 receptors mediate the anxiolytic-like actions of neuropeptide Y in the social interaction test. Eur. J. Pharmacol. 1999, 368, 143–147. [Google Scholar] [CrossRef]
  25. Broqua, P.; Wettstein, J.G.; Rocher, M.N.; Gauthier-Martin, B.; Junien, J.L. Behavioral effects of neuropeptide Y receptor agonists in the elevated plus-maze and fear-potentiated startle procedures. Behav. Pharmacol. 1995, 6, 215–222. [Google Scholar] [CrossRef] [PubMed]
  26. Karlsson, R.M.; Holmes, A.; Heilig, M.; Crawley, J.N. Anxiolytic-like actions of centrally-administered neuropeptide Y, but not galanin, in C57BL/6J mice. Pharmacol. Biochem. Behav. 2005, 80, 427–436. [Google Scholar] [CrossRef]
  27. Lach, G.; de Lima, T.C. Role of NPY Y1 receptor on acquisition, consolidation and extinction on contextual fear conditioning: Dissociation between anxiety, locomotion and non-emotional memory behavior. Neurobiol. Learn. Mem. 2013, 103, 26–33. [Google Scholar] [CrossRef]
  28. Gutman, A.R.; Yang, Y.; Ressler, K.J.; Davis, M. The role of neuropeptide Y in the expression and extinction of fear-potentiated startle. J. Neurosci. 2008, 28, 12682–12690. [Google Scholar] [CrossRef] [Green Version]
  29. Fendt, M.; Bürki, H.; Imobersteg, S.; Lingenhöhl, K.; McAllister, K.H.; Orain, D.; Uzunov, D.P.; Chaperon, F. Fear-reducing effects of intra-amygdala neuropeptide Y infusion in animal models of conditioned fear: An NPY Y1 receptor independent effect. Psychopharmacology 2009, 206, 291–301. [Google Scholar] [CrossRef]
  30. Pickens, C.L.; Adams-Deutsch, T.; Nair, S.G.; Navarre, B.M.; Heilig, M.; Shaham, Y. Effect of pharmacological manipulations of neuropeptide Y and corticotropin-releasing factor neurotransmission on incubation of conditioned fear. Neuroscience 2009, 164, 1398–1406. [Google Scholar] [CrossRef] [Green Version]
  31. Verma, D.; Tasan, R.O.; Herzog, H.; Sperk, G. NPY controls fear conditioning and fear extinction by combined action on Y1 and Y2 receptors. Br. J. Pharmacol. 2012, 166, 1461–1473. [Google Scholar] [CrossRef] [Green Version]
  32. Verma, D.; Wood, J.; Lach, G.; Mietzsch, M.; Weger, S.; Heilbronn, R.; Herzog, H.; Bonaventure, P.; Sperk, G.; Tasan, R.O. NPY Y2 receptors in the central amygdala reduce cued but not contextual fear. Neuropharmacology 2015, 99, 665–674. [Google Scholar] [CrossRef] [Green Version]
  33. Verma, D.; Hörmer, B.; Bellmann-Sickert, K.; Thieme, V.; Beck-Sickinger, A.G.; Herzog, H.; Sperk, G.; Tasan, R.O. Pancreatic polypeptide and its central Y4 receptors are essential for cued fear extinction and permanent suppression of fear. Br. J. Pharmacol. 2016, 173, 1925–1938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kornhuber, J.; Zoicas, I. Neuropeptide Y reduces expression of social fear via simultaneous activation of Y1 and Y2 receptors. J. Psychopharmacol. 2019, 33, 1533–1539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kornhuber, J.; Zoicas, I. Neuropeptide Y as alternative pharmacotherapy for antidepressant-resistant social fear. Int. J. Mol. Sci. 2020, 21, 8220. [Google Scholar] [CrossRef] [PubMed]
  36. Kask, A.; Harro, J.; von Hörsten, S.; Redrobe, J.P.; Dumont, Y.; Quirion, R. The neurocircuitry and receptor subtypes mediating anxiolytic-like effects of neuropeptide Y. Neurosci. Biobehav. Rev. 2002, 26, 259–283. [Google Scholar] [CrossRef]
  37. LeDoux, J.E.; Muller, J. Emotional memory and psychopathology. Philos. Trans. R Soc. Lond. B Biol. Sci. 1997, 352, 1719–1726. [Google Scholar]
  38. Reichmann, F.; Holzer, P. Neuropeptide Y: A stressful review. Neuropeptides 2016, 55, 99–109. [Google Scholar] [CrossRef] [Green Version]
  39. Tasan, R.O.; Verma, D.; Wood, J.; Lach, G.; Hörmer, B.; de Lima, T.C.; Herzog, H.; Sperk, G. The role of Neuropeptide Y in fear conditioning and extinction. Neuropeptides 2016, 55, 111–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Fanselow, M.S.; LeDoux, J.E. Why we think plasticity underlying Pavlovian fear conditioning occurs in the basolateral amygdala. Neuron 1999, 23, 229–232. [Google Scholar] [CrossRef] [Green Version]
  41. LeDoux, J.E. The amygdala. Curr. Biol. 2007, 17, R868–R874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Corcoran, K.A.; Maren, S. Hippocampal inactivation disrupts contextual retrieval of fear memory after extinction. J. Neurosci. 2001, 21, 1720–1726. [Google Scholar] [CrossRef] [PubMed]
  43. Maren, S.; Phan, K.L.; Liberzon, I. The contextual brain: Implications for fear conditioning, extinction and psychopathology. Nat. Rev. Neurosci. 2013, 14, 417–428. [Google Scholar] [CrossRef] [PubMed]
  44. Zoicas, I.; Slattery, D.A.; Neumann, I.D. Brain oxytocin in social fear conditioning and its extinction: Involvement of the lateral septum. Neuropsychopharmacology 2014, 39, 3027–3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zoicas, I.; Menon, R.; Neumann, I.D. Neuropeptide S reduces fear and avoidance of con-specifics induced by social fear conditioning and social defeat, respectively. Neuropharmacology 2016, 108, 284–291. [Google Scholar] [CrossRef] [PubMed]
  46. Sajdyk, T.J.; Schober, D.A.; Smiley, D.L.; Gehlert, D.R. Neuropeptide Y-Y2 receptors mediate anxiety in the amygdala. Pharmacol. Biochem. Behav. 2002, 71, 419–423. [Google Scholar] [CrossRef]
  47. File, S.E.; Hyde, J.R. Can social interaction be used to measure anxiety? Br. J. Pharmacol. 1978, 62, 19–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Toth, I.; Neumann, I.D. Animal models of social avoidance and social fear. Cell Tissue Res. 2013, 354, 107–118. [Google Scholar] [CrossRef]
  49. Kornhuber, J.; Zoicas, I. Neuropeptide Y prolongs non-social memory in a brain region- and receptor-specific way in male mice. Neuropharmacology 2020, 175, 108199. [Google Scholar] [CrossRef] [PubMed]
  50. Kornhuber, J.; Zoicas, I. Social fear memory requires two stages of protein synthesis in mice. Int. J. Mol. Sci. 2020, 21, 5537. [Google Scholar] [CrossRef]
  51. Toth, I.; Neumann, I.D.; Slattery, D.A. Social fear conditioning: A novel and specific animal model to study social anxiety disorder. Neuropsychopharmacology 2012, 37, 1433–1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Toth, I.; Neumann, I.D.; Slattery, D.A. Social fear conditioning as an animal model of social anxiety disorder. Curr. Protoc. Neurosci. 2013, 63, 9–42. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Neuropeptide Y (NPY) does not affect the expression of social fear when administered into the dorsal hippocampus (DH). (a) Pre-conditioned investigation of the non-social stimulus (empty cage) during social fear conditioning (SFC) on day 1. (b) Investigation of the non-social (ns1–ns3) and social (cages with mice; s1–s6) stimuli during social fear extinction on day 2. Unconditioned (SFC) and conditioned (SFC+) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before social fear extinction. Data represent means ± SEM and numbers in parentheses indicate group sizes. p < 0.05 * vs. respective SFC controls.
Figure 1. Neuropeptide Y (NPY) does not affect the expression of social fear when administered into the dorsal hippocampus (DH). (a) Pre-conditioned investigation of the non-social stimulus (empty cage) during social fear conditioning (SFC) on day 1. (b) Investigation of the non-social (ns1–ns3) and social (cages with mice; s1–s6) stimuli during social fear extinction on day 2. Unconditioned (SFC) and conditioned (SFC+) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before social fear extinction. Data represent means ± SEM and numbers in parentheses indicate group sizes. p < 0.05 * vs. respective SFC controls.
Ijms 22 03695 g001
Figure 2. NPY reduces the expression of social fear when administered into the dorsolateral septum (DLS). (a) Pre-conditioned investigation of the non-social stimulus (empty cage) during SFC on day 1. (b) Investigation of the non-social (ns1–ns3) and social (cages with mice; s1–s6) stimuli during social fear extinction on day 2. Unconditioned (SFC) and conditioned (SFC+) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before social fear extinction. Data represent means ± SEM and numbers in parentheses indicate group sizes. p < 0.05 * vs. all groups.
Figure 2. NPY reduces the expression of social fear when administered into the dorsolateral septum (DLS). (a) Pre-conditioned investigation of the non-social stimulus (empty cage) during SFC on day 1. (b) Investigation of the non-social (ns1–ns3) and social (cages with mice; s1–s6) stimuli during social fear extinction on day 2. Unconditioned (SFC) and conditioned (SFC+) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before social fear extinction. Data represent means ± SEM and numbers in parentheses indicate group sizes. p < 0.05 * vs. all groups.
Ijms 22 03695 g002
Figure 3. NPY reduces the expression of social fear when administered into the central amygdala (CeA), but not when administered into the medial (MeA) or basolateral amygdala (BLA). (a,c,e) Pre-conditioning investigation of the non-social stimulus (empty cage) during SFC on day 1. (b,d,f) Investigation of the non-social (ns1–ns3) and social (cages with mice; s1–s6) stimuli during social fear extinction on day 2. Unconditioned (SFC) and conditioned (SFC+) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before social fear extinction. Data represent means ± SEM and numbers in parentheses indicate group sizes. p < 0.05 * vs. all groups; # vs. respective SFC controls.
Figure 3. NPY reduces the expression of social fear when administered into the central amygdala (CeA), but not when administered into the medial (MeA) or basolateral amygdala (BLA). (a,c,e) Pre-conditioning investigation of the non-social stimulus (empty cage) during SFC on day 1. (b,d,f) Investigation of the non-social (ns1–ns3) and social (cages with mice; s1–s6) stimuli during social fear extinction on day 2. Unconditioned (SFC) and conditioned (SFC+) mice were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before social fear extinction. Data represent means ± SEM and numbers in parentheses indicate group sizes. p < 0.05 * vs. all groups; # vs. respective SFC controls.
Ijms 22 03695 g003
Figure 4. NPY exerts anxiolytic-like effects when administered into the dorsal hippocampus (a), dorsolateral septum (b), central amygdala (c) and basolateral amygdala (e), but not when administered into the medial amygdala (d). Mice (n = 8 per group for each brain region) were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before being tested on the elevated plus-maze. Data represent means + SEM. p < 0.05 * vs. Veh-treated mice.
Figure 4. NPY exerts anxiolytic-like effects when administered into the dorsal hippocampus (a), dorsolateral septum (b), central amygdala (c) and basolateral amygdala (e), but not when administered into the medial amygdala (d). Mice (n = 8 per group for each brain region) were administered either vehicle (Veh; 0.2 µL/side) or NPY (0.1 nmol/0.2 µL/side) 10 min before being tested on the elevated plus-maze. Data represent means + SEM. p < 0.05 * vs. Veh-treated mice.
Ijms 22 03695 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kornhuber, J.; Zoicas, I. Brain Region-Dependent Effects of Neuropeptide Y on Conditioned Social Fear and Anxiety-Like Behavior in Male Mice. Int. J. Mol. Sci. 2021, 22, 3695. https://doi.org/10.3390/ijms22073695

AMA Style

Kornhuber J, Zoicas I. Brain Region-Dependent Effects of Neuropeptide Y on Conditioned Social Fear and Anxiety-Like Behavior in Male Mice. International Journal of Molecular Sciences. 2021; 22(7):3695. https://doi.org/10.3390/ijms22073695

Chicago/Turabian Style

Kornhuber, Johannes, and Iulia Zoicas. 2021. "Brain Region-Dependent Effects of Neuropeptide Y on Conditioned Social Fear and Anxiety-Like Behavior in Male Mice" International Journal of Molecular Sciences 22, no. 7: 3695. https://doi.org/10.3390/ijms22073695

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop