Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Anxiolytic-Like Effects of Antisauvagine-30 in Mice Are Not Mediated by CRF2 Receptors

  • Eric P. Zorrilla ,

    ezorrilla@scripps.edu (EPZ); gkoob@scripps.edu (GFK)

    Affiliations Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California, United States of America, Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America

  • Amanda J. Roberts,

    Affiliation Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, United States of America

  • Jean E. Rivier,

    Affiliation The Clayton Foundation Laboratories for Peptide Biology and Structural Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America

  • George F. Koob

    ezorrilla@scripps.edu (EPZ); gkoob@scripps.edu (GFK)

    Affiliation Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California, United States of America

Abstract

The role of brain corticotropin-releasing factor type 2 (CRF2) receptors in behavioral stress responses remains controversial. Conflicting findings suggest pro-stress, anti-stress or no effects of impeding CRF2 signaling. Previous studies have used antisauvagine-30 as a selective CRF2 antagonist. The present study tested the hypotheses that 1) potential anxiolytic-like actions of intracerebroventricular (i.c.v.) administration of antisauvagine-30 also are present in mice lacking CRF2 receptors and 2) potential anxiolytic-like effects of antisauvagine-30 are not shared by the more selective CRF2 antagonist astressin2-B. Cannulated, male CRF2 receptor knockout (n = 22) and wildtype littermate mice (n = 21) backcrossed onto a C57BL/6J genetic background were tested in the marble burying, elevated plus-maze, and shock-induced freezing tests following pretreatment (i.c.v.) with vehicle, antisauvagine-30 or astressin2-B. Antisauvagine-30 reduced shock-induced freezing equally in wildtype and CRF2 knockout mice. In contrast, neither astressin2-B nor CRF2 genotype influenced shock-induced freezing. Neither CRF antagonist nor CRF2 genotype influenced anxiety-like behavior in the plus-maze or marble burying tests. A literature review showed that the typical antisauvagine-30 concentration infused in previous intracranial studies (∼1 mM) was 3 orders greater than its IC50 to block CRF1-mediated cAMP responses and 4 orders greater than its binding constants (Kd, Ki) for CRF1 receptors. Thus, increasing, previously used doses of antisauvagine-30 also exert non-CRF2-mediated effects, perhaps via CRF1. The results do not support the hypothesis that brain CRF2 receptors tonically promote anxiogenic-like behavior. Utilization of CRF2 antagonists, such as astressin2-B, at doses that are more subtype-selective, can better clarify the significance of brain CRF2 systems in stress-related behavior.

Introduction

In mammals, the stress-related peptide corticotropin-releasing factor (CRF) and its paralogs urocortins 1, 2, and 3 (Ucn 1, Ucn 2, Ucn 3), activate two CRF receptor subtypes, CRF1 and CRF2, to varying degrees [1]. CRF1 receptors mediate endocrine, behavioral, and autonomic responses to stress, which has spurred the development of drug-like CRF1 antagonists [2]. In contrast, the role of brain CRF2 receptors in stress responses remains controversial. Studies have implicated anti-stress-like actions, pro-stress-like actions, or a lack of involvement of CRF2 receptors [1]. Part of this uncertainty may reflect that, unlike the case with CRF1 antagonists [2], highly selective (>10,000-fold selectivity), small molecule CRF2 antagonists remain unavailable. Researchers have instead used truncated CRF2-preferring (100–1000-fold selectivity) peptide fragments as CRF2 antagonists, principally [D-Phe11,His12]sauvagine(11–40)NH2 (antisauvagine-30; [3] and cyclo(31–34)[D-Phe11,His12,CαMeLeu13,39,Nle17,Glu31,Lys34]Ac-sauvagine(8–40) (astressin2-B; [4]).

Antisauvagine-30 has been described as a selective CRF2 antagonist in the literature (1530 hits in Google Scholar as of August 2012). Antisauvagine-30 potently displaces radioiodinated CRF-related ligands from HEK293 cell membranes expressing recombinant mCRF2b (Kd = 1.4 nM; [3], hCRF2a (Ki = 0.8 nM; [5], or mCRF2b receptors (Ki = 0.41 nM; [6] and has lower affinity for HEK293 membranes expressing CRF1 receptors. Several findings suggest, however, that antisauvagine-30 may block CRF1 receptors at doses that have been used in vivo. First, antisauvagine-30 can displace [125I]-oCRF from HEK293-rCRF1 membranes (Ki = 154–166 nM; [3], [6] and [125I]-sauvagine from HEK293-hCRF1 membranes (Ki = 100 nM; [7]). Similarly, antisauvagine-30 competes with [125I]-astressin to bind rat and human uncoupled CRF1 receptors (Ki = 66 and 170 nM; [7], [8]. Yet, many intracerebroventricular and intracerebral studies have infused antisauvagine-30 at ∼4 orders greater concentrations (1–2 mM) (e.g., see Table 1). Moreover, in its original characterization, antisauvagine-30 showed ∼30% of the rCRF1 antagonist potency of astressin [3], a potent CRF1 antagonist. Accordingly, antisauvagine-30 blocks oCRF-induced cAMP accumulation in HEK293-rCRF1 cells [9] and oCRF-induced cAMP responses in human retinoblastoma Y79 cells [10] with IC50s = 1–2 µM, concentrations 3 orders lower than those that have been injected. The incomplete selectivity of antisauvagine-30 raises concern that some putative anxiolytic/anti-stress-like actions of antisauvagine-30 previously attributed to antagonism of brain CRF2 receptors may involve a non-CRF2 target, such as CRF1 receptors.

thumbnail
Table 1. Intracerebroventricular (ICV) studies of antisauvagine-30 effects on stress- or anxiety-related endpoints.

https://doi.org/10.1371/journal.pone.0063942.t001

Many antibodies [11] and antagonists [12] were subsequently found to have off-target binding or activity when evaluated in knockout (KO) mice. Here, we tested the hypotheses that any potential anxiolytic-like actions of antisauvagine-30 would 1) be present in mice lacking functional CRF2 receptors, and 2) not be shared by the more selective CRF2 antagonist astressin2-B. Astressin2-B binds to CRF2 receptors in vitro with similar potency as does anti-sauvagine-30 (e.g., displacement of [125I]sauvagine from CHO-hCRF2a membranes (Ki = 0.49 vs 0.29 nM), from intrinsic rCRF2b in A7r5 cells (Ki = 0.17 vs 0.77 nM), and from CRF2a in rat olfactory bulb (Ki = 0.50 vs 0.84 nM) [8]. But, astressin2-B shows one order less affinity for CRF1 receptors (Ki>1000 nM and 890 nM, respectively) [7], [8] than does antisavuagine-30 (Ki = 100 nM) [3], [6], [7], [8].

A secondary goal of the present study was to evaluate the anxiety-related phenotype of CRF2 KO mice backcrossed to C57BL/6J background. Previous studies that reported an anxiogenic-like phenotype of CRF2 knockout mice were performed on a hybrid 129SvJ-C57BL/6J genetic background [13], [14]. However, mixed genetic background transgenic mice can lead to spurious or inconsistent results due to the confounding (due to genetic linkage) and interactive influence of mixed genetic background on observed phenotypes [15]. The CRF2 null mutation was introduced into embryonic stem cells of the 129Sv genetic background. Due to genetic linkage, CRF2 null mutant mice studied on a hybrid background will overrepresent the 129Sv genetic background as compared to wildtype mice, which will show comparatively more C57BL/6 background [15]. Anxiogenic-like behavior is greater in 129Sv strain mice than in C57BL/6 mice, however [16], [17], [18], [19]. As a result, it is not clear whether the previously reported anxiogenic-like CRF2 KO phenotype is actually due to the null mutation as opposed to linked 129Sv genetic material. Potentially consistent with the latter possibility, no anxiogenic-like phenotype in elevated plus-maze or open field behavior was seen in CRF2 KO mice backcrossed 3 generations (∼87.5%) to a C57BL/6J background (Coste et al., 2000). Therefore, we here revisit the anxiety-related phenotype of CRF2 KO mice that were previously reported to show anxiogenic-like behavior on a hybrid background [13], but now studied after being backcrossed extensively (>99.975%) onto a C57BL/6J background.

Materials and Methods

Ethics Statement

Procedures adhered to the National Institutes of Health Guide for the Care and Use of Laboratory Animals (NIH publication no. 85–23, 1996) and Principles of Laboratory Animal Care and were approved by the Institutional Animal Care and Use Committee of The Scripps Research Institute (protocol #08-0010). All surgery was performed under isoflurane anesthesia, and all efforts were made to minimize suffering.

Subjects

Subjects were adult (26.5–32.3 g at study onset), male CRF2 receptor KO (n = 22; Crhr2tm1Klee/Crhr2tm1Klee; [13] and wildtype littermate mice (n = 21; WT, ≥12 generations C57BL/6J backcrossing; ≥99.9755869% consomy) offspring of heterozygote breeding. Mice were group-housed under a reverse 12 h/12 h light/dark cycle in a humidity- (60%) and temperature-controlled (22°C) vivarium with chow (LM-485 Diet 7012, Harlan, Madison, WI) and water available ad libitum.

Surgery

Anaesthetized (isoflurane, 1–3%) mice were stereotaxically (David Kopf, Tujunga, CA) implanted with a 27-gauge, 7.5 mm stainless steel guide cannula 1 mm above the lateral ventricle. Coordinates (in mm) were (anterior/posterior: −0.1, medial/lateral: ±1.0 from bregma, dorsal/ventral: −1.5 from skull; [20]. A 30-gauge obturator maintained patency. Mice recovered ≥7 days before testing. Cannula placement was inferred from successful gravity injection and from ventricular spread of injected dye in randomly tested mice.

Drugs and injection

Antisauvagine-30 and astressin2-B were synthesized using solid-phase methodology, purified using HPLC and characterized using capillary zone electrophoresis, HPLC and MS [4]. Peptides were dissolved in 0.5× PBS before testing and kept on ice. For intracerebroventricular (i.c.v.) infusions, the 30-gauge injector extended 1 mm beyond the cannula and was attached to tubing (0.01 i.d., 0.03 o.d. inches) from which 2 µl solution was delivered into the ventricle by gravity over 30 sec. The injector was left in place for 60 sec. The pretreatment intervals, during which the mouse was returned to its home cage were 15 min for the marble burying test and 30 min for the plus-maze and shock-induced freezing tests.

Study design

Mice were tested during the dark phase in the marble burying, elevated plus-maze, and shock-induced freezing tests using a between-subjects design for treatment. The same set of mice were subjects in the 3 tests. Experiments involved a 2 (Genotype: WT vs. KO)×3 (Antagonist: vehicle vs. antisauvagine-30 vs. astressin2-B) factorial design. The dose of antisauvagine-30 (i.c.v. ∼3 nmol, or 10.7 µg) was representative of doses used in previous studies of stress- or anxiety-related endpoints (Table 1). Astressin2-B was administered at the same dose. Tests were spaced by one week, and mice received a given drug treatment no more than twice across the three tests.

Marble burying

For marble burying testing [21], mice were individually placed in a polycarbonate cage (29×18×12 cm) containing 20 marbles (1.5 cm diameter) evenly spaced on 5-cm deep bedding. Marbles covered at least two-thirds by bedding, an index of anxiogenic-like behavior, were counted 30 min later.

Elevated plus-maze

The plus-maze apparatus has four arms (5×30 cm) at right angles to each other, elevated 30 cm from the floor. Two arms have 16-cm black plastic walls (closed arms), and two arms have 16-cm clear plastic walls (more open arms). Controls tested in this modified apparatus spend 35–40% of their time on the open arms, allowing changes to be detected bidirectionally; mice tested in the original plus-maze (open arms with no wall) typically spend 10–15% of their time on the open arms, making it difficult to detect anxiogenic-like effects. Mice were placed on the center of the maze, and behavior was videorecorded for 5 min. Decreases in % open arm time, calculated as: 100*open arm time/(open arm time+closed arm time) [22], indicate increased anxiety-like behavior. More total arm entries indicate increased locomotor activity [22].

Shock-induced freezing

Mice were placed in a Mouse NIR Video Fear Conditioning System (Med Associates, St. Albans, VT) housed in a soundproofed box, allowed to habituate for 2 min and then exposed to three 1.5 mA, 1-sec footshocks, separated by 20 sec. Freezing, a CRF/CRF1-dependent defensive response [23], was measured automatically from real-time video recordings (30 frames per second) across 15 min using Video Fear Conditioning Software (Med Associates) that distinguishes between subtle movements, such as whisker twitches, tail flicks and freezing behavior.

Statistics

Analysis of variance (ANOVA) was used to evaluate effects of Genotype, Antagonist and their interaction. Fisher's protected least significant difference tests identified pairwise differences. The software used was Systat 12.0 (SPSS, Chicago, IL).

Results

Figure 1 shows that antisauvagine-30 reduced the duration of shock-induced freezing in both WT and CRF2 KO mice (Antagonist: F2,37 = 4.17, p<0.05). Antisauvagine-30-treated mice froze less than mice pretreated with either vehicle or astressin2-B (ps<0.05), which did not differ from one another (p = 0.96). No Genotype (F1,37 = 0.03, p>0.85) or Genotype×Antagonist effects (F2,37 = 0.39, p>0.68) were seen.

thumbnail
Figure 1. Effects of antisauvagine-30, astressin2-B and CRF2 genotype on shock-induced freezing.

The data are expressed as M ± SEM. Antisauvagine-30 (i.c.v., 3 nmol) significantly and equally reduced the duration of shock-induced freezing in both wildtype and CRF2 knockout mice. In contrast, the same dose of astressin2B, a selective CRF2 antagonist, and CRF2 null genotype did not alter shock-induced freezing (n = 6–9/group). *p<0.05, differs from vehicle and astressin2-B-treated mice (Fisher's protected least significant difference test).

https://doi.org/10.1371/journal.pone.0063942.g001

Table 2 shows that there were no significant Genotype, Antagonist or Genotype×Antagonist effects on raw open arm time (F1,37 = 1.07, F2,37 = 1.22, F2,37 = 2.41, all ps>0.1), % open arm time calculated as a function of total arm time (F1,37 = 1.12, F2,37 = 1.16, F2,37 = 2.13, all ps>0.1), or the total number of arm entries in the elevated plus-maze (F1,37 = 0.42, F2,37 = 1.54, F2,37 = 0.15, all ps>0.2). There also were no significant Genotype (F1,37 = 0.35, p>0.55), Antagonist (F2,37 = 0.12, ps>0.89) or Genotype×Antagonist (F2,37 = 1.52, p>0.23) effects on the number of marbles buried in the marble burying test. A priori analysis in vehicle-treated mice considered separately also indicated no significant Genotype effect on shock-induced freezing (p>0.15); plus maze measures of % open arm time (p>0.15), open arm time (p>0.14), or total arm entries (p>0.72); or marbles buried (p>0.24).

thumbnail
Table 2. Effects of genotype and CRF antagonist on behavior in the elevated plus-maze and marble burying tests.

https://doi.org/10.1371/journal.pone.0063942.t002

Table 3 lists published studies in which antisauvagine-30 was administered site-specifically to discrete brain regions as a CRF2 antagonist. As can be seen, the concentrations that have been infused locally range from 137–2000 µM, on the order of those given i.c.v. previously (Table 1) and in the present study. The median concentration infused, 1050 µM is ∼3 orders greater than the reviewed IC50 of antisavuagine-30 to block CRF1-mediated cAMP responses (∼1–2 µM) and ∼4 orders greater than reviewed binding constants (Kd, Ki∼0.066–0.166 µM) of antisauvagine-30 for CRF1 receptors.

thumbnail
Table 3. Intracerebral (IC) site-specific studies of antisauvagine-30 effects on stress- or anxiety-related endpoints.

https://doi.org/10.1371/journal.pone.0063942.t003

Discussion

The present study found that i.c.v. infusion of a dose of antisauvagine-30 intermediate to those used in the literature reduced shock-induced freezing in both wild-type and CRF2 KO mice, unlike the CRF2 antagonist astressin2-B, which did not mitigate shock-induced freezing in either genotype. The present study also found that neither CRF2 KO nor i.c.v. astressin2-B infusion produced anxiolytic-like effects in 3 tests of anxiety-like behavior. Altogether, the results indicate that increasing doses of antisauvagine-30 lose their specificity and can exert non-CRF2-mediated effects at doses previously used. The collective results do not support the hypothesis that activation of brain CRF2 receptors tonically promotes anxiogenic-like behavior.

Antagonism of CRF1 receptors is a plausible mechanism for the non-CRF2 mediated anxiolytic-like actions of antisauvagine-30 seen here on shock-induced freezing. The low-moderate CRF1 binding affinities (∼100 nM) of antisauvagine-30 are not shared by the other widely used CRF2 antagonist, astressin2-B (Ki>1000 nM and 890 nM, respectively; [7], [8], which is similarly potent to antisauvagine-30 at binding CRF2 receptors (e.g., displacement of [125I]sauvagine from CHO-hCRF2a membranes [Ki = 0.49 vs. 0.29 nM], from intrinsic rCRF2b in A7r5 cells [Ki = 0.17 vs. 0.77 nM], and from CRF2a in rat olfactory bulb [Ki = 0.50 vs. 0.84 nM]; [8]. Accordingly, the i.c.v. dose of astressin2-B used here, which can block anorexia induced by urocortin 3, a selective CRF2 agonist [24], did not reduce shock-induced freezing. The results suggest that astressin2-B is more CRF2-selective than antisauvagine-30.

Many previous studies using antisauvagine-30 have interpreted that its effects were not CRF1 mediated because central administration of small molecule, selective CRF1 antagonists did not produce the same effects. Unfortunately, these comparisons have involved excessively lipophilic CRF1 antagonists, such as NBI27914, CP-154,526, or antalarmin, which are water insoluble, precipitate upon central administration and may therefore not diffuse to target sites or be available for pharmacological activity. Better controls would involve less hydrophobic, recently developed CRF1 antagonists more suitable for intracerebral administration, such as NBI-35965, GW-876008, pexacerfont or BMS-561,388.

Neither CRF2 KO nor selective CRF2 antagonism via astressin2-B altered behavior in three anxiety models, suggesting that CRF2 signaling is not a key modulator of anxiety-like behavior under basal conditions. Two previous studies that reported a basal anxiogenic-like phenotype of CRF2 knockout mice were performed on a hybrid 129SvJ-C57BL/6J genetic background [13], [14]. In contrast, similar to the present results in mice fully backcrossed onto a C57BL/6J background, no significant anxiety-like phenotype was seen in CRF2 knockout mice backcrossed 3 generations toward a C57BL/6J background [25]. Thus, because 129Sv and C57BL/6J mice differ in anxiety-like behavior [16], [17], [18], [19], genetic background may have interacted with the effect of CRF2 null mutation on behavioral measures in previous studies [15]. However, these results should not be prematurely concluded to mean that CRF2 receptors do not modulate anxiety-like behavior. Consistent with an anxiolytic-like action of CRF2 activation, i.c.v. administration of type 2 urocortins, selective CRF2 agonists, can produce anxiolytic-like and anti-stress-like behavioral effects [26], [27], [28], [29], [30], [31], [32], [33], [34]. Perhaps CRF2 receptors are normally quiescent under basal conditions, but are recruited in compensatory opposition to high or more sustained stress, as brought out following stressors or the anxiogenic-like 129Sv genetic background. Consistent with this hypothesis, CRF2 KO mice previously showed an anxiogenic-like phenotype in the light-dark box test following 30-min immobilization stress, but not under basal conditions (see Fig. 6A in [35]). Under this view, the stressful aspects of the 3 tests used in the present study (novelty, brief shock) may have been too brief in duration (<5 min), mild in magnitude, or initiated too soon before the behavioral assessment to allow a putative compensatory CRF2 response to be observed. Finally, it cannot be ruled out that a larger sample size might have led to a statistically significant p-value. For example, a trend for an anxiogenic-like effect of CRF2 null mutation, as reported previously [13], [14], was present in vehicle-treated subjects of the elevated plus-maze that, if considered separately, would have attained significance with a sample size of 16/group (standardized Cohen's d = −0.73).

While antisauvagine-30 exerted non-CRF2 actions at the tested dose, this does not mean that it is intrinsically non-selective. Lower in vivo doses or concentrations might be shown via a KO control study to be adequately selective for functional studies. Indeed, the finding that a low central dose of antisauvagine-30 (i.c.v., 400 ng) previously produced an anxiogenic-like effect, opposite to those seen with increasing doses of the antagonist (see Table 1), is consistent with the interpretation that antisauvagine-30 may lose specificity with increasing doses. The present result with a 3 nmol dose of antisauvagine-30 suggests that many (if not most) previous intracranial administration studies used a dose that can exert non-CRF2 mediated effects, complicating their interpretation (Table 1). Utilization of CRF2 antagonists at doses validated to be subtype-selective in knockout mice can help further clarify the biological significance of brain CRF2 systems in stress-related behavior.

Acknowledgments

We thank Lindsay Cates, Lindsey Casal, and Chelsea Cates-Gatto for expert technical assistance, Mary Gichuhi for administrative assistance and Mike Arends for editorial assistance.

Author Contributions

Conceived and designed the experiments: EPZ GFK AJR. Performed the experiments: AJR. Analyzed the data: EPZ GFK AJR. Contributed reagents/materials/analysis tools: JER AJR. Wrote the paper: EPZ GFK AJR JER.

References

  1. 1. Fekete EM, Zorrilla EP (2007) Physiology, pharmacology, and therapeutic relevance of urocortins in mammals: ancient CRF paralogs. Front Neuroendocrinol 28: 1–27.
  2. 2. Zorrilla EP, Koob GF (2010) Progress in corticotropin-releasing factor-1 antagonist development. Drug Discov Today 15: 371–383.
  3. 3. Ruhmann A, Bonk I, Lin CR, Rosenfeld MG, Spiess J (1998) Structural requirements for peptidic antagonists of the corticotropin-releasing factor receptor (CRFR): development of CRFR2beta-selective antisauvagine-30. Proc Natl Acad Sci U S A 95: 15264–15269.
  4. 4. Rivier J, Gulyas J, Kirby D, Low W, Perrin MH, et al. (2002) Potent and long-acting corticotropin releasing factor (CRF) receptor 2 selective peptide competitive antagonists. J Med Chem 45: 4737–4747.
  5. 5. Higelin J, Py-Lang G, Paternoster C, Ellis GJ, Patel A, et al. (2001) 125I-Antisauvagine-30: a novel and specific high-affinity radioligand for the characterization of corticotropin-releasing factor type 2 receptors. Neuropharmacology 40: 114–122.
  6. 6. Brauns O, Brauns S, Zimmermann B, Jahn O, Spiess J (2002) Differential responsiveness of CRF receptor subtypes to N-terminal truncation of peptidic ligands. Peptides 23: 881–888.
  7. 7. Hoare SR, Sullivan SK, Schwarz DA, Ling N, Vale WW, et al. (2004) Ligand affinity for amino-terminal and juxtamembrane domains of the corticotropin releasing factor type I receptor: regulation by G-protein and nonpeptide antagonists. Biochemistry 43: 3996–4011.
  8. 8. Hoare SR, Sullivan SK, Fan J, Khongsaly K, Grigoriadis DE (2005) Peptide ligand binding properties of the corticotropin-releasing factor (CRF) type 2 receptor: pharmacology of endogenously expressed receptors, G-protein-coupling sensitivity and determinants of CRF2 receptor selectivity. Peptides 26: 457–470.
  9. 9. Brauns O, Liepold T, Radulovic J, Spiess J (2001) Pharmacological and chemical properties of astressin, antisauvagine-30 and alpha-helCRF: significance for behavioral experiments. Neuropharmacology 41: 507–516.
  10. 10. Gutknecht E, Hauger RL, Van der Linden I, Vauquelin G, Dautzenberg FM (2008) Expression, binding, and signaling properties of CRF2(a) receptors endogenously expressed in human retinoblastoma Y79 cells: passage-dependent regulation of functional receptors. J Neurochem 104: 926–936.
  11. 11. Refojo D, Schweizer M, Kuehne C, Ehrenberg S, Thoeringer C, et al. (2011) Glutamatergic and dopaminergic neurons mediate anxiogenic and anxiolytic effects of CRHR1. Science 333: 1903–1907.
  12. 12. Della-Zuana O, Revereault L, Beck-Sickinger A, Monge A, Caignard DH, et al. (2004) A potent and selective NPY Y5 antagonist reduces food intake but not through blockade of the NPY Y5 receptor. Int J Obes Relat Metab Disord 28: 628–639.
  13. 13. Bale TL, Contarino A, Smith GW, Chan R, Gold LH, et al. (2000) Mice deficient for corticotropin-releasing hormone receptor-2 display anxiety-like behaviour and are hypersensitive to stress. Nat Genet 24: 410–414.
  14. 14. Kishimoto T, Radulovic J, Radulovic M, Lin CR, Schrick C, et al. (2000) Deletion of crhr2 reveals an anxiolytic role for corticotropin-releasing hormone receptor-2. Nat Genet 24: 415–419.
  15. 15. Gerlai R (1996) Gene-targeting studies of mammalian behavior: is it the mutation or the background genotype? Trends Neurosci 19: 177–181.
  16. 16. Contet C, Rawlins JN, Deacon RM (2001) A comparison of 129S2/SvHsd and C57BL/6JOlaHsd mice on a test battery assessing sensorimotor, affective and cognitive behaviours: implications for the study of genetically modified mice. Behav Brain Res 124: 33–46.
  17. 17. Voikar V, Koks S, Vasar E, Rauvala H (2001) Strain and gender differences in the behavior of mouse lines commonly used in transgenic studies. Physiol Behav 72: 271–281.
  18. 18. 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: 9077–9081.
  19. 19. Rodgers RJ, Boullier E, Chatzimichalaki P, Cooper GD, Shorten A (2002) Contrasting phenotypes of C57BL/6JOlaHsd, 129S2/SvHsd and 129/SvEv mice in two exploration-based tests of anxiety-related behaviour. Physiol Behav 77: 301–310.
  20. 20. Franklin K, Paxinos G (2007) The Mouse Brain in Stereotaxic Coordinates, 3rd Edition San Diego: Academic Press.
  21. 21. Njung'e K, Handley SL (1991) Evaluation of marble-burying behavior as a model of anxiety. Pharmacol Biochem Behav 38: 63–67.
  22. 22. File SE (2001) Factors controlling measures of anxiety and responses to novelty in the mouse. Behav Brain Res 125: 151–157.
  23. 23. Kalin NH, Sherman JE, Takahashi LK (1988) Antagonism of endogenous CRH systems attenuates stress-induced freezing behavior in rats. Brain Res 457: 130–135.
  24. 24. Fekete EM, Inoue K, Zhao Y, Rivier JE, Vale WW, et al. (2007) Delayed satiety-like actions and altered feeding microstructure by a selective type 2 corticotropin-releasing factor agonist in rats: intra-hypothalamic urocortin 3 administration reduces food intake by prolonging the post-meal interval. Neuropsychopharmacology 32: 1052–1068.
  25. 25. Coste SC, Kesterson RA, Heldwein KA, Stevens SL, Heard AD, et al. (2000) Abnormal adaptations to stress and impaired cardiovascular function in mice lacking corticotropin-releasing hormone receptor-2. Nat Genet 24: 403–409.
  26. 26. Valdez GR, Inoue K, Koob GF, Rivier J, Vale W, et al. (2002) Human urocortin II: mild locomotor suppressive and delayed anxiolytic-like effects of a novel corticotropin-releasing factor related peptide. Brain Res 943: 142–150.
  27. 27. Valdez GR, Zorrilla EP, Rivier J, Vale WW, Koob GF (2003) Locomotor suppressive and anxiolytic-like effects of urocortin 3, a highly selective type 2 corticotropin-releasing factor agonist. Brain Res 980: 206–212.
  28. 28. Zorrilla EP, Reinhardt LE, Valdez GR, Inoue K, Rivier JE, et al. (2004) Human urocortin 2, a corticotropin-releasing factor (CRF)2 agonist, and ovine CRF, a CRF1 agonist, differentially alter feeding and motor activity. J Pharmacol Exp Ther 310: 1027–1034.
  29. 29. Zhao Y, Valdez GR, Fekete EM, Rivier JE, Vale WW, et al. (2007) Subtype-selective corticotropin-releasing factor receptor agonists exert contrasting, but not opposite, effects on anxiety-related behavior in rats. J Pharmacol Exp Ther 323: 846–854.
  30. 30. Ohata H, Shibasaki T (2004) Effects of urocortin 2 and 3 on motor activity and food intake in rats. Peptides 25: 1703–1709.
  31. 31. Venihaki M, Sakihara S, Subramanian S, Dikkes P, Weninger SC, et al. (2004) Urocortin III, a brain neuropeptide of the corticotropin-releasing hormone family: modulation by stress and attenuation of some anxiety-like behaviours. J Neuroendocrinol 16: 411–422.
  32. 32. Telegdy G, Kadar K, Toth G (2011) Anxiolytic action of urocortin 3 fragments in mice. Behav Brain Res 222: 295–298.
  33. 33. Tanaka M, Kadar K, Toth G, Telegdy G (2011) Antidepressant-like effects of urocortin 3 fragments. Brain Res Bull 84: 414–418.
  34. 34. Tanaka M, Telegdy G (2008) Antidepressant-like effects of the CRF family peptides, urocortin 1, urocortin 2 and urocortin 3 in a modified forced swimming test in mice. Brain Res Bull 75: 509–512.
  35. 35. Henry B, Vale W, Markou A (2006) The effect of lateral septum corticotropin-releasing factor receptor 2 activation on anxiety is modulated by stress. J Neurosci 26: 9142–9152.
  36. 36. Takahashi LK, Ho SP, Livanov V, Graciani N, Arneric SP (2001) Antagonism of CRF(2) receptors produces anxiolytic behavior in animal models of anxiety. Brain Res 902: 135–142.
  37. 37. Pelleymounter MA, Joppa M, Carmouche M, Cullen MJ, Brown B, et al. (2000) Role of corticotropin-releasing factor (CRF) receptors in the anorexic syndrome induced by CRF. J Pharmacol Exp Ther 293: 799–806.
  38. 38. Miragaya JR, Harris RB (2008) Antagonism of corticotrophin-releasing factor receptors in the fourth ventricle modifies responses to mild but not restraint stress. Am J Physiol Regul Integr Comp Physiol 295: R404–416.
  39. 39. Sutherland JE, Conti LH (2011) Restraint stress-induced reduction in prepulse inhibition in Brown Norway rats: role of the CRF2 receptor. Neuropharmacology 60: 561–571.
  40. 40. Chance WT, Dayal R, Friend LA, Thomas I, Sheriff S (2007) Mediation of burn-induced hypermetabolism by CRF receptor-2 activity. Life Sci 80: 1064–1072.
  41. 41. Staub DR, Evans AK, Lowry CA (2006) Evidence supporting a role for corticotropin-releasing factor type 2 (CRF2) receptors in the regulation of subpopulations of serotonergic neurons. Brain Res 1070: 77–89.
  42. 42. Risbrough VB, Hauger RL, Roberts AL, Vale WW, Geyer MA (2004) Corticotropin-releasing factor receptors CRF1 and CRF2 exert both additive and opposing influences on defensive startle behavior. J Neurosci 24: 6545–6552.
  43. 43. Risbrough VB, Hauger RL, Pelleymounter MA, Geyer MA (2003) Role of corticotropin releasing factor (CRF) receptors 1 and 2 in CRF-potentiated acoustic startle in mice. Psychopharmacology (Berl) 170: 178–187.
  44. 44. Pelleymounter MA, Joppa M, Ling N, Foster AC (2002) Pharmacological evidence supporting a role for central corticotropin-releasing factor(2) receptors in behavioral, but not endocrine, response to environmental stress. J Pharmacol Exp Ther 302: 145–152.
  45. 45. Takahashi C, Ohata H, Shibasaki T (2011) Corticotropin-releasing factor (CRF) receptor subtypes in mediating neuronal activation of brain areas involved in responses to intracerebroventricular CRF and stress in rats. Peptides 32: 2384–2393.
  46. 46. Sekino A, Ohata H, Mano-Otagiri A, Arai K, Shibasaki T (2004) Both corticotropin-releasing factor receptor type 1 and type 2 are involved in stress-induced inhibition of food intake in rats. Psychopharmacology (Berl) 176: 30–38.
  47. 47. Maruyama H, Makino S, Noguchi T, Nishioka T, Hashimoto K (2007) Central type 2 corticotropin-releasing hormone receptor mediates hypothalamic-pituitary-adrenocortical axis activation in the rat. Neuroendocrinology 86: 1–16.
  48. 48. Navarro-Zaragoza J, Nunez C, Ruiz-Medina J, Laorden ML, Valverde O, et al. (2011) CRF(2) mediates the increased noradrenergic activity in the hypothalamic paraventricular nucleus and the negative state of morphine withdrawal in rats. Br J Pharmacol 162: 851–862.
  49. 49. Cooper MA, Huhman KL (2010) Blocking corticotropin-releasing factor-2 receptors, but not corticotropin-releasing factor-1 receptors or glucocorticoid feedback, disrupts the development of conditioned defeat. Physiol Behav 101: 527–532.
  50. 50. Chen CY, Inui A, Asakawa A, Fujino K, Kato I, et al. (2005) Des-acyl ghrelin acts by CRF type 2 receptors to disrupt fasted stomach motility in conscious rats. Gastroenterology 129: 8–25.
  51. 51. Cullen MJ, Ling N, Foster AC, Pelleymounter MA (2001) Urocortin, corticotropin releasing factor-2 receptors and energy balance. Endocrinology 142: 992–999.
  52. 52. Lam MP, Gianoulakis C (2011) Effects of corticotropin-releasing hormone receptor antagonists on the ethanol-induced increase of dynorphin A1-8 release in the rat central amygdala. Alcohol 45: 621–630.
  53. 53. Lam MP, Gianoulakis C (2011) Effects of acute ethanol on corticotropin-releasing hormone and beta-endorphin systems at the level of the rat central amygdala. Psychopharmacology (Berl) 218: 229–239.
  54. 54. Sananbenesi F, Fischer A, Schrick C, Spiess J, Radulovic J (2003) Mitogen-activated protein kinase signaling in the hippocampus and its modulation by corticotropin-releasing factor receptor 2: a possible link between stress and fear memory. J Neurosci 23: 11436–11443.
  55. 55. Radulovic J, Ruhmann A, Liepold T, Spiess J (1999) Modulation of learning and anxiety by corticotropin-releasing factor (CRF) and stress: differential roles of CRF receptors 1 and 2. J Neurosci 19: 5016–5025.
  56. 56. Todorovic C, Radulovic J, Jahn O, Radulovic M, Sherrin T, et al. (2007) Differential activation of CRF receptor subtypes removes stress-induced memory deficit and anxiety. Eur J Neurosci 25: 3385–3397.
  57. 57. Sahuque LL, Kullberg EF, McGeehan AJ, Kinder JR, Hicks MP, et al. (2006) Anxiogenic and aversive effects of corticotropin-releasing factor (CRF) in the bed nucleus of the stria terminalis in the rat: role of CRF receptor subtypes. Psychopharmacology (Berl) 186: 122–132.
  58. 58. Cooper MA, Huhman KL (2007) Corticotropin-releasing factor receptors in the dorsal raphe nucleus modulate social behavior in Syrian hamsters. Psychopharmacology (Berl) 194: 297–307.
  59. 59. Hammack SE, Schmid MJ, LoPresti ML, Der-Avakian A, Pellymounter MA, et al. (2003) Corticotropin releasing hormone type 2 receptors in the dorsal raphe nucleus mediate the behavioral consequences of uncontrollable stress. J Neurosci 23: 1019–1025.
  60. 60. Turek VF, Ryabinin AE (2005) Ethanol versus lipopolysaccharide-induced hypothermia: involvement of urocortin. Neuroscience 133: 1021–1028.
  61. 61. Bledsoe AC, Oliver KM, Scholl JL, Forster GL (2011) Anxiety states induced by post-weaning social isolation are mediated by CRF receptors in the dorsal raphe nucleus. Brain Res Bull 85: 117–122.
  62. 62. Scholl JL, Vuong SM, Forster GL (2010) Chronic amphetamine treatment enhances corticotropin-releasing factor-induced serotonin release in the amygdala. Eur J Pharmacol 644: 80–87.
  63. 63. Vuong SM, Oliver HA, Scholl JL, Oliver KM, Forster GL (2010) Increased anxiety-like behavior of rats during amphetamine withdrawal is reversed by CRF2 receptor antagonism. Behav Brain Res 208: 278–281.
  64. 64. Forster GL, Pringle RB, Mouw NJ, Vuong SM, Watt MJ, et al. (2008) Corticotropin-releasing factor in the dorsal raphe nucleus increases medial prefrontal cortical serotonin via type 2 receptors and median raphe nucleus activity. Eur J Neurosci 28: 299–310.
  65. 65. Lukkes JL, Forster GL, Renner KJ, Summers CH (2008) Corticotropin-releasing factor 1 and 2 receptors in the dorsal raphe differentially affect serotonin release in the nucleus accumbens. Eur J Pharmacol 578: 185–193.
  66. 66. Ohata H, Shibasaki T (2011) Involvement of CRF2 receptor in the brain regions in restraint-induced anorexia. Neuroreport 22: 494–498.
  67. 67. Amat J, Tamblyn JP, Paul ED, Bland ST, Amat P, et al. (2004) Microinjection of urocortin 2 into the dorsal raphe nucleus activates serotonergic neurons and increases extracellular serotonin in the basolateral amygdala. Neuroscience 129: 509–519.