125I-Antisauvagine-30: a novel and specific high-affinity radioligand for the characterization of corticotropin-releasing factor type 2 receptors

https://doi.org/10.1016/S0028-3908(00)00105-2Get rights and content

Abstract

Corticotropin-releasing factor (CRF) receptors type 1 (CRF1) and type 2 (CRF2) differ from each other in their pharmacological properties. The human and ovine CRF versions bind to CRF1 receptors with significantly higher affinity than to CRF2 receptors. Recently antisauvagine-30, an N-terminally truncated version of the CRF analog sauvagine, was characterized as a specific antagonist to mouse CRF2B. We have synthesized the radiolabeled version 125I-antisauvagine-30 and tested it for its affinity at human CRF1 (hCRF1), hCRF2A, Xenopus CRF1 (xCRF1) and xCRF2 receptors. In control binding studies 125I-labeled hCRF, sauvagine and astressin were also bound to these receptors. 125I-antisauvagine-30 exclusively bound to hCRF2A and xCRF2 but not to hCRF1 and xCRF1 receptors. 125I-antisauvagine-30 binding to hCRF2A and xCRF2 receptors was saturable and of high affinity (hCRF2A: Kd=125 pM; xCRF2: Kd=1.1 nM). In displacement binding experiments using 125I-antisauvagine-30 as radioligand several CRF analogs bound to hCRF2A and xCRF2 receptors with similar rank orders as reported with other CRF radioligands. Finally, preliminary studies using 125I-antisauvagine-30 binding to membrane homogenates prepared from different rat brain structures showed that the peptide bound specifically to brain areas expressing CRF2 receptors. These data demonstrate that 125I-antisauvagine-30 is the first high-affinity ligand to specifically label CRF2 receptors.

Introduction

The 41-amino acid peptide corticotropin-releasing factor (CRF), a main regulator of the body's stress response (for a review see Vale et al., 1997), is assumed to play a major role in several psychiatric disorders (Dunn and Berridge, 1990, Arborelius et al., 1999). CRF and its structurally related analogs mammalian urocortin (UCN), amphibian sauvagine (SVG) and fish urotensin I (URO) mediate their effects through two receptors, both of which belong to the class B subfamily of G protein-coupled receptors (GPCR) (reviewed in Kilpatrick et al., 1999). Two principal subtypes of the CRF receptor, type 1 (CRF1) and type 2 (CRF2), which are 70% identical on amino acid level, have been cloned from a variety of species (reviewed in Holsboer, 1999).

Despite their high degree of sequence homology, CRF1 and CRF2 receptors differ markedly from each other in their pharmacological properties and tissue distribution. The mammalian CRF1 receptor is nonselective for CRF, UCN, URO and SVG. These peptides are bound with similar affinity and are equipotent in their ability to stimulate cAMP accumulation in cells expressing recombinant mammalian CRF1 receptors (Vaughan et al., 1995, Donaldson et al., 1996, Dautzenberg et al., 1997, Dautzenberg et al., 1999a, Palchaudhuri et al., 1998). CRF2 receptors, however, are ligand-selective (Donaldson et al., 1996, Dautzenberg et al., 1997, Dautzenberg et al., 1999a, Ardati et al., 1999, Palchaudhuri et al., 1999). While UCN, URO and SVG are bound by CRF2 receptors with high affinity, CRF isolated from different species are bound with significantly lower affinity and thereby are CRF1 receptor-specific ligands.

The tissue distribution of CRF receptors has mainly been studied by detection of their mRNAs using in situ hybridization or RNAse protection analyses. Thereby, CRF1 receptor mRNA was found to be abundantly expressed in the brain, especially in cortex and cerebellum, and in the pituitary (Chalmers et al., 1995, Palchaudhuri et al., 1998). In the periphery, on the other hand, CRF1 receptor mRNA is restricted to low level expression in a few organs (Palchaudhuri et al., 1998). CRF2 receptor mRNA expression in the brain, in contrast to the CRF1 receptor, is very discrete, occurring only in the hypothalamus, lateral septum, hippocampus and dorsal raphe (Chalmers et al., 1995, Palchaudhuri et al., 1999, Sanchez et al., 1999), loci of the neuroendocrine stress response, behavioral components of the startle response, contextual fear conditioning, and explicit memory acquisition (reviewed in Chalmers et al., 1996, Hauger and Dautzenberg, 1999). In addition to its expression in the brain, CRF2 receptor mRNA is abundantly expressed in the skeletal muscle and the heart (Chalmers et al., 1995, Palchaudhuri et al., 1999).

Another method to study the distribution of CRF receptors in the brain is receptor autoradiography. For CRF1 receptors, this has been accomplished using 125I-labeled oCRF (Wynn et al., 1984), a radioligand displaying subnanomolar affinity to CRF1 receptors but >100 nM affinity to CRF2 receptors (Perrin et al., 1999). Although high-affinity 125I-labeled ligands displaying equal affinity to CRF1 and CRF2 receptors have been developed (Grigoriadis et al., 1996, Perrin et al., 1999), there is still the need for a high-affinity CRF2 receptor selective radioligand. Recently, [D-Phe11, His12]SVG(11–40), named antisauvagine-30 (aSVG-30), has been reported as the first CRF antagonist with ∼100-fold higher affinity to CRF2 than to CRF1 receptors (Rühmann et al., 1998).

In this study, we report the characterization of the labeled compound, (125I-aSVG-30) that has high affinity to human CRF2A (hCRF2A) and Xenopus CRF2 (xCRF2) receptors but is devoid of any activity at hCRF1 and xCRF1 receptors, allowing for the first time the exclusive labeling of the CRF2 receptor subtypes in vitro and providing a potential high-quality pharmacological tool for the study of this receptor in mixed populations of cells or tissues expressing both CRF receptor subtypes.

Section snippets

Materials, peptides and reagents

All cell culture reagents were purchased from Gibco/BRL. Aprotinin was obtained from Roche Molecular Biochemicals (Mannheim, Germany) and the CRF peptides were from Bachem (Bubendorf, Switzerland). The purity was greater than 95%.

Synthesis and radioiodination of aSVG-30

The peptide aSVG-30 was synthesized de novo. 125I-aSVG-30 was radiolabeled at the histidine residue by direct iodination with sodium 125I-iodide using the chloramine-T method. The product was purified by high-performance liquid chromatography using a linear

Binding of 125I-aSVG-30 and other 125I-labeled CRF analogs to hCRF1, hCRF2A, xCRF1 and xCRF2 receptors

To determine the relative potency of 125I-aSVG-30 to label CRF receptors, the radioligand (∼100 pM) was bound to membrane preparations of HEK293 cells stably expressing hCRF1, hCRF2A, xCRF1 or xCRF2 receptors. In control experiments, binding of the CRF analogs 125I-Tyr0-hCRF, 125I-Tyr0-SVG and 125I-astressin to the CRF receptor preparations was tested. Specific binding represented 77% (125I-Tyr0-hCRF at hCRF1 and xCRF1 receptors), 87% (125I-Tyr0-SVG at hCRF1, hCRF2A and xCRF2 receptors), 93% (

Discussion

In this study, we introduce the novel radioligand 125I-aSVG-30 for the specific labeling of native and recombinant CRF2 receptors. Although 125I- or 3H-labeled CRF analogs have been reported to bind CRF1 and CRF2 receptors with high affinity (Grigoriadis et al., 1996, Gottowik et al., 1997, Perrin et al., 1999), none of these ligands is specific for CRF2 receptors. The synthesis of the CRF analog aSVG-30 (Rühmann et al., 1998), therefore, was a major breakthrough for the development of a CRF2

References (32)

  • P.C. Wynn et al.

    Brain and pituitary receptors for corticotropin releasing factor: localization and differential regulation after adrenalectomy

    Peptides

    (1984)
  • L. Arborelius et al.

    The role of corticotropin-releasing factor in depression and anxiety disorders

    Journal of Endocrinology

    (1999)
  • L. Birnbaumer

    G proteins in signal transduction

    Annual Review in Pharmacology and Toxicology

    (1990)
  • D.T. Chalmers et al.

    Localization of novel corticotropin-releasing factor receptor (CRF2) mRNA expression to specific subcortical nuclei in rat brain: comparison with CRF1 receptor mRNA expression

    Journal of Neuroscience

    (1995)
  • F.M. Dautzenberg et al.

    Identification of two corticotropin-releasing factor receptors with high ligand selectivity from Xenopus laevis: unusual pharmacology of the type 1 receptor

    Journal of Neurochemistry

    (1997)
  • F.M. Dautzenberg et al.

    Mapping of the ligand-selective domain of the Xenopus laevis corticotropin-releasing factor receptor 1: implications for the ligand-binding site

    Proceedings of the National Academy of Science USA

    (1998)
  • Cited by (67)

    • Restraint stress-induced reduction in prepulse inhibition in Brown Norway rats: Role of the CRF<inf>2</inf> receptor

      2011, Neuropharmacology
      Citation Excerpt :

      Ten minutes after ICV infusion, rats were observed in the elevated plus maze for 5 min. In experiment 6, rats received an ICV infusion of the selective CRF2 receptor antagonist, antisauvagine-30 (ASV-30; 10.0 μg in 5.0 μl saline) (Higelin et al., 2001), or 5.0 μl saline. Ten minutes after ICV infusion, rats were restrained for 2 h or were handled briefly but not restrained.

    • Chronic amphetamine treatment enhances corticotropin-releasing factor-induced serotonin release in the amygdala

      2010, European Journal of Pharmacology
      Citation Excerpt :

      Given that CRF2 receptors are up-regulated in amphetamine pretreated rats (Pringle et al., 2008), the role of CRF2 receptors was assessed by pretreatment of the dRN with the selective CRF2 receptor antagonist antisauvagine-30 (ASV-30; Lukkes et al., 2008; Forster et al., 2008). This antagonist has 1000–10,000 fold selectivity for the vertebrate CRF2 receptor over the CRF1 receptor (Higelin et al., 2001), and does not affect CRF1 receptor mediated 5-HT release at this concentration infused into the dRN (2 μg; Lukkes et al., 2008). Using a separate group of rats from Experiment 1, amphetamine (2.5 mg/kg, i.p. n = 31) or saline (n = 14) was injected into each rat once-daily for 14 days during the dark (active) phase of the light cycle.

    • Placental urocortin-2 and -3: Endocrine or Paracrine Functioning During Healthy Pregnancy?

      2010, Placenta
      Citation Excerpt :

      Subsequently, the DHEAS-culture medium was after 4 hours replaced by DHEAS-culture medium supplemented with either vehicle (incubation medium with DHEAS), human Ucn-2 (up to 10−7 M; Phoenix Pharmaceuticals Inc. Belmont, USA), human Ucn-2 [10−7 M] and the specific CRF-R2 antagonist [23] antisauvagine1–30 (5 × 10−7 M; Polypeptide Lab. Inc., Torrance, USA), or with antisauvagine1–30 [5 × 10−7 M].

    View all citing articles on Scopus
    View full text