Elsevier

Neuropharmacology

Volume 52, Issue 7, June 2007, Pages 1472-1481
Neuropharmacology

Discovery of a dual action first-in-class peptide that mimics and enhances CNS-mediated actions of thyrotropin-releasing hormone

https://doi.org/10.1016/j.neuropharm.2007.02.003Get rights and content

Abstract

Thyrotropin-releasing hormone (TRH) displays multiple CNS-mediated actions that have long been recognized to have therapeutic potential in treating a wide range of neurological disorders. Investigations of CNS functions and clinical use of TRH are hindered, however, due to its rapid degradation by TRH-degrading ectoenzyme (TRH-DE). We now report the discovery of a set of first-in-class compounds that display unique ability to both potently inhibit TRH-DE and bind to central TRH receptors with unparalleled affinity. This dual pharmacological activity within one molecular entity was found through selective manipulation of peptide stereochemistry. Notably, the lead compound of this set, l-pyroglutamyl-l-asparaginyl-l-prolyl-d-tyrosyl-d-tryptophan amide (Glp-Asn-Pro-d-Tyr-d-TrpNH2), is effective in vivo at producing and potentiating central actions of TRH without evoking release of thyroid-stimulating hormone (TSH). Specifically, this peptide displayed high plasma stability and combined potent inhibition of TRH-DE (Ki 151 nM) with high affinity binding to central TRH receptors (Ki 6.8 nM). Moreover, intraperitoneal injection of this peptide mimicked and augmented the effects of TRH on behavioural activity in rat. Analogous to TRH, it also antagonized pentobarbital-induced narcosis when administered intravenously. This discovery provides new opportunities for probing the role of TRH actions in the CNS and a basis for development of novel TRH-based neurotherapeutics.

Introduction

Thyrotropin-releasing hormone (TRH) is an important multifunctional signaling molecule in the CNS (Bauer et al., 1999). It is recognized that this naturally occurring tripeptide (Glp-His-ProNH2) has considerable influence on the activity of a number of neurobiological systems through CNS-mediated actions that are independent of the hypothalamic-pituitary-thyroid (HPT) axis (Gary et al., 2003). These central effects of TRH have attracted widespread attention particularly because of their potential therapeutic application across a broad platform of neurological conditions, such as CNS trauma, epilepsy, depression, spinocerebellar degeneration (SCD), Alzheimer's Disease and feeding disorders, for which there exists a pressing need for new and effective therapies (for review see Faden and Salzman, 1992, Kubek and Garg, 2002, Vetulani and Nalepa, 2000, Luo et al., 2002, Bennett et al., 1997, Nillni and Sevarino, 1999, Horita, 1998, Steward et al., 2003).

Critically, the rapid inactivation of TRH by the neuropeptide-specific ectopeptidase TRH-degrading ectoenzyme (TRH-DE, EC 3.4.19.6), also known as pyroglutamyl aminopeptidase II (PPII), impedes both therapeutic use of TRH (Kelly, 1995) and research into the biological functions of TRH in the CNS. In attempts to overcome this constraint much emphasis has been placed on the development of degradation-stabilized agonist analogs of TRH (Gary et al., 2003, Kelly, 1995, Horita, 1998, Metcalf, 1982). To accentuate CNS-mediated actions of TRH, a major goal of agonist analog design has been to separate central from endocrine actions; accordingly, such analogs would ideally bind with high affinity to central TRH receptors but have minimal effect on TSH release (Gary et al., 2003, Kelly, 1995, Horita, 1998, Metcalf, 1982). To date, all reported degradation-stabilized TRH analogs have reduced affinity compared to TRH for central TRH receptors and most retain some TSH-releasing activity. Interestingly, several TRH-like peptides, in which the central histidyl residue is replaced by another amino acid, have been shown to display central TRH effects without evoking TSH release (Vonhof et al., 1990, Lloyd et al., 2001, Pekary et al., 2005). Such peptides, however, have been shown to display poor affinity for central TRH receptors in competitive radioligand binding studies (Kelly et al., 2002, Hinkle et al., 2002). Further, it has been reported that their properties limit CNS access and hence neuropharmacological application (Prokai et al., 1999). As a consequence of their low affinity for known TRH receptors, it has been suggested that the central actions of these TRH-like peptides may be mediated by hitherto unidentified receptors (Hinkle et al., 2002, Colson and Gershengorn, 2006). Two TRH receptor subtypes have been identified in rat thus far: TRHR1 and TRHR2 (Cao et al., 1998, Itadani et al., 1998). A third TRH receptor subtype has recently been cloned in Xenopus laevis (xTRHR3) (Bidaud et al., 2002) and possibly a mammalian homologue of this receptor, or some other orphan receptor, may mediate CNS effects of TRH-related peptides (Lu et al., 2003). TRHR1 and TRHR2 have a distinct amino acid sequence and distribution pattern, but both display a similar high-affinity for [3H][3-Me-His2]TRH. Hence, [3H][3-Me-His2]TRH cannot be used to discriminate between these two known TRH receptor subtypes. TRHR1 is believed to be responsible for mediating the endocrine actions of TRH and TRHR2 appears to be strategically located to mediate central TRH actions, though it has not been detected in humans (Colson and Gershengorn, 2006). Subtype-selective analogues would be useful for probing the functions of these two receptors. Recently, analogues with reduced binding affinity in comparison to TRH have been identified that display a degree of selectivity for TRHR2 (Kaur et al., 2007).

The use of TRH-DE inhibitors represents an alternative approach to enhance the neuropharmacological actions of TRH (Charli et al., 1989, Kelly et al., 2000). TRH-DE is distributed primarily in serum and CNS tissues; peripheral organs, with the exception of lung and liver, are almost devoid of activity (Bauer et al., 1999). This enzyme is a particularly attractive therapeutic target because it displays extraordinary functional selectivity for TRH. Moreover, TRH is not degraded by any other enzyme in a position to affect TRH signaling (Turner, 1997). Consequently, TRH-DE inhibitors should enhance the biological actions of TRH exclusively.

We have previously shown that replacement of the central His residue in TRH with Asn confers resistance to proteolysis by TRH-DE and efficient inhibition of this enzyme (Kelly et al., 2000). Further, we have demonstrated that addition of hydrophobic l-amino acids to the C-terminus of Glp-Asn-ProNH2 substantially improves inhibitory potency (Kelly et al., 2005). Although Glp-Asn-ProNH2 is not susceptible to hydrolysis by general amino- and carboxy-peptidases because of the presence of an N-terminal Glp residue and an amidated C-terminus (Turner, 1997), inclusion of l-amino acids in the extension may be expected to render such peptides susceptible to degradation. Replacement of l-amino acids by d-amino acids is a common method used to confer stability to proteolysis, as well as to stabilize bioactive conformations (Hruby, 2002). Indeed, replacement of the hydrophobic l-amino acid residues with their d-isomers in C-terminally extended analogs of Glp-Asn-ProNH2 was a key development leading to the discovery of the set of compounds reported herein, which display unique capability not only to inhibit TRH-DE with high potency, but also bind to central native TRH receptors with an affinity greater than that of all other previously described TRH analogs. Compounds with such potent dual-action have not been described before and are of both experimental and therapeutic interest. Moreover, we demonstrate that one member of this new class of compound, Glp-Asn-Pro-d-Tyr-d-TrpNH2, is effective in producing central TRH actions in vivo while not evoking TSH release, thereby obviating any changes in thyroid status. Thus, this peptide not only contains the dual functionalities of an ectopeptidase inhibitor and receptor binding ligand within one molecular structure, but also displays properties in vivo that are highly desirable for a TRH analog with investigative and therapeutic application.

Section snippets

Materials and methods

All animal experiments were carried out under license in accordance with EC directive 86/609/EC. All efforts were made to minimize animal suffering and to reduce the number of animals used.

Inhibition of TRH-DE and binding to central native TRH receptors

Enzyme inhibition and receptor binding constants for each peptide were determined using kinetic and radioligand binding assays. Table 1 summarizes the affinity of C-terminally extended analogs of Glp-Asn-ProNH2 for TRH-DE and native rat brain TRH receptors in comparison to those of TRH and its agonist analog [3-Me-His2]TRH. The data show that peptides containing d-amino acids in the C-terminal extension of Glp-Asn-Pro retained high inhibitory potency for TRH-DE compared to their all-l

Discussion

The susceptibility of TRH to degradation by TRH-DE is recognized to be a major factor undermining the investigation of the neurobiological functions and therapeutic use of TRH. Thus, despite all research efforts over the last thirty years, the functions of CNS-mediated actions of TRH are still poorly understood. There remains, therefore, a need to develop strategies to enhance or simulate central TRH actions, both to facilitate investigations into the role of these actions and to provide

Acknowledgements

This work was supported by the Wellcome Trust (061881/Z/00), the Health Research Board, Ireland (DIP 12/2002), and Enterprise Ireland (SC/2002/0496).

References (72)

  • P.M. Hinkle et al.

    Role of TRH receptors as possible mediators of analeptic actions of TRH-like peptides

    Brain Res.

    (2002)
  • A. Horita

    An update on the CNS actions of TRH and its analogs

    Life Sci.

    (1998)
  • H. Itadani et al.

    Cloning and characterization of a new subtype of thyrotropin-releasing hormone receptors

    Biochem. Biophys. Res. Commun.

    (1998)
  • H.C. Jackson

    Behavioral profile of the TRH analogue RX77368 in developing rats

    Peptides

    (1990)
  • N. Kaur et al.

    Modifications of the pyroglutamic acid and histidine residues in thyrotropin-releasing hormone (TRH) yield analogs with selectivity for TRH receptor type 2 over type 1

    Bioorg. Med. Chem.

    (2007)
  • A.J. Kastin et al.

    Improvement in mental depression with decreased thyrotropin response after administration of thyrotropin-releasing hormone

    Lancet

    (1972)
  • J.A. Kelly et al.

    Kinetic investigation of the specificity of porcine brain thyrotropin-releasing hormone-degrading ectoenzyme for thyrotropin-releasing hormone-like peptides

    J. Biol. Chem.

    (2000)
  • J.A. Kelly et al.

    Pharmacologically distinct binding sites in rat brain for [3H]thyrotropin-releasing hormone (TRH) and [3H][3-methyl-histidine2]TRH

    Biochem. Pharmacol.

    (2002)
  • C.S. Konkoy et al.

    Ectoenzymes as sites of peptide regulation

    Trends Pharmacol. Sci.

    (1996)
  • M.J. Kubek et al.

    Thyrotropin-releasing hormone in the treatment of intractable epilepsy

    Pediatr. Neurol.

    (2002)
  • L. Lankiewicz et al.

    Biological activities of thionated thyrotropin-releasing hormone analogs

    Biochem. Biophys. Res. Commun.

    (1992)
  • R.L. Lloyd et al.

    Antidepressant effects of thyrotropin-releasing hormone analogues using a rodent model of depression

    Pharmacol. Biochem. Behav.

    (2001)
  • M. Matsushita et al.

    Effect of JTP-2942, a novel thyrotropin-releasing hormone analogue, on pentobarbital-induced anesthesia in rats

    Eur. J. Pharmacol.

    (1995)
  • G. Metcalf

    Regulatory peptides as a source of new drugs-the clinical prospects for analogues of TRH which are resistant to metabolic degradation

    Brain Res.

    (1982)
  • G. Metcalf et al.

    Is thyrotropin releasing hormone an endogenous ergotropic substance in the brain?

    Lancet

    (1981)
  • Y. Miwa et al.

    YM-14673, a thyrotropin-releasing hormone analogue, injected into the nucleus accumbens and the striatum produces repetitive jaw movements in rats

    Eur. J. Pharmacol.

    (1995)
  • M. Miyamoto et al.

    A TRH analog (DN-1417): motor stimulation with rearing related to catecholaminergic mechanisms in rats

    Neuropharmacology

    (1984)
  • R. Morphy et al.

    From magic bullets to designed multiple ligands. Drug Discov

    Today

    (2004)
  • A.E. Pekary et al.

    Electroconvulsive seizures modulate levels of thyrotropin releasing hormone and related peptides in rat hypothalamus, cingulate and lateral cerebellum

    Brain Res.

    (2000)
  • A.E. Pekary et al.

    Valproate modulates TRH receptor, TRH and TRH-like peptide levels in rat brain

    Peptides

    (2004)
  • A.J. Prange et al.

    Effects of thyrotropin-releasing hormone in depression

    Lancet

    (1972)
  • A. Sattin et al.

    Lithium modulates expression of TRH receptors and TRH-related peptides in rat brain

    Neuroscience

    (2002)
  • J. Vetulani et al.

    Antidepressants: past, present and future

    Eur. J. Pharmacol.

    (2000)
  • S. Vonhof et al.

    Norvaline2-TRH: binding to TRH receptors in rat brain homogenates

    Eur. J. Pharmacol.

    (1990)
  • J.L. Waddington et al.

    Behavioural pharmacology of ‘D-1-like’ dopamine receptors: further subtyping, new pharmacological probes and interactions with ‘D-2-like’ receptors

    Prog. Neuropsychopharmacol. Biol. Psychiatry.

    (1995)
  • M.B. Youdim et al.

    Multi-functional drugs for various CNS targets in the treatment of neurodegenerative disorders

    Trends Pharmacol. Sci.

    (2005)
  • Cited by (29)

    • Effect of L-pGlu-(1-benzyl)-l-His-l-Pro-NH<inf>2</inf> against in-vitro and in-vivo models of cerebral ischemia and associated neurological disorders

      2016, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      Indeed the neuroprotection in cerebral ischemia; several TRH-analogues have been evaluated including YM-14673 (Azetirelin), RX77368 and CG3509 (NS-3, Montirelin). However, the specific molecular interaction of some analogues including YM-14673 (Azetirelin), RX77368 have not reported whereas others including CG3509 (NS-3, Montirelin) has found to be responsible for increased release of dopamine in cerebral cortex [9,17,40–42]. Moreover, TAO910 (Taltirelin, Ceredist) and JTP-2942 were found to be sensible for stimulating acetylcholine release, dopamine synthesis and inhibition of glutamate associated cell death.

    • Mediobasal hypothalamic and adenohypophyseal TRH-degrading enzyme (PPII) is down-regulated by zinc deficiency

      2015, International Journal of Developmental Neuroscience
      Citation Excerpt :

      However, the systemic administration of the inhibitor does not allow to determine whether the effect is due to a low activity of the enzyme located in the AH or the one synthesized in the MBH. An in vivo i.p. administration of TRH is also able to raise TSH serum levels, which remain elevated for longer time when a PPII inhibitor is administered (Scalabrino et al., 2007). In the present study however, we observed that with no exogenous administration of TRH, the down-regulation of the AH PPII was enough to increase TSH serum levels in Zn-deficient animals.

    • First-in-class thyrotropin-releasing hormone (TRH)-based compound binds to a pharmacologically distinct TRH receptor subtype in human brain and is effective in neurodegenerative models

      2015, Neuropharmacology
      Citation Excerpt :

      However, since the Ki for the receptor is in the nanomolar range and that for the enzyme is two orders of magnitude higher it may be anticipated that the interaction with the receptor would predominate. As we have noted previously, inhibition of TRH-DE by JAK4D might also be anticipated to increase the amount of endogenous TRH available to the pituitary (Scalabrino et al., 2007). As anterior pituitary basal TRH-DE levels are very low, TRH-DE inhibition is expected to have minimal influence on pituitary hormone secretion (Scalabrino et al., 2007).

    • TRH

      2013, Handbook of Biologically Active Peptides
    • Protective effects of l-pGlu-(2-propyl)-l-His-l-ProNH<inf>2</inf>, a newer thyrotropin releasing hormone analog in in vitro and in vivo models of cerebral ischemia

      2011, Peptides
      Citation Excerpt :

      TRH and TRH analogs have been investigated in different models of ischemia, providing promising results [17,30,40,48,52–54]. Over time, various TRH analogs have been synthesized to get better half life and CNS selectivity [7,15,20,21,26,31,37]. Yet, most of the reported analogs have reduced affinity for the central TRH receptors and retain some TSH activity.

    View all citing articles on Scopus
    1

    G.A.S. and N.H. contributed equally to this work.

    View full text