Elsevier

Neuropharmacology

Volume 51, Issue 2, August 2006, Pages 386-396
Neuropharmacology

Inhibition of the striatum-enriched phosphodiesterase PDE10A: A novel approach to the treatment of psychosis

https://doi.org/10.1016/j.neuropharm.2006.04.013Get rights and content

Abstract

Phosphodiesterase 10A (PDE10A) is a recently identified cyclic nucleotide phosphodiesterase expressed primarily in dopaminoreceptive medium spiny neurons of the striatum. We report that papaverine is a potent, specific inhibitor of PDE10A and use this compound to explore the role of PDE10A in regulating striatal function. Papaverine administration produces an increase in striatal tissue levels of cGMP and an increase in extracellular cAMP measured by microdialysis. These cyclic nucleotide changes are accompanied by increases in the phosphorylation of CREB and ERK, downstream markers of neuronal activation. In rats, papaverine potentiates haloperidol-induced catalepsy, consistent with the hypothesis that inhibition of PDE10A can increase striatal output and prompting a further evaluation of papaverine in models predictive of antipsychotic activity. Papaverine is found to inhibit conditioned avoidance responding in rats and mice and to inhibit PCP- and amphetamine-stimulated locomotor activity in rats. The effects of papaverine on striatal cGMP and CREB and ERK phosphorylation, as well as on conditioned avoidance responding, were absent in PDE10A knockout mice, indicating that the effects of the compound are the result of PDE10A inhibition. These results indicate that PDE10A regulates the activation of striatal medium spiny neurons through effects on cAMP- and cGMP-dependent signaling cascades. Furthermore, the present results demonstrate that papaverine has efficacy in behavioral models predictive of antipsychotic activity. Thus, inhibition of PDE10A may represent a novel approach to the treatment of psychosis.

Introduction

The phosphodiesterases (PDEs) are the enzymes that metabolically inactivate the ubiquitous intracellular second messengers cAMP and cGMP, a function that serves to regulate and compartmentalize the cyclic nucleotide signaling cascades. The PDEs are divided into 11 families based on sequence homology within the catalytic domain, which results in similar substrate specificity and sensitivity to pharmacological inhibitors within a family. Because inhibition of these enzymes can have a profound effect on cellular function, there is considerable interest in identifying family-specific PDE inhibitors to probe the physiology of these enzymes and to explore therapeutic utilities for such agents.

Of the 11 PDE families, PDE10A has the most restricted distribution. PDE10A mRNA is highly expressed only in brain and testes (Fujishige et al., 1999a, Fujishige et al., 1999b). In the brain, mRNA and protein are highly enriched in one population of neurons, the medium spiny projections neurons (MSNs) of the striatum (Fujishige et al., 1999a, Seeger et al., 2003). Within these neurons, the enzyme is distributed throughout the cell body, dendrites and axons. The striatal MSNs function as the principal input site of the basal ganglia, a series of interconnected subcortical nuclei that integrate cortical and dopaminergic inputs to facilitate planning and execution of relevant motor and cognitive patterns while suppressing unwanted or irrelevant patterns (Graybiel, 2000). The high level of expression of PDE10A in these neurons suggests that inhibition of this enzyme will result in changes in behaviors regulated by the activity of this circuit. This hypothesis is supported by results of studies in mice in which the gene for PDE10A is disrupted (PDE10A−/− mice; Siuciak et al., 2006—preceding report). PDE10A−/− mice are characterized by reduced locomotor activity in a number of behavioral paradigms and a reduced rate of acquisition of a conditioned avoidance response. This phenotype is interpreted to suggest that disruption of the PDE10A gene leads to increased activation of the MSNs and reduced behavioral responsiveness, which is consistent with the role of these neurons and the basal ganglia in regulating behavior.

The present studies were aimed at further investigating the function of PDE10A, in this case using a pharmacological approach. We identify papaverine as a selective and potent inhibitor of PDE10A, which was then used as a tool to study the effects of acute PDE10A inhibition in vivo. The results indicate that PDE10A regulates both cAMP and cGMP signaling in the MSNs and that inhibition of this enzyme causes behavioral effects consistent with an activation of striatal output. These results are discussed in regard to the hypothesis that PDE10A inhibitors may represent a novel approach to the treatment of psychosis.

Section snippets

PDE enzyme assay

Phosphodiesterases 1–4 and 7–11 were generated from recombinant clones transfected into SF9 cells as previously described (Fisher et al., 1998a, Fisher et al., 1998b, Fawcett et al., 2000, Seeger et al., 2003). PDE5 was isolated from human platelets and PDE6 from bovine retina as described (Boolell et al., 1996). PDE activity was measured using a scintillation proximity assay (SPA)-based method as previously described (Seeger et al., 2003). The effect of PDE inhibitors was investigated by

Papaverine is a selective inhibitor of rat PDE10A in vitro

To search for a useful inhibitor of PDE10A, a group of commercially available PDE inhibitors were screened for inhibition of cyclic nucleotide hydrolysis by recombinant rat PDE10A. As reported previously, dipyridamole and IBMX inhibited PDE10A with micromolar potency (Soderling et al., 1999). Papaverine, however, was found to be a particularly potent inhibitor of rat PDE10A with an IC50 of 36 nM. Inhibition by papaverine was competitive using either cAMP or cGMP as substrates (data not shown).

Discussion

In the preceding report (Siuciak et al., 2006) we began to investigate the function of PDE10A by evaluating the behavioral consequences of disrupting the PDE10A gene in mice. The most apparent phenotype of PDE10A−/− mice was a significant reduction in exploratory activity. Although the mice were slower at acquiring a conditioned avoidance task, there was no evidence of a general learning deficit in either passive avoidance or the Morris water maze. We proposed that this phenotype is consistent

Acknowledgements

We gratefully acknowledge the assistance of the following individuals involved with the making, validation and breeding of the PDE10A knockout mice: John McNeish, Jeffrey Stock, Christine Strick, Larry C. James, Robert Williams, Tina Walsh-Spivey and Linda Loverro. Portions of this work have been previously presented at the Society for Neuroscience 2002 and 2004 Meetings and the International Congress of Schizophrenia Research, March 2003.

References (47)

  • C. Marin et al.

    Dopamine D1 receptor stimulation but not dopamine D2 receptor stimulation attenuates haloperidol-induced behavioral sensitivity and receptor up-regulation

    Eur. J. Pharmacol.

    (1993)
  • K.P. Minneman et al.

    Receptor-linked cyclic AMP systems in rat neostriatum: differential localization revealed by kainic acid injection

    Brain Res.

    (1978)
  • D.R. Repaske et al.

    A cyclic GMP-stimulated cyclic nucleotide phosphodiesterase gene is highly expressed in the limbic system of the rat brain

    Neuroscience

    (1993)
  • T.F. Seeger et al.

    Immunohistochemical localization of PDE10A in the rat brain

    Brain Res.

    (2003)
  • J.A. Siuciak et al.

    Genetic deletion of the striatum-enriched phosphodiesterase PDE10A: Evidence for altered striatal function

    Neuropharmacology

    (2006)
  • H. Son et al.

    Long-term potentiation is reduced in mice that are doubly mutant in endothelial and neuronal nitric oxide synthase

    Cell

    (1996)
  • M.L. Wadenberg et al.

    The conditioned avoidance response test re-evaluated: is it a sensitive test for the detection of potentially atypical antipsychotics?

    Neurosci. Biobehav. Rev.

    (1999)
  • P. Zanassi et al.

    cAMP-dependent protein kinase induced cAMP-response element-binding protein phosphorylation via an intracellular calcium release/ERK dependent pathways in striatal neurons

    J Biol. Chem.

    (2001)
  • M. Adams et al.

    Loss of haloperidol induced gene expression and catalepsy in protein kinase A-deficient mice

    Proc. Natl. Acad. Sci. U.S.A.

    (1997)
  • C.A. Altar et al.

    Discriminatory roles for D1 and D2 dopamine receptors subtypes in the in vivo control of neostriatal cyclic GMP

    Eur. J. Pharmacol.

    (1990)
  • M. Boolell et al.

    Sildenafil: an orally active type 5 cyclic GMP-specific phosphodiesterase inhibitor for the treatment of penile erectile dysfunction

    Int. J. Impotence Res.

    (1996)
  • J.L. Bos et al.

    RAP1 signalling: adhering to new models

    Nat. Rev.

    (2001)
  • K. Brami-Cherrier et al.

    Dopamine induce a PI3-kinase independent activation of Akt in striatal neurons: a new route to cAMP response element-binding protein phosphorylation

    J. Neurosci.

    (2002)
  • Cited by (253)

    • Antiparkinsonian-like effects of CPL500036, a novel selective inhibitor of phosphodiesterase 10A, in the unilateral rat model of Parkinson's disease

      2021, European Journal of Pharmacology
      Citation Excerpt :

      Previous data concerning an influence of PDE10A inhibitors on the catalepsy or sedation induced by pharmacological tools i.e. neuroleptics or dopamine depleting agents were conflicting. While Megens and coworkers (2014) reported a reduction of such behavior after some inhibitors (JNJ-42311415, TP-10, MP-10), others showed an opposite effect (MP-10 or papaverine) (Siuciak et al., 2006a; Gentzel et al., 2015). In the present paper, we did not observe any enhancement of the lesion-induced catalepsy by CPL500036.

    • Medicinal chemistry strategies for the development of phosphodiesterase 10A (PDE10A) inhibitors - An update of recent progress

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Papaverine (1) is a potent PDE10A inhibitor. The efficacy of Papaverine in the conditioned avoidance response (CAR) model and inefficacy in PDE10A knock-out model revealed that the antipsychotic effect was due to PDE10A inhibition [42]. Papaverine also exhibited a positive effect in the social approach/social avoidance model (model of negative symptoms), Phencyclidine (PCP) and Amphetamine (AMPH) induced model, MK-801 induced model and novel object recognition (NOR) test [43].

    View all citing articles on Scopus
    View full text