Elsevier

Neuropharmacology

Volume 47, Issue 7, December 2004, Pages 1081-1092
Neuropharmacology

Inhibition of phosphodiesterase 2 increases neuronal cGMP, synaptic plasticity and memory performance

https://doi.org/10.1016/j.neuropharm.2004.07.040Get rights and content

Abstract

An essential element of the signalling cascade leading to synaptic plasticity is the intracellular second messenger molecule guanosine 3′,5′-cyclic monophosphate (cGMP). Using the novel, potent, and selective inhibitor Bay 60-7550, we show that the enzyme 3′,5′-cyclic nucleotide phosphodiesterase type 2 (PDE2) is responsible for the degradation of newly synthesized cGMP in cultured neurons and hippocampal slices. Inhibition of PDE2 enhanced long-term potentiation of synaptic transmission without altering basal synaptic transmission. Inhibition of PDE2 also improved the performance of rats in social and object recognition memory tasks, and reversed MK801-induced deficits in spontaneous alternation in mice in a T-maze. Our data provide strong evidence that inhibition of PDE2 can improve memory functions by enhancing neuronal plasticity.

Introduction

The nitric oxide/guanosine 3′,5′-cyclic monophosphate (NO/cGMP) signal transduction pathway has an important role during synaptic plasticity. Nitric oxide synthase (NOS), the enzyme that generates NO, is activated by Ca2+-influx through glutamate receptors of the N-methyl-d-aspartate (NMDA)-type on postsynaptic neurons (Garthwaite, 1991). NO stimulates both post- and presynaptic guanylyl cyclase (GC), leading to the formation of the second messenger cGMP (Boulton et al., 1995, De Vente and Steinbusch, 1992). In turn, cGMP directly opens cyclic nucleotide-gated ion channels (Savchenko et al., 1997) and activates cGMP-dependent protein kinase (PKG), which phosphorylates synaptic proteins (Wang and Robinson, 1997). Inhibitors of NOS, GC, or PKG block long-term potentiation (LTP) of hippocampal synaptic transmission, a physiological correlate of learning and memory processes (Böhme et al., 1991, O’Dell et al., 1991, Schuman and Madison, 1991, Zhuo et al., 1994, Boulton et al., 1995, Lu et al., 1999). Thus, NO may serve as a retrograde signal that closes an important feedback loop during use-dependent synaptic plasticity in the hippocampus (reviewed by Collingridge and Bliss, 1995, Hawkins et al., 1998). While marked reductions in LTP have been demonstrated in knock-out mice that lack both endothelial and neuronal NO synthase (Son et al., 1996), the role of NO as a retrograde messenger in LTP is controversial (discussed in Holscher, 1997, Hawkins et al., 1998). Changes in the strength of the postsynaptic response during LTP as a consequence of the activation of cAMP-dependent protein kinase (PKA) and calcium/calmodulin-dependent kinase include increased glutamate receptor density and function (reviewed by Lynch, 2004). Several substrate proteins of these kinases are also phosphorylated by PKG which could result in similar changes in postsynaptic function through the NO/cGMP/PKG pathway (Wang and Robinson, 1997, Ahem et al., 2002).

NOS inhibitors impair memory performance in one-trial passive avoidance in chicks (Holscher and Rose, 1993) and spontaneous alternation in mice (Yamada et al., 1996). Local injection of cGMP analogues can ameliorate the deficit induced by NOS-inhibition (Yamada et al., 1996). Intra-hippocampal injections of 8-Br cGMP improved the ability of rats to remember objects (Prickaerts et al., 2002a), as well as retention performance in a passive avoidance task (Bernabeu et al., 1996). Furthermore, cGMP levels in the hippocampus of rats increased transiently during the first 30 min of a passive avoidance task (Bernabeu et al., 1996). These findings support an essential role of cGMP in early memory formation.

An important part of the signal transduction process is the rapid degradation of cGMP by cyclic nucleotide phosphodiesterases (PDEs). At least 21 PDE genes have been identified and subgrouped into 11 families (Soderling and Beavo, 2000, O’Donnell, 2000). PDE4, PDE7 and PDE8 hydrolyse cAMP, while PDE5, PDE6 and PDE9 hydrolyse cGMP; PDE1, PDE2, PDE3, PDE10 and PDE11 can hydrolyse both cAMP and cGMP. The strong expression of PDE2 in neurons of the hippocampus and cortex (Repaske et al., 1993) suggests that this enzyme may control intraneuronal cGMP and cAMP levels in areas that are important for memory formation and storage. A characteristic feature of PDE2 is the positive cooperativity of the substrate cGMP—low levels of cGMP enhance the rate of cAMP hydrolysis by a factor of 5–6 (Martins et al., 1982). Under basal conditions, PDE2 activity in neurons is low, but it is stimulated by the acute increase in cGMP levels following GC activation during signal transduction. Therefore, inhibition of brain PDE2 may selectively increase cGMP and cAMP levels in active synapses and could thus influence synaptic plasticity and memory formation.

We investigated the role of PDE2 in neuronal signal transduction and memory, using the highly potent and selective PDE2 inhibitor 2-(3,4-dimethoxybenzyl)-7-{(1R)-1-[(1R)-1-hydroxyethyl]-4-phenylbutyl}-5-methyllimidazo[5,1-f][1,2,4]triazin-4(3H)-one (Bay 60-7550). To this end, we determined the effects of Bay 60-7550 on cGMP levels in neurons in primary culture and in slices, on hippocampal LTP, and on cognitive function in rats and mice, assessed using an object recognition task, social memory, and a T-maze spontaneous alternation task.

Section snippets

Materials

The novel PDE2 inhibitor 2-(3,4-dimethoxybenzyl)-7-{(1R)-1-[(1R)-1-hydroxyethyl]-4-phenylbutyl}-5-methyl imidazo[5,1-f][1,2,4]triazin-4(3H)-one (Bay 60-7550) (Table 1) and the GC stimulator Bay 41-8543 (Stasch et al., 2002) were synthesized by the Chemistry Department of Pharma Research, Bayer AG (Wuppertal, Germany).

PDE inhibition and selectivity assays

PDE2 was purified from bovine heart, PDE1 from bovine aorta and PDE5 from human platelets (Saenz de Tejada et al., 2001). Human recombinant PDEs were expressed in SF9 cells using

Bay 60-7550—a selective PDE2 inhibitor

Bay 60-7550 inhibited the activity of PDE2 purified from bovine heart with an IC50 value of 2.0±0.7 nM (n=12) and human recombinant PDE2 with an IC50 value of 4.7±1.0 nM (n=7). Bay 60-7550 showed 50-fold selectivity for PDE2 compared to PDE1 and more than 100-fold selectivity compared to PDE5 and the other PDEs tested (PDE3B, PDE4B, PDE7B, PDE8A, PDE9A, PDE10A, PDE11A, see Table 1, Fig. 1A). Bay 60-7550 had an IC50 of >10 μM (less than 50% inhibition at 10 μM) for the following receptors and

Discussion

Owing to its high potency and excellent selectivity profile, Bay 60-7550 is a very useful tool for the study of PDE2-dependent processes. To date, the physiological actions of PDE2 have been characterized with erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) (Mery and Fischmeister, 1994, Podzuweit et al., 1995), which inhibits PDE2 with an IC50 of ∼1 μM. However, in contrast to Bay 60-7550, EHNA is also a very potent adenosine deaminase inhibitor (IC50∼2 nM), which complicates the interpretation of

Acknowledgements

The LTP studies were conducted in the laboratories of Dr. U.H. Schröder and Professor K. Reymann, Forschungsinstitut Angewandte Neurowissenschaften (FAN), Magdeburg, Germany. A. Sik, Maastricht, is acknowledged for contributions to the object recognition studies. K. Selbach is acknowledged for contributions to the social recognition studies. This work is dedicated to our former colleague, the late Ulrich Niewoehner.

References (74)

  • J.D. De Vente et al.

    On the stimulation of soluble and particulate guanylate cyclase in the rat brain and the involvement of nitric oxide as studied by cGMP immunocytochemistry

    Acta Histochem

    (1992)
  • A. Ennaceur et al.

    A new one-trial test for neurobiological studies of memory in rats 1: behavioral data

    Behav. Brain Res

    (1988)
  • J. Garthwaite

    Glutamate, nitric oxide and cell–cell signalling in the nervous system

    Trends Neurosci

    (1991)
  • R. Gerlai

    A new continuous alternation task in T-maze detects hippocampal dysfunction in mice. A strain comparison and lesion study

    Behav. Brain Res

    (1998)
  • R.D. Hawkins et al.

    Nitric oxide as a retrograde messenger during long-term potentiation in hippocampus

    Prog. Brain Res

    (1998)
  • C. Holscher

    Nitric oxide, the enigmatic neuronal messenger: its role in synaptic plasticity

    Trends Neurosci

    (1997)
  • C. Holscher et al.

    Inhibiting synthesis of the putative retrograde messenger nitric oxide results in amnesia in a passive avoidance task in the chick

    Brain Res

    (1993)
  • F. Libert et al.

    Cloning and functional characterization of a human A1 adenosine receptor

    Biochem. Biophys. Res. Commun

    (1992)
  • K. Loughney et al.

    Isolation and characterization of cDNAs encoding PDE5A, a human cGMP-binding, cGMP-specific 3′,5′-cyclic nucleotide phosphodiesterase

    Gene

    (1998)
  • G.R. Martin et al.

    Receptors for 5-hydroxytryptamine: current perspectives on classification and nomenclature

    Neuropharmacology

    (1994)
  • T.J. Martins et al.

    Purification and characterization of a cyclic GMP-stimulated cyclic nucleotide phosphodiesterase from bovine tissues

    J. Biol. Chem

    (1982)
  • R.W. Olsen et al.

    [3H]AMPA binding to glutamate receptor subpopulations in rat brain

    Brain Res

    (1987)
  • T. Podzuweit et al.

    Isozyme selective inhibition of cGMP-stimulated cyclic nucleotide phosphodiesterase by EHNA (erythro-9-(2-hydroxy-3-nonyl)adenine)

    Cell Sign

    (1995)
  • J. Prickaerts et al.

    Possible role of nitric oxide-cyclic GMP pathway in object recognition, effects of 7-nitroindazole and zaprinast

    Eur. J. Pharmacol

    (1997)
  • J. Prickaerts et al.

    Cyclic GMP, but not cyclic AMP, in rats hippocampus is involved in early stages of object memory consolidation

    Eur. J. Pharmacol

    (2002)
  • 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)
  • Y. Shen et al.

    Molecular cloning and expression of a 5-hydroxytryptamine7 serotonin receptor subtype

    J. Biol. Chem

    (1993)
  • M.A. Sills et al.

    [3H]CGP39653: a new N-methyl-d-aspartate antagonist radioligand with low nanomolar affinity in rat brain

    Eur. J. Pharmacol

    (1991)
  • S.H. Soderling et al.

    Regulation of cAMP and cGMP signaling: new phosphodiesterases and new functions

    Curr. Opin. Cell Biol

    (2000)
  • H. Son et al.

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

    Cell

    (1996)
  • R.C. Speth et al.

    Benzodiazepine receptors: temperature dependence of [3H]flunitrazepam binding

    Life Sci

    (1979)
  • B. Suchanek et al.

    The 5-HT1A receptor agonist Bay x 3702 prevents staurosporine-induced apoptosis

    Eur. J. Pharmacol

    (1998)
  • P. Urban et al.

    Comparative membrane locations and activities of human monoamine oxidases expressed in yeast

    FEBS Lett

    (1991)
  • W.C.G. Van Staveren et al.

    The effects of phosphodiesterase inhibition on cyclic GMP and cyclic AMP accumulation in the hippocampus of the rat

    Brain Res

    (2001)
  • K. Yamada et al.

    Role of nitric oxide and cyclic-GMP in the dizolcipine-induced impairment of spontaneous-alternation behavior in mice

    Neuroscience

    (1996)
  • K. Yanai et al.

    Binding characteristics of a histamine H3-receptor antagonist, [3H]S-methylthioperamide: comparison with [3H](R)alpha-methylhistamine binding to rat tissues

    Jap. J. Pharmacol

    (1994)
  • G.P. Ahem et al.

    cGMP and S-nitrosylation: two routes for modulation of neuronal excitability by NO

    Trends Neurosci

    (2002)
  • Cited by (272)

    View all citing articles on Scopus
    1

    Present address: Neuroscience Discovery Research, Eli Lilly and Company Limited, Lilly Research Centre, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK.

    2

    Present address: ID Lelystad, Institute for Animal Science and Health BV, Lelystad 8200 AB, The Netherlands.

    3

    Present address: Merz GmbH, 60318 Frankfurt, Germany.

    4

    Present address: Roche Pharmaceuticals, Palo Alto, CA 94304, USA.

    5

    Present address: Johnson & Johnson PRD, B2340 Beerse, Belgium.

    6

    Present address: Envivo Pharmaceuticals, Cambridge, MA 02139, USA.

    View full text