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

A Biphasic and Brain-Region Selective Down-Regulation of Cyclic Adenosine Monophosphate Concentrations Supports Object Recognition in the Rat

  • Maïte Hotte,

    Affiliations Université de Caen Basse–Normandie, Groupe Mémoire et Plasticité comportementale (GMPc), EA4259, IFR 146, Caen, France, Université de Rouen, NeoVasc, EA 4309, IFRMP23, IHURBM, Rouen, France

  • François Dauphin,

    Affiliation Université de Caen Basse–Normandie, Groupe Mémoire et Plasticité comportementale (GMPc), EA4259, IFR 146, Caen, France

  • Thomas Freret,

    Affiliation Université de Caen Basse–Normandie, Groupe Mémoire et Plasticité comportementale (GMPc), EA4259, IFR 146, Caen, France

  • Michel Boulouard,

    Affiliation Université de Caen Basse–Normandie, Groupe Mémoire et Plasticité comportementale (GMPc), EA4259, IFR 146, Caen, France

  • Guenaëlle Levallet

    levallet-g@chu-caen.fr

    Affiliations Université de Caen Basse–Normandie, Groupe Mémoire et Plasticité comportementale (GMPc), EA4259, IFR 146, Caen, France, CHU de Caen, Service d'Anatomie Pathologie, Caen, France

Abstract

Background

We aimed to further understand the relationship between cAMP concentration and mnesic performance.

Methods and Findings

Rats were injected with milrinone (PDE3 inhibitor, 0.3 mg/kg, i.p.), rolipram (PDE4 inhibitor, 0.3 mg/kg, i.p.) and/or the selective 5-HT4R agonist RS 67333 (1 mg/kg, i.p.) before testing in the object recognition paradigm. Cyclic AMP concentrations were measured in brain structures linked to episodic-like memory (i.e. hippocampus, prefrontal and perirhinal cortices) before or after either the sample or the testing phase. Except in the hippocampus of rolipram treated-rats, all treatment increased cAMP levels in each brain sub-region studied before the sample phase. After the sample phase, cAMP levels were significantly increased in hippocampus (1.8 fold), prefrontal (1.3 fold) and perirhinal (1.3 fold) cortices from controls rat while decreased in prefrontal cortex (∼0.83 to 0.62 fold) from drug-treated rats (except for milrinone+RS 67333 treatment). After the testing phase, cAMP concentrations were still increased in both the hippocampus (2.76 fold) and the perirhinal cortex (2.1 fold) from controls animals. Minor increase were reported in hippocampus and perirhinal cortex from both rolipram (respectively, 1.44 fold and 1.70 fold) and milrinone (respectively 1.46 fold and 1.56 fold)-treated rat. Following the paradigm, cAMP levels were significantly lower in the hippocampus, prefrontal and perirhinal cortices from drug-treated rat when compared to controls animals, however, only drug-treated rats spent longer time exploring the novel object during the testing phase (inter-phase interval of 4 h).

Conclusions

Our results strongly suggest that a “pre-sample” early increase in cAMP levels followed by a specific lowering of cAMP concentrations in each brain sub-region linked to the object recognition paradigm support learning efficacy after a middle-term delay.

Introduction

Most modern theories of learning and memory postulate that memory processes require cyclic adenosine monophosphate (cAMP) synthesis [1]; however, there is little evidence concerning the mechanisms by which memory affects adenylyl cyclase activity (cAMP synthesis) and/or phosphodiesterase (PDE) activity (cAMP degradation). Literature reports indicate that activation of the cAMP-PKA pathway cascade by memory processes triggers activation of transcription factors such as CREB [2], leading to neural processes that underlie learning and memory [1], [3][5]. Consequently, several studies argue that artificial cAMP-PKA cascade activation through intra-hippocampal infusion of 8Br-cAMP, adenylyl cyclase or PKA activation improves memory performance [4], [6][9] whereas pharmacological inhibition of PKA disrupts hippocampal long term potentiation and hippocampus-based long-term memory [6], [10], [11]. Memory efficiency seems, however, to require a restricted or selective cAMP production; high cAMP levels do not necessarily improve memory. Indeed, studies on flies and mice show that increases in adenylyl cyclase activity can result in memory deficits [12], [13]. Similarly, increasing PKA activity impairs prefrontal cortex-dependent memory in mice and expression of a constitutively active isoform of the G-protein subunit Gαs impairs mice behavioural performance in a fear-conditioning task [14]. These works clearly demonstrate the complexity of cAMP-dependent responses.

Mnesic mechanisms may be investigated through the use of an object recognition memory task, a one-phase task based on spontaneous activity and the natural preference that rodents display to explore a novel object rather than a familiar one [15]. With this paradigm, memory performances were demonstrated to be enhanced by the activation of serotonin 5-HT4 receptors (5-HT4R) [16][19], receptors that have been also demonstrated to be implicated in short- and long-term memory processes in laboratory animals [20][30] (for review see [31]). Activation of 5-HT4R, positively coupled to adenylyl cyclase, induce increases in cAMP concentrations that can be regulated by activation of cAMP phosphodiesterases (PDE) isoforms from families 1, 2, 3, and 4 [18]. Involvement of PDE4 inhibitors in working and reference memory [5], [32][34] has already been well investigated through the use of rolipram, a selective PDE4 inhibitor. In fact, several studies have already reported a positive effect of PDE4 inhibition on spatial memory [3], [5], [33][37], inhibitory avoidance learning [5], [33], [38], contextual fear conditioning [4], [39], and object recognition [40][42]. The PDE4 isoenzymes are encoded by four independent genes (Pde4a to Pde4d), which generate more than 25 splice variants [43], [44]. Each splice variant exhibits unique properties leading to specific control of cAMP levels [45], [46]. Few studies deal with the involvement of each PDE4 isoforms in memory performance as few studies have also investigated the effect of others cAMP-PDE families on memory performance, especially in the object recognition task [42], [47]. Thus, from the literature, little is known about the behavioural consequence and especially memory performance following PDE4 [18], PDE3 [48] or PDE2 [47] inhibition although these PDE families accounts for a major part of the total cAMP-PDE-hydrolysing activities in the hippocampus, the prefrontal and perirhinal cortices, brain structures involved in recognition memory [18].

A way to enhance cAMP signaling and consequently influence the pathways involved in object recognition (episodic-like) memory, is to stimulate 5-HT4Rs and/or inhibit PDE enzymes, especially PDE3 and 4 [18]. Here, we further characterize the respective role of PDE3 and PDE4 in the processes of recognition memory and assayed the relationship between cAMP concentrations and mnesic performance. With this aim, we injected rats before the acquisition phase, with milrinone (a selective PDE3 inhibitor currently used in heart failure studies [49], with a half-life of 1.5–2.3 h [50], [51], 0.3 mg/kg), or rolipram (a selective PDE4 inhibitor with good brain penetration and a relatively short half-life of 1–3 h [52], 0.3 mg/kg) both alone and in combination or not with the selective 5-HT4R agonist RS 67333 (1 mg/kg) [17], [18]. The half-life of RS 67333 in the rat is not reported in the literature, but the data from behavioral studies [17], [18] suggest that this is comparable (1–3 hours) to those of the two PDE inhibitors.

Before or after either the sample or the testing phase, cAMP concentrations were measured in the hippocampus, prefrontal and perirhinal cortices. We show that a “pre-sample” early increase in cAMP levels followed by a specific lowering of cAMP concentrations in each brain sub-region linked to the object recognition paradigm support learning efficacy after a middle-term delay. Following the different treatments and at the end of the testing trial, we also examined in these brain sub-regions i) the PDE activities to validate efficiency of PDE3 or PDE4 inhibition by their respective inhibitor and ii) the phosphoprotein phosphatase (PP) type 2 activities since cAMP concentrations have been shown to be transiently up- or down-regulated by PP2A activation in various cell types [53][55]. In fact, as cAMP-PDE limit excessive cAMP production by catalysing its hydrolysis; PP1 and PP2 (accounting for more than 90% of total phosphatase activity in brain [56]) limit PDE-induced excessive catabolism of cAMP by reversing PP2A phosphorylation of phosphorylated PPE3B [57] or particulate PDE4 activities [58]. We also demonstrate here, that milrinone alters type PP2 activities in anatomical structures linked to object recognition memory in rat.

Materials and Methods

1. Subjects

A total of 172 adult male Sprague-Dawley rats (300–350 g, René Janvier, France) were used in these experiments. Rats were housed in groups of three in a temperature controlled room under a 12L∶12D cycle (lights on at 8:00 pm), with food and water provided ad libitum. All procedures were performed in conformity with National (JO 887–848) and European (86/609/EEC) legislations on animal experimentation. Behavioural procedures received approval from the Ethics Committee for Animal Experimentation of Normandy (Approval number 1009-01).

2. Behavioural experiments

2.1 Apparatus.

The apparatus consisted of an open-box (100×100×60 cm) made of wood with the inside painted in black. The objects to be discriminated were made of plastic, or glass (all 5 cm height) and were available in four copies. The objects were fixed (Patafix) on the floor in the box, to ensure that they could not be displaced by the rats.

2.2 Handling and habituation.

Rats were handled daily for one week prior to the study and then habituated to the apparatus and the test room. The first two days, rats were put together as a group of 3 to explore the empty arena for 10 min. On the third day, rats were put individually in the empty box for 3 min and the next two days, in the presence of an object that will not be used for the experimental task. Testing began on day 6.

2.3 Object recognition task.

Animals were tested in the object recognition task as described previously [15], [18]. The test session consisted of two phases with a duration of 3 min each on day 6. During the sample phase, each rat was placed in the box with two identical objects (placed close to the corners). After a delay of 4 h, during which the animal returned to its cage and both objects were replaced (one by its identical copy, the other by a new object in the same locations), the rat was returned to the box (testing phase). From rat to rat, the role (familiar or new object) as well as the relative position of the two objects were counterbalanced and randomly permuted.

The number of animals in each group was: saline-treated (n = 32), RS 67333 1 mg/kg (n = 32), milrinone 0.3 mg/kg (n = 27), milrinone+RS 67333 (n = 27), rolipram 0.3 mg/kg (n = 27), rolipram+RS 67333 (n = 27).

3. Drugs and drug administration

In all experiments, each rat was given an i.p injection of either saline (NaCl 0.9%) or RS 67333 (1 mg/kg) 30 minutes prior to the sample phase as previously described [18]. We have not tested other steps of memorization or lower doses of RS 67333 because i) RS6733-induced enhancement was reported only for the acquisition phase of information processing and ii) doses of 0.001 or 0.01 mg/kg were ineffective to enhance recognition memory [16], [17]. Milrinone (PDE3 inhibitor) or rolipram (PDE4 inhibitor) was injected each at the dose of 0.3 mg/kg i.p. 45 min prior to the sample phase. Higher doses of PDE inhibitors were not tested since at high dosage, milrinone can have vasodilatory and arrhythmogenic effects [59] and rolipram can have sedative side-effects [60], [61]. Efficiency of PDE3 or PDE4 inhibition was confirmed by specific PDE3 or 4 activity measures in hippocampus, prefrontal and perirhinal cortices at the end of the behavioural task. Moreover, since object recognition performance can only be determined if the animals show sufficient exploration [62], we concomitantly evaluated the exploration levels of the animals. In our experiment, a dose of 0.3 mg/kg rolipram or 0.3 mg/kg milrinone, given 45 min before the sample phase, resulted in a substantial decrease in locomotor activity but not in exploratory behaviour. A total of 148 rats were used to perform the object recognition task (tested animals), and 24 rats received the different injections, without being subjected to the behavioural task (untested animals).

4. Biochemical analysis

4.1 Cyclic AMP measurement.

Cyclic AMP extraction was performed according to a procedure adapted from Rodriguez [63]. Immediately after the testing phase or at the corresponding delay following the different injections (for untested animals), rats were subjected to euthanasia by decapitation without prior anesthesia. Intact brains were dissected on ice into prefrontal cortex, perirhinal cortex and hippocampus, taken systematically in this order and in less than 3 min following euthanasia. Brain sub-regions were rinsed with cold saline and dried. Each sub-region was homogenized in cold 100% ethanol in an ice bath and the homogenate centrifuged at 20,000 g for 15 min. The supernatant was recovered and, the pellet resuspended in 1 ml of 2∶1 ethanol∶water solution and centrifuged as before. The combined supernatants were evaporated to dryness in a 60°C bath under a stream of nitrogen gas. The final residue was dissolved in 0.5 ml of assay buffer (0.05 M sodium acetate, pH 5.8, containing sodium azide). Cyclic AMP levels were determined with a radioimmunoassay (Amersham). This assay measures the competitive binding of 3H-labeled cAMP to a cAMP-specific antibody.

4.2 Preparation of rat brain membranes and soluble fractions.

Sub-cellular fractionation of the brain regions was also performed immediately after euthanasia as detailed previously [18]. Briefly, each cerebral tissue was placed, immediately upon isolation, into ice-cold homogenization buffer (20 mMTris-HCl pH 7.2, 1 mM EDTA, 250 mM sucrose, supplemented with 0.1 mMphenylmethanesulfonyl fluoride, 2 mMbenzamidin, and a mixture of antiproteases (antipain, aprotinin, leupeptin, pepstatin A) at a final concentration of 1 microg/ml), homogenized by several passages through 25-G needle. Homogenates were centrifuged at 1,000 g, 4°C for 5 min and the supernatants decanted and centrifuged at 100,000 g, 4°C for 1 h. Each supernatant (soluble fraction) and the respective pellet (particulate fraction, re-suspended in ice-cold complete homogenization buffer) were then stored at −20°C. Protein content of each fraction was determined by the method of Bradford with BSA as a standard [64]. Purity of each subcellular fraction was assayed by both lactate deshydrogenase (soluble activity) and alkaline phosphatase (membrane-associated activities) as already reported [18].

4.3 PDE Assay.

Phosphodiesterase activities were assayed according to the two-step modified procedure of Thompson and Applemann [65] as already described [18]. To discriminate PDE2, PDE3 or PDE4 activities from other PDE activities, protein from each sample were incubated either in the absence (total PDE activities) or in the presence of specific inhibitors of each family: 20 µM erythro-9-(2-hydroxy-3-nonyl)-adenine (EHNA), 20 µM milrinone or 10 µM rolipram for PDE2, PDE3 and PDE4, respectively, according to their respective IC50 described elsewhere [18]. Differences between total and selective inhibitor-insensitive PDE activities were considered as corresponding PDE activities.

4.4 Phosphatase Assay.

Total PP2 activities in subcellular fractions of the different rat brain structures were determined by Serine/Threonine Phosphatase Assay (Promega, Charbonière-les-Bains, France) which used a specific substrate for PP2. Assays were conducted according to the manufacturer's procedure. Free phosphate was then quantified by a colorimetric method.

4.5 SDS-PAGE Western Blot Analysis.

Subcellular fraction protein from hippocampus, prefrontal and perirhinal cortices were boiled for 5 min and separated by 8% SDS-PAGE. The proteins were transferred onto a nitrocellulose membrane (1 h at 100 V and 4°C). Western blotting was then performed using an affinity-purified goat polyclonal antibody raised against a peptide that maps near the C-terminus of the human PDE4D (Santa Cruz Biotechnology). Immunoblotting with antibody that was pre-incubated with an excess of the peptide used for immunization (Santa Cruz Biotechnology) was performed as a negative control, following the instructions of the supplier. Immunoreactive bands were detected using a donkey anti-goat IgG-horseradish peroxidase (HRP) complex and an enhanced chemiluminescence (ECL) Advance Western Blotting Detection Kit (Amersham Biosciences). For β-actin detection, the blots were stripped in a stripping buffer that contained 62.5 mM Tris-HCl (pH 6.7) 2% SDS, and 100 mM β-mercaptoethanol at 58°C for 30 min, and reprobed for actin with monoclonal mouse anti-actin antibody and goat anti-mouse IgG-HRP (Calbiochem). The immunoblots were scanned on the ProXPRESS Proteomic Imaging System (Perkin Elmer Life Science, Boston, MA) and analyzed with the TotalLab Image Analysis software (Nonlinear Dynamics Ltd., Newcastle, UK).

5. Data scoring and analysis

5.1 Behavioural analysis.

The experimenter sat in front of the box. Total time spent exploring each object in both the sample and the testing phases were recorded. Exploration of an object was defined as follows: directing the nose to the object at a distance <2 cm. Overall exploration times across phases were analyzed by a two-way ANOVA (phase and treatment as factors) with repeated measures. For testing phase data, exploration of each object was analyzed using a two-way repeated-measurements ANOVA with object and treatment as factors. When appropriates, post-hoc testing was performed using Fisher's least significant difference (LSD) test. We calculated discrimination indexes as D1, which is the difference in time spent exploring the two objects in testing trial (i.e. time with novel object minus time with familiar object); and D2, the discrimination ratio, which is the difference in exploration time (D1) expressed as a ratio of the total time spent exploring the two objects in the testing trail (e.g. novel-familiar/novel+familiar). This ratio makes it possible to adjust for individual or group differences in the total amount of exploration time. Comparisons were made using one-way ANOVA with treatment as factor and post-hoc testing was performed using Fisher's least significant difference (LSD) test.

Locomotor activity was measured during the test session through videotaping. The arena was divided into 9 squares (32×32 cm). During each phase, the number of entries in each square was measured. Analysis was performed using two-way repeated-measurements ANOVA with entry and treatment as factors, followed by Fisher's LSD test when necessary.

5.2 Biochemical analysis.

After construction of a standard curve, cAMP levels were determined directly from the counts (in duplicate for each brain region of each animal) in nanomoles per milligram of tissue wet weight. PDE activities (in triplicates) were expressed in pmol of cAMP hydrolyzed per min and mg of protein. PP2 activities were expressed as nmol of phosphate released per min.

Statistical differences were determined through non-parametric tests adapted to small size data (Friedman and Kruskal-Wallis, followed by a post-hoc Mann-Whitney U-test; Sigma Stat software SPSS Inc, Chicago, IL).

Results

Hippocampus, prefrontal and perirhinal cortices exhibit different patterns of particulate PDE4D isoforms

Cyclic AMP-PDE was assayed in subcellular fraction from hippocampus, prefrontal and perirhinal cortices. Here, we confirmed our previous work [18] reporting that the total cAMP-PDE-hydrolysing activities of the particulate fraction from hippocampus, prefrontal and perirhinal cortices are mainly composed by PDE3 (38,2%, 34,8% and 43,4% respectively in hippocampus, prefrontal and perirhinal cortices) and PDE4 (26,7%, 43,4% and 21,4% respectively in hippocampus, prefrontal and perirhinal cortices) (data not shown). Since each PDE4D isoform plays specific roles on the cAMP concentration feedback [45], [46], we furthermore characterized by western blotting the pattern of PDE4D isoforms present in these brain sub-region (Fig. 1). We demonstrated that PDE4D protein expression differed according to the subcellular fraction and the brain sub-region. In the particulate fraction from prefrontal cortex, the presence of nine immunoreactive proteins suggests that all nine PDE4D isoforms (i.e. PDE4D1 to PDE4D9) are expressed whereas particulate fraction of hippocampus did not exhibit PDE4D6 and of perirhinal cortex neither particulate PDE4D8/9 nor particulate PDE4D3. Nevertheless, in the particulate fraction from hippocampus, prefrontal or perirhinal cortices, both the short PDE4D1 and the long PDE4D4 isoforms are the mainly PDE4D isoforms expressed. Finally, PDE4D1, PDE4D2, PDE4D4 and PDE4D6 were the isoforms revealed in the soluble fraction from prefrontal or perirhinal cortices. A similar panel of PDE4D isoform was revealed in the soluble fraction from the hippocampus, except that no immunoreactive band matches with PDE4D2. As reflected by the densitometric analyses (right panel of Fig. 1), the patterns of the putative PDE4D isoforms did not display any significant structure-related differences.

thumbnail
Figure 1. Expression of PDE4D proteins in the particulate and soluble fractions of rat hippocampus, prefrontal and perirhinal cortices.

Particulate and soluble fractions from the rat hippocampus, the prefrontal cortex and perirhinal cortex were isolated and proteins extracted as described in Materials and Methods section. The left panel shows representative immunoblots of particulate (25 µg) and soluble (25 µg) protein fractions probed with goat polyclonal human anti-PDE4D antibody in the hippocampus, prefrontal and perirhinal cortices. Arrowheads indicate the molecular weights of the immunoreactive proteins. The right panel shows quantification; the intensities of the immunoreactive bands in the particulate and soluble fractions from hippocampus, prefrontal and perirhinal cortices were determined and normalized to those of actin. The densitometry values are the mean ± SEM (n = 3).

https://doi.org/10.1371/journal.pone.0032244.g001

RS 67333 enhances particulate PDE3 activity from the perirhinal cortex in rat

To identify which cAMP-PDE family support the rolipram-insensitive PDE activities increased in the perirhinal cortex following the selective activation of 5-HT4R (RS 67333; [18]), rats were injected with a saline solution or RS 67333 (1 mg/kg, i.p.) before the object recognition paradigm (inter-phase interval of 4 h). Immediately after the testing phase, rats were subjected to euthanasia and PDE activities were assayed. As shown in Fig. 2, RS 67333 elevated PDE3 activities by 71% (P<0.01) in the perirhinal cortex lightening this family as a key regulator of cAMP concentration in this structure linked to object recognition. No variation was measured in the supernatant fraction (data not shown).

thumbnail
Figure 2. Effect of RS 67333 (1 mg/kg, i.p.) on PDE activities in the perirhinal cortex from rats performing the object recognition task with a 4-h delay.

Rats were injected with saline or the 5-HT4 receptor agonist RS 67333 (1 mg/kg, i.p.), 30 minutes before exposure to the sample trial of he object recognition task. Immediately after the testing trial, particulate fractions from the hippocampus, prefrontal and perirhinal cortices were isolated and assayed for EHNA- (20 µM), milrinone- (20 µM) and rolipram- (10 µM) sensitive PDE activities, respective inhibitors of PDE2, PDE3 and PDE4 families. PDE inhibitor-sensitive and –insensitive PDE activities were each expressed as pmolcAMPhydrolysed/min/mg protein. Results are means ± SEM of four independent subcellular fractionations performed in triplicate. Within each subcellular compartment, * indicates a significant difference of PDE activity as compared with saline treatment within a family of PDE activity (PDE2, 3, 4 or other PDE) (**, P<0.01, ANOVA followed by Fisher's LSD test).

https://doi.org/10.1371/journal.pone.0032244.g002

5-HT4 receptor stimulation, PDE3- or PDE4-inhibition improve familiar object recognition after a 4-h delay in rat

Rats were then injected with milrinone (PDE3 inhibitor, 0.3 mg/kg, i.p.), rolipram (PDE4 inhibitor, 0.3 mg/kg, i.p.) and/or the selective 5-HT4R agonist RS 67333 (1 mg/kg, i.p.) before the object recognition paradigm sample phase.

We first validate the efficiency of the treatments with PDE inhibitors; rats were immediately subjected to euthanasia after the testing phase, and PDE activities from the hippocampus, prefrontal and perirhinal cortices, were assessed. We especially measured PDE3 activity for milrinone-treated animals (Fig. 3) and PDE4 activity for rolipram-treated animals (Fig. 4). Concerning the measurements of particulate PDE3 activities in milrinone-treated rats (Fig. 3), we showed that PDE3 activity was inhibited in the hippocampus (−30%, P<0.01) and prefrontal cortex (−63%, P<0.001), but not in the perirhinal cortex, when compared to saline-treated rats. However, pre-treatment of rats by milrinone before RS 67333 prevented the RS 67333-induced increase in particulate PDE3 in the perirhinal cortex. Finally, milrinone did not affect significantly cAMP-PDE activities supported by other families than PDE3. As illustrated in Fig. 4, in rolipram-treated rats, particulate PDE4 activity was lower than in saline-treated rats, in the hippocampus (−60%, P<0.05) and the prefrontal (−42%, P<0.05) cortex, while tend to be lower in the perirhinal cortex (−26%). Similar decrease when compared to RS 67333-treated group was also observed for rolipram+RS 67333-treated animals in the prefrontal cortex (−42%, P<0.05) and, despite no significant, in the perirhinal cortex (−47%).

thumbnail
Figure 3. Milrinone (0.3 mg/kg, i.p.) specifically inhibits PDE3 activities in hippocampus, prefrontal cortex and perirhinal cortex from rats.

Rats were injected with the PDE3 inhibitor (milrinone, 0.3 mg/kg, i.p.) and then with saline or the 5-HT4 receptor agonist (RS 67333, 1 mg/kg, i.p.), respectively 45 minutes and 30 minutes before the sample phase of the object recognition task. Immediately after the testing phase, both particulate and soluble fractions from the hippocampus, the prefrontal cortex and perirhinal cortex were isolated and particulate fraction was assayed for milrinone (20 µM)-sensitive PDE activities. Milrinone-sensitive and –insensitive PDE activities were expressed as pmolcAMPhydrolysed/min/mg protein. Results are means ± SEM of four independent subcellular fractionations performed in triplicate. Within each subcellular compartment, * indicated significant differences of PDE activity as compared with other treatment within a type of PDE activity (PDE3 or other PDE) (*, P<0.05, **, P<0.01, ***, P<0.01, ANOVA followed by Fisher's LSD test).

https://doi.org/10.1371/journal.pone.0032244.g003

thumbnail
Figure 4. Rolipram (0.3 mg/kg, i.p.) specifically inhibits PDE4 activities in hippocampus, prefrontal cortex and perirhinal cortex from rats.

Rats were injected with the PDE4 inhibitor (rolipram, 0.3 mg/kg), and then with saline or the 5-HT4 receptor agonist (RS 67333, 1 mg/kg), respectively 45 minutes and 30 minutes before the sample phase of the object recognition task. Immediately after the testing phase, both particulate and soluble fractions from the hippocampus, the prefrontal cortex and perirhinal cortex were isolated and the particulate fraction was assayed for rolipram (10 µM)-sensitive PDE activities. Rolipram-sensitive and –insensitive PDE activities were expressed as pmolcAMPhydrolysed/min/mg protein. Results are means ± SEM of four independent subcellular fractionations performed in triplicate. Within each subcellular compartment, * indicated significant differences of PDE activity as compared with other treatment within a type of PDE activity (PDE4 or other PDE) (*, P<0.05, **, P<0.01, ANOVA followed by Fisher's LSD test).

https://doi.org/10.1371/journal.pone.0032244.g004

As shown in Table 1, all rats spent a similar total time exploring both objects during either the sample or the testing phase after a 4-h delay (P>0.05). Comparison of locomotor activities revealed an overall significant effect of treatment during the sample phase (F (5, 112) = 9.25, P<0.001). Post hoc analyses showed that rolipram- or rolipram+RS 67333-treated rats had a smaller number of entries compared to saline-treated (respectively P<0.001, P<0.01), RS 67333-treated (P<0.001), milrinone-treated (respectively P<0.001, P<0.05), milrinone+RS 67333- treated animals (respectively P<0.001, P<0.01) (Table 2). Finally, we found that milrinone-treated animals explore less than RS 67333-treated ones (P<0.01). Analysis of the testing phase revealed no significant treatment effect on the exploratory behaviour.

thumbnail
Table 1. Time of exploration of objects measured during the sample and the testing trails in the object recognition task.

https://doi.org/10.1371/journal.pone.0032244.t001

thumbnail
Table 2. Locomotor activity measured during the sample and the testing trails in the object recognition task.

https://doi.org/10.1371/journal.pone.0032244.t002

The repeated-measures ANOVA revealed i) for saline-treated rats, no significant difference of novel object exploration time (Fig. 5); ii) for drug-treated rats, both an overall significant effect of time spent exploring each object (F(1, 112 = 109.3, P<0.001) and an interaction between time exploring each object and treatment (F 5, 122) = 5.4, P<0.001). Post hoc analyses showed that all drug-treated rats significantly spent more time exploring the novel object, when compared to saline-treated rats (RS 67333-treated, P<0.001, rolipram-treated, P<0.001; rolipram+RS 67333-treated, P<0.01, P<0.001; milrinone-treated, P<0.001; milrinone+RS 67333-treated, P<0.05) (Fig. 5). This result is also confirmed by analysis of discrimination indexes (Table 3). ANOVA performed on D1 and D2 showed a significant treatment effect [for both D1 and D2: (F(5, 112) = 4.5, P<0.001)]. Post-hoc analysis revealed that all treated animals had a greater discrimination index (D1) compared to saline-treated animals (RS 67333-treated (P<0.001), rolipram-treated (P<0.001), rolipram+RS 67333 treated (p<0.01), milrinone-treated (P<0.01) milrinone+RS 67333- treated animals (P<0.05). These results are also confirmed by post-hoc analysis of the discrimination ratio (D2), compared to saline-treated rats (RS 67333-treated (P<0.001), rolipram-treated (P<0.001), rolipram+RS 67333 treated (p<0.05) milrinone-treated (P<0.01) milrinone+RS 67333- treated animals (P<0.05).

thumbnail
Figure 5. Object recognition task after a 4 h-delay in saline, rolipram- or milrinone- treated rats with or without RS 67333 co-treatment.

Time of exploration of the familiar and novel objects during the testing phase of the object recognition memory task of saline (n = 32), RS 67333 (n = 32), rolipram (n = 27), rolipram+RS 67333 (n = 27), milrinone (n = 27), or milrinone+RS 67333 (n = 27)-treated rats. Rats were injected with the PDE inhibitor (0.3 mg/kg, i.p.) solution 45 minutes before the sample phase and with saline or RS 67333 (1 mg/kg, i.p.) solution 30 minutes before the sample phase. Object recognition was assayed after a 4 h-delay. Values are means in s ± SEM. NS: non significant, (*) indicates a significant difference in comparison with saline treatment. (*, P<0.05, **, P<0.01, ***, P<0.001, ANOVA followed by Fisher's PLSD test).

https://doi.org/10.1371/journal.pone.0032244.g005

thumbnail
Table 3. Value of the index measures of discrimination in the object recognition task.

https://doi.org/10.1371/journal.pone.0032244.t003

Familiar object recognition is associated with a “pre-sample” early increase in cAMP levels in hippocampus, prefrontal and perirhinal cortices

To further characterize the cellular mechanisms involved after a 4 h-delay, rats were subjected to euthanasia before or after the sample or the testing phase of the paradigm, and cAMP was measured in the anatomical structures linked to the object recognition task (i.e. hippocampus, prefrontal and perirhinal cortices) (Fig. 6).

thumbnail
Figure 6. Rolipram and/or RS 67333 induce a biphasic modulation of cAMP concentrations in the hippocampus, prefrontal and perirhinal cortices of rats performing an object recognition task with a 4-h delay.

Rats were injected with the inhibitor of PDE4 (rolipram, 0.3 mg/kg, i.p.) and then with saline or the 5-HT4 receptor agonist (RS 67333, 1 mg/kg, i.p.), respectively 45 minutes and 30 minutes before to the sample phase of the object recognition task. Rats were euthanized before or after the sample phase, or before or after the testing phase. Cyclic AMP was extracted from the hippocampus, prefrontal and perirhinal cortices and then assayed. Cyclic AMP was expressed as pmolcAMP/mg of weight tissue. Results were means ± SEM of three independent extractions performed in duplicate. (0) indicated significant differences in comparison with other steps of the paradigm in each brain sub-region, Mann-Whitney test, P<0.05. (*) indicated a significant difference in comparison with saline treatment in each brain sub-region. (0,*, P<0.05).

https://doi.org/10.1371/journal.pone.0032244.g006

As illustrated in Fig. 6, cAMP concentrations measured before the sample phase (white bars), demonstrate, except in the hippocampus of rolipram treated-rats, the efficiency of RS 67333 (1 mg/kg, i.p.), rolipram (0.3 mg/kg, i.p.) and milrinone (0.3 mg/kg, i.p., data not illustrated for more readability) to increase cAMP levels in the three brain structures studied, i.e. hippocampus (∼1.3 fold), prefrontal (∼1.5 fold) and perirhinal cortices (∼1.6 fold) when compared to saline-treated rats (Mann-Whitney test versus saline group, P<0.05).

RS 67333, milrinone and rolipram treatments prevent the sample phase-induced increase in cAMP levels in the rat central nervous system

Analysis of the cAMP concentrations reveal that, in saline-treated rats, the sample phase induces an increase in cAMP levels in all brain regions studied (Fig. 6, grey bars versus white bars; ∼1.8 fold, ∼1.3 fold and ∼1.3 fold for the hippocampus, prefrontal and perirhinal cortices, respectively; Mann-Whitney test, P<0.05). Such a sample phase-induced increase could not be observed for the drug-treated animals; a tendency to a decreased level of cAMP was even noticed in the prefrontal cortex of these animals (∼0.86 to 0.62 fold according to drug treatment, Fig. 6) except those treated with milrinone+RS67333.

Drug treatments lower cAMP levels in hippocampus, prefrontal cortex and perirhinal cortex after the object recognition test

After the testing phase (black bars, Fig. 6) when compared with the situation before the testing phase (hatched bar), cAMP concentrations in saline –treated rats were once more increased in both the hippocampus (2.76 fold, P<0.05) and the perirhinal cortex (2.10 fold, P<0.05) but not in the prefrontal cortex. Nevertheless cAMP concentrations in the prefrontal cortex from saline-treated animals still tend to be higher than before the sample phase (P>0.05). Thus, between the beginning and the end of the paradigm, we reported a 4.1, 1.3 and 3.2 fold increase in cAMP concentrations, respectively in the hippocampus, prefrontal cortex and perirhinal cortex from the saline group (Mann-Whitney test, respectively P<0.05; P>0.05 and P<0.05).

In the drug-treated animals, minor increases in cAMP levels (lower than in the saline-treated group) were also reported after the testing phase. Both rolipram and milrinone alone or administered before RS 67333 induced an almost 1.4 fold increase in cAMP concentrations in the hippocampus between the beginning and the end of the paradigm (P<0.05). Similarly, in the perirhinal cortex, a 1.6 fold increase of cAMP levels in the rolipram group (Fig. 6, P<0.05) and a 1.5 fold increase in the milrinone group (P<0.05) were reported after the testing phase. Finally, in the prefrontal cortex from drug-treated rats, cAMP levels still tend to decrease (∼−8 to −22% according to drug treatment).

At the end of the paradigm, cAMP concentrations from rats injected with a PDE inhibitor alone or in combination with RS 67333 were markedly lower when compared to saline-treated rats in the hippocampus (−54 to −61% according to the treatment, P<0.05), the prefrontal (−13 to −31% according to the treatment) or perirhinal cortices (−13 to −35% according to the treatment) (Mann-Whitney test versus saline group, P<0.05).

Milrinone altered PP2 activities in anatomical structures linked to object recognition memory in rat

We assessed PP2 activities in both subcellular compartments (soluble and particulate) of the hippocampus, prefrontal and perirhinal cortices since excessive cAMP catabolism is limited by reversing phosphorylation of particulate PDE4 activities [53][55]. PP2 activity was measured in both the soluble and particulate fraction (Table 4); PP2 activity was however mainly present in the soluble fraction (68.1±2.1% to 75.3±1.7% of total PP2 activity according to the structure considered). Milrinone significantly altered PP2 activities in the soluble fraction of the hippocampus (Fig. 7a) and perirhinal cortex (Fig. 7c) as well as the soluble and particulate fractions of the prefrontal cortex (Fig. 7b). In fact, a slight decrease was evidenced in the soluble fraction of the hippocampus from RS 67333-treated rats (−20%, P<0.01, Fig. 7a). Thus, in milrinone-treated rats, soluble PP2 activities decrease (−44% when compared to saline rats, P<0.001) was significantly strengthened (P<0.01) when rats were also injected with RS 67333 (PP2 activities was diminished by −70% when compared to saline rats, P<0.001). Similar observations were done in the prefrontal cortex of milrinone-treated rats (Fig. 7b), in which PP2 activities decrease in the soluble fraction (−53% when compared to saline rats, P<0.001) was significantly strengthened (P<0.01) when rats were also injected with RS 67333 (PP2 activities was diminished by −79% when compared to saline rats, P<0.001). PP2 activities of the particulate compartment were altered in milrinone-treated rat (−79%, P<0.001) but this decrease was not strengthened in milrinone+RS 67333 treated animals. Finally, milrinone affected soluble PP2 activities in the perirhinal cortex, when administrated alone or in association with RS 67333 (−75% when compared to saline-treated rats, P<0.001; Fig. 7c).

thumbnail
Figure 7. Effect of milrinone (0.3 mg/kg) on PP2 activities in hippocampus, prefrontal and perirhinal cortices from rats performing the object recognition task with a 4-h delay.

Rats were injected with the inhibitor of PDE3 (milrinone, 0.3 mg/kg) 45 minutes before exposure then with saline or the 5-HT4 receptor agonist (RS 67333, 1 mg/kg), 30 minutes before exposure to the sample trial of the object recognition task. Immediately after the testing trial, both particulate (white bar) and soluble (black bar) fractions from the hippocampus (a), the prefrontal cortex (b) and perirhinal cortex (c) were isolated and were assayed for PP2 activity. PP2 activities were pmol of phosphate released by min and mg protein. Results are means ± SEM of four independent subcellular fractionations performed in triplicate. Within each subcellular compartment, # indicated significant differences of PP2 activity as compared with other treatment (#, P<0.05, ##, P<0.01, ###, P<0.001, ANOVA followed by Fisher's LSD test).

https://doi.org/10.1371/journal.pone.0032244.g007

thumbnail
Table 4. Distribution of the total PP2 activities between particulate and soluble fractions from rat hippocampus, prefrontal cortex and perirhinal cortex.

https://doi.org/10.1371/journal.pone.0032244.t004

Such modifications of PP2 activities could not be evidenced in rolipram-treated animals (data not shown).

Discussion

Object recognition memory in rodents and primates is thought to be mediated, at least in part, by interactions between the perirhinal cortex, prefrontal cortex and hippocampus [66][70]. Interestingly, 5-HT4R, known to be involved in learning and memory [31], including acquisition of information [18] are widely distributed among these brain structures [71][73]. Since 5-HT4R stimulation induces an increase in cAMP that has been associated to memory processes [6], [8], [9], [11], we first hypothesized that the drug-induced increase in cAMP might support the improvement of object recognition memory performance. Data presented here strongly suggest that the “pre-sample” early increase in cAMP levels followed by a specific lowering of cAMP concentrations in each brain sub-region involved to the object recognition paradigm improve learning efficacy after a middle-term delay.

We first confirmed the major role of PDE3 and PDE4 in the control of cAMP levels in the anatomical structures linked to the object recognition task [18]. Indeed, we reported that both the stimulation of cAMP production (RS 67333) and the inhibition of its hydrolysis (milrinone, PDE3 inhibitor or rolipram, PDE4 inhibitor) in the rat, improve familiar object recognition after a 4-h delay. Besides, we observed similar effects of PDE3 and PDE4 inhibitors. In line with previous studies, we reported a higher sensitivity of PDE3- than PDE4-regulated cAMP pools to PKA activation, associated to a lower efficacy of PDE3 to hydrolyze cAMP [74][77]. We hypothesized this familiar object recognition improvement to be linked to the early cAMP levels increase measured before the sample phase in the hippocampus, prefrontal and perirhinal cortices from these animals. Early cAMP activations in the prefrontal cortex have already been described to be beneficial for working memory under conditions that require hippocampal-prefrontal cortex interactions [14], [78]. Moreover, the observation that the drug-induced early increase in cAMP in the brain sub-regions improves the mnesic trace is consistent with data reporting that activation of the cAMP-PKA pathway cascade improves memory processes [2], [4], [6], [8], [9] unlike inhibition of PKA [6], [10], [11], [79]. Indeed, according to the model of Frankland and Bontempi, experience is both initially encoded in hippocampal and cortical networks. Subsequent reactivation of the hippocampal network reinstates activity in different cortical networks. This coordinated replay across hippocampal–cortical networks leads to gradual strengthening of cortico-cortical connections, progressively disengaging the memory trace from the hippocampus [80]. Thus, the higher cAMP levels induced by drug treatments could support a better acquisition of the mnesic trace that will, in turn, benefit to the animal during the testing phase.

We also observed that drug treatments induce a lowering, and/or a reduction of the awaited increases, in cAMP levels in all brains regions studied after the testing phase, when compared to saline injected animals. Hence, cAMP concentrations were systematically lower in both the hippocampus and perirhinal cortex from animals that have increased behavioural performances than in the saline-treated rats that exhibit poor object recognition performances. Appearing at a first glance as a discrepancy from data from the literature, this situation can easily be reconciled by recent studies suggesting that memory requires a restricted, or selective cAMP production rather than a large and widespread increase in cAMP levels [28], [81]. Indeed, Kelly and co-workers (2008) observed an impairment of memory consolidation and/or retrieval in a fear-conditioning task in mice that express a constitutively active isoform of the G-protein subunit Gαs in the forebrain [81]. Perez-Garcia and Meneses, (2008) also demonstrate that the hippocampal production of cAMP was higher in untrained rats than in rats subjected to a behavioral task [28]. In this respect, low cAMP levels might be optimal to convert temporary memory during acquisition to long-term memory (4 h-delay) while high cAMP levels might disturb such a conversion of short-term memory to long-term memory, resulting in low performances in controls animals.

These few elements point out the complexity of cAMP-dependent responses and the putative interactions between behaviour- and drug-induced effects on cellular signaling. By rapidly degrading cAMP from selected compartments, PDEs can fix the boundaries for cAMP diffusion, shape the intracellular gradients of the second messenger and thereby modulate defined sets of PKA-mediated intracellular events. Hence, PDE alteration may affect cAMP compartmentalization, leading to untargeted cAMP signals, aberrant phosphorylation of target proteins and thus contribute to dysfunction. We report here that hippocampus, prefrontal and perirhinal cortices exhibit different patterns of particulate PDE4D isoforms. One can therefore hypothesize differential implications of PDE isoforms, keys mediators of memory and learning processes in the limitation of cAMP increase [36], [37], [82], [83]. For example, PDE4D8 found only in the particulate fraction from the hippocampus or the prefrontal cortex, has been shown to be responsible for controlling local cAMP concentrations and PKA activity in the vicinity of β1 adrenergic receptors [46]. PDE4D3 that we only described in the particulate fraction from the hippocampus or the prefrontal cortex, was reported to bind to muscle- specific A-kinase anchoring protein (mAKAP), which in turns controls perinuclear AMP levels and recruits the MAP kinases MEK5 and ERK5 [84]. Hence, each PDE isoform plays a critical role in the specificity of cAMP-signaling, effectively creating cyclic nucleotide microdomains and/or cAMP gradients that can be sensed by the cell [46], [85][88]. Accordingly, some PDE4D isoforms precisely regulate the coupling between GPCR and the Gs protein. A desensitization of 5-HT4R, through an over-stimulation by RS67333 and/or an alteration of PDE, could thus lead to a lowering of cAMP concentrations in brain regions expressing 5-HT4R. Indeed, PKA activated after a 5-HT4R stimulation, will phosphorylate 5-HT4R leading to cell membrane recruitment of GRK2, which in turn will phosphorylate i) the associated GPCR [89] inhibiting its coupling with Gs, and ii) PDE4, which locally attenuates the PKA activity by lowering local cAMP levels [90][92].

Effects induced by 5-HT4R stimulation are time-limited since PKA phosphorylation of 5-HT4R, GRK2 or PDE can be reversed by phosphatase (PP) activity [58]. PP1 and PP2, compartmentalized inside the mammalian cell [93], [94], account for the major phosphatase activities [56]; PP2 is however more particularly investigated because of its ability to dephosphorylate many signaling proteins [93]. Since PP2A is activated by cAMP level increases [53][55], we suggested that phosphatase activity may have been raised in the brain sub-region structures from drug-treated rats, but it is not what we found here. While inhibition of PDE4 activity failed to alter PP2 activity, milrinone administration induced an alteration of PP2 activity, especially in the supernatant fractions from the brain regions investigated; such an alteration could in turn alter dephosphorylation of the 5-HT4R, prevent the efficiency of coupling between this receptor and the Gs protein, and thus lead to a lowered cAMP production. Such a functional contrast following selective inhibition of PDE3 and PDE4 has been already observed in many cell types [95][97]. Interestingly, the cell membrane recruited PDE4 also desensitizes the switched coupling of the β2AR to activation of Gi induced by the PKA-mediated phosphorylation [98], defining thus an appropriate coupling of the GPCR [99]. Hence, in our opinion, drugs injected before the sample phase rapidly increase cAMP levels leading to the uncoupling of 5-HT4R. However, during the test phase, further 5-HT4R stimulation does not raise cAMP level, probably because of 5-HT4R uncoupling. 5-HT4R uncoupling could thus be an adaptative mechanism to reduce cAMP levels in the presence of an excessive stimulation of 5-HT4R or an absence of PDE3 or 4 activities thus avoiding an excessive accumulation of cAMP. If such threshold of cAMP level exists and is reached by either the stimulation of 5-HT4R or inhibition of PDE3 or PDE4 alone, thus no further improvement of memory performance could be induced by the pharmacological treatments by the combination of RS 67 333 and PDE inhibitor.

Finally, early increases in cAMP levels followed by an immediate drop in cAMP concentrations have already been well described in cell differentiation, particularly in Sertoli cells [58]. Indeed, before the cAMP increase, stimulation of Sertoli cells by gonadotropin leads to an activation of the ERK pathway, while following the peak of cAMP, gonadotropin activates the PKA pathway. Interestingly, ERK pathway could prolong activation of the cAMP signaling system in cells by having both short and long term effects on PDE4D activity by respectively inactivating long PDE4D isoform (the ones to exhibits a site that allows phosphorylation by ERK) and altering PDE4D mRNA stability (for review [99]). Hence, by analogy to differentiation mechanisms, another hypothesis is that “cellular learning” may result from the crossing of a milestone, resulting in the subsequent activation of alternative intracellular signalling pathways. Increases in cAMP levels but also their subsequent declines account in mnesic performance improvement. The part of ERK pathway in these processes should be addressed in furthers works.

Our results show that a “pre-sample” early increase in cAMP levels followed by both a “post-sample” lowering of cAMP concentrations in the prefrontal cortex and a “post-test” lowering of cAMP concentrations in the hippocampus and perirhinal cortex support improved learning efficacy after a middle-term delay. If cAMP triggers a temporally defined cellular response, a major question that should be addressed in future works is to clarify how such a functionally ubiquitous signaling pathway may be involved in memory formation.

Acknowledgments

The authors thank Dr. S. Carreau (Laboratoire Œstrogènes et Reproduction, EA 2608, INRA USC 2006, University of Caen) and Dr. P. Barbey (LAMARE) for giving us access to the ultracentrifuge and the radioactivity laboratory, respectively, as well as Pr. B. Cox and Dr M. Dacher (Uniformed Service University, Bethesda, MD, USA) for english editing of the manuscript.

Author Contributions

Conceived and designed the experiments: MH GL FD MB. Performed the experiments: MH GL. Analyzed the data: MH TF FD MB GL. Contributed reagents/materials/analysis tools: MH TF GL. Wrote the paper: TF FD MB GL MH. Statistical analysis: TF.

References

  1. 1. Monti B, Berteotti C, Contestabile A (2006) Subchronic rolipram delivery activates hippocampal CREB and arc, enhances retention and slows down extinction of conditioned fear. Neuropsychopharmacology 31: 278–286.
  2. 2. Hoyer D, Pompe B, Friedrich H, Zwiener U, Baranowski R, et al. (2004) Autonomic Information Flow during awakeness, sleep, and multiple organ dysfunction syndrome assessed by mutual information function of heart rate fluctuations. Conf Proc IEEE Eng Med Biol Soc 1: 628–630.
  3. 3. Bach ME, Barad M, Son H, Zhuo M, Lu YF, et al. (1999) Age-related defects in spatial memory are correlated with defects in the late phase of hippocampal long-term potentiation in vitro and are attenuated by drugs that enhance the cAMP signaling pathway. Proc Natl Acad Sci U S A 96: 5280–5285.
  4. 4. Barad M, Bourtchouladze R, Winder DG, Golan H, Kandel E (1998) Rolipram, a type IV-specific phosphodiesterase inhibitor, facilitates the establishment of long-lasting long-term potentiation and improves memory. Proc Natl Acad Sci U S A 95: 15020–15025.
  5. 5. Zhang HT, O'Donnell JM (2000) Effects of rolipram on scopolamine-induced impairment of working and reference memory in the radial-arm maze tests in rats. Psychopharmacology (Berl) 150: 311–316.
  6. 6. Bernabeu R, Cammarota M, Izquierdo I, Medina JH (1997) Involvement of hippocampal AMPA glutamate receptor changes and the cAMP/protein kinase A/CREB-P signalling pathway in memory consolidation of an avoidance task in rats. Braz J Med Biol Res 30: 961–965.
  7. 7. Bernabeu R, Schmitz P, Faillace MP, Izquierdo I, Medina JH (1996) Hippocampal cGMP and cAMP are differentially involved in memory processing of inhibitory avoidance learning. Neuroreport 7: 585–588.
  8. 8. Prickaerts J, de Vente J, Honig W, Steinbusch HW, Blokland A (2002) cGMP, but not cAMP, in rat hippocampus is involved in early stages of object memory consolidation. Eur J Pharmacol 436: 83–87.
  9. 9. Rutten K, Prickaerts J, Hendrix M, van der Staay FJ, Sik A, et al. (2007) Time-dependent involvement of cAMP and cGMP in consolidation of object memory: Studies using selective phosphodiesterase type 2, 4 and 5 inhibitors. Eur J Pharmacol 558: 107–112.
  10. 10. Bourtchouladze R, Abel T, Berman N, Gordon R, Lapidus K, et al. (1998) Different training procedures recruit either one or two critical periods for contextual memory consolidation, each of which requires protein synthesis and PKA. Learn Mem 5: 365–374.
  11. 11. Taylor JR, Birnbaum S, Ubriani R, Arnsten AF (1999) Activation of cAMP-dependent protein kinase A in prefrontal cortex impairs working memory performance. J Neurosci 19: RC23.
  12. 12. Connolly DA, Hockley WE, Pratt MW (1996) A developmental evaluation of frequency memory for actions presented in lists, scripts, and stories. Memory 4: 243–263.
  13. 13. Pineda VV, Athos JI, Wang H, Celver J, Ippolito D, et al. (2004) Removal of G(ialpha1) constraints on adenylyl cyclase in the hippocampus enhances LTP and impairs memory formation. Neuron 41: 153–163.
  14. 14. Ramos BP, Birnbaum SG, Lindenmayer I, Newton SS, Duman RS, et al. (2003) Dysregulation of protein kinase a signaling in the aged prefrontal cortex: new strategy for treating age-related cognitive decline. Neuron 40: 835–845.
  15. 15. Ennaceur A, Delacour J (1988) A new one-trial test for neurobiological studies of memory in rats. 1: Behavioral data. Behav Brain Res 31: 47–59.
  16. 16. Lamirault L, Guillou C, Thal C, Simon H (2003) Combined treatment with galanthaminium bromide, a new cholinesterase inhibitor, and RS 67333, a partial agonist of 5-HT4 receptors, enhances place and object recognition in young adult and old rats. Prog Neuropsychopharmacol Biol Psychiatry 27: 185–195.
  17. 17. Lamirault L, Simon H (2001) Enhancement of place and object recognition memory in young adult and old rats by RS 67333, a partial agonist of 5-HT4 receptors. Neuropharmacology 41: 844–853.
  18. 18. Levallet G, Hotte M, Boulouard M, Dauphin F (2009) Increased particulate phosphodiesterase 4 in the prefrontal cortex supports 5-HT4 receptor-induced improvement of object recognition memory in the rat. Psychopharmacology (Berl) 202: 125–139.
  19. 19. Moser PC, Bergis OE, Jegham S, Lochead A, Duconseille E, et al. (2002) SL65.0155, a novel 5-hydroxytryptamine(4) receptor partial agonist with potent cognition-enhancing properties. J Pharmacol Exp Ther 302: 731–741.
  20. 20. Fontana DJ, Daniels SE, Wong EH, Clark RD, Eglen RM (1997) The effects of novel, selective 5-hydroxytryptamine (5-HT)4 receptor ligands in rat spatial navigation. Neuropharmacology 36: 689–696.
  21. 21. Galeotti N, Ghelardini C, Bartolini A (1998) Role of 5-HT4 receptors in the mouse passive avoidance test. J Pharmacol Exp Ther 286: 1115–1121.
  22. 22. Lelong V, Dauphin F, Boulouard M (2001) RS 67333 and D-cycloserine accelerate learning acquisition in the rat. Neuropharmacology 41: 517–522.
  23. 23. Letty S, Child R, Dumuis A, Pantaloni A, Bockaert J, et al. (1997) 5-HT4 receptors improve social olfactory memory in the rat. Neuropharmacology 36: 681–687.
  24. 24. Marchetti E, Dumuis A, Bockaert J, Soumireu-Mourat B, Roman FS (2000) Differential modulation of the 5-HT(4) receptor agonists and antagonist on rat learning and memory. Neuropharmacology 39: 2017–2027.
  25. 25. Marchetti E, Jacquet M, Jeltsch H, Migliorati M, Nivet E, et al. (2008) Complete recovery of olfactory associative learning by activation of 5-HT4 receptors after dentate granule cell damage in rats. Neurobiol Learn Mem 90: 185–191.
  26. 26. Meneses A (2007) Stimulation of 5-HT1A, 5-HT1B, 5-HT2A/2C, 5-HT3 and 5-HT4 receptors or 5-HT uptake inhibition: short- and long-term memory. Behav Brain Res 184: 81–90.
  27. 27. Orsetti M, Dellarole A, Ferri S, Ghi P (2003) Acquisition, retention, and recall of memory after injection of RS67333, a 5-HT(4) receptor agonist, into the nucleus basalis magnocellularis of the rat. Learn Mem 10: 420–426.
  28. 28. Perez-Garcia G, Meneses A (2008) Ex vivo study of 5-HT(1A) and 5-HT(7) receptor agonists and antagonists on cAMP accumulation during memory formation and amnesia. Behav Brain Res 195: 139–146.
  29. 29. Restivo L, Roman F, Dumuis A, Bockaert J, Marchetti E, et al. (2008) The promnesic effect of G-protein-coupled 5-HT4 receptors activation is mediated by a potentiation of learning-induced spine growth in the mouse hippocampus. Neuropsychopharmacology 33: 2427–2434.
  30. 30. Terry AV Jr, Buccafusco JJ, Jackson WJ, Prendergast MA, Fontana DJ, et al. (1998) Enhanced delayed matching performance in younger and older macaques administered the 5-HT4 receptor agonist, RS 17017. Psychopharmacology (Berl) 135: 407–415.
  31. 31. King MV, Marsden CA, Fone KC (2008) A role for the 5-HT(1A), 5-HT4 and 5-HT6 receptors in learning and memory. Trends Pharmacol Sci 29: 482–492.
  32. 32. Egawa T, Mishima K, Matsumoto Y, Iwasaki K, Fujiwara M (1997) Rolipram and its optical isomers, phosphodiesterase 4 inhibitors, attenuated the scopolamine-induced impairments of learning and memory in rats. Jpn J Pharmacol 75: 275–281.
  33. 33. Zhang HT, Crissman AM, Dorairaj NR, Chandler LJ, O'Donnell JM (2000) Inhibition of cyclic AMP phosphodiesterase (PDE4) reverses memory deficits associated with NMDA receptor antagonism. Neuropsychopharmacology 23: 198–204.
  34. 34. Zhang HT, Zhao Y, Huang Y, Dorairaj NR, Chandler LJ, et al. (2004) Inhibition of the phosphodiesterase 4 (PDE4) enzyme reverses memory deficits produced by infusion of the MEK inhibitor U0126 into the CA1 subregion of the rat hippocampus. Neuropsychopharmacology 29: 1432–1439.
  35. 35. Rutten K, Wallace TL, Works M, Prickaerts J, Blokland A, et al. (2011) Enhanced long-term depression and impaired reversal learning in phosphodiesterase 4B-knockout (PDE4B(−/−)) mice. Neuropharmacology 61: 138–147.
  36. 36. Burgin AB, Magnusson OT, Singh J, Witte P, Staker BL, et al. (2010) Design of phosphodiesterase 4D (PDE4D) allosteric modulators for enhancing cognition with improved safety. Nat Biotechnol 28: 63–70.
  37. 37. Li YF, Cheng YF, Huang Y, Conti M, Wilson SP, et al. (2011) Phosphodiesterase-4D knock-out and RNA interference-mediated knock-down enhance memory and increase hippocampal neurogenesis via increased cAMP signaling. J Neurosci 31: 172–183.
  38. 38. Randt CT, Judge ME, Bonnet KA, Quartermain D (1982) Brain cyclic AMP and memory in mice. Pharmacol Biochem Behav 17: 677–680.
  39. 39. Gong B, Vitolo OV, Trinchese F, Liu S, Shelanski M, et al. (2004) Persistent improvement in synaptic and cognitive functions in an Alzheimer mouse model after rolipram treatment. J Clin Invest 114: 1624–1634.
  40. 40. Bourtchouladze R, Lidge R, Catapano R, Stanley J, Gossweiler S, et al. (2003) A mouse model of Rubinstein-Taybi syndrome: defective long-term memory is ameliorated by inhibitors of phosphodiesterase 4. Proc Natl Acad Sci U S A 100: 10518–10522.
  41. 41. Rutten K, Prickaerts J, Blokland A (2006) Rolipram reverses scopolamine-induced and time-dependent memory deficits in object recognition by different mechanisms of action. Neurobiol Learn Mem 85: 132–138.
  42. 42. Rutten K, Van Donkelaar EL, Ferrington L, Blokland A, Bollen E, et al. (2009) Phosphodiesterase inhibitors enhance object memory independent of cerebral blood flow and glucose utilization in rats. Neuropsychopharmacology 34: 1914–1925.
  43. 43. Houslay MD (2001) PDE4 cAMP-specific phosphodiesterases. Prog Nucleic Acid Res Mol Biol 69: 249–315.
  44. 44. Zhang HT (2009) Cyclic AMP-specific phosphodiesterase-4 as a target for the development of antidepressant drugs. Curr Pharm Des 15: 1688–1698.
  45. 45. Hoffmann R, Baillie GS, MacKenzie SJ, Yarwood SJ, Houslay MD (1999) The MAP kinase ERK2 inhibits the cyclic AMP-specific phosphodiesterase HSPDE4D3 by phosphorylating it at Ser579. EMBO J 18: 893–903.
  46. 46. Richter W, Day P, Agrawal R, Bruss MD, Granier S, et al. (2008) Signaling from beta1- and beta2-adrenergic receptors is defined by differential interactions with PDE4. EMBO J 27: 384–393.
  47. 47. van Donkelaar EL, Rutten K, Blokland A, Akkerman S, Steinbusch HW, et al. (2008) Phosphodiesterase 2 and 5 inhibition attenuates the object memory deficit induced by acute tryptophan depletion. Eur J Pharmacol 600: 98–104.
  48. 48. Zhao J, Harada N, Kurihara H, Nakagata N, Okajima K (2010) Cilostazol improves cognitive function in mice by increasing the production of insulin-like growth factor-I in the hippocampus. Neuropharmacology 58: 774–783.
  49. 49. O'Donnell JM, Frith S (1999) Behavioral effects of family-selective inhibitors of cyclic nucleotide phosphodiesterases. Pharmacol Biochem Behav 63: 185–192.
  50. 50. Edelson J, Stroshane R, Benziger DP, Cody R, Benotti J, et al. (1986) Pharmacokinetics of the bipyridines amrinone and milrinone. Circulation 73: III145–152.
  51. 51. Lindsay CA, Barton P, Lawless S, Kitchen L, Zorka A, et al. (1998) Pharmacokinetics and pharmacodynamics of milrinone lactate in pediatric patients with septic shock. J Pediatr 132: 329–334.
  52. 52. Krause W, Kuhne G (1988) Pharmacokinetics of rolipram in the rhesus and cynomolgus monkeys, the rat and the rabbit. Studies on species differences. Xenobiotica 18: 561–571.
  53. 53. Feschenko MS, Stevenson E, Nairn AC, Sweadner KJ (2002) A novel cAMP-stimulated pathway in protein phosphatase 2A activation. J Pharmacol Exp Ther 302: 111–118.
  54. 54. Moon EY, Lerner A (2003) PDE4 inhibitors activate a mitochondrial apoptotic pathway in chronic lymphocytic leukemia cells that is regulated by protein phosphatase 2A. Blood 101: 4122–4130.
  55. 55. Pullar CE, Grahn JC, Liu W, Isseroff RR (2006) Beta2-adrenergic receptor activation delays wound healing. FASEB J 20: 76–86.
  56. 56. Oliver CJ, Shenolikar S (1998) Physiologic importance of protein phosphatase inhibitors. Front Biosci 3: D961–972.
  57. 57. Resjo S, Oknianska A, Zolnierowicz S, Manganiello V, Degerman E (1999) Phosphorylation and activation of phosphodiesterase type 3B (PDE3B) in adipocytes in response to serine/threonine phosphatase inhibitors: deactivation of PDE3B in vitro by protein phosphatase type 2A. Biochem J 341(Pt 3): 839–845.
  58. 58. Levallet G, Levallet J, Bonnamy PJ (2008) FSH-induced phosphoprotein phosphatase 2A-mediated deactivation of particulate phosphodiesterase-4 activities is abolished after alteration in proteoglycan synthesis in immature rat Sertoli cells. J Endocrinol 197: 45–54.
  59. 59. Desjardins S, Cauchy MJ (1995) Comparative cardiac effects of milrinone and sodium nitroprusside in conscious rats. Drug Chem Toxicol 18: 43–59.
  60. 60. Griebel G, Misslin R, Vogel E, Bourguignon JJ (1991) Behavioral effects of rolipram and structurally related compounds in mice: behavioral sedation of cAMP phosphodiesterase inhibitors. Pharmacol Biochem Behav 39: 321–323.
  61. 61. Silvestre JS, Fernandez AG, Palacios JM (1999) Effects of rolipram on the elevated plus-maze test in rats: a preliminary study. J Psychopharmacol 13: 274–277.
  62. 62. Sik A, van Nieuwehuyzen P, Prickaerts J, Blokland A (2003) Performance of different mouse strains in an object recognition task. Behav Brain Res 147: 49–54.
  63. 63. Rodriguez R, Molino B, Weiss HR, Scholz PM (2004) Negative metabolic and coronary flow effects of decreases in cAMP and increases in cGMP in control and renal hypertensive rabbit hearts. J Appl Physiol 97: 439–445.
  64. 64. Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72: 248–254.
  65. 65. Thompson WJ, Appleman MM (1971) Multiple cyclic nucleotide phosphodiesterase activities from rat brain. Biochemistry 10: 311–316.
  66. 66. Bussey TJ, Duck J, Muir JL, Aggleton JP (2000) Distinct patterns of behavioural impairments resulting from fornix transection or neurotoxic lesions of the perirhinal and postrhinal cortices in the rat. Behav Brain Res 111: 187–202.
  67. 67. Ennaceur A, Neave N, Aggleton JP (1997) Spontaneous object recognition and object location memory in rats: the effects of lesions in the cingulate cortices, the medial prefrontal cortex, the cingulum bundle and the fornix. Exp Brain Res 113: 509–519.
  68. 68. Hotte M, Thuault S, Lachaise F, Dineley KT, Hemmings HC, et al. (2006) D1 receptor modulation of memory retrieval performance is associated with changes in pCREB and pDARPP-32 in rat prefrontal cortex. Behav Brain Res 171: 127–133.
  69. 69. Parker A, Gaffan D (1998) Interaction of frontal and perirhinal cortices in visual object recognition memory in monkeys. Eur J Neurosci 10: 3044–3057.
  70. 70. Warburton EC, Brown MW (2010) Findings from animals concerning when interactions between perirhinal cortex, hippocampus and medial prefrontal cortex are necessary for recognition memory. Neuropsychologia 48: 2262–2272.
  71. 71. Bonaventure P, Hall H, Gommeren W, Cras P, Langlois X, et al. (2000) Mapping of serotonin 5-HT(4) receptor mRNA and ligand binding sites in the post-mortem human brain. Synapse 36: 35–46.
  72. 72. Eglen RM, Bonhaus DW, Johnson LG, Leung E, Clark RD (1995) Pharmacological characterization of two novel and potent 5-HT4 receptor agonists, RS 67333 and RS 67506, in vitro and in vivo. Br J Pharmacol 115: 1387–1392.
  73. 73. Vilaro MT, Cortes R, Gerald C, Branchek TA, Palacios JM, et al. (1996) Localization of 5-HT4 receptor mRNA in rat brain by in situ hybridization histochemistry. Brain Res Mol Brain Res 43: 356–360.
  74. 74. Chini CC, Grande JP, Chini EN, Dousa TP (1997) Compartmentalization of cAMP signaling in mesangial cells by phosphodiesterase isozymes PDE3 and PDE4. Regulation of superoxidation and mitogenesis. J Biol Chem 272: 9854–9859.
  75. 75. Dousa TP (1998) Signaling role of PDE isozymes in pathobiology of glomerular mesangial cells. Studies in vitro and in vivo. Cell Biochem Biophys 29: 19–34.
  76. 76. Dousa TP (1999) Cyclic-3′,5′-nucleotide phosphodiesterase isozymes in cell biology and pathophysiology of the kidney. Kidney Int 55: 29–62.
  77. 77. Matousovic K, Grande JP, Chini CC, Chini EN, Dousa TP (1995) Inhibitors of cyclic nucleotide phosphodiesterase isozymes type-III and type-IV suppress mitogenesis of rat mesangial cells. J Clin Invest 96: 401–410.
  78. 78. Aujla H, Beninger RJ (2001) Hippocampal-prefrontocortical circuits: PKA inhibition in the prefrontal cortex impairs delayed nonmatching in the radial maze in rats. Behav Neurosci 115: 1204–1211.
  79. 79. Abel T, Nguyen PV, Barad M, Deuel TA, Kandel ER, et al. (1997) Genetic demonstration of a role for PKA in the late phase of LTP and in hippocampus-based long-term memory. Cell 88: 615–626.
  80. 80. Frankland PW, Bontempi B (2005) The organization of recent and remote memories. Nat Rev Neurosci 6: 119–130.
  81. 81. Kelly MP, Cheung YF, Favilla C, Siegel SJ, Kanes SJ, et al. (2008) Constitutive activation of the G-protein subunit Galphas within forebrain neurons causes PKA-dependent alterations in fear conditioning and cortical Arc mRNA expression. Learn Mem 15: 75–83.
  82. 82. Giorgi M, Modica A, Pompili A, Pacitti C, Gasbarri A (2004) The induction of cyclic nucleotide phosphodiesterase 4 gene (PDE4D) impairs memory in a water maze task. Behav Brain Res 154: 99–106.
  83. 83. Rutten K, Basile JL, Prickaerts J, Blokland A, Vivian JA (2008) Selective PDE inhibitors rolipram and sildenafil improve object retrieval performance in adult cynomolgus macaques. Psychopharmacology (Berl) 196: 643–648.
  84. 84. Dodge-Kafka KL, Bauman A, Mayer N, Henson E, Heredia L, et al. (2010) cAMP-stimulated protein phosphatase 2A activity associated with muscle A kinase-anchoring protein (mAKAP) signaling complexes inhibits the phosphorylation and activity of the cAMP-specific phosphodiesterase PDE4D3. J Biol Chem 285: 11078–11086.
  85. 85. Besheer J, Jensen HC, Bevins RA (1999) Dopamine antagonism in a novel-object recognition and a novel-object place conditioning preparation with rats. Behav Brain Res 103: 35–44.
  86. 86. Fischmeister R, Castro LR, Abi-Gerges A, Rochais F, Jurevicius J, et al. (2006) Compartmentation of cyclic nucleotide signaling in the heart: the role of cyclic nucleotide phosphodiesterases. Circ Res 99: 816–828.
  87. 87. Xiang Y, Naro F, Zoudilova M, Jin SL, Conti M, et al. (2005) Phosphodiesterase 4D is required for beta2 adrenoceptor subtype-specific signaling in cardiac myocytes. Proc Natl Acad Sci U S A 102: 909–914.
  88. 88. Zaccolo M, Pozzan T (2002) Discrete microdomains with high concentration of cAMP in stimulated rat neonatal cardiac myocytes. Science 295: 1711–1715.
  89. 89. Cong M, Perry SJ, Lin FT, Fraser ID, Hu LA, et al. (2001) Regulation of membrane targeting of the G protein-coupled receptor kinase 2 by protein kinase A and its anchoring protein AKAP79. J Biol Chem 276: 15192–15199.
  90. 90. Baillie GS, Sood A, McPhee I, Gall I, Perry SJ, et al. (2003) beta-Arrestin-mediated PDE4 cAMP phosphodiesterase recruitment regulates beta-adrenoceptor switching from Gs to Gi. Proc Natl Acad Sci U S A 100: 940–945.
  91. 91. Li X, Huston E, Lynch MJ, Houslay MD, Baillie GS (2006) Phosphodiesterase-4 influences the PKA phosphorylation status and membrane translocation of G-protein receptor kinase 2 (GRK2) in HEK-293beta2 cells and cardiac myocytes. Biochem J 394: 427–435.
  92. 92. Xin W, Tran TM, Richter W, Clark RB, Rich TC (2008) Roles of GRK and PDE4 activities in the regulation of beta2 adrenergic signaling. J Gen Physiol 131: 349–364.
  93. 93. Janssens V, Goris J (2001) Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem J 353: 417–439.
  94. 94. Sim AT, Ludowyke RI, Verrills NM (2006) Mast cell function: regulation of degranulation by serine/threonine phosphatases. Pharmacol Ther 112: 425–439.
  95. 95. Huang Z, Ducharme Y, Macdonald D, Robichaud A (2001) The next generation of PDE4 inhibitors. Curr Opin Chem Biol 5: 432–438.
  96. 96. Lugnier C (2006) Cyclic nucleotide phosphodiesterase (PDE) superfamily: a new target for the development of specific therapeutic agents. Pharmacol Ther 109: 366–398.
  97. 97. Maurice DH, Palmer D, Tilley DG, Dunkerley HA, Netherton SJ, et al. (2003) Cyclic nucleotide phosphodiesterase activity, expression, and targeting in cells of the cardiovascular system. Mol Pharmacol 64: 533–546.
  98. 98. Zamah AM, Delahunty M, Luttrell LM, Lefkowitz RJ (2002) Protein kinase A-mediated phosphorylation of the beta 2-adrenergic receptor regulates its coupling to Gs and Gi. Demonstration in a reconstituted system. J Biol Chem 277: 31249–31256.
  99. 99. Houslay MD, Baillie GS (2003) The role of ERK2 docking and phosphorylation of PDE4 cAMP phosphodiesterase isoforms in mediating cross-talk between the cAMP and ERK signalling pathways. Biochem Soc Trans 31: 1186–1190.