Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • For Subscribers
    • For Advertisers
    • Terms & Conditions of Use
    • Permissions
  • About Us
    • About JPET
    • Feedback
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • For Subscribers
    • For Advertisers
    • Terms & Conditions of Use
    • Permissions
  • About Us
    • About JPET
    • Feedback
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleNEUROPHARMACOLOGY

Correlation between ex Vivo Receptor Occupancy and Wake-Promoting Activity of Selective H3 Receptor Antagonists

Siyuan Le, John A. Gruner, Joanne R. Mathiasen, Michael J. Marino and Hervé Schaffhauser
Journal of Pharmacology and Experimental Therapeutics June 2008, 325 (3) 902-909; DOI: https://doi.org/10.1124/jpet.107.135343
Siyuan Le
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
John A. Gruner
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joanne R. Mathiasen
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael J. Marino
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hervé Schaffhauser
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

The histamine H3 receptor (H3R) modulates the release of neurotransmitters that are involved in vigilance, cognition, and sleep-wake regulation. H3R antagonism has been proposed as a novel approach to the treatment of cognitive and attention deficit as well as sleep disorders. It is apparent that H3R antagonists produce pharmacological effects in preclinical animal models across a wide dose range. Several H3R antagonists were reported to be effective at producing cognitive enhancing effects at low doses, while producing robust wake enhancement at higher doses. To better understand the effect of H3R antagonists across a broad dose range, an ex vivo receptor binding assay has been used to estimate the degree of H3R occupancy in vivo. The H3R antagonists ciproxifan, thioperamide, GSK189254 (6-[(3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride), and ABT-239 ([4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile) produced wake-promoting activity in vivo and a dose-dependent inhibition of H3R binding ex vivo. For ciproxifan, thioperamide, and GSK189254, a relatively low level of cumulative wake activity was linearly correlated with up to 80% of the receptor occupancy. In contrast, an abrupt break from linearity and a robust increase of waking activity was observed at doses that produce greater than 80% occupancy. Our results suggest a relatively small increase of waking activity at low levels of receptor occupancy that may be consistent with reported enhancement of attention and cognitive function. Robust waking activity at higher levels of H3R occupancy may be mechanistically different from activities at low levels of H3R occupancy.

The neurotransmitter histamine is synthesized by neurons originating in the tuberomammillary nucleus of the posterior hypothalamus. These neurons project widely throughout the cortex, hippocampus, amygdala, and striatum (Brown et al., 2001). Histamine exerts its action by interacting with a group of G protein-coupled histamine receptors and plays important roles in mediating a variety of functions in the central nervous system (Brown et al., 2001). The H3Ris presynaptically localized and functions both as an autoreceptor and a heteroreceptor by modulating the release of neurotransmitters, including histamine (Arrang et al., 1985), dopamine, acetylcholine, serotonin, and norepinephrine (Schlicker et al., 1994; Blandina et al., 1996; Brown et al., 2001). Activation of the H3R results in the inhibition of neurotransmitter release. In contrast, blockade of the H3R by selective antagonists or inverse agonists can reverse the histamine-mediated inhibition of neurotransmitter release. By virtue of the unique central nervous system localization of the H3R (Drutel et al., 2001; Leurs et al., 2005) and its ability to regulate a variety of neurotransmitters that are thought to be involved in vigilance, cognition, and wakefulness, H3R antagonists and inverse agonists are suggested to hold promise for a number of therapeutic applications (Hancock and Fox, 2004).

Selective H3R antagonists have been extensively evaluated in several preclinical rodent models to establish their role in cognitive function and sleep-wake regulation. These models include social recognition (Fox et al., 2003a,b, 2005), novel object recognition (Fox et al., 2005; Ligneau et al., 2007; Medhurst et al., 2007a), spatial learning and memory in an adult rat water maze test (Fox et al., 2005), scopolamine-induced deficits in memory consolidation in a passive avoidance test (Medhurst et al., 2007b), and cortical electroencephalographic (EEG) recording (Barbier et al., 2004; Ligneau et al., 2007).

It has also been suggested that histamine plays an important role in the modulation of the sleep-wake cycle. Histaminergic neurons exhibit an increase in firing rate during waking (Brown et al., 2001). Depletion of histamine has been shown to lead to the reduction of awake time (Monti et al., 1988), whereas activation of the H3R by selective agonists results in an increase in slow-wave sleep (Lin et al., 1990; Monti, 1993; Monti et al., 1996). In contrast, H3R antagonists have been shown to increase wakefulness in cats, rats, and mice (Lin et al., 1990; Monti et al., 1991, 1996; Barbier et al., 2004; Ligneau et al., 2007; Parmentier et al., 2007). Recent studies with the H3R antagonist thioperamide and JNJ-5207852 in H3R knockout mice have confirmed the involvement of the H3R in the wake-promoting activity of these compounds (Toyota et al., 2002; Barbier et al., 2004).

It is apparent that H3R antagonists produce in vivo effects in preclinical models across a wide dose range. Several selective H3R antagonists exhibit potent efficacy in social recognition models (Fox et al., 2003b, 2005). However, studies suggest that much higher doses of H3R antagonists are required to produce robust wake activity. For example, the minimal effective i.p. dose for ciproxifan to reduce slow-wave activity was 10-fold higher than that required for the improvement of social memory, whereas the dose difference was 30-fold for A-304121 (Fox et al., 2003b). In the case of ABT-239, there was no detectable change in slow-wave EEG at 30 mg/kg, whereas it was effective in social recognition at 0.01 mg/kg (Fox et al., 2005). The present study sought to examine this phenomenon by correlating the wake-promoting activity of a number of H3R antagonists with an ex vivo estimate of receptor occupancy. Our results with ciproxifan, thioperamide, and GSK189254 suggest that the relatively low levels of cumulative wake activity are linearly correlated with up to 80% of H3R occupancy, after which an abrupt increase of waking activity is observed. These observations suggest that the relatively small increase in waking EEG activity observed at low levels of H3R occupancy may be consistent with literature reports of enhanced attention and cognitive function. Robust waking activity is associated with higher levels of H3R occupancy and may be mechanistically different from activities observed at low occupancy.

Materials and Methods

Animals. Adult Long Evans rats (350 g; Harlan, Indianapolis, IN) were used in these studies. All animals were maintained on a 24-h light/dark cycle (on at 7:00 AM/off at 7:00 PM), with food and water available ad libitum. All animals except those in the sleep study (see below) were group housed. All experimental animal procedures were conducted in accordance with the Institute of Laboratory Animal Resources (1996) and were approved by Cephalon's Institutional Animal Care and Use Committee.

Compounds. Ciproxifan and GSK189254 were purchased from ValuTek (Princeton, NJ). Thioperamide and d-amphetamine were purchased from Sigma-Aldrich (St. Louis, MO). [3H]N-α-Methylhistamine ([3H]NAMH, catalog no. NET 1027, 83 Ci/mmol) was purchased from PerkinElmer Life and Analytical Sciences (Waltham, MA).

Ex Vivo [3H]NAMH Binding Assay in Rat Cortical Homogenate. Animals were dosed with vehicle (0.5% methylcellulose/0.2% Tween 80) or various concentrations of compound by i.p. injection. The animals were killed by decapitation 1 h after injection, and the cerebral cortex was dissected. The 1-h time point was chosen based on pharmacokinetic studies demonstrating that, after i.p. dosing, all of the compounds evaluated in the present study reached a plateau plasma level by 1 h that was maintained for at least 4 h. Therefore, 1 h represents the earliest time at which steady-state occupancy could be assessed. The cortices were homogenized in 50 mM HEPES, pH 7.4 (7.5 ml/2 g) with a polytron for 10 s. The cortical homogenate was added to a deep-well plate containing [3H]NAMH (0.15 nM), in a volume of 0.4 ml (final protein concentration 0.8 mg/assay) as described previously (Taylor et al., 1992). Reactions were incubated at 25°C for 1 h and stopped by rapid filtration onto glass filter paper (GF/B Whatman, catalog no. FPLR-24) using a 48-well cell harvester (Brandel Inc., Gaithersburg, MD). The filters were washed three times with ice-cold assay buffer and mixed with 5 ml of liquid scintillation cocktail (Ultima Gold; PerkinElmer Life and Analytical Sciences). Radioactivity trapped on the filters was measured in a liquid scintillation counter (PerkinElmer Life and Analytical Sciences). A limitation of ex vivo binding is the in vitro manipulation of the tissue, an issue that has been described in great detail for corticotrophin-releasing factor 1 (CRF1) binding (Li et al., 2003). Care was taken in our ex vivo assays to choose a minimal incubation time that allowed binding to reach equilibrium by assessing the time course of receptor binding for H3 antagonists from 15 min to 3 h. These studies showed that receptor binding reached equilibrium at 1-h incubation and was stable for up to 3 h for compounds used.

Nonspecific binding was determined in the presence of 10 μM thioperamide. The nonspecific binding was 10% of total binding in vehicle-treated samples, indicating that 90% of the [3H]NAMH binding was specific. Each compound was tested in three independent experiments with a total of at least four animals per dose in the dose-response curve. The inhibition of specific [3H]NAMH binding, calculated as relative to vehicle-treated samples, was determined to provide an indication of receptor occupancy by the compound. The dose-response curve was fit to a four parameter logistic equation to yield the IC50 value using Prism 4.0 (GraphPad Software Inc., San Diego, CA).

Compound Quantification. Plasma and brain samples were collected from the study animals in the ex vivo binding experiments and were submitted for quantitative analysis to determine the respective compound concentrations. Blood samples were collected into heparinized tubes and placed on wet ice until centrifuged (16,000g, 5 min) to separate plasma. The supernatant was collected and stored at -20°C pending analysis. At the time of analysis, 10 volumes of cold acetonitrile containing an internal standard (Alprenolol) was added to each sample, which was then vortexed and centrifuged. The supernatant was removed and placed into an auto sampler vial, and the amount of compound present in the samples was analyzed by liquid chromatography/mass spectrometry. The concentration of compound in the samples was quantified against either a rat plasma or brain standard curve made via serial dilution in a concentration range from 10 to 5000 ng/ml. Samples containing concentrations greater than 5% above the top of the standard curve were diluted 1:10 with acetonitrile. The brain/plasma ratio was calculated accordingly.

Determination of Sleep/Wake Activity. Cortical EEG and electromyographic (EMG) recordings from neck muscles were used to determine whether the reference H3R antagonists altered sleep-wake activity in rats. Ciproxifan (0.3, 1, 3, 10, and 100 mg/kg), thioperamide (1, 3, 10, 30, and 100 mg/kg), GSK189254 (1, 3, 10, 20, and 30 mg/kg), or ABT-239 (1, 2, 3, and 10 mg/kg) was dosed by i.p. injection. EEG and EMG electrodes were chronically implanted under Nembutal anesthesia. EEG activity was recorded from screw electrodes over the frontal cortex (+3.0 mm AP from bregma, ±2.0 mm mediolateral) and hippocampus (-4.0 mm AP from bregma, ±2.0 mm mediolateral). The animals were allowed to recover from surgery for 2 weeks before recording, during which time they were housed in pairs in standard rat cages. The day before recording, rats were placed in individual containers (31 × 31 × 31 cm) in sound-attenuating cabinets and connected by cables to the recording equipment. Each cabinet contained a fan, a light (12-h light/dark cycle, ∼30 lux), and a speaker to provide background noise. The animals were dosed the following day 5 h after lights on (circadian time-5) and removed 22 h later and were otherwise not disturbed. EEG and EMG signals were amplified (10,000 and 1000, respectively), band pass filtered between 0.3 and 500 Hz for EEG and between 10 and 500 Hz for EMG, and digitized at 128 samples/s. Sleep/wake activity was evaluated using ICELUS sleep scoring software (M. Opp, University of Michigan, Ann Arbor, MI) by scoring EEG and EMG activity into wake, slow-wave sleep, and rapid-eye movement sleep in 6-s epochs according to standard criteria (Opp and Krueger, 1994; Edgar and Seidel, 1997). The EEG/EMG data were scored for 8 h postdosing and converted to percentage time awake. Given the 1-h occupancy determination time, wake would ideally be measured at 1 h. However, the manipulation and injection procedure produces a transient increase in wake activity that lasts for approximately 30 min (evident in vehicle curve in Fig. 3). The 2-h cumulative wake time was therefore chosen as the most representative measure of wake activity. Values of cumulative time awake were evaluated for 2 h postdosing for comparison with receptor occupancy.

Statistical Analysis. Values are listed as mean ± S.E.M. where appropriate. Treatment effects were evaluated using ANOVA. Treatment group comparisons were made using unpaired, two-tailed Student's t tests or Dunnett's test for multiple comparisons as appropriate.

Results

Ex Vivo Binding. An ex vivo binding assay was used to estimate in vivo occupancy by H3R antagonists. Ciproxifan (0.1–10 mg/kg), thioperamide (0.3–100 mg/kg), GSK189254 (0.1–30 mg/kg), or ABT-239 (0.3–30 mg/kg) were administered by the i.p. route as described under Materials and Methods. All four compounds tested produced dose-dependent inhibition of specific ex vivo binding of [3H]NAMH to cortical homogenate with the following rank order of potency: ciproxifan > GSK189254 > ABT-239 > thioperamide (Fig. 1; Table 1). Maximal inhibition was obtained with all compounds and IC50 values were between 0.44 and 5.7 mg/kg i.p. (Table 1). The plasma and brain concentrations were determined by liquid chromatography/mass spectrometry. All compounds showed a linear correlation between the inhibition of specific [3H]N-α-methylhistamine binding and plasma or brain compound concentration (Fig. 2). These data were used to calculate the OC50 (brain concentration at 50% occupancy) for each compound (Table 1). Although these numbers are significantly higher than the Ki values typically reported for these compounds in rat cortex (for example, see Medhurst et al., 2007a), multiple factors other than binding affinity such as the distribution of the compound in brain tissue (Fox et al., 2005) and free fraction of the compound play a role in determining the OC50. d-Amphetamine was also tested in the ex vivo H3R binding assay. Although 0.3 and 1 mg/kg i.p. doses of d-amphetamine produced robust wake promotion activity, no detectable inhibition of the specific [3H]N-α-methylhistamine binding was observed from 0.003 up to 1 mg/kg (data not shown).

View this table:
  • View inline
  • View popup
TABLE 1

IC50 and OC50 of H3R antagonists measured by ex vivo binding Data from Fig. 1 were fit to a four-parameter logistic equation giving an IC50 value for each compound. The number of animals tested per dose is also indicated. Data are shown as mean ± S.E.M. from at least three independent experiments. The brain concentration at which 50% occupancy was observed is shown.

Wake Promotion of H3R Antagonists. EEG/EMG recording was used to determine the ability of H3R antagonists to affect arousal and sleep-wake states. Ciproxifan, thioperamide, GSK189254, or ABT-239 was administered i.p. The time course of wake activity of ABT-239 is shown in Fig. 3. A dose-dependent increase in cumulative wake activity for 2 h postdosing [2-h area under the curve (AUC)] was observed from 1 to 10 mg/kg i.p. for this compound (Fig. 4D). In contrast, slow-wave sleep and rapid-eye movement sleep showed cumulative decreases from 2 to 10 mg/kg (data not shown). Cumulative wake activity for the H3R antagonists ciproxifan, thioperamide, and GSK189254 are also shown in Fig. 4. All four compounds enhanced wake in this assay: ciproxifan (0.3–30 mg/kg i.p.), thioperamide (30 and 100 mg/kg i.p.), GSK189254 (20–30 mg/kg i.p.), and ABT-239 (1–10 mg/kg i.p.) (p < 0.001, ANOVA).

    Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Inhibition of ex vivo [3H]NAMH binding in the rat cortex after i.p. administration of ciproxifan (A), thioperamide (B), GSK189254 (C), and ABT-239 (D) for 1 h. Data points are compared with vehicle-treated animals and expressed as percentage inhibition of specific binding. Data are expressed as mean ± S.E.M. of at least four animals per dose from a minimum of three independent experiments.

Correlation of Receptor Occupancy and Waking Activity of H3R Antagonists. The fact that H3R antagonists are reported to promote wakefulness at doses that are 10- to 100-fold higher than the minimal effective dose in other preclinical models prompted us to investigate the relationship between receptor occupancy and waking activity. For this purpose, we correlated the 2-h cumulative wake time of each H3 compound with the respective ex vivo binding values. As shown in the Fig. 5, a modest increase in cumulative wake activity (2-h AUC) with increasing receptor occupancy was observed for ciproxifan, thioperamide, and GSK189254 up to relatively high occupancy values (Fig. 5). All four compounds showed highly linear correlations (R2 > 0.8) over this portion of the occupancy versus wake activity curve. The slopes for ciproxifan, thioperamide, and GSK189254 were comparable and yielded an average increase of approximately 2 min/h of wake for every 10% increase in occupancy. In contrast to the modest increase in wake at low levels of H3R occupancy for these compounds, a robust increase in wakefulness was observed at a particular level of receptor occupancy for each compound. For ciproxifan, thioperamide, or GSK189254, this upward increase occurred when the H3R was at least 80% occupied. In the case of ABT-239, a slope of 8.8 min/h for every 10% increase in occupancy was observed, which was approximately 4-fold greater than the others. For ABT-239, a similar level of wake activity (∼100 min) was reached at around 80% of the H3R occupancy without an abrupt increase up to 2 h postdosing.

Discussion

The results obtained in the present study demonstrated a consistent relationship between the in vivo effects and the level of H3R occupancy produced by several selective H3R antagonists. The ex vivo receptor binding assay has been useful in examining the ability of compounds to penetrate the blood-brain barrier and to bind receptors (Taylor et al., 1992). This approach has been used in previous reports for assessing the in vivo receptor binding of a variety of compounds, including H3R antagonists thioperamide (Barnes et al., 1993; Bertoni et al., 2007), VUF 9153 (Barnes et al., 1993), JNJ-5207852 (Barbier et al., 2004), GSK189254 (Medhurst et al., 2007a), GSK207040, and GSK334429 (Medhurst et al., 2007b). In the present study, the data demonstrate a dose-dependent inhibition of the ex vivo binding by H3R antagonists in rat cortical homogenate. As a negative control, d-amphetamine, a wake-promoting agent and a known inhibitor of dopamine transporter activity (Nishino et al., 1998; Wayment et al., 1998) and a dopamine release agent (Sulzer et al., 1993), does not inhibit specific binding of [3H]NAMH. We also demonstrate that receptor occupancy is correlated with the plasma and brain level of the compound. Ex vivo binding studies with the same H3R antagonists employed in the present studies have been described previously (Medhurst et al., 2007a). Although there are significant methodological differences between these studies and the studies presented here (dosing route, dosing time, tissue preparation, and binding conditions), it is of interest to compare the results. Despite the differences in protocol, our results agree reasonably well. The OC50 for GSK189254 in the two studies was within 3-fold (60 nM in Medhurst et al. and 167 nM in the present study). In addition, although Medhurst et al. did not report an OC50 for the other H3R antagonists, the percentage inhibition at the highest dose tested for ciproxifan (97% at 10 mg/kg compared with 99 ± 0.2% in the present study), thioperamide (1.3-fold difference with 80% at 10 mg/kg reported compared with 60 ± 5% in the present study), and ABT-239 (3.7-fold difference with 68% at 1 mg/kg reported compared with 18 ± 6% in the present study) all compare favorably. Although it could be argued that the present studies underestimate or that the previous studies overestimate in vivo occupancy, a true measure of in vivo occupancy must await the use of a sensitive PET ligand.

The relationship between receptor occupancy and behavioral effects of several targets has been studied, including CRF1 (Li et al., 2003), cannabinoid (Gifford et al., 1999), and dopamine D2 receptor (Wadenberg et al., 2000). In the case of the CRF1 receptor, it was shown that some compounds produced a minimal anxiolytic effort at 50% occupancy (Heinrichs et al., 2002) but were ineffective in a defensive withdrawal test until higher level of receptor occupancy was reached (Arborelius et al., 2000). Likewise, studies with D2 receptor showed that suppression of conditioned avoidance response by raclopride occurred at a D2 occupancy of around 70 to 75%, but catalepsy occurred at D2 occupancy > 80% in rats (Wadenberg et al., 2000). Here, we find that relatively high H3R occupancy is required to produce robust waking activity. GSK189254 required 92% occupancy (20 mg/kg i.p.) to significantly increase waking, whereas thioperamide required 85% occupancy (30 mg/kg i.p.). Notably, we demonstrate here a significant wake-enhancing effect for ABT-239 that was not observed at similar doses previously (Fox et al., 2005). A possible reason for this discrepancy is that only delta power was used to evaluate changes in sleep/wake activity previously. Our data suggest that the increase in waking activity and receptor occupancy is linear up to relatively high occupancy. The slopes for the linear correlations shown in Fig. 5 are modest, indicating a small effect on wake at levels of H3R occupancy below 80% for ciproxifan, thioperamide, and GSK189254 (2 min/h for every 10% increase in occupancy). Only when H3R occupancy exceeded a certain threshold (∼80%) was an abrupt increase in waking activity detected. It is interesting to note that ABT-239 seems to have a waking versus occupancy profile different from the other antagonists. First, although the increase in wake was proportional to occupancy up to 80%, the rate of increase was greater (8.8 min/h per 10% increase in occupancy) (Fig. 5). Second, there was no abrupt increase in waking activity produced by ABT-239. However, there is evidently a distinct increase in excitability at higher levels. Tremor and seizures have been reported at above 28 mg/kg i.p. in rats (Fox et al., 2005). Although these observations cannot be explained based on the current data, the possibility exists that ABT-239 may possess additional activities that distinguish it from the other H3R antagonists. Alternatively, other properties may contribute to the differential activity of the compound. For example, H3Rs are known to exhibit constitutive activity (Rouleau et al., 2002). The degree of inverse agonism exhibited by an H3R ligand, as well as variations in the degree of constitutive activity in various brain regions could produce significantly different in vivo behavioral effects at identical occupancy levels. Other factors including the pharmacokinetic and pharmacodynamic properties of the compounds may also play a role in the observed differential in vivo activity.

    Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Relationship between ex vivo H3R binding and plasma (A) and brain (B) concentration of ciproxifan (a), thioperamide (b), GSK189254 (c), and ABT-239 (d) in individual animals. The curves were fit by linear regression with R2 = 0.87, 0.90, 0.88, and 0.94 for ciproxifan, thioperamide, GSK189254, and ABT-239 in plasma and R2 = 0.87, 0.91, 0.79, and 0.95 for ciproxifan, thioperamide, GSK189254, and ABT-239 in brain, respectively.

    Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Time course of wake enhancement produced by ABT-239 dosed at 1 to 10 mg/kg i.p., 5 h after lights on. Dosing occurs at time = 0, which wakes animals up for approximately 30 min. Time of lights off shown by black bar on time axis. N for each group shown in parentheses in legend. Values = mean + S.E.M. *, p < 0.05 versus vehicle, unpaired Student's t test for individual 0.5-h time points.

    Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Cumulative wake activity (2 h AUC) for ciproxifan (A), thioperamide (B), GSK189254 (C), and ABT-239 (D). Values = mean + S.E.M.; the number of animals in each treatment group is shown at bottom of the bar. Each compound showed a significant treatment effect (p < 0.001, ANOVA). *, p < 0.05 versus vehicle, Dunnett's test.

Our results suggest that the H3R system is finely tuned to discriminate changes in receptor occupancy. A relatively small increase of EEG activity at low levels of receptor occupancy observed with ciproxifan, thioperamide, and GSK189254. However, these effects may be consistent with previously reported enhancement of attention and cognitive function by H3R antagonists. Robust waking activity at higher occupancy may be mechanistically different from activities at low occupancy.

    Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Relationship between receptor occupancy and cumulative waking (2-h AUC): A, ciproxifan; B, thioperamide; C, GSK189254; and D, ABT-239. Solid line, linear correlation over selected points. Corresponding slope, R2, and p value indicating difference of slope from zero are indicated for each compound. *, p < 0.05 for 2-h wake AUC versus vehicle for each point, Dunnett's test.

The blockade of H3R is known to promote the release of a variety of neurotransmitters, most of which play important roles in arousal, cognition and sleep-wake regulation (Parmentier et al., 2007). Synaptic levels of dopamine, norepinephrine, and serotonin are regulated by autoreceptors, their respective transporters (Schmitz et al., 2001; Torres et al., 2003) and by recurrent inhibitory circuits (Le Roux et al., 2006), which would tend to oppose increased excitability. The sudden enhancement of waking at higher occupancy levels suggests that the homeostatic regulation of transmitter release may change or even be overwhelmed under these conditions. Alternatively, the enhanced excitability could result from the release of particular neurotransmitters over different occupancy ranges. It will therefore be informative to correlate receptor occupancy with the amounts of particular neurotransmitters released during in vivo microdialysis studies.

It is important to note that, although the compounds used in these studies are reported in the literature to be selective for the H3R, we cannot completely exclude the possibility that the highest concentrations of H3R antagonists could exert effects by binding to other receptors. The fact that these compounds represent multiple chemical scaffolds makes this an unlikely possibility; however, future studies with additional structurally diverse H3R antagonists are needed.

Our observations suggest that the difference in efficacy produced by H3R antagonists observed in the in vivo models can be explained by the degree of H3R occupancy. This information can be valuable in understanding in vivo pharmacodynamics and is potentially useful in providing the guideline for the selection of dose regimens in clinical studies.

Acknowledgments

We thank Rebecca Morey and Deborah Galinis for conducting compound quantification studies and Val Marcy and Yinguo Lin for technical assistance in conducting the animal EEG studies.

Footnotes

  • This study was supported by Cephalon, Inc.

  • Article, publication date, and citation information can be found at http://jpet.aspetjournals.org.

  • doi:10.1124/jpet.107.135343.

  • ABBREVIATIONS: H3R, histamine H3 receptor; EEG, electroencephalographic; JNJ-5207852, 1-[4-(3-piperidin-1-yl-propoxy)-benzyl]-piperidine; A-304121, [4-(3-((2R)-2-aminopropanoyl-1-piperazinyl)propoxy)phenyl)cyclopropylmethanone]; ABT-239, [4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile; GSK189254, 6-[(3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride; [3H]NAMH, [3H]N-α-methylhistamine; CRF1, corticotrophin-releasing factor 1; EMG, electromyographic; ANOVA, analysis of variance; AUC, area under the curve.

    • Received December 14, 2007.
    • Accepted February 26, 2008.
  • The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    Arborelius L, Skelton KH, Thrivikraman KV, Plotsky PM, Schulz DW, and Owens MJ (2000) Chronic administration of the selective corticotropin-releasing factor 1 receptor antagonist CP-154,526: behavioral, endocrine and neurochemical effects in the rat. J Pharmacol Exp Ther 294: 588-597.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Arrang JM, Garbarg M, and Schwartz JC (1985) Autoregulation of histamine release in brain by presynaptic H3-receptors. Neuroscience 15: 553-562.
    OpenUrlCrossRefPubMed
  3. ↵
    Barbier AJ, Berridge C, Dugovic C, Laposky AD, Wilson SJ, Boggs J, Aluisio L, Lord B, Mazur C, Pudiak CM, et al. (2004) Acute wake-promoting actions of JNJ-5207852, a novel, diamine-based H3 antagonist. Br J Pharmacol 143: 649-661.
    OpenUrlCrossRefPubMed
  4. ↵
    Barnes JC, Brown JD, Clarke NP, Clapham J, Evans DJ, and O'Shaughnessy CT (1993) Pharmacological activity of VUF 9153, an isothiourea histamine H3 receptor antagonist. Eur J Pharmacol 250: 147-152.
    OpenUrlCrossRefPubMed
  5. ↵
    Bertoni S, Flammini L, Manenti V, Ballabeni V, Morini G, Comini M, and Barocelli E (2007) In vitro pharmacology at human histamine H3 receptors and brain access of non-imidazole alkylpiperidine derivatives. Pharmacol Res 55: 111-116.
    OpenUrlCrossRefPubMed
  6. ↵
    Blandina P, Giorgetti M, Bartolini L, Cecchi M, Timmerman H, Leurs R, Pepeu G, and Giovannini MG (1996) Inhibition of cortical acetylcholine release and cognitive performance by histamine H3 receptor activation in rats. Br J Pharmacol 119: 1656-1664.
    OpenUrlPubMed
  7. ↵
    Brown RE, Stevens DR, and Haas HL (2001) The physiology of brain histamine. Prog Neurobiol 63: 637-672.
    OpenUrlCrossRefPubMed
  8. ↵
    Drutel G, Peitsaro N, Karlstedt K, Wieland K, Smit MJ, Timmerman H, Panula P, and Leurs R (2001) Identification of rat H3 receptor isoforms with different brain expression and signaling properties. Mol Pharmacol 59: 1-8.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Edgar DM and Seidel WF (1997) Modafinil induces wakefulness without intensifying motor activity or subsequent rebound hypersomnolence in the rat. J Pharmacol Exp Ther 283: 757-769.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Fox GB, Esbenshade TA, Pan JB, Radek RJ, Krueger KM, Yao BB, Browman KE, Buckley MJ, Ballard ME, Komater VA, et al. (2005) Pharmacological properties of ABT-239 [4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile]: II. Neurophysiological characterization and broad preclinical efficacy in cognition and schizophrenia of a potent and selective histamine H3 receptor antagonist. J Pharmacol Exp Ther 313: 176-190.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Fox GB, Pan JB, Faghih R, Esbenshade TA, Lewis A, Bitner RS, Black LA, Bennani YL, Decker MW, and Hancock AA (2003a) Identification of novel H3 receptor (H3R) antagonists with cognition enhancing properties in rats. Inflamm Res 52 (Suppl 1): S31-S32.
    OpenUrlCrossRefPubMed
  12. ↵
    Fox GB, Pan JB, Radek RJ, Lewis AM, Bitner RS, Esbenshade TA, Faghih R, Bennani YL, Williams M, Yao BB, et al. (2003b) Two novel and selective nonimidazole H3 receptor antagonists A-304121 and A-317920: II. In vivo behavioral and neurophysiological characterization. J Pharmacol Exp Ther 305: 897-908.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Gifford AN, Bruneus M, Gatley SJ, Lan R, Makriyannis A, and Volkow ND (1999) Large receptor reserve for cannabinoid actions in the central nervous system. J Pharmacol Exp Ther 288: 478-483.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Hancock AA and Fox GB (2004) Perspectives on cognitive domains, H3 receptor ligands and neurological disease. Expert Opin Investig Drugs 13: 1237-1248.
    OpenUrlCrossRefPubMed
  15. ↵
    Heinrichs SC, De Souza EB, Schulteis G, Lapsansky JL, and Grigoriadis DE (2002) Brain penetrance, receptor occupancy and antistress in vivo efficacy of a small molecule corticotropin releasing factor type I receptor selective antagonist. Neuropsychopharmacology 27: 194-202.
    OpenUrlCrossRefPubMed
  16. ↵
    Institute of Laboratory Animal Resources (1996) Guide for the Care and Use of Laboratory Animals 7th ed. Institute of Laboratory Animal Resources, Commission on Life Sciences, National Research Council, Washington DC.
  17. ↵
    Le Roux N, Amar M, Baux G, and Fossier P (2006) Homeostatic control of the excitation-inhibition balance in cortical layer 5 pyramidal neurons. Eur J Neurosci 24: 3507-3518.
    OpenUrlCrossRefPubMed
  18. ↵
    Leurs R, Bakker RA, Timmerman H, and de Esch IJ (2005) The histamine H3 receptor: from gene cloning to H3 receptor drugs. Nat Rev Drug Discov 4: 107-120.
    OpenUrlCrossRefPubMed
  19. ↵
    Li YW, Hill G, Wong H, Kelly N, Ward K, Pierdomenico M, Ren S, Gilligan P, Grossman S, Trainor G, et al. (2003) Receptor occupancy of nonpeptide corticotropin-releasing factor 1 antagonist DMP696: correlation with drug exposure and anxiolytic efficacy. J Pharmacol Exp Ther 305: 86-96.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Ligneau X, Perrin D, Landais L, Camelin JC, Calmels TP, Berrebi-Bertrand I, Lecomte JM, Parmentier R, Anaclet C, Lin JS, et al. (2007) BF2.649 [1-{3-[3-(4-Chlorophenyl)propoxy]propyl}piperidine, hydrochloride], a nonimidazole inverse agonist/antagonist at the human histamine H3 receptor: preclinical pharmacology. J Pharmacol Exp Ther 320: 365-375.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Lin JS, Sakai K, Vanni-Mercier G, Arrang JM, Garbarg M, Schwartz JC, and Jouvet M (1990) Involvement of histaminergic neurons in arousal mechanisms demonstrated with H3-receptor ligands in the cat. Brain Res 523: 325-330.
    OpenUrlCrossRefPubMed
  22. ↵
    Medhurst AD, Atkins AR, Beresford IJ, Brackenborough K, Briggs MA, Calver AR, Cilia J, Cluderay JE, Crook B, Davis JB, et al. (2007a) GSK189254, a novel H3 receptor antagonist that binds to histamine H3 receptors in Alzheimer's disease brain and improves cognitive performance in preclinical models. J Pharmacol Exp Ther 321: 1032-1045.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Medhurst AD, Briggs MA, Bruton G, Calver AR, Chessell I, Crook B, Davis JB, Davis RP, Foley AG, Heslop T, et al. (2007b) Structurally novel histamine H3 receptor antagonists GSK207040 and GSK334429 improve scopolamine-induced memory impairment and capsaicin-induced secondary allodynia in rats. Biochem Pharmacol 73: 1182-1194.
    OpenUrlCrossRefPubMed
  24. ↵
    Monti JM (1993) Involvement of histamine in the control of the waking state. Life Sci 53: 1331-1338.
    OpenUrlCrossRefPubMed
  25. ↵
    Monti JM, D'Angelo L, Jantos H, and Pazos S (1988) Effects of α-fluoromethylhistidine on sleep and wakefulness in the rat: short note. J Neural Transm 72: 141-145.
    OpenUrlCrossRefPubMed
  26. ↵
    Monti JM, Jantos H, Boussard M, Altier H, Orellana C, and Olivera S (1991) Effects of selective activation or blockade of the histamine H3 receptor on sleep and wakefulness. Eur J Pharmacol 205: 283-287.
    OpenUrlCrossRefPubMed
  27. ↵
    Monti JM, Jantos H, Ponzoni A, and Monti D (1996) Sleep and waking during acute histamine H3 agonist BP 2.94 or H3 antagonist carboperamide (MR 16155) administration in rats. Neuropsychopharmacology 15: 31-35.
    OpenUrlPubMed
  28. ↵
    Nishino S, Mao J, Sampathkumaran R, and Shelton J (1998) Increased dopaminergic transmission mediates the wake-promoting effects of CNS stimulants. Sleep Res Online 1: 49-61.
    OpenUrlPubMed
  29. ↵
    Opp MR and Krueger JM (1994) Anti-interleukin-1 beta reduces sleep and sleep rebound after sleep deprivation in rats. Am J Physiol Regul Integr Comp Physiol 266: R688-R695.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Parmentier R, Anaclet C, Guhennec C, Brousseau E, Bricout D, Giboulot T, Bozyczko-Coyne D, Spiegel K, Ohtsu H, Williams M, et al. (2007) The brain H3-receptor as a novel therapeutic target for vigilance and sleep-wake disorders. Biochem Pharmacol 73: 1157-1171.
    OpenUrlCrossRefPubMed
  31. ↵
    Rouleau A, Ligneau X, Tardivel-Lacombe J, Morisset S, Gbahou F, Schwartz JC, and Arrang JM (2002) Histamine H3-receptor-mediated [35S]GTP gamma[S] binding: evidence for constitutive activity of the recombinant and native rat and human H3 receptors. Br J Pharmacol 135: 383-392.
    OpenUrlCrossRefPubMed
  32. ↵
    Schlicker E, Malinowska B, Kathmann M, and Gothert M (1994) Modulation of neurotransmitter release via histamine H3 heteroreceptors. Fundam Clin Pharmacol 8: 128-137.
    OpenUrlCrossRefPubMed
  33. ↵
    Schmitz Y, Lee CJ, Schmauss C, Gonon F, and Sulzer D (2001) Amphetamine distorts stimulation-dependent dopamine overflow: effects on D2 autoreceptors, transporters, and synaptic vesicle stores. J Neurosci 21: 5916-5924.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Sulzer D, Maidment NT, and Rayport S (1993) Amphetamine and other weak bases act to promote reverse transport of dopamine in ventral midbrain neurons. J Neurochem 60: 527-535.
    OpenUrlCrossRefPubMed
  35. ↵
    Taylor SJ, Michel AD, and Kilpatrick GJ (1992) In vivo occupancy of histamine H3 receptors by thioperamide and (R)-alpha-methylhistamine measured using histamine turnover and an ex vivo labeling technique. Biochem Pharmacol 44: 1261-1267.
    OpenUrlCrossRefPubMed
  36. ↵
    Torres GE, Gainetdinov RR, and Caron MG (2003) Plasma membrane monoamine transporters: structure, regulation and function. Nat Rev Neurosci 4: 13-25.
    OpenUrlCrossRefPubMed
  37. ↵
    Toyota H, Dugovic C, Koehl M, Laposky AD, Weber C, Ngo K, Wu Y, Lee DH, Yanai K, Sakurai E, et al. (2002) Behavioral characterization of mice lacking histamine H(3) receptors. Mol Pharmacol 62: 389-397.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Wadenberg ML, Kapur S, Soliman A, Jones C, and Vaccarino F (2000) Dopamine D2 receptor occupancy predicts catalepsy and the suppression of conditioned avoidance response behavior in rats. Psychopharmacology (Berl) 150: 422-429.
    OpenUrlCrossRefPubMed
  39. ↵
    Wayment H, Meiergerd SM, and Schenk JO (1998) Relationships between the catechol substrate binding site and amphetamine, cocaine, and mazindol binding sites in a kinetic model of the striatal transporter of dopamine in vitro. J Neurochem 70: 1941-1949.
    OpenUrlPubMed
View Abstract
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 365 (2)
Journal of Pharmacology and Experimental Therapeutics
Vol. 365, Issue 2
1 May 2018
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Correlation between ex Vivo Receptor Occupancy and Wake-Promoting Activity of Selective H3 Receptor Antagonists
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
Citation Tools
Research ArticleNEUROPHARMACOLOGY

Correlation between ex Vivo Receptor Occupancy and Wake-Promoting Activity of Selective H3 Receptor Antagonists

Siyuan Le, John A. Gruner, Joanne R. Mathiasen, Michael J. Marino and Hervé Schaffhauser
Journal of Pharmacology and Experimental Therapeutics June 1, 2008, 325 (3) 902-909; DOI: https://doi.org/10.1124/jpet.107.135343

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleNEUROPHARMACOLOGY

Correlation between ex Vivo Receptor Occupancy and Wake-Promoting Activity of Selective H3 Receptor Antagonists

Siyuan Le, John A. Gruner, Joanne R. Mathiasen, Michael J. Marino and Hervé Schaffhauser
Journal of Pharmacology and Experimental Therapeutics June 1, 2008, 325 (3) 902-909; DOI: https://doi.org/10.1124/jpet.107.135343
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Pharmacological Characterization of Synthetic Cannabinoids
  • The Nurr1 ligand,1,1-bis(3'-indolyl)-1-(p-chlorophenyl)methane, modulates glial reactivity and is neuroprotective in MPTP-induced parkinsonism
  • Pharmacological characterizations of H05, a novel potent serotonin and noradrenaline reuptake inhibitor with moderate 5-HT2A antagonist activity for anti-depression
Show more Neuropharmacology

Similar Articles

  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2018 by the American Society for Pharmacology and Experimental Therapeutics