2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxylic acids: novel 5-ht 3 receptor...

7
ARTICLE 2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxylic acids: Novel 5-HT 3 receptor antagonists with anxiolytic-like activity in rodent behavioral models Radhakrishnan Mahesh, Arghya Kusum Dhar, Ankur Jindal, and Shvetank Bhatt Abstract: The aim of this study was to investigate the anxiolytic potential of a series of novel carboxylic acid based 1,8 naphthyridines as 5-HT 3 receptor antagonists. The pA 2 values of all the compounds were determined against agonist 2-methyl- 5-hydroxytryptamine in longitudinal muscle myenteric plexus preparations from guinea pig ileum. Compounds with higher pA 2 values, particularly those greater than ondansetron, a standard 5-HT 3 receptor antagonist, and optimal log P values were screened in mice by using behavioral tests such as a light–dark (L/D) aversion test, elevated plus maze (EPM) test, and an open field test (OFT). In the L/D test, compounds 7a, 7b, 7d, 7e, and 7i (2 mg/kg body mass, intraperitoneal) significantly (P < 0.05) increased the latency time to leave the light compartment, total time spent in the light compartment, and the number of transitions between the light and dark compartments. Compounds 7a, 7d, 7f, 7h, and 7i (2 mg/kg, i.p.) significantly (P < 0.05) increased the time spent in the open arms and the number of entries into the open arms in the EPM test. In addition, compounds 7a, 7d, 7e, 7f, and 7h (2 mg/kg, i.p.) significantly (P < 0.05) increased the ambulation scores and the frequency of rearing in the OFT. Key words: 5-HT 3 antagonist, anxiolytic, 1;8-naphthyridine, L/D, EPM, OFT. Résumé : Le but de cette étude était d'examiner le potentiel anxiolytique d'une série de nouveaux carboxylates de 1,8- naphthyridines en leur qualité d'antagonistes du récepteur 5-HT 3 . Les valeurs de pA 2 de tous les composés ont été déterminées par rapport a ` l'agoniste 2-méthyl-5-hydroxytryptamine dans une préparation de plexus myentérique du muscle longitudinal de l'iléon de cobaye. Les composés dont les valeurs de pA 2 étaient plus élevées, particulièrement ceux dont les valeurs étaient supérieures a ` celle de l'ondasétron, un antagoniste standard du récepteur 5-HT 3 , et dont les valeurs du log P étaient optimales, ont été criblés chez la souris a ` l'aide de tests comportementaux, notamment le test d'aversion lumière/noirceur (light–dark aversion test, L/D), le labyrinthe en croix surélevé (elevated plus maze, EPM) et le test d'espace ouvert (open field test, OFT). Lors du test L/D, les composés 7a, 7b, 7d, 7e et 7i (2 mg/kg de masse corporelle, intrapéritonéale (i.p.) accroissaient significativement (P < 0,05) le temps de latence pour quitter le compartiment éclairé, le temps total passé dans le compartiment éclairé et le nombre de transitions entre les compartiments éclairés et sombres. Les composés 7a, 7d, 7f, 7h et 7i (2 mg/kg, i.p.) accroissaient significa- tivement le temps passés dans les corridors ouverts et le nombre d'entrées dans les corridors ouverts lors du test EPM. De plus, les composés 7a, 7d, 7e, 7f et 7h (2 mg/kg, i.p.) accroissaient significativement (p < 0,05) les scores d'ambulation et le nombre de cabrages lors de l'OFT. [Traduit par la Rédaction] Mots-clés : antagoniste du 5-HT 3 , anxiolytique, 1;8-naphthyridine, L/D, EPM, OFT. Introduction Anxiety, a normal response to stress, is a cardinal symptom of many psychiatric illnesses closely allied with appropriate fear that in many cases can become an important pathological, dis- abling, and lasting disorder (Barkow et al. 2004; Airaksinen et al. 2005). Anxiety disorders are the most common psychiatric disor- ders in the US, with lifetime prevalence in approximately 30% of the population, and affecting 1/8 of the total population world- wide (Kessler et al. 2005). Benzodiazepines (BZDs) (diazepam and related drugs) are a class of drug generally prescribed as medica- tion for the treatment of anxiety disorders. However, this class of drugs has a wide side effects profile, including sedation and de- pendence, as well as cognitive and psychomotor impairment (Lader and Morton 1991; Whiting 2006). Besides BZDs, selective serotonin reuptake inhibitors (SSRIs) (Hidalgo and Davidson 2000; Borsini et al. 2002) and MAO-A inhibitors (De Angelis and Furlan 2000; Mai et al. 2003) are effective in treating a wide spectrum of anxiety disorders. Despite their therapeutic actions, weight changes, insomnia, drowsiness or sedation, headache, and sexual dysfunction (Dording et al. 2002; Priest et al. 1992) are the reported side effects associated with SSRIs and MAO inhibitors. This prompted many researchers to explore for new anxiolytics with fewer unwanted effects. The pathophysiology and precise mechanism of anxiety re- mains unclear. Nevertheless, monoaminergic system (serotonin (5-HT) and nor-epinephrine (NE)) in the brain have been postulated to play an important role in the pathophysiology of anxiety disor- ders (Zhang et al. 2004). Few preclinical or clinical studies provide evidence to support the hypothesis that anxiety may be associated with an alteration or dysfunction of the serotonergic system in the central nervous system (CNS) (Aghajanian et al. 1990; Revelli et al. 1998; Jindal et al. 2013). Thus, modulation of the serotonergic system may be implicated as a promising mechanism for the ac- tion of anxiolytic drugs (Zhang et al. 2004; Jindal et al. 2013). The 5-HT 3 receptor is a ligand-gated ion channel that mediates the release of monoamine neurotransmitters (Wolf 2000). The 5-HT 3 receptor has been identified in the CNS (Pandey et al. 2008). An- tagonists of this receptor, such as ondansetron, granisetron, Received 3 April 2013. Accepted 29 May 2013. R. Mahesh, A.K. Dhar, A. Jindal, and S. Bhatt. Department of Pharmacy, FD-III, Birla Institute of Technology & Science, Pilani, Rajasthan 333 031, India. Corresponding author: Arghya Kusum Dhar (e-mail: [email protected]). 848 Can. J. Physiol. Pharmacol. 91: 848–854 (2013) dx.doi.org/10.1139/cjpp-2013-0134 Published at www.nrcresearchpress.com/cjpp on 11 June 2013. Can. J. Physiol. Pharmacol. Downloaded from www.nrcresearchpress.com by Laurentian University on 11/01/13 For personal use only.

Upload: shvetank

Post on 20-Dec-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

ARTICLE

2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxylic acids:Novel 5-HT3 receptor antagonists with anxiolytic-like activity in rodentbehavioral modelsRadhakrishnan Mahesh, Arghya Kusum Dhar, Ankur Jindal, and Shvetank Bhatt

Abstract: The aim of this study was to investigate the anxiolytic potential of a series of novel carboxylic acid based 1,8naphthyridines as 5-HT3 receptor antagonists. The pA2 values of all the compounds were determined against agonist 2-methyl-5-hydroxytryptamine in longitudinalmusclemyenteric plexus preparations from guinea pig ileum. Compounds with higher pA2

values, particularly those greater than ondansetron, a standard 5-HT3 receptor antagonist, and optimal log P values werescreened in mice by using behavioral tests such as a light–dark (L/D) aversion test, elevated plus maze (EPM) test, and an openfield test (OFT). In the L/D test, compounds 7a, 7b, 7d, 7e, and 7i (2 mg/kg body mass, intraperitoneal) significantly (P < 0.05)increased the latency time to leave the light compartment, total time spent in the light compartment, and the number oftransitions between the light and dark compartments. Compounds 7a, 7d, 7f, 7h, and 7i (2 mg/kg, i.p.) significantly (P < 0.05)increased the time spent in the open arms and the number of entries into the open arms in the EPM test. In addition, compounds7a, 7d, 7e, 7f, and 7h (2 mg/kg, i.p.) significantly (P < 0.05) increased the ambulation scores and the frequency of rearing in the OFT.

Key words: 5-HT3 antagonist, anxiolytic, 1;8-naphthyridine, L/D, EPM, OFT.

Résumé : Le but de cette étude était d'examiner le potentiel anxiolytique d'une série de nouveaux carboxylates de 1,8-naphthyridines en leur qualité d'antagonistes du récepteur 5-HT3. Les valeurs de pA2 de tous les composés ont été déterminéespar rapport a l'agoniste 2-méthyl-5-hydroxytryptamine dans une préparation de plexus myentérique du muscle longitudinal del'iléon de cobaye. Les composés dont les valeurs de pA2 étaient plus élevées, particulièrement ceux dont les valeurs étaientsupérieures a celle de l'ondasétron, un antagoniste standard du récepteur 5-HT3, et dont les valeurs du log P étaient optimales,ont été criblés chez la souris a l'aide de tests comportementaux, notamment le test d'aversion lumière/noirceur (light–darkaversion test, L/D), le labyrinthe en croix surélevé (elevated plus maze, EPM) et le test d'espace ouvert (open field test, OFT). Lors du testL/D, les composés 7a, 7b, 7d, 7e et 7i (2mg/kg demasse corporelle, intrapéritonéale (i.p.) accroissaient significativement (P< 0,05)le temps de latence pour quitter le compartiment éclairé, le temps total passé dans le compartiment éclairé et le nombre detransitions entre les compartiments éclairés et sombres. Les composés 7a, 7d, 7f, 7h et 7i (2 mg/kg, i.p.) accroissaient significa-tivement le temps passés dans les corridors ouverts et le nombre d'entrées dans les corridors ouverts lors du test EPM. De plus,les composés 7a, 7d, 7e, 7f et 7h (2 mg/kg, i.p.) accroissaient significativement (p < 0,05) les scores d'ambulation et le nombre decabrages lors de l'OFT. [Traduit par la Rédaction]

Mots-clés : antagoniste du 5-HT3, anxiolytique, 1;8-naphthyridine, L/D, EPM, OFT.

IntroductionAnxiety, a normal response to stress, is a cardinal symptom of

many psychiatric illnesses closely allied with appropriate fearthat in many cases can become an important pathological, dis-abling, and lasting disorder (Barkow et al. 2004; Airaksinen et al.2005). Anxiety disorders are the most common psychiatric disor-ders in the US, with lifetime prevalence in approximately 30% ofthe population, and affecting 1/8 of the total population world-wide (Kessler et al. 2005). Benzodiazepines (BZDs) (diazepam andrelated drugs) are a class of drug generally prescribed as medica-tion for the treatment of anxiety disorders. However, this class ofdrugs has a wide side effects profile, including sedation and de-pendence, as well as cognitive and psychomotor impairment(Lader and Morton 1991; Whiting 2006). Besides BZDs, selectiveserotonin reuptake inhibitors (SSRIs) (Hidalgo andDavidson 2000;Borsini et al. 2002) and MAO-A inhibitors (De Angelis and Furlan2000; Mai et al. 2003) are effective in treating a wide spectrum ofanxiety disorders. Despite their therapeutic actions, weightchanges, insomnia, drowsiness or sedation, headache, and sexual

dysfunction (Dording et al. 2002; Priest et al. 1992) are the reportedside effects associated with SSRIs and MAO inhibitors. Thisprompted many researchers to explore for new anxiolytics withfewer unwanted effects.

The pathophysiology and precise mechanism of anxiety re-mains unclear. Nevertheless, monoaminergic system (serotonin(5-HT) and nor-epinephrine (NE)) in the brain have been postulatedto play an important role in the pathophysiology of anxiety disor-ders (Zhang et al. 2004). Few preclinical or clinical studies provideevidence to support the hypothesis that anxietymay be associatedwith an alteration or dysfunction of the serotonergic system inthe central nervous system (CNS) (Aghajanian et al. 1990; Revelliet al. 1998; Jindal et al. 2013). Thus, modulation of the serotonergicsystem may be implicated as a promising mechanism for the ac-tion of anxiolytic drugs (Zhang et al. 2004; Jindal et al. 2013). The5-HT3 receptor is a ligand-gated ion channel that mediates therelease of monoamine neurotransmitters (Wolf 2000). The 5-HT3receptor has been identified in the CNS (Pandey et al. 2008). An-tagonists of this receptor, such as ondansetron, granisetron,

Received 3 April 2013. Accepted 29 May 2013.

R. Mahesh, A.K. Dhar, A. Jindal, and S. Bhatt. Department of Pharmacy, FD-III, Birla Institute of Technology & Science, Pilani, Rajasthan 333 031, India.

Corresponding author: Arghya Kusum Dhar (e-mail: [email protected]).

848

Can. J. Physiol. Pharmacol. 91: 848–854 (2013) dx.doi.org/10.1139/cjpp-2013-0134 Published at www.nrcresearchpress.com/cjpp on 11 June 2013.

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.

dolasetron, and tropisetron are commonly used for the treatmentof radiotherapy- or chemotherapy-induced nausea and vomiting(CINV) in patients with cancer (Israili 2001; Walstab et al. 2010).

Numerous studies have indicated that 5-HT3 receptors are ex-pressed in the areas of the brain implicated in anxiety disorders.(Jones 1990; Costall and Naylor 1992; Simpson et al. 1992; Olivieret al. 2000; Walstab et al. 2010). Animal studies have revealed that5-HT3 receptor antagonists exert an anxiolytic effect by blockinglimbic (hippocampus and amygdale) hyperactivity responses (Griebel1995; Millan 2003; Rajkumar and Mahesh 2010). In addition, stud-ies of 5-HT3 knockout mice also supported the involvement of5-HT3 receptors in anxiety, which revealed that 5-HT3 receptorregulates anxiety-related behaviour (Kelley et al. 2003).

Thus, this study was intended to investigate the anxiolytic-likeeffect of a series of carboxylic acid based 1,8 naphthyridines thatwere designed and synthesized as bioisosteric analogs of our ear-lier synthesized nitrile-based 1,8 naphthyridines (Mahesh et al.2004). The compounds were tested in our laboratory for their5-HT3 receptor antagonistic potential in isolated guinea pig il-eum, and the pA2 values were determined against 2-methyl-5-hydroxytryptamine, a 5HT3 receptor agonist (Table 1). In thisstudy, the compounds were selected from the series based ontheir optimal log P and pA2 value, particularly if it was greaterthan the standard 5-HT3 receptor antagonist, ondansetron(Table 1). A behavioral test battery, including the light–dark (L/D)test, elevated plus maze (EPM) test, and the open field exploratorytest (OFT) were used to explore the anxiolytic-like activities of theselected compounds.

Materials and methods

AnimalsSwiss albino mice (25–35 g) were obtained from Hissar Agricul-

tural University (Hissar, Haryana, India) and maintained under

standard conditions: 12.00 h (light) −12.00 h (dark) cycle, temper-ature of 23 ± 2 °C, and relative humidity of 60% ± 10%. The rodentswere housed in standard polycarbonate cages and given free ac-cess to food (standard pellet chow) and filtered water. The animalswere used only once for each experiment and were acclimatizedto the experimental room for 1 h before testing. Experiments onanimals were approved by the Institutional Animal Ethics Com-mittee of the Birla Institute of Technology & Science, Pilani, India(Protocol No. IAEC/RES/14/04).

DrugsDiazepam was obtained from Lancaster chemicals (UK). Ondanse-

tron was obtained from Natco Pharmaceuticals, Hyderabad, India.All of the other chemicals used for synthesis were of analyticalgrade. Diazepam and ondansetron were used as reference stan-dards. All of the NCEs, diazepam, and ondansetron were freshlyprepared before use by dissolving in distilled water. Diazepam(2mg/kg bodymass), ondansetron (2mg/kg), and NCEs (1 or 2mg/kg)were administered intraperitoneally (i.p.) to mice 30 min beforestarting the behavioural observation in each test.

ChemistryThe title compounds 2-(4-substituted piperazin-1-yl)-1,8-

naphthyridine-3-carboxylic acids were designed as bioisostericanalogs of previously synthesized 2-(4-substituted piperazine1-yl)-1,8-naphthyridine-3-carbonitriles (Mahesh et al. 2004). Encouraged bythe promising 5-HT3 antagonism profile of our earlier synthesizednitrile-based 1,8 naphthyridines, it was proposed to design ana-logs. Consequently, the nitrile group of the previously synthe-sized molecules was replaced with its nonclassical bioisosterecarboxylic acid group (Lima and Barreiro 2005) to generate 2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxlic acid. Thecompounds were synthesized via the sequence of reactions de-picted in the Supplementary data1 (Fig. S1, Table S1). The startingmaterial, 2-aminonicotinaldehyde (3), was synthesized by heatingnicotinamide and ammonium sulphamate in a neat solvent-freecondition to obtain a pyrido-pyrimidine intermediate, which washydrolyzed by heating with dilute HCl, according to the methodsreported in the literature (Majewicz and Caluwe 1974). Refluxing2-aminonicotinaldehyde with diethyl malonate in ethanol, with afew drops of piperidine as the catalyst, afforded the intermediateethyl 2-oxo-1,2-dihydro-1,8-naphthyridine-3-carboxylate (4) (Xiaoet al. 2010). Compound 4was subjected to refluxing in POCl3 witha catalytic amount of DMF to furnish ethyl 2-chloro-1,2-dihydro-1,8-naphthyridine-3-carboxylate (5). Nucleophilic displacementreaction of the chlorine atom of compound 5 was carried outby various substituted piperazines in refluxing acetonitrilewith K2CO3 to obtain the compounds 6a–6o, i.e., substitutionwith phenyl piperazine, resulted in ethyl 2-(4-phenylpiperazin-1-yl)-1,8-naphthyridine-3-carboxylate. Finally, the ester derivatives(6a–6o) were stirred with aqueous NaOH in ethanol. Upon comple-tion of the reaction, the alcohol was evaporated and the reactionmixture was acidified very carefully with a weak acid, such as citricacid, up to pH 4–5 to avoid protonation at the N4 nitrogen of thepiperazine ring, and this furnished the title compounds 7a–7o.

The structures of the synthesized compounds were confirmedby their spectral data.

The spectral data for one representative compound, 2-(4-(4-nitrophenyl) piperazin-1-yl)-1, 8-naphthyridine-3-carboxylic acid(7d), are presented below.

FT-IR (KBr, cm−1): 3290 (broad str of acid –OH), 2931, 2850, 2644,2357, 1719 (broad str of acid C=O), 1625, 1510, 1462, 1350, 1296, and1240. 1H NMR, CDCl3, � (ppm): 8.75–8.73 (dd, 1H, J = 4.8, 2 Hz, C7-Hof naphthyridine ring), 8.12 (s, 1H, C4-H of naphthyridine ring),8.07–8.04 (m, 2H, C6-H of naphthyridine ring), 7.98–7.96 (m, 1H,

1Supplementary data are available with the article through the journal Web site at http://nrcresearchpress.com/doi/suppl/10.1139/cjpp-2013-0134.

Table 1. The pA2 and log P values of novel 2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxylic acids.

(7a–7o) Basic structure

Compounda Rb pA2c Log Pd

7a C6H5 7.6 3.357b CH2C6H5 6.7 2.897c CH2CH2C6H5 6.5 3.057d p-NO2C6H4 7.2 3.247e m-ClC6H4 7.3 4.017f p-ClC6H4 6.0 4.017g o-ClC6H4 4.6 4.017h o-OCH3C6H4 6.6 3.267i p-OCH3C6H4 7.0 3.267j m-OCH3C6H4 5.0 3.267k p-CH3C6H4 5.9 3.577l o-CH3C6H4 4.4 3.577m CH3 6.3 1.187n CH2CH3 5.4 1.607o CH(CH3)2 5.2 2.06

aA series of compounds synthesized in the laboratory and coded 7a to 7o.bGroup attached to the N4 of piperazine ring of the basic structure.cThe pA2 values are calculated against 2-methyl-5-hydroxytryptamine.dThe log P values are calculated using JMEMolecular Editor (Courtesy of Peter

Ertl, Novartis).

Mahesh et al. 849

Published by NRC Research Press

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.

C5-H of naphthyridine ring), 7.18–7.15 (m, 2H, phenyl), 6.84–6.82(m, 2H, phenyl), 4.0–3.95 (m, 4 H, methylene protons adjacent toN1 of piperazine ring), 3.62–3.58 (m, 4H, methylene protons adja-cent to N4 of piperazine ring). MS (ESI –ve mode): m/z (%) = 378.32(M-H+, 100).

Behavioural screening

Light–dark aversion testThe L/D apparatus comprised a box divided into 2 separate com-

partments, occupying 2/3 and 1/3 of the total size, respectively.The larger compartment (light compartment) was illuminated bya 60 W bulb, while the smaller (dark compartment) was entirelyblack and enclosed under a dark cover. The L/D compartmentswere separated by a partition with a tunnel to allow the mousepassage from one compartment to the other (Mi et al. 2005). At thebeginning of the test, the mouse was placed individually at thecenter of the light compartment facing towards the tunnel andwas allowed to explore the entire apparatus for 5 min. The behav-ioral parameters such as latency time to leave the light compart-ment, total time spent in the light compartment, and number oftransitions between the light and dark compartments were ob-served and recorded. A compartment entry was considered validwhen all 4 of the animal's paws were inside that chamber. Theapparatus was thoroughly cleaned with 70% ethanol after eachtrial.

Elevated plus mazeThe EPM test was first evaluated for rats (Pellow et al. 1985) and

later adapted for mice (Lister 1987). In brief, the apparatus con-sisted of a wooden maze with 2 enclosed arms (30 cm × 5 cm ×15 cm) and 2 open arms (30 cm × 5 cm × 0.25 cm) that extendedfrom a central platform (5 cm × 5 cm) to form a “plus sign”. Theplus-maze apparatus was elevated to a height of 25 cm and placedinside a sound-attenuated room. The trial was started by placing amouse on the central platformof themaze facing its head towardsan open arm. The behavioral performances recorded during a5 min test period were as follows: percentage open arm entries,percentage time spent in open arm, and total entries (Klodzinskaet al. 2004). Entry into an arm was considered valid only when all4 paws of the mouse were inside that arm (Biala and Kruk 2008).

The animal's activities were observed and readings were recorded.The apparatus was thoroughly cleaned with 70% ethanol aftereach trial.

Open field testThe apparatus consisted of a wooden box (60 cm × 60 cm ×

30 cm) with the floor divided into 16 squares (15 cm × 15 cm)squares by black parallel and intersecting lines. The apparatuswas illuminated with 60Wbulb suspended 100 cm above it. At thebeginning of the test, the mouse was placed individually at thecenter of the square arena. The ambulation scores (number ofsquares crossed) and frequency of rearing (standing upright onthe hind legs) were recorded for a 5 min period. After each indi-vidual test session the floor was thoroughly cleaned with 70%ethanol.

Statistical analysisResults are the mean ± SEM. The data were analyzed using

one-way analysis of variance (ANOVA) followed by Dunnett's testin Graph pad prism 3. The level of statistical significancewas fixedat P < 0.05.

Results

Light–dark aversion testThe effects of NCEs, ondansetron, and diazepam on the behav-

iour of mice in the L/D test are presented in Table 2. Treatmentwith compounds 7a–7f, 7h, 7i, and 7m (1 or 2 mg/kg body mass,i.p.), ondansetron (2 mg/kg, i.p.), and diazepam (2 mg/kg, i.p.) sig-nificantly (P < 0.05) increased the time spent by the mice in the litarea as well as the number of transitions, compared with thevehicle control. In addition, diazepam (2 mg/kg, i.p.) treatmentsignificantly (P < 0.05) increased the latency to enter the darkchamber. Treatment with compounds 7a, 7b, 7d, 7e, or 7i (only at2 mg/kg, i.p.) significantly (P < 0.05) increased the latency to enterthe dark chamber. The lower dose of NCEs (1 mg/kg, i.p.) did notproduce any significant change in the latency to leave the litcompartment, when compared with the vehicle control. Theanxiolytic-like effect was more pronounced at the higher dose(2 mg/kg) for all of the tested compounds.

Table 2. Effect of 2-(4-substituted piperazin-1-yl) 1,8-naphthyridine 3-carboxylic acids, ondansetron, and diazepam in a light–darkmouse model.

Latency (s)Time spent in lightchamber (s) Number of transitions

Dose (mg/kg) Dose (mg/kg) Dose (mg/kg)

Compound 2.0 1.0 2.0 1.0 2.0 1.0

7a 48.17±19.45* 40.5±13.84 127.67±12.76* 116.83±7.5* 13.33±1.40* 12.67±0.88*7b 46±7.76* 27.17±9.78 146.33±9.88* 131.75±22.15* 11.20±1.74* 8.83±2.42*7c 6.00±2.07 7.17±2.84 136.67±30.36* 107.17±26.33* 13.5±1.84* 15.6±2.22*7d 36.60±5.15* 26.17±7.80 141.67±13.91* 124.33±10.79* 15.00±1.65* 13.83±1.12*7e 49.20±9.35* 35.20±10.40 131.33±12.14* 129.33±10.44* 16.67±2.48* 15.33±1.37*7f 12.50±3.38 10.00±1.83 147.00±18.64* 90.67±15.03* 17.67±2.21* 13.17±0.83*7g — — — — — —7h 23.33±1.87 19.76±6.84 98.33±10.67* 87.76±16.04* 16.67±2.10* 11±2.19*7i 45.50±8.28* 27.17±9.34 158.67±19.45* 96.33±20.09* 14.00±1.31* 10.00±2.29*7j — — — — — —7k 12.00±2.79 12.50±4.05 87.40±15.12* 71.33±14.61 9.60±1.94* 13.40±2.12*7l — — — — — —7m 10.33±3.91 9.83±1.91 124.75±16.03* 83.50±10.21* 13.17±2.62* 14.60±3.19*7n — — — — — —7o — — — — — —Ondansetron 18±5.21 — 123±5.50* — 11.23±4.00* —Diazepam 73.83±8.90* — 164.83±5.50* — 17.16±9.00* —Control 24.17±3.06 — 63.40±5.29 — 4.67±0.87 —

Note: Values are the mean ± SEM; *, P < 0.05 when compared with the vehicle-treated group (control); n = 6 mice per group.

850 Can. J. Physiol. Pharmacol. Vol. 91, 2013

Published by NRC Research Press

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.

Elevated plus mazeThe effects of NCEs, ondansetron, or diazepam on the behavior

ofmice in the EPM test are summarized in Table 3. Treatmentwithcompounds 7a, 7e, 7f, and 7h (1 or 2 mg/kg, i.p.), ondansetron(2 mg/kg, i.p.), or diazepam (2 mg/kg, i.p.) significantly (P < 0.05)increased the time spent in the open arm, when compared withthe vehicle control. Compounds 7d, 7i, and 7m (2mg/kg, i.p., only)significantly (P < 0.05) increased the open-arm entries as wellas the time spent in the open arm. Treatment with diazepam(2mg/kg, i.p.) or ondansetron (2 mg/kg, i.p.) significantly (P < 0.05)increased the open-arm entries and time spent in the open arm,when compared with the vehicle control. Compound 7c onlyshowed a significant (P < 0.05) increase in time spent in the openarms at 2 mg/kg, i.p. The lower dose of the test compounds(1 mg/kg, i.p.) did not induce a significant (P < 0.05) change in theopen-arm entries, when compared with the vehicle control. Treat-ment with compound 7a (1 or 2 mg/kg, i.p.) significantly increasedthe percentage of open-arm entries as well as the percentage of timespent in the open arm, when compared with the vehicle control.

Open field testThe results of the OFT are shown in Table 4. Treatment with

compounds 7a or 7h (1 or 2 mg/kg, i.p. for both), 7d, 7e, or 7f(2 mg/kg, i.p., only), diazepam (2 mg/kg, i.p.), or ondansetron(2 mg/kg, i.p.) significantly (P < 0.05) increased the ambulationscores, when compared with the vehicle control. Moreover, com-pounds 7f and 7h (1 or 2 mg/kg, i.p., for both) and compounds 7a,7d, 7i, and 7m (2mg/kg, i.p., only) significantly (P< 0.05) increasedthe frequency of rearing, compared with the vehicle control.

DiscussionThe present behavioral investigation with various animal

models of anxiety revealed the anxiolytic like effect of novel1,8 naphthyridine derivatives as 5-HT3 receptor antagonists. A3-component model for the interaction of arylpiperazine type li-gands with the 5-HT3 receptor was derived (Cappelli et al. 2002)and it demonstrated that themost essential structural features for

the interaction of arylpiperazines with 5-HT3 receptor were asfollows: (i) the N4 piperazine nitrogen atom, (ii) the heterocylicnitrogen atom, and (iii) the bicyclic heteroaromatic system(Cappelli et al. 2002). All of the compounds (Table 1) were synthe-sized according to a previously described pharmacophore (Rosenet al. 1990; Cappelli et al. 2002; Klodzinska et al. 2004) for the5-HT3 receptor antagonists. The putative anxiolytic activities ofthe synthesized compounds were determined by L/D test, EPMtest, and OFT. The time spent in the lit chamber and in the open

Table 3. Effect of 2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxylic acids, ondansetron, and diazepam on the behav-ior of mice in an elevated plus maze test.

No. of entries Time spent (s)

Treatmentgroups

Dose(mg/kg) Open arm Closed arm Open arm Closed arm % OAE % TSOA

7a 2 4.33±0.48* 6.23±0.50 107.13±4.94* 192.87±2.14 41.62±3.89* 35.71±2.71*1 2.83±0.51 5.50±0.96 105.17±6.90* 194.83±4.90 33.97±2.56* 35.05±3.64*

7b 2 3.17±0.68 6.83±0.93 35.83±2.28 264.17±2.28 30.89±2.32 11.94±1.041 2.29±1.08 6.86±1.03 32.14±2.22 267.86±3.95 25.02±3.53 10.71±0.98*

7c 2 2.04±1.12 6.21±1.86 53.15±12.29* 246.85±4.72 24.72±3.66 17.71±6.09*1 1.21±0.67 5.50±1.01 32.13±10.14 267.87±10.11 18.03±7.51 10.71±4.04

7d 2 5.83±1.04* 10.83±0.83 54.5±5.74* 245.5±8.95 34.09±4.13 18.16±1.91*1 2.00±0.57 3.33±0.56 26.33±7.80 273.67±10.11 30.73±7.64 8.78±2.60

7e 2 3.09±1.48 7.16±2.13 51.14±5.52* 248.86±6.95 30.14±5.53 17.04±2.98*1 2.83±2.67 7.10±1.51 44.83±2.04* 255.17±1.11 28.49±7.51 14.94±4.04

7f 2 3.83±0.44* 7.17±1.28 44.33±4.69* 255.67±4.69 36.18±1.78 14.77±1.56*1 2.33±0.67 5.50±2.01 37.83±0.04* 262.17±2.11 29.75±7.51 12.61±2.04

7h 2 5.33±0.99* 7.17±1.07 69.00±13.38* 231.00±8.35 39.84±2.32* 23.00±3.48*1 3.11±0.67 6.50±1.01 54.83±1.14* 245.17±2.51 32.36±7.51 18.27±4.04

7i 2 3.5±0.61* 5.50±1.02 49.17±11.03* 250.83±4.95 30.61±3.45 16.39±3.67*1 3.0±0.50 5.93±1.01 28.00±13.93 272.00±15.11 33.59±4.11 9.33±1.04

7k 2 3.29±1.28 5.16±1.53 41.14±5.42 258.86±8.95 38.93±5.53 13.71±2.98*1 2.43±1.67 5.10±1.21 34.83±2.14 265.17±1.11 32.27±7.51 11.61±2.34

7m 2 3.20±0.08* 7.86±1.03 51.14±5.52* 248.86±6.95 28.93±5.53 17.04±2.18*1 2.13±0.67 7.10±1.51 30.83±12.14 269.17±12.11 23.07±3.51 10.27±2.04

Ondansetron 2 4.83±0.37* 6.67±1.22 41.50±4.35* 258.50±2.32 39.84±3.51 13.83±1.44*Diazepam 2 6.5±1.23* 4.33±0.71 124.5±11.8* 175.5±8.15 60.01±5.53* 41.50±4.38*Control 2.29±0.68 4.4±0.73 26.30±9.94 281.70±9.94 34.23±2.53 8.76±2.16

Note: All values are the mean ± SEM; *, P < 0.05 when compared with the vehicle-treated group; n = 6 mice per group; %OAE, percentage ofopen-arm entries; %TSOA, percent time spent in the open arm in seconds.

Table 4. Effect of 2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carboxylic acids, ondansetron, and diazepam on the behavior of micein an open-field test.

Ambulation score Rearing number

Dose (mg/kg) Dose (mg/kg)

Compound 2.0 1.0 2.0 1.0

7a 331.46±9.81* 319.00±8.30* 17.62±2.89* 14.83±6.007b 249.00±1.48 236.43±3.30 15.17±7.50 08.26±5.537c 304.49±2.92 302.08±8.39 15.32±3.66* 15.17±7.517d 351.50±15.60* 285.00±7.36 21.83±2.45* 18.17±5.517e 328.23±3.50* 253.66±2.35 14.86±6.95 11.17±3.517f 327.00±9.79* 280.16±6.45 19.83±4.04* 15.17±3.11*7g — — — —7h 319.66±7.09* 313.58±5.99* 16.50±2.11* 21.17±7.51*7i 303.67±2.17 291.17±7.80 18.50±2.61* 11.17±7.517j — — — —7k 283.15±2.61 277.60±5.89 12.26±5.53 05.17±2.517l — — — —7m 281.06±4.06 279.30±6.39 18.26±2.43* 12.17±7.917n — — — —7o — — — —Ondansetron 333.17±25.32* — 20.5±3.31* —Diazepam 467.50±15.19* — 40.50±1.02* —Control 276.50±29.23 — 9.83±2.18 —

Note: Values are themean ± SEM; *, P < 0.05 when compared with the vehicle-treated group (control); n = 6 mice per group.

Mahesh et al. 851

Published by NRC Research Press

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.

arm was measured during the L/D test and EPM test respectively,whereas the ambulation score and rearing frequency wererecorded in the OFT. All of these models of anxiety are quitesensitive and relatively specific to anxiolytics, and have beenwidely used to screen NCEs for their anxiolytic potential (Borsiniet al. 1989; Costall et al. 1989, 1993; Kilfoil et al. 1989; Barnes et al.1992; Hogg 1996).

The L/D test is based on the aversive properties of light (Angelisand Furlan 2000; Belzuing et al. 1987) and uses as its index ofanxiety the time spent in the lit area as opposed to the time spentin the dark area. The data from this study is consistent with pre-vious studies (Young and Johnson 1991) showing that the best wayto determine the effect of an anxiolytic agent in mice is the timespent in the lit area, while a decrease in the time spent in the litarea as well as decrease in the number of transitions between litand dark area are characteristic of an anxiogenic response (Laland Emmett-Oglesby 1983; De Angelis 1992). Treatment with com-pounds 7a, 7d, 7e, 7f, diazepam, or ondansetron significantlyincreased the number of transitions as well as the time spent inthe lit area, suggesting anti-anxiety-like effect that is also shownby various anxiolytics and 5-HT3 receptor antagonists (Olivieret al. 2000). It can be observed that the compounds containingelectron withdrawing substituents in the phenyl ring, such as4-nitro (compound 7d), 3-chloro (compound 7e), 4-chloro (com-pound 7f), and an electron releasing substituent such as the4-methoxy (compound 7i) group, showed better activity thanother compounds in the L/D test.

EPM is considered a well established model of unconditionedanxiety for detecting anxiolytic/anxiogenic-like activity by inves-tigating aspects of physiological and pharmacological behavior.In the EPM test, increased number of entries and time spent in theopen arms of the maze are the most reliable indicators of de-creased anxiety or for indicating the anxiolytic-like activity of acompound, while anxiogenic substances have the opposite effect(Fernandes and File 1996; Dubinsky et al. 2002; Sienkiewicz-Jaroszet al. 2003). In our study, treatment with these 1,8 naphthyridinederivatives produced anxiolytic-like effects in the EPM test, asevidenced by a significant increase in both open-arm entries andtime spent in open arms, compared with the vehicle control.These results are consistent with previous studies that suggest5-HT3 receptor antagonists increased the number of entries andtime spent in the open arms, and exhibited anxiolytic activity inthe EPM test (Blackburn et al. 1993; Artaiz et al. 1995; Hewlett et al.2001; Zhang et al. 2001). In addition, diazepam was used as a stan-dard reference, and it also showed an anxiolytic effect in theEPM test. It can be noted that analogs containing an electron-withdrawing group such as 4-nitro (compound 7d), 3-chloro (com-pound 7e), and 4-chloro (compound 7f), as well as an electronreleasing group such as 2-methoxy (compound 7h), 4-methoxy(compound 7i), and 4-methyl (compound 7k) groups in the phenylring, showed lower activity compared with the unsubstitutedphenyl ring counterpart (compound 7a) in the EPM test. The openfield test is widely used for the screening of anxiolytic/anxiogenicdrugs. Normal aversion of a rodent to the brightly lit open areaproduces anxiety and fear, which is characterized by alteration inthe behavioral parameters of the animal in the open field. Previ-ous reports have suggested that anxiolytic compounds have atendency to reduce the fearful behaviors of rodents in the OFT(Mechan et al. 2002). Treatment with compounds 7a, 7d, or 7hincreased the ambulation score and rearing frequency in the OFT,indicating the anxiolytic effect of these compounds. Analogs thatcontain electron-withdrawing substituents in the phenyl ring,such as 4-nitro (compound 7d), 3-chloro (compound 7e), 4-chloro(compound 7f), and electron releasing substituents such as2-methoxy (compound 7h), induced better anxiolytic-like activitythan other compounds, in the OFT. In general, compounds withconsiderably higher pA2 values, such as 7a (pA2:7.6), 7d (pA2:7.2),7e (pA2:7.3), 7h (pA2:6.6), and 7i (pA2:7.0) exhibited significant

anxiolytic-like activity in the L/D test, EPM test, and OFT. Thepreliminary attributes of the anxiolytic-like activities of thesenovel 1,8-naphthyridine derivatives could be (i) log P values of 2.01to 3.35, which are optimal for blood–brain barrier permeability(Ter Laak et al. 1994), (ii) 5-HT3 receptor antagonism (pA2 value)greater than ondansetron (6.6), which is a standard 5-HT3 receptorantagonist.

In conclusion, the results from this study demonstrate the an-xiolytic activity of 1,8 naphthyridine carboxylic acid derivatives in3 validated animal models of anxiety. The precise mechanismbehind the anti-anxiety like effect of these 1,8 naphthyridine de-rivatives remains to be fully explored. However, an increase in thelevel of serotonin through blockade of the 5-HT3 receptor (Schafer1999) could possibly contribute to the anxiolytic-like effect ofthese compounds (Greenshaw 1992). The 5-HT3 receptor locatedwithin a neuro-anatomical region might be indirectly regulatingthe 5-HT transmission (Kilpatrick et al. 1989). The blockade of5-HT3 receptors facilitates 5-HT neurotransmission (Rajkumar andMahesh 2010). 5-HT3 receptor antagonists block the postsynaptic5-HT3 receptors that mediate a fast excitatory potential in thelimbic brain regions (Sugita et al. 1992). Although the cascade ofcellular events that follow this fast transmission blockade re-mains unclear, an overall anti-anxiety-like behaviour is plausible(Rajkumar andMahesh 2010). Therefore, our findings reiterate thedevelopment of 5-HT3 receptor antagonists as a possible alterna-tive approach for the treatment of anxiety disorders. Hopefully,future studies with molecular techniques will help in better un-derstanding of the possible mechanism underlying the anxiolyticlike effect of these 1,8 naphthyridine carboxylic acid derivatives atthe cellular level, which may lead to agents that are useful forcontrolling anxiety disorders.

AcknowledgementsThis study was financially supported by grants from the Indian

Council of Medical Research (ICMR), New Delhi, India.

ReferencesAghajanian, G.K., Sprouse, J.S., Sheldon, P., and Rasmussen, K. 1990. Electro-

physiology of the central serotonin system: receptor subtypes and transducermechanisms. Ann. N.Y. Acad. Sci. 600: 93–103. doi:10.1111/j.1749-6632.1990.tb16875.x. PMID:2123618.

Airaksinen, E., Larsson,M., and Forsell, Y. 2005. Neuropsychological functions inanxiety disorders in population-based samples: evidence of episodicmemorydysfunction. J. Psychiatr. Res. 39(2): 207–214. doi:10.1016/j.jpsychires.2004.06.001. PMID:15589570.

Angelis, L.D., and Furlan, C. 2000. The anxiolytic-like properties of two selectiveMAOIs, moclobemide and selegiline, in a standard and an enhanced light/dark aversion test. Pharmacol. Biochem. Behav. 65(4): 649. doi:10.1016/S0091-3057(99)00237-3. PMID:10764917.

Artaiz, I., Romero, G., Zazpe, A., Monge, A., Calderó, J.M., Roca, J., et al. 1995. Thepharmacology of VA21B7: an atypical 5-HT3 receptor antagonist withanxiolytic-like properties in animal models. Psychopharmacology, 117(2):137–148. doi:10.1007/BF02245179. PMID:7753959.

Barkow, K., Heun, R., Ustun, T.B., Berger, M., Bermejo, I., Gaebel, W., et al. 2004.Identification of somatic and anxiety symptoms which contribute to thedetection of depression in primary health care. Eur. Psychiatry, 19(5): 250–257. doi:10.1016/j.eurpsy.2004.04.015. PMID:15276656.

Barnes, J.M., Barnes, N.M., and Cooper, S.J. 1992. Behavioural pharmacology of5-HT receptor ligands. Neurosci. Biobehav. Rev. 16(1): 107–113. PMID:1553101.

Belzuing, C., Misslin, R., Vogel, E., Dodd, R.H., and Chapounthier, G. 1987. Anx-iogenic effects of methyl-b-carboline-3-carboxylate in a light dark choicesituation. Pharmacol. Biochem. Behav. 28(1): 29–33. doi:10.1016/0091-3057(87)90006-2. PMID:3116561.

Biala, G., and Kruk, M. 2008. Calcium channel antagonists suppress cross-tolerance to the anxiogenic effects of D-amphetamine and nicotine in themouse elevated plus maze test. Prog. Neuropsychopharmacol. Biol. Psychia-try, 32(1): 54–61. doi:10.1016/j.pnpbp.2007.07.006. PMID:17761379.

Blackburn, T.P., Baxter, G.S., Kennett, G.A., King, F.D., Piper, D.C., Sanger, G.J.,et al. 1993. BRL-46470A: a highly potent, selective and long acting 5-HT3receptor antagonist with anxiolytic-like properties. Psychopharmacology,110(3): 257–264. doi:10.1007/BF02251279. PMID:7831418.

Borsini, F., Lecci, A., Volterra, G., and Meli, A. 1989. A model to measure antici-patory anxiety in mice? Psychopharmacology, 98(2): 207–211. doi:10.1007/BF00444693. PMID:2502791.

Borsini, F., Podhorna, J., and Marazziti, D. 2002. Do animal models of anxiety

852 Can. J. Physiol. Pharmacol. Vol. 91, 2013

Published by NRC Research Press

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.

predict anxiolytic-like effects of antidepressants? Psychopharmacology, 163:121–141. doi:10.1007/s00213-002-1155-6. PMID:12202959.

Cappelli, A., Anzini, M., Vomero, S., Mennuni, L., Makovec, F., Hamon, M., et al.2002. The interactions of the 5-HT3 receptor with arylpiperazine, tropane,and quinuclidine ligands. Curr. Top. Med. Chem. 2(6): 599–624. doi:10.2174/1568026023393813. PMID:12052196.

Costall, B., and Naylor, R.J. 1992. Anxiolytic potential of 5-HT3 antagonists. Phar-macol. Toxicol. 70(3): 157–162. doi:10.1111/j.1600-0773.1992.tb00448.x. PMID:1349746.

Costall, B., Jones, B.J., Kelly, M.E., Naylor, R.J., and Tomkins, D.M. 1989. Explora-tion of mice in a black and white test box: validation as a model of anxiety.Pharmacol. Biochem. Behav. 32(3): 777–785. doi:10.1016/0091-3057(89)90033-6.PMID:2740429.

Costall, B., Domeney, A.M., Kelly, M.E., Tomkins, D.M., Naylor, R.J.,Wong, E.H.F.,et al. 1993. The effect of the 5-HT3 receptor antagonist, RS-42358-197, inanimal models of anxiety. Eur. J. Pharmacol. 234(1): 91–99. doi:10.1016/0014-2999(93)90710-Y. PMID:8097165.

De Angelis, L. 1992. The anxiogenic-like effects of pentylenetetrazole in micetreated chronically with carbamazepine or valproate. Methods Find. Exp.Clin. Pharmacol. 14(10): 767–771. PMID:1297893.

De Angelis, L., and Furlan, C. 2000. The anxiolytic-like properties of two selectiveMAOIs, moclobemide and selegiline, in a standard and an enhanced light/dark aversion test. Pharmacol. Biochem. Behav. 65: 649–653. doi:10.1016/S0091-3057(99)00237-3. PMID:10764917.

Dording, C.M., Mischoulon, D., Petersen, T.J., Kornblum, R., Gordon, J.,Nierenberg, A.A., et al. 2002. The pharmacologic management of SSRI-induced side effects: a survey of psychiatrists. Ann. Clin. Psychiatry, 14: 143–147. doi:10.3109/10401230209147450. PMID:12585563.

Dubinsky, B., Vaidya, A.H., Rosenthal, D.I., Hochman, C., Crooke, J.J., Deluca, S.,et al. 2002. 5-ethoxymethyl-7-fluoro-3-oxo-1,2,3,5-tetrahydrobenzo(4,5)imidazo(1,2a)pyridine-4-N-(2-fluorophenyl) carboxamide (RWJ-51204), a new nonbenzo-diazepine anxiolytic. J. Pharmacol. Exp. Ther. 303(2): 777–790. doi:10.1124/jpet.102.036954. PMID:12388665.

Fernandes, C., and File, S.E. 1996. The influence of open arm ledges and mazeexperience in the elevated plus-maze. Pharmacol. Biochem. Behav. 54(1):31–40. doi:10.1016/0091-3057(95)02171-X. PMID:8728536.

Greenshaw, A.J. 1992. Behavioral pharmacology of 5-HT3 receptor antagonists: Acritical uptake on therapeutic potential. Trends Pharmacol. Sci. 14(7): 265–270. PMID:8105596.

Griebel, G. 1995. 5-Hydroxytryptamine-interacting drugs in animal models ofanxiety disorders: more than 30 years of research. Pharmacol. Ther. 65(3):319–395. doi:10.1016/0163-7258(95)98597-J. PMID:7644567.

Hewlett, W.A., Zhang, Z.-J., de Paulis, T., Schmidt, D.E., Trivedi, B.L., andEbert, M.H. 2001. Anxiolytic-like effects of DAIZAC, a selective high-affinity5-HT (3) receptor antagonist, in the mouse elevated plus-maze. Pharmacol.Biochem. Behav. 69: 571–578 doi:10.1016/S0091-3057(01)00566-4. PMID:11509218.

Hidalgo, R.B., and Davidson, J.R.T. 2000. Selective serotonin reuptake inhibitorsin post-traumatic stress disorder. J. Psychopharmacol. 14: 70. doi:10.1177/026988110001400110. PMID:10757257.

Hogg, S. 1996. A review of the validity and variability of the elevated plus-mazeas an animal model of anxiety. Pharmacol. Biochem. Behav. 54(1): 21–30.doi:10.1016/0091-3057(95)02126-4. PMID:8728535.

Israili, Z.H. 2001. Clinical pharmacology of serotonin receptor type 3 (5-HT3)antagonists. Curr. Med. Chem. CNS Agents, 1: 171–199. doi:10.2174/1568015013358572.

Jindal, A., Mahesh, R., and Kumar, B. 2013. Anxiolytic-like effect of linezolid inexperimental mouse models of anxiety. Prog. Neuro-Psychopharmacol. Biol.Psychiatry, 40: 47–53. doi:10.1016/j.pnpbp.2012.09.006. PMID:23021974.

Jones, B. 1990. 5-HT3 receptor antagonists in anxiety and schizophrenia. DrugNews Persp. 3: 1061.

Kelley, S.P., Bratt, A.M., and Hodge, C.W. 2003a. Targeted gene deletion of the5-HT3A receptor subunit produces an anxiolytic phenotype in mice. Eur. J.Pharmacol. 461(1): 19–25. doi:10.1016/S0014-2999(02)02960-6. PMID:12568911.

Kessler, R.C., Berglund, P., Demler, O., Jin, R., Merikangas, K.R., andWalters, E.E.2005. Lifetime prevalence and age-of-onset distributions of DSM-IV disordersin the national comorbidity, survey replication. Arch. Gen. Psychiatry, 62:593–602. doi:10.1001/archpsyc.62.6.593. PMID:15939837.

Kilfoil, T., Michel, A., Montgomery, D., and Whiting, R.L. 1989. Effects of anxio-lytic and anxiogenic drugs on exploratory activity in a simple model ofanxiety in mice. Neuropharmacology, 28(9): 901–905. doi:10.1016/0028-3908(89)90188-3. PMID:2572995.

Kilpatrick, G.J., Butler, A., Ireland, S.J., Michel, A.D., and Tyers, M.B. 1989. Affin-ities of 5-HT uptake inhibitors for 5-HT receptors in both binding and func-tional studies. Br. J. Pharmacol. 98(Suppl.): 859P.

Klodzinska, A., Tatarczynska, E., Chojnacka-Wójcik, E., Nowak, G.,Cosford, N.D., and Pilc, A. 2004. Anxiolytic-like effects of MTEP, a potent andselective mGlu5 receptor agonist does not involve GABA (A) signaling.Neuropharmacology, 47(3): 342–350. doi:10.1016/j.neuropharm.2004.04.013.PMID:15275823.

Lader, M., and Morton, S. 1991. Benzodiazepine problems. Br. J. Addict. 86(7):823–828. doi:10.1111/j.1360-0443.1991.tb01831.x. PMID:1680514.

Laguerre, M., Dubost, J.P., Kummer, E., and Carpy, A. 1994. Molecular modeling

of 5-HT3 receptor antagonists: geometrical, electronic and lipophilic featuresof the pharmacophore and 3D-QSAR study. Drug Des. Discov. 11(3): 205–222.PMID:7548636.

Lal, H., and Emmett-Oglesby, M.W. 1983. Behavioural analogues of anxiety. Neu-ropharmacology, 22(12B): 1423–1441. doi:10.1016/0028-3908(83)90111-9. PMID:6142426.

Lima, L.M., and Barreiro, E.J. 2005. Bioisosterism: a useful strategy for molecularmodification and drug design. Curr. Med. Chem. 12(1): 23–49. doi:10.2174/0929867053363540. PMID:15638729.

Lister, R.G. 1987. The use of a plus-maze to measure anxiety in the mouse.Psychopharmacology, 92(2): 180–185. doi:10.1007/BF00177912. PMID:3110839.

Mahesh, R., Perumal, R.V., and Pandi, P.V. 2004. Microwave assisted synthesis of2-(4-substituted piperazin-1-yl)-1,8-naphthyridine-3-carbonitrile as a newclass of serotonin 5-HT3 receptor antagonists, Bioorg. Med. Chem. Lett.14(20): 5179–5181. doi:10.1016/j.bmcl.2004.07.060. PMID:15380223.

Mai, A., Artico, M., Esposito, M., Ragno, R., Sbardella, G., and Massa, S. 2003.Synthesis and biological evaluation of enantiomerically pure pyrro-lyloxazolidinones as a new class of potent and selective monoamine oxidasetype A inhibitors. Farmaco, 58: 231–241. doi:10.1016/S0014-827X(03)00016-8.PMID:12620419.

Majewicz, T.G., and Caluwe, P. 1974. A facile synthesis of 2-ami-nonicotinaldehyde. J. Org. Chem. 39(5): 720–721. doi:10.1021/jo00919a033.

Mechan, A.O., Moran, P.M., Elliott, M., Young, A.J., Joseph, M.H., and Green, R.2002. A comparison between dark agouti and Sprague–Dawley rats in theirbehaviour on the elevated plus-maze, open field apparatus and activity me-ters and their response to diazepam. Psychopharmacology (Berl.), 159(2):188–195. doi:10.1007/s002130100902. PMID:11862348.

Mi, X.J., Chen, S.W., Wang, W.J., Wang, R., Zhang, Y.J., Li, W.J., and Li, Y.L. 2005.Anxiolytic-like effect of paeonol in mice. Pharmacol. Biochem. Behav. 81(3):683–687. doi:10.1016/j.pbb.2005.04.016. PMID:15970315.

Millan, M.J. 2003. The neurobiology and control of anxious states. Prog. Neuro-biol. 70(2): 83–244. doi:10.1016/S0301-0082(03)00087-X. PMID:12927745.

Olivier, B., Wijngaarden, I.V., and Soudijn, W. 2000. 5-HT receptor antagonistsand anxiety; a preclinical and clinical review. Eur. Neuropsychopharm. 10(2):77–95. doi:10.1016/S0924-977X(99)00065-6. PMID:10706989.

Pandey, D.K., Ramamoorthy, R., Mahesh, R., and Radha, R. 2008. Depressant-likeeffects of parthenolide in rodent behavioural antidepressant test battery.J. Pharm. Pharmacol. 60(12): 1643–1650. doi:10.1211/jpp.60.12.0010. PMID:19000369.

Pellow, S., Chopin, P., File, S.E., and Briley, M. 1985. Validation of open:closedarm entries in an elevated plus maze as a measure of anxiety in the rat. J.Neurosci. Methods, 14(3): 149–167. doi:10.1016/0165-0270(85)90031-7. PMID:2864480.

Priest, R.G., Baldwin, D.S., Bullock, T., Kibel, D., Smeyatsky, N., and Steinert, J.1992. Recent advances in antidepressant drugs. S. Afr. Med. J. 6(Suppl.): 1–4.PMID:1609337.

Rajkumar, R., and Mahesh, R. 2010. The auspicious role of the 5-HT3 receptor indepression: a probable neuronal target? J. Psychopharmacol. 24(4): 455–469.doi:10.1177/0269881109348161. PMID:20123937.

Revelli, A., Tesarik, J., and Massobrio, M. 1998. Nongenomic effects of neuros-teroids. Gynecol. Endocrinol. 12(1): 61–67. doi:10.3109/09513599809024971.PMID:9526711.

Rosen, T., Nagel, A.A., Rizzi, J.P., Ives, J.L., Daffeh, J.B., Ganong, A.H., et al. 1990.Thiazole as a carbonyl bioisostere. A novel class of highly potent and selective5-HT3 receptor antagonists. J. Med. Chem. 33(10): 2715–2720. doi:10.1021/jm00172a006. PMID:2213824.

Schafer,W.R. 1999. How does antidepressant work? Prospect for genetic analysisof drug mechanism. Cell, 98(5): 551–554. PMID:10490094.

Sienkiewicz-Jarosz, E., Szyndler, J., Członkowska, A.I., Atkowski, M.S.,Maciejak, P.,Wisłowska, A., et al. 2003. Rat behavior in twomodels of anxietyand brain (3H) muscimol binding: pharmacological, correlation, and multi-factor analysis. Behav. Brain Res. 145(1–2): 17–22. doi:10.1016/S0166-4328(03)00096-2. PMID:14529801.

Simpson, K.H., Murphy, P., Colthup, P.V., andWhelan, P. 1992. Concentration ofondansetron in cerebrospinal fluid following oral dosing in volunteers. Psy-chopharmacology (Berl.), 109(4): 497–498. doi:10.1007/BF02247730. PMID:1365869.

Sugita, S., Shen, K.Z., and North, R.A. 1992. 5-hydroxytryptamine is a fast excit-atory transmitter at 5-HT3 receptors in rat amygdala. Neuron, 8: 199–203.doi:10.1016/0896-6273(92)90121-S. PMID:1346089.

Ter Laak, A.M., Tsai, R.S., Donne-Op den Kelder, G.M., Carrupt, P.A., Testa, B., andTimmerman, H. 1994. Lipophilicity and hydrogen-bonding capacity of H1-antihistaminic agents in relation to their central sedative side-effects. Eur. J.Pharm. Sci. 2: 373. doi:10.1016/0928-0987(94)00065-4.

Walstab, J., Gudrun, R., and Beate, N. 2010. 5-HT3 receptors: Role in disease andtarget of drugs. Pharmacol. Ther. 128(1): 146–169. doi:10.1016/j.pharmthera.2010.07.001. PMID:20621123.

Whiting, P. 2006. GABA-A receptors: a viable target for novel anxiolytics? Curr.Opin. Pharmacol. 6(1): 24–29. doi:10.1016/j.coph.2005.08.005. PMID:16359919.

Wolf, H. 2000. Preclinical and clinical pharmacology of the 5-HT3 receptor antag-onists. Scand. J. Rheumatol. Suppl. 113: 37–45. doi:10.1080/030097400446625.PMID:11028830.

Mahesh et al. 853

Published by NRC Research Press

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.

Xiao, Y., Chen, X., Karra, S.R., Huck, B.R., Sutton, A.E., and Goutopoulos, A. 2010.Naphthyridininones as aurora kinase inhibitors. Patent WO 2010002779 A2.Available from http://www.google.com/patents/WO2010002779A2?cl=en.

Young, R., and Johnson, D.N. 1991. A fully automated light/dark apparatus usefulfor comparing anxiolytic agents. Pharmacol. Biochem. Behav. 40(4): 739–743.doi:.10.1016/0091-3057(91)90078-G. PMID:1687762.

Zhang, Z.J., Schmidt, D.E., De Paulis, T., Trivedi, B.L., Onaivi, E.S., Ebert, M.H.,

et al. 2001. Anxiolytic-like effects of DAIZAC, a selective high-affinity5-HT3 receptor antagonist, in the mouse elevated plus-maze. Pharmacol.Biochem. Behav. 69(3–4): 571–578. doi:10.1016/S0091-3057(01)00566-4. PMID:11509218.

Zhang, Y., Wang, W., and Zhang, J. 2004. Effects of novel anxiolytic 4-butyl-alpha-agarofuran on levels of monoamine neurotransmitters in rats.Eur. J. Pharmacol. 504(1–2): 39–44. doi:10.1016/j.ejphar.2004.09.051. PMID:15507219.

854 Can. J. Physiol. Pharmacol. Vol. 91, 2013

Published by NRC Research Press

Can

. J. P

hysi

ol. P

harm

acol

. Dow

nloa

ded

from

ww

w.n

rcre

sear

chpr

ess.

com

by

Lau

rent

ian

Uni

vers

ity o

n 11

/01/

13Fo

r pe

rson

al u

se o

nly.