Anticonvulsant profile of the alkaloids (+)-erythravine and (+)-11-α-hydroxy-erythravine isolated...

6
Anticonvulsant prole of the alkaloids (+)-erythravine and (+)-11-α-hydroxy-erythravine isolated from the owers of Erythrina mulungu Mart ex Benth (LeguminosaePapilionaceae) Silmara Aparecida Faggion a , Alexandra Olimpio Siqueira Cunha b , Helene Aparecida Fachim b , Amanda Salomão Gavin a , Wagner Ferreira dos Santos b , Ana Maria Soares Pereira a , Renê Oliveira Beleboni a, a Department of Biotechnology, University of Ribeirão Preto (UNAERP), São Paulo, Brazil b Department of Biology, FFCLRP, University of São Paulo (USP), São Paulo, Brazil abstract article info Article history: Received 9 September 2010 Revised 17 November 2010 Accepted 22 December 2010 Available online 1 February 2011 Keywords: Alkaloids Anticonvulsants Erythrina mulungu (+)-erythravine (+)-11-α-hydroxyerythravine Natural products Neural mechanisms underlying the onset and maintenance of epileptic seizures involve alterations in inhibitory and/or excitatory neurotransmitter pathways. Thus, the prospecting of novel molecules from natural products that target both inhibition and excitation systems has deserved interest in the rational design of new anticonvulsants. We isolated the alkaloids (+)-erythravine and (+)-11-α-hydroxy- erythravine from the owers of Erythrina mulungu and evaluated the action of these compounds against chemically induced seizures in rats. Our results showed that the administration of different doses of (+)- erythravine inhibited seizures evoked by bicuculline, pentylenetetrazole, and kainic acid at maximum of 80, 100, and 100%, respectively, whereas different doses of (+)-11-α-hydroxy-erythravine inhibited seizures at a maximum of 100% when induced by bicuculline, NMDA, and kainic acid, and, to a lesser extent, PTZ (60%). The analysis of mean latency to seizure onset of nonprotected animals, for specic doses of alkaloids, showed that (+)-erythravine increased latencies to seizures induced by bicuculline. Although (+)-erythravine exhibited very weak anticonvulsant action against seizures induced by NMDA, this alkaloid increased the latency in this assay. The increase in latency to onset of seizures promoted by (+)-11-α-hydroxy-erythravine reached a maximum of threefold in the bicuculline test. All animals were protected against death when treated with different doses of (+)-11-α-hydroxy-erythravine in the tests using the four chemical convulsants. Identical results were obtained when using (+)-erythravine in the tests of bicuculline, NMDA, and PTZ, and, to a lesser extent, kainic acid. Therefore, these data validate the anticonvulsant properties of the tested alkaloids, which is of relevance in consideration of the ethnopharmacological/biotechnological potential of E. mulungu. © 2010 Elsevier Inc. All rights reserved. 1. Introduction Epilepsy is a set of chronic disorders that modify brain function and are associated with the occurrence of sudden and unexpected seizures of convulsive or nonconvulsive nature. Most epileptic syndromes have particular neurophysiological and clinical character- istics, the seizures being the result of abnormal, hyperactive, or hypersynchronous neuronal discharges [1]. Moreover, neuronal mechanisms underlying the onset and maintenance of the epilepsies involve marked alterations in the balance between excitatory and inhibitory neurotransmission pathways in the central nervous system (CNS) [1,2]. In most cases, treatment of the epilepsies consists of maintaining the patient seizure free with anticonvulsant drugs. These drugs suppress synaptic hyperactivation by acting on ion channels, metabolic enzymes, or neurotransmitter transporters or receptors, the latter involving chiey GABA and/or glutamate [3]. Despite the considerable number of antiepileptic drugs, there is a need for novel medicines, as not all patients with epilepsy respond satisfactorily to the usual treatments, and also because these drugs have side effects such as cognitive impairment, sedation, teratogenesis, and hirsutism with chronic use [47]. In light of these facts, the search for novel anticonvulsant compounds of different chemical nature and the modication of the structure of traditional drugs may lead to the generation of more effective and better tolerated drugs, as well as to a better understanding of the mechanisms through which the brain becomes epileptic [6]. Erythrina mulungu (Sin. Corallodendron mulungu (Martius) Kuntze, Erythrina ammea Herzog, Erythrina mulungu Mart. ex Benth.) is a deciduous tree found mainly in Brazilian cerrados and other tropical regions [8,9]. The owers, fruits, seeds, and bark are frequently used in folk medicine because of their effects on the CNS; for example, Epilepsy & Behavior 20 (2011) 441446 Corresponding author. Unidade de Biotecnologia, Av. Costábile Romano, 2.201, Zip Code 14096-300, Ribeirão Preto, São Paulo, Brazil. E-mail addresses: [email protected], [email protected] (R.O. Beleboni). 1525-5050/$ see front matter © 2010 Elsevier Inc. All rights reserved. doi:10.1016/j.yebeh.2010.12.037 Contents lists available at ScienceDirect Epilepsy & Behavior journal homepage: www.elsevier.com/locate/yebeh

Transcript of Anticonvulsant profile of the alkaloids (+)-erythravine and (+)-11-α-hydroxy-erythravine isolated...

Epilepsy & Behavior 20 (2011) 441–446

Contents lists available at ScienceDirect

Epilepsy & Behavior

j ourna l homepage: www.e lsev ie r.com/ locate /yebeh

Anticonvulsant profile of the alkaloids (+)-erythravine and(+)-11-α-hydroxy-erythravine isolated from the flowers ofErythrina mulungu Mart ex Benth (Leguminosae–Papilionaceae)

Silmara Aparecida Faggion a, Alexandra Olimpio Siqueira Cunha b, Helene Aparecida Fachim b,Amanda Salomão Gavin a, Wagner Ferreira dos Santos b,Ana Maria Soares Pereira a, Renê Oliveira Beleboni a,⁎a Department of Biotechnology, University of Ribeirão Preto (UNAERP), São Paulo, Brazilb Department of Biology, FFCLRP, University of São Paulo (USP), São Paulo, Brazil

⁎ Corresponding author. Unidade de Biotecnologia, AvCode 14096-300, Ribeirão Preto, São Paulo, Brazil.

E-mail addresses: [email protected], rbeleboni@u

1525-5050/$ – see front matter © 2010 Elsevier Inc. Aldoi:10.1016/j.yebeh.2010.12.037

a b s t r a c t

a r t i c l e i n f o

Article history:Received 9 September 2010Revised 17 November 2010Accepted 22 December 2010Available online 1 February 2011

Keywords:AlkaloidsAnticonvulsantsErythrina mulungu(+)-erythravine(+)-11-α-hydroxyerythravineNatural products

Neural mechanisms underlying the onset and maintenance of epileptic seizures involve alterations ininhibitory and/or excitatory neurotransmitter pathways. Thus, the prospecting of novel molecules fromnatural products that target both inhibition and excitation systems has deserved interest in the rationaldesign of new anticonvulsants. We isolated the alkaloids (+)-erythravine and (+)-11-α-hydroxy-erythravine from the flowers of Erythrina mulungu and evaluated the action of these compounds againstchemically induced seizures in rats. Our results showed that the administration of different doses of (+)-erythravine inhibited seizures evoked by bicuculline, pentylenetetrazole, and kainic acid at maximum of 80,100, and 100%, respectively, whereas different doses of (+)-11-α-hydroxy-erythravine inhibited seizures at amaximum of 100%when induced by bicuculline, NMDA, and kainic acid, and, to a lesser extent, PTZ (60%). Theanalysis of mean latency to seizure onset of nonprotected animals, for specific doses of alkaloids, showed that(+)-erythravine increased latencies to seizures induced by bicuculline. Although (+)-erythravine exhibitedvery weak anticonvulsant action against seizures induced by NMDA, this alkaloid increased the latency in thisassay. The increase in latency to onset of seizures promoted by (+)-11-α-hydroxy-erythravine reached amaximum of threefold in the bicuculline test. All animals were protected against death when treated withdifferent doses of (+)-11-α-hydroxy-erythravine in the tests using the four chemical convulsants. Identicalresults were obtained when using (+)-erythravine in the tests of bicuculline, NMDA, and PTZ, and, to a lesserextent, kainic acid. Therefore, these data validate the anticonvulsant properties of the tested alkaloids, whichis of relevance in consideration of the ethnopharmacological/biotechnological potential of E. mulungu.

. Costábile Romano, 2.201, Zip

naerp.br (R.O. Beleboni).

l rights reserved.

© 2010 Elsevier Inc. All rights reserved.

1. Introduction

Epilepsy is a set of chronic disorders that modify brain functionand are associated with the occurrence of sudden and unexpectedseizures of convulsive or nonconvulsive nature. Most epilepticsyndromes have particular neurophysiological and clinical character-istics, the seizures being the result of abnormal, hyperactive, orhypersynchronous neuronal discharges [1]. Moreover, neuronalmechanisms underlying the onset and maintenance of the epilepsiesinvolve marked alterations in the balance between excitatory andinhibitory neurotransmission pathways in the central nervous system(CNS) [1,2].

In most cases, treatment of the epilepsies consists of maintainingthe patient seizure free with anticonvulsant drugs. These drugs

suppress synaptic hyperactivation by acting on ion channels,metabolic enzymes, or neurotransmitter transporters or receptors,the latter involving chiefly GABA and/or glutamate [3]. Despite theconsiderable number of antiepileptic drugs, there is a need for novelmedicines, as not all patients with epilepsy respond satisfactorily tothe usual treatments, and also because these drugs have side effectssuch as cognitive impairment, sedation, teratogenesis, and hirsutismwith chronic use [4–7]. In light of these facts, the search for novelanticonvulsant compounds of different chemical nature and themodification of the structure of traditional drugs may lead to thegeneration of more effective and better tolerated drugs, as well as to abetter understanding of the mechanisms through which the brainbecomes epileptic [6].

Erythrina mulungu (Sin. Corallodendron mulungu (Martius) Kuntze,Erythrina flammea Herzog, Erythrina mulungu Mart. ex Benth.) is adeciduous tree found mainly in Brazilian cerrados and other tropicalregions [8,9]. The flowers, fruits, seeds, and bark are frequently used infolk medicine because of their effects on the CNS; for example,

442 S.A. Faggion et al. / Epilepsy & Behavior 20 (2011) 441–446

anticonvulsant, antidepressant, analgesic, sedative, and hypnoticeffects are some of those actions validated by different experimentalmodels [8,10–12]. Indeed, experiments have pointed to the effectiveanxiolytic activity of E.mulunguhydroalcoholic extract [13–15] and alsoto its anticonvulsant effects against seizures induced by differentchemoconvulsants [16]. Chromatographic purification of the E.mulunguhydroalcoholic extract led to the identification of some alkaloidswith effective anxiolytic activity, among which are (+)-erythravineand (+)-11α-hydroxy-erythravine [17,18].

As anxiolytics may also function as antiepileptic drugs and based onthe popular use of E. mulungu as well as on previous reports pointing tothe anticonvulsant properties of the hydroalchoolic extract of this plant,the aim of the present work was to investigate the anticonvulsantproperties of (+)-erythravine and (+)-11α-hydroxy-erythravinealkaloids, analyzing their effectiveness in the blockade of seizuresinduced in rats by two GABAergic antagonists, bicuculline andpentylenetetrazole (PTZ), and two glutamatergic agonists, NMDA andkainic acid.

2. Material and Methods

2.1. Chemicals

Most of the chemical agents, reagents, and solvents used in thisstudy were purchased from Sigma–Aldrich (USA) or Merck (USA),unless otherwise specified. Diazepam, ketamine, and xylazine,acquired from Germed (Brazil), Agener União (Brazil), and HertapeCallier (Brazil), respectively, were injected in a final volume of0.3 mL (intraperitoneally). All reagents were of analytical grade.(+)-Erythravine and (+)-11-α-hydroxy-erythravine were dilutedin saline solution before bioassay and administered in a total volumeof 1 μL (intracerebroventricularly).

2.2. Animals

Male Wistar rats (200–250 g) were acquired from animal housingat the University of São Paulo in Ribeirão Preto-SP, Brazil. Animalswere maintained under controlled conditions of light (12-hour dark–light cycle, lights on at 7:00 AM), temperature (25 °C), and humidity(55%). This work was approved by the Ethics Committee forExperimental Animals at UNAERP (006/08). Experimental procedureswere performed according to the rules in the Handbook of AnimalExperimentation and Care from the Brazilian Society of Neurosciencesand Behavior. Furthermore, all efforts were made to avoid unneces-sary stress and pain for the animals.

2.3. Isolation and identification of (+)-erythravine and(+)-11α-hydroxy-erythravine from Erythrina mulungu Mart. Ex Benth.

Flowers from E. mulungu (5.0 kg) were collected in Rifaina, SãoPaulo, Brazil (latitude 20°07'41.7'', longitude 47°281'54.5'', 1037-GPS).Flowered branches were herborized according to the guide of theMedicinal Plants Herbarium in UNAERP and labeled with a vouchernumber (HPMU-1330).

Purification procedures followed those described by Flausino andcolleagues [17]. Briefly, freshflowersweremacerated in a 50 L solutionof ethanol:water (7:3, v/v). After 15 days, the extract was filtered andthe solvent submitted to rotary evaporation. The aqueous extract waslyophilized, resulting in 86.905 g of dry extract,whichwas dissolved ina solution of acetic acid:water (10%, v/v) and submitted to liquid–liquid extractionusing absolute chloroform(CHCl3). The organic phasewas separated and the solvent was evaporated, resulting in fraction 1(F1). Next, ammonium hydroxide was added to the aqueous phase toincrease the pH to the range 9–10 and this solution was thenresubmitted to extraction with CHCl3. The organic phase wasseparated and the organic solvent evaporated, resulting in fraction

2 (F2). The remaining aqueous fraction was named F3. To identify theactive compounds and to check the grade of purity, these threefractions were analyzed with thin-layer chromatography in silicausing CHCl3:MeOH (7:1, v/v) as the mobile phase and revelation withthe Dragendorff reagent.

Fraction 1 (5.6 mg) was applied to a chromatography column(50×150 mm) containing silica (0.035–0.070 mm) as stationaryphase and CHCl3:MeOH (10:0 and 8:2, v/v) as mobile phase. At thispoint, 169 fractions were eluted and grouped as follows: F1.1 (1–27,0.738 g); F1.2 (26–39, 0.982 g); F1.3 (40–49, 0.401 g); F1.4 (50–73,0.467 g); F1.5 (74–135, 230 g); F1.6 (136–157, 0.188 g); F1.7 (158–168,0.029 g); and F1.8 (169, 0.515 g). Fractions F1.2 and F1.8 (1.497 g)provided a novel pool fractionated in a chromatography column(20×425 mm), using silica (0.035–0.070 mm) and CHCl3/MeOH(9:1, 8:2, 7:3 and 6:4; v/v) as mobile phase, resulting in four fractions:F.1.2/8.1, F.1.2/8.2, F.1.2/8.3 and F.1.2/8.4. Next, F1.2/8.2was applied to asilica–fluoroscein preparative plate (Merck, USA) for alkaloid isolationusing as solvents toluene, acetone, ethanol, and ammonium hydroxide(45:45:7:3, v/v), resulting in the elution of (+)-erythravine (2.4 mg).Fractionation of F2 was conducted using a preparative plate(200×200 mm) and samples were eluted in CHCl3/MeOH (7:1; v/v),resulting in the isolation of (+)-11-α-hydroxy-erythravine (15.2 mg).

Isolated compounds were checked for grade of purity usingnuclear magnetic resonance (Varian Inc., USA) with scans at500 MHz and tetramethylsilate as internal marker and deuteratedchloroform (CD-CHCl3) as solvent.

2.4. Surgery

Stainless-steel guide cannulas (0.7×10 mm) were implanted inanimals (n=6–8, each group) on anesthesia with ketamine andxylazine (60 and 8 mg/kg, ip, respectively). Animals were placed in astereotaxic device (Stoelting-Standard, USA) and received a localinjection of 2% lidocaine to expose the calvaria and place the guidecannula in the right lateral ventricle. The coordinates used were0.9 mmposterior to bregma, 1.6 mm lateral frommidline, and 3.4 mmventral from the surface of the skull according to Paxinos andWatson[19]. The cannula was fixed to the skull using dental acrylate and wassealed with a stainless-steel wire to avoid obstruction. After thesurgery, animals received an injection of Flunixin meglumine anti-inflammatory (1.1 mg/kp, im). The animals were then allowed to restfor 5–7 days to recover from surgery.

2.5. Anticonvulsant screening

On theday of the experiments,with the aid of a digital infusion pump(Insight, Brazil) connected toaHamilton syringe (10 μL) andan injectioncannula, each animal was microinjected intracerebroventricularly (icv)with saline or each of the alkaloids in concentrations ranging from0.25 to 3 μg/μL. Animals used as a positive control received diazepam(2mg/kg, ip). After 10 minutes, chemoconvulsants were injected indoses adjusted to induce seizures in 97% of animals (CD97), as follows:bicuculline (0.9 μg/μL, icv), NMDA (17 μg/μL, icv), kainic acid (0.8 μg/μL,icv), and PTZ (80 mg/kg, ip). Intracerebroventricular injections wereadministered in a total volume of 1 μL over 1 minute, whereasintraperitoneal injections were delivered in a volume of 0.3 mL.

Animals were observed for 30 minutes for the occurrence ofseizures, and latency to onset or death was recorded. Seizures wereclassified using the limbic seizure scale [20] as follows: 0 = noseizure; 1 = orofacial movements; 2 = head myoclonus; 3 =hindlimb myoclonus; 4 = elevation; 5 = elevation and limbic fall;6 = head and ear myoclonus; 7 = rotation, jumps, and vocalization;8 = class 7 behaviors and hypertonus.

One day after the assays, animals were euthanized with anoverdose of thiopental (100 mg/kg, ip; Cristalia, Brazil) and theninjected with toluidine blue (1 μL, icv). Brains were removed, frozen,

443S.A. Faggion et al. / Epilepsy & Behavior 20 (2011) 441–446

andmanually cut to check placement of the cannula. Animals inwhichthere was inaccurate placement (≅5%) were not considered in theanalysis.

2.6. Data analysis

Number of animals protected against generalized chemicallyinduced tonic–clonic seizures was plotted in relation to eachconcentration of the alkaloids used. These frequencies were thencompared using the χ2 test. Latency to seizure onset in animals notfully protected by the alkaloids was analyzed using one-way analysisof variance (ANOVA), followed by the Tukey post test. A P valueb0.05was considered statistically significant. Data were analyzed and fittedusing the software Graph Prism (Version 4.0, GraphPad Software,USA).

3. Results

3.1. Isolation and identification of alkaloids

Alkaloid purification was effective in providing the appropriateamount of alkaloids to perform bioassays. In this regard, we obtained15.2 and2.4 mgof (+)-11-α-hydroxy-erythravineand (+)-erythravine,respectively. The degree of purity for both alkaloids was confirmed byNMR spectra (see Appendix—Supplementary Material).

Bicuculline

Saline 1 µg/µL 2 µg/µL 3 µg/µL

1 µg/µL 2 µg/µL 3 µg/µL

0

25

50

75

100

*

A

Non

seiz

ing

anim

als

(%)

B

PTZ

Saline

0

25

50

75

100

C** **

Non

seiz

ing

anim

als

(%)

Fig. 1. Percentages of animals protected against seizures induced with the chemoconvulsant(D) kainic acid (0.8 μg/μL, icv) after intracerebroventricular microinjection of different dose

3.2. Anticonvulsant screening

Data from anticonvulsant screening of (+)-erythravine forprotecting animals against seizures are summarized in Fig. 1.The data indicate that intracerebroventricular microinjection of(+)-erythravine into rats prior to the administration of bicucullineinhibited seizures in 40, 60, and 80% of animals at doses of 1, 2, and3 μg/μL, respectively (χ2[3]=10.40, P=0.015) (Fig. 1A). In the NMDAassay, treatment with (+)-erythravine at 3 μg/μL (icv) blockedseizures in only 20% of animals (χ2[3]=3.158, P=0.3679) (Fig. 1B).Systemic administration of PTZ (80 mg/kg, ip) induced generalizedseizures in all injected animals, which were blocked by pre-administration of (+)-erythravine in 60 and 100% of animals atdoses of 1 and 2 or 3 μg/μL, respectively (χ2[3]=14.73, P=0.0021)(Fig. 1C). In the kainic acid assay, pretreatment of animals with(+)-erythravine inhibited seizures in 40 and 100% of animals atdoses of 1 and 2 or 3 μg/μL, respectively (χ2[4]=20.19, P=0.0005);0.5 μg/μL was not protective (Fig. 1D). The anticonvulsant activity of(+)-erythravine exhibited a dose-dependent profile in all experiments.

Data from anticonvulsant screening of (+)-11-α-erythravinefor protecting animals against seizures are illustrated in Fig. 2.Our results show that this alkaloid, when centrally applied to ratsprior to the administration of bicuculline, inhibited seizures in allanimals at 1 and 2 μg/μL, and inhibited seizures in 60% of animals at0.5 μg/μL. At 0.25 μg/μL, (+)-11-α-erythravine exhibited no anticon-vulsant activity (χ2[4]=20.19, P=0.0005) (Fig. 2A). Tonic–clonicseizures induced by the glutamatergic agonist NMDAwere blocked by

NMDA

0

25

50

75

100

Non

seiz

ing

anim

als

(%)

1 µg/µL 2 µg/µL 3 µg/µLSaline

1 µg/µL0.5 µg/µL 2 µg/µL 3 µg/µLSaline

Kainic acid

0

25

50

75

100

D** **

Non

seiz

ing

anim

als

(%)

s (A) bicuculline (0.9 μg/μL, icv); (B) NMDA (17 μg/μL, icv); (C) PTZ (80 mg/kg, ip); ands of (+)-erythravine (0.12–3.0 μg/μL). *Pb0.05, **Pb0.01.

Bicuculline

Saline 0.25 µg/µL 0.5 µg/µL 1 µg/µL 2 µg/µL Saline 0.25 µg/µL 0.5 µg/µL 1 µg/µL 2 µg/µL

0.5 µg/µL 1 µg/µL 2 µg/µL1 µg/µL 2 µg/µL 3 µg/µL

0

25

50

75

100 *** ***

AN

onse

izin

g an

imal

s (%

)NMDA

0

25

50

75

100 *** ***C

Non

seiz

ing

anim

als

(%)

PTZ

Saline0

25

50

75

100D

Non

seiz

ing

anim

als

(%)

Kainic acid

Saline0

25

50

75

100 ***B

Non

seiz

ing

anim

als

(%)

Fig. 2. Percentages of animals protected against seizures induced with the chemoconvulsants (A) bicuculline (0.9 μg/μL, icv), (B) NMDA (17 μg/μL, icv), (C) PTZ (80 mg/kg, ip), and(D) kainic acid (0.8 μg/μL, icv) after intracerebroventricular microinjection of different doses of (+)-11α-hydroxy-erythravine (0.12–3.0 μg/μL). *Pb0.05, **Pb0.01, ***Pb0.001.

Table 1Mean latency to onset of seizures evoked by the administration of chemoconvulsants.

Latency to seizure onset (s)

Bicuculline PTZ NMDA Kainic acid

Saline 74.63±7.018 50.0±8.25 40.50±6.61 318±9.7Diazepam NS NS NS NSErythravine

0.5 μg/μL NA NA NA 18±3.7a

1 μg/μL 140.0±52.92 165.0±5.0b 31.4±14.5 320±722 μg/μL 143.0±3.00 NS 138±14.7b NS3 μg/μL 300c NS 218.8±16.6a NS

11-α-Hydroxy-erythravine0.25 μg/μL 94.0±10.42 NA 66.6±23.5 NA0.5 μg/μL 244.5±8.5b NA 300c 100±33.7b

1 μg/μL NS 289.5±185.5 NS 136.7±22b

2 μg/μL NS 360±272.9 NS NS3 μg/μL NA 269±31.0 NA NS

Note. Data represent means±SEM. NA, not applied; NS, no seizure.a Pb0.001.b Pb0.01.c Refers to one seizing animal; therefore, statistical analysis did not include this

datum.

444 S.A. Faggion et al. / Epilepsy & Behavior 20 (2011) 441–446

(+)-11-α-hydroxyerythravine in 80% of animals at 0.5 μg/μL, whereas100% of animals were protected from those seizures by theadministration of 1 and 2 μg/μL (+)-11-α-hydroxy-erythravine(χ2[4]=21.75, P=0.0002) (Fig. 2B). Seizures induced by systemicadministration of PTZ were blocked by the pre-administration of(+)-11-α-hydroxy-erythravine in only 40 and 60% of animals at 2and 3 μg/μL, respectively (χ2[3]=7.200, P=0.0658) (Fig. 2C). In thecase of kainic acid, 40 and 100% of animals were protected at 1 and2 μg/μL, respectively (χ2[3]=17.33, P=0.0006) (Fig. 2D). As observedfor (+)-erythravine, the anticonvulsant activity of (+)-11-α-hydroxy-erythravine exhibited a dose-dependent profile in all experiments.

In all experiments, microinjections of bicuculline (0.9 μg/μL, icv),NMDA(17 μg/μL, icv), PTZ (80 mg/kg, ip), and kainic acid (0.8 μg/μL, icv)induced tonic–clonic seizures in all animals pre-injected with saline.Conversely, diazepam protected 100% of animals against seizures.

Analysis of mean latency to seizure onset in unprotected animalsfor both alkaloids is summarized in Table 1.

With respect to unprotected animals treated with (+)-erythravine,we observed an increase in latency to onset of seizures inducedwith bicuculline, although this increase was not statistically significant(F[2,12]=3.486, P=0.07). Analysis of animals treatedwithPTZ showedthat at the only nonprotective dose (1 μg/μL), (+)-erythravineincreased the mean latency to onset compared with that of animalsinjected with saline (t=9.014, P=0.0008). Although (+)-erythravineshowedveryweakanticonvulsant activity against seizures inducedwithNMDA, in this assay, this alkaloid increased the latency to onset ofseizures at doses of 2 and 3 μg/μL (F[3,17]=38.4, Pb0.0001). Admin-istration of (+)-erythravineat0.5 μg/μLbeforekainic aciddecreased thelatency to seizure onset as comparedwith that for animals injectedwith(+)-erythravine 1 μg/μL as well as saline, pointing to a proconvulsanteffect at the lowest dose used (F[2,17]=38.4, Pb0.0001].

With respect to mean latency to seizure onset after treatmentwith (+)-11-α-hydroxy-erythravine, our findings showed that this

alkaloid, at a dose of 0.5 μg/μL, increased the latency to onset ofseizures evoked with bicuculline relative to the group injected withsaline as well as the group injected with the lowest dose of thealkaloid (0.25 μg/μL) (F[2,14]=55.46, Pb0.0001). The unprotectedanimals administered PTZ after (+)-11-α-hydroxy-erythravineexhibited seizures with heterogeneous latencies to seizure onset,and thus we did not observe statistical differences among means inthese groups of animals (F[3,13]=0.6887, P=0.5814).When injectedbefore NMDA, (+)-11-α-hydroxy-erythravine 0.25 μg/μL did not

445S.A. Faggion et al. / Epilepsy & Behavior 20 (2011) 441–446

increase the latency to seizure onset (t=0.95, P=0.37). However,administration of 0.5 μg/μL 11-α-hydroxy-erythravine induced asevenfold increase in the latency to seizure onset of the onlyseizing animal. Regarding kainic acid-induced seizures, we observeda significant decrease in the latency to seizure onset in animalstreated with (+)-11-α-hydroxy-erythravine 0.5 and 1 μg/μL (F[2,12]=23.76, Pb0.0001). These findings are similar to those obtained for(+)-erythravine 0.5 μg/μL.

All animals were protected against death when treated withdifferent doses of (+)-11-α-hydroxy-erythravine before bicuculline,NMDA, PTZ, and kainic acid. Identical results were obtained whenusing (+)-erythravine in the tests of bicuculline, NMDA, and PTZ.However, the protection against death afforded by (+)-erythravinewas 60, 80, and 100% at 0.5, 1, and 2 μg/μL (icv), respectively, in thekainic acid test. Mortality was 100% in animals injected only withsaline before NMDA, PTZ, and kainic acid and 80% in animals injectedonly with saline before bicuculline.

4. Discussion

According to the World Health Organization (WHO), from 65 to80% of the world's population, partially or totally, depends onnonconventional medicines for primary health care [21]. Plantextracts have been used in traditional folk medicine for centuries inthe treatment of CNS disorders. With formalization of a pharma-cological therapy approach, the pharmaceutical industry has tried toisolate and design neuroactive drugs from natural probes present inplants [22].

In Brazilian traditional medicine, E. mulungu extracts are commonlyused to treat anxiety and pain, as sedatives, and for blood pressurecontrol [8,10]. Scientific investigations have reported the neurobiolog-ical activity, including anticonvulsant, anxiolytic, and analgesic proper-ties, of the hydroalcoholic extract of parts of E. mulungu [12–16]. Theanalgesic activity appears to be independent of opioidmechanisms [12],and according to Vasconcelos and colleagues [16], the hydroalcoholicextract of the stem bark from specimens of E. mulungu inhibits seizuresinduced in mice by the administration of strychnine and PTZ.Fractionation of E. mulungu extract revealed the presence of twomajor alkaloids: (+)-11-α-hydroxy-erythravine and (+)-erythravine.These alkaloids promote anxiolytic effects when administered to micesubmitted to a light–dark choice apparatus and elevated plus maze[17,18].

Anxiety and epilepsy can be explained on the basis of similarneurobiological concepts, involving, in particular, alterations in theneurotransmitter balance in inhibitory and excitatory pathways. Infact, some anxiolytic drugs also act as anticonvulsants, such as thebenzodiazepines. On the basis of the results presented, both anxiolyticalkaloids exhibited significant anticonvulsant activity against chem-ically induced seizures in rats. However, with respect to blockinggeneralized tonic–clonic seizures induced by two GABAergic antago-nists, bicuculline and PTZ, and two glutamatergic agonists, NMDA andkainic acid, (+)-erythravine and (+)-11α-hydroxy-erythravine didnot differ enough to predict a preferential action of these alkaloids oninhibitory or excitatory pathways. It has been suggested that alkaloidsfrom Erythrina species act on the CNS by modifying GABA neurotrans-mission. It should be pointed out that agonists of GABA/benzodiaze-pine receptor typically promote motor impairment, particularly athigher doses [18,23]. As suggested by Flausino and colleagues [18],because the E. mulungu extract and (+)-erythravine/(+)-11-α-hydroxy-erythravine did not cause motor disruptions in mice [11], itis reasonable to assume that these alkaloids could be not active only bya prevalent action onGABAneurotransmission. The characterization ofthemodeof action of these erythrinian alkaloids is currently underwayin our laboratory.

Once again, as demonstrated by Vasconcelos and colleagues [11],systemic administration of the E.mulunguhydroalcholic extract in doses

up to 400 mg/kg does not impair motor coordination in female micesubmitted to the rotarod test. Moreover, behavioral analysis of theseanimals in the open field pointed to a depressant effect on rearingand other parameters of exploratory behavior, which, according tothe authors, could account for the sedative properties of the extract.We thus could consider the extract well tolerated. The importanceof these data lies in the continuous need for novel and better toleratedantiepileptic drugs as therapy for the epilepsies as well as othersyndromesof convulsive symptomatology [7]. As in theworkof Flausinoand colleagues [18], the alkaloids (+)-erythravine and (+)-11-α-hydroxy-erythravine were orally administered to animals with result-ing anxiolytic action; it is important to note that these alkaloids arewell absorbed in the gastrointestinal tract and may be able to cross theblood–brain barrier, which is of relevance in the development of newdrugs acting on the CNS, including antiepileptic drugs.

This work validates oncemore the ethnopharmacological potentialof E. mulungu in the treatment of neurological disorders and alsocomplies with biotechnological implications, as E. mulungu extract hasbeen used in phytotherapeutic compositions/medicines in Brazil andthe United States. In this context, continuing investigation of themode of action of these alkaloids could lead to novel probes that couldbe used in rational drug design, as well as tools in the investigation ofneural substrates that could be involved in the onset andmaintenanceof epileptic seizures.

Acknowledgments

The authors are grateful to FAPESP for Grants 08/52627-9 and 09/11357-1 and to Mrs. Sarazetti Pereira and Karen Lanza Iamamura(UNAERP) for technical assistance.

Appendix A. Supplementary data

Supplementary material associated with this article can be found,in the online version, at doi:10.1016/j.yebeh.2010.12.037.

References

[1] Engel Jr J. Surgical treatment of the epilepsies. 2nd ed. New York: Raven Press;1993.

[2] Delgado-Escueta AV, Wilson WA, Olsen RW, Porter RJ. New waves of research inthe epilepsies: crossing into the third millennium. In: Delgado-Escueta AV, WilsonWA, Olsen RW, Porter RJ, editors. Jasper's basic mechanisms of the epilepsies.Advances in Neurology. vol. 79, 3rd ed. Philadelphia: Lippincott Williams &Wilkins; 1999.

[3] Bialer M, White HS. Key factors in the discovery and development of newantiepileptic drugs. Nat Rev Drug Discovery 2010;9:68–82.

[4] Krogsgaard-Larsen P, Frolund BF, Falch E. Inhibitors of gamma-aminobutyric acidtransport as experimental tools and therapeutic agents. Meth Enzymol 1998;296:165–75.

[5] Andersen KE, Lau J, Lundt BF, et al. Synthesis of novel GABA uptake inhibitors:Part 6: Preparation and evaluation of N-omega asymmetrically substitutednipecotic acid derivatives. Bioorg Med Chem 2001;9:2773–85.

[6] Perucca E. An introduction to antiepileptic drugs. Epilepsia 2005;46:31–7.[7] Mortari MR, Cunha AO, Ferreira LB, dos Santos WF. Neurotoxins from

invertebrates as anticonvulsants: from basic research to therapeutic application.Pharmacol Ther 2007;114:171–83.

[8] Lourenzi H. Árvores Brasileiras: Manual de Identificação e Cultivo de Plantas. SãoPaulo: Plantarum; 1992.

[9] Neill DA. Experimental studies on species relationships in Erytrina (Leguminosae:Papilionoidae). Ann Mo Bot Gard 1988;75:886–969.

[10] Rodrigues VEG, Carvalho DA, editors. Plantas Medicinais do Cerrado. Lavras:Editora da Universidade Federal de Lavras; 2001.

[11] Vasconcelos SM, Macedo DS, De Melo CT, et al. Central activity of hydroalcoholicextracts from Erythrina velutina and Erythrina mulungu in mice. J Pharm Pharmacol2004;56:389–93.

[12] Vasconcelos SM, Rebouças OG, De Carvalho M, et al. Antinociceptive activities ofthe hydroalcoholic extracts from Erythrina velutina and Erythrina mulungu in mice.Biol Pharm Bull 2003;6:946–9.

[13] Onusic GM, Nogueira RL, Pereira AM, Viana MB. Effect of acute treatment with awater–alcohol extract of Erythrina mulungu on anxiety-related responses in rats.Braz J Med Biol Res 2002;35:473–8.

[14] Onusic GM, Nogueira RL, Pereira AM, Flausino Jr OA, Viana MDEB. Effects ofchronic treatment with a water–alcohol extract from Erythrina mulungu onanxiety-related responses in rats. Biol Pharm Bull 2003;26:1538–42.

446 S.A. Faggion et al. / Epilepsy & Behavior 20 (2011) 441–446

[15] Ribeiro MD, Onusic GM, Poltronieri SC, Viana MB. Effect of Erythrina velutina andErythrina mulungu in rats submitted to animal models of anxiety and depression.Braz J Med Biol Res 2006;39:263–70.

[16] Vasconcelos SM, Lima NM, Sales GT, et al. Anticonvulsant activity of hydroalcoholicextracts from Erythrina velutina and Erythrinamulungu. J Ethnopharmacol 2007;110:271–4.

[17] Flausino JrOA, Santos LA,VerliH, PereiraAM,BolzaniVS,Nunes-De-SouzaRL.Anxiolyticeffects of erythrinian alkaloids from Erythrina mulungu. J Nat Prod 2007;70:48–53.

[18] Flausino Jr OA, Pereira AM, Bolzani VS, Nunes-De-Souza RL. Effects of erythrinianalkaloids isolated from Erythrina mulungu (Papilionaceae) in mice submitted toanimal models of anxiety. Biol Pharm Bull 2007;30:375–8.

[19] Paxinos G, Watson C. The rat brain in stereotaxic coordinates. 2nd ed. San Diego:Academic Press; 1986.

[20] Pinel JP, Rovner LI. Electrode placement and kindling: induced experimentalepilepsy. Exp Neurol 1978;58:335–46.

[21] Akerele O. Nature's medicinal bounty: don't throw it away. World Health Forum1993;14:390–5.

[22] Quintans Júnior LJ, Almeida JRGS, Lima JT, et al. Plants with anticonvulsantproperties: a review. Braz J Pharmacognosy 2008;18(Suppl):798–819.

[23] Garín-Aguilar ME, Luna JE, Soto-Hernández M, Valencia del Toro G, Vázquez MM.Efect of crude extracts of Erythrina americana Mill. on aggressive behavior in rats.J Ethnopharmacol 2000;69:189–96.