journal of ethnopharmacology - core.ac.uk · cytotoxicity and apoptogenic effects of lafoensia...

8
Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondes a , Cristiane Loiva Reichert a , Lucas Ferrari de Andrade b , Cid Aimbiré de Moraes Santos a,c , Almeriane Maria Weffort-Santos a,dn a Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Paraná, Av. Lothário Meissner, 632, Jardim Botânico, 80210-170 Curitiba, PR, Brazil b Programa de Pós-graduação em Biologia Celular e Molecular, Departamento de Biologia Celular, Universidade Federal do Paraná, Rua Francisco H. dos Santos, s/n, Jd. das Américas, 81531-990 Curitiba, PR, Brazil c Laboratório de Farmacognosia, Departamento de Farmácia, Universidade Federal do Paraná, Av. Lothário Meissner, 632, Jardim Botânico, 80210-170 Curitiba, PR, Brazil d Laboratório de Hematologia, Departamento de Patologia Médica, Universidade Federal do Paraná, Av. Lothário Meissner, 632, Jardim Botânico, 80210-170 Curitiba, PR, Brazil article info Article history: Received 26 April 2014 Received in revised form 6 September 2014 Accepted 10 September 2014 Available online 13 October 2014 Keywords: Lafoensia pacari Apoptosis Cytotoxicity Cell cycle Tumor lineages Brazilian Cerrado plants abstract Ethnopharmacological relevance: The stem barks of Lafoensia pacari have been traditionally used not only by South Amerindians but also by Brazilian and Paraguayan populations for treating a variety of unhealthy conditions to which their biological potential has been scientically documented in several reports over the last decade. Although its anticancer usage is also popular, no scientic support for such activity has been found. Aim: To provide scientic evidence for the anticancer popularity of Lafoensia pacari. Materials and methods: Extracts prepared according to the popular use along with a methanol extract and its four fractions were produced from Lafoensia pacari stem barks. The chromatogram prole of each one was obtained by HPLC. Several tumor cell lines were exposed to these solutions in in vitro assays and the effects evaluated by morphological, growth, and cell cycle status changes. Results: High toxicity determined by the lactate dehydrogenase levels with a signicant drop in the cell proliferation index were found for all cell lines included in this study after exposition to Lafoensia pacari extract and fractions. The morphological features along with the expression of annexin V have strongly suggested apoptosis induction, which has been conrmed by G0/G1 cell cycle arrest. Conclusions: The data have clearly shown that exposition of human tumor cell lines to Lafoensia pacari stem barks extract leads to apoptosis induction due to cell cycle arrest in G0/G1 phases, supporting its anticancer use. & 2014 Elsevier Ireland Ltd. All rights reserved. 1. Introduction Lafoensia pacari A. St.-Hil., Lythraceae, is a decidual tree that grows mostly in the Pantanal and Cerrado biomas of Brazil. Known as dedaleiraor mangava-brava, its stem barks have been traditionally used as a tonic and for treating inammation condi- tions particularly related to gastric ulcers, wounds or fevers (Tamashiro Filho et al., 2012). Named precious woodby the Tupi-Guarani Amerindians and morosyvóby Eastern Paraguayans, Lafoensia pacari barks have also been taken to treat several types of cancer (de Melo et al., 2011). Interestingly, the folk remedy has at least two ways of preparation: bark aqueous decoction (DEC) and cold-water long-standing maceration (MAC) (Solon et al., 2000). The amount of data published over the last decade or so concerning to the biological potential of Lafoensia pacari has been considerably extensive in which its anti-inammatory (Guimaraes et al., 2010; Rogerio et al., 2008), anti-allergic (Rogerio et al., 2010), analgesic (Nascimento et al., 2011), anti-edematous (Rogerio et al., 2006), microbicidal (de Lima et al., 2006; Porrio et al., 2009; Silva et al., 2010), antiviral (Müller et al., 2007), antioxidant (Galano et al., 2014; Solon et al., 2000), sedating, and antidepressant-like effects (de Matos et al., 2008; Galdino et al., 2009) have been scientically documented, and some of them even elucidated at a mechanistical level, corroborating its many traditional uses. Some of these and other studies have also provided information that places the ellagic acid (EA), the phenolic phytochemical marker of the species, as the activities´ major protagonist including the anti- tumor effects (Berni et al., 2012; Narayanan et al., 1999; Nascimento Contents lists available at ScienceDirect journal homepage: www.elsevier.com/locate/jep Journal of Ethnopharmacology http://dx.doi.org/10.1016/j.jep.2014.09.018 0378-8741/& 2014 Elsevier Ireland Ltd. All rights reserved. n Corresponding author at: Departamento de Patologia Médica, Universidade Federal do Paraná, Av. Lothário Meissner, 632. JardimBotânico, 80210-170 Curitiba, PR, Brazil. Tel.: þ55 41 3360 4087; fax: þ55 41 3360 4062. E-mail addresses: [email protected] (D.B. da Silva Marcondes), [email protected] (C.L. Reichert), [email protected] (L.F. de Andrade), [email protected] (C.A. de Moraes Santos), [email protected] (A.M. Weffort-Santos). Journal of Ethnopharmacology 157 (2014) 243250

Upload: hoangcong

Post on 29-Nov-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

Cytotoxicity and apoptogenic effects of Lafoensia pacari

Daniela Beck da Silva Marcondes a, Cristiane Loiva Reichert a, Lucas Ferrari de Andrade b,Cid Aimbiré de Moraes Santos a,c, Almeriane Maria Weffort-Santos a,dn

a Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Federal do Paraná, Av. Lothário Meissner, 632, Jardim Botânico,80210-170 Curitiba, PR, Brazilb Programa de Pós-graduação em Biologia Celular e Molecular, Departamento de Biologia Celular, Universidade Federal do Paraná,Rua Francisco H. dos Santos, s/n, Jd. das Américas, 81531-990 Curitiba, PR, Brazilc Laboratório de Farmacognosia, Departamento de Farmácia, Universidade Federal do Paraná, Av. Lothário Meissner, 632, Jardim Botânico,80210-170 Curitiba, PR, Brazild Laboratório de Hematologia, Departamento de Patologia Médica, Universidade Federal do Paraná, Av. Lothário Meissner, 632, Jardim Botânico,80210-170 Curitiba, PR, Brazil

a r t i c l e i n f o

Article history:Received 26 April 2014Received in revised form6 September 2014Accepted 10 September 2014Available online 13 October 2014

Keywords:Lafoensia pacariApoptosisCytotoxicityCell cycleTumor lineagesBrazilian Cerrado plants

a b s t r a c t

Ethnopharmacological relevance: The stem barks of Lafoensia pacari have been traditionally used not onlyby South Amerindians but also by Brazilian and Paraguayan populations for treating a variety ofunhealthy conditions to which their biological potential has been scientifically documented in severalreports over the last decade. Although its anticancer usage is also popular, no scientific support for suchactivity has been found.Aim: To provide scientific evidence for the anticancer popularity of Lafoensia pacari.Materials and methods: Extracts prepared according to the popular use along with a methanol extractand its four fractions were produced from Lafoensia pacari stem barks. The chromatogram profile of eachone was obtained by HPLC. Several tumor cell lines were exposed to these solutions in in vitro assays andthe effects evaluated by morphological, growth, and cell cycle status changes.Results: High toxicity determined by the lactate dehydrogenase levels with a significant drop in the cellproliferation index were found for all cell lines included in this study after exposition to Lafoensia pacariextract and fractions. The morphological features along with the expression of annexin V have stronglysuggested apoptosis induction, which has been confirmed by G0/G1 cell cycle arrest.Conclusions: The data have clearly shown that exposition of human tumor cell lines to Lafoensia pacaristem barks extract leads to apoptosis induction due to cell cycle arrest in G0/G1 phases, supporting itsanticancer use.

& 2014 Elsevier Ireland Ltd. All rights reserved.

1. Introduction

Lafoensia pacari A. St.-Hil., Lythraceae, is a decidual tree thatgrows mostly in the Pantanal and Cerrado biomas of Brazil. Knownas “dedaleira” or “mangava-brava”, its stem barks have beentraditionally used as a tonic and for treating inflammation condi-tions particularly related to gastric ulcers, wounds or fevers(Tamashiro Filho et al., 2012).

Named “precious wood” by the Tupi-Guarani Amerindians and“morosyvó” by Eastern Paraguayans, Lafoensia pacari barks have alsobeen taken to treat several types of cancer (de Melo et al., 2011).

Interestingly, the folk remedy has at least two ways of preparation:bark aqueous decoction (DEC) and cold-water long-standingmaceration (MAC) (Solon et al., 2000).

The amount of data published over the last decade or soconcerning to the biological potential of Lafoensia pacari has beenconsiderably extensive in which its anti-inflammatory (Guimaraeset al., 2010; Rogerio et al., 2008), anti-allergic (Rogerio et al., 2010),analgesic (Nascimento et al., 2011), anti-edematous (Rogerio et al.,2006), microbicidal (de Lima et al., 2006; Porfirio et al., 2009; Silvaet al., 2010), antiviral (Müller et al., 2007), antioxidant (Galanoet al., 2014; Solon et al., 2000), sedating, and antidepressant-likeeffects (de Matos et al., 2008; Galdino et al., 2009) have beenscientifically documented, and some of them even elucidated at amechanistical level, corroborating its many traditional uses.

Some of these and other studies have also provided informationthat places the ellagic acid (EA), the phenolic phytochemical marker ofthe species, as the activities´ major protagonist including the anti-tumor effects (Berni et al., 2012; Narayanan et al., 1999; Nascimento

Contents lists available at ScienceDirect

journal homepage: www.elsevier.com/locate/jep

Journal of Ethnopharmacology

http://dx.doi.org/10.1016/j.jep.2014.09.0180378-8741/& 2014 Elsevier Ireland Ltd. All rights reserved.

n Corresponding author at: Departamento de Patologia Médica, UniversidadeFederal do Paraná, Av. Lothário Meissner, 632. Jardim Botânico, 80210-170 Curitiba,PR, Brazil. Tel.: þ55 41 3360 4087; fax: þ55 41 3360 4062.

E-mail addresses: [email protected] (D.B. da Silva Marcondes),[email protected] (C.L. Reichert), [email protected] (L.F. de Andrade),[email protected] (C.A. de Moraes Santos), [email protected] (A.M. Weffort-Santos).

Journal of Ethnopharmacology 157 (2014) 243–250

Page 2: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

et al., 2011; Pari and Sivasankari, 2008; Rogerio et al., 2008, 2006;Solon et al., 2000; Umesalma and Sudhandiran, 2010, 2011).

Although it has been recently reported that an ethanolic extractprepared from its stem barks is not mutagenic according to thein vitro Ames assay, and genotoxicity could be achieved only athigh doses using the in vivo mouse bone marrow micronucleustest (Lima et al., 2013), studies concerning to Lafoensia pacaricancer-related uses are lacking. As such and aiming to clarify andsupport the anticancer popular usage of Lafoensia pacari, we haveconducted an in vitro study using several tumor cell lines.

The results herein presented have clearly shown, for the firsttime, that exposition of human leukemic cell lines as well as othersrepresentatives of cervical, colon, and neuroblastoma humancancers to Lafoensia pacari stem barks extract leads to changescharacteristics of apoptosis induction, including morphologicalaspects and alteration of the cell cycle status.

2. Material and methods

2.1. Reagents and chemicals

Unless otherwise specified, all solid and liquid chemicals usedin this work were purchased from Sigma (São Paulo, BR) or Merck(São Paulo, BR) representatives.

2.2. Plant material, extracts and fractions preparation

Stem barks of Lafoensia pacari A. St.-Hil. were collected in thesurroundings areas of Maracaju (21145037.4″S,55121081.9″WO),Mato Grosso do Sul state, Brazil, in May 2011, under the govern-mental Normative Instruction No. 154/2007/SISBio regulation,receiving the 29123-1 authorization. They were identified by theBotanist Gerdt Hatschbach and a voucher specimen under theProtocol MBM11065 has been deposited at the Per Karl DusénCollection of the Museu Botânico de Curitiba, Paraná State, Brazil.The plant material was air dried at room temperature for five daysfollowed by three days at 60 1C. Dried and fragmented barks (10 g)were macerated in double distilled water for 24 h in a domesticrefrigerator (4–8 1C) or boiled for 10 min to prepare the cold-waterlong standing macerate (MAC) and the aqueous decocted (DEC)extracts, respectively. Powdered bark material (300 g/0.425 mmmesh) was also exhaustively macerated in MeOH for 72 h in thedark, at room temperature, filtered, and evaporated under pres-sure to dryness at 60 1C (26.5% w/w yield). The methanolic extract(ExtMeOH—60 g) was kept in cold water (900 mL) for 8 h underagitation, and a precipitate gave rise to the hydrophobic fraction(F1; 5.64% w/w yield). The dissolved material was partitioned(3�100 mL) with ethyl acetate (F2; 0.39% w/w yield), n-butanol(F3; 3.78% w/w yield), and water (F4; 7.96% w/w yield). Eachextract and fractions were lyophilized and kept at minus 25 1C. Forthe biological experiments, extracts and fractions were preparedat 4% (w/v) in DMSO and further diluted to 1% directly into RPMI1640 (Himedia Laboratories; Mumbal, India) or IMDM (Gibco;Paisley, UK) media supplemented with 10% heat-inactivated fetalcalf serum (FCS—Cultilab; Campinas, São Paulo, BR). After sterilizedthrough a 0.22 mm pore size filter, 1 mL aliquots were stored atminus 25 1C. Final concentrations varying from 0.1 to 1000 μg/mLwere obtained by thawing and diluting the 1% stock solution in theappropriate culture medium just before the experiments.

2.3. HPLC-PAD analyses

The analysis was performed by a Varian ProStar Gradient highperformance liquid chromatography coupled with photodiodearray detector (HPLC-PAD) consisting of a Solvent Delivery Module

(Mod. 230), an Auto Sampler (Mod. 410) injector, and a PhotodiodeArray Detector (Mod. 335) coupled to the chromatograph. A 5 μmKromasil C-18, 4.6�250 mm analytical column maintained at roomtemperature was used. Samples of the DEC, MAC, and ExtMeOHextracts as well as of the four fractions and ellagic acid (12.48 mg/mL),were dissolved in MeOH, filtered with 0.45 μm nylon filters andinjected in the HPLC-PAD instrument using a solvent system con-sisting of (A) acidulated water purified by a Milli-Q system (MiliporeMilford, MA), with 0.1% phosphoric acid, and (B) methanol. Thesolvents were run at a flow rate of 1.0 mL/min using the followinglinear gradient: 5% B to 65% B for 40 min, 65% B to 95% B for 5 min,5% B for 5 min. Detection was monitored at 270 nm to comparechromatographic profiles. After that, triplicate samples of theExtMeOH (1 mg/mL) and of the hydrophobic (F1; 1 mg/mL), ethylacetate (F2; 0.08 mg/mL), n-butanol (F3; 0.4 mg/mL), and aqueous(F4; 1 mg/mL) fractions were prepared, filtered with 0.45 μm nylonfilters, injected using de same HPLC-PAD conditions, and detectionfor ellagic acid was monitored at 270 nm and expressed in μg/mg(or μM). Chromatographic peak has been identified by comparing theretention time with standard recorded under the same conditions.A calibration curve was obtained with a commercial ellagic acidstandard after dilution in methanol (1.04 to 12.48 μg/mL). Thecorrelation coefficient obtained for ellagic acid was r2¼0.999.

2.4. Cell lines and maintenance

The U-937, Jurkat, and Daudi leukemic cell lines were pur-chased from Rio de Janeiro Cell Bank, Universidade Federal do Riode Janeiro (RJBC; Rio de Janeiro, BR). HeLa and HRT-18 cell lineswere kindly provided by Dr. K. S. Paludo (Departamento deBiologia Estrutural, Molecular e Genética, Universidade Estadualde Ponta Grossa, Ponta Grossa, Brazil); the SH-SY5Y cell line waskindly provided by Dr. R. F. Bachinski (Centro de Ciências Médicas,Universidade Federal Fluminense, Rio de Janeiro, Brazil). All celllines were kept at 37 1C in a 5% CO2 humidified atmosphere inRPMI 1640 or IMDM full medium, consisting of 10% FCS, 2 mML-glutamine, 3 g/L sodium bicarbonate, 100 IU/mL G-penicillin,and 50 mg/mL streptomycin sulphate. Twice a week, the cells weresplit at 105 cells/mL ratio. For the experiments, all cell lines wereharvested at the exponential growth phase and checked forviability using the Trypan Blue assay, in which the number ofviable cells always exceeded 90%.

2.5. Cell viability and toxicity

Toxic effects of the extract and fractions from 0.1 to 1000 mg/mLand of all solutions used in this study on peripheral blood, bonemarrow cells, and cell lines were evaluated using the Trypan Blueassay. Viability was taken as the percentage of cells not incorpor-ating the stain7one standard deviation (1 SD), counted in ahemocytometer after directly diluting the cell suspension with0.4% Trypan Blue solution.

2.6. Proliferation assay

For each cell line, cell suspensions in full medium weredistributed into flat-bottomed 24- or 96-well culture dishes(TPP; Switzerland) and left for 24 h at 37 1C in a fully humidified,5% CO2 incubator. Extract solution was then added to give finalconcentrations ranging from 0.1 to 1000 mg/mL in a 1000 or 200 mLfinal volume/well and further incubated for 6 h to five days. Formouse bone marrow cells, a 100 mL containing 2�104 MNC in fullIMDM mediumwere seeded per well and simultaneously added ofextract (0.1 to 1000 mg/mL) in a 200 mL final volume/well for aweek. The number of cells recovered was evaluated in a hemo-cytometer. Each experiment was performed in triplicates and the

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250244

Page 3: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

results were expressed as the mean cell number recovered71 SDafter incubation or as a proliferation index, of at least threeindependent experiments.

2.7. Lactate dehydrogenase (LDH) assay

LDH assay was performed with U-937 cells treated with 10,100, and 1000 mg/mL of extract for 24 h at 37 1C in a fullyhumidified, 5% CO2 incubator by assessing the enzyme contentreleased into the media as a marker of dead cells and the enzymepresent in the cell lysates (LDHCL)) as a marker of remaining livingcells using the LDH Liquiform commercial kit (Labtest DiagnósticaS.A., Curitiba, Brazil). After incubation, the culture supernatantswere separated by spinning down at 400 g/5 min and used tomeasure the LDH released into the media (LDHSPN) while the cellpellets were treated with lysing buffer (0.1 M Tris–HCl pH 7.2 sup-plemented with 1% Nonidet P-40, 0.01% SDS, 100 μM PMSF, and1 μg/mL aprotinin) giving rise to cell lysates. The absorbancesobtained at 340 nm in a Cobas Mira Classic instrument (Roche)were used to obtain the % of final LDH according to the followingequations: Free LDH (%)¼(LDHSPN�100)/(LDHSPNþLDHCL);100�(Free LDH %)¼% Final LDH.

2.8. Human mononuclear cells and phytohemagglutinin-elicitationassay

The protocol used for human peripheral blood collection hasbeen approved by the Health Sciences Ethical Committee of theFederal University of Paraná, CEP/SD, No. 039.SI003/04-01, whichis in accordance with the Declaration of Helsinki. The isolation ofthe mononuclear cells (MNC) as well as the phytohemagglutinin-elicitation assay were both performed essentially as describedelsewhere (Nardin et al., 2010).

2.9. Morphological assessment

Whereas morphology was accessed, 5–8�104 cells cytocentri-fuged using a CytoproTM instrument (Wescor, Utah, USA) werestained with May-Grunwald–Giemsa protocol. At least 200 cellsper assay were observed and differentiated for each experimentunder immersion oil light microscopy.

2.10. Annexin V and 7-AAD co-expression by flow cytometry

After 24 h of subculture at 37 1C, U-937, HeLa, HRT-18, andSH-SY5Y cell lines as well as human peripheral MNC (106 cells/mL)were treated with extract (10–1000 mg/mL) for 6, 12 or 24 h. Thetreated cells were then washed with cold D-PBS, stained simulta-neously with Annexin V-FITC (BD Bioscience, San Jose, CA, USA)and 7-AAD (BD Bioscience, San Jose, CA, USA), and analyzed byflow cytometry using an FACS Calibur instrument (Becton &Dickinson, San Diego, California, USA) and the strategies describedelsewhere (de Lima et al., 2010) in which at least 105 events wereacquired in the gated populations. Data were analysed using theWinMDI 2.8 software. The results were expressed as the meanpercentage of positive cells for each population from at least threeindependent experiments.

2.11. Cell cycle analysis

U-937 cells (106) exposed to 100 and 1000 mg/mL of extractfor 24 h at 37 1C were washed with cold D-PBS and fixed for30 min with 1% paraformaldehyde. After washing with cold D-PBS,the cells were stained directly with 20 mL of a solution containingpropidium iodide (PI; 2 mg/mL), 300 mL RNase (10 mg/mL) andTriton X-100 (0.3%) for 15 min in the dark at room temperature

followed by DNA content analysis in a FACS Calibur instrument,in which 105 events per assay were acquired in a linear scale.Histograms based on PI-stained cells were plotted by the WinMDI2.8 software program that provides the estimate of percentage ofcells with fractional DNA content (apoptotic cells: Ap) and cells inG1, S, and G2/M phases of the cycle.

2.12. Colony-forming assays

The clonogenic potential of U-937, Daudi, and Jurkat cells wasassayed by counting colonies grown in the commercial viscousmedium MethoCult™ (StemCell Technologies Inc.; Vancouver, CA)supplemented with Stem Cell Factor (50 ng/mL), GM-CSF (10 ng/mL),IL-3 (10 ng/mL), and erythropoietin (3 IU/mL) recombinant humancytokines. The cells were first washed with PBS, suspended intoIMDM medium supplemented with 10% FCS, and the concentrationadjusted to a density of 2�105 cells/mL. To 1 mL of MethoCult, 103

cells were added. The mixture was plated in three aliquots of 0.3 mLeach into 24-well plastic culture dishes (TPP, Switzerland). Lafoensiapacari methanol extract (1–100 mg/mL) was added to the platingmedium just before the cells. The dishes were incubated at 37 1C in afully humidified atmosphere supplemented with 5% CO2 in air. After7–10 days, the colonies were scored by direct observation under aninverted microscope. Each independent experiment was performedin triplicates and the results were expressed as the mean colonynumber71 SD.

2.13. Data analysis

Where applicable, differences between groups were analyzedusing Student’s t-test and one-way analysis of variance followedby the Tukey HSD �α¼0.05 multiple comparison test (PRISMsoftware, Graph Pad, La Jolla, CA). P values o0.05 were consideredstatistically significant.

3. Results and discussion

3.1. Plant extracts, fractions, and HPLC profiles

According to the popular usage, we have initially prepared twoextracts: one involving bark decoction (DEC) and the other basedon cold-water long-standing maceration (MAC). In parallel, amethanol extract (ExtMeOH) following classical extraction proce-dures was also prepared. HPLC analyses of these three solutionshave shown peaks with similar retention time (Fig. 1). Thestandard biomarker ellagic acid (EA) at 12.48 mg/mL (41.1 μM)resolved a peak at retention time of 34.5 min, and was identifiedin all, with 5.24 mg/mg (17.3 μM) in DEC, 9.89 mg/mg (32.6 μM) inMAC, and 9.05 mg/mg (29.9 μM) in ExtMeOH.

Partition of the ExtMeOH with solvents within a broad polarityrange led to four fractions (F1: hydrophobic; F2: EtOAc; F3:n-BuOH; F4: aqueous) whose HPLC profiles were quite interest-ingly as they all resembled the one presented by the mothersolution. EA was also present in these fractions, differing only inconcentration (28.1 μM in F1; 45.7 μM in F3, and 3.14 μM in F4),being the predominant compound of F2 (253.4 μM). These resultsshowed that temperatures, time exposure or solvent polarity havenot influenced the ending compounds present in the extract.

3.2. Comparative effects of extracts, fractions, and ellagic acidon U-937 cells behavior

To observe the effects on cell growth, U-937 cells, a mono-blastic human leukemic cell line, were exposed to all thesesolutions in concentrations ranging from 0.1 to 1000 mg/mL

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250 245

Page 4: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

for 24 h. As shown in Fig. 2A, no interference on cell proliferationstatus up to 10 mg/mL was observed in these short-term tests.However, when compared to control values, a concentration-related toxicity was present at higher concentrations, with asignificant drop in the cell proliferation index. No cells could berecovered from cultures in which the final concentration was1000 mg/mL. These findings were corroborated by the significantcytotoxicity determined by the lactate dehydrogenase (LDH) levels(Fig. 2B).

Interestingly, EA assayed under the same protocol within abroad concentration range corresponding to its presence withinsolutions (0.3 to 3300 μM) showed significantly lesser effects onthese parameters, and no toxicity up to 330 μM was observed(Fig. 2A and B).

3.3. Influence of Lafoensia pacari extract on U-937 cells’ physicalproperties

Once U-937 cells showed highly intoxicated with the ExtMeOHextract, and based on the concept that when cells go into a deathprocess, their mechanical properties, generally represented by sizeand inner complexity, change and the cells are able to alter theircapture position in a flow cytometer, with a decrease in the opticaldetection of these parameters, we exposed them to 100 mg/mL ofthis extract for 24 h and measured their size (FSC) and inner

complexity (SSC). The results confirmed the previous observations,with significant toxic effects promoted by the ExtMeOH treatment(Fig. 2C). In contrast, no changes in the FSC and SSC parametersobtained by flow cytometry were detected in EA treated cells.

In addition, among the fractions tested, F2, which presentedthe highest amount of EA, showed a pattern of effects not assignificant as did the ExtMeOH or the other fractions, suggestingthat EA, at least as it is, and using the experimental settingsincluded in this study, is not the sole or the main compoundresponsible for the effects described for the crude extract. Becauseseveral studies have described the anti-proliferative andapoptotic-induced potential of EA that naturally occurs in plants,fruits, and vegetables, through modulation of several subcellularsignaling pathways, and anti-oxidant defense mechanisms(Edderkaoui et al., 2013; Losso et al., 2004; Mishra and Vinayak,2014a, b; Narayanan et al., 1999; Vanella et al., 2013; Zhang et al.,2014), these results are of particular relevance for the potential ofthe Lafoensia pacari extract itself as an anti-cancer remedy, open-ing the possibility that one or more compounds apart from EA inthe extract are responsible for such a strong activity.

3.4. Morphological and proliferation effects of ExtMeOH on humanleukemic cell lines

As the MAC and DEC traditional folk remedies along with thefractions have all showed similar HPLC profiles of the ExtMeOH,have also evoked similar effects elicited by the methanolicpreparation alone, and are technically reproducible, we decidedto use only this extract to further study the Lafoensia pacari effectsupon other two leukemic cell lines in similar short-term assays,but expanding the exposition time from 6 to 48 h.

Thus, Daudi and Jurkat cells, both of lymphoid origin, weresubjected to the same treatment performed with U-937 cells. Asshowed in Fig. 3D, just after 6 h of treatment, cell viability andrecovery were both significantly affected, even at lower concen-trations such as 1 mg/mL. Once again, no cells at 1000 mg/mL wererecovered. The distinct effects on each cell line after extractexposure may be better visualized in the panels A, B, and Cof Fig. 3, in which dramatic changes on cell phenotype wereobserved in cytospun preparations stained with May-Grunwald–Giemsa. Typical features of apoptosis induction were found incontrast with the original morphology maintained by theuntreated population (panels 1-A, B and C). Within this context,the presence of cytoplasmic vacuoli, chromatin condensation,nuclear fragmentation, and formation of apoptotic bodies wereevident in the treated populations. After 48 h, the variable numberof naturally occurring cytoplasmic vacuoli had almost disappeared,giving space to a prominent and intensely blue, reactive-likecytoplasms in which, when present, nuclei with a highly con-densed chromatin were immersed (panels 3-A, B and C).

3.5. Effects of ExtMeOH on human leukemic cells´ clonogenicpotential

These leukemic cell lines are able to generate colonies in softsystems when appropriately stimulated. In order to investigatewhether the inhibitory effect would affect their clonogenicresponse, they were exposed to 1 and 10 mg/mL of ExtMeOH. Itresulted in a concentration-dependent inhibition of colony forma-tion for all when compared to the respective untreated popula-tions. For U-937 cells the number of colonies increased from100.0719.9 (control) to 124.0712.2 at 1.0 mg/mL followed by asignificant decrease to 72.377.5 at 10 mg/mL (Po0.01). For Daudicells, the values decreased from 103.071.0 (control) to 86.0721.8and 80.776.5, respectively for the same dose range (Pr0.01).Finally, for Jurkat cells, the colony number significantly declined

Fig. 1. Representative HPLC chromatograms of Lafoensia pacari extracts, fractions,and ellagic acid (EA). Bark decoction (DEC); cold-water long-standing maceration(MAC); methanol extract (ExtMeOH) and its hydrophobic (F1), ethyl acetate (F2),n-butanol (F3), and aqueous (F4) fractions.

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250246

Page 5: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

from 84.776.7 (control) to 71.379.1 and 26.3726.3 at 1 and10 mg/mL (Po0.05), respectively. At a 100 mg/mL, complete abro-gation of colony formation response was observed for all cell lines.It is important to note that, although the number of colonies wasaffected, their size and morphology were not different from thecontrol experiments.

3.6. Apoptosis evaluation by flow cytometry

Apoptosis can be detected not only on the basis of cellmorphology but also by flow cytometric analysis of heterogeneouscell populations, in which cell viability is preserved, allowing thesimultaneous detection of intact cells, cells undergoing apoptosis,and dead cells resulting from apoptotic and/or necrotic processesin the same sample and at the same time. To confirm whether theeffects observed for leukemic cells were compatible with apoptosisinduction, we labelled U-937 cells exposed to Lafoensia pacariExtMeOH for 6 to 24 h with annexin V and 7-AAD and analysedthem by flow cytometry.

Annexin V is a Ca2þ dependent phospholipid-binding proteinwith high affinity for phosphatidylserine (PS) present in the innerside of the plasma membrane. In the early stages of apoptosischanges difficult to recognize occur at the cell surface, includingthe translocation of PS from the inner side of the plasmamembrane to the outer layer, by which it becomes exposed atthe external cell surface. This phenomenon is present not onlyduring apoptosis, but occurs also during necrosis. The differencebetween these two forms of cell death is that during the initialstages of apoptosis the cell membrane remains intact, while at themoment that necrosis occurs, the cell membrane integrity is lostand the cell becomes leaky, allowing the entrance of molecules aslarge as 7-AAD, which can be used to mark necrotic cells.

Therefore, in a flow cytometric dot plot chart, the way cellsbind to annexin V (x axis) and/or 7-AAD (y axis) can discriminateviable, intact cells (double negative staining/lower left quadrant)from apoptotic cells (annexin V positive cells/lower right quad-rant) and necrotic cells (double positive staining/upper rightquadrant), corresponding to the different stages of the apoptoticcascade in vitro.

Fig. 2. Effects of Lafoensia pacari on U-937 cells. Exponentially growing U-937 cells were exposed to the indicated concentrations of Lafoensia pacari methanol extract(ExtMeOH) or its hydrophobic (F1), ActOEt (F2), n-BuOH (F3) or aqueous (F4) fractions, and to ellagic acid (EA) for 24 h. (A) Each line represents the mean proliferationindex7SD of recovered cells estimated in a hemocytometer in relation to the untreated group (CT). (B) Mean percentage7SD of LDH content measured in U-937 cell lysates,both obtained from at least three independent experiments, each one performed in triplicates. (C) Alteration of size (FSC) and inner complexity (SSC) morphological aspectsof ExtMeOH- and EA-treated U-937 cells optically captured by flow cytometry when compared to untreated cells (CT). nnPo0.01; nnnPo0.001.

Fig. 3. Lafoensia pacari methanol extract effects on leukemic cell lines. Characteristic phenotypes of U-937 (A), Daudi (B), and Jurkat cells (C) stained with May-Grünwald–Giemsa before (1) and after 6 (2) and 48 h (3) exposition to 1000 μg/mL of Lafoensia pacari methanol extract. Cell proliferation and viability of these cell lines (D) after 48 hexposition to the indicated concentrations of extract. The vertical bars indicate the mean number7SD of the surviving fraction of leukemic cells counted in ahemocytometer, and the lines indicate the mean percentage7SD of cell viability determined by the Trypan Blue exclusion assay in relation to the untreated population (CT)from at least three independent experiments. nPo0.05, nnPo0.01, nnnPo0.001. (For interpretation of the references to color in this figure legend, the reader is referred to theweb version of this article.)

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250 247

Page 6: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

Using this strategy, the majority (495%) of the untreated intactU-937 population (CT) was composed of healthy, viable cells at 0 h,showing annexin V and 7-AAD double negative staining (Fig. 4A).After 6 h and over, ExtMeOH treatment resulted in the appearanceof two distinct populations: one presenting annexin V singlepositive expression, which was assigned for early apoptotic cells(eAp), and other showing annexin V and 7-AAD double positivestaining (Fig. 4A/upper right quadrant), that we have referred aslate apoptotic cells (lAp). In Fig. 4B, it is possible to visualize thatthese populations were also accompanied by a progressive andsignificant increase of a necrotic cell population (N).

Probably, the double positive labeled U-937 population wascomposed of cells with the two distinct morphologies observedin Fig. 3A.2 and A.3: one containing small, irregularly-shaped cellswith condensed and fragmented nuclei, consistent with cells inthe late stage of apoptosis, and one containing larger cells withunfragmented nuclei that probably stained with 7-AAD, and weresuggestive of cells undergoing necrosis.

3.7. Effects of ExtMeOH on U-937 cell cycle

The results of the kinetics of apoptotic death of U-937 cells werealso compatible with the cell cycle analysis of these populations, inwhich the cells were prepared to assess their relative DNA contentin different phases of the cell cycle by flow cytometry (Fig. 5A).

In this assay the concentration-related increasing of the U-937apoptotic population could be easily observed (Fig. 5B), varyingfrom 17.7072.34% in the control population (CT) to 33.3271.99%at a 100 μg/mL, and 48.1575.15% at a 1000 μg/mL. Interestingly,the significant accumulation of cells in G0/G1 (36.0273.72%,34.6970.52%, and 34.6474.74% for untreated cells, cells treatedwith 100 and 1000 μg/mL, respectively) with the concomitantdecrease of cells in S phase (from 41.8574.35% to 34.7377.22and 24.8173.88% respectively for control cells, cells treated with100 and 1000 μg/mL) provide evidence that the observed apopto-tic cell death induced by Lafoensia pacari ExtMeOH may be partlydue to cell cycle arrest in G0/G1 phase.

3.8. Effects of ExtMeOH on human cancer cell lines

Significant results of apoptosis induction were also demon-strated by HRT-18, SH-SY5Y, and HeLa human cell lines whenexposed to Lafoensia pacari ExtMeOH extract followed by annexinV and 7-AAD treatment (Fig. 6A), broadening the anti-proliferativeeffects of the plant. Worthy of note is that the apoptosis-inductioneffects on these cell lines were observed at lower concentrations,and only a few cells were observed in the late apoptosis gate (lAp)for HeLa cells, suggesting a more prolonged retention on G1/G0phase for these cells.

Fig. 4. Effects of Lafoensia pacari extract on U-937 cells. (A) Exponentially growing U-937 cells exposed to 1000 μg/mL of Lafoensia pacari stem barks extract for 6 to 24 hwere labelled with annexin-V and 7-AAD and subject to flow cytometric analyses to identify subpopulations undergoing early (lower right quadrant/eAp), late (upper rightquadrant/ lAp) apoptosis or necrosis (upper left quadrant/N). Each bar represents the mean percentage71 SD of cells detected in each quadrant. nnnPo0.001 compared tountreated (CT) population.

Fig. 5. Effects of Lafoensia pacari extract on U-937 cell cycle phases. Exponential growing U-937 cells were exposed to the indicated concentrations of Lafoensia pacari extractfor 24 h and analysed for cell cycle status by flow cytometry. (A) The histograms are representative of one of at least three similar experiments of the dose-related DNAcontent detected in each cell cycling phase. (B) Each bar represents the mean percentage71 SD of cells detected in each phase. Significant differences were comparedbetween control (CT) and treated cells (nnPo0.01; nnnPo0.001).

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250248

Page 7: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

3.9. Effects of ExtMeOH on human peripheral mononuclear cells

Because cancer cells have different proliferative profile whencompared to normal cells, we have used peripheral mononuclearcells (MNC) from healthy donors, in which the majority arelymphocytes that are not cycling under normal circumstances, totest whether the apoptotic effects of Lafoensia pacari ExtMeOH ontumor cells would also affect normal cells.

Exposition to 10 and 100 mg/mL of Lafoensia pacari ExtMeOHfor 24 h also triggered a concentration-related apoptosis in thesecells (Fig. 6B). However, this phenomenon was observed mainlywhen MNC were previously stimulated to proliferate with phyto-hemagglutinin (PHA-MNC), in which the number of late apoptoticcells were significantly higher than those observed for the unsti-mulated population (Fig. 6C), suggesting that the effect of inducingapoptosis rests on cells that are cycling and/or under intensemetabolic activity.

4. Conclusion

Mainly as an infusion or a decoct, many plants have beentraditionally used for the treatment of several diseases and havegained recognition as biological response modifiers. A relevant

concern is the detection of effects present in these preparationsthat may interfere with cell division. In this context, plants havebeen a source of clinically relevant anticancer compounds andnowadays a variety of natural products and derivatives belong tothe repertoire of cancer chemotherapeutic agents.

Because preparations of Lafoensia pacari stem barks have beenreferred in traditional Brazilian and Paraguayan medicines forcancer treatment, and rare studies concerning their antitumoractivity have been reported, it prompted us to investigate thebehavior of some leukemic cell lines and others representatives ofhuman cancers when exposed to scaled concentrations of theirextract.

The experimental results of this study clearly showed that thestem barks of Lafoensia pacari, independently of its preparationway (as a methanol extract, as a decoction or a macerate), have thesame components’ profile, and exhibit significant inhibitoryin vitro effects against the growth and proliferation of severalleukemic and tumor cell lines by triggering cell cycle arrest thatleads to apoptogenic events. They also induced morphologicalchanges characteristic of apoptosis, which were confirmed by theirhigh incorporation of annexin V. More interestingly, these effectswere directly related to the concentration used.

Cell cycle arrest and induction of apoptosis are linked events inwhich numerous signaling molecules and regulatory proteins are

Fig. 6. Effects of Lafoensia pacari extract on human cell lines. (A) Exponentially growing HRT-18, SH-SY5Y, HeLa cells as well as mononuclear cells (MNC/B) from peripheralblood exposed to the indicated concentrations of Lafoensia pacari stem barks extract for 24 h were labelled with annexin V and 7-AAD and subject to flow cytometricanalyses to identify subpopulations undergoing early (lower right quadrant/eAp), late (upper right quadrant/lAp) apoptosis or necrosis (upper left quadrant/N). A fraction ofperipheral MNC were pre-treated with phytohemagglutin for 120 h (PHA-MNC) before exposure to extract. (C) Each bar represents the percentage of cells detected in eachquadrant of a representative experiment. nPo0.05; nnPo0.01; nnnPo0.001, all compared to untreated population; #Po0.01 when compared to 10 mg/mL.

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250 249

Page 8: Journal of Ethnopharmacology - core.ac.uk · Cytotoxicity and apoptogenic effects of Lafoensia pacari Daniela Beck da Silva Marcondesa, Cristiane Loiva Reicherta, Lucas Ferrari de

involved in a highly regulated process that occurs as part ofdifferentiation, proliferation, and growth of normal and malig-nant cells.

Many medicinal plants possess significant anticancer activity,in which this beneficial health effect may be attributed to one ormore of their constituents. As such, these molecules may interactwith cellular mechanisms, including interference with membraneand receptors, modulation of signaling cascades, interaction withenzymes involved in tumor promotion/progression, interactionwith oncogenes and oncoproteins, or direct or indirect interactionswith nucleic acids and nucleoproteins.

Although we did not explore these events at a mechanisticallevel and further investigations to identify whether one or moremolecules are responsible for the Lafoensia pacari stem barkseffects are under way, no data concerning to its anticancerpotential as reported in this study are available in the literature.Therefore, the overall results herein presented are relevant as theyprovide the first scientific evidence to support the traditionalanticancer applications of Lafoensia pacari, confirming the indi-genous wisdom. Moreover, they place this plant as a source ofcompounds with the potential to be further exploited for thediscovery and development of new anticancer drugs.

Acknowledgements

This work was supported by grants and fellowships from theCNPq (Grants 479191/2011-5 and 311787/2012-6), Brazil. DBSMand CLR thank CAPES for scholarship support. CAMS is a recipientof a CNPq productivity fellowship.

References

Berni, A., Grossi, M.R., Pepe, G., Filippi, S., Muthukumar, S., Papeschi, C., Natarajan, A.T.,Palitti, F., 2012. Protective effect of ellagic acid (EA) on micronucleus formationinduced by N-methyl-N0-nitro-N-nitrosoguanidine (MNNG) in mammalian cells, inin vitro assays and in vivo. Mutation Research 746, 60–65.

de Lima, M.P., Hilst, L.F., Mattana, F.V.R., Santos, C.A.D., Weffort-Santos, A.M., 2010.Alkaloid-rich fraction of Himatanthus lancifolius contains anti-tumor agentsagainst leukemic cells. Brazilian Journal of Pharmaceutical Sciences 46,273–280.

de Lima, M.R., de Souza Luna, J., dos Santos, A.F., de Andrade, M.C., Sant’Ana, A.E.,Genet, J.P., Marquez, B., Neuville, L., Moreau, N., 2006. Anti-bacterial activity ofsome Brazilian medicinal plants. Journal of Ethnopharmacology 105, 137–147.

de Matos, L.G., Santos, L.D., Ferreira, R.N., Pontes, I.S., de Paula, J.R., Costa, E.A., 2008.Anti-inflammatory, antinociceptive, and sedating effects of Lafoensia pacariaqueous extract. Pharmaceutical Biology 46, 341–346.

de Melo, J.G., Santos, A.G., de Amorim, E.L., do Nascimento, S.C., de Albuquerque, U.P.,2011. Medicinal plants used as antitumor agents in Brazil: an ethnobotanicalapproach. Evidence Based Complementary and Alternative Medicine 2011,Article ID 365359, 14.

Edderkaoui, M., Lugea, A., Hui, H., Eibl, G., Lu, Q.Y., Moro, A., Lu, X., Li, G., Go, V.L.,Pandol, S.J., 2013. Ellagic acid and embelin affect key cellular components ofpancreatic adenocarcinoma, cancer, and stellate cells. Nutrition and Cancer 65,1232–1244.

Galano, A., Francisco Marquez, M., Perez-Gonzalez, A., 2014. Ellagic acid: anunusually versatile protector against oxidative stress. Chemical Research inToxicology 27, 904–918.

Galdino, P.M., Nascimento, M.V., Sampaio, B.L., Ferreira, R.N., Paula, J.R., Costa, E.A.,2009. Antidepressant-like effect of Lafoensia pacari A. St.-Hil. ethanolic extractand fractions in mice. Journal of Ethnopharmacology 124, 581–585.

Guimaraes, H.A., Nascimento, M.V.M., Tavares, A., Galdino, P.M., de Paula, J.R., Costa, E.A.,2010. Effects of ethanolic extract of leaves of Lafoensia pacari A. St.-Hil., Lythraceae

(pacari), in pain and inflammation models. Revista Brasileira de Farmacognosia 20,328–333.

Lima, D.C., Silva, C.R., Sampaio, B.L., de Paula, J.R., Chen-Chen, L., 2013. Antigeno-toxic, and anticytotoxic activities of an ethanolic extract of Lafoensia pacari(Lythraceae) stem bark in bacteria and mice. Genetics and Molecular Research12, 3887–3896.

Losso, J.N., Bansode, R.R., Trappey 2nd, A., Bawadi, H.A., Truax, R., 2004. In vitro anti-proliferative activities of ellagic acid. Journal of Nutritional Biochemistry 15,672–678.

Mishra, S., Vinayak, M., 2014a. Ellagic acid induces novel and atypical PKC isoformsand promotes caspase-3 dependent apoptosis by blocking energy metabolism.Nutrition and Cancer 66, 675–681.

Mishra, S., Vinayak, M., 2014b. Ellagic acid inhibits PKC signaling by improvingantioxidant defense system in murine T cell lymphoma. Molecular BiologyReports 41, 4187–4197.

Müller, V., Chávez, J.H., Reginatto, F.H., Zucolotto, S.M., Niero, R., Navarro, D., Yunes, R.A.,Schenkel, E.P., Barardi, C.R.M., Zanetti, C.R., Simões, C.M.O., 2007. Evaluation ofantiviral activity of South American plant extracts against herpes simplex virus type1 and rabies virus. Phytotherapy Research 21, 970–974.

Narayanan, B.A., Geoffroy, O., Willingham, M.C., Re, G.G., Nixon, D.W., 1999. p53/p21(WAF1/CIP1) expression and its possible role in G1 arrest and apoptosis inellagic acid treated cancer cells. Cancer Letters 136, 215–221.

Nardin, J.M., Lima, M.P., Machado, J.C., Hilst, L.F., Santos, C.A.D., Weffort-Santos, A.M.,2010. The uleine-rich fraction of Himatanthus lancifolius blocks proliferativeresponses of human lymphoid cells. Planta Medica 76, 697–700.

Nascimento, M.V., Galdino, P.M., Florentino, I.F., Sampaio, B.L., Vanderlinde, F.A.,de Paula, J.R., Costa, E.A., 2011. Antinociceptive effect of Lafoensia pacari A. St.-Hil. independent of anti-inflammatory activity of ellagic acid. Journal of NaturalMedicines 65, 448–454.

Pari, L., Sivasankari, R., 2008. Effect of ellagic acid on cyclosporine A-inducedoxidative damage in the liver of rats. Fundamental and Clinical Pharmacology22, 395–401.

Porfirio, Z., Melo-Filho, G.C., Alvino, V., Lima, M.R.F., Sant’Ana, A.E.G., 2009.Antibacterial activity of the hydro-alcoholic extracts of Lafoensia pacari SaintHil. against multiresistant bacterial strains from hospital source. RevistaBrasileira de Farmacognosia 19, 785–789.

Rogerio, A.P., Fontanari, C., Borducchi, E., Keller, A.C., Russo, M., Soares, E.G.,Albuquerque, D.A., Faccioli, L.H., 2008. Anti-inflammatory effects of Lafoensiapacari and ellagic acid in a murine model of asthma. European Journal ofPharmacolology 580, 262–270.

Rogerio, A.P., Fontanari, C., Melo, M.C., Ambrosio, S.R., de Souza, G.E., Pereira, P.S.,Franca, S.C., da Costa, F.B., Albuquerque, D.A., Faccioli, L.H., 2006. Anti-inflam-matory, analgesic and anti-oedematous effects of Lafoensia pacari extract andellagic acid. Journal of Pharmacy and Pharmacology 58 (1265-1273).

Rogerio, A.P., Sa-Nunes, A., Faccioli, L.H., 2010. The activity of medicinal plants andsecondary metabolites on eosinophilic inflammation. PharmacologicalResearch 62, 298–307.

Silva, I.F., Raimondi, M., Zacchino, S., Cechinel, V., Noldin, V.F., Rao, V.S., Lima, J.C.S.,Martins, D.T.O., 2010. Evaluation of the antifungal activity and mode of action ofLafoensia pacari A. St.-Hil., Lythraceae, stem-bark extracts, fractions and ellagicacid. Revista Brasileira de Farmacognosia 20, 422–428.

Solon, S., Lopes, L., de Sousa, P.T., Schmeda-Hirschmann, G., 2000. Free radicalscavenging activity of Lafoensia pacari. Journal of Ethnopharmacology 72,173–178.

Tamashiro Filho, P., Sikiru Olaitan, B., Tavares de Almeida, D.A., Lima, J.C., Marson-Ascencio, P.G., Donizeti Ascencio, S., Rios-Santos, F., Martins, D.T., 2012.Evaluation of antiulcer activity and mechanism of action of methanol stembark extract of Lafoensia pacari A. St.-Hil. (Lytraceae) in experimental animals.Journal of Ethnopharmacology 144, 497–505.

Umesalma, S., Sudhandiran, G., 2010. Differential inhibitory effects of the poly-phenol ellagic acid on inflammatory mediators NF-kappaB, iNOS, COX-2, TNF-alpha, and IL-6 in 1,2-dimethylhydrazine-induced rat colon carcinogenesis.Basic and Clinical Pharmacology and Toxicology 107, 650–655.

Umesalma, S., Sudhandiran, G., 2011. Ellagic acid prevents rat colon carcinogenesisinduced by 1, 2 dimethyl hydrazine through inhibition of AKT-phosphoinositide-3 kinase pathway. European Journal of Pharmacology 660,249–258.

Vanella, L., Di Giacomo, C., Acquaviva, R., Barbagallo, I., Cardile, V., Kim, D.H.,Abraham, N.G., Sorrenti, V., 2013. Apoptotic markers in a prostate cancer cellline: effect of ellagic acid. Oncology Reports 30, 2804–2810.

Zhang, T., Chen, H.S., Wang, L.F., Bai, M.H., Wang, Y.C., Jiang, X.F., Liu, M., 2014.Ellagic acid exerts anti-proliferation effects via modulation of TGF-beta/SMAD3signaling in MCF-7 breast cancer cells. Asian Pacific Journal of Cancer Preven-tion 15, 273–276.

D.B. da Silva Marcondes et al. / Journal of Ethnopharmacology 157 (2014) 243–250250