Next Article in Journal
Expression of SCD and FADS2 Is Lower in the Necrotic Core and Growing Tumor Area than in the Peritumoral Area of Glioblastoma Multiforme
Next Article in Special Issue
Ameliorative Effect of Gum Acacia on Hookah Smoke-Induced Testicular Impairment in Mice
Previous Article in Journal
Metabolic and Lipidomic Profiling of Vegetable Juices Fermented with Various Probiotics
Previous Article in Special Issue
Machaerium hirtum (Vell.) Stellfeld Alleviates Acute Pain and Inflammation: Potential Mechanisms of Action
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Brazilian Red Propolis: Extracts Production, Physicochemical Characterization, and Cytotoxicity Profile for Antitumor Activity

by
Felipe Mendes de Andrade de Carvalho
1,2,
Jaderson Kleveston Schneider
1,2,
Carla Viviane Freitas de Jesus
1,2,
Luciana Nalone de Andrade
3,
Ricardo Guimarães Amaral
3,
Jorge Maurício David
4,
Laíza Canielas Krause
1,2,
Patrícia Severino
1,2,5,
Cleide Mara Faria Soares
1,2,
Elina Caramão Bastos
1,2,
Francine Ferreira Padilha
1,2,
Silvana Vieira Flores Gomes
1,2,
Raffaele Capasso
6,
Antonello Santini
7,
Eliana Barbosa Souto
8,9,* and
Ricardo Luiz Cavalcanti de Albuquerque-Júnior
1,2,*
1
Tiradentes University (UNIT), Av. Murilo Dantas, 300, Aracaju 49010-390, Brazil
2
Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, Aracaju 49032-490, Brazil
3
Federal University of Sergipe (UFS), Avenida Marechal Rondon, São Cristovão 49100-000, Brazil
4
Federal University of Bahia (UFBA), Salvador/BA 40170-110, Brazil
5
Tiradentes Institute, 150 Mt Vernon St, Dorchester, MA 02125, USA
6
Department of Agricultural Sciences, University of Napoli Federico II, Via Università 100, 80055 Portici, Italy
7
Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
8
Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
9
CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
*
Authors to whom correspondence should be addressed.
Biomolecules 2020, 10(5), 726; https://doi.org/10.3390/biom10050726
Submission received: 20 March 2020 / Revised: 30 April 2020 / Accepted: 1 May 2020 / Published: 6 May 2020
(This article belongs to the Collection Pharmacology of Medicinal Plants)

Abstract

:
Brazilian red propolis has been proposed as a new source of compounds with cytotoxic activity. Red propolis is a resinous material of vegetal origin, synthesized from the bees of the Appis mellifera family, with recognized biological properties. To obtain actives of low polarity and high cytotoxic profile from red propolis, in this work, we proposed a new solvent accelerated extraction method. A complete 23 factorial design was carried out to evaluate the influence of the independent variables or factors (e.g., temperature, number of cycles, and extraction time) on the dependent variable or response (i.e., yield of production). The extracts were analyzed by gas chromatography coupled with mass spectrometry for the identification of chemical compounds. Gas chromatography analysis revealed the presence of hydrocarbons, alcohols, ketones, ethers, and terpenes, such as lupeol, lupenone, and lupeol acetate, in most of the obtained extracts. To evaluate the cytotoxicity profile of the obtained bioactives, the 3-(4,5-dimethyl-2-thiazole)-2,5-diphenyl-2-H-tetrazolium bromide colorimetric assay was performed in different tumor cell lines (HCT116 and PC3). The results show that the extract obtained from 70 °C and one cycle of extraction of 10 min exhibited the highest cytotoxic activity against the tested cell lines. The highest yield, however, did not indicate the highest cytotoxic activity, but the optimal extraction conditions were indeed dependent on the temperature (i.e., 70 °C).

1. Introduction

Cancer is a group of diseases characterized by disordered growth of cells that invade tissues and organs, with high morbidity and mortality, and represents one of the most fatal diseases worldwide [1]. About 21 million new cancers and 8.2 million cancer deaths worldwide are estimated until 2030 [2]. Surgery, radiotherapy, and chemotherapy are the most used approaches in the management and treatment of the disease [3]. Chemotherapy is commonly associated with the non-specific delivery of antineoplastic drugs capable of inhibiting mitosis, causing cell death and systemic toxicity. The drug targets are not only cancer cells but also healthy cells, thereby causing serious adverse side effects, e.g., diarrhea, hair loss, mucositis, immunosuppression, nausea, and vomiting, thus compromising the patients’ quality of life [4].
In developing and emerging countries, a large part of the population makes use of natural products in the treatment of health conditions, as these products are commonly related to a lower risk of inducing adverse side effects [5]. Propolis is a complex resinous mixture of vegetable origin, synthesized by bees from the collection of various natural substances. Depending on the flora at the collection site, propolis samples may vary in color and chemical composition [6]. A red variety of Brazilian propolis has been described in the last years, with the ethanolic extracts being enriched in isoflavones, mainly formononetin, daidzein, and biochanin A [7]. Studies have demonstrated that such hydroalcoholic and ethanolic extracts have potential antitumoral activity both in vitro [8,9] and in vivo [10,11,12,13,14,15].
In addition to the phenolic compounds often extracted with high-polar solvents, low-polar chemical constituents have also been described in samples of red propolis, including terpenes and others [6]. Some of these chemical compounds, such as methyl eugenol, a methylated ether previously reported as one the major constituents of the essential oil of red propolis, have been demonstrated to induce anticancer activity [16]. Furthermore, other substances of lower polarity present in samples of red propolis, such as sesquiterpenes, have also exhibited antitumor activity in vitro [17,18].
The solvent accelerated extraction (SAE) method is a fast process that uses solvent under high pressure to promote greater desorption of the analytes present in the matrix, offering less exposure to organic solvents while producing more selective extracts [19]. Pressure liquid extraction (PLE) has been successfully used to obtain compounds of low polarity from propolis samples [19,20]. Although high pressurized ethanol (in supercritical state) has been previously applied to extract chemical compounds from Brazilian red propolis [21], the use of PLE with low-polarity solvents (such as hexane) to extract chemical compounds with potential anticancer activity has not been studied so far. The aim of this study was to chemically characterize hexane extracts of Brazilian red propolis obtained using SAE and assess their cytotoxic activity against HCT116 and PC-3 cell lines derived from human colorectal carcinoma and prostate small cell carcinoma, respectively.

2. Materials and Methods

2.1. Collection and Processing of Red Propolis

The red propolis was obtained from the Santo Antônio apiary located in the municipality of Barra de Santo Antônio, Alagoas, Brazil, under coordinates 09°24′17” S 35°30′26”, in August 2017. The collected propolis samples were crushed in a grinder (Cadence, Piçarras, SC, Brazil) and then sieved (60 mesh) to obtain an adequate granulometry (approximately 0.250 mm), in order to increase the surface area and improve the homogenization of the starting material in the extraction process. The sample was then stored in a polyethylene container sealed and refrigerated between 2 and 8 °C.

2.2. Extraction by Pressurized Liquids

The extraction process was performed using a Dionex ASE™ 100 extractor (Dionex, Sunnyvale, CA, USA), with a 34-mL capacity stainless steel extraction chamber, a rinse bottle, and glass flasks with a capacity of 250 mL.

2.3. Full 23 Factorial Design Experiments

A complete 23 factorial design approach (Table 1) of three independent variables or factors (i.e., temperature, number of cycles, and extraction time) and three experimental levels (center point) was used to define the stage of preparation of the extract, totaling 11 trials. In this study, the levels were coded at the minimum (−1), central (0), and maximum (+1). The planning of Table 1 was performed, and the assay response was evaluated in terms of the extract yield (%).
For each extract, 10 g of the crude propolis were placed into the extraction chamber, hermetically sealed, and inserted into the equipment. The independent variables were adjusted in the extraction equipment, namely, a constant pressure of 1450 psi (100 bar) exerted by pneumatic pressurization by nitrogen (N2), flushing of 100%, purge of 60 s, and hexane as the solvent. Each extract was concentrated under reduced pressure on a rotary evaporator (RE801BW2, Yamato Scientific America Inc, Santa Clara, CA, USA) for 10 min at 55 °C. The obtained extract was then taken to a laminar flow oven for 72 h at a constant temperature of 45 °C. The dry extracts were passed through a thermogravimetry scale to determine the humidity and then stored at room temperature for further analysis. Experimental planning and analysis of the results were performed using Statistica Software 7.0 (Statsoft®, Tulsa, USA). The significance of the effects related to the studied variables was evaluated by analysis of variance (ANOVA) for each response and the effects were verified using p values less than 0.05 to indicate that the variable was significant within the experimental domain studied, with a confidence level of 95%.

2.4. Yield of Production

The yield of the extracted material was determined based on the percentage of mass obtained, using the initial mass of the propolis before the extraction and the material obtained after extraction and removal of the solvent as reference, according to the following equation:
Y i e l d   ( % ) = E M I M × 100 ,
where EM stands for the extract mass (g) and IM for the initial mass (g).

2.5. Gas Chromatography Coupled with Mass Spectrometry (GC/MS)

The extracts of red propolis were dissolved in 5 mL of distilled hexane. Samples were filtered on 0.45-μm nylon membranes (Merck KGaA, Darmstadt, Germany) prior to being injected into CG/qMS (QP2010 Plus, Shimadzu, Kyoto, Japan). Analyses were conducted on an Agilent capillary column coated with DB-5 (5% diphenyl and 95% dimethylpolysiloxane, 30 m × 0.25 mm ID, 0.25 µm film thickness) (Palo Alto, CA, USA). The flow rate of the carrier gas (He) was 1 mL/min. The injection mode was splitless in the automatic injector AOC20i, and the temperature of the injector, interface, and detector were 300 °C. The column temperature ramp was maintained at 40 °C for 1 min, and raised 3 °C/min to 300 °C (kept for 15 min). The analysis mode was scan mode (45 to 500 Da). All data were treated by the GCMS Solution software 2.6.1 (Shimadzu, Kyoto, Japan), using a signal/noise of 3. The compounds were tentatively identified using the spectral library database of the National Institute of Standards and Technology (NIST, Beta release STRBase 2.0). For the identification, the spectra comparison and detailed analysis of the MS spectra and position (retention time) in the chromatogram were used. Only the peaks where the similarity with the libraries was higher than 80% were identified, together with confirmation through the linear temperature programmed retention index (LTPRI) (considering a difference of up to 20 units).
A C6-C30 mixture of n-alkanes (1 μL) at the concentration of 10 mg/L was injected into the GC/qMS system under the same conditions used for the samples to calculate the LTPRI. We used the retention time of each standard n-alkane to carry out this analysis, since this is a quantitative parameter. The retention indices were calculated for each compound in the sample and the values were compared to those from the literature for the same column (DB-5 or similar).

2.6. In Vitro Cytotoxicity Analysis

To determine the cytotoxic effect of the obtained extracts against tumor cell lines, the method of the 3-(4,5-dimethyl-2-thiazole)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) (Sigma Aldrich, Brazil) salt was used [22]. For all experiments, in 96-well plates, tumor cell lines were plated (100 μL/well) at concentrations of 0.7 × 105 cells/mL HCT116 (colon cancer) and 1 × 105 cells/mL of PC3 cells/mL (prostate cancer). All cells were obtained from Fundação Oswaldo Cruz (Fiocruz, Brazil). The cell lines were selected based on the worldwide incidence of these two types of cancer. After 24 h, the extracts were dissolved in 0.3% dimethyl sulfoxide (DMSO) and added to each well in a single concentration of 50 μg/mL. The experiment was performed three independent times in triplicate, using doxorubicin 0.25 μg/mL and 0.3% DMSO as the positive and negative control, respectively. The plates were incubated for 72 h in a 5% CO2 oven at 37 °C. After incubation, the plates were centrifuged (15 g/15 min) at 4 °C and the supernatants were removed. Subsequently, 150 μL of the MTT solution (0.5 mg/mL) were added to the wells and the plates were incubated for 3 h. Following this period, the plates were again centrifuged (30 g/10 min) at 4 °C, the supernatants were discarded, and the precipitate was resuspended in 150 μL of pure sterile DMSO. For the quantification of formazan crystals formed by viable cells, the absorbance was read using a multi-plate reader (DTX 880 Multimode Detector, Beckman Coulter Inc., Packard, ON, Canada) at 595 nm. All values were converted to the percent inhibition of cell growth (% ICG) by the following equation:
I C G % = 100 [ T C N ] × 100 ,
where T is the absorbance of the red propolis extract and CN is the absorbance of the negative control.
For each tested cell line, the results were analyzed using an ICG% scale as follows: (i) Samples of low activity have ICG ≤ 50%, (ii) samples with moderate activity have 50% < ICG < 75%, and (iii) samples of high activity have ICG ≥ 75%. To determine the minimum inhibitory concentration (IC50, capable of causing 50% of its maximal effect), the same protocol as described for the determination of ICG%, was performed in the same cell lines but by varying the extract concentration (0.39–50 μg/mL) only. The percentage of cell death was analyzed according to the mean ± standard error. All the assays were carried out in triplicate. The percentage of inhibition x log of the concentration was recorded and their IC50 and their respective confidence intervals (IC 95%) were recorded from non-linear regression. The analyzes and graphs were elaborated using the GraphPad Prism version 7.0 software (San Diego, CA, USA).

2.7. Statistical Analysis

The quantitative variables, expressed as the mean ± standard error of the mean, were subjected to the test of homoscedasticity and normal distribution of values. For the normality test, the Shapiro–Wilk test was applied, whereas the Bartlett (assuming that the variable has a normal distribution) and Levene (assuming that the data have a continuous distribution) tests were applied for homoscedasticity analysis. Data with a normal distribution and homoscedasticity were compared using the ANOVA test, followed by post hoc Tukey’s multiple comparisons test. Data that did not obey these assumptions (normal distribution and/or homoscedasticity) were compared using the Kruskal–Wallis test with post hoc Dunn’s multiple comparisons test. Differences between means were considered significant when p values were lower than 0.05.

3. Results and Discussion

The extraction processes of chemical compounds from propolis samples commonly use ethanol, water, hexane, ethyl acetate, or chloroform as solvents. However, these conventional extraction processes have some drawbacks, such as a strong residual taste, risk of harmful effects to the environment, poor quality of the obtained extract, and the need for a long extraction time. High pressure has been applied in the extraction, fractionation, refinement, and deodorization of natural sample matrices at the laboratory scale and industrial scale, with supercritical CO2 and ethanol (usually used as a co-solvent) being the most frequently used solvents [23].
In this work, a full 23 factorial design was applied for the optimization of the extraction method of substances of low polarity from red propolis, aiming to be fast, of low expenditures, and high yield. This model allows a combination of three variables and one central point with three samples and is frequently used in optimization studies [24,25,26,27,28]. However, studies applying factorial design for the analysis of extractive processes of propolis samples using pressurized liquids have not yet been reported in the literature. The results of the comparisons between the variables tested to obtain the best yields are shown in Figure 1. The surface responses show that the temperature, cycles, and time significantly influenced the yields (p < 0.05) but not the combination of variables (p > 0.05).
It has been reported that different extraction conditions can generate different responses with respect to their yield and their chemical composition. Thus, yield can be considered an important response variable to be evaluated in the research of different methods of natural product extraction [29]. For the extraction of low-polarity compounds, low-polarity solvents are also used because of the higher affinity promoted by the interaction between the polarities of the solute and the solvent, which favors the dragging of similar substances. Among the solvents with these chemical characteristics, hexane has been selected. This solvent has been frequently used for extractions of compounds of low polarity [30]. In the current study, there was large variability in the yield of the extractive processes within the different conditions tested, varying from 18.2% to 64.63%. The purpose of the experimental planning used in this study was to develop the extractive process based on yield, with optimal conditions assumed as those that provided values equal to or greater than 11%. Due to the lack of similar studies in the literature reporting extracts of low polarity with red propolis, this percentage (11%) was based on the reference value suggested by the Brazilian Ministry of Agriculture for hydroethanolic and ethanolic extracts of green propolis samples [31].
As shown in Figure 2, the yield obtained in all the samples ranged from 18.34% to 64.63%. The yields obtained in the current study were considered satisfactory in comparison to those reported by Biscaia and Ferreira [20] (10.79%), Zordi et al. [32] (24.8%), and Machado et al. [33] (9.16%), using supercritical fluid extraction, with CO2 and ethanol as the co-solvent. However, none of the extraction procedures described above were performed within the conditions tested in the current study, which makes it difficult to establish a proper comparison and deeper analysis of these data.
In the present study, the highest yields were obtained at 70 °C. These results can be explained by the resinous nature of propolis. When this heterogeneous substance is subjected to high temperature and pressure, the material of low polarity can be solubilized and dragged together with the compounds of greater affinity with the low-polarity solvent [34]. Herrero et al. used an experimental design for antioxidant extraction from Spirulina platensis microalgae by ASE, using hexane, petroleum ether, ethanol, and water [35]. The authors reported that although a similar antioxidant capacity could be seen for the extracts obtained using the organic solvents, a slightly higher antioxidant profile was recorded with hexane-based extracts. This was attributed to the capacity of hexane to extract a higher amount of non-polar compounds (carotenoids, among others) that can be extracted using hexane, which increase the antioxidant activity.
Gas chromatography is often used in the characterization of the chemical compounds of natural products. This technique is usually employed to separate compounds that have low polarity and high volatility. These characteristics, associated with the availability of a large library of mass spectra information of each substance, allow the identification of the chemical compounds present in the samples [16,36]. Due to the advantages of this separation technique, and the low polarity of the extracts, gas chromatography coupled with mass spectrometry (GC/MS) was used in this study. Chromatograms with similar profiles were obtained for all extracts, regardless of the tested conditions. A total of 46 compounds were identified and are shown in Table 2. The identification was confirmed from the comparison with the retention index of the peaks, using the NIST Chemistry WebBook, SRD 69 as a reference.
A wide range of chemical compounds with no previously reported biological activity were found in the extracts of red propolis. The presence of aromatic hydrocarbons may be related to the contamination of the water and soil of the apicultural grass (collection sites of the bees), mainly by fuels, since these compounds are often found around fuel stations and account for a considerable part of air pollution [37]. On the other hand, unsaturated hydrocarbons, such as heptacos-1-ene and nonacos-1-ene, belonging to the family of alcohols, are often found in products derived from the fermentation of sugar in plants [38]. Regarding the chemical compounds belonging to the class of ethers, only methyl eugenol has been previously reported to show biological activity, as a promising agent against hemorrhoids, rheumatism, contusions, and seizures. Two compounds belonging to the class of ketones, 2-(3H)-furanone, 5-dodecyldihydro-, which do not present biological activity, and acetophenone, which presents anti-inflammatory and antioxidant activities [5]. Additionally, according to Favier et al. [39], acetophenone is a phenolic compound that represents a secondary metabolite often found in plants subjected to environmental stress.
Among the chemical compounds found in RPE presenting well-established biological activity, there were the terpenes lupeol (C30H50O), lupeol acetate (C32H52O2), and lupenone (C30H48O). Triterpenoids have been demonstrated to exert cytotoxic effects on human hepatocellular carcinoma (Hep-G2) and melanoma (MEL-2) cell lines by the inhibition of topoisomerase II and farnesyl transferase protein [40]. Moreover, triterpenes have been reported to reduce the number of aberrant cells, micronucleus DNA, and morphological signs of cytotoxicity (e.g., pyknosis, caryolysis, and karyorrhexis) in a benzo [α] pyrene-induced clastogenicity model [41]. Regarding in vivo experimental models, a lupeol-associated antitumor effect on epidermal tumors induced by TPA (12-tetradecanoylphorbol-13-acetate) has been previously reported, likely due to modulation of the NF-κB and PI3K/Akt pathways [42]. Furthermore, lupeol applied on the dorsum of rats displayed chemopreventive effects on the carcinogenesis induced by 7,12-dimethylbenz (a) anthracene (DMBA) [43]. These data suggest that substances presenting those compounds in their chemical composition, such as the RPE obtained in the current study, might display cytotoxicity or even antitumor activity.
For screening purposes, the cytotoxic activity of a given product against tumor cell lines, either a plant extract or an isolated molecule, is considered moderate when the viability index is between 25% and 50% and strong when this index is less than 25%, whereas cell viability above 50% means no consistent cytotoxic activity. In the current study, the cytotoxic potential of the 11 samples was assessed using two different tumor cell lines, colon cancer (HCT116) and prostate (PC3). The choice for these cell lines was based on the high frequency of colon and prostate cancers worldwide [44]. As demonstrated in Figure 3, most samples showed at least moderate cytotoxic activity against HCT116 cells, except for sample 8, and against PC3, excluding samples 1 and 8. However, only sample 5 showed strong cytotoxic activity against both tumor cell lines.
The cytotoxic activities of the RPE might be related to the presence of triterpenes in their chemical composition [45]. Supporting this hypothesis, different extracts rich in lupeol and derivatives have been demonstrated to exert cytotoxic activity against several malignant tumor cell lines, including NCI-H292 (lung mucoepidermoid carcinoma), MCF-7 (breast adenocarcinoma), and HEp-2 (squamous cell carcinoma of larynx) [46]. Besides, the use of lupeol has been shown to have potent cytotoxic activity against MCF-7 (breast adenocarcinoma) cells [47], U87MG (glioblastoma), CEM/ADR5000 (leukemia), and HCT116 (colon adenocarcinoma) [48]. On the other hand, lupeol acetate has cytotoxic activity against lung carcinoma (A549), colon adenocarcinoma (DLD-1), breast (MCF-7, MDA-MB468), pancreas (MIAPaCa-2), prostate (C-1) and ovary (SK-OV-3), cervical carcinoma (HeLa), hepatocellular carcinoma (PLC/PRF/5), and renal carcinoma (786-0, Caki-1) [49]. In addition, extracts containing lupenone have presented cytotoxic activity against MCF-7, HT-29 (colon) and HeLa cells (cervix) [50]. Thus, the presence of triterpenes, such as lupeol, lupenone, and lupeol acetate, in the RPE analyzed in the current study, although in apparently low concentrations, the results see to support the moderate cytotoxic activity of the majority of the samples. Furthermore, sample 5 demonstrated stronger cytotoxicity against HCT116 and PC3 cells, which could be explained by the presence of both lupeol and lupeol acetate in the chemical composition of the extract.
As sample 5 presented the strongest cytotoxic effects and HCT116 cells were the most sensitive to RPE, they were both selected to the IC 50 assessment assay (mean inhibitory concentration capable of causing 50% of maximal effect), as demonstrated in Figure 4. According to the guidelines for testing for cytotoxicity with plant-derived extracts (Gad-Shayne, 2009), IC 50 < 10 μg/mL is considered to be “very toxic”; IC 50 between 10 and 100 μg/mL would be “potentially toxic”; IC 50 100–1000 μg/mL “potentially harmful”; and IC50 > 1000 μg/mL “potentially non-toxic”. As the IC 50 value obtained in the current study was 31.53 μg/mL, sample 5 could be considered “potentially toxic”, which makes it a target for alternative treatments against cancer, either as chemotherapeutic or chemopreventive agents.
It is possible to estimate some hypotheses about the pathophysiological mechanisms that might be involved in the cytotoxic activity. Previous studies have asserted that the anticancer potential of triterpenes could be dependent on the generation of reactive oxygen species (ROS), which would later lead to mitochondrial membrane rupture and potential apoptosis of tumor cells [49]. Furthermore, triterpenes, such as lupeol, are able to downregulate Bcl-2 and Bcl-xL protein expression, and contribute to the induction of MCF-7 cell death by activating mitochondrial pathway apoptosis [47]. In addition to the activation of the intrinsic pathway of apoptosis by regulating the Bcl-2 family genes’ expression, the involvement of upregulation of proapoptotic proteins, such as Smac/DIABLO, can also be postulated. Smac/DIABLO is a protein that promotes the displacement of the inhibitor of apoptosis protein (XIAP) from caspase 9, thus neutralizing the ability of XIAP to suppress the activity of the effector caspase within the apoptosome complex [51]. In fact, a cytotoxic effect of methanol extract of Ficus Deltoidea (FD2c), rich in phytochemical lupeol, against PC-3 and LNCaP prostate cancer cells, with an IC50 of 29 and 23 ug/mL, respectively, mediated by downregulation of Bcl-2 and upregulation of Smac/DIABLO, and consequent activation of caspases 3, and 7, involved in the intrinsic pathway of apoptosis cascade, has been previously reported [52]. As these IC values are remarkably similar to the IC50 of RPE sample 5 against HCT116 cells (31.53 ug/mL), we speculate whether these two protein families might be involved in the pathogenesis of cytotoxic activity exerted by RPE. However, further studies are still necessary to clarify the biochemical and molecular mechanisms underlying the cytotoxic effects of RPE on tumor cell lines. Besides, in order to be site specific for tumor cells, the loading of such chemotherapeutic and/or chemopreventive agents or their extracts into nanoparticles has been proposed to minimize the risk of cytotoxic events in healthy cells [53,54]. Additional screening tests, such as genotoxicity [55,56], oxidative stress [57], and apoptosis [58], have been proposed.

4. Conclusions

In summary, the hexane-based extracts obtained in this study showed very a similar chemical composition, represented by saturated and unsaturated aromatic hydrocarbons, ketones, alcohols, ethers, and terpenes. Most of the extracts exhibited moderate cytotoxic activity. In addition, the extract obtained at 70 °C and using one cycle of 10 min presented the strongest cytotoxic activity against HCT116 and PC3 tumor cell lines, and such activity could be related to the presence of terpenes, such as lupeol and lupeol acetate, in its chemical composition.

Author Contributions

F.M.d.A.d.C., J.K.S., C.V.F.d.J. and L.N.d.A. have contributed for the conceptualization, methodology, software and data validation. R.G.A., J.M.D., L.C.K., P.S., C.M.F.S. have contributed for formal analysis, investigation, resources and data curation. F.M.d.A.d.C., E.C.B., F.F.P., S.V.F.G., R.C., A.S., E.B.S. and R.L.C.d.A.-J. have participated in the writing of the review, editing and visualization. P.S., E.B.S. and R.L.C.d.A.-J. have contributed for the supervision, project administration and funding acquisition. All authors have made a substantial contribution to this work and All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Coordination for Improvement of Higher Education Personnel (CAPES) and by the Foundation for Support to Research and Technological Innovation of Sergipe (FAPITEC). EBS acknowledges the sponsorship of the projects M-ERA-NET-0004/2015-PAIRED and UIDB/04469/2020 (strategic fund), received support from the Portuguese Science and Technology Foundation, Ministry of Science and Education (FCT/MEC) through national funds, and was co-financed by FEDER, under the Partnership Agreement PT2020.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Laryionava, K.; Winkler, E.C. Patients’ preferences in non-curable cancer disease. J. Oncol. Res. Treat. 2019, 42, 31–34. [Google Scholar] [CrossRef] [PubMed]
  2. Zaidi, S.A. Molecular imprinting prevents environmental contamination and body toxicity from anticancer drugs: An update. J. Crit. Rev. Anal. Chem. 2019, 49, 324–335. [Google Scholar] [CrossRef] [PubMed]
  3. Schildmann, J. Value and values in cancer care. Assessing the benefit of treatment in patients with advanced cancer. J. Oncol. Res. Treat. 2019, 42, 35–40. [Google Scholar] [CrossRef] [PubMed]
  4. Van Eenbergen, M.C.; van den Hurk, C.; Mols, F.; van de Poll-Franse, L.V. Usability of an online application for reporting the burden of side effects in cancer patients. J. Supportive Care Cancer 2019, 1–9. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, N.H.; Li, W.; Zhong, Y.L.; Niu, Q.W.; Li, Y.Y.; Zhang, Y.B.; Li, M.M.; Li, Y.L.; Wang, G.C. New Acetophenone Derivatives from Acronychia oligophlebia and Their Anti-inflammatory and Antioxidant Activities. J. Chem. Biodivers. 2018, 15, e18000080. [Google Scholar] [CrossRef]
  6. Devequi-Nunes, D.; Machado, B.A.S.; Barreto, G.A.; Reboucas Silva, J.; da Silva, D.F.; da Rocha, J.L.C.; Brandao, H.N.; Borges, V.M.; Umsza-Guez, M.A. Chemical characterization and biological activity of six different extracts of propolis through conventional methods and supercritical extraction. PLoS ONE 2018, 13, e0207676. [Google Scholar] [CrossRef]
  7. Rufatto, L.C.; Luchtenberg, P.; Garcia, C.; Thomassigny, C.; Bouttier, S.; Henriques, J.A.P.; Roesch-Ely, M.; Dumas, F.; Moura, S. Brazilian red propolis: Chemical composition and antibacterial activity determined using bioguided fractionation. J. Microbiol. Res. 2018, 214, 74–82. [Google Scholar] [CrossRef]
  8. Frozza, C.; Santos, D.A.; Rufatto, L.C.; Minetto, L.; Scariot, F.J.; Echeverrigaray, S.; Pich, C.T.; Moura, S.; Padilha, F.F.; Borsuk, S.; et al. Antitumor activity of Brazilian red propolis fractions against Hep-2 cancer cell line. Biomed. Pharmacother. Biomed. Pharmacother. 2017, 91, 951–963. [Google Scholar] [CrossRef]
  9. Silva, F.R.G.; Matias, T.M.S.; Souza, L.I.O.; Matos-Rocha, T.J.; Fonseca, S.A.; Mousinho, K.C.; Santos, A.F. Phytochemical screening and in vitro antibacterial, antifungal, antioxidant and antitumor activities of the red propolis Alagoas. Braz. J. Biol. Rev. Brasleira De Biol. 2019, 79, 452–459. [Google Scholar] [CrossRef]
  10. Pinheiro, K.S.; Ribeiro, D.R.; Alves, A.V.; Pereira-Filho, R.N.; Oliveira, C.R.; Lima, S.O.; Reis, F.P.; Cardoso, J.C.; Albuquerque-Junior, R.L. Modulatory activity of Brazilian red propolis on chemically induced dermal carcinogenesis. Acta Cir Bras 2014, 29, 111–117. [Google Scholar] [CrossRef]
  11. Ribeiro, D.R.; Alves, A.V.; dos Santos, E.P.; Padilha, F.F.; Gomes, M.Z.; Rabelo, A.S.; Cardoso, J.C.; Massarioli, A.P.; de Alencar, S.M.; de Albuquerque-Junior, R.L. Inhibition of DMBA-induced Oral Squamous Cells Carcinoma Growth by Brazilian Red Propolis in Rodent Model. Basic Clin. Pharm. Toxicol. 2015, 117, 85–95. [Google Scholar] [CrossRef] [PubMed]
  12. Lucarini, M.; Durazzo, A.; Kiefer, J.; Santini, A.; Lombardi-Boccia, G.; Souto, E.B.; Romani, A.; Lampe, A.; Ferrari Nicoli, S.; Gabrielli, P.; et al. Grape Seeds: Chromatographic Profile of Fatty Acids and Phenolic Compounds and Qualitative Analysis by FTIR-ATR Spectroscopy. Foods 2019, 9, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Durazzo, A.; Lucarini, M.; Novellino, E.; Souto, E.B.; Daliu, P.; Santini, A. Abelmoschus esculentus (L.): Bioactive Components’ Beneficial Properties-Focused on Antidiabetic Role-For Sustainable Health Applications. Molecules 2018, 24, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cefali, L.C.; Ataide, J.A.; Fernandes, A.R.; Sousa, I.M.O.; Goncalves, F.; Eberlin, S.; Davila, J.L.; Jozala, A.F.; Chaud, M.V.; Sanchez-Lopez, E.; et al. Flavonoid-Enriched Plant-Extract-Loaded Emulsion: A Novel Phytocosmetic Sunscreen Formulation with Antioxidant Properties. Antioxidants 2019, 8, 443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cefali, L.C.; Ataide, J.A.; Fernandes, A.R.; Sanchez-Lopez, E.; Sousa, I.M.O.; Figueiredo, M.C.; Ruiz, A.; Foglio, M.A.; Mazzola, P.G.; Souto, E.B. Evaluation of In Vitro Solar Protection Factor (SPF), Antioxidant Activity, and Cell Viability of Mixed Vegetable Extracts from Dirmophandra mollis Benth, Ginkgo biloba L., Ruta graveolens L., and Vitis vinifera L. Plants 2019, 8, 453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sena-Lopes, A.; Bezerra, F.S.B.; das Neves, R.N.; de Pinho, R.B.; Silva, M.T.O.; Savegnago, L.; Collares, T.; Seixas, F.; Begnini, K.; Henriques, J.A.P.; et al. Chemical composition, immunostimulatory, cytotoxic and antiparasitic activities of the essential oil from Brazilian red propolis. PLoS ONE 2018, 13, e0191797. [Google Scholar] [CrossRef] [PubMed]
  17. Babaei, G.; Aliarab, A.; Abroon, S.; Rasmi, Y.; Aziz, S.G. Application of sesquiterpene lactone: A new promising way for cancer therapy based on anticancer activity. Biomed. Pharmacother. Biomed. Pharmacother. 2018, 106, 239–246. [Google Scholar] [CrossRef]
  18. Xu, D.D.; Yan, Y.; Jiang, C.X.; Liang, J.J.; Li, H.F.; Wu, Q.X.; Zhu, Y. Sesquiterpenes and diterpenes with cytotoxic activities from the aerial parts of Carpesium humile. Fitoterapia 2018, 128, 50–56. [Google Scholar] [CrossRef]
  19. Monroy, Y.M.; Rodrigues, R.A.; Rodrigues, M.V.; Cabral, F.A. Fractionation of ethanolic and hydroalcoholic extracts of green propolis using supercritical carbon dioxide as an anti-solvent to obtain artepillin rich-extract. J. Supercrit. Fluids 2018, 138, 167–173. [Google Scholar] [CrossRef]
  20. Biscaia, D.; Ferreira, S.R. Propolis extracts obtained by low pressure methods and supercritical fluid extraction. J. Supercrit. Fluids 2009, 51, 17–23. [Google Scholar] [CrossRef]
  21. Saito, É.; Sacoda, P.; Paviani, L.C.; Paula, J.T.; Cabral, F.A. Conventional and supercritical extraction of phenolic compounds from Brazilian red and green propolis. Sep. Sci. Technol. 2020, 1–8. [Google Scholar] [CrossRef]
  22. Barbosa, T.C.; Nascimento, L.E.D.; Bani, C.; Almeida, T.; Nery, M.; Santos, R.S.; Menezes, L.R.O.; Zielinska, A.; Fernandes, A.R.; Cardoso, J.C.; et al. Development, Cytotoxicity and Eye Irritation Profile of a New Sunscreen Formulation Based on Benzophenone-3-poly(epsilon-caprolactone) Nanocapsules. Toxics 2019, 7, 51. [Google Scholar] [CrossRef] [Green Version]
  23. Idrus, N.F.M.; Yian, L.N.; Idham, Z.; Aris, N.A.; Putra, N.R.; Aziz, A.H.A.; Yunus, M.A.C. Mini review: Application of supercritical carbon dioxide in extraction of propolis extract. J. Malays. J. Fundam. Appl. Sci. 2018, 14, 387–396. [Google Scholar] [CrossRef]
  24. Mullane, K.; Curtis, M.J.; Williams, M. Experimental planning and execution. In Research in the Biomedical Sciences; Elsevier: Amsterdam, The Netherlands, 2018; pp. 67–106. [Google Scholar]
  25. Campos, J.R.; Fernandes, A.R.; Sousa, R.; Fangueiro, J.F.; Boonme, P.; Garcia, M.L.; Silva, A.M.; Naveros, B.C.; Souto, E.B. Optimization of nimesulide-loaded solid lipid nanoparticles (SLN) by factorial design, release profile and cytotoxicity in human Colon adenocarcinoma cell line. Pharm. Dev. Technol. 2019, 24, 616–622. [Google Scholar] [CrossRef] [PubMed]
  26. Rigon, R.B.; Goncalez, M.L.; Severino, P.; Alves, D.A.; Santana, M.H.A.; Souto, E.B.; Chorilli, M. Solid lipid nanoparticles optimized by 2(2) factorial design for skin administration: Cytotoxicity in NIH3T3 fibroblasts. Colloids Surf. B Biointerfaces 2018, 171, 501–505. [Google Scholar] [CrossRef] [Green Version]
  27. Severino, P.; Santana, M.H.; Souto, E.B. Optimizing SLN and NLC by 2(2) full factorial design: Effect of homogenization technique. Mater. Sci. Eng. C Mater. Biol. Appl. 2012, 32, 1375–1379. [Google Scholar] [CrossRef]
  28. Zielinska, A.; Ferreira, N.R.; Durazzo, A.; Lucarini, M.; Cicero, N.; Mamouni, S.E.; Silva, A.M.; Nowak, I.; Santini, A.; Souto, E.B. Development and Optimization of Alpha-Pinene-Loaded Solid Lipid Nanoparticles (SLN) Using Experimental Factorial Design and Dispersion Analysis. Molecules 2019, 24, 2683. [Google Scholar] [CrossRef] [Green Version]
  29. Yahyaoui, M.; Ghazouani, N.; Saoudi, S.; Sifaoui, I.; Chammem, N.; Abderrabba, M. Experimental design methodologyapplication in the optimization of phytochemical compoundsextraction and antioxidant activity of Thymelaea hirsuta L. extracts. J. Mater. Environ. Sci. 2018, 9, 1551–1561. [Google Scholar]
  30. Jiang, Z.; Kempinski, C.; Chappell, J. Extraction and Analysis of Terpenes/Terpenoids. Curr. Protoc. Plant Biol. 2016, 1, 345–358. [Google Scholar] [CrossRef] [Green Version]
  31. De Almeida, M.F., Mel, Instrução Normativa Nº11, Ministerio da Agricultura e Abastecimento do Brasil, de 20 de Outubro de 2000. Available online: https://bibliodigital.unijui.edu.br:8443/xmlui/handle/123456789/5029 (accessed on 8 October 2019).
  32. De Zordi, N.; Cortesi, A.; Kikic, I.; Moneghini, M.; Solinas, D.; Innocenti, G.; Portolan, A.; Baratto, G.; Dall’Acqua, S. The supercritical carbon dioxide extraction of polyphenols from propolis: A central composite design approach. J. Supercrit. Fluids 2014, 95, 491–498. [Google Scholar] [CrossRef]
  33. Machado, B.A.; Silva, R.P.; Barreto Gde, A.; Costa, S.S.; Silva, D.F.; Brandao, H.N.; Rocha, J.L.; Dellagostin, O.A.; Henriques, J.A.; Umsza-Guez, M.A.; et al. Chemical Composition and Biological Activity of Extracts Obtained by Supercritical Extraction and Ethanolic Extraction of Brown, Green and Red Propolis Derived from Different Geographic Regions in Brazil. PLoS ONE 2016, 11, e0145954. [Google Scholar] [CrossRef] [PubMed]
  34. Kubiliene, L.; Laugaliene, V.; Pavilonis, A.; Maruska, A.; Majiene, D.; Barcauskaite, K.; Kubilius, R.; Kasparaviciene, G.; Savickas, A. Alternative preparation of propolis extracts: Comparison of their composition and biological activities. BMC Compl. Altern. Med. 2015, 15, 156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Herrero, M.; Martín-Álvarez, P.J.; Señoráns, F.J.; Cifuentes, A.; Ibáñez, E. Optimization of accelerated solvent extraction of antioxidants from Spirulina platensis microalga. Food Chem. 2005, 93, 417–423. [Google Scholar] [CrossRef] [Green Version]
  36. Pellati, F.; Prencipe, F.P.; Benvenuti, S. Headspace solid-phase microextraction-gas chromatography-mass spectrometry characterization of propolis volatile compounds. J. Pharm. Biomed. Anal. 2013, 84, 103–111. [Google Scholar] [CrossRef] [PubMed]
  37. Mosaed, H.P.; Sobhanardakani, S.; Merrikhpour, H.; Farmany, A.; Cheraghi, M.; Ashorlo, S. The effect of urban fuel stations on soil contamination with petroleum hydrocarbons. Iranian J. Toxicol. 2015, 9. [Google Scholar]
  38. Alcarde, A.R.; Souza, P.A.d.; Belluco, A.E.d.S. Chemical profile of sugarcane spirits produced by double distillation methodologies in rectifying still. J. Food Sci. Technol. 2011, 31, 355–360. [Google Scholar] [CrossRef] [Green Version]
  39. Favier, L.; Tonn, C.; Guerreiro, E.; Rotelli, A.; Pelzer, L. Anti-inflammatory activity of acetophenones from Ophryosporus axilliflorus. Planta Med. 1998, 64, 657–659. [Google Scholar] [CrossRef]
  40. Setzer, W.N.; Setzer, M.C. Plant-derived triterpenoids as potential antineoplastic agents. Mini Rev. Med. Chem. 2003, 3, 540–556. [Google Scholar] [CrossRef]
  41. Gallo, M.B.C.; Sarachine, J. Biological activities of lupeol. Int. J. Res. Pharm. Biomed. Sci. 2009, 3, 46–66. [Google Scholar]
  42. Saleem, M.; Afaq, F.; Adhami, V.M.; Mukhtar, H. Lupeol modulates NF-kappaB and PI3K/Akt pathways and inhibits skin cancer in CD-1 mice. Oncogene 2004, 23, 5203–5214. [Google Scholar] [CrossRef] [Green Version]
  43. Nigam, N.; Prasad, S.; Shukla, Y. Preventive effects of lupeol on DMBA induced DNA alkylation damage in mouse skin. Food Chem. Toxicol. 2007, 45, 2331–2335. [Google Scholar] [CrossRef] [PubMed]
  44. Culp, M.B.; Soerjomataram, I.; Efstathiou, J.A.; Bray, F.; Jemal, A. Recent Global Patterns in Prostate Cancer Incidence and Mortality Rates. Eur. Urol. 2020, 77, 38–52. [Google Scholar] [CrossRef] [PubMed]
  45. Chudzik, M.; Korzonek-Szlacheta, I.; Krol, W. Triterpenes as potentially cytotoxic compounds. Molecules 2015, 20, 1610–1625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Bastos, I.; Oliveira, T.B.; Rodrigues, M.D.; Militao, G.C.G.; Silva, T.G.D.; Turatti, I.C.C.; Lopes, N.P.; Melo, S.J. Use of GC/MS to identify chemical constituents and cytotoxic activity of the leaves of Phoradendron mucronatum and Phoradendron microphyllum (Viscaceae). An. Da Acad. Bras. De Cienc. 2017, 89, 991–1001. [Google Scholar] [CrossRef] [Green Version]
  47. Pitchai, D.; Roy, A.; Ignatius, C. In vitro evaluation of anticancer potentials of lupeol isolated from Elephantopus scaber L. on MCF-7 cell line. J. Adv. Pharm. Technol. Res. 2014, 5, 179–184. [Google Scholar] [CrossRef]
  48. Nyaboke, H.O.; Moraa, M.; Omosa, L.K.; Mbaveng, A.T.; Vaderament-Alexe, N.-N.; Masila, V.; Okemwa, E.; Heydenreich, M.; Efferth, T.; Kuete, V. Cytotoxicity of lupeol from the stem bark of Zanthoxylum gilletii against multi-factorial drug resistant cancer cell lines. Invest. Med.l Chem. Pharmacol. 2018, 1, 10. [Google Scholar]
  49. Mishra, T.; Arya, R.K.; Meena, S.; Joshi, P.; Pal, M.; Meena, B.; Upreti, D.K.; Rana, T.S.; Datta, D. Isolation, Characterization and Anticancer Potential of Cytotoxic Triterpenes from Betula utilis Bark. PLoS ONE 2016, 11, e0159430. [Google Scholar] [CrossRef]
  50. Ahmad, S.; Sukari, M.A.; Ismail, N.; Ismail, I.S.; Abdul, A.B.; Abu Bakar, M.F.; Kifli, N.; Ee, G.C. Phytochemicals from Mangifera pajang Kosterm and their biological activities. BMC Compl. Altern. Med. 2015, 15, 83. [Google Scholar] [CrossRef] [Green Version]
  51. Vucic, D.; Deshayes, K.; Ackerly, H.; Pisabarro, M.T.; Kadkhodayan, S.; Fairbrother, W.J.; Dixit, V.M. SMAC negatively regulates the anti-apoptotic activity of melanoma inhibitor of apoptosis (ML-IAP). J. Biol. Chem. 2002, 277, 12275–12279. [Google Scholar] [CrossRef] [Green Version]
  52. Hanafi, M.M.M.; Afzan, A.; Yaakob, H.; Aziz, R.; Sarmidi, M.R.; Wolfender, J.L.; Prieto, J.M. In Vitro Pro-apoptotic and Anti-migratory Effects of Ficus deltoidea L. Plant Extracts on the Human Prostate Cancer Cell Lines PC3. Front. Pharm. 2017, 8, 895. [Google Scholar] [CrossRef] [Green Version]
  53. Souto, E.B.; Zielinska, A.; Souto, S.B.; Durazzo, A.; Lucarini, M.; Santini, A.; Silva, A.M.; Atanasov, A.G.; Marques, C.; Andrade, L.N.; et al. (+)-Limonene 1,2-epoxide-loaded SLN: Evaluation of drug release, antioxidant activity and cytotoxicity in HaCaT cell line. Int. J. Mol. Sci. 2020, 21, 1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Souto, E.B.; Souto, S.B.; Zielinska, A.; Durazzo, A.; Lucarini, M.; Santini, A.; Horbańczuk, O.K.; Atanasov, A.G.; Marques, C.; Andrade, L.N.; et al. Perillaldehyde 1,2-epoxide loaded SLN-tailored mAb: Production, physicochemical characterization and in vitro cytotoxicity profile in MCF-7 cell lines. Pharmaceutics 2020, 12, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Souto, E.B.; Campos, J.R.; Da Ana, R.; Martins-Gomes, C.; Silva, A.M.; Souto, S.B.; Lucarini, M.; Durazzo, A.; Santini, A. Ocular Cell Lines and Genotoxicity Assessment. Int. J. Env. Res. Public Health 2020, 17, 46. [Google Scholar] [CrossRef] [Green Version]
  56. Doktorovova, S.; Silva, A.M.; Gaivao, I.; Souto, E.B.; Teixeira, J.P.; Martins-Lopes, P. Comet assay reveals no genotoxicity risk of cationic solid lipid nanoparticles. J. Appl. Toxicol. 2014, 34, 395–403. [Google Scholar] [CrossRef] [PubMed]
  57. Doktorovova, S.; Santos, D.L.; Costa, I.; Andreani, T.; Souto, E.B.; Silva, A.M. Cationic solid lipid nanoparticles interfere with the activity of antioxidant enzymes in hepatocellular carcinoma cells. Int. J. Pharm. 2014, 471, 18–27. [Google Scholar] [CrossRef] [PubMed]
  58. Scarpa, E.S.; Emanuelli, M.; Frati, A.; Pozzi, V.; Antonini, E.; Diamantini, G.; Di Ruscio, G.; Sartini, D.; Armeni, T.; Palma, F.; et al. Betacyanins enhance vitexin-2-O-xyloside mediated inhibition of proliferation of T24 bladder cancer cells. Food Funct. 2016, 7, 4772–4780. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (a) Surface response chart of the 23 factorial design, representing the interactions of two distinct factors (time and cycle) favoring the determination of the best extraction conditions. (b) Surface response chart representing the interactions between the temperature and number of cycles. (c) Surface response chart representing the interactions between the time and temperature variables. (d) Pareto graph of 2 interactions between the variables applied to determine the yield as a response of the developed factorial planning. The vertical interrupted red line (in d) represents the significance of the compared variables; the right-hand side of the cut has significant representativity and the left-hand side does not present significance.
Figure 1. (a) Surface response chart of the 23 factorial design, representing the interactions of two distinct factors (time and cycle) favoring the determination of the best extraction conditions. (b) Surface response chart representing the interactions between the temperature and number of cycles. (c) Surface response chart representing the interactions between the time and temperature variables. (d) Pareto graph of 2 interactions between the variables applied to determine the yield as a response of the developed factorial planning. The vertical interrupted red line (in d) represents the significance of the compared variables; the right-hand side of the cut has significant representativity and the left-hand side does not present significance.
Biomolecules 10 00726 g001
Figure 2. Evaluation of the yield of red propolis extract samples obtained by the solvent accelerated extraction method, where the vertical interrupted red line represents the cutoff of 11%.
Figure 2. Evaluation of the yield of red propolis extract samples obtained by the solvent accelerated extraction method, where the vertical interrupted red line represents the cutoff of 11%.
Biomolecules 10 00726 g002
Figure 3. Cytotoxic analysis against tumor cell lines derived from human malignant neoplasms HCT116 (colon adenocarcinoma) and PC3 (prostate adenocarcinoma). - - - - Minimum percentage limit of cytotoxic activity considered to be at least moderate. ______ Minimal percentage limit of cytotoxic activity considered strong.
Figure 3. Cytotoxic analysis against tumor cell lines derived from human malignant neoplasms HCT116 (colon adenocarcinoma) and PC3 (prostate adenocarcinoma). - - - - Minimum percentage limit of cytotoxic activity considered to be at least moderate. ______ Minimal percentage limit of cytotoxic activity considered strong.
Biomolecules 10 00726 g003
Figure 4. Average inhibitory concentration graph capable of causing 50% of the maximal effect in HCT116 cells.
Figure 4. Average inhibitory concentration graph capable of causing 50% of the maximal effect in HCT116 cells.
Biomolecules 10 00726 g004
Table 1. Experimental variables used in the 23 full factorial design.
Table 1. Experimental variables used in the 23 full factorial design.
FactorsLevels
Minimum Value (−)Central Point (0)Maximum Value (+)
Number of cycles123
Extraction time (min)101520
Temperature (°C)405570
AssayNumber of cyclesExtraction time (min)Temperature (°C)
1−1 (1)−1 (10)−1 (40)
21 (3)−1 (10)−1 (40)
3−1 (1)1 (20)−1 (40)
41 (3)1 (20)−1 (40)
5−1 (1)−1 (10)1 (70)
61 (3)−1 (10)1 (70)
7−1 (1)1 (20)1 (70)
81 (3)1 (20)1 (70)
9 (PC)0 (2)0 (15)0 (55)
10 (PC)0 (2)0 (15)0 (55)
11 (PC)0 (2)0 (15)0 (55)
Table 2. Compounds identified in the extract of red propolis by gas chromatography coupled with mass spectrometry (GC/MS) with the relative area (%) and linear temperature programmed retention index (LTPRI) of the compounds.
Table 2. Compounds identified in the extract of red propolis by gas chromatography coupled with mass spectrometry (GC/MS) with the relative area (%) and linear temperature programmed retention index (LTPRI) of the compounds.
CompoundsSamplesLTPRI
1234567891011
Aromatic HydrocarbonsArea (%)TheoreticalCalculatedDifference
Toluene8.02.52.62.113.23.37.817.9-5.54.07747740
Benzene, 1,2,3-trimethyl- 0.56.10.64.4-3.62.97.2--2.09909922
Benzene, 1,2,4-trimethyl- 3.32.05.41.18.70.90.81.75.32.70.79769760
Indane0.60.3--0.4------10311032.41,4
Benzene, 1,2-diethyl- 0.3----------10461045−1
Benzene, 1-methyl-3-propyl- 0.40.3--0.4------104810491
Benzene, 1,4-diethyl- 0.40.5--0.5------105210531
Benzene, 1,3-diethyl- ----0.5------105410540
Benzene, 2-ethyl-1,4-dimethyl-0.60.2--0.4------10821085.24,3
Benzene, 4-ethyl-1,2-dimethyl- ----0.3------108310830
Benzene, 1,2,3-trimetoxy-5-(2-propenyl)---0.70.50.40.40.3----155415540
Saturated hydrocarbonsArea (%)
Tricosane--4.02.42.03.00.23.04.03.50.4229923001
Pentacosane15.110.514.58.46.17.90.610.511.110.30.8249925001
Hexacosane-0.41.40.91.00.80.80.21.50.60.4259926001
Heptacosane0.224.625.019.119.9-----1.227012700−1
Octacosane0.51.00.70.80.80.70.41.30.40.30.1279928001
Nonacosane0.12.51.811.910.216.613.016.221.015.814.2289929001
Triacontane-1.520.40.9-0.90.70.71.11.01.0299830002
Untriacontane0.223.0-17.6-18.520.022.431.021.319.93108 3100−8
Unsaturated HydrocarbonsArea (%)
Heptacos-1-ene----0.30.92.10.30.80.520.226742684.210.2
Nonacos-1-ene--1.41.1-------287528772
AlcoholsArea (%)
1-Pentanol, 2,3-dimethyl-----0.4--0.7---80982718
2-Hexyn-1-ol----2.4------8468471
1-Butanol, 3-methyl-, acetate3.21.01.60.95.01.02.44.22.71.5-8728720
Behenic alcohol2.10.6-0.3-0.47.20.40.50.38.624732470−3
Octacosanol---3.7-4.83.82.71.13.75.130983110.612.6
1-Heptacosanol---16.0--15.62.1-15.8-32973307−10
1-Triacontanol-----17.6-----330633060
EthersArea (%)
Methyleugenol--0.60.40.60.30.2----140114010
Isopropyl tetracosyl ether----0.7------299830002
KetonesArea (%)
Acetofenone----0.5---3.0-0.8106110610
2 (3H) -furanone, 5-dodecildi-hidro-1.23.65.1-2.05.42.73.65.46.24.621002104.34.3
TerpenesArea (%)
Lupenone--------11.910.812.233843384.20,2
Lupeol----20.36.68.80.1---349935001
Lupeol acetate16.417.510.66.13.03.43.70.2---35333525−8

Share and Cite

MDPI and ACS Style

de Carvalho, F.M.d.A.; Schneider, J.K.; de Jesus, C.V.F.; de Andrade, L.N.; Amaral, R.G.; David, J.M.; Krause, L.C.; Severino, P.; Soares, C.M.F.; Caramão Bastos, E.; et al. Brazilian Red Propolis: Extracts Production, Physicochemical Characterization, and Cytotoxicity Profile for Antitumor Activity. Biomolecules 2020, 10, 726. https://doi.org/10.3390/biom10050726

AMA Style

de Carvalho FMdA, Schneider JK, de Jesus CVF, de Andrade LN, Amaral RG, David JM, Krause LC, Severino P, Soares CMF, Caramão Bastos E, et al. Brazilian Red Propolis: Extracts Production, Physicochemical Characterization, and Cytotoxicity Profile for Antitumor Activity. Biomolecules. 2020; 10(5):726. https://doi.org/10.3390/biom10050726

Chicago/Turabian Style

de Carvalho, Felipe Mendes de Andrade, Jaderson Kleveston Schneider, Carla Viviane Freitas de Jesus, Luciana Nalone de Andrade, Ricardo Guimarães Amaral, Jorge Maurício David, Laíza Canielas Krause, Patrícia Severino, Cleide Mara Faria Soares, Elina Caramão Bastos, and et al. 2020. "Brazilian Red Propolis: Extracts Production, Physicochemical Characterization, and Cytotoxicity Profile for Antitumor Activity" Biomolecules 10, no. 5: 726. https://doi.org/10.3390/biom10050726

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop