Next Article in Journal
The Importance of Monitoring Cortisol in the Agri-Food Sector—A Systematic Review
Next Article in Special Issue
In Vitro and In Silico Anti-Picornavirus Triterpene Alkanoic Acid Ester from Saudi Collection of Rhazya stricta Decne
Previous Article in Journal
Associations between Plasma Lipid Mediators and Chronic Daily Headache Outcomes in Patients Randomized to a Low Linoleic Acid Diet with or without Added Omega-3 Fatty Acids
Previous Article in Special Issue
Development and Characterization of Yellow Passion Fruit Peel Flour (Passiflora edulis f. flavicarpa)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Elaboration and Characterization of Pereskia aculeate Miller Extracts Obtained from Multiple Ultrasound-Assisted Extraction Conditions

1
Departamento de Alimentos, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
2
Departamento de Ciências Exatas e Biológicas, Campus Sete Lagoas, Universidade Federal de São João del-Rei, Sete Lagoas 35702-031, Brazil
3
Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus Belo Horizonte, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
4
Departamento de Engenharia e Tecnologia de Alimentos, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, Campinas 130862-862, Brazil
5
Universidade Federal do Rio Grande do Norte, Distrito de Macaíba 59280-000, Brazil
6
Departamento de Pedagogia, Instituto de Educação Ciência e Tecnologia do Paraná, Palmas 85555-000, Brazil
7
Departamento de Química, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
8
Departamento de Ciências Agrárias, Campus Bambuí, Instituto Federal de Educação, Ciência e Tecnologia de Minas Gerais, Bambuí 38900-000, Brazil
9
Departamento de Engenharia de Alimentos, Campus Sete Lagoas, Universidade Federal de São João del-Rei, Sete Lagoas 35702-031, Brazil
*
Author to whom correspondence should be addressed.
Metabolites 2023, 13(6), 691; https://doi.org/10.3390/metabo13060691
Submission received: 30 December 2022 / Revised: 31 January 2023 / Accepted: 5 February 2023 / Published: 26 May 2023

Abstract

:
Pereskia aculeata Miller, is an unconventional food plant native to South America. This study aimed to investigate the influence of different ultrasonic extraction times (10, 20, 30, and 40 min) on the phytochemical profile, antioxidant and antibacterial activities of ethanolic extracts obtained from lyophilized Pereskia aculeate Miller (ora-pro-nobis) leaves, an under-researched plant. Morphological structure and chemical group evaluations were also conducted for the lyophilized P. aculeate leaves. The different extraction times resulted in distinct phenolic content and Antioxidant Activity (ATT) values. Different extraction time conditions resulted in phenolic compound contents ranging from 2.07 to 2.60 mg EAG.g−1 of extract and different ATT values. The ATT evaluated by DPPH was significantly higher (from 61.20 to 70.20 μM of TE.g−1 of extract) in extraction times of 30 and 40 min, respectively. For ABTS, it varied between 6.38 and 10.24 μM of TE.g−1 of extract and 24.34 and 32.12 μM ferrous sulp.g−1 of extract. All of the obtained extracts inhibited the growth of Staphylococcus aureus, particularly the treatment employing 20 min of extraction at the highest dilution (1.56 mg.mL−1). Although liquid chromatography analyses showed that chlorogenic acid was the primary compound detected for all extracts, Paper Spray Mass Spectrometry (PS-MS) suggested the extracts contained 53 substances, such as organic, fatty, and phenolic acids, sugars, flavonoids, terpenes, phytosterols, and other components. The PS-MS proved to be a valuable technique to obtain the P. aculeate leaves extract chemical profile. It was observed that the freeze-drying process enhanced the conservation of morphological structures of P. aculeate leaves, as evidenced by scanning electron microscopy (SEM). Fourier transform infrared spectroscopy (FTIR) identified carboxyl functional groups and proteins between the 1000 and 1500 cm−1 bands in the P. aculeate leaves, thus favoring water interaction and contributing to gel formation. To the best of our knowledge, this is the first study to evaluate different times (10, 20, 30 and 40 min) for ultrasound extraction of P. aculeate leaves. The polyphenols improved extraction, and high antioxidant activity demonstrates the potential for applying P. aculeate leaves and their extract as functional ingredients or additives in the food and pharmaceutical industries.

1. Introduction

Plant materials are natural sources of phytochemicals. In the human body, these compounds perform crucial roles such as protection, antioxidant, anti-inflammatory, antifungal, and anti-bacterial activities. On top of that, phytochemicals act in the immune system stimulation [1,2]. A Pereskia aculeate Miller, known worldwide as ora-pro-nobis, is a representative of phytochemical-rich plants. P. aculeate is a South American plant species, known by the acronym UFP(Unconventional Food Plant) (PANC, in Portuguese), belonging to the Cactaceae family and Pereskioideae subfamily. The plant’s leaves, its non-toxic edible portion, are used in traditional medicine and cooking [1,2].
The P. aculeate leaves are a nutritional complement primarily because of their high protein, fiber, calcium, and iron contents. Furthermore, P. aculeate extracts and leaves were also found to be rich in carotenoids and phenolic compounds, valuable phytochemicals which provide healing, anti-inflammatory, antifungal, and antioxidant activities, as well as analgesic potential [3,4,5,6,7].
While Cruz et al. [8] reported significant antioxidant and anti-hemolytic activities for ora-pro-nobis leaves, Torres et al. [9] identified anti-inflammatory and anticholinergics activities. The extracts obtained by Massocatto et al. [10] from P. aculeate leaves and fruit presented anticholinesterasic, cytotoxic, and antiproliferative effects. Pinto et al. [11,12] observed that ora-pro-nobis methanolic extract and hexanic fractios containing gels enhanced the excisional wound healing in mice, as well as analgesic effect of hydromethanolic fractions. These publications indicated the P. aculeate extract’s potential for applications in the pharmaceutical industry. According to Souza et al. [7], the essential oil obtained from P. aculeate leaves presented antioxidant, antimicrobial, and antifungical activities in in vitro assays. Considering this vast literature, one may infer that the ora-pro-nobis has a considerable potential to be used as a valuable raw-material, ingredient, or additive in the food/pharmaceutical industries.
Currently, the consumption of nutraceuticals and food supplements has increased, as the consumers have become more aware of the benefits of consuming products rich in bioactive compounds, which represent health benefits to the human body (e.g., disease prevention) [13]. Bioactive compounds with antioxidant capacity can be found in complex vegetable matrices. Hence, to obtain such compounds, it is necessary to employ more efficient and selective extraction techniques to the analyte of interest.
The most known traditional extraction methods are maceration, Soxhlet, and mechanical agitation. These methods are based on high temperature, long process times, and high solvent consuming techniques [14]. Moreover, in general, the solvents used in these methods are toxic and harmful to health and the environment.
Specific extraction conditions such as temperature, may directly affect the antioxidant and biological potential of compounds obtained from natural extracts [15]. Thus, more efficient extraction techniques providing satisfactory yields at mild temperatures such as the ultrasound-assisted extraction are promising alternatives. Compared to conventional methods for bioactive compound extraction, Ultrasound-Assisted Extraction (UAE) offers significant benefits regarding extraction time and process security [16,17].
The UAE technique is based on the formation of mechanical waves that form small bubbles. These bubbles grow and implode, resulting in the cavitation phenomenon. Then, the disruption of the vegetable material cell wall takes place, increasing its surface contact and allowing the entrance of the solvent in the matrix, consequently intensifying the mass transfer. UAE is a rapid technique which produces extracts with high yields. Furthermore, it is considered to be an environmentally friendly method because of its shorter extraction time and solvent consumption compared to conventional techniques [15,16]. Furthermore, the UAE enhances the phytomolecules yields and extracts/isolated biological efficiency [16,18].
Recent studies have shown that UAE is efficient for recovering different compounds (e.g., anthocyanins, phenolic compounds, flavonoids, carotenoids, and antioxidants) from various vegetal matrices [15,17]. Nevertheless, we could not find publications using UAE to obtain bioactive compounds from P. aculeate leaves and there are just few publications in the literature identifying chemical compounds found in P. aculeate leaves [19]. Moreover, it is crucial to understand the impact of extraction techniques on the proportion of bioactive compounds found in the extracts and leaves. It is also important to mention that natural extracts such as those obtained from ora-pro-nobis leaves may act as food preservatives, besides their health-beneficial effects, when applied in food production. These extracts can be substitutes for broadly used synthetic preservatives in food products [20,21].
In this context, the present paper aims to investigate the influence of different ultrasonic extraction times on the phytochemical profile and antioxidant and antibacterial activities of ethanolic extracts obtained from lyophilized P. aculeate leaves. In addition, a characterization of the morphological structure and presence of chemical groups was conducted. This study is intended to contribute to the scientific advance of the area by allowing the obtaining of optimized ultrasound-assisted extraction time of bioactive compounds form ora-pro-nobis leaves. The obtained extracts may be used as potential additives in food and pharmaceutical products.

2. Materials and Methods

2.1. Chemical Reagents and Bacterial Strains

The Folin–Ciocalteu, 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) diamonic salt (ABTS), 2,4,6-Tris(2-pyridyl)-s-triazine (TPTZ), 2,2-diphenyl-1-picrylhydrazyl (DPPH), 6-Hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox), catechin, caffeic acid, ellagic acid, quercetin, and chlorogenic acid reagents were supplied by from Sigma-Aldrich (San Luis, MO, USA,). All reagents were of analytical grade.
The reference strains of the Staphylococcus aureus ATCC 29213, Escherichia coli (ATCC 35218; beta-lactamase producer) and methicillin resistant Staphylococcus aureus Staphylococcus aureus resistant to methicillin (MRSA; ATCC 43300) bacteria were gently provided by the Clinical Microbiology Laboratory from the Faculty of Pharmacy at UFMG.

2.2. Plant Sampling

The P. aculeate leaves were collected from a crop at the Federal University of São João del-Rei (UFSJ, Sete Lagoas, Brazil) in March 2019.

2.3. Lyophilization

The P. aculeate leaves were selected according to their visual appearance (typical green color and integrity). After selection, the leaves were sanitized with flowing water and dried with paper towels. Thus, the material was submitted to freeze in an Ultra Low-Temperature Freezer at −55 °C for 24 h.
The lyophilization was carried out by a lyophilizer (LS-6000-A model, Terroni Equipamentos Científicos, São Carlos, Brazil) for 12 h. The leaves were conditioned in plastic bags at −18 °C until further analyses when the material was unfrozen, crushed in a blender (Philips Walita, São Paulo, Brazil), and sifted in a 16-mesh sieve.

2.4. Extracts Preparation

For the extract preparation, 5 g of the sample was mixed with 50 mL of a 50% ethyl alcohol solution. The mixture was weighed in an Erlenmeyer wrapped with aluminum foil and covered with plastic paraffin film (Parafilm®, São Paulo, Brazil) and aluminum foil to avoid alcohol evaporation during extraction.
The extracts were prepared using a digital ultrasonic washer (SoniClean 2, Sanders Medical, Santa Rita do Sapucaí, Brazil) at room temperature (20 °C), frequency 42 kHz, and power 160 W. Four different extraction times were evaluated, namely, 10, 20, 30, and 40 min.
After extractions, filtration was carried out with qualitative filter paper. The obtained extracts were vaporized in a rotary evaporator (R-215, Büchi, Valinhos, Brazil) at 45 °C until the alcohol removal, resulting in aqueous extracts, named EO10, EO20, EO30 and EO40 corresponding to 10, 20, 30 and 40 min ultrasound treatment, respectively.

2.5. Total Phenolic Compounds and Antioxidant Activity

Following the Folin–Ciocalteu method [22], the total phenolic compounds content was determined, with results expressed in equivalents of gallic acid (mg EAG.g−1 of the extract). The Antioxidant Activity (ATT) was evaluated using three distinct methodologies: (1) reaction with 2,2-diphenyl-1-picrylhydrazyl (DPPH) [23]; (2) free radical capture 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) [24]; (3) Ferric Reducing Antioxidant Power (FRAP) [24]. The analyses were carried out in a dark room and triplicate.

2.6. Phenolic Compounds Identification and Quantification

High-Performance Liquid Chromatography (HPLC) analyses were conducted according to the method described by Eça et al. [25]. The four extracts were filtered in a 0.22 μm nylon filter syringe and injected in HPLC equipment (Acquity UPLC® Class, Milford, Waters, MA, USA), equipped with a UV detector by diode array, quarternary bomb, degasser, autosampler, and column (Acquity UPLC ® BEH C18—2.1 × 100 mm; 1.7 µm, Waters, Milford, MA, USA). The flow was kept constant at 0.3 mL.min−1, with two mobile phases (A = acetonitrile and B = water : formic acid, 99.75:0.25).
Calibration curves used to identify the compounds were prepared with the following external standards: gallic acid, catechin, and chlorogenic acid (diluted in water for complete dissolution); caffeic acid, ellagic acid, and quercetin (diluted in methanol for complete dissolution). While the water diluted standards were run for 10 min in an isocratic elution mode of 5:95 v/v (A : B), a linear gradient was applied to the other standards: 0 min, 8% (A); 8 min, 15% (A); 14 min, 25% (A); 20 min, 8% (A). The UV spectra were obtained between 200 and 400 nm. Chromatograms were processed at 253, 271, 320, and 372 nm. The calibration curves were built within the following ranges: gallic acid and quercetin (21.2 to 212 μg.g−1), catechin (24.4 to 244 μg.g−1), chlorogenic acid (18.4 to 184 μg.g−1), and ellagic acid and caffeic acid (20 to 200 μg.g−1). All data acquired from these analyses were processed in the Empower® software, and the results were expressed in µg.g−1 of extract.

2.7. Antibacterial Activity

Antibacterial activity of the four extracts (E10, E20, E30 and E40) was assessed by the broth microdilution method in 96-well microplates, according to the Clinical and Laboratory Standards Institute [26]. First, 200 µL of each extract was added to 100 µL of Mueller Hinton broth (MHB). Serial dilutions (100 µL) were made in order to reach concentrations from 50 to 1.56%, that is, 50 to 1.56 mg.mL−1, after adding each bacterial suspension (100 μL) containing 1.5 × 105 colony forming units per milliliter (CFU.mL−1). A growth inhibition controls (0.1 µg.mL−1 of penicillin for S. aureus, 2.5 µg.mL−1 of gentamicin for E. coli and 2.5 µg.mL−1 of streptomycin for MRSA), a cell viability control (bacteria only), and a sterility control (MHB only) were used in triplicate in each assay. After incubation at 37 °C for 24 h, microplates were inspected visually for inhibition of bacterial growth, and wells with no visible growth were considered as the minimal inhibitory concentration (MIC), that is, the lowest concentration (mg.mL−1) of the extracts in which no visible bacterial growth was evidenced. All conditions were tested in triplicate in at least two independent assays.

2.8. Fourier-Transform Infrared (FTIR) Spectroscopy

The lyophilized P. aculeate leaves spectrum was acquired in the 4000 range at 500 cm−1, with 20 scannings by experiment (resolution of 4 cm−1). The FTIR spectroscopy was performed in a spectrometer (Shimadzu IRAffinity—1) with DLaTGS detector (Deuterated Triglycine Sulfate and Doped with L-alanine) and total attenuated reflectance accessory (ATR) with zinc crystal.

2.9. Scanning Electron Microscopy (SEM)

The microstructure of the lyophilized P. aculeate leaves fragments was evaluated using a scanning electron microscope with X-rays dispersive energy detector (Leo 440i, EDS: 6070, LEO Electron Microscopy/Oxford, Cambridge, UK). Before the analyses, the lyophilized fragments were dried and kept for 24 h in a vacuum. Subsequently, they were coated with a gold layer of 200 A° in a Sputter Coater EMITECH spray applicator (Model: K450, Kent, UK). The sample surfaces were assessed under vacuum, using an acceleration tension of 10 kV and 500×, 800×, 2000×, and 3000× magnification.

2.10. Chemical Profile Determination by Paper Spray Mass Spectrometry (PS—MS)

The extract presenting the best results for antioxidant and antimicrobial activities was evaluated in terms of chemical profile using an LCQ Fleet mass spectrometer (Thermo Scientific, Waltham, MA, USA), equipped with a paper spray ionization source, according to the methodology reported by Silva et al. [27]. The positive and negative ionization modes were used in triplicate. The equipment was used with source voltage equal to +5.0 kV (positive ionization mode) and −3.0 kV (negative ionization mode); capilar voltage of 4.0 V; transference tube temperature of 275 °C; tube lens voltage of 115 V; mass range from 100 to 1000 m/z.
An equilateral triangle-shaped chromatography paper (1.5 cm) was positioned in front of the mass spectrometer entrance to carry out the analyses. The paper was supported by a metallic connection and placed at a distance of 0.5 cm with a mobile platform (XYZ). This device was connected to a high-tension mass spectrometer source by a copper wire. Then, 2.0 μL of the extract was applied to the tip of the equilateral triangle-shaped chromatography paper, 40 μL of methanol was transferred to the paper, and the voltage source was switched on for data acquisition. The m/z was compared with the literature data and the subsequent fragmentation by sequential mass spectrometry for compound identification. Collision energies for compound fragmentation varied from 15 to 30 eV. The obtained mass spectra were processed with the Xcalibur software (Thermo Scientific, Waltham, MA, USA).

2.11. Statistical Analysis

The results were expressed with the replica means and their respective standard deviations. Results normalization, variance homogeneity, analysis of variance, and the mean comparison by Tukey test at 5% were carried out by the SPSS 15.0 program (SPSS Inc., Chicago, IL, USA).

3. Results and Discussion

3.1. Total Phenolic Compounds and Antioxidant Activity

Table 1 shows the P. aculeate leaves total phenolic compounds content and antioxidant activities.
It is essential to mention that the protocols employed in the antioxidant activity assessment were chosen because of their complementary features. Hence, it was possible to identify a multitude of bioactive compounds performing this activity in the samples.
In this study, the total phenolic compounds contents ranged from 2.07 to 2.60 mg EAG.g−1 of extract. Overall, it was possible to observe that different extraction times led to different phenolic content and antioxidant activity values. On the other hand, Sim et al. [28] examined the phenolic content of Pereskia grandifolia leaves extracts. The authors reported results of 19.08 mg EAG.g−1 using hexane as a solvent. Souza et al. [7], who also evaluated P. aculeate leaves chemical composition and biological activity, detected 5.17 mg EAG.g−1, 11.78 mg EAG.g−1, and 15.04 mg EAG.g−1 of phenolic compounds in the extracts, using chloroform, petroleum ether, and methanol as solvents. Additionally, different technologies of pressurized fluids were used by Torres et al. [9] to obtain antioxidante-rich extracts from P. aculeate leaves. The highest yields were obtained using Soxhlet extraction with ethanol (38.18 mg EAG.g−1) and Pressurized Liquid Extraction (PLE) (60.09 mg EAG.g−1) using ethanol as solvent at 110 °C. Cruz et al. [8] achieved high total phenolic values (64–65 mg EAG.g−1) using mechanical agitation conventional extraction and various mixture proportions of water, acetone, and ethanol.
Those variations of total phenolic compounds might result from diverse factors such as climate, cultivar, maturation stage and plant genetics. Moreover, the samples preparation techniques such as drying methods and type/condition of extraction also affect the recovery yield of phenolic compounds [14,15]. Solvents such as hexane, chloroform, petroleum ether, and methanol provide high extraction yields, but are toxic and restricted to food applications, as well as generate environmentally dangerous waste.
In the extraction aided by ultrasound, the bubbles formed during the process are responsible for the matrix cavitation. When in contact with the vegetal cell structure, they can disrupt the polymers by mechanical effect, creating microspores and allowing the extraction of polyphenols [29,30]. Therefore, it can be inferred that shorter ultrasound extraction times are also effective for disrupting the vegetal membrane, thus, releasing bioactive compounds.
P. aculeate leaves antioxidant activities are assigned mainly to secondary metabolites and some primary metabolites, particularly phenolic compounds and flavonoids [31]. Table 1 shows that AAT by DPPH was significantly higher for longer extraction times. This result may be explained by the higher extraction of bioactive compounds that have the ability to scavenge free radicals (DPPH), which was not possible in the shorter exposure times for this same sample. Souza et al. [7], when evaluating the P. aculeate leaves chemical composition and biological activity, also found higher antioxidant activity using DPPH in the extracts presenting higher phenolic compound content, similar to extract EO40. Cruz et al. [8] demonstrated that mixtures between water + ethanol and water + acetone provided high antioxidant capacities, measured by FRAP and DPPH. Torres et al. [9,32] evaluated PLE with pure water and ethanol at 80 °C, reporting antioxidant capacity of 0.25 mmol of TE.g−1 of extract (FRAP) and 1315 µmol of TE.g−1 of extract (ABTS), respectively. The authors reported that the highest ora-pro-nobis extracts antioxidant activities, measured with FRAP, were obtained using polar solvents, demonstrating the efficacy of solvents such as water and ethanol for extracting bioactive compounds. According to Mustafa and Turner [33], binary systems may favor the recovery of bioactive compounds because they promote the solute release while increase their solubility, thus, enhancing the extraction yield. This phenomenon was also observed by Cruz et al. [8] during the extraction of bioactive compounds from P. aculeate leaves using different solvent mixtures.
The results show that innovative e technologies, such as UAE, are feasible for obtaining bioactive compounds with antioxidant properties from vegetal matrices [34]. In addition, antioxidants obtained from P. aculeate leaves are natural and can be utilized as an alternative to synthetic antioxidants. It is also important to mention that previous studies regarding the evaluation of Pereskia spp. antioxidant activity presented in the literature did not employ a set of different antioxidant methods, as used in this research, emphasizing the importance of this work since antioxidant compounds can act by distinct mechanisms.

3.2. Identification and Quantification of Phenolic Compounds

The phenolic compounds identified and quantified by liquid chromatography corresponded to phenolic acids (chlorogenic acid, gallic acid, caffeic acid, and ellagic acid) and flavonoid (quercetin) groups. Table 2 shows the obtained results.
Figure 1 shows the chromatograms of identified phenolic compounds extracted from the lyophilized material with ethanol solution (50%) and UAE for 20 min. This treatment presented the highest concentrations of chlorogenic acid and the lowest antibacterial activity result value.
The results showed that phenolic acids are found at high concentrations in the P. aculeate leaves extracts. The chlorogenic acid, in particular, is the main compound identified in all extracts. High levels of chlorogenic acid were also verified in Pereskia aculeate by Agostini-Costa [35] for several genus of Cactoideae and Pereskioideae subfamilies.
On the other hand, extracts EOR30 and EOR40 presented the highest contents of gallic and ellagic acids. In addition, caffeic acid was detected only in these treatments. Therefore, the longer exposure time to ultrasound positively contributed to the gallic, caffeic, and ellagic acids extraction efficiency.
Compared to other compounds, the low content of quercetin found in the extracts may be explained by its high instability [36]. This result was also verified by publications in the literature where P. aculeate and Pereskia bleo (Kunth) leaves extracts were evaluated [19,37]. Finally, catechin was the only compound not detected in any of the P. aculeate leaves extracts assessed in this study. Nevertheless, it is important to mention that previous publications described the identification and quantification of this component (9.18 mg.g−1) for P. bleo ethanolic extract [37]. The absence of catechin in the extracts evaluated here may be explained by factors such as the geographical location of plant cultivation, species, harvest condition, leaves drying, and extraction methods. Moreover, the type of solvent used in the extraction process may directly affect the extraction efficiency because of the solvent polarity and sample profile [38].

3.3. Antibacterial Activity Evaluation

Table 3 shows the Minimal Inhibitory Concentration (MIC) of P. aculeate leaves extracts against S. aureus, E. coli, and MRSA bacterial strains. Extracts were evaluated in six different dilutions (50 to 1.56% v/v), i.e., from 50 to 1.56 mg.mL−1.
As one may notice from Table 3, the extracts presented divergent behaviors for bacterial growth inhibition against Gram-positive bacteria, S. aureus, and MRSA. On the other hand, none of them showed antibacterial activity against E. coli. Although all the extracts were elaborated using the same raw material and solvent, differences in inhibitory activity between S. aureus and MRSA might be related to the resistant phenotype of MRSA and the extraction method used. This result may be associated with the influence of different ultrasound-assisted extraction times as they appear to determine the type and content of P. aculeate leaves extracts compounds [39].
All extracts inhibited the S. aureus growth, particularly the EO20 treatment that inhibited the growth even at the highest dilution assessed (1.56 mg.mL−1). It is important to observe that EO20 treatment produced extracts with higher concentrations of chlorogenic acid (524.54 ± 0.00 μg.g−1), in comparison to E30 (459.38 ± 1.00 μg.g−1), E40 (364.48 ± 0.03 μg.g−1), and E10 (350.93 ± 0.01 μg.g−1). Promising results regarding the chlorogenic acid anti-bacterial activity were also obtained in the literature against S. aureus and Streptococcus mutans [40,41]. These results suggest that chlorogenic acid had an essential role in anti-bacterial activity against S. aureus (E20 showed lower MIC values compared to E10). Furthermore, other bioactive compounds such as caffeic acid (not identified for E10 and E20 treatments) and gallic acid could also benefit MIC values.
The mechanisms that confer MRSA resistance to penicillin, methicillin, and oxacillin (structural alterations of the penicillin-binding proteins—PBP) possibly reduced the compounds’ interaction affinity with the anti-bacterial activity present in the extracts, as observed by MIC values (12.5 to 50 mg.mL−1). Gram-negative bacteria, on the other hand, are known because of their high resistance to natural and conventional antibiotics (outer membranes), decreasing the penetration of antibacterial agents [42]. These results are also corroborated by publications in the literature using different types of plant extracts, including P. aculeate leaves, where better results for extracts of anti-bacterial activity were registered against Gram-positive bacteria, in comparison with those obtained against Gram-negative ones such as E. coli [7,19,43,44,45,46].
Souza et al. [7] also verified the antibacterial activity of P. aculeate leaves extracts using disk-diffusion assays. In this publication, the authors found that petroleum ether extract exhibited potent antibacterial activity against E. coli, while the chloroformic extract showed inhibitory activity against Bacillus cereus and S. aureus. The inhibition of E. coli growth presented by P. aculeate leaves petroleum ether extracts might be related to this solvent’s higher capacity of extracting low molecular weight phenolic compounds with antibacterial activity. However, it is essential to mention that although presenting better performance, those two solvents are toxic and environmental pollutants [47].
Recent studies approaching MIC analysis of P. aculeate leaves are scarce. Thus, this study shows that P. aculeate leaves extracts present anti-bacterial activity against Gram-positive bacteria. However, the inhibition of microbial growth may be linked to different factors such as the microorganism characteristics, the strains used, extraction conditions, and extracts composition.

3.4. Fourier-Transform Infrared (FTIR) Spectroscopy

The FTIR spectroscopy is a diagnostic analysis that provides evidence regarding the presence of functional groups in substances’ chemical structures. FTIR spectroscopy has been widely used for fast multicomponent analyses such as compound identification and structural determination. Thus, the infrared spectrum was obtained for functional groups verification in the lyophilized P. aculeate leaves (Figure 2). Similarly, Amaral et al. [48] used this technique to evaluate activated carbon-based structures on P. aculeate residue.
As one may notice in Figure 2, N-H bonds (amino group) can be observed at 1646 cm−1. The same pattern was observed by Reinas et al. [49] in chitosan infrared spectrograms. Bands at 1440 and 1320 cm−1 can be related to CH3, CH2, flavonoids, and aromatic groups, with C-H vibration and the enlargement of aromatic compounds vibration [50]. The band at 2916 cm−1 was characterized as C-H stretch vibrations in cellulose and hemicellulose [51,52]. The bands at 1000 cm−1 and 1050 cm−1 indicate the gallic acid structural conformation [53], as identified and quantified by chromatography in this study.
The peak observed within the band of 3267 cm−1 is characterized by the OH- bond. A similar result has been found in the literature [54], where lyophilized P. aculeate fruit mucilage infrared spectrum was evaluated. The presence of OH- bond conferred affinity for polymers in P. aculeate leaves mucilage with water molecules, a typical property of hydrophilic compounds [55]. The peak at 1234 cm−1 shows lignin’s C-O stretch bond [56].
The peak at the 890 cm−1 wave number was identified as characteristic of polysaccharide structures that originated from β-glycosidic bonds between the sugar units. Furthermore, 518 and 942 cm−1 bands can be associated with pectins and lignins [57,58]. Finally, the presence of functional carboxyl groups and proteins in the region the between 1000 and 1500 cm−1 bands [54,58,59] in P. aculeate leaves is evidence of the favoring interaction with water, contributing to crucial characteristics for colloid obtaining, such as gel formation.

3.5. Scanning Electron Microscopy (SEM) Analysis

Figure 3 shows SEM images of lyophilized P. aculeate leaf’s morphological structure (FOPN). One may notice that the leaves presented an irregular and porous surface, giving the material a broader contact area and contributing to water retention in the product [60,61]. This characteristic is particularly desirable for food production and emulsion formation, as observed by Sato et al. [62] and Conceição et al. [59]. As reported by the authors, this material feature is specifically beneficial for pasta production and obtaining emulsions from P. aculeate leaves. The FOPN microscopic analysis showed the presence of paracytic and anomocytic stomata (represented by arrows in Figure 3A,B). The publication by Squena et al. [63] which performed a morpho-anatomical analysis of P. aculeate aerial vegetative parts, also showed these structures.
Strong attraction and adhesion of minor and significant particles also indicate an excellent ordination obtained by the lyophilization process. This structure is characteristic of hygroscopic materials such as chitosan and guar gum [64,65]. The FOPN microstructures, such as porous and irregular surfaces, revealed characteristics similar to those of Monteiro et al. [66] in lyophilized P. aculeate leaves. The obtained microstructure is typical of ice sublimation formed in intracellular spaces. These microstructures are responsible for maintaining the leaf cellular structure, leading to particles with irregular surfaces formed by large pores [66].
As presented in Figure 3, the lyophilized P. aculeate leaves presented a rough surface. It was observed that more prolonged exposure to ultrasound-assisted extraction mediated the cavitation process in the vegetal matrix [30], releasing more significant quantities of phenolic acids. Considering these results, one may infer that the lyophilization process promoted the conservation of morphological structures of P. aculeate leaves, granting more versatility and stability for their components, mainly used for formulation purposes as an additive, for instance, in pharmaceutical and food industries.

3.6. Chemical Profile by Paper Spray Mass Spectrometry (PS-MS)

The PS-MS analysis was used to obtain the chemical fingerprint of complex matrices present at P. aculeate. This technique provides a super quick and low-cost methodology for assessing the overall quality of the material without generating chemical residues. Thus, the extract obtained by ethanol solution (50%) and ultrasound-assisted extraction for 20 min was chosen for chemical profile evaluation. The chosen extract, presenting the best results for antibacterial activity, was analyzed by PS-MS in positive and negative ionization modes, as shown in Figure 4.
The characterization in negative and positive ionization modes revealed 53 chemical compounds present, including organic, fatty, and phenolic acids. Additionally, sugars, flavonoids, terpenes, phytosterols, and other components were identified. Comparably, Garcia et al. [19] identified minor compounds in the P. aculeate leaves hydroethanolic extract (10 substances). The authors reported two phenolic acids (caffeic acid derivatives) and eight flavonoids (quercetin, kaempferol, and isorhamnetin glycoside derivatives) were identified. Table 4 shows the 33 substances identified for EO20 treatment using the negative ionization mode.
As one may notice from Table 4, the group of flavonoids was the major compound group quantified in the negative ionization mode. Similar results were reported in the publication by Silva et al. [47], where the Eriobotrya japonica Lindl chemical profile was characterized by PS-MS. This result is in accordance with the class of secondary plant metabolites represented by flavonoids. These substances have important health benefits, such as antioxidant activity [79].
According to Li et al. [76], the quercetin-3-O-glucoside (m/z 463 [M-162-H]-) and rutin (m/z 609 [M-308-H]-) MS/MS fragmentation generated the m/z 301 ions, corresponding to quercetin without the glucoside and rutinoside units, respectively. In the present study, the main flavonoids found in P. aculeate leaves were detected by PS-MS, namely quercetin, isorhamnetin, and kaempferol [35]. However, kaempferol-xylose is classified as a glycoside, a common characteristic of P. aculeate in different parts, which also presents various aglycones.
Although HPLC did not detect caffeic acid and quercetin, the PS-MS analysis identified those compounds. This result highlights the technique’s ability to evaluate compound fingerprints, covering an extensive molecular mass range, and to identify compounds that often are not detected by other methods. Souza et al. [78] also found caffeic acid (m/z 179) and ferulic acid (m/z 193) in P. aculeate in natura leaves. 5-feruloylquinic acid was characterized as an ester of ferulic acid and quinic acid.
To the best of our knowledge, this is the first study that identified substances from each chemical class of organic acids, fatty acids, phenolic acids, sugars, flavonoids, and terpenes in lyophilized P. aculeate leaves extract obtained by an environmentally friendly process technology.
Using the positive ionization mode, 20 distinct compounds were identified, including flavonoids, phytosterols, terpenes, and sugar chemical classes. Table 5 shows the identified compounds using positive ionization mode for EO20 samples.
Again, the primary group identified in the samples was the group of flavonoids. Most of these flavonoids were found to be conjugated derivatives of glycosides. As expected, the conjugated form of these compounds is a typical characteristic of the leaves because it provides better plant protection [81].
Pinto et al. [11] used the positive mode of HPLC-MS/MS to detect only seven different compounds (tryptamine, abrine, mescaline, hordenine, petunidin, di-tert-butylphenol isomers, and quercetin) in P. aculeate leaves extract samples. The authors also evaluated the antinociceptive activity of extracts obtained by hydromethanolical solutions. The discrepancy found in the results, using the same positive ionization mode, indicated that the use of ultrasound-assisted extraction enhanced the bioactive compounds obtained, in comparison to traditional extraction processes [34,87].

4. Conclusions

The present work presented a set of methods for evaluating antioxidant compounds that have not yet been verified in studies of Pereskia aculeate Miller leaves. We were able to verify that the content of phenolic compounds and antioxidant activity are influenced by different extraction times in an ultrasonic bath. The ora-pro-nobis leaf extracts showed antibacterial activity against S. aureus and MRSA strains, also demonstrating a potential use in formulations with against these Gram-positive bacteria. EO20 extract, for example, inhibited S. aureus up to the last dilution evaluated (1.56 mg.mL−1) and showed the highest content of chlorogenic acid, a substance with described antibacterial activity.
A total of 53 compounds were found in P. aculeate leaves extracts applying the PS-MS technique, proving to be a source of organic, fatty, and phenolic acids, flavonoids, terpenes, phytosterols, sugars among other substances with important antioxidant and antimicrobial activities. Employing FTIR and SEM, it was possible to verify the presence of functional groups that help the formation of colloids, which are desirable in pharmaceutical and food formulations in industries, and that lyophilization was able to promote greater conservation of the morphological structures of ora-pro-nobis leaves. As a perspective, extracts from the leaves of P. aculeate Miller can be used in nutraceuticals and functional foods, due to the high levels of polyphenols, including their antioxidant and antibacterial activities.

Author Contributions

Conceptualization, M.C.C.M., C.V.P., A.C.C.F.F.d.P., J.O.F.M., R.A. and C.A.F.; methodology, M.C.C.M., V.D.M.S., M.S.M.S., V.T.d.V.C. and D.T.V.P.; formal analysis, M.C.C.M.; investigation, M.C.C.M. and P.R.A.; resources, C.V.P., A.C.C.F.F.d.P., J.O.F.M., R.A. and C.A.F.; data curation, M.C.C.M.; writing—original draft preparation, M.C.C.M. and A.S.J.d.S.; writing—review and editing C.V.P., A.C.C.F.F.d.P., J.O.F.M., R.A. and C.A.F.; visualization, M.C.C.M., V.D.M.S., M.S.M.S., V.T.d.V.C., H.d.O.P.M. and D.T.V.P.; supervision, C.V.P., A.C.C.F.F.d.P., J.O.F.M., R.A. and C.A.F.; project administration, C.V.P., A.C.C.F.F.d.P., J.O.F.M., R.A. and C.A.F.; funding acquisition, C.V.P., A.C.C.F.F.d.P., J.O.F.M., R.A. and C.A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank the Universidade Federal de Minas Gerais (UFMG), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—CAPES, Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG), Pró-Reitoria de Pesquisa (PRPq) da Universidade Federal de Minas Gerais (UFMG), Universidade Federal de São João del-Rei (UFSJ) and Instituto Federal de Educação Ciência e Tecnologia de Minas Gerais (IFMG) for financial support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Silva, D.O.; Seifert, M.; Nora, F.R.; Bobrowski, V.L.; Freitag, R.A.; Kucera, H.R.; Nora, L.; Gaikwad, N.W.; Al, S.E.T. Acute Toxicity and Cytotoxicity of Pereskia aculeata, a Highly Nutritious Cactaceae Plant. J. Med. Food 2017, 20, 403–409. [Google Scholar] [CrossRef] [PubMed]
  2. Soares, S.; Barbalho, S.M.; Guiguer, E.L.; Araújo, A.C.; Cincotto, P.; Farinazzi-machado, F.M.V.; Maria, L.; Lima, D.L.; Clivati, B.; Mendes, C.G. Effects of Pereskia aculeata Miller on the Biochemical Profiles and Body Composition of Wistar Rats. J. Biosci. Med. 2015, 3, 82–89. [Google Scholar]
  3. Agostini-Costa, T.D.S.; Wondraceck, D.C.; Rocha, W.D.S.; Silva, D.B.D. Carotenoids Profile and Total Polyphenols in Fruits of Pereskia aculeata Miller. Rev. Bras. Frutic. 2012, 34, 234–238. [Google Scholar] [CrossRef]
  4. Amado, A.; Alves, R.; Freitas, D.; Lanzi, G.; Henrique, P.; Evangelista, L.; Rita, M. Chemical Structure and Physical-Chemical Properties of Mucilage from the Leaves of Pereskia aculeata. Food Hydrocoll. 2017, 70, 20–28. [Google Scholar] [CrossRef]
  5. De Castro Campos Pinto, N.; Scio, E. The Biological Activities and Chemical Composition of Pereskia Species (Cactaceae)-A Review. Plant Foods Hum. Nutr. 2014, 69, 189–195. [Google Scholar] [CrossRef]
  6. Agostini-Costa, T.S.; Pêssoa, G.K.A.; Silva, D.B.; Gomes, I.S.; Silva, J.P. Carotenoid Composition of Berries and Leaves from a Cactaceae—Pereskia Sp. J. Funct. Foods 2014, 11, 178–184. [Google Scholar] [CrossRef]
  7. Souza, L.F.; Caputo, L.; De Barros, I.B.I.; Fratianni, F.; Nazzaro, F.; De Feo, V. Pereskia aculeata Muller (Cactaceae) Leaves: Chemical Composition and Biological Activities. Int. J. Mol. Sci. 2016, 17, 1478. [Google Scholar] [CrossRef]
  8. Cruz, T.M.; Santos, J.S.; do Carmo, M.A.V.; Hellström, J.; Pihlava, J.M.; Azevedo, L.; Granato, D.; Marques, M.B. Extraction Optimization of Bioactive Compounds from Ora-pro-Nobis (Pereskia aculeata Miller) Leaves and Their in Vitro Antioxidant and Antihemolytic Activities. Food Chem. 2021, 361, 130078. [Google Scholar] [CrossRef]
  9. Torres, T.M.S.; Álvarez-Rivera, G.; Mazzutti, S.; Sánchez-Martínez, J.D.; Cifuentes, A.; Ibáñez, E.; Ferreira, S.R.S. Neuroprotective Potential of Extracts from Leaves of Ora-pro-Nobis (Pereskia aculeata) Recovered by Clean Compressed Fluids. J. Supercrit. Fluids 2021, 179, 105390. [Google Scholar] [CrossRef]
  10. Massocatto, A.M.; de Souza Silva, N.F.; Kazama, C.C.; Dal Bem Pires, M.; Takemura, O.S.; Jacomassi, E.; Gois Ruiz, A.L.T.; Laverde, A. Biological Activity Survey of Pereskia aculeata Mill. and Pereskia grandifolia Haw. (Cactaceae). Pharm. Sci. 2022, 28, 156–165. [Google Scholar] [CrossRef]
  11. Pinto, N.D.C.C.; Duque, A.P.D.N.; Pacheco, N.R.; Mendes, R.D.F.; Motta, E.V.D.S.; Bellozi, P.M.Q.; Ribeiro, A.; Salvador, M.J.; Scio, E. Pereskia aculeata: A Plant Food with Antinociceptive Activity. Pharm. Biol. 2015, 53, 1780–1785. [Google Scholar] [CrossRef] [PubMed]
  12. Pinto, N.D.C.C.; Cassini-Vieira, P.; de Souza-Fagundes, E.M.; Barcelos, L.S.; Castañon, M.C.M.N.; Scio, E. Pereskia aculeata Miller Leaves Accelerate Excisional Wound Healing in Mice. J. Ethnopharmacol. 2016, 194, 131–136. [Google Scholar] [CrossRef] [PubMed]
  13. Martins, Z.E.; Pinho, O.; Ferreira, I.M.P.L.V.O. Food Industry By-Products Used as Functional Ingredients of Bakery Products. Trends Food Sci. Technol. 2017, 67, 106–128. [Google Scholar] [CrossRef]
  14. Wang, L.; Weller, C.L. Recent Advances in Extraction of Nutraceuticals from Plants. Trends Food Sci. Technol. 2006, 17, 300–312. [Google Scholar] [CrossRef]
  15. Chemat, F.; Rombaut, N.; Sicaire, A.G.; Meullemiestre, A.; Fabiano-Tixier, A.S.; Abert-Vian, M. Ultrasound Assisted Extraction of Food and Natural Products. Mechanisms, Techniques, Combinations, Protocols and Applications. A Review. Ultrason. Sonochem. 2017, 34, 540–560. [Google Scholar] [CrossRef]
  16. Natiéli, C.P.B.; Bromberger, S.M.; Barcelos, F.D.C.; Nichelle, L.B.; Barin, J.S.; Rosa, C.S.D.; Terra, N.N. Ultrasonic Assisted Extraction to Obtain Bioactive, Antioxidant and Antimicrobial Compounds from Marcela. Ciência Rural 2018, 48, 1–6. [Google Scholar]
  17. Yusoff, I.M.; Taher, Z.M.; Rahmat, Z.; Chua, L.S. A Review of Ultrasound-Assisted Extraction for Plant Bioactive Compounds: Phenolics, Flavonoids, Thymols, Saponins and Proteins. Food Res. Int. 2022, 157, 111268. [Google Scholar] [CrossRef]
  18. Heinrich, M. Ethnopharmacology in the 21st Century—Grand Challenges. Front. Pharmacol. 2010, 1, 1980–1982. [Google Scholar] [CrossRef]
  19. Garcia, J.A.A.; Corrêa, R.C.G.; Barros, L.; Pereira, C.; Abreu, R.M.V.; Alves, M.J.; Calhelha, R.C.; Bracht, A.; Peralta, R.M.; Ferreira, I.C.F.R. Phytochemical Profile and Biological Activities of “Ora-pro-Nobis” Leaves (Pereskia aculeata Miller), an Underexploited Superfood from the Brazilian Atlantic Forest. Food Chem. 2019, 294, 302–308. [Google Scholar] [CrossRef]
  20. Atanasov, A.G.; Waltenberger, B.; Pferschy-wenzig, E. Europe PMC Funders Group Discovery and Resupply of Pharmacologically Active Plant-Derived Natural Products: A Review. Biotechnol. Adv. 2016, 33, 1582–1614. [Google Scholar] [CrossRef]
  21. Gonçalves, G.A.; Corrêa, R.C.G.; Barros, L.; Dias, M.I.; Calhelha, R.C.; Correa, V.G.; Bracht, A.; Peralta, R.M.; Ferreira, I.C.F.R. Effects of in Vitro Gastrointestinal Digestion and Colonic Fermentation on a Rosemary (Rosmarinus officinalis L.) Extract Rich in Rosmarinic Acid. Food Chem. 2019, 271, 393–400. [Google Scholar] [CrossRef] [PubMed]
  22. Sahan, Y.; Cansev, A.; Gulen, H. Effect of Processing Techniques on Antioxidative Enzyme Activities, Antioxidant Capacity, Phenolic Compounds, and Fatty Acids of Table Olives. Food Sci. Biotechnol. 2013, 22, 613–620. [Google Scholar] [CrossRef]
  23. Official Methods of Analysis of the Association of Official Analytical Chemists, 19th ed.; Horwitz, W. (Ed.) AOAC: Gaithersburg, MD, USA, 2012. [Google Scholar]
  24. Rufino, M.D.S.M.; Alves, R.E.; de Brito, E.S.; Pérez-Jiménez, J.; Saura-Calixto, F.; Mancini-Filho, J. Bioactive Compounds and Antioxidant Capacities of 18 Non-Traditional Tropical Fruits from Brazil. Food Chem. 2010, 121, 996–1002. [Google Scholar] [CrossRef]
  25. Eça, K.S.; Machado, M.T.C.; Hubinger, M.D.; Menegalli, F.C. Development of Active Films From Pectin and Fruit Extracts: Light Protection, Antioxidant Capacity, and Compounds Stability. J. Food Sci. 2015, 80, C2389–C2396. [Google Scholar] [CrossRef]
  26. Performance Standards for Antimicrobial Susceptibility Testing; Supplement. CLSI document M100-S25; CLSI: Wayne, PA, USA, 2017.
  27. Silva, M.R.; Freitas, L.G.; Souza, A.G.; Araújo, R.L.B.; Lacerda, I.C.A.; Pereira, H.V.; Augusti, R.; Melo, J.O.F. Antioxidant Activity and Metabolomic Analysis of Cagaitas (Eugenia dysenterica) Using Paper Spray Mass Spectrometry. J. Braz. Chem. Soc. 2019, 30, 1034–1044. [Google Scholar] [CrossRef]
  28. Sri Nurestri, A.; Norhanom, A.; Sim, K. Phenolic Content and Antioxidant Activity of Pereskia grandifolia Haw. (Cactaceae) Extracts. Pharmacogn. Mag. 2010, 6, 248. [Google Scholar] [CrossRef]
  29. Medina-torres, N.; Ayora-talavera, T.; Espinosa-andrews, H. Ultrasound Assisted Extraction for the Recovery of Phenolic Compounds from Vegetable Sources. Agronomy 2017, 7, 47. [Google Scholar] [CrossRef]
  30. Wang, J.; Jia, Z.; Zhang, Z.; Wang, Y.; Liu, X.; Wang, L.; Lin, R.; Schmidt, T.J. Analysis of Chemical Constituents of Melastoma dodecandrum Lour. By UPLC-ESI-Q-Exactive Focus-MS/MS. Molecules 2017, 22, 476. [Google Scholar] [CrossRef]
  31. Andrade, T.C.; De Freitas, P.H.S.; Ribeiro, J.M.; Pinto, P.F.; de Souza-Fagundes, E.M.; Scio, E.; Ribeiro, A. Avaliação Da Atividade Antioxidante e Imunomoduladora Dos Metabólitos Primários de Pereskia aculeata Miller. J. Biol. Pharm. Agric. Manag. 2021, 17, 358–376. [Google Scholar]
  32. Torres, T.M.S.; Guedes, J.A.C.; de Brito, E.S.; Mazzutti, S.; Ferreira, S.R.S. High-Pressure Biorefining of Ora-pro-Nobis (Pereskia aculeata). J. Supercrit. Fluids 2022, 181, 105514. [Google Scholar] [CrossRef]
  33. Mustafa, A.; Turner, C. Pressurized Liquid Extraction as a Green Approach in Food and Herbal Plants Extraction: A Review. Anal. Chim. Acta 2011, 703, 8–18. [Google Scholar] [CrossRef] [PubMed]
  34. Balasubramaniam, V.G.; Sathvika, P.A.S.; Antony, U. Effect of Enzyme Pretreatment in the Ultrasound Assisted Extraction of Finger Millet Polyphenols. J. Food Sci. Technol. 2019, 56, 1583–1594. [Google Scholar] [CrossRef] [PubMed]
  35. Agostini-costa, T.S. Bioactive Compounds and Health Benefits of Pereskioideae and Cactoideae: A Review. Food Chem. 2020, 327, 126961. [Google Scholar] [CrossRef]
  36. Dos Reis, L.C.R.; Facco, E.M.P.; Salvador, M.; Flôres, S.H.; Rios, A.D.O. Characterization of Orange Passion Fruit Peel Flour and Its Use as an Ingredient in Bakery Products. J. Culin. Sci. Technol. 2020, 18, 214–230. [Google Scholar] [CrossRef]
  37. Hassanbaglou, B. Antioxidant Activity of Different Extracts from Leaves of Pereskia bleo (Cactaceae). J. Med. Plants Res. 2012, 6, 2932–2937. [Google Scholar] [CrossRef]
  38. Chirinos, R.; Pedreschi, R.; Rogez, H.; Larondelle, Y.; Campos, D. Phenolic Compound Contents and Antioxidant Activity in Plants with Nutritional and/or Medicinal Properties from the Peruvian Andean Region. Ind. Crops Prod. 2013, 47, 145–152. [Google Scholar] [CrossRef]
  39. Rabelo, S.V.; Da Costa, M.M.; Libório, R.C.; Almeida, J.R.G.D.S. Atividade Antioxidante e Antimicrobiana de Extratos de Atemoia (Annona cherimola Mill. × A. squamosa L.). Rev. Bras. Frutic. 2014, 36, 265–271. [Google Scholar] [CrossRef]
  40. Cerqueira, M.Â.P.R.; Souza, B.; Martins, J.T.; Teixeira, J.A.; Vicente, A.A. Functional Properties of Gleditsia Triacanthos Seeds Extracts and Their Incorporation into Galactomannan Films for Food Applications. In Total Food: Sustainability of the Agri-Food Chain; Royal Society of Chemistry: London, UK, 2010; pp. 238–243. ISBN 978-1-84755-750-6. [Google Scholar]
  41. Soni, H.; Malik, J.; Singhai, A.K.; Sharma, S. Antimicrobial and Antiinflammatory Activity of the Hydrogels Containing Rutin Delivery. Asian J. Chem. 2013, 25, 8371–8373. [Google Scholar] [CrossRef]
  42. Ribeiro, A.F.C.; Telles, T.C.; Ferraz, V.P.; Souza-Fagundes, E.M.; Cassali, G.D.; Carvalho, A.T.; Melo, M.M. Effect of Arrabidaea Chica Extracts on the Ehrlich Solid Tumor Development. Rev. Bras. Farmacogn. Braz. J. Pharmacogn. 2012, 22, 364–373. [Google Scholar] [CrossRef]
  43. Bazargani, M.M.; Rohloff, J. Antibiofilm Activity of Essential Oils and Plant Extracts against Staphylococcus aureus and Escherichia coli Biofilms. Food Control 2016, 61, 156–164. [Google Scholar] [CrossRef]
  44. Miranda, G.S.; Santana, G.S.; Machado, B.B.; Coelho, F.P.; Carvalho, C.A. Atividade Antibacteriana in Vitro de Quatro Espécies Vegetais Em Diferentes Graduações Alcoólicas. Rev. Bras. Plantas Med. 2013, 15, 104–111. [Google Scholar] [CrossRef]
  45. Muruzović, M.Ž.; Mladenović, K.G.; Stefanović, O.D.; Vasić, S.M.; Čomić, L.R. Extracts of Agrimonia eupatoria L. as Sources of Biologically Active Compounds and Evaluation of Their Antioxidant, Antimicrobial, and Antibiofilm Activities. J. Food Drug Anal. 2016, 24, 539–547. [Google Scholar] [CrossRef] [PubMed]
  46. Nostro, A.; Guerrini, A.; Marino, A.; Tacchini, M.; Di Giulio, M.; Grandini, A.; Akin, M.; Cellini, L.; Bisignano, G.; Sarac, H.T. In Vitro Activity of Plant Extracts against Biofilm-Producing Food-Related Bacteria. Int. J. Food Microbiol. 2016, 238, 33–39. [Google Scholar] [CrossRef] [PubMed]
  47. Silva, V.D.M.; Macedo, M.C.C.; dos Santos, A.N.; Silva, M.R.; Augusti, R.; Lacerda, I.C.A.; Melo, J.O.F.; Fante, C.A. Bioactive Activities and Chemical Profile Characterization Using Paper Spray Mass Spectrometry of Extracts of Eriobotrya japonica Lindl. Leaves. Rapid Commun. Mass Spectrom. 2020, 34, e8883. [Google Scholar] [CrossRef] [PubMed]
  48. Amaral, T.N.; Junque Ora, L.A.; Alves, C.C.D.; Dloveora, N.L.; Pradd, M.E.T.; Resende, J.V. De Extraction of Hydrocolloids from Pereskia aculeata Miller: Reuse of Process Residue as Activated Carbon for the Pigment-Removal Phase. Food Sci. Technol. 2018, 38, 77–85. [Google Scholar] [CrossRef]
  49. Reinas, A.E.; Hoscheid, J.; Outuki, P.M.; Cardoso, M.L.C. Preparation and Characterization of Microcapsules of Pterodon Pubescens Benth. By Using Natural Polymers. Braz. J. Pharm. Sci. 2014, 50, 919–930. [Google Scholar] [CrossRef]
  50. Oliveira, R.N.; Mancini, M.C.; de Oliveira, F.C.S.; Passos, T.M.; Quilty, B.; Thiré, R.M.d.S.M.; McGuinness, G.B. FTIR Analysis and Quantification of Phenols and Flavonoids of Five Commercially Available Plants Extracts Used in Wound Healing. Rev. Mater. 2016, 21, 767–779. [Google Scholar] [CrossRef]
  51. Oliveira, T.Í.S.; Rosa, M.F.; Cavalcante, F.L.; Pereira, P.H.F.; Moates, G.K.; Wellner, N.; Mazzetto, S.E.; Waldron, K.W.; Azeredo, H.M.C. Optimization of Pectin Extraction from Banana Peels with Citric Acid by Using Response Surface Methodology. Food Chem. 2016, 198, 113–118. [Google Scholar] [CrossRef]
  52. Pelissari, F.M.; Sobral, P.J.d.A.; Menegalli, F.C. Isolation and Characterization of Cellulose Nanofibers from Banana Peels. Cellulose 2014, 21, 417–432. [Google Scholar] [CrossRef]
  53. Ragupathi Raja Kannan, R.; Arumugam, R.; Anantharaman, P. Fourier Transform Infrared Spectroscopy Analysis of Seagrass Polyphenols. Curr. Bioact. Compd. 2011, 7, 118–125. [Google Scholar] [CrossRef]
  54. Silva, S.H.; Neves, I.C.O.; Oliveira, N.L.; de Oliveira, A.C.F.; Lago, A.M.T.; de Oliveira Giarola, T.M.; de Resende, J.V. Extraction Processes and Characterization of the Mucilage Obtained from Green Fruits of Pereskia aculeata Miller. Ind. Crops Prod. 2019, 140, 111716. [Google Scholar] [CrossRef]
  55. Saha, D.; Bhattacharya, S. Hydrocolloids as Thickening and Gelling Agents in Food: A Critical Review. J. Food Sci. Technol. 2010, 47, 587–597. [Google Scholar] [CrossRef] [PubMed]
  56. El-din, G.A.; Amer, A.A.; Malsh, G.; Hussein, M. Study on the Use of Banana Peels for Oil Spill Removal. Alex. Eng. J. 2018, 57, 2061–2068. [Google Scholar] [CrossRef]
  57. Lupoi, J.S.; Gjersing, E.; Davis, M.F. Evaluating Lignocellulosic Biomass, Its Derivatives, and Downstream Products with Raman Spectroscopy. Front. Bioeng. Biotechnol. 2015, 3, 50. [Google Scholar] [CrossRef] [PubMed]
  58. Martins, Q.; Felix Arinos, N.; Aguirre, C.; Brito de Faria, J. Pereskia aculeata Vibrational Model by Raman Characterization and DFT Method. Int. J. Innov. Educ. Res. 2021, 9, 485–505. [Google Scholar] [CrossRef]
  59. Conceição, M.C.; Junqueira, L.A.; Silva, K.C.G.; Prado, M.E.T.; de Resende, J.V. Thermal and Microstructural Stability of a Powdered Gum Derived from Pereskia aculeata Miller Leaves. Food Hydrocoll. 2014, 40, 104–114. [Google Scholar] [CrossRef]
  60. Alves, P.L.D.S.; Ascheri, J.L.R. Analise Por Imagem e Microscopia Electrônica Por Varredura Das Farinhas Extrudadas de Arroz e Maracujá. Hig. Aliment 2016, 30, 144–148. [Google Scholar]
  61. Liew, S.Q.; Ngoh, G.C.; Yusoff, R.; Teoh, W.H. Sequential Ultrasound-Microwave Assisted Acid Extraction (UMAE) of Pectin from Pomelo Peels. Int. J. Biol. Macromol. 2016, 93, 426–435. [Google Scholar] [CrossRef]
  62. Sato, R.; Cilli, L.P.D.L.; de Oliveira, B.E.; Maciel, V.B.V.; Venturini, A.C.; Yoshida, C.M.P. Nutritional Improvement of Pasta with Pereskia aculeata Miller: A Non-Conventional Edible Vegetable. Food Sci. Technol. 2019, 39, 28–34. [Google Scholar] [CrossRef]
  63. Squena, A.P.; dos Santos, V.L.P.; Franco, C.R.C.; Budel, J.M. Morpho-Anatomy Analysis of Aerial Vegetative Parts of Pereskia aculeata Mill. CactaceaE. Cad. Esc. Saúde 2012, 8, 189–207. [Google Scholar]
  64. Renault, F.; Sancey, B.; Badot, P.M.; Crini, G. Chitosan for Coagulation/Flocculation Processes—An Eco-Friendly Approach. Eur. Polym. J. 2009, 45, 1337–1348. [Google Scholar] [CrossRef]
  65. Wang, Q.; Ellis, P.R.; Ross-Murphy, S.B. Dissolution Kinetics of Guar Gum Powders. I. Methods for Commercial Polydisperse Samples. Carbohydr. Polym. 2002, 49, 131–137. [Google Scholar] [CrossRef]
  66. Monteiro, R.L.; Garcia, A.H.; Tribuzi, G.; Mattar Carciofi, B.A.; Laurindo, J.B. Microwave Vacuum Drying of Pereskia aculeata Miller Leaves: Powder Production and Characterization. J. Food Process Eng. 2021, 44, e13612. [Google Scholar] [CrossRef]
  67. Al Kadhi, O.; Melchini, A.; Mithen, R.; Saha, S. Development of a LC-MS/MS Method for the Simultaneous Detection of Tricarboxylic Acid Cycle Intermediates in a Range of Biological Matrices. J. Anal. Methods Chem. 2017, 2017, 1–12. [Google Scholar] [CrossRef] [PubMed]
  68. Abu-reidah, I.M.; Ali-shtayeh, M.S.; Jamous, R.M.; Arráez-román, D.; Segura-carretero, A. HPLC—DAD—ESI-MS/MS Screening of Bioactive Components from Rhus coriaria L. (Sumac) Fruits. Food Chem. 2015, 166, 179–191. [Google Scholar] [CrossRef]
  69. Sun, J.; Liang, F.; Bin, Y.; Li, P.; Duan, C. Screening Non-Colored Phenolics in Red Wines Using Liquid Chromatography/Ultraviolet and Mass Spectrometry/Mass Spectrometry Libraries. Molecules 2007, 12, 679–693. [Google Scholar] [CrossRef]
  70. Aaby, K.; Ekeberg, D.; Skrede, G. Characterization of Phenolic Compounds in Strawberry (Fragaria × ananassa) Fruits by Different HPLC Detectors and Contribution of Individual Compounds to Total Antioxidant Capacity. J. Agric. Food Chem. 2007, 55, 4395–4406. [Google Scholar] [CrossRef]
  71. Kang, J.; Price, W.E.; Ashton, J.; Tapsell, L.C.; Johnson, S. Identification and Characterization of Phenolic Compounds in Hydromethanolic Extracts of Sorghum Wholegrains by LC-ESI-MSn. Food Chem. 2016, 211, 215–226. [Google Scholar] [CrossRef]
  72. Chen, B.; Long, P.; Sun, Y.; Meng, Q.; Liu, X.; Cui, H.; Lv, Q.; Zhang, L. The Chemical Profiling of Loquat Leaf Extract by HPLC-DAD-ESI-MS and Its Effects on Hyperlipidemia and Hyperglycemia in Rats Induced by a High-Fat and Fructose Diet. Food Funct. 2017, 8, 687–694. [Google Scholar] [CrossRef]
  73. Zhang, L.; Zhu, M.; Shi, T.; Guo, C.; Huang, Y.; Chen, Y.; Xie, M. Recovery of Dietary Fiber and Polyphenol from Grape Juice Pomace and Evaluation of Their Functional Properties and Polyphenol Compositions. Food Funct. 2017, 8, 341–351. [Google Scholar] [CrossRef]
  74. Simirgiotis, M.J.; Schmeda-Hirschmann, G.; Bórquez, J.; Kennelly, E.J. The Passiflora Tripartita (Banana Passion) Fruit: A Source of Bioactive Flavonoid C-Glycosides Isolated by HSCCC and Characterized by HPLC-DAD-ESI/MS/MS. Molecules 2013, 18, 1672–1692. [Google Scholar] [CrossRef] [PubMed]
  75. Guo, Y.; Gu, Z.; Liu, X.; Liu, J.; Ma, M.; Chen, B.; Wang, L. Rapid Analysis of Corni Fructus Using Paper Spray-Mass Spectrometry. Phytochem. Anal. 2017, 28, 344–350. [Google Scholar] [CrossRef] [PubMed]
  76. Li, Z.H.; Guo, H.; Xu, W.B.; Ge, J.; Li, X.; Alimu, M.; He, D.J. Rapid Identification of Flavonoid Constituents Directly from PTP1B Inhibitive Extract of Raspberry (Rubus idaeus L.) Leaves by HPLC-ESI-QTOF-MS-MS. J. Chromatogr. Sci. 2016, 54, 805–810. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, W.; Zhao, X.; Sun, C.; Li, X.; Chen, K. Phenolic Composition from Different Loquat (Eriobotrya japonica Lindl.) Cultivars Grown in China and Their Antioxidant Properties. Molecules 2015, 20, 542–555. [Google Scholar] [CrossRef]
  78. Souza, A.H.D.; Silva, B.M.; Silva, E.C.D.; Augusti, R.; Melo, J.O.F.; Carlos, L.D.A. Influence of Thermal Processing on the Characteristics and Chemical Profile of Orapro- Nobis by PS/MS Paper Spray. Res. Soc. Dev. 2021, 10, e12110212119. [Google Scholar] [CrossRef]
  79. Liskova, A.; Koklesova, L.; Samec, M.; Smejkal, K.; Samuel, S.M.; Varghese, E.; Abotaleb, M.; Biringer, K.; Kudela, E.; Danko, J.; et al. Flavonoids in Cancer Metastasis. Cancers 2020, 12, 1498. [Google Scholar] [CrossRef]
  80. Gobbo-Neto, L.; Lopes, N.P. Online Identification of Chlorogenic Acids, Sesquiterpene Lactones, and Flavonoids in the Brazilian Arnica Lychnophora Ericoides Mart. (Asteraceae) Leaves by HPLC-DAD-MS and HPLC-DAD-MS/MS and a Validated HPLC-DAD Method for Their Simultaneous Analysis. J. Agric. Food Chem. 2008, 56, 1193–1204. [Google Scholar] [CrossRef]
  81. Silva, V.; Arquelau, P.; Silva, M.; Augusti, R.; Melo, J.; Fante, C. Use of Paper Spray-Mass Spectrometry to Determine the Chemical Profile of Ripe Banana Peel Flour and Evaluation of Its Physicochemical and Antioxidant Properties. Quim. Nova 2020, 43, 579–585. [Google Scholar] [CrossRef]
  82. Jia, C.; Zhu, A.Y.; Zhang, J.; Yang, J.; Xu, C.; Mao, D. Identification of Glycoside Compounds from Tobacco by High Performance Liquid Chromatography/Electrospray Ionization Linear Ion-Trap Tandem Mass Spectrometry Coupled with Electrospray Ionization Orbitrap Mass Spectrometry. J. Braz. Chem. Soc. 2017, 28, 629–640. [Google Scholar] [CrossRef]
  83. Ren, Q.; Long, S.S. Chemical Identification and Quantification of HU-GU Capsule by UHPLC–Q-TOF-MS and HPLC-DAD. Rev. Bras. Farmacogn. 2017, 27, 557–563. [Google Scholar] [CrossRef]
  84. Cavaliere, C.; Foglia, P.; Pastorini, E.; Samperi, R.; Laganà, A. Identification and Mass Spectrometric Characterization of Glycosylated Flavonoids in Triticum Durum Plants by High-Performance Liquid Chromatography with Tandem Mass Spectrometry. Rapid Commun. Mass Spectrom. 2005, 19, 3143–3158. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, W.W.; Luo, J.G.; Wang, J.S.; Lu, Y.Y.; Kong, L.Y. LC-DAD-ESI-MS-MS for Characterization and Quantitative Analysis of Diterpenoids from Coleus forskohlii. Chromatographia 2009, 70, 1635–1643. [Google Scholar] [CrossRef]
  86. Xiong, S.; Li, J.; Mu, Y.; Zhu, X.; Wang, X.; Zhang, Z. The Effects of Gender and Multiple Oral Dosing on the Pharmacokinetics and Bioavailability of Morroniside in Beagle Dogs: A Pilot Study. Indian J. Pharm. Sci. 2017, 79, 303–306. [Google Scholar] [CrossRef]
  87. Singh, B.; Singh, N.; Thakur, S.; Kaur, A. Ultrasound Assisted Extraction of Polyphenols and Their Distribution in Whole Mung Bean, Hull and Cotyledon. J. Food Sci. Technol. 2016, 54, 921–932. [Google Scholar] [CrossRef]
Figure 1. Identified phenolic compounds chromatograms in the lyophilized P. aculeate leaves extracted with ethanol (50%) and ultrasound for 20 min (EO20). (A): spectrum of standard pattern diluted in water; 1: gallic acid; 2: chlorogenic acid; (B): spectrum of standard pattern diluted in methanol; 3: ellagic acid; 4: quercetin.
Figure 1. Identified phenolic compounds chromatograms in the lyophilized P. aculeate leaves extracted with ethanol (50%) and ultrasound for 20 min (EO20). (A): spectrum of standard pattern diluted in water; 1: gallic acid; 2: chlorogenic acid; (B): spectrum of standard pattern diluted in methanol; 3: ellagic acid; 4: quercetin.
Metabolites 13 00691 g001
Figure 2. Lyophilized P. aculeate leaves Infrared spectrum with Fourier transform.
Figure 2. Lyophilized P. aculeate leaves Infrared spectrum with Fourier transform.
Metabolites 13 00691 g002
Figure 3. Lyophilized P. aculeate leaves SEM images. * sign for changed multiplication. The legend is unique in that it is the same figure with different sources of extension, as evidenced in each subfigure (AD).
Figure 3. Lyophilized P. aculeate leaves SEM images. * sign for changed multiplication. The legend is unique in that it is the same figure with different sources of extension, as evidenced in each subfigure (AD).
Metabolites 13 00691 g003
Figure 4. Paper spray mass spectra representation. (A) PS-MS spectrogram in negative ionization mode and (B) PS-MS spectrogram in positive ionization mode of the lyophilized P. aculeate leaves extract extracted with ethanol (50%) by ultrasound for 20 min (EO20).
Figure 4. Paper spray mass spectra representation. (A) PS-MS spectrogram in negative ionization mode and (B) PS-MS spectrogram in positive ionization mode of the lyophilized P. aculeate leaves extract extracted with ethanol (50%) by ultrasound for 20 min (EO20).
Metabolites 13 00691 g004
Table 1. Total phenolic compounds (TPC) and antioxidant activity (ATT) of P. aculeate leaves hydroalcoholic extracts.
Table 1. Total phenolic compounds (TPC) and antioxidant activity (ATT) of P. aculeate leaves hydroalcoholic extracts.
MethodEO10 *EO20 *EO30 *EO40 *
TPC
mg EAG.g−1 of extract
2.35 ± 0.03 ab2.14 ± 0.04 bc2.07 ± 0.07 c2.60 ± 0.06 a
AAT by ABTS *+
(μM de TE.g−1 of extract)
10.24 ± 0.40 a9.00 ± 0.08 b6.38 ± 0.17 c7.14 ± 0.17 c
AAT by FRAP
(μM ferrous sulp.g−1 of extract)
30.00 ± 0.27 ab24.34 ± 0.87 c32.13 ± 0.60 a27.37 ± 0.83 bc
AAT by DPPH
(μM of TE.g−1 of extract)
24.48 ± 0.15 b28.22 ± 0.99 b70.20 ± 1.05 a61.20 ± 1.12 a
* Means value ± standard deviation; n = 3. Equal letters on the same line do not differ significantly from each other at 5%, by Tukey’s test (p < 0.05). Aqueous extracts, named EO10, EO20, EO30 and EO40 corresponding to 10, 20, 30 and 40 min ultrasound treatment, respectively; AAT: total antioxidant activity; TE: Trolox equivalent; EAG: equivalents acid gallic.
Table 2. Phenolics profile (μg.g−1 of extract) in P. aculeate leaves hydroalcoholic extracts.
Table 2. Phenolics profile (μg.g−1 of extract) in P. aculeate leaves hydroalcoholic extracts.
CompoundEO10 *EO20 *EO30 *EO40 *
Chlorogenic acid350.93 ± 0.01 d524.54 ± 0.00 a459.38 ±1.00 b364.48 ±0.03 c
Gallic acid0.49 ± 0.17 d0.31± 0.09 c12.01 ± 0.20 b14.66 ± 0.11 a
Caffeic acidNDND29.03 ± 0.04 b31.23 ± 0.02 a
Ellagic acid1.00 ± 0.25 d1.35 ± 0.02 c6.32 ± 0.00 b10.01 ± 0.01 a
Quercetin0.05 ± 0.03 d0.10 ± 0.05 a0.08 ±0.01 c0.09 ± 0.08 b
CatechinNDNDNDND
* Mean value ± standard deviation; n = 3. Equal letters on the same line do not differ significantly from each other at 5%, by Tukey’s test (p < 0.05). Aqueous extracts, named EO10, EO20, EO30 and EO40 corresponding to 10, 20, 30 and 40 min ultrasound treatment, respectively; ND: not detected.
Table 3. Minimal inhibitory concentration (mg.mL−1) of the P. aculeate leaves hydroalcoholic extracts against Staphylococcus aureus, Escherichia coli and methicillin resistant Staphylococcus aureus (MRSA).
Table 3. Minimal inhibitory concentration (mg.mL−1) of the P. aculeate leaves hydroalcoholic extracts against Staphylococcus aureus, Escherichia coli and methicillin resistant Staphylococcus aureus (MRSA).
Staphylococcus aureus
(ATCC 29213)
MRSA
(ATCC 43300)
Escherichia coli
(ATCC 35218)
EO10EO20EO30EO40EO10EO20EO30EO40EO10EO20EO30EO40
6.251.563.136.252512.550NANANANANA
NA: Not active. EO10, EO20, EO30 and EO40 correspond to 10, 20, 30 and 40 min ultrasound treatment, respectively.
Table 4. Identified ions in lyophilized P. aculeate leaves extract obtained from extraction with ethanol (50%) by ultrasound for 20 min (EO20) by PS-MS in negative ionization mode.
Table 4. Identified ions in lyophilized P. aculeate leaves extract obtained from extraction with ethanol (50%) by ultrasound for 20 min (EO20) by PS-MS in negative ionization mode.
Compoundm/zMS/MSReference
Organic acids
Fumaric acid11571(Al Kadhi et al. [67])
Mallic acid133115(Abu-Reidah et al. [68]; Silva et al. [27])
Cumaric acid163119(Abu-Reidah et al. [68]; Sun et al. [69])
Ferulic acid193149(Wang et al. [30])
Fertaric acid325163,193(Aaby et al. [70];
Abu-Reidah et al. [68])
Fatty acids
Stearic acid283237(Wang et al. [30])
Ricinoleic acid297183(Wang et al. [30])
Eicosanoic acid311293(Wang et al. [30])
Trihydroxy-octadecadienoic acid327291,309(Kang et al. [71])
Phenolic acids
Caffeic acid179135(Kang et al. [71])
Quinic acid191127,173(Abu-Reidah et al. [68];
Chen et al. [72])
5-feruloylquinic acid367175(Zhang et al. [73])
Apigenin-6-C-glucoside431431(Kang et al. [71])
Eriodictiol 6,8 di-C-glucoside
flavonoid
611491,593(Simirgiotis et al. [74])
Sugars
Hexose215179(Guo et al. [75]; Silva et al. [27])
Saccharide371113,121,231,249(Kang et al. [71])
Flavonoids
Kaempferol–xylose417152,285(Chen et al. [72])
Quercetin-3-rhamnoside447447,300(Zhang et al. [73])
Verbonol453435(Abu-Reidah et al. [68])
Quercetin-3-O-glucoside463300,301,343(Li et al. [76]; Zhang et al. [77])
Taxifolin hexoside465303,447(Kang et al. [71])
Dimethyl Ellagic acid hexoside491454(Silva et al. [27])
Caffeoyl derivative hexose499337(Kang et al. [71])
Kaempferol-3-O-rutinoside593285,447(Silva et al. [27]; Wang et al. [30])
Rutin609255,271,301,463(Chen et al. [72]; Silva et al. [27];
Wang et al. [30])
Naringin579271,459(Sun et al. [69])
Isorhamnetin-3-O-rutinoside623315(Souza et al. [78])
Vicenin II derivative629593(Simirgiotis et al. [74])
Terpenes
Sorhamnetin-7-O-glucopyranoside477243,343(Wang et al. [30])
Metil corosolate485423,467(Chen et al. [72])
Others
Spinochrome A265235(Abu-Reidah et al. [56])
Plastoquinone 3339135,163,203(Souza et al. [78])
Vaccihein A377289,347,361(Souza et al. [78])
Table 5. Identified ions in lyophilized P. aculeate leaves extract obtained from extraction with ethanol (50%) by ultrasound for 20 min (EO20) by PS-MS in positive ionization mode.
Table 5. Identified ions in lyophilized P. aculeate leaves extract obtained from extraction with ethanol (50%) by ultrasound for 20 min (EO20) by PS-MS in positive ionization mode.
Compoundsm/zMS/MSReference
Flavonoids
3-O-methylquercetin317121,193,245,274(Gobbo-Neto and Lopes [80];
Silva et al. [81])
6,8-di-C-β-glicopiranosil cristine579495,507,543,561(Gobbo-Neto and Lopes [80])
Lucenine-2 (6,8-di-C-β-glucopiranosilluteoline611527,593(Gobbo-Neto and Lopes [80])
Rutin633331, 487,615(Jia et al. [82])
Tricin di-O,O-hexosídeo655493(Silva et al. [81])
Sagittatoside A677531(Ren and Long [83])
3-Hidroxicariine-O-glucose-rhamnose693547(Ren and Long [83])
lolidide-β-D-glucopyranoside711549,651,693(Jia et al. [82])
Chrysoeriol O,O-malonyl hexoside797711(Cavaliere et al. [84])
Phytosterols
Sitosterol397247(Wang et al. [30])
Terpenes
Deacetylforskolin369235,431(Abu-Reidah et al. [68];
Zhang et al. [85])
Vomifoliol β-D-409353,391,394(Jia et al. [71])
Dihidroisovaltrato425365(Abu-Reidah et al. [68])
Ononin431269,431(Ren and Long [83])
Sugars
Sacarose365185(Guo et al. [75])
Morroniside429267(Guo et al. [75]; Xiong et al. [86])
Phenolic Acids
Licanic acid293257,275(Wang et al. [30])
Fatty Acids
Magnoflorine342282,342(Ren and Long [83])
Others
L arginine175129(Silva et al. [27])
Diallyl phthalate247187(Ren and Long [83])
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Macedo, M.C.C.; Silva, V.D.M.; Serafim, M.S.M.; da Veiga Correia, V.T.; Pereira, D.T.V.; Amante, P.R.; da Silva, A.S.J.; de Oliveira Prata Mendonça, H.; Augusti, R.; de Paula, A.C.C.F.F.; et al. Elaboration and Characterization of Pereskia aculeate Miller Extracts Obtained from Multiple Ultrasound-Assisted Extraction Conditions. Metabolites 2023, 13, 691. https://doi.org/10.3390/metabo13060691

AMA Style

Macedo MCC, Silva VDM, Serafim MSM, da Veiga Correia VT, Pereira DTV, Amante PR, da Silva ASJ, de Oliveira Prata Mendonça H, Augusti R, de Paula ACCFF, et al. Elaboration and Characterization of Pereskia aculeate Miller Extracts Obtained from Multiple Ultrasound-Assisted Extraction Conditions. Metabolites. 2023; 13(6):691. https://doi.org/10.3390/metabo13060691

Chicago/Turabian Style

Macedo, Maria Clara Coutinho, Viviane Dias Medeiros Silva, Mateus Sá Magalhães Serafim, Vinícius Tadeu da Veiga Correia, Débora Tamires Vitor Pereira, Patrícia Regina Amante, Antônio Soares Júnior da Silva, Henrique de Oliveira Prata Mendonça, Rodinei Augusti, Ana Cardoso Clemente Filha Ferreira de Paula, and et al. 2023. "Elaboration and Characterization of Pereskia aculeate Miller Extracts Obtained from Multiple Ultrasound-Assisted Extraction Conditions" Metabolites 13, no. 6: 691. https://doi.org/10.3390/metabo13060691

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