Next Article in Journal
Development of Pharmacological Strategies with Therapeutic Potential in Ischemic Stroke
Next Article in Special Issue
Bioactives and Technological Quality of Functional Biscuits Containing Flour and Liquid Extracts from Broccoli By-Products
Previous Article in Journal
17β-Estradiol (E2) Upregulates the ERα/SIRT1/PGC-1α Signaling Pathway and Protects Mitochondrial Function to Prevent Bilateral Oophorectomy (OVX)-Induced Nonalcoholic Fatty Liver Disease (NAFLD)
Previous Article in Special Issue
Memory Recovery Effect of a New Bioactive Innovative Combination in Rats with Experimental Dementia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Metabolite Profiling, through LC-ESI/LTQOrbitrap/MS Analysis, of Antioxidant Extracts from Physalis alkekengi L.

1
Department of Pharmacy, University of the Study of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
2
Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy
3
Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(12), 2101; https://doi.org/10.3390/antiox12122101
Submission received: 20 November 2023 / Revised: 5 December 2023 / Accepted: 10 December 2023 / Published: 12 December 2023

Abstract

:
Due to the increasing use of Physalis alkekengi L. as a food supplement and starting material for tea preparation, a comprehensive analysis of green extracts was performed. Two different extraction methods were applied to yellow Physalis alkekengi L. fruit and calyx and compared: hydroalcoholic extraction and decoction. Characterization of the metabolome of the calyx and fruit of yellow Physalis alkekengi L. was performed by LC-ESI/LTQOrbitrap/MS followed by LC-ESI/LTQOrbitrap/MS/MS to identify 58 phytocompounds using the two different extraction techniques. Subsequently, through preliminary spectrophotometric assays followed by cell studies, the antioxidant activity of the different Physalis alkekengi L. extracts were evaluated. It was found that Physalis alkekengi L. extracts are a good source of metabolites such as flavonoids, organic acids, phenylpropanoids, physalins and carotenoids, with various biological activities, in particular, antioxidant activity capable of reducing the production of free radicals in intestinal Caco-2 cells. For the first time, an integrated approach (metabolomics approach and antioxidant evaluation) was applied to the study of Physalis alkekengi green extracts and decoctions, the green extraction method mostly used in herbal preparations. An interesting finding was the high antioxidant activity shown by these extracts.

Graphical Abstract

1. Introduction

Physalis alkekengi L. (family Solanaceae) is a plant native to Asia and southern Europe. It is also called Chinese lantern, Japanese lantern, bladder cherry, and winter cherry. It is now spread all over the world [1].
It is characterized by an upright, tufted growth habit and can reach a height of up to 60 cm when fully grown. The light green leaves are alternate, oval, and slightly hairy. Between July and August (in the northern hemisphere), usually from the second year after planting, the plant bears fruit filled with berries about 17 mm in diameter, which are covered in a papery calyx; they can be eaten both cooked and raw, and are the only edible part of the plant [2].
The formation of the fruit is fascinating: the calyx swells to a length of 5–7 cm, then swells like a balloon of parchment paper and changes color, turning from the initial green to a bright red of autumn. If the fruit remains on the plant during the winter season, the husk slowly disintegrates, and only the fibrous scaffold remains intertwined with the berry inside [1].
Physalis alkekengi L. is rich in steroids, flavonoids, terpenoids, alkaloids, phenylpropanoids, phenolic acids, etc. As for many other species of the genus Physalis, it contains a wide variety of physalins [3,4].
It has medicinal properties, so has been cultivated since ancient times [5,6]. It must be noted that it is a toxic plant in all its parts except the berry, which is very juicy, and has a fresh and somewhat bitter taste. Bitterness tends to decrease if the fruit is frozen [7]. The berries contain large amounts of vitamin C, citric acid, tannin, and sugar [2,8]. Among its therapeutic properties, its use in the treatment of kidney and bladder stones and as a diuretic stand out.
The Chinese lantern has many biological activities such as anti-inflammatory, antibacterial, antiseptic, sedative, laxative, diuretic, hypoglycemic, and spasmolytic, which is why it is widely used in traditional medicine. In Chinese medicine, Physalis alkekengi L. is a remedy for several diseases [9].
Extracts and complex fractions isolated from Physalis plants are known in the literature to show biological and pharmacological activities, and most are associated with the presence of the above-mentioned secondary metabolites. The total data disclose the potential of Physalis spp. as highly functional foods, profitable crops, and as sources of valuable secondary metabolites, novel medicine and cosmetics [8].
It has been shown in the literature that the phenolic compounds identified in Physalis alkekengi L. are able to inhibit the growth of breast and colon cancer cells [10]. In addition, the fruit peel extract showed anti-inflammatory activity by relieving inflammation in mice with inflammatory bowel disease [11]. The fruit contains compounds called withanolides that may have anti-inflammatory effects, potentially protecting against colon cancer [12]. Physalis alkekengi L. is a good source of antioxidants partly due to its high vitamin C content, but also due to the presence of bioactive compounds such as physalines and flavonoids [10]. For this reason, its intake can help the body counteract the activity of free radicals. These compounds, which are a by-product of human metabolism, are often the cause of serious diseases such as cancer. Etzbach et al. demonstrated that the skin of the berries (332.00 µg/g dw) has almost three times the amount of antioxidants as their pulp (118.50 µg/g dw) [13]. In addition, antioxidant levels peak when the fruit is ripe [10].
The present study was aimed at characterizing the metabolome by LC-ESI/LTQOrbitrap/MS analysis of the calyx and fruit to identify the majority of phytocompounds of Physalis alkekengi L. Subsequently, through preliminary spectrophotometric assays and then by cell studies, the antioxidant activity of various Physalis alkekengi L. extracts was evaluated. It was found that Physalis alkekengi L. extracts are a good source of metabolites with various biological activities, in particular, antioxidant activity capable of reducing the production of free radicals in the intestinal Caco-2 cell line.

2. Materials and Methods

2.1. Raw Materials

P. alkekengi L. was purchased online from Frutta Web (https://www.fruttaweb.com/) on 10 November 2019. The plant matrix was divided into two parts: leaves that comprise the calyx, and fruit. Two types of extraction were performed on these plant parts: decoction and hydroalcoholic extraction. On this basis, the samples were divided as follows: PACD (P. alkekengi L. calyx decoction), PAFD (P. alkekengi L. fruit decoction), PACE (P. alkekengi L. calyx hydroalcoholic extract), and PAFE (P. alkekengi L. fruit hydroalcoholic extract).

2.2. Reagents and Solvents

Ethanol (≥99.9%) and water (≥99.9%) employed for the extractions were bought from VWR (Milan, Italy). Acetonitrile (ACN), formic acid, water and methanol of LC-MS grade were purchased from Merk (Merk KGaA, Darmstadt, Germany). ABTS (2,20-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid)), Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid), DPPH (2,2-Diphenyl-1-picrylhydrazyl), NaOH (sodium hydroxide), NaNO2 (sodium nitrite), AlCl3 (aluminum chloride), K2S2O8 (potassium persulfate), PBS (phosphate buffered saline), vitamin C and quercetin were purchased from Sigma-Aldrich (Milan, Italy).
2′,7′-dichlorodihydrofluoresceindiacetate (H2-DCF-DA) and MTT powder were purchased from Sigma Aldrich.

2.3. Sample Preparation

After the various parts of the Physalis alkekengi L. were divided, they were stored at −80 °C and then freeze-dried by a freeze dryer (LIO 5Pascal) that applied vacuum pressure and freezing temperature (−48 °C) to remove 95–98% of the water content of the plant materials while retaining their nutritional value and composition. The freeze-dried plant material was extracted using two extraction methods: by sonication with an ethanol/water solution (70:30) and by water decoction. For hydroalcoholic extractions, 1 g of freeze-dried matrix was used with 40 mL of ethanol/water solution (70/30) for the calyx, and 20 mL for the fruit. The extractions were performed in an ultrasonic bath (Grant, ultrasonic bath XUBA3) for 15 min at room temperature, repeating the extractions three times and then filtering the extracts with paper filters.
Amounts of 1 g of calyx and 2.5 g of fruit were used for decoctions with 50 mL of water, following EP pharmacopeia guidelines.

2.4. LC-ESI/LTQOrbitrap/MS and LC-ESI/LTQOrbitrap/MS/MS Analysis

A method was developed for metabolite profiling from decoctions and ethanolic extractions by coupling an HPLC with a hybrid mass spectrometer combining the linear trap quadrupole (LTQ) and the Orbitrap mass analyzer. Experiments were performed using a Thermo Scientific liquid chromatography system equipped with an Accela 600 quaternary pump, and an Accela autosampler coupled to a linear trap–Orbitrap hybrid mass spectrometer (LTQ-Orbitrap XL, Thermo Fisher Scientific, Bremen, Germany) with an electrospray ionization (ESI) source. A Phenomenex Luna C18 5 µm (150 mm × 2.00 mm) column (Phenomenex, Aschaffenburg, Germany) was used for the separation. The mobile phases used were water + 0.1% formic acid (A) and acetonitrile + 0.1% formic acid (B). An increasing linear gradient (v/v) at a flow rate of 0.200 mL/min of solvent B was used: 0–35 min, from 5 to 95%; 35–36 min, from 95 to 5%; and maintaining this percentage until 40 min. The experiments were performed in positive ion mode and 5 µL of each sample was injected. The parameters of the ESI source were as follows: capillary voltage, 49 V; tube lens voltage, 120 V; ion source temperature, 280 °C; sheath and auxiliary gas (N2) flow rates, 30 and 5; sweep gas, 0. The full m/z range suitable for recording MS spectra was 180–1600. To study the fragmentation pattern, a data-dependent scan was performed in which the precursor ions corresponding to the most intense peaks were fragmented in LC–MS analysis with a collision energy of 30%. Xcalibur software version 2.2 was used for instrument control, data acquisition and data analysis.

2.5. DPPH· Radical Scavenging Activity

The antioxidant activity of Physalis alkekengi L. was investigated by evaluating the anti-radical activity of the stables 1,1-diphenyl-2-picrylhydrazyl radical (DPPH·) according to the previously published method [14].
The first step was to prepare a methanolic solution of DPPH· with a concentration of 0.025 g/L. The samples to be studied were prepared by adding an aliquot (37.5 µL) of the methanolic solution containing different amounts of each extract (50, 100, and 200 µg/mL) to 1.5 mL of the previously prepared DPPH· solution.
The controls consisted of 37.5 µL and 1.5 mL of DPPH· solution. After 10 min, the absorbance was measured at 517 nm using a UV spectrophotometer (Spectrophotometer Multiskan Go, Thermo Scientific). All experiments were performed in triplicate.
The antioxidant extracts reduced the DPPH radical in the compound DPPH-H, resulting in a decrease in absorbance. Thus, the antiradical activity of the extracts was evaluated as a decrease in absorbance at 517 nm, more precisely expressed as a percentage of radical inhibition of DPPH according to Equation (1):
%   I n h i b i t i o n   D P P H = A 0 A e A e 100
where A0 is the average of the absorbances of the control in triplicate and Ae is the average of the absorbances of each concentration of the various extracts in triplicate. Vitamin C was used as the reference compound.

2.6. Trolox Equivalent Antioxidant Capacity (TEAC) Assay

Trolox equivalent antioxidant capacity assay was set up as a further test following a protocol already published [14]. The extracts were diluted with water and methanol to obtain final concentrations of 250, 500, 750 and 1000 µg/mL. The assay was set up in 96-well plates, adding 15 µL of each sample to 150 µL of ABTS. The absorbance was measured at 734 nm. The experiments were performed in triplicate.
The percentage diminution in absorbance was calculated for each concentration relative to a blank absorbance (methanol/water) and plotted as a function of the concentration of the compound or standard 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox).
The antioxidant activities were indicated as a TEAC value, i.e., the concentration of a Trolox standard solution with the same antioxidant activity as 1 mg/mL of the tested extract; quercetin-3-O-glucoside was used as the reference compound.

2.7. Cell Culture

The Caco-2 cell line was obtained from ECACC (Salisbury, UK). Caco-2 cells are derived from human colorectal adenocarcinoma which, once reaching confluence, spontaneously differentiate into normal enterocytes. Dulbecco’s modified Eagle’s medium (DMEM) with low glucose and with L-Arginin, phosphate-buffered saline (PBS) without MgCl2 and CaCl2, fetal bovine serum (FBS) and penicillin/streptomycin 1X were purchased from Euroclone (Milano, Italy). Caco-2 cells were grown in T75 flasks until their confluence reached 80%, at 37 °C in a 5% CO2 humidified atmosphere in DMEM supplemented with 10% FBS, and 1% antibiotic/antimycotic solution (100 U/mL penicillin, 100 mg/mL streptomycin) [15]. At passage 45–60, cells were removed from flasks by adding a trypsin solution at 1% and incubating at 37 °C for 5–10 min; Caco-2 cells were then collected, centrifuged and counted in a Bürker chamber and then seeded into 24- or 96-well plates at a concentration of 5 × 104 cells/mL for different experiments. Cells were cultured replacing the medium twice weekly, to allow their spontaneous differentiation [16].

2.8. MTT Viability Test

To ascertain any cytotoxic activity of the phenolic extracts in normal differentiated (21 days post-seeding) cells and in intestinal cancer cells (three days post-seeding), Caco-2 cell viability was assessed using an MTT assay. The cells were seeded in 96-well plates (5 × 103 cells/well in 100 μL) incubated with different concentrations (5–100 μg/mL) of methanolic extracts or to an equivalent volume of methanol for the controls, and were incubated for 24 h. At 24 h before treatment, the 10% serum supplemented medium was discarded and replaced with a medium with 2.5% serum. After incubation, the medium was removed and 100 μL of the MTT solution (5 mg/mL of MTT in PBS, 8%, in fresh serum free medium, 92%) was added and left for 4 h at 37 °C. The MTT solution was then discarded and 100 μL of DMSO was added to each well. Afterwards, the absorbance of each well was measured at 570 nm using a microplate reader (Infinite F200, Tecan, Salzburg, Austria). Cell survival was expressed as percentage of control (0 μg/mL) values.

2.9. Determination of Intracellular ROS Production

ROS production in Caco-2 cells was evaluated using the fluorescent probe H2-DCF-DA as reported by Gil et al. [17], with minor modifications. Cells were seeded in 96-well plates and once differentiated, were incubated with 10 μM of H2-DCF-DA in 100 μL of PBS for 30 min. Afterwards, H2-DCF-DA was replaced by the PBS solution containing the phenolic extract (5, 10, 25, 50 and 100 μg/mL), 30 min prior to adding tert-Butyl hydroperoxide (TBH) 2.5 mM to induce ROS release and lipid peroxidation. Control cells were treated only with PBS. The increase in cell fluorescence was determined using an Infinite F200 (Tecan, Salzburg, Austria) microplate reader at 485 and 530 nm (excitation and emission wavelengths, respectively). ROS production was monitored by reading the fluorescence emitted after 120 min of incubation. Moreover, cells treated with PAFE were analyzed as representative samples using a ZOETM fluorescent cell imager (Bio-Rad Laboratories, Hercules, CA, U.S.A.), using the green channel with a blue LED (excitation: 480/17 nm; emission: 517/23 nm) to the size of 100 μm.

2.10. Statistical Analyses

Data were analyzed by means of software GraphPad Prism 5 (GraphPad software, San Diego, CA, USA), using one-way analysis of variance (ANOVA) followed by post-hoc Tukey’s test. A level of p < 0.05 was considered statistically significant.

3. Results

3.1. Identification of Metabolites in Physalis alkekengi L. Fruit and Calyx Extracts by LC-ESI/LTQOrbitrap/MS and LC-ESI-LTQOrbitrap/MS/MS Analysis

Metabolite profiles of hydroalcoholic and decoction extracts obtained from Physalis alkekengi L. fruit and calyx analyzed by LC-ESI-LTQ-MS/MS and investigated by Xcalibur Software, and the total ion current LC-MS chromatograms for the two different extraction methods applied to the two different parts of the plant under investigation, are reported in Figure 1. Accurate mass measurement (ppm ≤ 5) and fragmentation experiments (MS/MS) obtained under data dependent scan mode, associated with research in specific databases on spectral data for natural substances as KNApSAcK (www.knapsackfamily.com) on 12 November 2023 and in the literature for the species Physalis alkekengi L., allowed the putative identification of 58 secondary metabolites mainly belonging to flavonoid glycosides, steroids, phenolic glycosides, coumarins, phenolic acids, carotenoids, and N-containing compounds (Table 1). Flavonoids were among the major components of Physalis alkekengi L., with a common C6–C3–C6 tricyclic skeleton [18]. Compounds 1, 7, 11, 16, 18, 19, 21, 23, 24 were identified as flavonoid glycosides, occurring both in the fruit and calyx of Physalis alkekengi L., and were observed as the major compounds when the extraction method was decoction. Flavonoid aglycons quercetin (37) and luteolin (41) were recovered in fruit and calyx by using both extraction methods, while apigenin (50) was identified in the calyx and not the fruit. The interesting presence of flavonoids in the fruit and calyx of Physalis alkekengi L. was reviewed in 2018 by Li et al. [6].
In addition to the presence of organic acids, p-coumaric acid (2), quinic acid (6), 1,3-dicaffeoylquinic acid (9), 4,5-dicaffeoylquinic acid (12), 3-caffeoylquinic acid (22), 5-caffeoylquinic acid (43), and caffeic acid (25) were revealed together with their derivatives, as suberic acid (4), feruloyl dihexoside (5), feruloyl hexoside (32), caffeoylglycerol (36), and coumaroyl dihexoside (2 isomers, 45 and 46). Coumaroyl and feruloyl glycosides largely contribute to the antioxidant activity of diverse plant materials [19].
The presence of these compounds is probably involved in the antioxidant activity of the calyx and fruit of Physalis alkekengi L. In addition, in the Physalis alkekengi L. fruit and calyx, in agreement with Wen and co-authors [20], these compounds were found in the final part of the chromatograms of hydroalcoholic extracts compounds identified as carotenoids (52, 53, 54, 55, 56, 57, 58). The same compounds were not identified in the decoction extracts, due to the polarity of the extraction solvent (water).
Table 1. Metabolite profiling of yellow Physalys alkekengi L. fruit and calyx by LC-ESI-LTQ-Orbitrap-MS/MS. PACE (Physalis alkekengi L. calyx hydroalcoholic extract), PAFE (Physalis alkekengi L. fruit hydroalcoholic extract) PACD (Physalis alkekengi L. calyx decoction extract), PAFD (Physalis alkekengi L. fruit decoction extract).
Table 1. Metabolite profiling of yellow Physalys alkekengi L. fruit and calyx by LC-ESI-LTQ-Orbitrap-MS/MS. PACE (Physalis alkekengi L. calyx hydroalcoholic extract), PAFE (Physalis alkekengi L. fruit hydroalcoholic extract) PACD (Physalis alkekengi L. calyx decoction extract), PAFD (Physalis alkekengi L. fruit decoction extract).
RtIdentityMolecular
Formula
[M + H]+ΔppmMS/MSReferencesPACEPAFEPACDPAFD
17.81quercetin 3-O-rutinoside-7-O-glucosideC33H40O20773.21623.5611; 449; 303;Mass Bankndndxx
28.56p-coumaric acidC9H6O3163.0391−2.5165; 147[21]ndndxx
38.57hydroxycoumarinC9H6O3163.03950.9163; 145[21]xxxx
48.92suberic acidC8H13O4175.0816−1.8157; 139[21]ndndxx
59.15feruloyl dihexoside (2)C22H30O14519.17130.7195[22]xxxx
69.25quinic acidC7H12O6193.05562.1135; 117[21]xxxx
710.96rutinC27H30O16611.1534−0.6465; 303[23]xxxx
811.265,6α-epoxy-physalin CC28H30O10527.1937−1.5509; 499; 483[21]xndxnd
911.491,3-dicaffeoylquinic acidC25H24O12517.13440.6193[21]xxxx
1011.684-hydroxyneophysalin A isomerC28H30O11543.1866−0.5511; 497; 449[21]xndndnd
1111.82luteolin-rutinosideC27H30O15595.1584−0.7449; 287Mass Bankxxxx
1212.664,5-dicaffeoylquinic acidC25H24O12517.13441284; 269; 141[21]xxxx
1313.13L-tryptophanC11H12N2O2205.07072.3171; 147[22]xxxx
1414.914-hydroxyneophysalin A isomerC28H29O11541.1713−1513; 497; 465[21]xndxnd
1515.594-hydroxyneophysalin AC28H30O11541.18660.5513; 497; 465[21]xxxx
1615.95isolariciresinol O-glucosideC26H34O11523.21791505; 184[21]xxxx
1715.965,6-O-epoxy-physalin C isomerC28H30O10527.1937−1.5509; 499; 483[21]xndxx
1816.14quercetin 3-O-glucosideC21H20O12465.1033−0.9303; 285[21]xxxx
1916.67luteolin 7-O-glucosideC21H20O11449.1083−0.9287; 285[21]xxxx
2016.915,6-O-epoxy-physalin CC28H30O10527.19170.8509; 499; 483[21]xndxnd
2116.99quercetin O-rhamnosideC21H20O11449.1083−0.9303; 287[21]xxxx
2217.333-caffeoylquinic acidC16H18O9355.08720.5337; 284[21]xxxx
2317.92vitexinC21H20O10433.11341.2433; 417; 379[21]xxxx
2417.92apigenin O-glucosideC21H20O10433.11341.2271[21]xxxx
2517.94caffeic acidC9H8O4181.0500−2.2163[21]xxxx
2618.214-hydroxyneophysalin A isomerC28H30O11541.18660.8284; 198; 141[21]xxxx
2719.824,7-didehydro-7-deoxyneophysalin AC28H28O9509.18110.7497; 465[21]xxxnd
2819.82N,N′-dicaffeoylspermidine (1)C25H31N3O6470.22110.5333; 289[22]xxxx
2919.865-ethoxy-6-hydroxy-5,6-dihydrophysalin BC30H36O11573.23350.7555; 523[21]xndxnd
3019.97physalin ZC28H30O10527.19170.6509; 499; 483[21]xxxx
3119.97physalin AC28H30O10527.19170.8509; 499; 483[21]xxxx
3219.97feruloyl hexosideC16H20O9355.10401.8195; 179[22]xxxx
3320.71physalin CC28H30O9511.19680.6493; 481; 423[21]xxxx
3420.73physalin OC28H32O10529.19170.4511; 483[21]ndndxnd
3521.85physalin NC28H30O10527.19170.6509; 499; 483[21]xxxx
3621.87caffeoylglycerolC12H14O6255.07120.8214[21]ndxndx
3721.9quercetinC15H10O7302.23030.7257; 229Mass Bank
3821.99isophysalin BC28H30O9511.19680.6493; 467; 423[21]xxxnd
3923.28N,N′-dicaffeoylspermidine (2)C25H31N3O6470.22110.5333; 289[22]xxxx
4023.55sonchuionoside CC19H30O8387.2018−0.7331; 198[21]xxxx
4123.9luteolinC15H10O6285.03831.3153; 135[21]xxxx
4223.914,7-didehydro-physalin BC28H28O9509.18110.7497; 465[21]xxxnd
4324.025-caffeoylquinic acidC16H18O9355.08711.5198; 141; 109[21]xxxx
4424.337β-hydroxy-25,27-didehydrophysalin LC28H30O10527.19170.6509; 499; 483[21]xxxx
4524.81coumaroyl dihexosideC21H28O13489.16081.7327[22]xxxx
4625.46coumaroyl hexosideC15H18O8327.10792.1188; 165[22]xxxx
4726.144,7-didehydrophysalin BC28H28O9509.18110.7509; 499; 483[21]xxxnd
4826.41physalin MC28H32O9513.21240.8495; 451; 135[21]xndxx
4926.824,7-didehydroneophysalin BC28H28O9509.18110.7497; 465[21]xxxnd
5028.41apigeninC15H10O5271.06061.6153[21]xndxnd
5128.45physalin BC28H30O9511.19680.6493; 467; 423[21]xxxnd
5230.63(all-E)-antheraxanthinC40H56O3585.43071.3567; 549; 493[20]xxndnd
5330.91zeaxanthin myristateC54H82O3779.63420.6761; 687; 669; 551; 533[20]xxndnd
5431.18(13Z)-β-caroteneC40H56537.44601.3445; 347; 281; 255[20]xxndnd
5531.18(all-E)-α-caroteneC40H56537.44601.3481; 399[20]xxndnd
5631.46(all-E)-luteinC40H56O2569.43581.4551[20]xxndnd
5732.49(15Z)-zeaxanthinC40H56O2569.43581.4551[20]xxndnd
5833.96(all-E)-zeaxanthinC40H56O2569.43581.4551[20]xxndnd
nd: not determined.
Metabolites characteristic of the genus Physalis are a group of steroids bearing 13, 14-seco-16,24-cycloergostane skeletons, named as physalins. Physalin A (31), isolated from the leaves of P. alkekengi var. franchetii in 1969, was the first member of this group [24]. Subsequently, a series of physalins with complex and diverse structures have been reported from the genus Physalis [6,25,26].
In recent decades, about fifty physalins have been identified and reported to exert anti-inflammatory, antimicrobial, anti-diabetes, cytotoxic, and quinone reductase induction activities [27,28].
The present metabolomics approach allowed the putative identification of 21 compounds pertaining to this group of metabolites (8, 10, 14, 15, 17, 20, 26, 27, 29, 30, 31, 33, 34, 35, 38, 42, 44, 47, 48, 49, 51), showing that these compounds have the ability to give good [M + H]+ ions for LC-MS identification. Until now they have been identified mainly by LC-MS in negative ionization mode [25].
Other identified compounds in the metabolite profiling approach were metabolites pertaining to various different classes: coumarins (3), amino acids (13), lignans (16), nitrogen containing compounds (39), terpene glycosides (40); all compounds that have been reported previously in genus Physalis [6].
Although a quantitative targeted approach was not performed due to the unavailability of standard compounds, by visual comparison of profiles it was possible to note substancial differences in the fruit and calyx profiles, but even more evident were differences in extracts obtained with different extraction modes. Observing Figure 1, it is easy to evaluate that in decoctions, the most evident metabolites were those eluted at retention times between 4 and 18 min. Comparing this evidence with the table of characterization, these compounds were phenolic acids and flavonoids. On the other hand, physalins start to be eluted after 18 min. Physalins were identified in the decocted extract too, but in very low amounts. This finding means that decoction is an extraction method that permits an extract enriched in polyphenols—which are highly involved in antioxidant activity—with a lower amount of potentially toxic physalins.

3.2. Antioxidant Activity Evaluated by Spectrophotometric Assays

Plants of the genus Physalis are widely used as medicinal plants for the treatment of asthma, dermatitis, and rheumatism, and to prevent cancer [29]. Given the high content of compounds with antioxidant activity such as physalins and glicosylated flavonoids, the antioxidant activities of Physalis alkekengi L. extracts were investigated by spectrophotometric assays (Table 2).
The Physalis alkekengi L. extracts tested showed good antioxidant activity, regardless of the type of extraction performed, confirming that both hydroalcoholic extraction and decoction allow the extraction of compounds with antioxidant activity. Data obtained for the samples were compared with the positive controls, quercetin 3-O-glucoside for TEAC assay and vitamin C for DPPH assay. However, the decoctions were slightly more active than the hydroalcoholic extracts, possibly due to a higher content of extracted glycosylated flavonoids, as indicated by the metabolite profiling performed on the samples.
On the other hand, as far as the analyzed part of the plant is concerned, the calyx extracts were certainly more active than those derived from fruit. In fact, calyx extracts reached TEAC values (equivalent antioxidant capacity in Trolox expressed in mg/mL) of 2.382 for PACD and 1.749 for PACE and, respectively, an IC50 for DPPH (expressed in mg/mL) of 0.017 for PACD and 0.033 for PACE.
The finding was in agreement with antioxidant activity measured for Physalis alkekengi fruit extract by Vicas et al. (2020) by DPPH test, and Liu et al. (2023) by ABTS and DPPH chemical tests [30,31].

3.3. Cell Viability

The possible cytotoxic effect of the four extracts was assessed on cancer or normal differentiated Caco-2 cells at concentrations ranging from 5 to 100 μg/mL using the MTT assay. Figure 2 summarizes the cell viability results on differentiated cells. As shown, the tested extracts did not cause a decrease in the cellular viability of Caco-2 cells, showing results of around 100% of the control (0 μL extract/mL) with the exception of the concentration 100 μg/mL, which resulted as being significantly cytotoxic for all four extracts. The same treatment was also carried out on non-differentiated cancer cells, to explore the anti-tumoral and anti-proliferative potential of these extracts (Figure 3).
It was observed how extracts derived from the Physalis alkekengi L. fruit extract and decoction were significantly cytotoxic in Caco-2 starting from 25 μg/mL, while the fruit decoction extract was toxic only at the highest concentration tested. Among the extracts tested, the most effective in exerting toxic effects on Caco-2 cells was the one derived from calyx decoction, which was found to be significantly toxic starting from the minimum concentration tested (5 μg/mL). Given the results obtained in normal cells, we decided to use 50 μg/mL as the maximum concentration, being the the highest non-toxic one, in the subsequent experiments.

3.4. ROS Production

The ability of the four different extracts to scavenge reactive species was also tested in Caco-2 cell cultures (Figure 4).
Determination of intracellular ROS production and subsequent oxidative damage to cell membranes was induced by TBH at the concentration of 2.5 mM. In cells pretreated with all four phenolic extracts, inhibition of ROS formation was observed, as indicated by the lower emission of fluorescence. This inhibition was observed starting from a concentration of 10 μg/mL in the extracts derived from the Physalis alkekengi L. fruit (p < 0.05), while in those derived from calyces, minimum concentrations of 25 μg/mL for the decoction and 50 μg/mL for the extract were needed to observe a significant inhibition against TBH-induced peroxidation. To better highlight the antioxidant action of the extracts, we captured as a representative example the fluorescence emitted by cells treated with fruit extract by ZOETM, as can be observed in Figure 5, in which it is possible to more clearly see the inhibition of ROS production.

4. Discussion

The fruit and calyx from Physalis alkekengi L. are considered to be the only edible parts of the plants. The interesting results show the possibility of using the fruit and calyx for the preparation of tea infusion. To date, a few chemical investigations have been performed on this cultivar, mainly focusing on the analysis of methanol extracts. Due to the increasing use of Physalis as a food supplement and starting material for tea preparation, in this study a comprehensive analysis of green extracts was performed.
The comprehensive metabolomics analysis performed by LC-LTQ-Orbitrap-MS and LC-LTQ-Orbitrap-MS/MS showed no relevant differences in the composition of calyx and fruit. On the other hand, the extracts obtained by decoction showed some differences with respect to EtOH:H2O extracts: in fact, they presented the same composition with regard to flavonoids, phenylpropanoids, organic acids, but were characterized by a low amount of physalins, and they were not characterized by the presence of carotenoids, identified in hydroalcoholic extracts. Previous studies have found that carotenoids in Physalis alkekengi L. fruit have good antioxidant effects and safety [32].
The metabolomics approach allowed the identification of 58 compounds confirming the data reported in different previous phytochemical studies, extensively reviewed in 2018 by Li et al. [6].
Comparison of antioxidant activity of calyx and fruit extracts shown in the preliminary chemical tests revealed higher activity for the calyx.
Despite the absence of carotenoids, calyx extract obtained by decoction showed a higher antioxidant activity in both TEAC and DPPH assays, due to the presence of a higher content of flavonoid glycosides; in fact, reducing and free radical scavenging abilities are always positively correlated with total flavonoids content.
The cytotoxicity studies on intestinal Caco-2 cancer cells showed a difference between fruit and calyx, with major toxicity induced by calyx extracts and significantly less toxicity for extracts obtained by decoction. This may be due to the different composition in compounds that may exert higher cytotoxic effects or can be better absorbed by intestinal cancer cells [33]. It is notable that this toxicity towards cancer cells did not occur, with the exception of the highest concentration (100 µg/mL), in normal intestinal cells, suggesting a safety profile in regard to all extracts tested.
Regarding the inhibition of ROS production, in addition to the better activity of the extracts obtained by decoction with respect to the hydroalcoholic extracts, we observed the best activity for decoction extract when derived from fruit. In this specific case, we can speculate that probably in fruit, in addition to flavonoids, we may find the presence of polysaccharides, that participate in the scavenging capacity with synergistic effect when the antioxidant activity is measured on cells [34]. The activity carried out by the antioxidant compounds present in Physalis alkekengi L. extracts may merely be a free radical scavenging action, but also, modulatory effects towards redox-sensitive pathways which regulate the redox state of the cell may occur [35]. In particular, it has been widely observed how some polyphenols such as quercetin or cinnamic acid derivatives are able to modulate at intestinal level the MAP kinases signaling pathway and regulate the expression of Nrf-2, a transcription factor that induces the expression of antioxidant enzymes such as heme oxygenase (HO)-1, NAD(P)H:quinone oxidoreductase (NQO1), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) [29,36]. Furthermore, it must be considered that many of the compounds present in the extracts can easily be absorbed and metabolized by intestinal cells, forming a pool of equally active phase I/II metabolites. These metabolites can act as primary antioxidants, but are also able to modulate signaling pathways and induce the expression of antioxidant enzymes [37].
In particular, physalins are characterized by fast absorption, wide distribution, and rapid excretion. Our findings indicated that physalins are extremely unstable and have low bioavailability in the intestine, which reduces their long-term toxicity. On the other hand, it should be noted that few studies have investigated the pharmacokinetics of extracts and active compounds involved in antioxidant activity, i.e., flavonoids. Consequently, further pharmacokinetics studies in the laboratory and clinic analyses should be performed for a comprehensive approach to the use of Physalis alkekengi for its antioxidant effect in therapy [7].

5. Conclusions

An LC-ESI-LTQ/OrbitrapMS and LC-ESI-LTQ/OrbitrapMS/MS metabolomics approach allowed the characterization of metabolites present in different parts of Physalis alkekengi L. The preparation of two different types of extraction—hydroalcoholic extraction and decoction—showed that the highest detection of metabolites was obtained through hydroethanol extraction, with the putative identification of 58 metabolites. Subsequently, through preliminary spectrophotometric assays and then with cell studies, the antioxidant activity of various Physalis alkekengi L. extracts was evaluated. It was found that Physalis alkekengi L. extracts are a good source of metabolites with various biological activities, in particular, antioxidant activity capable of reducing the production of free radicals in Caco-2 cell-line.
An interesting result was the antioxidant activity of extracts obtained by decoction, that allows the suggestion of a potential beneficial use of Physalis alkekengi L. fruit and calyx in the preparation of tea infusions.
The innovative aspect of the present study was connected with the preparation of green extracts. For the first time, an integrated approach (metabolomics approach and antioxidant evaluation) was applied to the study of Physalys alkekengi’s decoctions, the most often used green extraction method in herbal preparations.

Author Contributions

Conceptualization, M.D., P.M. and S.P.; data curation, M.A.C. and G.S.; formal analysis, M.A.C. and G.S.; funding acquisition, S.P.; investigation, M.D., P.M. and S.P.; methodology, M.A.C., G.S., C.I.G.T. and P.M.; project administration, P.M. and S.P.; resources, C.I.G.T. and S.P.; software, M.A.C. and G.S.; supervision, M.D. and S.P.; validation, M.A.C. and G.S.; visualization, C.I.G.T. and P.M.; writing—original draft, M.A.C., G.S. and P.M.; writing—review and editing, M.D., C.I.G.T., P.M. and S.P. 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

The data used to support the findings of this study can be made available by the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liang, L.; Li, C.; Wang, Y.; Yue, Y.; Zhang, H.; Yang, M.; Cao, X.; Zhao, M.; Du, J.; Peng, M.; et al. Physalis alkekengi L. var. franchetii (Mast.) Makino: A review of the pharmacognosy, chemical constituents, pharmacological effects, quality control, and applications. Phytomedicine 2022, 105, 154328. [Google Scholar] [CrossRef] [PubMed]
  2. Popova, V.; Petkova, Z.; Mazova, N.; Ivanova, T.; Petkova, N.; Stoyanova, M.; Stoyanova, A.; Ercisli, S.; Okcu, Z.; Skrovankova, S.; et al. Chemical Composition Assessment of Structural Parts (Seeds, Peel, Pulp) of Physalis alkekengi L. Fruits. Molecules 2022, 27, 5787. [Google Scholar] [CrossRef] [PubMed]
  3. Popova, V.; Ivanova, T.; Stoyanova, M.; Mazova, N.; Dimitrova, I.; Stoyanova, A.; Assouguem, A.; Kara, M.; Topçu, H.; Farah, A.; et al. Phytochemical analysis of leaves and stems of Physalis alkekengi L. (Solanaceae). Open Chem. 2022, 20, 1292–1303. [Google Scholar] [CrossRef]
  4. Zheng, Y.; Chen, Y.; Ren, Y.; Luan, L.; Wu, Y. Quantitative and transformation product analysis of major active physalins from Physalis alkekengi var. franchetii (Chinese lantern) using ultraperformance liquid chromatography with electrospray ionisation tandem mass spectrometry and time-of-flight mass spectrometry. Phytochem. Anal. 2012, 23, 337–344. [Google Scholar] [CrossRef] [PubMed]
  5. Bahmani, M.; Rafieian-kopaei, M.; Naghdi, N.; Mozaffari Nejad, A.S.; Afsordeh, O. Physalis alkekengi: A review of Its Therapeutic Effects. J. Chem. Pharm. Sci. 2016, 9, 1472–1485. [Google Scholar]
  6. Li, A.-L.; Chen, B.-J.; Li, G.-H.; Zhou, M.-X.; Li, Y.-R.; Ren, D.-M.; Lou, H.-X.; Wang, X.-N.; Shen, T. Physalis alkekengi L. var. franchetii (Mast.) Makino: An ethnomedical, phytochemical and pharmacological review. J. Ethnopharmacol. 2018, 210, 260–274. [Google Scholar] [CrossRef] [PubMed]
  7. Yang, J.; Sun, Y.; Cao, F.; Yang, B.; Kuang, H. Natural Products from Physalis alkekengi L. var. franchetii (Mast.) Makino: A Review on Their Structural Analysis, Quality Control, Pharmacology, and Pharmacokinetics. Molecules 2022, 27, 695. [Google Scholar] [CrossRef] [PubMed]
  8. Popova, V.; Stoyanova, A.; Mazova, N. Phytochemical composition and biological activity of Physalis spp.: A mini-review. Food Sci. Appl. Biotechnol. 2020, 3, 56. [Google Scholar] [CrossRef]
  9. Teng, Y.A.-O.; Yuan, Q.A.-O.X.; Wu, Y.; Wu, S.; Su, J.; Zhang, P.; Zhang, Y.A.-O. Research on the Chemical Constituents against Alzheimer’s Disease of the Fruits of Physalis alkekengi L. var. franchetii (Mast.) Makino. Chem. Biodivers. 2023, 20, e202301075. [Google Scholar] [CrossRef]
  10. El-Ghorab, A.; Ramadana, M.; El-Ghoraba, A.; Ghanemb, K. Volatile compounds, antioxidants, and anticancer activities of Cape gooseberry fruit (Physalis peruviana L.): An in-vitro study. J. Arab. Soc. Med. Res. 2015, 10, 56–64. [Google Scholar] [CrossRef]
  11. Castro, J.; Ocampo, Y.; Franco, L. Cape Gooseberry [Physalis peruviana L.] Calyces Ameliorate TNBS Acid-induced Colitis in Rats. J. Crohns Colitis 2015, 9, 1004–1015. [Google Scholar] [CrossRef] [PubMed]
  12. Chang, L.C.; Sang-Ngern, M.; Pezzuto, J.M.; Ma, C. The Daniel K. Inouye College of Pharmacy Scripts: Poha Berry (Physalis peruviana) with Potential Anti-inflammatory and Cancer Prevention Activities. Hawai’i J. Med. Public Health 2016, 75, 353. [Google Scholar]
  13. Etzbach, L.; Pfeiffer, A.; Weber, F.; Schieber, A. Characterization of carotenoid profiles in goldenberry (Physalis peruviana L.) fruits at various ripening stages and in different plant tissues by HPLC-DAD-APCI-MS(n). Food Chem. 2018, 245, 508–517. [Google Scholar] [CrossRef] [PubMed]
  14. Crescenzi, M.A.; D’Urso, G.; Piacente, S.; Montoro, P. LC-ESI/LTQOrbitrap/MS Metabolomic Analysis of Fennel Waste (Foeniculum vulgare Mill.) as a Byproduct Rich in Bioactive Compounds. Foods 2021, 10, 1893. [Google Scholar] [CrossRef] [PubMed]
  15. Santoru, M.L.; Piras, C.; Murgia, F.; Spada, M.; Tronci, L.; Leoni, V.P.; Serreli, G.; Deiana, M.; Atzori, L. Modulatory Effect of Nicotinic Acid on the Metabolism of Caco-2 Cells Exposed to IL-1β and LPS. Metabolites 2020, 10, 204. [Google Scholar] [CrossRef] [PubMed]
  16. Deiana, M.; Montoro, P.; Jerković, I.; Atzeri, A.; Marijanović, Z.; Serreli, G.; Piacente, S.; Tuberoso, C.I.G. First characterization of Pompia intrea candied fruit: The headspace chemical profile, polar extract composition and its biological activities. Food Res. Int. 2019, 120, 620–630. [Google Scholar] [CrossRef] [PubMed]
  17. Gil, K.A.; Nowicka, P.; Wojdyło, A.; Serreli, G.; Deiana, M.; Tuberoso, C.I.G. Antioxidant Activity and Inhibition of Digestive Enzymes of New Strawberry Tree Fruit/Apple Smoothies. Antioxidants 2023, 12, 805. [Google Scholar] [CrossRef] [PubMed]
  18. Qiu, L.; Jiang, Z.H.; Liu, H.X.; Chen, L.X.; Qu, G.X.; Qiu, F. Flavonoid glycosides of the calyx Physalis. J. Shenyang Pharm. Univ. 2007, 24, 744–747. [Google Scholar]
  19. Kylli, P.; Nousiainen, P.; Biely, P.; Sipilä, J.; Tenkanen, M.; Heinonen, M. Antioxidant potential of hydroxycinnamic acid glycoside esters. J. Agric. Food Chem. 2008, 56, 4797–4805. [Google Scholar] [CrossRef]
  20. Wen, X.; Hempel, J.; Schweiggert, R.M.; Ni, Y.; Carle, R. Carotenoids and Carotenoid Esters of Red and Yellow Physalis (Physalis alkekengi L. and P. pubescens L.) Fruits and Calyces. J. Agric. Food Chem. 2017, 65, 6140–6151. [Google Scholar] [CrossRef]
  21. Zhang, Q.; Hu, X.F.; Xin, M.M.; Liu, H.B.; Sun, L.J.; Morris-Natschke, S.L.; Chen, Y.; Lee, K.H. Antidiabetic potential of the ethyl acetate extract of Physalis alkekengi and chemical constituents identified by HPLC-ESI-QTOF-MS. J. Ethnopharmacol. 2018, 225, 202–210. [Google Scholar] [CrossRef]
  22. Wen, X.; Erşan, S.; Li, M.; Wang, K.; Steingass, C.B.; Schweiggert, R.M.; Ni, Y.; Carle, R. Physicochemical characteristics and phytochemical profiles of yellow and red Physalis (Physalis alkekengi L. and P. pubescens L.) fruits cultivated in China. Food Res. Int. 2019, 120, 389–398. [Google Scholar] [CrossRef]
  23. Liu, M.; Qiu, Z.; Zhao, X.; Sun, L.; Wang, L.; Ren, X.; Deng, Y. Assessing the Quality of Calyx of Physalis alkekengi L. var. franchetii Based on Quantitative Analysis of Q-Marker Combined with Chemometrics and Machine Learning Algorithms. J. Chem. 2021, 2021, 8502929. [Google Scholar] [CrossRef]
  24. Matsuura, T.; Kawai, M.; Nakashima, R.; Butsugan, Y. Bitter principles of Physalis Alkekengi var Francheti: Structure of physalin a. Tetrahedron Lett. 1969, 10, 1083–1086. [Google Scholar] [CrossRef]
  25. Huang, C.; Xu, Q.; Chen, C.; Song, C.; Xu, Y.; Xiang, Y.; Feng, Y.; Ouyang, H.; Zhang, Y.; Jiang, H. The rapid discovery and identification of physalins in the calyx of Physalis alkekengi L. var. franchetii (Mast.) Makino using ultra-high performance liquid chromatography–quadrupole time of flight tandem mass spectrometry together with a novel three-step data mining strategy. J. Chromatogr. A 2014, 1361, 139–152. [Google Scholar]
  26. Qiu, L.; Zhao, F.; Jiang, Z.H.; Chen, L.X.; Zhao, Q.; Liu, H.X.; Yao, X.S.; Qiu, F. Steroids and flavonoids from Physalis alkekengi var. franchetii and their inhibitory effects on nitric oxide production. J. Nat. Prod. 2008, 71, 642–646. [Google Scholar] [CrossRef]
  27. Shu, Z.; Xing, N.; Wang, Q.; Li, X.; Xu, B.; Li, Z.; Kuang, H. Antibacterial and Anti-Inflammatory Activities of Physalis Alkekengi var. franchetii and Its Main Constituents. Evid. Based Complement. Altern. Med. 2016, 2016, 4359394. [Google Scholar] [CrossRef]
  28. Zhang, W.N.; Tong, W.Y. Chemical Constituents and Biological Activities of Plants from the Genus Physalis. Chem. Biodivers. 2016, 13, 48–65. [Google Scholar] [CrossRef]
  29. He, B.W.; Wang, F.F.; Qu, L.P. Anti-Inflammatory and Antioxidant Properties of Physalis alkekengi L. Extracts In Vitro and In Vivo: Potential Application for Skin Care. Evid. Based Complement. Altern. Med. 2022, 2022, 7579572. [Google Scholar] [CrossRef]
  30. Vicas, L.G.; Jurca, T.; Baldea, I.; Filip, G.A.; Olteanu, D.; Clichici, S.V.; Pallag, A.; Marian, E.; Micle, O.; Crivii, C.B.; et al. Physalis alkekengi L. Extract Reduces the Oxidative Stress, Inflammation and Apoptosis in Endothelial Vascular Cells Exposed to Hyperglycemia. Molecules 2020, 25, 3747. [Google Scholar] [CrossRef]
  31. Liu, M.; Liu, G.; Ma, Z.; Wen, J.L.; Liu, Y.; Sun, L.; Ren, X. A comprehensive quality evaluation method of different medicinal parts of Physalis Calyx seu Fructus by fingerprints, chemometrics, antioxidant activity, network pharmacology and molecular docking. Biomed. Chromatogr. 2023, 37, e5701. [Google Scholar] [CrossRef] [PubMed]
  32. Chedea, V.S.; Pintea, A.; Bunea, A.; Braicu, C.; Stanila, A.; Socaciu, C. Physalis alkekengi carotenoidic extract inhibitor of soybean lipoxygenase-1 activity. Biomed Res. Int. 2014, 2014, 589168. [Google Scholar] [CrossRef] [PubMed]
  33. Sak, K. Cytotoxicity of dietary flavonoids on different human cancer types. Pharmacogn. Rev. 2014, 8, 122–146. [Google Scholar] [CrossRef] [PubMed]
  34. Tong, H.; Zhu, M.E.I.; Feng, K.A.I.; Sun, L. Purification, characterization and in vitro antioxidant activities of polysaccharide fractions isolated from the fruits of Physalis alkekengi L. J. Food Biochem. 2011, 35, 524–541. [Google Scholar] [CrossRef]
  35. Rahman, M.M.; Rahaman, M.S.; Islam, M.R.; Rahman, F.; Mithi, F.M.; Alqahtani, T.; Almikhlafi, M.A.; Alghamdi, S.Q.; Alruwaili, A.S.; Hossain, M.S.; et al. Role of Phenolic Compounds in Human Disease: Current Knowledge and Future Prospects. Molecules 2022, 27, 233. [Google Scholar] [CrossRef]
  36. Sun, L.; Xu, G.; Dong, Y.; Li, M.; Yang, L.; Lu, W. Quercetin Protects Against Lipopolysaccharide-Induced Intestinal Oxidative Stress in Broiler Chickens through Activation of Nrf2 Pathway. Molecules 2020, 25, 1053. [Google Scholar] [CrossRef]
  37. Boronat, A.; Rodriguez-Morató, J.; Serreli, G.; Fitó, M.; Tyndale, R.F.; Deiana, M.; de la Torre, R. Contribution of Biotransformations Carried Out by the Microbiota, Drug-Metabolizing Enzymes, and Transport Proteins to the Biological Activities of Phytochemicals Found in the Diet. Adv. Nutr. 2021, 12, 2172–2189. [Google Scholar] [CrossRef]
Figure 1. Positive ion mode profiles of Physalis alkekengi L. ethanolic extracts obtained by LC-ESI/LTQ/Orbitrap MS: PAFE (Physalis alkekengi L. fruit ethanolic extraction), PACE (Physalis alkekengi L. calyx ethanolic extraction), PAFD (Physalis alkekengi L. fruit decoction), and PACD (Physalis alkekengi L. calyx decoction).
Figure 1. Positive ion mode profiles of Physalis alkekengi L. ethanolic extracts obtained by LC-ESI/LTQ/Orbitrap MS: PAFE (Physalis alkekengi L. fruit ethanolic extraction), PACE (Physalis alkekengi L. calyx ethanolic extraction), PAFD (Physalis alkekengi L. fruit decoction), and PACD (Physalis alkekengi L. calyx decoction).
Antioxidants 12 02101 g001
Figure 2. Percentage of cell viability compared with the control (0 μg/mL, 100% viable cells) of differentiated Caco-2 cells incubated for 48 h with different concentrations (5–100 μg/mL) of phenolic extracts derived from fruit extract (PAFE), fruit decoction (PAFD), calyx extract (PACE) and calyx decoction (PACD). Each column represents the mean ± SD of the independent experiments (n = 16). *** = p < 0.001 vs. 0 μg/mL; ** = p < 0.01 vs. 0 μg/mL; * = p < 0.05 vs. 0 μg/mL.
Figure 2. Percentage of cell viability compared with the control (0 μg/mL, 100% viable cells) of differentiated Caco-2 cells incubated for 48 h with different concentrations (5–100 μg/mL) of phenolic extracts derived from fruit extract (PAFE), fruit decoction (PAFD), calyx extract (PACE) and calyx decoction (PACD). Each column represents the mean ± SD of the independent experiments (n = 16). *** = p < 0.001 vs. 0 μg/mL; ** = p < 0.01 vs. 0 μg/mL; * = p < 0.05 vs. 0 μg/mL.
Antioxidants 12 02101 g002
Figure 3. Percentage of cell viability compared with the control (0 μg/mL, 100% viable cells) of cancer Caco-2 cells incubated for 48 h with different concentrations (5–100 μg/mL) of phenolic extracts derived from fruit extract (PAFE), fruit decoction (PAFD), calyx extract (PACE) and calyx decoction (PACD). Each column represents the mean ± SD of the independent experiments (n = 16). *** = p < 0.001 vs. 0 μg/mL; ** = p < 0.01 vs. 0 μg/mL; * = p < 0.05 vs. 0 μg/mL.
Figure 3. Percentage of cell viability compared with the control (0 μg/mL, 100% viable cells) of cancer Caco-2 cells incubated for 48 h with different concentrations (5–100 μg/mL) of phenolic extracts derived from fruit extract (PAFE), fruit decoction (PAFD), calyx extract (PACE) and calyx decoction (PACD). Each column represents the mean ± SD of the independent experiments (n = 16). *** = p < 0.001 vs. 0 μg/mL; ** = p < 0.01 vs. 0 μg/mL; * = p < 0.05 vs. 0 μg/mL.
Antioxidants 12 02101 g003
Figure 4. Determination of intracellular ROS production. ROS level, visualized as H2-DCF-DA fluorescence and expressed as a % of the control samples (non-oxidized nor pretreated samples), in Caco-2 after 120 min incubation with TBH 2.5 mM and pretreated with the four extracts (5–50 μg/mL). °°° = p < 0.001 vs. control; *** = p < 0.001 vs. TBH 2.5 mM; ** = p < 0.01 vs. TBH 2.5 mM; * = p < 0.05 vs. TBH 2.5 mM.
Figure 4. Determination of intracellular ROS production. ROS level, visualized as H2-DCF-DA fluorescence and expressed as a % of the control samples (non-oxidized nor pretreated samples), in Caco-2 after 120 min incubation with TBH 2.5 mM and pretreated with the four extracts (5–50 μg/mL). °°° = p < 0.001 vs. control; *** = p < 0.001 vs. TBH 2.5 mM; ** = p < 0.01 vs. TBH 2.5 mM; * = p < 0.05 vs. TBH 2.5 mM.
Antioxidants 12 02101 g004
Figure 5. ROS levels visualized as H2-DCF-DA fluorescence in differentiated Caco-2 after 30 min incubation with TBH 2.5 mM and pretreated with the phenolic extract from PAFE (30 min).
Figure 5. ROS levels visualized as H2-DCF-DA fluorescence in differentiated Caco-2 after 30 min incubation with TBH 2.5 mM and pretreated with the phenolic extract from PAFE (30 min).
Antioxidants 12 02101 g005
Table 2. Antioxidant activity of extracts of Physalis alkekengi L. evaluated by Trolox equivalent antioxidant capacity (TEAC) and DPPH• radical scavenging activity assays.
Table 2. Antioxidant activity of extracts of Physalis alkekengi L. evaluated by Trolox equivalent antioxidant capacity (TEAC) and DPPH• radical scavenging activity assays.
Physalis alkekengi L. ExtractsTEAC
[mg/mL ± SDa] b
DPPH
[IC50 (mg/mL) ± SDa]
PACD2.382 ± 0.0200.017 ± 0.003
PAFD0.720 ± 0.0040.502 ± 0.010
PACE1.749 ± 0.0100.033 ± 0.003
PAFE0.731 ± 0.0020.670 ± 0.020
Quercetin 3-O-glucoside2.426 ± 0.010/
Vitamin C/0.170 ± 0.020
SDa, standard deviation of three independent experiments; b, antioxidant activity determined by TEAC assay, and expressed as antioxidant capacity equivalent in Trolox in mg/mL. PAFE (Physalis alkekengi L. fruit ethanolic extraction), PACE (Physalis alkekengi L. calyx ethanolic extraction), PAFD (Physalis alkekengi L. fruit decoction), and PACD (Physalis alkekengi L. calyx decoction).
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

Crescenzi, M.A.; Serreli, G.; Deiana, M.; Tuberoso, C.I.G.; Montoro, P.; Piacente, S. Metabolite Profiling, through LC-ESI/LTQOrbitrap/MS Analysis, of Antioxidant Extracts from Physalis alkekengi L. Antioxidants 2023, 12, 2101. https://doi.org/10.3390/antiox12122101

AMA Style

Crescenzi MA, Serreli G, Deiana M, Tuberoso CIG, Montoro P, Piacente S. Metabolite Profiling, through LC-ESI/LTQOrbitrap/MS Analysis, of Antioxidant Extracts from Physalis alkekengi L. Antioxidants. 2023; 12(12):2101. https://doi.org/10.3390/antiox12122101

Chicago/Turabian Style

Crescenzi, Maria Assunta, Gabriele Serreli, Monica Deiana, Carlo I. G. Tuberoso, Paola Montoro, and Sonia Piacente. 2023. "Metabolite Profiling, through LC-ESI/LTQOrbitrap/MS Analysis, of Antioxidant Extracts from Physalis alkekengi L." Antioxidants 12, no. 12: 2101. https://doi.org/10.3390/antiox12122101

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