Next Article in Journal
Bar Adsorptive Microextraction Coated with Carbon-Based Phase Mixtures for Performance-Enhancement to Monitor Selected Benzotriazoles, Benzothiazoles, and Benzenesulfonamides in Environmental Water Matrices
Next Article in Special Issue
Comparative Analysis of Milk Fat Globular Membrane (MFGM) Proteome between Saudi Arabia Camelus dromedary Safra and Wadha Breeds
Previous Article in Journal
An Unexpected Reaction of Isodehydracetic Acid with Amines in the Presence of 1-Ethyl-3-(3-dimethylaminopropyl) Carbodiimide Hydrochloride Yields a New Type of β-Enaminones
Previous Article in Special Issue
Cannabis sativa L. Inflorescences from Monoecious Cultivars Grown in Central Italy: An Untargeted Chemical Characterization from Early Flowering to Ripening
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Bioaccessibility and Antioxidant Activity of Coffee Silverskin Polyphenolic Extract and Characterization of Bioactive Compounds Using UHPLC-Q-Orbitrap HRMS

1
Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Napoli, Italy
2
Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
3
Staff of UNESCO Chair on Health Education and Sustainable Development, Federico II University, 80131 Naples, Italy
*
Author to whom correspondence should be addressed.
Submission received: 14 April 2020 / Revised: 29 April 2020 / Accepted: 1 May 2020 / Published: 2 May 2020
(This article belongs to the Collection Advances in Food Chemistry)

Abstract

:
Coffee silverskin (CS), the main by-product in the coffee industry, contains a vast number of human health-related compounds, which may justify its exploitation as a functional food ingredient. This study aimed to provide a comprehensive analysis of the polyphenolic and alkaloid profile through UHPLC-Q-Orbitrap HRMS analysis. The bioaccessibility of total phenolic compounds and changes in the antioxidant activity during an in vitro gastrointestinal digestion were also evaluated through spectrophotometric tests (TPC by Folin-Ciocalteu, ABTS, DPPH, and FRAP), to elucidate their efficacy for future applications in the nutraceutical industry. Caffeoylquinic and feruloylquinic acids were the most representative polyphenols, with a mean concentration of 5.93 and 4.25 mg/g, respectively. Results showed a high content of caffeine in the analyzed CS extracts, with a mean value of 31.2 mg/g, meaning a two-fold increase when compared to coffee brews. Our findings highlighted that both the bioaccessibility and antioxidant activity of CS polyphenols significantly increased in each in vitro gastrointestinal digestion stage. In addition, the colon stage might constitute the main biological site of action of these antioxidant compounds. These results suggest that in vivo, the dietary polyphenols from CS might be metabolized by human colonic microflora, generating metabolites with a greater antioxidant activity, increasing their well-known beneficial effects.

1. Introduction

Coffee is one of the most popular and appreciated beverages around the world due to its pleasant flavor and stimulating properties [1]. The coffee industry has steadily grown in the Western countries and represents one of the most highly traded commodities [2]. According to the latest data reported by the International Coffee Organization (ICO), global coffee production reached 168.9 million of 60 kg bags in the year 2019/2020 [3]. Consequently, huge amounts of waste and by-products are generated in the coffee industry, which are often poured into the environment, contributing to soil and water pollution [4].
Currently, by-product valorization is an innovative and eco-friendly way to contribute to a sustainable development, promoting the use of alternative sources for obtaining bioactive compounds with potential market value [5]. Natural high value ingredients recovered from agro-industrial by-products, such as antioxidants, may be an economically attractive solution, which can be used effectively in nutraceutical formulations, functional foods, dietary supplements, fortified foods, or for their health-promoting properties [6].
Coffee silverskin (CS) is a thin tegument of green coffee beans and constitutes the main by-product in the coffee industry [7]. Despite the efforts made by some research groups to find an alternative use for this coffee by-product, CS remains underutilized as a raw material for industrial processes [8]. Previous studies indicated that CS contains a wide range of active molecules that may affect human health, which include alkaloids, polyphenols, and other high molecular weight compounds generated in the late stages of the Maillard reaction during the roasting process, such as melanoidins [9]. These active molecules are well recognized as powerful compounds involved in the prevention of lifestyle-related diseases [10,11,12].
Among alkaloids, caffeine is considered the most active component in coffee, as well as CS samples [13]. Scientific evidence supports a protective role of caffeine against several chronic diseases, including type 2 diabetes mellitus and Parkinson’s disease [14], attributing many beneficial effects derived from coffee consumption to the polyphenolic fraction. Dietary polyphenols are secondary metabolites present in many plants, namely in tea, coffee, fruits, vegetables, and red wine, among others [15]. An ever-expanding amount of scientific data suggests that the habitual dietary intake of polyphenols can contribute to maintaining human health, preventing from a wide range of age-related pathologies [16]. Chlorogenic acids (CGAs) represent the major polyphenols found in CS samples. CGAs are plant-derived phytochemicals with antioxidant properties [17] formed by the esterification of cinnamic acids, namely caffeic, p-coumaric, and ferulic acids with a common skeleton of quinic acid [18], which give rise to caffeoylquinic (CQA), dicaffeoylquinic acid (diCQA), p-coumaroylquinic (pCoQA), feruloylquinic (FQA), caffeoyl-feruloylquinic (CFQA), and feruloyl-caffeoylquinic acids (FCQA) [19]. Caffeoylquinic, p-coumaroylquinic, and feruloylquinic acids are some of the most important polyphenols reported in CS, as well as coffee brews. As reported by several investigations, CGA shows high bioaccessibility in humans and a wide range of important biological activities such as antioxidant and anti-inflammatory properties, becoming good candidates for medicinal applications [20,21,22]. As regards melanoidins, recent studies have also reported significant antioxidant potential due to the presence of low molecular compounds, such as CGAs, incorporated through non-covalent bonds during roasting of coffee beans [23].
According to Tagliazzucchi et al. [24] bioaccessibility represents one of the most relevant factors concerning the beneficial activity of polyphenols. Bioaccessibility is defined as the amount of a compound present in the gastrointestinal tract that may be considered available for absorption [25]. The actions of both human digestive and gut microbial enzymes represent an essential mechanism for releasing native polyphenols from components of the food matrix. Previous studies have suggested that polyphenols play a role in modulation of the gut microflora and activity, which may have positive implications in maintaining human health [26]. Campos-Vega et al. [27] investigated the bioaccessibility and antioxidant capacity of spent coffee after gastrointestinal digestion and colonic fermentation. The authors concluded that gastrointestinal digestion significantly increased the antioxidant activity compared to their respective non-digested samples. Fernandez-Gomez et al. [28] evaluated the in vitro bioaccessibility of CGAs extracted from CS samples using a simulated gastrointestinal digestion protocol, excluding the colon stage in the analysis. However, it has been reported that in the large intestine, coffee melanoidins might be metabolized by gut microbiota enzymes generating antioxidant compounds linked to them. In this line, a recent study reported that, during in vitro gastrointestinal digestion, low molecular weight compounds were released from melanoidins in the colon stage, resulting in a higher antioxidant activity compared to their respective non-digested sample [29]. On the other hand, there is a lack of information regarding the bioaccessibility of total phenolic compounds of CS extracts and changes in the antioxidant activity displayed during each stage of the gastrointestinal digestion, including the colon fermentation phase.
As far as the analytical methods are concerned, the determination of both CGAs and alkaloids in vegetal matrices is mainly based on liquid chromatography (LC) coupled to mass spectrometry (MS) [30]. In the last decade, there have been improvements in the LC technique with the development of ultra-high performance LC (UHPLC), which has led to shorter analysis time, higher peak efficiency, and higher resolution [31,32]. Moreover, high resolution mass spectrometers (HRMS), such as the quadrupole orbital ion trap analyzer (Q-Orbitrap), have been used coupled to UHPLC for the determination of polyphenols and alkaloids profile in several food matrices including cocoa, tea, coffee, and coffee by-products [33,34,35]. This methodology stands as a powerful tool for the identification of natural products in plant extracts due to its high sensitivity and specificity, allowing precise quantification based on exact mass measurement.
Therefore, this study aimed to provide a comprehensive analysis of CGAs (n = 11) and alkaloids (n = 4) contained in four different polar coffee silverskin extracts obtained from Coffea arabica L. through ultra-high performance liquid chromatography coupled to a high resolution Orbitrap mass spectrometry (UHPLC-Q-Orbitrap HRMS). Besides, the bioaccessibility of total phenol compounds and changes in the antioxidant activity during an in vitro gastrointestinal digestion were also evaluated, in order to elucidate their efficacy for future applications in the nutraceutical industry.

2. Results

2.1. High Molecular Weight Melanoidins’ Content

The content in high molecular weight melanoidins (HMWM) obtained from four different CS samples by an ultrafiltration technique is summarized in Table 1. Total content of HMWM found in different types of CS extracts ranged from 173.8 to 234.9 mg/g, with a mean value of 204.6 mg/g for all samples.

2.2. Identification of CGAs and Alkaloids in the Analyzed CS Extracts though UHPLC-Q-Orbitrap HRMS

Identification of individual CGAs and alkaloids was conducted through UHPLC-Q-Orbitrap HRMS. For the purpose of achieving a good separation of the studied analytes, three different gradient programs were tested. By starting with 0% of Phase B, the authors obtained satisfactory results in terms of separation and peak shape for all target analytes. Accurately, the authors selected a C18 column with a polar modified surface and a stable stationary phase under 100% aqueous condition, as reported by the manufacturer [36], compatible with the chosen gradient. A total of 15 compounds were identified from different samples of CS. The exact mass for the studied compounds including ion assignment, theoretical and measured mass (m/z), accuracy, and sensitivity are shown in Table 2.
Experiments were achieved both in negative (ESI−) and in positive (ESI+) ion mode. All CGAs exhibited better fragmentation patterns producing the deprotonated molecule [M−H], whereas the ionization in the positive mode generated the protonated molecule [M + H]+ for alkaloids.
Full-scan HRMS data acquisition captured all sample data, enabling the identification of untargeted compounds and retrospective data analysis without the need to re-run samples. The confirmation was based on the accurate mass measurement, elemental composition assignment, and MS/MS spectrum interpretation (Figure S1). The identification of structural isomers caffeoylquinic acid (CQA, m/z 353.08780), p-coumaroylquinic acid (pCoQA, m/z 337.09289), dicaffeoylquinic acid (diCQA, m/z 515.11950), and feruloyl-caffeoylquinic and caffeoyl-feruloylquinic acids (FCQA and CFQA, m/z 529.13245) was achieved by comparison of their fragmentation pattern and the retention times previously reported in the literature [33,37]. Separation of all the investigated analytes was carried out in a total run time of 9 min. Due to insufficient chromatographic separation, 4- and 5-FQA were quantified together.

2.3. Quantification and Semi-Quantification of CGAs and Alkaloids in the Analyzed CS Extracts

The CGAs and alkaloids found in extracted concentration from the CS extracts were analyzed through the UHPLC-Q-Orbitrap HRMS method. The quantitative determination of target analytes was performed using calibration curves at eight concentration levels. We obtained regression coefficients >0.990. Semi-quantification of compounds that had no standard to generate a curve was based on a representative standard of the same group.
In the herein analyzed samples, eleven CGAs were identified and semi-quantified. Table 3 shows the results expressed as the mean value ± SD of the CGAs detected in different analyzed CS extracts. CQA isomers represented from 13.5 to 45.6% of total CGAs in a concentration range between 1.62 and 5.93 mg/g. Moreover, 4-CQA was found as the most commonly detected CQA ranging from 0.45 up to 2.65 mg/g. As far as FQAs were concerned, the levels of the three isomers represented from 24.8 to 69.0% of total CGAs and semi-quantified at a concentration range from 4.21 to 8.30 mg/g. On the other hand, pCoQA, mainly represented by 3- and 5-pCoQA, was detected at mean values of 1.17 and 0.84 mg/g for all samples, respectively. Regarding 3-pCoQA, the CS4 sample showed a ten-fold increase compared with other samples assayed. As shown in Table 3, among diCQA, 3,5-diCQA was the most commonly detected compound, ranging from 0.08 to 1.4 mg/g, with a mean value of 0.72 mg/g. On the other hand, isomers 4,5-CFQA and 3,4-FCQA were less relevant compounds, being only found in one and two samples, respectively, at low concentrations.
Apart from those, some important alkaloids including trigonelline, theobromine, theophylline, and caffeine were quantified and semi-quantified in the assayed samples, as shown in Table 4. Caffeine was the most commonly detected alkaloid, ranging from 27.7 up to 34.3 mg/g, with a mean value of 31.2 mg/g for all samples. Among the alkaloids, trigonelline was detected in all samples with a mean of 20.8 mg/g (range from 16.3 to 23.7 mg/g). Theobromine and theophylline were semi-quantified in the lowest amount for all the analyzed samples with a mean value of 0.1 and 0.2 mg/g, respectively.

2.4. In Vitro Bioaccessibility of Total Phenolic Compounds in the Analyzed CS Extracts

The bioaccessibility of total phenol compounds of the analyzed CS extracts was measured by using a simulated gastrointestinal digestion and colonic fermentation. Therefore, TPC by the Folin-Ciocalteu assay was evaluated during all stages of the gastrointestinal digestion and compared to non-digested samples in order to obtain an overview of their bioaccessibility. Table 5 shows the mean values (mg GAE/g) ± SD of TPC for all samples in each stage of the in vitro gastrointestinal digestion and the percentage of increase in TPC (not digested vs. digested stage). Digested samples showed significantly higher TPC values through all the gastrointestinal digestion stages when compared with the non-digested ones (p-value < 0.05). As shown in Table 5, the percentage of increase in TPC during gastrointestinal digestion was between 20.9 and 169.5%, with the highest value after the colon stage (considered as Pronase E stage + Viscozyme L stage).

2.5. Antioxidant Activity of the Polyphenolic Extract after In Vitro Gastrointestinal Digestion

The antioxidant activity of soluble fractions obtained during the gastrointestinal digestion (oral stage (OS), gastric stage (GS), duodenal stage (DS), Pronase E stage (PS), and Viscozyme stage (VS)) and non-digested extract was assessed and compared by three different procedures (ABTS, DPPH, and FRAP). Table 6 shows the results obtained (mean value and SD), expressed as mmol of Trolox equivalent per kg of extract.
Digested samples showed significantly higher antioxidant activity through all the gastrointestinal digestion stages when compared with the not digested ones (p-value < 0.05). The results are summarized in Table 6. The percentage of increase in the antioxidant activity during gastrointestinal digestion ranged from 19.4 to 162.6%, from 19.1 to 211.1%, and from −4.5 to 134.2% for ABTS, DPPH, and FRAP, respectively.
The highest percentage of increase in the antioxidant activity was found in the colon stage for all samples investigated. The ABTS, DPPH, and FRAP values measured during the gastrointestinal digestion against their corresponding total phenolic content values were correlated, except for the FRAP assay in the oral stage, as shown in Table 7.

3. Discussion

The aim of the present study was to provide useful information regarding the bioactive components present in CS material such as polyphenols, alkaloids, and melanoidins in order to add value to this coffee by-product. The polyphenolic profile of aqueous CS extracts was determined through UHPLC-HRMS with an Orbitrap mass analyzer. In detail, eleven CGAs and four alkaloids were identified and semi-quantified in four different CS extracts. Concerning the occurrence of CGAs in CS extracts, the levels found in assayed samples showed a two- to three-fold increase when compared to aqueous CS extract previously analyzed through the HPLC-MS/MS method by Nzekoue et al. [21], who reported a total concentration up to 5.44 mg/g. The monitored compounds were three of 11 CGAs (3,5-diCQA, 3-, and 5-CQA) revealed in the analyzed extracts, which explained the different results observed. CGAs detected in the extracts were mainly composed of CQAs and FQAs. The biological activity of these molecules has been related to specific functions involved in maintaining health status, like increasing plasma levels of glutathione, delayed glucose absorption, reducing the LDL oxidative susceptibility, and the inflammatory process, helping to prevent from several disorders [38,39,40]. Among alkaloids, caffeine was the most commonly detected compound, with a mean value of 31.2 mg/g. Our data revealed a two-fold increase in the mentioned alkaloid compared with regular coffee brews (ranging from 10.9 to 16.5 mg/g) [41]. Currently, caffeine represents the main ingredient in most energy drinks and energy bars. These products usually contain added synthetic caffeine, due to the significant price differences between natural sources of caffeine (coffee beans, tea leaves, and guarana, among others) and synthetic caffeine sources [42]. Caffeine-enriched extracts from CS, at a low purchase cost, could represent a natural ideal source to be used in these formulations. Melanoidins appear as one of the most relevant compounds found in CS samples. Several studies indicated that the melanoidins may exert a wide range of important biological properties including antioxidant and prebiotic activity [43]. It has been reported that the phenolic compounds found in coffee green beans can be linked with carbohydrates, dietary fiber, and proteins during the Maillard reaction, which contribute to the formation of coffee melanoidins [44]. According to Tores de la Cruz et al. [45], low molecular weight phenolic compounds, such as CGA, are incorporated in the melanoidin skeleton during the roasting process, forming a fiber–antioxidant complex. Our results showed a very high melanoidin content in aqueous extracts from CS, suggesting their potential use in the design of new healthy foods or as functional ingredients.
The bioaccessibility of total phenolic compounds and changes in the antioxidant activity during an in vitro gastrointestinal digestion were also evaluated. The digestion simulated in vitro was performed according to the INFOGEST method [46] until the duodenal stage, while the combined action of Pronase E and Viscozyme L was used to reproduce the action of microbiota occurring in the colon stage. On the other hand, our findings highlighted that both the bioaccessibility and antioxidant activity of CS polyphenols significantly increased after the colonic stage. These results suggested that the dietary polyphenols might be metabolized by human colonic microflora, generating metabolites with a greater antioxidant activity, resulting in an increase of their beneficial effects. Moreover, a good correlation existed among data obtained from the ABTS, DPPH, and FRAP tests and TPC measured during the gastrointestinal digestion, suggesting that these methods could provide some reliable information on the antioxidant properties of compounds released during in vitro digestion. Similar results have also been observed by Campos-Vega [27], who reported a higher bioaccessibility of phenolic compounds during the colon fermentation in spent coffee samples. Some epidemiology studies and meta-analysis showed an inverse association between coffee consumption and the incidence of colon cancer [47,48]. This outcome appeared to be attributable to the content of the melanoidins and CGAs in coffee brews, the same compounds found in analyzed samples [49]. Recently, several studies proposed the melanoidins as a type of soluble Maillardized dietary fiber that passes through the upper gastrointestinal tract without being absorbed and during the colon stage should be metabolized by gut microbiota, resulting in the release of phenolic compounds linked to them [50]. On the other hand, a previous study [44] reported that CS extract efficiently modulated gut microbiota composition; in particular, CS extract rich in melanoidins was able to induce preferential growth of bifidobacteria, rather than clostridia and Bacteroides spp. with well-known beneficial effects, suggesting that the consumption of CS melanoidin may exert a prebiotic effect. Results highlighted a high content of melanoidins, as well as CGAs and alkaloids, suggesting that these extracts may represent a suitable raw material for bioactive compounds to be used in nutraceutical formulations, functional foods, dietary supplements, fortified foods, or for their health-promoting properties.

4. Materials and Methods

4.1. Reagents and Materials

A total of four different samples of coffee silverskin (CS) produced by roasting coffee beans (Coffea arabica L.) of different origins (Congo: CS1, Nicaragua: CS2, Uganda: CS3, and Brazil: CS4) were provided from Evra Srl, Basilicata region, Italy. All materials were dried at 60 °C using a laboratory oven until the sample moisture reached a level from 6.6% to 6.9% (w/w). After drying, all samples were milled into powder using a laboratory mill and stored at room temperature in a cool and dry place, away from direct light.
The standards of polyphenols (purity >98%), namely chlorogenic acid, 3,4-dicaffeoylquinic acid, and caffeine, were purchased from Sigma-Aldrich (Milan, Italy). For the antioxidant assays, gallic acid, 2,2’-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid (ABTS), 6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid (Trolox), potassium persulfate, 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,3,5-triphenyltetrazolio chloride (TPTZ), anhydrous ferric chloride, hydrochloric acid, and sodium acetate were purchased from Sigma-Aldrich (Milan, Italy).
The enzymes used to simulate gastrointestinal digestion: α-amylase (1000–3000 U/mg solid) from human saliva, pepsin (≥2500 U/mg solid) from porcine gastric mucosa, pancreatin (8× USP) from porcine pancreas, bacterial protease from Streptomyces griseus (called also Pronase E, ≥3.5 U/mg solid), Viscozyme L, and also the chemicals calcium chloride dihydrate (CaCl2·2 H2O), sodium hydroxide (NaOH), potassium chloride (KCl), potassium thiocyanate (KCNS), monosodium phosphate (NaH2PO4), sodium sulphate (Na2SO4), sodium chloride (NaCl), and sodium bicarbonate (NaHCO3) were purchased from Sigma-Aldrich (Milan, Italy).
Methanol (MeOH) and water (LC-MS grade) were acquired from Carlo Erba reagents (Milan, Italy), whereas formic acid (98–100%) was purchased from Fluka (Milan, Italy).

4.2. Polyphenols Extraction

The polyphenols extraction procedure developed by Mesías et al. [51] was selected as a starting point and then slightly modified, as follows: 20 g of sample were suspended in 200 mL of water at 80 °C. The mixture was stirred using a horizontal shaker (KS130 Basic IKA, Argo Lab, Milan, Italy) for 30 min at 300× g and then centrifuged (X3R Heraeus Multifuge, Thermo Fisher Scientific LED GmbH, Kalkberg, Germany) at 4900× g at 24 °C for 10 min. The supernatant was recovered, and the pellet was re-extracted using the same procedure above reported. The pooled supernatants were freeze-dried and stored at −18 °C until analysis.

4.3. High Molecular Weight Melanoidins’ Content

Quantification of HMWM was carried out according to the procedure described by Rufian-Henares [52] with minor modifications. Briefly, four milliliters 4 mL of water extract (5 mg/mL) was subjected to ultrafiltration using an Amicon Ultra-4, regenerated cellulose 10 kDa (Millipore, Milan, Italy) at 7500× g for 70 min at 4 °C. The retentate was refilled with 4 mL of water and washed three times in order to separate the low molecular weight compounds from the retentate. The content of HMWM was determined by weighing the freeze dried retentate obtained after dialysis. The results were expressed as mg/g CS extracts.

4.4. Ultra-High Performance Liquid Chromatography and Orbitrap High Resolution Mass Spectrometry Analysis

Chromatographic analysis was performed through an UHPLC system (Thermo Fisher Scientific, Waltham, MA, USA) equipped with a degassing system, a Dionex Ultimate 3000, a Quaternary UHPLC pump with a pressure tolerance of 1250 bar, an autosampler device, and a thermostated (25 °C) Gemini 1.7 µm (50 mm × 2.1 mm, Phenomenex) column. The injection volume was of 5 μL. The eluent phases were: Phase A (water with 0.1% formic acid v/v) and Phase B (acetonitrile with 0.1% formic acid v/v). The separation gradient was applied as follows: initial 0% B for 1 min, increased to 95% B in 1 min. The gradient was held for 0.5 min at 95% B and linearly decreased to 75% B in 2.5 min and decreased again to 60% B in 1 min. Afterward, the gradient switched back to 0% B in 0.5 min and was held for 2.5 min for column re-equilibration. The flow rate was 0.4 mL/min. The autosampler was set at 10 °C. The UHPLC system was coupled to a Q-Exactive Orbitrap mass spectrometer (UHPLC, Thermo Fischer Scientific, Waltham, MA, USA) equipped with an electrospray (ESI) source simultaneously operating in fast negative/positive ion switching mode (Thermo Scientific, Bremen, Germany). The acquisitions were conducted by setting full MS/all ion fragmentation (AIF) mode that used a full MS scan (without higher-energy collisional dissociation HCD fragmentation), followed by an AIF scan (with fragmentation energy applied). Full MS/AIF experiments were carried out with the settings: microscans, 1; automatic gain control (AGC) target, 1e6; maximum injection time, 200 ms; mass resolution, 35,000 full width at half maximum (FWHM) at m/z 200 for full MS analysis; whereas AIF scan conditions were: microscans, 1; AGC target, 1e5; maximum injection time, 200 ms; mass resolution, 17,500 FWHM at m/z 200, HCD energy, at 10, 20, and 45. In both cases, the instrument was set to spray voltage 3.5 kV, capillary temperature 275 °C, sheath gas 45 (arbitrary units), auxiliary gas 10 (arbitrary units), m/z range 80–1200, data acquisition in profile mode. The accuracy and calibration of the Q Exactive Orbitrap LC-MS/MS were checked daily using a reference standard mixture provided by the manufacturer. The mass tolerance window was set to 5 ppm in both full scan MS and AIF modes. Data analysis and processing were performed using Xcalibur software v. 3.0.63 (Xcalibur, Thermo Fisher Scientific, Waltham, MA, USA).

4.5. In Vitro Simulated Gastrointestinal Digestion

The in vitro gastrointestinal digestion was performed according to the harmonized protocol recently developed in the COST action INFOGEST network [46]. The solutions used to simulate gastrointestinal digestion fluids (SSF: simulated salivary fluid, SGF: simulated gastric fluid, and SIF: simulated intestinal fluid) were prepared according to the procedure described by Minekus et al. [46] and shown in Table 8.
Briefly, five grams of aqueous extract were suspended in 3.5 mL of SSF. After 1 min of stirring, zero-point-five milliliters of α-amylase solution (made up in SSF, 75 U/mL), 25 µL of 0.3 M calcium chloride, and 975 µL water were added. Then, the pH of the mixture was adjusted to 7 with HCl 1 M, and the solution was incubated in a shaker bath at 100× g at 37 °C for 2 min. Afterward, 7.5 mL SGF, 1.6 mL pepsin solution (made up in SGF, 2000 U/mL), 5 μL of 0.3 M calcium chloride, and 695 µL of water were added and thoroughly mixed. The pH of the solution was adjusted to 3 with HCl 1 M, and the mixture was incubated in a shaker bath at 100× g at 37 °C for 2 h.
Then, in order to simulate the intestinal condition, eleven milliliters SIF, 5 mL pancreatin solution (made up in SIF, 100 U/mL of trypsin activity), 40 μL of 0.3 M calcium chloride, and 2.5 mL bile salt solution (65 mg/mL) were added, and the volume was adjusted to 20 mL with water. Then, the mixture was thoroughly mixed, and the pH of the solution was adjusted to 7 with NaOH 1 M. The mixture was incubated in a shaker bath at 100× g at 37 °C for 2 h and then centrifuged at 4900× g at 37 °C for 10 min. The supernatant was collected, and the remaining pellet was treated in order to determine the polyphenols’ bioaccessibility and antioxidant activity in the colon stage according to the procedure described by Annunziata et al. [53]. Therefore, in order to simulate the actions of the gut microbiota in the colon stage, five milliliters Pronase E solution (1 mg/mL) were added, and the pH was adjusted to 8 with NaOH 1 M. The mixture was incubated in a shaker bath at 100× g at 37 °C for 1 h. Finally, the solution was centrifuged at 4900× g at 37 °C for 10 min. The supernatant was collected, and the remaining pellet was treated with 150 µL Viscozyme L. The mixture was adjusted to pH 4 and incubated in a shaker bath at 100× g at 37 °C for 16 h, then centrifuged at 4900× g at 24 °C for 10 min.
At each in vitro digestion stage (OP: oral phase, GP: gastric phase, IP: intestinal phase, PO: Pronase E phase, and VO: Viscozyme L phase) the supernatants were collected and freeze-dried.

4.6. Determination of the Antioxidant Activity

The antioxidant activity of soluble fractions obtained during the gastrointestinal digestion (OP, GP, IP, PO, and VO) and non-digested extract was assessed and compared spectrophotometrically by three different procedures (ABTS, DPPH, and FRAP).

4.6.1. Determination of 2,2’-Azino-bis-3-ethylbenzthiazoline-6-sulphonic Acid Free Radical-Scavenging Activity

The ABTS assay was conducted based on the method reported by Re et al. [54] with minor modifications. Briefly, forty-four microliters of 2.45 mM aqueous potassium persulfate were added to 2.5 mL of aqueous ABTS (7 mM), following 16 h of incubation at room temperature in the dark. The ABTS solution was diluted with ethanol until an absorbance value of 0.75 (± 0.02) at 734 nm to obtain an ABTS radical working solution. The analysis was performed by adding 100 μL of the sample to 1 mL of ABTS radical working solution. The absorbance was monitored after 3 min at 734 nm. Results were expressed as mmol Trolox equivalents (TE) per kg of sample extract.

4.6.2. Determination of 2,2-Diphenyl-1-picrylhydrazyl Free Radical-Scavenging Activity

Determination of DPPH free radical-scavenging activity was carried out according to the procedure described by Brand-Williams et al. [55] with minor modifications. Briefly, methanolic DPPH (4 mg in 10 mL) was diluted with methanol to an absorbance value of 0.90 (± 0.02) at 517 nm to obtain a DPPH radical working solution. The analysis was performed by adding 200 μL of sample to 1 mL of DPPH radical working solution. The decrease in absorbance was monitored after 10 min at 517 nm. Results were expressed as mmol Trolox equivalents (TE) per kg of sample extract.

4.6.3. Determination of Ferric Reducing/Antioxidant Power Assay

The FRAP assay was conducted based on the method reported by Rajurkar et al. [56] with slight adaptations. Briefly, the FRAP solution contained 1.25 mL of TPTZ (2,4,6-tris(2-pyridyl)-s-triazine) solution (10 mM) in HCl (40 mM), 1.25 mL of FeCl3 (20 mM) in water, and 12.5 mL of acetate buffer (0.3 M, pH 3.6). Then, one-hundred-fifty microliters of sample were added to 2.850 mL of FRAP solution. The absorbance was monitored after 4 min at 593 nm. Results were expressed as mmol Trolox equivalents (TE) per kg of extract.

4.7. Determination of Total Phenolic Content

Total phenolic content (TPC) was measured using the Folin-Ciocalteu colorimetric method described previously by Singleton et al. [57] with slight modifications. In brief, zero-point-one-two-five milliliters of properly diluted extract were mixed with 0.125 mL of Folin-Ciocalteu reagent, 0.5 mL of deionized water, and then incubated at room temperature for 6 min. Afterward, one-point-two-five milliliters of a 7.5% sodium carbonate solution were added to the mixture and adjusted to 3 mL with deionized water. The absorbance was measured at 760 nm after 90 min of incubation at room temperature. The results were expressed as mg of gallic acid equivalents (GAE)/g of sample.

4.8. Statistics and Data Analysis

Values were expressed as the average values ± standard deviation (SD) of at least triplicate experiments. Statistical analysis of the results was performed using STATA 12 (STATACorp LP, College Station, TX, USA). The differences between average values were evaluated by using Tukey’s test at the level of significance p < 0.05. The correlation coefficients among means was determined using Pearson’s method.

5. Conclusions

In summary, the characterization of the main bioactive compounds including CGAs (n = 11) and alkaloids (n = 4) in four coffee silverskin samples through UHPLC-Q-Orbitrap spectrometry measurement was carried out. Notably, the caffeine content found in the analyzed CS extracts was similar to the concentration reported in coffee brews and roasted coffee. Results highlighted that CQAs and FQAs were the most common CGAs detected in the analyzed samples.
Moreover, our results suggested that CS polyphenol bioaccessibility and antioxidant activity increased during gastrointestinal digestion, and the colon stage might constitute the main biological site of action of these antioxidant compounds, suggesting their potential health benefits and justifying their exploitation as a functional food ingredient. The results herein obtained could represent a starting point for further in-depth studies regarding compounds released during in vitro digestion stages and after microbiota fermentation in order to evaluate the potential health benefits.

Supplementary Materials

The supplementary materials are available online: Figure S1: MS/MS mass spectra of investigated compounds (n = 15) extracted of coffee silverskin produced by roasting coffee beans (Coffea arabica L.).

Author Contributions

Conceptualization, A.R. and L.C.; methodology, L.C.; formal analysis, L.C.; investigation, L.C. and G.G.; resources, A.R.; writing, original draft preparation, L.C.; writing, review and editing, L.C., L.I., and A.N.; supervision, A.R.; project administration, A.R.; funding acquisition, A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the technical support of Mario Rubino, Maria Cristina Tricarico, and Paola Giannini.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bułdak, R.J.; Hejmo, T.; Osowski, M.; Bułdak, Ł.; Kukla, M.; Polaniak, R.; Birkner, E. The Impact of Coffee and Its Selected Bioactive Compounds on the Development and Progression of Colorectal Cancer In Vivo and In Vitro. Molecules 2018, 23, 3309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Sung, S.H.; Chang, Y.; Han, J. Development of polylactic acid nanocomposite films reinforced with cellulose nanocrystals derived from coffee silverskin. Carbohydr. Polym. 2017, 169, 495–503. [Google Scholar] [CrossRef] [PubMed]
  3. International Coffee Organization-(ICO). 2020. Available online: http://www.ico.org/documents/cy2019-20/cmr-1019-e.pdf (accessed on 10 April 2020).
  4. Jiménez-Zamora, A.; Pastoriza, S.; Rufián-Henares, J.A. Revalorization of coffee by-products. Prebiotic, antimicrobial and antioxidant properties. LWT-Food Sci. Technol. 2015, 61, 12–18. [Google Scholar]
  5. Costa, A.S.; Alves, R.C.; Vinha, A.F.; Costa, E.; Costa, C.S.; Nunes, M.A.; Almeida, A.A.; Santos-Silva, A.; Oliveira, M.B.P. Nutritional, chemical and antioxidant/pro-oxidant profiles of silverskin, a coffee roasting by-product. Food Chem. 2018, 267, 28–35. [Google Scholar] [CrossRef]
  6. Oreopoulou, V.; Tzia, C. Utilization of plant by-products for the recovery of proteins, dietary fibers, antioxidants, and colorants. In Utilization of By-Products and Treatment of Waste in the Food Industry; Springer: Boston, MA, USA, 2007; pp. 209–232. [Google Scholar]
  7. Narita, Y.; Inouye, K. High antioxidant activity of coffee silverskin extracts obtained by the treatment of coffee silverskin with subcritical water. Food Chem. 2012, 135, 943–949. [Google Scholar] [CrossRef] [Green Version]
  8. Ballesteros, L.F.; Teixeira, J.A.; Mussatto, S.I. Chemical, functional, and structural properties of spent coffee grounds and coffee silverskin. Food Bioprocess Technol. 2014, 7, 3493–3503. [Google Scholar] [CrossRef] [Green Version]
  9. Bresciani, L.; Calani, L.; Bruni, R.; Brighenti, F.; Del Rio, D. Phenolic composition, caffeine content and antioxidant capacity of coffee silverskin. Food Res. Int. 2014, 61, 196–201. [Google Scholar] [CrossRef]
  10. Bhupathiraju, S.N.; Pan, A.; Malik, V.S.; Manson, J.E.; Willett, W.C.; van Dam, R.M.; Hu, F.B. Caffeinated and caffeine-free beverages and risk of type 2 diabetes. Am. J. Clin. Nutr. 2013, 97, 155–166. [Google Scholar] [CrossRef] [Green Version]
  11. Iwai, K.; Narita, Y.; Fukunaga, T.; Nakagiri, O.; Kamiya, T.; Ikeguchi, M.; Kikuchi, Y. Study on the postprandial glucose responses to a chlorogenic acid-rich extract of decaffeinated green coffee beans in rats and healthy human subjects. Food Sci. Technol. Res. 2012, 18, 849–860. [Google Scholar] [CrossRef] [Green Version]
  12. Martinez-Gomez, A.; Caballero, I.; Blanco, C.A. Phenols and Melanoidins as Natural Antioxidants in Beer. Structure, Reactivity and Antioxidant Activity. Biomolecules 2020, 10, 400. [Google Scholar] [CrossRef] [Green Version]
  13. Regazzoni, L.; Saligari, F.; Marinello, C.; Rossoni, G.; Aldini, G.; Carini, M.; Orioli, M. Coffee silver skin as a source of polyphenols: High resolution mass spectrometric profiling of components and antioxidant activity. J. Funct. Foods 2016, 20, 472–485. [Google Scholar] [CrossRef]
  14. Grosso, G.; Godos, J.; Galvano, F.; Giovannucci, E.L. Coffee, caffeine, and health outcomes: An umbrella review. Annu. Rev. Nutr. 2017, 37, 131–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Izzo, L.; Castaldo, L.; Narváez, A.; Graziani, G.; Gaspari, A.; Rodríguez-Carrasco, Y.; Ritieni, A. Analysis of Phenolic Compounds in Commercial Cannabis sativa L. Inflorescences Using UHPLC-Q-Orbitrap HRMS. Molecules 2020, 25, 631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Gaspari, A.; Di Minno, G.; Ritieni, A. Red Wine Consumption and Cardiovascular Health. Molecules 2019, 24, 3626. [Google Scholar] [CrossRef] [Green Version]
  17. Gonçalves, B.; Moeenfard, M.; Rocha, F.; Alves, A.; Estevinho, B.N.; Santos, L. Microencapsulation of a natural antioxidant from coffee—Chlorogenic acid (3-caffeoylquinic acid). Food Bioprocess Technol. 2017, 10, 1521–1530. [Google Scholar] [CrossRef]
  18. Ouyang, H.; Li, J.; Wu, B.; Zhang, X.; Li, Y.; Yang, S.; He, M.; Feng, Y. A robust platform based on ultra-high performance liquid chromatography Quadrupole time of flight tandem mass spectrometry with a two-step data mining strategy in the investigation, classification, and identification of chlorogenic acids in Ainsliaea fragrans Champ. J. Chromatogr. A 2017, 1502, 38–50. [Google Scholar]
  19. Makita, C.; Chimuka, L.; Cukrowska, E.; Steenkamp, P.; Kandawa-Schutz, M.; Ndhlala, A.; Madala, N. UPLC-qTOF-MS profiling of pharmacologically important chlorogenic acids and associated glycosides in Moringa ovalifolia leaf extracts. S. Afr. J. Bot. 2017, 108, 193–199. [Google Scholar] [CrossRef]
  20. Clifford, M.N.; Jaganath, I.B.; Ludwig, I.A.; Crozier, A. Chlorogenic acids and the acyl-quinic acids: Discovery, biosynthesis, bioavailability and bioactivity. Nat. Prod. Rep. 2017, 34, 1391–1421. [Google Scholar] [CrossRef] [Green Version]
  21. Nzekoue, F.K.; Angeloni, S.; Navarini, L.; Angeloni, C.; Freschi, M.; Hrelia, S.; Vitali, L.A.; Sagratini, G.; Vittori, S.; Caprioli, G. Coffee silverskin extracts: Quantification of 30 bioactive compounds by a new HPLC-MS/MS method and evaluation of their antioxidant and antibacterial activities. Food Res. Int. 2020, 133, 109128. [Google Scholar] [CrossRef]
  22. Williamson, G.; Dionisi, F.; Renouf, M. Flavanols from green tea and phenolic acids from coffee: Critical quantitative evaluation of the pharmacokinetic data in humans after consumption of single doses of beverages. Mol. Nutr. Food Res. 2011, 55, 864–873. [Google Scholar] [CrossRef]
  23. Yu, M.; He, S.; Tang, M.; Zhang, Z.; Zhu, Y.; Sun, H. Antioxidant activity and sensory characteristics of Maillard reaction products derived from different peptide fractions of soybean meal hydrolysate. Food Chem. 2018, 243, 249–257. [Google Scholar] [CrossRef] [PubMed]
  24. Tagliazucchi, D.; Verzelloni, E.; Conte, A. Effect of dietary melanoidins on lipid peroxidation during simulated gastric digestion: Their possible role in the prevention of oxidative damage. J. Agric. Food Chem. 2010, 58, 2513–2519. [Google Scholar] [CrossRef] [PubMed]
  25. Gutiérrez-Grijalva, E.P.; Ambriz-Pére, D.L.; Leyva-López, N.; Castillo-López, R.I.; Heredia, J.B. Dietary phenolic compounds, health benefits and bioaccessibility. Arch. Latinoam. Nutr. 2016, 66, 87–100. [Google Scholar]
  26. Kumar Singh, A.; Cabral, C.; Kumar, R.; Ganguly, R.; Kumar Rana, H.; Gupta, A.; Rosaria Lauro, M.; Carbone, C.; Reis, F.; Pandey, A.K. Beneficial effects of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency. Nutrients 2019, 11, 2216. [Google Scholar] [CrossRef] [Green Version]
  27. Campos-Vega, R.; Vázquez-Sánchez, K.; López-Barrera, D.; Loarca-Piña, G.; Mendoza-Díaz, S.; Oomah, B.D. Simulated gastrointestinal digestion and in vitro colonic fermentation of spent coffee (Coffea arabica L.): Bioaccessibility and intestinal permeability. Food Res. Int. 2015, 77, 156–161. [Google Scholar] [CrossRef]
  28. Fernandez-Gomez, B.; Lezama, A.; Amigo-Benavent, M.; Ullate, M.; Herrero, M.; Martín, M.Á.; Mesa, M.D.; del Castillo, M.D. Insights on the health benefits of the bioactive compounds of coffee silverskin extract. J. Funct. Foods 2016, 25, 197–207. [Google Scholar] [CrossRef] [Green Version]
  29. Rufián-Henares, J.A.; Morales, F.J. Effect of in vitro enzymatic digestion on antioxidant activity of coffee melanoidins and fractions. J. Agric. Food Chem. 2007, 55, 10016–10021. [Google Scholar] [CrossRef] [Green Version]
  30. Jeszka-Skowron, M.; Zgoła-Grześkowiak, A.; Grześkowiak, T. Analytical methods applied for the characterization and the determination of bioactive compounds in coffee. Eur. Food Res. Technol. 2015, 240, 19–31. [Google Scholar] [CrossRef]
  31. Cooper, K.A.; Campos-Giménez, E.; Jiménez Alvarez, D.; Nagy, K.; Donovan, J.L.; Williamson, G. Rapid reversed phase ultra-performance liquid chromatography analysis of the major cocoa polyphenols and inter-relationships of their concentrations in chocolate. J. Agric. Food Chem. 2007, 55, 2841–2847. [Google Scholar] [CrossRef]
  32. Rodríguez-Carrasco, Y.; Castaldo, L.; Gaspari, A.; Graziani, G.; Ritieni, A. Development of an UHPLC-Q-Orbitrap HRMS method for simultaneous determination of mycotoxins and isoflavones in soy-based burgers. LWT-Food Sci. Technol. 2019, 99, 34–42. [Google Scholar]
  33. Castaldo, L.; Graziani, G.; Gaspari, A.; Izzo, L.; Luz, C.; Mañes, J.; Rubino, M.; Meca, G.; Ritieni, A. Study of the chemical components, bioactivity and antifungal properties of the coffee husk. J. Food Res. 2018, 7, 43–54. [Google Scholar] [CrossRef] [Green Version]
  34. Sánchez-Rabaneda, F.; Jáuregui, O.; Casals, I.; Andrés-Lacueva, C.; Izquierdo-Pulido, M.; Lamuela-Raventós, R.M. Liquid chromatographic/electrospray ionization tandem mass spectrometric study of the phenolic composition of cocoa (Theobroma cacao). J. Mass Spectrom. 2003, 38, 35–42. [Google Scholar] [CrossRef] [PubMed]
  35. López-Gutiérrez, N.; Romero-González, R.; Plaza-Bolaños, P.; Vidal, J.L.M.; Frenich, A.G. Identification and quantification of phytochemicals in nutraceutical products from green tea by UHPLC–Orbitrap-MS. Food Chem. 2015, 173, 607–618. [Google Scholar] [CrossRef] [PubMed]
  36. Phenomenex. The Ultimate Guide to HPLC/UHPLC Reversed Phase Selectivity. Available online: https://phenomenex.blob.core.windows.net/documents/4e38c0e4-ddf3-44df-aab7-da72332eef8d.pdf (accessed on 10 April 2020).
  37. Mullen, W.; Nemzer, B.; Stalmach, A.; Ali, S.; Combet, E. Polyphenolic and hydroxycinnamate contents of whole coffee fruits from China, India, and Mexico. J. Agric. Food Chem. 2013, 61, 5298–5309. [Google Scholar] [CrossRef] [Green Version]
  38. McCarty, M.F. A chlorogenic acid-induced increase in GLP-1 production may mediate the impact of heavy coffee consumption on diabetes risk. Med. Hypotheses 2005, 64, 848–853. [Google Scholar] [CrossRef]
  39. Cornelis, M.C.; El-Sohemy, A. Coffee, caffeine, and coronary heart disease. Curr. Opin. Lipidol. 2007, 18, 13–19. [Google Scholar] [CrossRef]
  40. Liang, N.; Kitts, D.D. Role of chlorogenic acids in controlling oxidative and inflammatory stress conditions. Nutrients 2016, 8, 16. [Google Scholar] [CrossRef] [Green Version]
  41. Fujioka, K.; Shibamoto, T. Chlorogenic acid and caffeine contents in various commercial brewed coffees. Food Chem. 2008, 106, 217–221. [Google Scholar] [CrossRef]
  42. Zhang, L.; Kujawinski, D.M.; Federherr, E.; Schmidt, T.C.; Jochmann, M.A. Caffeine in your drink: Natural or synthetic? Anal. Chem. 2012, 84, 2805–2810. [Google Scholar] [CrossRef]
  43. Wang, H.-Y.; Qian, H.; Yao, W.-R. Melanoidins produced by the Maillard reaction: Structure and biological activity. Food Chem. 2011, 128, 573–584. [Google Scholar] [CrossRef]
  44. Borrelli, R.C.; Esposito, F.; Napolitano, A.; Ritieni, A.; Fogliano, V. Characterization of a new potential functional ingredient: Coffee silverskin. J. Agric. Food Chem. 2004, 52, 1338–1343. [Google Scholar] [CrossRef] [PubMed]
  45. Tores de la Cruz, S.; Iriondo-DeHond, A.; Herrera, T.; Lopez-Tofiño, Y.; Galvez-Robleño, C.; Prodanov, M.; Velazquez-Escobar, F.; Abalo, R.; del Castillo, M.D. An Assessment of the Bioactivity of Coffee Silverskin Melanoidins. Foods 2019, 8, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Minekus, M.; Alminger, M.; Alvito, P.; Ballance, S.; Bohn, T.; Bourlieu, C.; Carriere, F.; Boutrou, R.; Corredig, M.; Dupont, D. A standardised static in vitro digestion method suitable for food–an international consensus. Food Funct. 2014, 5, 1113–1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Galeone, C.; Turati, F.; La Vecchia, C.; Tavani, A. Coffee consumption and risk of colorectal cancer: A meta-analysis of case–control studies. Cancer Causes Control 2010, 21, 1949–1959. [Google Scholar] [CrossRef]
  48. Schmit, S.L.; Rennert, H.S.; Rennert, G.; Gruber, S.B. Coffee consumption and the risk of colorectal cancer. Cancer Epidemiol. Prev. Biomark. 2016, 25, 634–639. [Google Scholar] [CrossRef] [Green Version]
  49. Vitaglione, P.; Fogliano, V.; Pellegrini, N. Coffee, colon function and colorectal cancer. Food Funct. 2012, 3, 916–922. [Google Scholar] [CrossRef]
  50. Pérez-Burillo, S.; Mehta, T.; Esteban-Muñoz, A.; Pastoriza, S.; Paliy, O.; Rufián-Henares, J.Á. Effect of in vitro digestion-fermentation on green and roasted coffee bioactivity: The role of the gut microbiota. Food Chem. 2019, 279, 252–259. [Google Scholar] [CrossRef]
  51. Mesías, M.; Navarro, M.; Martínez-Saez, N.; Ullate, M.; Del Castillo, M.; Morales, F. Antiglycative and carbonyl trapping properties of the water soluble fraction of coffee silverskin. Food Res. Int. 2014, 62, 1120–1126. [Google Scholar] [CrossRef] [Green Version]
  52. Rufian-Henares, J.A.; de la Cueva, S.P. Antimicrobial Activity of Coffee Melanoidins—A Study of Their Metal-Chelating Properties. J. Agric. Food Chem. 2009, 57, 432–438. [Google Scholar] [CrossRef]
  53. Annunziata, G.; Maisto, M.; Schisano, C.; Ciampaglia, R.; Daliu, P.; Narciso, V.; Tenore, G.C.; Novellino, E. Colon bioaccessibility and antioxidant activity of white, green and black tea polyphenols extract after in vitro simulated gastrointestinal digestion. Nutrients 2018, 10, 1711. [Google Scholar] [CrossRef] [Green Version]
  54. Re, R.; Pellegrini, N.; Proteggente, A.; Pannala, A.; Yang, M.; Rice-Evans, C.J.F. Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radic. Bio. Med. 1999, 26, 1231–1237. [Google Scholar] [CrossRef]
  55. Brand-Williams, W.; Cuvelier, M.-E.; Berset, C. Use of a free radical method to evaluate antioxidant activity. LWT-Food Sci. Technol. 1995, 28, 25–30. [Google Scholar] [CrossRef]
  56. Rajurkar, N.S.; Hande, S. Estimation of phytochemical content and antioxidant activity of some selected traditional Indian medicinal plants. Indian J. Pharm. Sci. 2011, 73, 146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Singleton, V.L.; Rossi, J.A. Colorimetry of total phenolics with phosphomolybdic-phosphotungstic acid reagents. Am. J. Enol. Vitic. 1965, 16, 144–158. [Google Scholar]
Sample Availability: Not available.
Table 1. Melanoidin content in different analyzed coffee silverskin (CS) extracts.
Table 1. Melanoidin content in different analyzed coffee silverskin (CS) extracts.
SamplesMelanoidins
(mg/g) ± SD
CS1194.4 ± 2.1
CS2173.8 ± 1.3
CS3215.4 ± 2.6
CS4234.9 ± 3.1
Table 2. Chromatographic and spectrometric parameters including ion assignment, theoretical and measured mass (m/z), retention time, accuracy, and sensitivity for the investigated analytes (n = 15). FCQA, feruloyl-caffeoylquinic acid; pCoQA, p-coumaroylquinic acid; CFQA, caffeoyl-feruloylquinic.
Table 2. Chromatographic and spectrometric parameters including ion assignment, theoretical and measured mass (m/z), retention time, accuracy, and sensitivity for the investigated analytes (n = 15). FCQA, feruloyl-caffeoylquinic acid; pCoQA, p-coumaroylquinic acid; CFQA, caffeoyl-feruloylquinic.
CompoundsChemicalAdductTheoreticalMeasuredAccuracyRetention TimeLODLOQ
FormulaMass (m/z)Mass (m/z)(Δ mg/kg)(min)(mg/kg)(mg/kg)
TrigonellineC7H7NO2[M + H]+138.05495138.05487−0.580.40.050.16
TheophyllineC7H8N4O2[M + H]+181.07200181.07193−0.393.630.050.16
TheobromineC7H8N4O2[M + H]+181.07200181.072040.223.740.050.16
5-CQAC16H18O9[M − H]353.08780353.087800.003.810.050.14
CaffeineC8H10N4O2[M + H]+195.08765195.08757−0.413.840.050.16
3,4-FCQAC26H26O12[M − H]529.13245529.132450.003.850.050.16
4-CQAC16H18O9[M − H]353.08780353.08768−0.343.90.050.14
3-CQAC16H18O9[M − H]353.08780353.08762−0.513.940.050.14
3-pCoQAC16H18O8[M − H]337.09289337.09232−1.693.960.050.14
5-pCoQAC16H18O8[M − H]337.09289337.092900.033.990.050.14
3-FQAC17H20O9[M − H]367.10346367.10303−1.173.960.050.14
4 + 5-FQAC17H20O9[M − H]367.10346367.10309−1.014.010.050.14
3,5-diCQAC25H24O12[M − H]515.11950515.119930.834.110.050.16
4,5-CFQAC26H26O12[M − H]529.13245529.134954.724.140.050.16
3,4-diCQAC25H24O12[M − H]515.11950515.121032.974.220.050.16
Table 3. Chlorogenic acids’ (CGAs) content in the analyzed CS extracts. Results are shown as the mean value ± SD (n = 3).
Table 3. Chlorogenic acids’ (CGAs) content in the analyzed CS extracts. Results are shown as the mean value ± SD (n = 3).
CompoundsCS1CS2CS3CS4
Average (mg/g) ± SDAverage (mg/g) ± SDAverage (mg/g) ± SDAverage (mg/g) ± SD
3-CQA0.21 ± 0.020.40 ± 0.040.51 ± 0.031.54 ± 0.06
4-CQA0.45 ± 0.032.46 ± 0.122.65 ± 0.112.61 ± 0.04
5-CQA0.96 ± 0.052.38 ± 0.610.85 ± 0.091.78 ± 0.06
3-pCoQA0.04 ± 0.0010.21 ± 0.040.15 ± 0.034.30 ± 0.31
5-pCoQA0.31 ± 0.040.24 ± 0.080.13 ± 0.012.67 ± 0.04
3-FQA1.51 ± 0.160.87 ± 0.090.75 ± 0.040.79 ± 0.05
4 + 5-FQA6.79 ± 0.425.36 ± 0.193.46 ± 0.203.46 ± 0.09
3,4-diCQA0.05 ± 0.01n. d.n. d.n. d.
3,5-diCQA1.40 ± 0.110.69 ± 0.080.08 ± 0.01n. d.
3,4-FCQA0.25 ± 0.06n. d.0.19 ± 0.03n. d.
4,5-CFQA0.07 ± 0.01n. d.n. d.n. d.
Total12.0412.618.7717.15
The differences between average values were evaluated by using Tukey’s test at the level of significance p < 0.05.
Table 4. Alkaloids’ content in the analyzed CS extracts. Results are shown as the mean value ± SD (n = 3).
Table 4. Alkaloids’ content in the analyzed CS extracts. Results are shown as the mean value ± SD (n = 3).
AlkaloidsCS1CS2CS3CS4
Average (mg/g) ± SDAverage (mg/g) ± SDAverage (mg/kg) ± SDAverage (mg/g) ± SD
Trigonelline22.5 ± 0.316.3 ± 0.220.4 ± 0.323.7 ± 0.3
Theobromine0.3 ± 0.010.2 ± 0.020.3 ± 0.030.0 ± 0.01
Theophylline0.1 ± 0.020.1 ± 0.010.1 ± 0.020.1 ± 0.01
Caffeine33.6 ± 0.429.2 ± 0.527.7 ± 0.434.3 ± 0.6
Total56.3945.7348.4758.19
The differences between average values were evaluated by using Tukey’s test at the level of significance p < 0.05.
Table 5. Bioaccessibility of total phenolic content evaluated by the Folin-Ciocalteu method in non-digested samples and during the simulated in vitro digestion.
Table 5. Bioaccessibility of total phenolic content evaluated by the Folin-Ciocalteu method in non-digested samples and during the simulated in vitro digestion.
SamplesTPC in CS1TPC in CS2TPC in CS3TPC in CS4
mg GAE/g ± SD%mg GAE/g ± SD%mg GAE/g ± SD%mg GAE/g ± SD%
Not digested5.7 ± 0.1 5.8 ± 0.2 6.5 ± 0.2 6.9 ± 0.3
Digestion Stage
OS7.4 ± 0.328.27.3 ± 0.324.78.5 ± 0.333.08.3 ± 0.320.9
GS7.6 ± 0.332.07.8 ± 0.434.510.1 ± 0.559.19.4 ± 0.637.0
DS8.4 ± 0.446.78.7 ± 0.448.810.3 ± 0.462.210.1 ± 0.247.2
PS8.1 ± 0.540.98.3 ± 0.542.79.7 ± 0.352.710.4 ± 0.451.5
VS6.2 ± 0.58.06.9 ± 0.618.67.4 ± 0.416.28.1 ± 0.318.0
Total colon stage14.3 ± 0.5148.915.2 ± 0.6161.317.1 ± 0.4168.918.5 ± 0.4169.5
OS: oral stage; GS: gastric stage; DS: duodenal stage; PS: Pronase E stage; VS: Viscozyme stage; GAE: gallic acid equivalents; TPC: total phenolic content; %: percentage of increase or decrease in TPC (not digested vs. digested stage). The differences between average values were evaluated by using Tukey’s test at the level of significance p < 0.05.
Table 6. Antioxidant activity of digested samples evaluated by ABTS, DPPH, and FRAP assays expressed as mmol Trolox/kg of CS extract.
Table 6. Antioxidant activity of digested samples evaluated by ABTS, DPPH, and FRAP assays expressed as mmol Trolox/kg of CS extract.
ABTS mmol Trolox/kg ± SDDPPH mmol Trolox/kg ± SDFRAP mmol Trolox/kg ± SD
CS1CS2CS3CS4CS1CS2CS3CS4CS1CS2CS3CS4
Not digested23.5 ± 0.219.1 ± 0.328.3 ± 0.329.1 ± 0.48.1 ± 0.16.7 ± 0.19.5 ± 0.211.2 ± 0.214.4 ± 0.315.8 ± 0.318.3 ± 0.517.2 ± 0.4
Digestion Stage
OS30.5 ± 0.328.7 ± 0.433.8 ± 0.437.0 ± 0.510.3 ± 0.210.0 ± 0.112.0 ± 0.113.6 ± 0.217.2 ± 0.119.2 ± 0.222.9 ± 0.321.8 ± 0.4
GS30.8 ± 0.324.7 ± 0.336.8 ± 0.436.9 ± 0.59.7 ± 0.28.1 ± 0.114.0 ± 0.313.5 ± 0.416.2 ± 0.418.7 ± 0.317.5 ± 0.524.0 ± 0.6
DS43.4 ± 1.635.3 ± 0.849.4 ± 1.152.4 ± 0.620.5 ± 0.210.7 ± 0.322.9 ± 0.322.8 ± 0.416.5 ± 0.516.7 ± 0.219.3 ± 0.322.2 ± 0.3
PS36.0 ± 0.422.1 ± 0.232.7 ± 0.442.6 ± 0.816.5 ± 0.113.0 ± 0.318.5 ± 0.221.2 ± 0.317.5 ± 0.116.6 ± 0.319.7 ± 0.521.6 ± 0.4
VS25.7 ± 0.720.4 ± 0.329.3 ± 0.431.9 ± 0.58.7 ± 0.17.5 ± 0.110.5 ± 0.312.2 ± 0.311.7 ± 0.110.6 ± 0.213.7 ± 0.118.6 ± 0.2
Total colon stage61.7 ± 0.342.6 ± 0.261.9 ± 0.474.5 ± 0.725.2 ± 0.120.5 ± 0.229.0 ± 0.333.5 ± 0.329.2 ± 0.127.2 ± 0.327.2 ± 0.340.3 ± 0.3
OS: oral stage; GS: gastric stage; DS: duodenal stage; PS: Pronase E stage; VS: Viscozyme stage; ABTS and DPPH: free-radical scavenging; FRAP ferric reducing/antioxidant power; Trolox equivalent antioxidant capacity (TEAC). The differences between average values were evaluated by using Tukey’s test at the level of significance p < 0.05.
Table 7. Correlation between TPC evaluated by Folin-Ciocalteu and antioxidant activity evaluated by the ABTS, DPPH, and FRAP methods. The correlation coefficients among means were determined using Pearson’s method.
Table 7. Correlation between TPC evaluated by Folin-Ciocalteu and antioxidant activity evaluated by the ABTS, DPPH, and FRAP methods. The correlation coefficients among means were determined using Pearson’s method.
AssayOral Stage R2Gastric Stage R2Duodenal Stage R2Pronase E Stage R2Viscozyme L Stage R2
ABTS0.8850.840.8230.6310.677
DPPH0.8820.9410.6520.8750.82
FRAP0.5570.3790.8610.9680.861
Table 8. Composition of stock solutions of simulated digestion fluid. SSF: simulated salivary fluid, SGF: simulated gastric fluid, and SIF: simulated intestinal fluid.
Table 8. Composition of stock solutions of simulated digestion fluid. SSF: simulated salivary fluid, SGF: simulated gastric fluid, and SIF: simulated intestinal fluid.
SSF (pH 7)SGF (pH 3)SIF (pH 7)
Salt SolutionStock Concentration (mol/L)mL of Stock Added to Prepare 0.4 L (mL)Final Salt Concentration in Sample (mmol/L)mL of Stock Added to Prepare 0.4 L (mL)Final Salt Concentration in Sample (mmol/L)mL of Stock Added to Prepare 0.4 L (mL)Final Salt Concentration in Sample (mmol/L)
KCl0.515.115.16.96.96.86.8
KH2PO40.53.71.350.90.90.80.8
NaHCO316.813.6812.52542.585
NaCl2--11.847.29.638.4
MgCl2(H2O)60.150.50.150.40.121.10.33
NH4(CO3)20.50.060.060.50.5--
CaCl2(H2O)20.3-1.5-0.15-0.6

Share and Cite

MDPI and ACS Style

Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Ritieni, A. In Vitro Bioaccessibility and Antioxidant Activity of Coffee Silverskin Polyphenolic Extract and Characterization of Bioactive Compounds Using UHPLC-Q-Orbitrap HRMS. Molecules 2020, 25, 2132. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25092132

AMA Style

Castaldo L, Narváez A, Izzo L, Graziani G, Ritieni A. In Vitro Bioaccessibility and Antioxidant Activity of Coffee Silverskin Polyphenolic Extract and Characterization of Bioactive Compounds Using UHPLC-Q-Orbitrap HRMS. Molecules. 2020; 25(9):2132. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25092132

Chicago/Turabian Style

Castaldo, Luigi, Alfonso Narváez, Luana Izzo, Giulia Graziani, and Alberto Ritieni. 2020. "In Vitro Bioaccessibility and Antioxidant Activity of Coffee Silverskin Polyphenolic Extract and Characterization of Bioactive Compounds Using UHPLC-Q-Orbitrap HRMS" Molecules 25, no. 9: 2132. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25092132

Article Metrics

Back to TopTop