Next Article in Journal
Interleukin-4-Mediated Oxidative Stress Is Harmful to Hippocampal Neurons of Prothrombin Kringle-2-Lesioned Rat In Vivo
Next Article in Special Issue
HR-LCMS-Based Metabolite Profiling, Antioxidant, and Anticancer Properties of Teucrium polium L. Methanolic Extract: Computational and In Vitro Study
Previous Article in Journal
Cardiac and Metabolic Impact of Functional Foods with Antioxidant Properties Based on Whey Derived Proteins Enriched with Hemp Seed Oil
Previous Article in Special Issue
Allium subhirsutum L. as a Potential Source of Antioxidant and Anticancer Bioactive Molecules: HR-LCMS Phytochemical Profiling, In Vitro and In Vivo Pharmacological Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Bilberry (Vaccinium myrtillus L.) Extracts Comparative Analysis Regarding Their Phytonutrient Profiles, Antioxidant Capacity along with the In Vivo Rescue Effects Tested on a Drosophila melanogaster High-Sugar Diet Model

1
Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
2
Doctoral School of Molecular Cell Biology and Immunology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
3
Doctoral School of Animal Science, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary
4
Faculty of Agricultural Science, Food Industry and Environmental Protection, “Lucian Blaga” University of Sibiu, 550012 Sibiu, Romania
5
Faculty of Medicine, “Vasile Goldis” Western University of Arad, 310045 Arad, Romania
6
Faculty of Pharmacy, “Vasile Goldis” Western University of Arad, 310045 Arad, Romania
7
Doctoral School of Nutrition, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary
8
Agricultural and Molecular Research and Service Institute, University of Nyiregyhaza, H-4400 Nyíregyháza, Hungary
9
Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary
*
Authors to whom correspondence should be addressed.
Andreea-Adriana Neamtu and Rita Szoke-Kovacs contributed equally to this work and are both considered first author.
Submission received: 10 October 2020 / Revised: 23 October 2020 / Accepted: 26 October 2020 / Published: 30 October 2020
(This article belongs to the Special Issue Antioxidant and Biological Properties of Plant Extracts)

Abstract

:
Bilberries (Vaccinium myrtillus L.) have been reported to hold a plentitude of health-promoting properties beyond basic nutrition, mainly attributed to their anthocyanin content and antioxidant activity. In this article, we built the phytochemical profile of three wild bilberry fruit extract formulations (aqueous, methanolic, and hydro-methanolic) using UHPLC-ESI-MS/MS putative analysis, identifying 88 individual phytochemicals, mainly flavonoids (total content 8.41 ± 0.11 mg QE/g dw), free amino acids, polyphenols (total content 21.68 ± 0.19 mg GAE/g dw), carboxylic acids, and vitamins. Furthermore, the antioxidant activity of the extract was assessed, reaching 78.03 ± 0.16% DPPH free radical scavenging activity, comparable to literature values determined for bilberry extracts of other origin. Due to the increased prevalence of metabolic syndrome and based on the reviewed benefits of bilberries, we tested the most potent formulation of our bilberry extracts in this biological context. The in vivo rescue effect of a bilberry extract supplemented diet on Drosophila melanogaster was assessed by monitoring biochemical and genomic markers. Hemolymph trehalose levels were halved upon addition of 3% hydro-methanolic bilberry extract to a high-sugar (1.5 M sucrose) diet, as compared to the non-supplemented high-sugar diet. Noteworthy, the rescue seen for flies kept on the bilberry extract supplemented high-sugar diet appeared to parallel the trehalose levels observed in the case of the control diet (50 mM sucrose) flies. Moreover, next to the trehalose-lowering type of in vivo effects, other gene expression related rescues were also detected for genes such as InR, Akh, AstA, AstC, Irk, Npc2g, and CCHa2 upon supplementation of the high-sugar diet with our hydro-methanolic bilberry fruit extract. Our findings suggest that such a bilberry fruit extract could generate physiological and genomic type of compensatory mechanisms so that further translational approaches would advance the understanding of some human specific pathological conditions.

Graphical Abstract

1. Introduction

Bilberries (Vaccinium myrtillus L.) are the native European blueberries, closely related to the North American blueberry species (Vaccinium corymbosum L.), both part of the widespread genus Vaccinium containing over 200 species [1]. Berries constitute a large group of functional foods, also nowadays known as “superfoods”, whose consumption delivers several health benefits beyond basic nutrition, mainly attributed to their anthocyanin content holding mainly antioxidant but also recently characterized antiglycoxidant properties [2,3]. Despite their similarities, berries differ in their phytochemical content, such as flavonoids and anthocyanins, of both plants and fruit [4,5,6]. The quantity of the main anthocyanins found in bilberries is more than twice the one found in blueberries [7]. Furthermore, anthocyanin content seems to be higher in wild bilberries as compared to the cultivated ones [8,9,10].
The biological effects of individual anthocyanins have been studied in detail; however, that does not hold true for the whole bilberry fruit extract. Complex interactions of multiple components within the food matrix may differ significantly in outcome when compared to a single purified constituent or constituents [11]. Hence, there is a need to evaluate the observed biological properties as a result of not only additive but also complementary synergistic and/or antagonistic effects, or any combination thereof, of all phytochemicals present [11]. In line with this rationale, we decided to focus our study on the whole bilberry fruit extract.
Bilberries have been observed to hold a plentitude of health-promoting properties. Research studies support their beneficial effects, among which antioxidant, anti-obesity, anticarcinogenic, cardioprotective, anti-inflammatory, hypoglycemic, antimicrobial, and vision improvement [12,13,14].
Based on the health-promoting properties of bilberries, we decided to study the extract in a Drosophila melanogaster model for insulin resistance and type II diabetes [15]. The first paper that explicitly modelled type II diabetes in Drosophila tested the role of a high-sugar diet on the acquisition of insulin resistance [16]. Characteristics of insulin resistance were assessed as rearing flies on high-sugar diet resulted in the overexpression of dilp 2,3,5 RNA and circulating DILP2. Despite increased levels of DILP(s) in circulation, sugar levels remained elevated—a feature resembling mammalian insulin resistance. Moreover, decreased levels of phospho-Akt were observed in response to administration of exogenous insulin in adult flies reared on a high-sugar diet, suggesting a decreased capacity of response to insulin signalling following chronic uptake of dietary sugar. Finally, flies exposed to a high-sugar diet exhibited elevation of free fatty acid and triglyceride levels as compared to the flies raised on a control isocaloric diet, and the morphology of lipid storage changed, to exhibit fewer, larger droplets [17]. Together, these studies showed that flies kept on a chronic high-sugar diet are hyperglycemic, insulin-resistant, and obese, constituting a relevant model to assess our bilberry fruit extract health promoting properties [18,19].
This is highly relevant nowadays, as in recent years, people consume increasingly higher amounts of carbohydrates, to reach what Bovi et al. suggestively called “the pandemic of sucrose intake” [20]. Added sugars represent empty calories, as they only supply food energy but no other nutrients. They increase the risk of developing obesity, inflammation, cardiovascular disease, hypertension, diabetes, insulin resistance, obesity-related cancers, and metabolic syndrome, a non-communicable disease responsible for morbidity and mortality predominantly in the developed world [21]. It is defined by the World Health Organization as a pathology characterized by abdominal obesity, insulin resistance, hypertension, and hyperlipidemia [22]. The incidence of metabolic syndrome in the United States is about one in three people, reaching epidemic proportions [23].
With growing attention dedicated to harvesting and consuming local foods, we find it important to build the chemical profile, including chemo-mapping, antioxidant activity, and polyphenol and flavonoid content of bilberry fruit growing in the Carpathian Mountains, in the central part of Romania, which is a novel analysis and never before made the subject of a scientific article to the best of our knowledge. We also examine the rescue effects generated by our bilberry fruit extract in relation to the modifications caused by a high-sugar diet, using Drosophila melanogaster as a nutritional genetic model.

2. Materials and Methods

2.1. Chemicals and Reagents

Gallic acid, quercetin, formic acid, tryptophan, catechin, epigallocatechin, chlorogenic acid, vanillin, epicatechin, cyanidin-3-O-glucoside, 4-coumaric acid, isoquercitrin, myricetin, quercitrin, and naringenin, Folin-Ciocalteu reagent, DPPH (2,2-diphenyl-1-picrylhydrazyl), methanol analytical grade, aluminum chloride, sodium acetate, and sodium carbonate were purchased from Sigma Aldrich (Merck; Darmstadt, Germany).

2.2. Extracts Preparation

Extracts of bilberry (Vaccinium myrtillus L.) were prepared in three formulations from ripe fruit of wild bilberry collected in three consecutive years from the same alpine ecosystem in Făgăraș (South Carpathian) Mountains, Sibiu county, Romania, in July 2017, 2018, and 2019. The further described extracts were prepared from a mixture of samples of all three harvests, frozen and stored at −20 °C, then oven dried at 40 °C, until a constant mass was achieved, grinded on a domestic mill, and pooled together in equal quantities.
In the preparation of the extracts, the solvent varied as follows: E1—distilled water (aqueous extract); E2—methanol (methanolic extract); E3—distilled water:methanol mixture in 1:1 ratio (hydro-methanolic extract). For each extract, 500 mg of fruit powder was extracted using 10 mL of the specific solvent, in the ultrasound bath for 30 min, at 40 °C. Then, vacuum filtration was performed through Grade 1 (11 µm pore size) Whatman filter paper and brought to scale in 10 mL volumetric flasks.
Post-preparation, the extracts were stored in the fridge at 4 °C, in tinted glass bottles. Prior to experimental usage, the extracts were allowed to reach room temperature and shaken thoroughly.

2.3. Phytonutrient Profile Determination Via UHPLC-ESI-MS/MS Analysis

A Dionex Ultimate 3000RS ultrahigh pressure liquid chromatography (UHPLC) system equipped with a Thermo Scientific Accucore C18 column, L./I.D. of 100/2.1 mm, and a particle size of 2.6 μm, was coupled to a Thermo Q Exactive Orbitrap mass spectrometer (MS) using an electrospray ionization source (ESI), and the measurement accuracy was within 5 ppm. The mass spectrometer was operated at 320 °C capillary temperature, 4.0 kV in positive mode and 3.8 kV in negative mode of spray voltage, and a resolution of 35,000 in the case of MS, while it was 17,500 for MS/MS. The 100–1500 m/z was the scanned mass interval. For MS/MS scans, the collision energy was 35 NCE. The difference between measured and calculated molecular ion masses was always below 5 ppm.
For both the positive and negative ionization modes of UHPLC separation, an eluent A (1000 mL of water containing and 1 mL of formic acid) and eluent B (1000 mL of methanol and 1 mL of formic acid) combination was used.
The flow rate was set to 200 μL/min, and the same gradient elution program was used both positive and negative ionization mode type of determinations (0–3 min, 95% A, 3–43 min, 95→0% A; 43–61 min, 0% A; 61–62 min, 0→95% A; 62–70 min, 95% A). An amount of 5 μL of aqueous or hydro-methanolic ripe bilberry fruit extracts was injected at every run.
In the analysis, the following isolated compounds were used as standards: gallic acid (3,4,5-trihydroxybenzoic acid), tryptophan, catechin, epigallocatechin, chlorogenic acid (3-O-caffeoylquinic acid), vanillin, epicatechin, cyanidin-3-O-glucoside (kuromanin, asterin, chrysanthemin), 4-coumaric acid, isoquercitrin (hirsutrin, quercetin-3-O-glucoside), myricetin, quercitrin (quercetin-3-O-rhamnoside), and naringenin.
The data were acquired and processed using Thermo Trace Finder 2.1 software (Thermo Fisher Scientific Inc.; Waltham, MA, USA) based on own and internet databases: Metlin (La Jolla, CA, USA), Mass Bank of North America (Davis, CA, USA), m/z Cloud (HighChem; Bratislava, Slovakia). After processing, the results were manually checked using Thermo Xcalibur 4.0 software (Thermo Fisher Scientific Inc.; Waltham, MA, USA). The phytochemicals found in the extracts were identified on the basis of our previous published works or data found in literature using the exact molecular mass, isotopic pattern, characteristic fragmentation ions, and retention time.

2.4. Spectrophotometric Determication of the Total Polyphenol and Flavonoid Content, and the Antioxidant Activity of the Extracts

2.4.1. Total Polyphenol Content Determination

The total polyphenols content was determined by the Folin–Ciocalteu method [24]. An amount of 0.4 mL of extract was mixed with 1 mL Folin–Ciocalteu reagent, 15 mL distilled water, and 2 mL of sodium carbonate solution (290 g/L). The mixture was shaken and then kept in a water bath at 40 °C for 20 min. The absorbance was measured at 760 nm using a Cecil CE 1021 UV-Vis spectrophotometer. The calibration curve was determined, showing a linear range for concentrations of 0.9–4.3 µg/mL gallic acid. The total polyphenol content of extracts was expressed as mg gallic acid equivalents (GAE)/g dry weight (dw), using the following equation based on the calibration curve: y = 73.773x − 0.0198 (R2 = 0.9937), where x was the absorbance recorded at 760 nm and y was the concentration expressed as mg GAE/g dw.

2.4.2. Total Flavonoid Content

The total flavonoid content was determined by using a colorimetric method adapted after the Romanian Pharmacopoea [25]. In a 25 mL volumetric flask, 1 mL of the extract was mixed with 5 mL of a sodium acetate (100 g/L) and 3 mL of an aluminum chloride (25 g/L) solution and methanol to 25 mL. The mixture was shaken and left to stand for 15 min at room temperature before the analysis. The absorbance was recorded at 430 nm, using a Cecil CE 1021 UV-Vis spectrophotometer, and the results were expressed as mg quercetin equivalent (QE)/g dw. The calibration curve was determined, showing a linear range for concentrations 8.0–40.2 µg/mL quercetin. The equation based on the calibration curve: y = 27.627x − 0.0374 (R2 = 0.993), where x was the absorbance and y - the concentration expressed as mg QE/g dw.

2.4.3. Antioxidant Activity through DPPH Free Radical Scavenging Assay

The free radical 2,2-diphenyl-1-picrilhydrazyl (DPPH) was used to test the ability of our extracts to act as free radical scavengers or hydrogen donors and, hence, the evaluation of their antioxidant activity. DPPH free radical scavenging activity was determined by using the method proposed by Tylkowski and colleagues [26]. A stock solution of 25 µg/mL DPPH in methanol was prepared and kept at 4 °C in the dark for 2 h before usage. An amount of 970 µL of DPPH stock solution was added onto 30 µL of extract. The absorbance was recorded at 517 nm, using a Cecil CE 1021 UV-Vis spectrophotometer, and the results were expressed as percentages. The calibration curve was linear for the range of DPPH concentrations of 0.25–250 µg/mL. The equation based on the calibration curve: y = 0.0047x + 0.013 (R2 = 0.996), where x was the absorbance and y—the concentration expressed as µg DPPH per mL. The DPPH radical scavenging activity was determined by using the following formula: RSA (%) = [(C0 − C1)/C0]·100, where: RSA represents the DPPH radical scavenging activity (%), C0 represents the concentration of the DPPH stock solution (µg/mL), and C1 represents the DPPH concentration in the sample (µg/mL).

2.5. Drosophila Melanogaster Strains and Feeding Design

Wild type Drosophila melanogaster Canton Special flies were used in all experiments. The flies were acquired from Bloomington Drosophila Stock Center (Indiana University, USA). Flies were reared on a standard medium containing 6% (w/v) yeasts, 4% (v/v) molasses, 1.25% (w/v) agar, and 0.4% (v/v) propionic acid (as a mold growth inhibitor). For larvae cultivation, a bottle of flies (10–15 males + 10–15 females) was placed at 25 °C. The flies were allowed to lay eggs for 2–4 h, then the bottle was cleared of flies and incubated for 3–4 days at 25 °C, or until third instar larvae were visible. Some water was then added to the bottle with larvae and let sit for 10 min. The 3rd instar larvae crawled to the walls, were collected using a brush, and placed into a petri dish containing PBS. Then, larvae were gently washed and transferred to a mesh basket and placed tissue paper to dry. About 200 3rd instar larvae were transferred to Holidic medium, containing 50 mM sucrose, and prepared as described by Piper and colleagues [27]. After eclosion, the flies were transferred to experimental media, namely: Holidic media (control); Holidic media with additional 1.45 M sucrose (high-sugar diet); Holidic media and 3% (v/v) aqueous bilberry extract E1 (control + bilberries); Holidic media and additional 1.45 M sucrose and 3% (v/v) aqueous bilberry extract E1 (high-sugar diet + bilberries). The files were reared on such experimental media for 12 days at 25 °C, and further analyzed as described in the forthcoming paragraphs.

2.6. Drosophila Melanogaster Two-Choice Feeding Preference Assay

Flies were first starved for a day and then provided with the choice of two foods, presented on a Petri dish at concentrations that were used in our experiments. The two food choices also contained different tasteless colored substrates (sulforhodamine B dye—red and erioglaucine dye—blue). The feeding was carried out in darkness (to exclude any influence of color preference), and the abdomen of the flies was inspected visually on the next day. The flies feeding exclusively on one of the substrates were featuring the corresponding red or blue abdomens, while the flies feeding on both substrates presented purple coloured abdomens. Counting the flies can be used to determine the feeding preference index: PI (red) = [N(red) + 0.5N(purple)]/[N(red) + N(blue) + N(purple)].

2.7. Drosophila Melanogaster Hemolymph Glycemia Measurement

To determine the hemolymph specific trehalose concentration, 20 flies (10 females and 10 males) per sample were briefly knocked down with CO2, and then their thorax was poked under the wing or next to the legs. The fly bodies were then transferred to a haemolymph collection tube placed on ice and centrifuged for 6 min at 3000× g at 4 °C. A total of 0.5 μL of hemolymph was mixed with 100 μL of the Glucose (HK) Assay Kit (Sigma; G3293–50ML). A total of 0.5 μL of Porcine Trehalase (Sigma, T8778) per 10 μL of reaction was added to the mixture, incubated at 37 °C for 18 h, and analyzed with a fluorescent plate reader measuring absorption at 340 nm wavelength. Standard curves were generated from D-glucose (0–1000 mg/mL) standards for each trial.

2.8. Analysis of mRNA Levels of Neurohormonal Drosophila Melanogaster Genes by qRT-PCR

2.8.1. Total RNA Extraction and cDNA Synthesis

Male flies were decapitated after immersion in liquid nitrogen. We prepared three total RNA samples per treatment. For each sample 50 fly heads were placed into RNA stabilization solution (TRIzol) and kept at 4 °C until RNA extraction. The heads were homogenised in 1.5 mL RNase-free Eppendorf tubes containing 1 mL TRIzol reagent using a 1.5 mL pestle (Astral, RNase- and DNase-free; Thermo Fisher Scientific Inc.; Waltham, MA, USA). Samples were incubated for 15 min at room temperature and centrifuged for 10 min at 12,000× g at 4 °C. An amount of 800 µL of supernatant was decanted off and 200 µL of chloroform added. Tubes were shaken vigorously for 15 s, incubated at room temperature for 3 min and centrifuged for 20 min at 12,000× g at 4 °C. The aqueous phase was transferred to a Direct-zol™ RNA MiniPrep column (Zymo Research R2070; Freiburg, Germany), and all other steps were performed according to the manufacturer’s protocol. Extraction was followed by a DNase treatment (DNaseI, Invitrogen; Thermo Fisher Scientific Inc.; Waltham, MA, USA) to eliminate potential genomic DNA in the sample parameters. RNA was then stored at −80 °C before further processing. The quality and quantity of RNA were assessed by Qubit RNA HS Assay (Thermo Fisher Scientific Inc.; Waltham, MA, USA) and with an Agilent Bioanalyser RNA 6000 Pico Kit (Agilent Technologies, Inc.; Santa Clara, CA, USA). Synthesis of cDNA and subsequent quantification of mRNA levels for specific genes were performed using SuperScript™ III First-Strand Synthesis SuperMix for qRT-PCR (Thermo Scientific 11752050; Waltham, MA, USA). The cDNA samples were stored at −20 °C until further use.

2.8.2. Primer Design

The real-time qPCR assays were designed by using the Roche Universal Probe Library (Roche Molecular Systems, Inc.; Basel, Switzerland). Primers used for the analysis were designed by the Roche Probe Finder Assay Design Software [28]. The designed primers covered an exon–intron boundary to prevent genomic DNA amplification. The PCR primer sequences are shown in Table S7.

2.8.3. Reverse Transcription qPCR

On a Roche Light Cycler 480 Real Time PCR System, triplicate first strand cDNA aliquots for each sample served as templates for RT-qPCR using the Light Cycler 480 Probes Master PCR Master Mix (Roche Molecular Systems, Inc.; Basel, Switzerland). The amplification reactions were performed in 10 mL total volumes with 3 mL of cDNA (diluted 1:100) and 100 nM of each primer, in 384-well optical plates (LightCycler® 480 Multiwell Plate 384, Roche Molecular Systems, Inc.; Basel, Switzerland). The program conditions were: pre-incubation 1 cycle (95 °C, no acquisition, 10 min, 4.8 ramp rate), amplification 45 cycles (95 °C, no acquisition, 10 s, 4.8 ramp rate; 60 °C, single acquisition, 30 s, 2.5 ramp rate; 72 °C, no acquisition, 1 s, 4.8 ramp rate), and cooling 1 cycle (40 °C, no acquisition, 30 s, 2.5 ramp rate). Reverse transcription-qPCR efficiency was determined for each gene with the slope of a linear regression model. Relative standard curves for the gene transcripts were generated with serial dilutions of cDNA.

2.8.4. Data Mining and Selection of Reference Gene Candidates

Expression levels were determined as the number of cycles needed for the amplification to reach a fixed threshold in the exponential phase of the PCR reaction [29]. The number of cycles is referred to as the quantification cycle (Cq) value, the standard name for the threshold cycle or crossing point value according to the RDML guidelines [30]. The threshold was set at 0.045 for all genes, and the corresponding Cq values were transformed into quantities via the standard curve using PCR efficiencies according to [31].

3. Results

3.1. Chemical Analysis of the Extracts

The aqueous (E1), methanolic (E2), and hydro-methanolic with 1:1 water:methanol ratio (E3) extracts of bilberry ripe fruit underwent UHPLC-ESI-MS/MS qualitative analysis in order to determine their phytonutrient profile. The chromatograms in positive and negative ionization mode are shown in Figure 1 for all analyzed extracts, with labeled retention times of major peaks. The data recorded can be found in Table S1 (E1), Table S2 (E2), and Table S3 (E3). The fragmentation patterns used in the putative identification of the total of 88 individual phytochemicals are summarized in Table S4.
This analysis revealed several differences in the composition of the extracts of the bilberry ripe fruit depending on the applied extraction protocol used. The comparative analysis of the composition is presented in Table 1, and it also contains the chemical classification of the compounds. A total of 88 biomolecules were identified in the ripe bilberry fruit through our analysis. Out of these, the most phytochemicals, 81 different compounds, were identified in the hydro-methanolic extract, while 79 compounds were identified in the methanolic, and the lowest number, namely 65 compounds, were observed in the aqueous extract.
The chemo-mapping of the extracts based on the number of members of each chemical class is also summarized in Figure 2. More than half of the phytochemicals identified in the extracts are representatives of the flavonoid class—48 compounds out of 88. The classes of amino acids (12 compounds), polyphenols (6 compounds), carboxylic acids (5 compounds), and vitamins (4 compounds) were also revealed through the qualitative analysis of the composition of bilberry ripe fruit extracts.
Looking at the chemical classification and the identified number of the bilberry specific phytoconstituents, it seems obvious that the flavonoids represent the largest category followed by amino acids, polyphenols, carboxylic acids, and vitamins (Table 1, Figure 2).
It is also interesting that choline, the uniquely identified alkaloid in our study, was present only in the aqueous extract (E1), while the 7-deoxyloganic acid representing the iridoids was observed exclusively in the methanolic (E2) and the hydro-methanolic (E3) extracts. Other differences regarding the composition of the phytonutrient profiles of the assessed bilberry extracts were also evident (Table 1), confirming the importance of the applied extraction conditions.
To continue the phytonutrient profile characterization we have shown that from the three extraction formulations, the hydro-methanolic extract (E3) presented the highest amounts of total polyphenols (21.68 mg GAE/g dw) and flavonoids (8.41 mg QE/g dw), closely followed by the methanolic extract (E2), while the lowest quantities of these compounds were found in the aqueous extract (E1), at roughly half of the content. Thus, the hydro-alcoholic solvent was proven to extract the compounds of interest more efficiently than methanol or water alone (Table 2).
The in vitro antioxidant activity of the extracts was determined based on the DPPH free radical scavenging assay. The obtained antioxidant activity data suggest a trend that parallels the total polyphenolic and flavonoid content of the analyzed extracts (Table 2), meaning that the hydro-methanolic extract (E3) having the highest total polyphenol and flavonoid contents showed the strongest antioxidant activity, while the aqueous extract (E1) specific lowest total polyphenol and flavonoid content was associated with the most reduced antioxidant activity.

3.2. In Vivo Drosphila Melanogaster Studies of the Rescue-Effect of the Hydro-Methanolic Extract (E3)

Seeing the plenitude of phytonutrients associated with our bilberry extracts, and preparing to analyze the associated biological effects, we set to avoid the false positive data due to the preference or avoidance of the experimental fruit fly culture media. Therefore, a two-choice preference assay was carried out, and the feeding preference index was determined for each culture media. There was no preference or avoidance observed towards any of the experimental media used in further experiments, with an average preference index of about 0.5 (Table S9).
Experiemntal data concerning the individual phytonutrients present in biberry have been shown to generate a strong antidiabetic physiological effect (Table S6). Our data support this finding, so that flies reared on a high-sugar diet and supplemented with hydro-methanolic bilberry extract (E3) showed a reduction in hemolymph trehalose, reaching similar levels to those observed in the normal diet control (Figure 3), and significantly lower levels than those of flies reared on the high-sugar diet without extract supplementation. The choice for the hydro-methanolic bilberry ripe fruit extract (E3) formulation was based on its highest polyphenol and flavonoid content, highest antioxidant activity (Table 2), and highest number of phytochemicals present (Table 1).
Additional to the doubled hemolymph trehalose levels exhibited by flies reared on the high-sugar diet as compared to the normal diet control, such flies also featured modified gene expression profiles for a set of genes encoding for neuropeptides (Figure 4). Among the analyzed genes, 11 exhibited elevated mRNA levels, with the highest increase observed in the case of Akh (above 2-fold) followed by InR and AstA (about 1.5-fold). Genes such as Ilp6, Nplp2, AstC, tobi, Hr96, and usp exhibited foldchanges between 1.14 and 1.35, while pdf and Eip75B showed very slight overexpression, with values close to their constitutive levels. From the analyzed genes, pronounced downregulation of mRNA expression was observed in the case of Nplp3, CCAP, Npc2g, CCHa2, Irk1, Ide, and CG15618, with less than half of the gene expression levels when the flies were reared for 12 days on a high-sugar diet as compared to the normal diet control. Slightly downregulated expression levels (below 20% decrease) were also apparent for Ilp2, Ilp5, SIFa, CNMa, and Thor genes.
Upon high-sugar diet supplementation with 3% hydro-methanolic bilberry extract (E3), the foldchanges in mRNA expression of the analyzed neuropeptide-encoding genes were more dramatic (Figure 5). The Nplp2 and Nplp3 genes were more than 20-fold overexpressed, while for genes such as Npc2g, CCHa2, and Eip75B, the overexpression exceeded a 2-fold increment as compared to their corresponding controls. Moreover, we could observe the severe downregulation of other genes like Ilp2, Ilp5, Akh, CCAP, SIFa, tobi, and Ide, whose mRNA expression decreased below 30% of the respective control.

4. Discussion

The chemical evaluation of the three bilberry extracts revealed the hydro-methanolic extract (E3) containing the highest number of phytochemicals, and showing the highest content of polyphenols and flavonoids, along with the highest antioxidant activity, followed by the methanolic extract (E2) and, lastly, by the aqueous extract (E1) (Table 1 and Table 2, Figure 2). Our results are in line with previous studies, suggesting that the bioactive compound content varies upon altitude, habitat type, and site conditions [24,25,26]. For reference, our hydro-methanolic extract presented 21.68 mg GAE/g dw total polyphenols and 8.41 mg QE/g dw flavonoids, while a similar study on bilberries from another region of Romania reported a total polyphenolic content of 34,7 - 41,9 mg GAE/g dw [32]. Similarly, a study from Turkey revealed total polyphenols of 20.06 mg GAE/g dw and total flavonoids of 2.67 mg QE/g dw [33], while another study from Germany had total polyphenols of 29.7 mg GAE/g dw, and total flavonoids of 13.5 mg QE/g dw [34].
The chemo-mapping of our extracts offers new insights into their composition, as to our knowledge, the previously reported bilberry extracts were highly focused on the anthocyanin content [2,3,4,5,6], rather than the complete phytochemical profile (Table 1). The identified chemical classes (Figure 2) are in agreement with reported composition of Vaccinium myrtillus L. extracts (Table S5) [35,36,37,38,39,40,41,42,43,44]. Moreover, they are in accordance with reported composition of the bilberry fruit, based on the report of the European Medicines Agency [35].
Noteworthy, the polyphenol and flavonoid content decrease with the increase in the degree of fruit ripening [35]. Oppositely, anthocyanosides—more than half of the molecules giving the color in Vaccinium spp.—increase in concentration when the fruit approaches maturation [35]. Carboxylic acids, vitamins, and miscellaneous classes in our samples are highly similar in composition to those reported in previous studies [35]. Regarding alkaloids and iridoids, their presence is generally scarce, especially in ripe fruit, with as little as one and two representatives [35], which is in accordance with our findings.
Approximately 40% of the compounds identified in our extracts have not been previously reported in Vaccinium spp., while 3 phytochemicals (cinnamtannin B1, cinnamtannin D1, and quercetin-3-O-galactoside) were only described in other species, and not for Vaccinium myrtillus L. (Table S5; Table S6—superscripts A, B, C) [35,36,37,38,39,40,41,42,43,44]. Regarding the substances not reported earlier, it is, however, important to mention that the free amino acids presented in our study pertain to this category. This might be due to a mainly anthocyanin focused bilberry specific research, omitting, hence, the free amino acid class of compounds, though the later might confer substantial nutritional values to the bilberry extracts.
Furthermore, we summarized the already reported biological effects for each phytochemical identified in the studied bilberry extracts (Figure 6, Table S6) [45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254,255,256,257,258,259,260,261]. More than half of the characterized substances are reported in literature to behave like antioxidant agents. Such a category includes free radical scavengers; antilipid peroxidation agents; and molecules that inhibit or deter free radical formation, protect the target, or repair the damage produced due to oxidative stress (Table S6). The pronounced presence of various phytonutrients with antioxidant function strongly indicates that the bilberry extracts as a whole might also aid in the reduction in oxidative stress, despite the fact that the implicated coping mechanisms at cellular level are currently scarcely documented. Some already existing research data accentuate the antioxidant feature of bilberry extracts largely attributed to the rich anthocyanin content of bilberries [2,3,4,5,6,7]. Moreover, the in vitro antioxidant property of the assessed three bilberry extracts is further substantiated by the DPPH free radical scavenging assay, suggesting once again the superiority of the hydro-methanolic extract (E3) over the other formulations (Table 2).
Besides phytonutrients with antioxidant features, many others also had immune response modulator, antidiabetic, anticancer, cardiovascular protective, and neuroprotective properties (Figure 6). Interestingly, some bilberry extract-specific phytonutrients, when assessed individually using different experimental setups, have been shown to generate multiple in vivo effects. Therefore, the investigation of a bilberry extract associated with biological functions calls for a systemic approach that will let us combine genetic, nutritional, and physiological methodologies using in vivo animal models.
In line with the abovementioned considerations, we decided to take advantage of a Drosophila melanogaster-based nutritional genetic in vivo model for which normal media and high-sugar types of diets generated diabetic modifications could be assessed upon bilberry extract supplementation. Among the in vivo experiments, the most compellingantidiabetic rescue effect of the bilberry extract is related to the hemolymph trehalose levels that are restored to the levels of normal diet controls upon addition of current hydro-methanolic bilberry extract (E3) to the high-sugar diet (Figure 2, Table S8). When reared on high-sugar media, the level of hemolymph trehalose reached an average of 943 mg/L, while upon addition of the hydro-methanolic extract (E3), the level of hemolymph trehalose fell to an average of 447 mg/L, comparable to the levels in control normal diet averaging 457 mg/L. Similarly, Musselmann and colleagues demonstrated an approximate 2-fold increase in hemolymph trehalose levels induced by a high-sugar diet [16]; such an observation would strengthen our claim for the rescue effect exhibited by the hydro-methanolic bilberry extract (E3).
Furthermore, the hydro-methanolic bilberry extract (E3) was tested in the abovementioned dietary conditions regarding its putative rescue effects that might compensate for the high-sugar diet generated diabetic carbohydrate and lipid metabolisms, inflammation, neurodegeneration, and other Met-S associated pathologies. In this context, we chose to study the genetic control of neurohumoral regulation by assessing the mRNA expression level of several genes encoding for neuropeptides. Some of the genes in question were selected based on previously conducted studies of high-sugar diet experiments in Drosophila [16,17,18].
The Drosophila insulin-like proteins (Ilps) are released in response to high levels of circulating sugar, while a glucagon-like molecule, the adipokinetic hormone (AKH), is synthetized in response to low levels of circulating sugar [262,263,264,265]. Our result shows very slight differences, less than 15%, between the gene expression of Ilp(s) in flies reared for 12 days on a high-sugar diet, as compared to those fed on a normal diet control. In the case of hydro-methanolic bilberry extract (E3) supplementation, the Ilp(s) expression levels are observed to be drastically reduced for Ilp2 and Ilp5 reaching about 20% of the control levels, while they are about 50% increased for Ilp6. This corresponds to the observed activity of Ilps, as Ilp6 is an insulin receptor agonist which has been shown in other contexts to work in opposition to the major circulating Ilps [266,267]. Upon starvation, Ilp6 is induced in the fat body and acts on the insulin-producing cells to downregulate Ilp2 and Ilp5 [268]. In our study, the expression of Akh is raised by the chronic exposure to elevated dietary sugar, with a foldchange comparable to the previously reported findings [16,17]. As noted also by previous authors, the increase in expression levels of Akh could lead to a paradoxal mobilization of stored fat and carbohydrates [16,265], without affecting metabolic rate but reducing food intake [265]. The rescue effect of the bilberry extract is also remarkedly pronounced in the case of Akh gene, whose expression is reduced at about 95% as compared to the appropriate control, suggesting that the bilberry extract could compensate for the high-sugar diet affected Ilps and Akh genes.
InR, the fruit fly insulin receptor, mediates functions similar to those of insulin and insulin-like growth factor receptors in humans [269]. In our study, it presents a rescue effect upon bilberry extract supplementation of the high sugar diet, with above 60% overexpression observed when adult flies are reared on a high-sugar diet alone, and reduced overexpression to about 35% in the case of hydro-methanolic bilberry extract (E3) supplementation of the high-sugar diet as compared to the respective controls. The expression levels of InR oppose those of Ilp2 and Ilp5, corresponding to previously described trends, and reinforcing the suitability of Drosophila to study action mechanisms generated by bilberry in relation to diabetes, insulin resistance, and other associated disease [270].
Among the assessed genes, AstA, AstC, CCHa2, Nplp2, and Nplp3 are mainly expressed in the Drosophila gut, and the expression levels of these enteroendocrine peptides might fluctuate, depending on the nutritional state and dietary conditions [271]. Our results show a rescue effect upon addition of the hydro-methanolic extract (E3) to the diet for the gene expression levels of AstA, AstC, and CCHa2. The most dramatic foldchanges in expression levels were observed for Nplp2 and Nplp3, reaching above 20-fold increases upon extract supplementation to the high-sugar diet as compared to their controls. However, the exact functional relevance of Nplp2 and Nplp3 as neuropeptides remains elusive.
Starting 24 h from eclosion, only 2 residual neurons in Drosophila express CCAP (crustacean cardioactive peptide), a gene extensively studied in the context of ecdysis [272,273], recently proved to signal for food intake and regulate the triglyceride metabolism. The adult flies lacking the CCAP peptide had significantly lower triglyceride levels [274]. This correlates with the expression levels observed by us upon supplementation of the high-sugar diet with hydro-methanolic bilberry extract (E3). The CCAP expression levels shows a 2-fold decrease for the high-sugar diet, while a 30-fold decrease is seen for the extract-supplemented high-sugar diet, as compared to their appropriate controls. Our observations related to the expression of the CCAP gene suggest its possible implication in the triglyceride lowering effect of the bilberry extract.
Npc2g is a neuropeptide that plays a role in the cholesterol breakdown, absorption, and trafficking, along with Hr96 [275,276]. In our study, a counteracting rescue effect on the gene expression of Npc2g is observed, with a 3-fold increase in the case of hydro-methanolic bilberry extract (E3) supplementation to the high-sugar diet, while presenting a 3-fold decrease in the case of the high-sugar diet alone, as compared to their appropriate controls. The Npc2g specific gene expression profile indicates the bilberry extract possible implication in cholesterol homeostasis. However, no significant modifications could be observed in the case of the expression levels of Hr96 gene.
The SIFa gene has been observed to have a function in food uptake by enhancing the taste-guided and odor-guided appetitive behavior [277]. No change is observed in our experiments when comparing the expression levels of the SIFa gene in flies having a high-sugar diet or a normal diet control regimen; however, there is a 4-fold decrease in its expression observed upon hydro-methanolic bilberry extract (E3) addition to the high-sugar diet. However, the effect of this modification on the Drosophila metabolism is still unclear.
The pdf is another gene involved in the feeding behavior. While it is not described as regulating the carbohydrate or fat metabolisms in Drosophila, its function as a major synchronization and output factor of the circadian clock [278] is well studied as affecting the timing of sleep and feeding [279,280], with AstA-expressing PLP neurons as a downstream target of the clock output factor PDF [281]. While a rescue effect of of the hydro-methanolic bilberry extract (E3) is observed for the AstA gene expression levels, the same trend of downregulation is observed for the expression of pdf, as its expression changes only in the case of extract-supplemented high-sugar diet, decreasing 3-fold as compared to the respective control.
Despite the undetermined function of CNMa protein [282], it is noteworthy that the CNMa gene expression patterns under the test conditions are similar to those of pdf and SIFa, with very slight variations caused by the high-sugar diet, and about 3-fold decrease measured upon addition of hydro-methanolic bilberry extract (E3).
The tested transcript levels of Thor gene were previously considered as a readout of the peripheral insulin signaling [283]. Thor is a target of the evolutionarily conserved transcription box Forkhead, which is repressed by insulin signaling [284,285]. Hence, Thor is repressed by the active peripheral insulin signaling [284,285], and increased brain insulin levels are expected to reduce Thor expression levels. In contrast to this expectation, it was observed that Thor transcript levels are unaffected in Akh loss-of-function mutants, suggesting that the peripheral insulin signaling is impaired in response to AKH deficiency [283]. On the other hand, our results indicate no significant changes in the Thor expression levels as compared to the extent of Akh expression pattern. The high-sugar diet with or without extract supplementation experiments did not provide us relevant functional cues in relation to the Akh-Thor previously suggested interdependence on a normal diet.
Different from the Thor expression pattern, the transcription levels of tobi, another insulin target that encodes for a putative amyloglucosidase [283], shows a rescue effect due to the hydro-methanolic bilberry extract (E3) supplementation of the high-sugar diet as compared to the controls. Our observations are in contrast with the findings of Galikova and colleagues, where Akh over-expression led to the non-significant modification of the tobi mRNA levels [283]. Taken together, our data suggest that the expression pattern of the Akh-Thor and tobi genes seems to depend considerably on dietary conditions.
The gene called inwardly rectifying potassium channel1 (Irk1) is responsible for transepithelial ion transport in the Drosophila renal tubule and is additive to Na+/K+-ATPase-dependent pathways [286]. Its main functions are iono- and osmoregulation, and along with the intricate links with sugar and fat metabolism, in our study, there is a rescue effect exhibited by extract supplementation under these test conditions, where the Irk expression is downregulated by high-sugar diet alone, whilst it is overexpressed upon hydro-methanolic bilberry extract (E3) supplementation.
Insulin-degrading enzyme (IDE) is a neutral zinc and thiol-dependent metallopeptidase, for which increasing evidence suggests its dysfunction leads to pathogenesis of type II diabetes [287,288,289,290]. Moreover, the enzymatic activity of Ide targets, besides insulin, also other hormones, suggesting a complex contribution of Ide to the regulation of carbohydrate metabolism [291]. In our analysis, the high-sugar diet alone induces the downregulation of Ide expression, while supplementation of bilberry extract enhances the downregulation under tested conditions.
The gene Eip75B plays a role in steroid signaling, regulating the circadian rhythm and protecting the central clock against environmental stressors in Drosophila [292]. It was previously proven that nutrition rich in sugars, used as a stressor, can induce arrhythmic or weak circadian behavior upon Eip75B knockdown [292]. Hence, it could be hypothesized that the 2.75-fold upregulation of the Eip75B expression on our high-sugar diet supplemented with hydro-methanolic bilberry extract (E3) is either a result of additional stress induced by the extract, or a boost for the expression of Eip75B meant to help in compensating for the nutritional stressor.
The gene CG15618 has been identified as the Drosophila ortholog of THADA (Thyroid adenoma-associated protein homolog) [293], the latter being identified as one of the top risk loci for type 2 diabetes in human genome-wide association studies [294]. In Drosophila, the CG15618 acts like a metabolic regulator that would match fat storage and heat production. In our experiments, the obese and hyperphagic phenotypes specific for the reduced expression of CG15618 were not recapitulated (data not shown). Moreover, the similar level of downregulation of CG15618 expression, seen for both the bilberry extract-supplemented high-sugar diet and the high-sugar diet alone, would indicate that the applied dietary conditions do not affect or interfere with the functions of the gene in question.
The Usp (ultraspiracle) protein belongs to a family of nuclear hormone receptors that are ligand-inducible transcription regulators [295]. The usp gene is the insect ortholog of the vertebrate retinoid X receptor (RXR) [296], similarly to which it heterodimerizes with nuclear receptors to form active receptor complexes. Its main interaction partner is the ecdysone receptor (EcR), the receptor of ecdysteroids, insect steroid hormones that control development, reproduction, together with starvation response [297]. It is unclear what function it holds in the context of our experimental setup; however, it is observed that usp expression is about 30% upregulated for flies reared on a high-sugar diet, both with and without bilberry extract supplementation.
Taken together, the application of the high-sugar diet and Drosophila-based experimental approach to study the bilberry extract induced in vivo effects looks rather promising and recapitulates not only metabolic syndrome-related aspects but could also give a sneak peek at other diet influenceable pathologies. Besides the bilberry relatedantidiabetic aspects of neurohormonal regulation, it would be equally important to look at the genetic control of antioxidant andanticancer effects upon dietary conditions that could widen or restrict the preventive/therapeutic applicability of bilberries. For example, from the standpoint of colorectal cancer, the 3rd most common type of cancer in the world and the 2nd most responsible for the number of cancer-related deaths worldwide [298], the elevated levels of Thor expression represent a poor prognosis indicator, and hence it is considered a potential colorectal cancer biomarker [299]. We were able to demonstrate for Drosophila that the high-sugar diet alone or supplemented with bilberry extract could not significantly modify the expression levels of Thor, suggesting that the relevance of Thor expression level and the peripheral insulin sensitivity connection has to be carefully reconsidered in the context of future colorectal cancer studies. Noteworthy, the above presented picture would become even more complicated if we were to consider the Akh-Thor connections too.

5. Conclusions

In conclusion, our study on bilberry revealed a complex phytonutrient profile that could be associated withantidiabetic effects, such as efficiently reducing the haemolymph trehalose levels using a Drosophila melanogaster based high-sugar diet model. Moreover, the bilberry induced beneficial effect is substantiated by reversing the affected expression level of some genes implicated in the neuro-hormonal control of the high-sugar diet induced pathologies. Our results also suggest that the beneficial effects of diet-based supplementation of bilberry would generate multiple compensatory mechanisms as seen for Drosophila genes such as InR, Akh, AstA, AstC, Irk, Npc2g, and CCHa2. Defining the interdependence of the abovementioned genes and assessing the whole genome response upon diet supplementation with bilberry requires further transcriptomic studies that undoubtedly could generate rather advanced interactomes explaining the complexity of functional consequences. In a larger context, our data contribute to a better understanding of the metabolic syndrome associated pathologies, such as type II diabetes and various cancers [20,21,22,23,298], alongside the associated genetic and environmental risk factors, and the elucidation of the bilberry-based preventive/therapeutic diet-induced genetic and physiological consequences.

Supplementary Materials

Author Contributions

A.-A.N. and R.S.-K. contributed equally to this work and are both considered first authors. Conceptualization: E.M. (Endre Mathe), C.G., C.N., V.T. and N.K.O. Methodology: A.-A.N., R.S.-K., A.F., Z.C. and E.M. (Emoke Mihok). Validation: Z.C. and Z.S.-K. Formal analysis: A.-A.N., O.T., N.K.O. and Z.S.-K. Resources: E.M. (Endre Mathe), V.T., Z.C., O.T. and C.G. Writing—original draft preparation: A.-A.N., R.S.-K., and E.M. (Endre Mathe). Writing—review and editing: A.-A.N., R.S.-K., E.M. (Emoke Mihok), C.N., V.T. and N.K.O. Visualization: Z.C. and Z.S.-K. Supervision: C.G. and E.M. (Endre Mathe). Project administration: E.M. (Endre Mathe). All authors have read and agreed to the published version of the manuscript.

Funding

The research and publication is supported through the EFOP-3.6.3-VEKOP-16-2017-00008 grant. The project is co-financed by the European Union and the European Social Fund.

Acknowledgments

We are thankful to Adrian Maghiar (University of Oradea) for fruitful discussions and the interest in evidence-based effects of plant derived extracts, and their inclusion, as adjuvant treatment, in future therapeutic protocols.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Riihinen, K.; Jaakola, L.; Kärenlampi, S.; Hohtola, A. Organ-specific distribution of phenolic compounds in bilberry (Vaccinium myrtillus) and ‘northblue’ blueberry (Vaccinium corymbosum × V. angustifolium). Food Chem. 2008, 110, 156–160. [Google Scholar] [CrossRef] [PubMed]
  2. Directorate-General for Research and Innovation—European Commission. Publications Office of the European Union: Luxembourg, Luxembourg. Functional Foods. 2010. Available online: https://op.europa.eu/en/publication-detail/-/publication/238407ee-0301-4309-9fac-e180e33a3f89 (accessed on 10 June 2020).
  3. Fraisse, D.; Bred, A.; Felgines, C.; Senejoux, F. Screening and Characterization of Antiglycoxidant Anthocyanins from Vaccinium myrtillus Fruit Using DPPH and Methylglyoxal Pre-Column HPLC Assays. Antioxidants 2020, 9, 512. [Google Scholar] [CrossRef] [PubMed]
  4. Ștefănescu, B.-E.; Călinoiu, L.F.; Ranga, F.; Fetea, F.; Mocan, A.; Vodnar, D.C.; Crișan, G. Chemical Composition and Biological Activities of the Nord-West Romanian Wild Bilberry (Vaccinium myrtillus L.) and Lingonberry (Vaccinium vitis-idaea L.) Leaves. Antioxidants 2020, 9, 495. [Google Scholar] [CrossRef] [PubMed]
  5. Diaconeasa, Z.; Iuhas, C.I.; Ayvaz, H.; Rugină, D.; Stanilă, A.; Dulf, F.; Bunea, A.; Socaci, S.A.; Socaciu, C.; Pintea, A. Phytochemical Characterization of Commercial Processed Blueberry, Blackberry, Blackcurrant, Cranberry, and Raspberry and Their Antioxidant Activity. Antioxidants 2019, 8, 540. [Google Scholar] [CrossRef] [Green Version]
  6. Ștefănescu, B.E.; Szabo, K.; Mocan, A.; Crişan, G. Phenolic Compounds from Five Ericaceae Species Leaves and Their Related Bioavailability and Health Benefits. Molecules 2019, 24, 2046. [Google Scholar] [CrossRef] [Green Version]
  7. Muller, D.; Schantz, M.; Richling, E. High performance liquid chromatography analysis of anthocyanins in bilberries (Vaccinium myrtillus L.), blueberries (Vaccinium corymbosum L.), and corresponding juices. J. Food Sci. 2012, 77, 340–345. [Google Scholar] [CrossRef] [PubMed]
  8. Ştefănescu, R.E.; Eșianu, S.; Laczkó-Zöld, E.; Mare, A.; Tudor, B.; Dogaru, M.T. Short Period Storage Impact on Bioactive Constituents from Bilberries and Blueberries. Acta Med. Marisiensis 2017, 63, 87–90. [Google Scholar] [CrossRef] [Green Version]
  9. Rodriguez-Mateos, A.; Cifuentes-Gomez, T.; Tabatabaee, S.; Lecras, C.; Spencer, J.P. Procyanidin, anthocyanin, and chlorogenic acid contents of highbush and lowbush blueberries. J. Agric. Food Chem. 2012, 60, 5772–5778. [Google Scholar] [CrossRef]
  10. Åkerström, A. Factors affecting the Anthocyanidin Concentration in Fruits of Vaccinium myrtillus. Acta Uni Agric. Sued. Agrar. 2010, 52, 1652–6880. Available online: https://pub.epsilon.slu.se/2342/ (accessed on 29 September 2020).
  11. Seeram, N.P. Berry Fruits: Compositional Elements, Biochemical Activities, and the Impact of Their Intake on Human Health, Performance, and Disease. J. Agric. Food Chem. 2008, 56, 627–629. [Google Scholar] [CrossRef]
  12. Chan, S.W.; Tomlinson, B. Effects of Bilberry Supplementation on Metabolic and Cardiovascular Disease Risk. Molecules 2020, 25, 1653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bujor, O.-C.; Tanase, C.; Popa, M.E. Phenolic Antioxidants in Aerial Parts of Wild Vaccinium Species: Towards Pharmaceutical and Biological Properties. Antioxidants 2019, 8, 649. [Google Scholar] [CrossRef] [Green Version]
  14. Pires, T.C.S.P.; Caleja, C.; Santos-Buelga, C.; Barros, L.; Ferreira, I.C. Vaccinium myrtillus L. Fruits as a Novel Source of Phenolic Compounds with Health Benefits and Industrial Applications—A Review. Curr. Pharm. Des. 2020, 26, 1917–1928. [Google Scholar] [CrossRef] [PubMed]
  15. Brasanac-Vukanovic, S.; Mutic, J.; Stankovic, D.M.; Arsic, I.; Blagojevic, N.; Vukasinovic-Pesic, V.; Tadic, V.M. Wild Bilberry (Vaccinium myrtillus L.; Ericaceae) from Montenegro as a Source of Antioxidants for Use in the Production of Nutraceuticals. Molecules 2018, 23, 1864. [Google Scholar] [CrossRef] [Green Version]
  16. Musselman, L.P.; Fink, J.L.; Narzinski, K.; Ramachandran, P.V.; Hathiramani, S.S.; Cagan, R.L.; Baranski, T.J. A high-sugar diet produces obesity and insulin resistance in wild-type Drosophila. Dis. Model. Mech. 2011, 4, 842–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Musselman, L.P.; Fink, J.L.; Ramachandran, P.V.; Patterson, B.W.; Okunade, A.L.; Maier, E.; Baranski, T.J. Role of fat body lipogenesis in protection against the effects of caloric overload in Drosophila. J. Biol. Chem. 2013, 288, 8028–8042. [Google Scholar] [CrossRef] [Green Version]
  18. Graham, P.; Pick, L. Chapter Thirteen—Drosophila as a Model for Diabetes and Diseases of Insulin Resistance. Curr. Top. Dev. Biol. 2017, 121, 397–419. [Google Scholar] [CrossRef] [Green Version]
  19. Liu, H.-Y.; Walden, T.B.; Cai, D.; Ahl, D.; Bertilsson, S.; Phillipson, M.; Nyman, M.; Holm, L. Dietary Fiber in Bilberry Ameliorates Pre-Obesity Events in Rats by Regulating Lipid Depot, Cecal Short-Chain Fatty Acid Formation and Microbiota Composition. Nutrients 2019, 11, 1350. [Google Scholar] [CrossRef] [Green Version]
  20. Delli Bovi, A.P.; Di Michele, L.; Laino, G.; Vajro, P. Obesity and Obesity Related Diseases, Sugar Consumption and Bad Oral Health: A Fatal Epidemic Mixtures: The Pediatric and Odontologist Point of View. Transl. Med. UniSa 2017, 16, 11–16. Available online: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC5536157/pdf/tm-16-11.pdf (accessed on 18 September 2020).
  21. World Health Organization. Guidelines—Sugars Intake for Adults and Children. 2015. Available online: http://www.who.int/nutrition/publications/guidelines/sugars_intake/en/ (accessed on 18 September 2020).
  22. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [Green Version]
  23. National Center for Health Statistics, Division of Health Interview Statistics, Centers for Disease Control and Prevention, Division of Diabetes Translation. Crude and Age-adjusted Percentage of Civilian, Noninstitutionalized Adults with Diagnosed Diabetes, United States, 1980–2010; National Center for Chronic Disease Prevention and Health Promotion: Atlanta, GA, USA, 2012.
  24. Frum, A.; Georgescu, C.; Gligor, F.G.; Lengyel, E.; Stegarus, D.I.; Dobrea, C.M.; Tita, O. Identification and Quantification of Phenolic Compounds from Red Grape Pomace. Sci. Stud. Res. Chem. Chem. Eng. Biotechnol. Food Ind. 2018, 19, 1–8. Available online: http://pubs.ub.ro/dwnl.php?id=CSCC6201801V01S01A0005 (accessed on 7 May 2020).
  25. Soroceanu, V. Romanian Pharmacopoeia, 10th ed.; Medical Publishing House Bucharest: Bucharest, Romania, 1993. [Google Scholar]
  26. Tylkowski, B.; Tsibranska, I.; Kochanov, R.; Peev, G.; Giamberini, M. Concentration of Biologically Active Compounds Extracted from Sideritis Ssp. L. by Nanofiltration. Food Bioprod. Process. 2011, 89, 307–314. [Google Scholar] [CrossRef]
  27. Piper, M.D.W.; Blanc, E.; Leitão-Gonçalves, R.; Yang, M.; He, X.; Linford, N.J.; Hoddinott, M.P.; Hopfen, C.; Soultoukis, G.A.; Niemeyer, C.; et al. A holidic medium for Drosophila melanogaster. Nat. Methods 2014, 11, 100–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Roche LifeScience—Universal ProbeLibrary Assay Design Center. Available online: https://lifescience.roche.com/en_gb/brands/universal-probe-library.html#assay-design-center (accessed on 5 May 2019).
  29. Walker, N. A Technique Whose Time Has Come. Science 2002, 296, 557–559. [Google Scholar] [CrossRef] [PubMed]
  30. Lefever, S.; Hellemans, J.; Pattyn, F.; Przybylski, D.R.; Taylor, C.; Geurts, R. RDML Consortium. RDML: Structured language and reporting guidelines for real-time quantitative PCR data. Nucleic Acids Res. 2009, 37, 2065–2069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Vandesompele, J.; De Preter, K.; Pattyn, F.; Poppe, B.; Van Roy, N.; De Paepe, A.; Speleman, F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002, 3, 34–41. Available online: https://0-link-springer-com.brum.beds.ac.uk/protocol/10.1385/1-59745-229-7:205 (accessed on 22 September 2020). [CrossRef] [Green Version]
  32. Bujor, O.C.; Le Bourvellec, C.; Volf, I.; Popa, V.I.; Dufour, C. Seasonal Variations of the Phenolic Constituents in Bilberry (Vaccinium Myrtillus L.) Leaves, Stems and Fruits, and Their Antioxidant Activity. Food Chem. 2016, 213, 58–68. [Google Scholar] [CrossRef]
  33. Saral, Ö.; Ölmez, Z.; Şahin, H. Comparison of Antioxidant Properties of Wild Blueberries (Vaccinium Arctostaphylos L. and Vaccinium Myrtillus L.) with Cultivated Blueberry Varieties (Vaccinium Corymbosum L.) in Artvin Region of Turkey. TURJAF 2014, 3, 40–44. [Google Scholar] [CrossRef] [Green Version]
  34. Lima, E.C.; Baptista, J.B.; Albuquerque, L.M. Antioxidant Capacity versus Total Phenolic, Total Flavonoid and Anthocyanin Content of Endemic Azorean Vaccinium Cylindraceum: Comparison with Commercial Bilberry and Highbush Blueberry. Acta Hortic. 2009, 810, 901–910. [Google Scholar] [CrossRef]
  35. Committee on Herbal Medicinal Products. Assessment Report on Vaccinium myrtillus L., Fructus. Assessment Report of European Union; European Medicines Agency: London, UK, 2015; Available online: https://www.ema.europa.eu/en/documents/herbal-report/draft-assessment-report-vaccinium-myrtillus-l-fructus-recens_en.pdf (accessed on 23 September 2020).
  36. Morimoto, S.; Nonaka, G.; Nishioka, I. Tannins and Related Compounds. LX.: Isolation and Characterization of Proanthocyanidins with a Doubly-Linked Unit from Vaccinium vitis-idaea L. Chem. Pharm. Bull. 1988, 36, 33–38. Available online: https://www.jstage.jst.go.jp/article/cpb1958/36/1/36_1_33/_pdf (accessed on 9 September 2020). [CrossRef] [Green Version]
  37. Barizza, E.; Guzzo, F.; Fanton, P.; Lucchini, G.; Sacchi, G.; Lo Schiavo, F.; Nascimbene, J. Nutritional Profile and Productivity of Bilberry (Vaccinium myrtillus L.) in Different Habitats of a Protected Area of the Eastern Italian Alps. J. Food Sci. 2013, 78, C674–C678. [Google Scholar] [CrossRef]
  38. Du, Q.; Jerz, G.; Winterhalter, P. Isolation of two anthocyanin sambubiosides from bilberry (Vaccinium myrtillus) by high-speed counter-current chromatography. J. Chromatogr. A 2004, 1045, 59–63. [Google Scholar] [CrossRef]
  39. Seeram, N.; Adams, L.; Zhang, Y.; Lee, R.; Sand, D.; Scheuller, H.; Heber, D. Blackberry, Black Raspberry, Blueberry, Cranberry, Red Raspberry, and Strawberry Extracts Inhibit Growth and Stimulate Apoptosis of Human Cancer Cells In Vitro. J. Agric. Food Chem. 2006, 54, 9329–9339. [Google Scholar] [CrossRef] [PubMed]
  40. Laaksonen, O.; Sandell, M.; Kallio, H. Chemical factors contributing to orosensory profiles of bilberry (Vaccinium myrtillus) fractions. Eur. Food Res. Technol. 2010, 231, 271–285. [Google Scholar] [CrossRef]
  41. Koskimäki, J.; Hokkanen, J.; Jaakola, L.; Suorsa, M.; Tolonen, A.; Mattila, S.; Hohtola, A. Flavonoid biosynthesis and degradation play a role in early defence responses of bilberry (Vaccinium myrtillus) against biotic stress. Eur. J. Plant Pathol. 2009, 125, 629–637. [Google Scholar] [CrossRef]
  42. Karppinen, K.; Hirvelä, E.; Nevala, T.; Sipari, N.; Suokas, M.; Jaakola, L. Changes in the abscisic acid levels and related gene expression during fruit development and ripening in bilberry (Vaccinium myrtillus L.). Phytochemistry 2013, 95, 127–134. [Google Scholar] [CrossRef] [PubMed]
  43. Sadowska, B.; Paszkiewicz, M.; Podsędek, A.; Redzynia, M.; Różalska, B. Vaccinium myrtillus leaves and Frangula alnus bark derived extracts as potential antistaphylococcal agents. Acta Biochim. Pol. 2016, 61. [Google Scholar] [CrossRef] [Green Version]
  44. Korus, A.; Jaworska, G.; Bernaś, E.; Juszczak, L. Characteristics of physico-chemical properties of bilberry (Vaccinium myrtillus L.) jams with added herbs. JFST 2015, 52, 2815–2823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Chen, S.; Zhao, X.; Wan, J.; Ran, L.; Qin, Y.; Wang, X.; Zhang, Q. Dihydromyricetin improves glucose and lipid metabolism and exerts anti-inflammatory effects in nonalcoholic fatty liver disease: A randomized controlled trial. Pharmacol. Res. 2015, 99, 74–81. [Google Scholar] [CrossRef]
  46. Xiong, D.; Zhu, J.; Liu, C. Effect of ampelopsis extract from young stems and leaves on antimicrobial activities. J. Food Sci. 2000, 21, 48–50. [Google Scholar]
  47. Lin, X.; Zhang, R. Effect of ampelopsin on anti-inflammation and analgesic. Fujian J. Med. 1995, 17, 39–41. [Google Scholar]
  48. Ni, F.; Gong, Y.; Li, L.; Abdolmaleky, H.; Zhou, J. Flavonoid Ampelopsin Inhibits the Growth and Metastasis of Prostate Cancer In Vitro and in Mice. PLoS ONE 2012, 7, e38802. [Google Scholar] [CrossRef] [PubMed]
  49. Xuefen, Z.; Guifen, Z. Hypoglycemic Effect of Ampelopsin on Diabetic Rats Induced by Steptozotocin. Guangxi Sci. 2000, 3, 203–205. Available online: https://europepmc.org/article/cba/338628 (accessed on 1 July 2020).
  50. Hou, X.; Zhang, J.; Ahma, H.; Zhang, H.; Xu, Z.; Wang, T. Evaluation of Antioxidant Activities of Ampelopsin and Its Protective Effect in Lipopolysaccharide-Induced Oxidative Stress Piglets. PLoS ONE 2014, 9, e108314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Liu, J.; Zhou, T. A pharmacognostical study on “Tengcha”, bigdentate ampelopsis (Ampelopsis grossedentata). Zhong Cao Yao 1999, 30, 459–463. Available online: https://europepmc.org/article/cba/332003 (accessed on 1 July 2020).
  52. Zheng, Z.; Zeng, C.; Lin, Y. The protective effect of APS against acute chemical liver injury of the mouse. J. Guangxi Coll. TCM 2002, 5, 15–16. [Google Scholar]
  53. Ku, K.; Huang, Y.; Huang, Y.; Chiou, W. Miyabenol inhibits LPS-induced NO production via IKK/IkappaB inactivation in RAW 264.7 macrophages: Possible involvement of the p38 and PI3K pathways. J. Agric. Food Chem. 2008, 56, 8911–8918. [Google Scholar] [CrossRef]
  54. Li, Y.; Zhou, Y.; Zhou, C. Study of pharmacological function of ampelopsis composition. J. Hunan Norm. Univ. 2006, 3, 36–39. [Google Scholar]
  55. Fujimori, K.; Shibano, M. Avicularin, a Plant Flavonoid, Suppresses Lipid Accumulation through Repression of C/EBPα-Activated GLUT4-Mediated Glucose Uptake in 3T3-L1 Cells. J. Agric. Food Chem. 2013, 61, 5139–5147. [Google Scholar] [CrossRef]
  56. Feng, W. Antihistamine Composition. U.S. Patent 7022349, 4 April 2006. Available online: https://europepmc.org/article/pat/us7022349 (accessed on 1 July 2020).
  57. Guo, X.; Liu, J.; Ma, S.; Zhang, P.; Sun, W. Avicularin reversed multidrug-resistance in human gastric cancer through enhancing Bax and BOK expressions. Biomed. Pharmacother. 2018, 103, 67–74. [Google Scholar] [CrossRef]
  58. Dall’Agnola, R.; Ferraza, A.; Bernardia, A.; Albringa, D.; Nöra, C.; Sarmentob, L.; Schapoval, E. Antimicrobial activity of some Hypericum species. Phytomedicine 2003, 10, 511–516. [Google Scholar] [CrossRef] [PubMed]
  59. Butterweck, V.; Jürgenliemk, G.; Nahrstedt, A.; Winterhoff, H. Flavonoids from Hypericum perforatum show antidepressant activity in the forced swimming test. Planta Med. 2000, 66, 3–6. [Google Scholar] [CrossRef] [PubMed]
  60. Kim, S.; Kang, K.; Jho, E.; Jung, Y.; Nho, C.; Um, B.; Pan, C. Hepatoprotective effect of flavonoid glycosides from Lespedeza cuneata against oxidative stress induced by tert-butyl hyperoxide. Phytother. Res. 2011, 25, 1011–1017. [Google Scholar] [CrossRef]
  61. Kim, J.; Jung, Y.; Kim, M.; Yang, S.; Lee, S.; Kim, Y. Protective effect of components isolated from Lindera erythrocarpa against oxidative stress-induced apoptosis of H9c2 cardiomyocytes. Phytother. Res. 2011, 25, 1612–1617. [Google Scholar] [CrossRef]
  62. Van Anh Vo, J.W.; Chang, J.E.; Kim, J.Y.; Kim, N.H.; Lee, H.J.; Kim, S.S.; Chun, W.; Kwon, Y.S. Avicularin Inhibits Lipopolysaccharide-Induced Inflammatory Response by Suppressing ERK Phosphorylation in RAW 264.7 Macrophages. Biomol. Ther. 2012, 20, 532–537. [Google Scholar] [CrossRef] [Green Version]
  63. Crespy, V.; Williamson, G. A Review of the Health Effects of Green Tea Catechins in In Vivo Animal Models. J. Nutr. 2004, 134, 3431S–3440S. [Google Scholar] [CrossRef]
  64. Kashiwada, Y.; Nonaka, G.; Nishioka, I.; Chang, J.; Lee, K. Tannins and relatedcompounds as selective cytotoxic agents. Antitumor Agents 1992, 129, 1033–1043. Available online: https://0-pubs-acs-org.brum.beds.ac.uk/doi/pdf/10.1021/np50086a002 (accessed on 2 July 2020).
  65. Taher, M.; Abdul Majid, F.; Sarmidi, M. Cinnamtannin B1 activity on adipocytes formation. Med. J. Malays. 2004, 59 (Suppl. SB), 97–98. Available online: https://europepmc.org/article/med/15468836 (accessed on 2 July 2020).
  66. Balde, A.; van Hoof, L.; Pieters, L.; Vanden Berghe, D.; Vlietinck, A. Plant antiviral agents. VII. Antiviral and antibacterial proanthocyanidins from the bark of Pavetta owariensis. Phytother. Res. 1990, 4, 182–188. [Google Scholar] [CrossRef]
  67. Ho, K.; Huang, J.; Tsai, C.; Lin, T.; Hsu, Y.; Lin, C. Antioxidant Activity of Tannin Components from Vaccinium vitis-idaea L. J. Pharm. Pharmacol. 1999, 51, 1075–1078. [Google Scholar] [CrossRef]
  68. Way, T.; Tsai, S.; Wang, C.; Jhan, Y.; Ho, C.; Chou, C. Cinnamtannin D1 from Rhododendron formosanum induces autophagy via the inhibition of Akt/mTOR and activation of ERK1/2 in non-small-cell lung carcinoma cells. J. Agric. Food Chem. 2015, 63, 10407–10417. [Google Scholar] [CrossRef] [PubMed]
  69. Sun, P.; Wang, T.; Chen, L.; Yu, B.W.; Jia, Q.; Chen, K.X.; Fan, H.M.; Li, Y.M.; Wang, H.Y. Trimer procyanidin oligomers contribute to the protective effects of cinnamon extracts on pancreatic β-cells in vitro. Acta Pharmacol. Sin. 2006, 37, 1083–1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Huh, G.; Park, J.; Kang, J.H.; Jeong, T. Flavonoids from Lindera glauca Blume as low-density lipoprotein oxidation inhibitors. Nat. Prod. Res. 2014, 28, 831–834. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, L.; Yang, Y.; Yuan, P.; Yang, Y.; Chen, K.; Jia, Q.; Li, Y. Immunosuppressive Effects of A-Type Procyanidin Oligomers from Cinnamomum tamala. Evid. Based Complement. Altern. Med. 2014, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  72. Serraino, I.; Dugo, L.; Dugo, P.; Mondello, L.; Mazzon, E.; Dugo, G.; Caputi, A.P.; Cuzzocrea, S. Protective effects of cyanidin-3-O-glucoside from blackberry extract against peroxynitrite-induced endothelial dysfunction and vascular failure. Life Sci. 2003, 73, 1097–1114. [Google Scholar] [CrossRef]
  73. Guo, H.; Xia, M.; Zou, T.; Ling, W.; Zhong, R.; Zhang, W. Cyanidin 3-glucoside attenuates obesity-associated insulin resistance and hepatic steatosis in high-fat diet-fed and db/db mice via the transcription factor FoxO1. J. Nutr. Biochem. 2012, 23, 349–360. [Google Scholar] [CrossRef]
  74. Tsuda, T.; Watanabe, M.; Ohshima, K.; Norinobu, S.; Choi, S.; Kawakishi, S.; Osawa, T. Antioxidative activity of the anthocyanin pigments cyanidin 3-O-beta-D-glucoside and cyanidin. J. Agric. Food Chem. 1994, 42, 2407–2410. Available online: https://0-pubs-acs-org.brum.beds.ac.uk/doi/pdf/10.1021/jf00047a009 (accessed on 3 July 2020). [CrossRef]
  75. Sun, C.; Zheng, Y.; Chen, Q.; Tang, X.; Jiang, M.; Zhang, J.; Li, X.; Chen, K. Purification and anti-tumour activity of cyanidin-3-O-glucoside from Chinese bayberry fruit. Food Chem. 2012, 131, 1287–1294. [Google Scholar] [CrossRef]
  76. Scazzocchio, B.; Varì, R.; Filesi, C.; D’Archivio, M.; Santangelo, C.; Giovannini, C.; Iacovelli, A.; Silecchia, G.; Volti, G.L.; Galvano, F.; et al. Cyanidin-3-O-β-Glucoside and Protocatechuic Acid Exert Insulin-Like Effects by Upregulating PPARγ Activity in Human Omental Adipocytes. BMJ Open Diab. Res. CA 2011, 60, 2234–2244. [Google Scholar] [CrossRef] [Green Version]
  77. Jiang, X.; Guo, H.; Shen, T.; Tang, X.; Yang, Y.; Ling, W. Cyanidin-3-O-β-glucoside Purified from Black Rice Protects Mice against Hepatic Fibrosis Induced by Carbon Tetrachloride via Inhibiting Hepatic Stellate Cell Activation. J. Agric. Food Chem. 2015, 63, 6221–6230. [Google Scholar] [CrossRef]
  78. Speciale, A.; Canali, R.; Chirafisi, J.; Saija, A.; Virgili, F.; Cimino, F. Cyanidin-3-O-glucoside Protection against TNF-α-Induced Endothelial Dysfunction: Involvement of Nuclear Factor-κB Signaling. J. Agric. Food Chem. 2010, 58, 12048–12054. [Google Scholar] [CrossRef] [PubMed]
  79. Cimino, F.; Ambra, R.; Canali, R.; Saija, A.; Virgili, F. Effect of Cyanidin-3-O-glucoside on UVB-Induced Response in Human Keratinocytes. J. Agric. Food Chem. 2006, 54, 4041–4047. [Google Scholar] [CrossRef]
  80. Im, S.E.; Nam, T.G.; Lee, H.; Han, M.W.; Heo, H.J.; Koo, S.I.; Lee, C.Y.; Kim, D.O. Anthocyanins in the ripe fruits of Rubus coreanus Miquel and their protective effect on neuronal PC-12 cells. Food Chem. 2013, 139, 604–610. [Google Scholar] [CrossRef] [PubMed]
  81. Hernandez, L.M.R.; Fan, J.; Johnson, M.; Gonzalez de Mejia, E. Berry Phenolic Compounds Increase Expression of Hepatocyte Nuclear Factor-1α (HNF-1α) in Caco-2 and Normal Colon Cells Due to High Affinities with Transcription and Dimerization Domains of HNF-1α. PLoS ONE 2015, 10, e0138768. [Google Scholar] [CrossRef] [PubMed]
  82. Meiers, S.; Kemény, M.; Weyand, U.; Gastpar, R.; von Angerer, E.; Marko, D. The anthocyanidins cyanidin and delphinidin are potent inhibitors of the epidermal growth-factor receptor. J. Agric. Food Chem. 2001, 49, 958–962. [Google Scholar] [CrossRef]
  83. Inoue, H.; Maeda-Yamamoto, M.; Nesumi, A.; Murakami, A. Delphinidin-3-O-galactoside protects mouse hepatocytes from (-)-epigallocatechin-3-gallate-induced cytotoxicity via up-regulation of heme oxygenase-1 and heat shock protein 70. Nutr. Res. 2012, 32, 357–364. [Google Scholar] [CrossRef] [PubMed]
  84. Steinmetz, K.; Potter, J. Vegetables, fruit, and cancer. Epidemiol. Cancer Causes Control 1991, 2, 325–357. [Google Scholar] [CrossRef]
  85. Chakravarthy, B.; Gupta, S.; Gode, K. Functional beta cell regeneration in the islets of pancreas in alloxan induced diabetic rats by (−)-epicatechin. Life Sci. 1982, 31, 2693–2697. [Google Scholar] [CrossRef]
  86. Terao, J.; Piskula, M.; Yao, Q. Protective Effect of Epicatechin, Epicatechin Gallate, and Quercetin on Lipid Peroxidation in Phospholipid Bilayers. Arch. Biochem. Biophys. 1994, 308, 278–284. [Google Scholar] [CrossRef]
  87. Schroeter, H.; Heiss, C.; Balzer, J.; Kleinbongard, P.; Keen, C.; Hollenberg, N.; Kelm, M. (–)-Epicatechin mediates beneficial effects of flavanol-rich cocoa on vascular function in humans. PNAS 2006, 103, 1024–1029. [Google Scholar] [CrossRef] [Green Version]
  88. Hertog, M.; Feskens, E.; Hollman, P.; Katan, M.; Kromhout, D. Dietary antioxidant flavonoids and risk of coronary heart disease: The Zutphen Elderly Study. Lancet 1993, 342, 1007–1011. [Google Scholar] [CrossRef]
  89. Engler, M.B.; Engler, M.M.; Chen, C.Y.; Malloy, M.J.; Browne, A.; Chiu, E.Y.; Kwak, H.K.; Milbury, P.; Paul, S.M.; Blumberg, J.; et al. Flavonoid-Rich Dark Chocolate Improves Endothelial Function and Increases Plasma Epicatechin Concentrations in Healthy Adults. J. Am. Coll. Nutr. 2004, 23, 197–204. [Google Scholar] [CrossRef] [PubMed]
  90. Ahmad, N.; Feyes, D.; Agarwal, R.; Mukhtar, H.; Nieminen, A. Green Tea Constituent Epigallocatechin-3-Gallate and Induction of Apoptosis and Cell Cycle Arrest in Human Carcinoma Cells. JNCI 1997, 89, 1881–1886. [Google Scholar] [CrossRef] [Green Version]
  91. Reynolds, C.; McGillicuddy, F.; Harford, K.; Finucane, O.; Mills, K.; Roche, H. Dietary saturated fatty acids prime the NLRP3 inflammasome via TLR4 in dendritic cells-implications for diet-induced insulin resistance. Mol. Nutr. Food Res. 2012, 56, 1212–1222. [Google Scholar] [CrossRef] [PubMed]
  92. Sharangi, A. Medicinal and therapeutic potentialities of tea (Camellia sinensis L.)—A review. Food Res. Int. 2009, 42, 529–535. [Google Scholar] [CrossRef]
  93. Li, W.; Liu, M.; Xu, Y.; Feng, Y.; Che, J.; Wang, G.; Zheng, J. Combination of quercetin and hyperoside has anticancer effects on renal cancer cells through inhibition of oncogenic microRNA-27a. Oncol. Rep. 2014, 31, 117–124. [Google Scholar] [CrossRef] [Green Version]
  94. Verma, N.; Amresh, G.; Sahu, P.; Mishra, N.; Rao, C.; Singh, A. Pharmacological evaluation of hyperin for antihyperglycemic activity and effect on lipid profile in diabetic rats. Indian J. Exp. Biol. 2013, 51, 65–72. Available online: http://nopr.niscair.res.in/handle/123456789/15273 (accessed on 10 July 2020).
  95. Ku, S.; Kwak, S.; Kwon, O.; Bae, J. Hyperoside inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflammation 2014, 37, 1389–1400. [Google Scholar] [CrossRef]
  96. Zheng, M.; Liu, C.; Pan, F.; Shi, D.; Zhang, Y. Antidepressant- like effect of hyperoside isolated from Apocynum venetum leaves: Possible cellular mechanisms. Phytomedicine 2012, 19, 145–149. [Google Scholar] [CrossRef]
  97. Xing, H.; Liu, Y.; Chen, J.; Sun, F.; Shi, H.; Xia, P. Hyperoside attenuates hydrogen peroxide-induced L02 cell damage via MAPK-dependent Keap (1)-Nrf(2)-ARE signaling pathway. Biochem. Biophys. Res. Commun. 2011, 410, 759–765. [Google Scholar] [CrossRef]
  98. Geng, M.; Wang, J.; Chen, H.; Yang, X.; Huang, Z. Effects of hyperin on the cccDNA of duck hepatitis B virus and its immunological regulation. Acta Pharm. Sin. 2009, 44, 1440–1444. Available online: https://europepmc.org/article/med/21351483 (accessed on 10 July 2020).
  99. Li, Q.; Chou, G.; Chen, Z.; MA, C. Inhibitory mechanism of hyperin on the apoptosis in myocardial ischemia reperfusion in rats. Acta Pharm. Sin. 2002, 37, 849–852. Available online: https://europepmc.org/article/cba/379068 (accessed on 10 July 2020).
  100. Song, M.; Hong, M.; Lee, M.Y.; Jee, J.G.; Lee, Y.M.; Bae, J.S.; Jeong, T.C.; Lee, S. Selective inhibition of the cytochrome P450 isoform by hyperoside and its potent inhibition of CYP2D6. Food Chem. Toxicol. 2013, 59, 549–553. [Google Scholar] [CrossRef] [PubMed]
  101. Zhao, W.; Chen, Z.; Song, B.; Wang, Y.; Wang, Q.; Tang, M.; Jiang, Q. The protective effect of hyperin on gastric mucosal injury in mice and its mechanism. Acta Univ. Med. Nahui 1999, 34, 178–180. Available online: http://en.cnki.com.cn/Article_en/CJFDTotal-YIKE903.007.htm (accessed on 10 July 2020).
  102. Choi, J.; Kim, D.; Yun, N.; Choi, J.; Islam, M.; Kim, Y.; Lee, S. Protective effects of hyperoside against carbon tetrachloride-induced liver damage in mice. J. Nat. Prod. 2011, 74, 1055–1060. [Google Scholar] [CrossRef]
  103. Huang, K.; Yang, X.; Huang, Z.; Geng, M.; Chen, H.; Wang, J.; Yang, J. The effect of hyperin on immune function in normal mice. PJCPLA 2009, 25, 133–135. Available online: http://en.cnki.com.cn/Article_en/CJFDTotal-JFJN200902011.htm (accessed on 10 July 2020).
  104. Lee, S.; Son, K.; Chang, H.; Do, L.; Jung, Y.; Kang, S.; Kim, H. Antiinflammatory activity of naturally occurring flavone and fiavonol glycosides. APHRDQ 1993, 16, 25–28. [Google Scholar] [CrossRef]
  105. Zhang, L.; Cheng, X.; Hu, J.; Sun, L.; Du, G. Neuroprotective effects of hyperoside on sodium azide- induced apoptosis in pc12 cells. Chin. J. Nat. Med. 2011, 77, 450–455. [Google Scholar] [CrossRef]
  106. Zeng, K.; Wang, X.; Ko, H.; Kwon, H.; Cha, J.; Yang, H. Hyperoside protects primary rat cortical neurons from neurotoxicity induced by amyloid beta-protein via the PI3K/Akt/Bad/Bcl(XL)-regulated mitochondrial apoptotic pathway. Eur. J. Pharmacol. 2011, 672, 45–55. [Google Scholar] [CrossRef]
  107. Tsuda, T.; Horio, F.; Uchida, K.; Aoki, H.; Osawa, T. Dietary cyanidin 3-O-β-D-glucoside-rich purple corn color prevents obesity and ameliorates hyperglycemia in mice. J. Nutr. 2003, 133, 149–160. [Google Scholar] [CrossRef]
  108. Hou, D.; Douglas, L. Potential mechanisms of cancer chemopre- vention by anthocyanins. Curr. Mol. Med. 2003, 133, 2125–2130. [Google Scholar] [CrossRef]
  109. Rice-Evans, C.; Miller, N.; Paganga, G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic. Biol. Med. 1996, 20, 933–956. [Google Scholar] [CrossRef]
  110. Ramirez-Tortosa, C.; Andersen, Ø.M.; Gardner, P.T.; Morrice, P.C.; Wood, S.G.; Duthie, S.J.; Collins, A.R.; Duthie, G.G. Anthocyanin-rich extract decreases indices of lipid peroxidation and DNA damage in vitamin E-depleted rats. Free Radic. Biol. Med. 2001, 31, 1033–1037. [Google Scholar] [CrossRef]
  111. Joseph, J.; Denisova, N.; Arendash, G.; Gordon, M.; Diamond, D.; Shukitt-Hale, B.; Morgan, D. Blueberry supplementation enhances signaling and prevents behavioral deficits in an Alzheimer disease model. Nutr. Neurosci. 2003, 6, 153–162. [Google Scholar] [CrossRef] [PubMed]
  112. Fernandez, J.; Reyes, R.; Ponce, H.; Oropeza, M.; Vancalsteren, M.; Jankowski, C.; Campos, M. Isoquercitrin from Argemone platyceras inhibits carbachol and leukotriene D4-induced contraction in guinea-pig airways. Eur. J. Pharmacol. 2005, 522, 108–115. [Google Scholar] [CrossRef]
  113. Sumi, M.; Tateishi, N.; Shibata, H.; Ohki, T.; Sata, M. Quercetin glucosides promote ischemia-induced angiogenesis, but do not promote tumor growth. Life Sci. 2013, 93, 814–819. [Google Scholar] [CrossRef]
  114. Panda, S.; Kar, S. Antidiabetic and antioxidative effects of Annona squamosa leaves are possibly mediated through quercetin-3-O-glucoside. Biofactors 2007, 31, 201–210. [Google Scholar] [CrossRef]
  115. Li, R.; Yuan, C.; Dong, C.; Shuang, S.; Choi, M. In vivo antioxidative effect of isoquercitrin on cadmium-induced oxidative damage to mouse liver and kidney. Naunyn Schmiedeberg. Arch. Pharmacol. 2011, 383, 437–445. [Google Scholar] [CrossRef]
  116. Junior, A.G.; Prando, T.B.; Leme, T.D.; Gasparotto, F.M.; Lourenço, E.L.; Rattmann, Y.D.; Da Silva-Santos, J.E.; Kassuya, C.A.; Marques, M.C. Mechanisms underlying the diuretic effects of Tropaeolum majus L. extracts and its main component isoquercitrin. J. Ethnopharmacol. 2012, 141, 501–509. [Google Scholar] [CrossRef]
  117. Morikawa, K.; Nonaka, M.; Narahara, M.; Torii, I.; Kawaguchi, K.; Yoshikawa, T.; Kumazawa, Y.; Morikawa, S. Inhibitory effect of quercetin on carrageenan-induced inflammation in rats. Life Sci. 2003, 74, 709–721. [Google Scholar] [CrossRef]
  118. Loscalzo, L.; Wasowski, C.; Marder, M. Neuroactive flavonoid glycosides from Tilia petiolaris DC. extracts. Phytother. Res. 2009, 23, 1453–1457. [Google Scholar] [CrossRef] [PubMed]
  119. Park, J.; Kim, S.; Lee, H.; Kim, S.; Kwon, Y.; Chun, W. Isorhamnetin-3-O-Glucuronide Suppresses JNK and p38 Activation and Increases Heme-Oxygenase-1 in Lipopolysaccharide-Challenged RAW264.7 Cells. Drug Dev. Res. 2016, 77, 143–151. [Google Scholar] [CrossRef]
  120. Jimenez, R.; Lopez-Sepulveda, R.; Romero, M.; Toral, M.; Cogolludo, A.; Perez-Vizcaino, F.; Duarte, J. Quercetin and its metabolites inhibit the membrane NADPH oxidase activity in vascular smooth muscle cells from normotensive and spontaneously hypertensive rats. Food Funct. 2015, 6, 409–414. [Google Scholar] [CrossRef] [PubMed]
  121. Zern, T.; West, K.; Fernandez, M. Grape polyphenols decrease plasma triglycerides and cholesterol accumulation in the aorta of ovariectomized guinea pigs. J. Nutr. 2003, 133, 2268–2272. [Google Scholar] [CrossRef] [PubMed]
  122. Kameda, K.; Takaku, T.; Okuda, H.; Kimura, Y.; Okuda, T.; Hatano, T.; Agata, I.; Arichi, S. Inhibitory effects of various flavonoids isolated from leaves of persimmon on angiotensin- converting enzyme activity. J. Nat. Prod. 1987, 50, 680–683. [Google Scholar] [CrossRef] [PubMed]
  123. Huang, W.W.; Chiu, Y.J.; Fan, M.J.; Lu, H.F.; Yeh, H.F.; Li, K.H.; Chen, P.Y.; Chung, J.G.; Yang, J.S. Kaempferol induced apoptosis via endoplasmic reticulum stress and mitochondria-dependent pathway in human osteosarcoma U-2 OS cells. Mol. Nutr. Food Res. 2010, 54, 1585–1595. [Google Scholar] [CrossRef]
  124. Jorge, A.; Horst, H.; de Sousa, E.; Pizzolatti, M.; Silva, F. Insulinomimetic effects of kaempferitrin on glycaemia and on 14C- glucose uptake in rat soleus muscle. Chem. Biol. Interact. 2004, 149, 89–96. [Google Scholar] [CrossRef]
  125. Kataoka, M.; Hirata, K.; Kunikata, T.; Ushio, S.; Iwaki, K.; Ohashi, K.; Ikeda, M.; Kurimoto, M. Antibacterial action of tryptanthrin and kaempferol, isolated from the indigo plant (Polygonum tinctorium Lour.), against Helicobacter pylori-infected Mongolian gerbils. J. Gastroenterol. 2001, 36, 5–9. Available online: https://0-link-springer-com.brum.beds.ac.uk/content/pdf/10.1007/s005350170147.pdf (accessed on 12 July 2020). [CrossRef]
  126. Kampkotter, A.; Gombitang, N.; Zurawski, R.; Timpel, C.; Chovolou, Y.; Watjen, W.; Kahl, R. Effects of the flavonoids kaempferol and fisetin on thermotolerance, oxidative stress and FoxO transcription factor DAF-16 in the model organism Caenorhabditis elegans. Arch. Toxicol. 2007, 81, 849–858. [Google Scholar] [CrossRef]
  127. Tu, Y.; Lian, T.; Yen, J.; Chen, Z.; Wu, M. Antiatherogenic effects of kaempferol and rhamnocitrin. J. Agric. Food Chem. 2007, 55, 9969–9976. [Google Scholar] [CrossRef]
  128. Chung, M.; Gan, K.; Lin, C.; Ko, F.; Teng, C. Antiplatelet effects and vasorelaxing action of some constituents of Formosan plants. J. Nat. Prod. 1993, 56, 929–934. [Google Scholar] [CrossRef] [PubMed]
  129. Wang, L.; Tu, Y.; Lian, T.; Hung, J.; Yen, J.; Wu, M. Distinctive antioxidant and antiinflammatory effects of flavonols. J. Agric. Food Chem. 2006, 54, 9798–9804. [Google Scholar] [CrossRef] [PubMed]
  130. Hsu, Y.; Laricitrin, A. Polyphenolic Compound Of Red Grape, Inhibits Lung Cancer-Mediated Dendritic Cell Suppression By Decreasing Signal Transducer And Activator Of Transcription 3 Pathway. Am. J. Resp. Crit. Care Med. 2016, 193, S2564. Available online: https://www.atsjournals.org/doi/abs/10.1164/ajrccm-conference.2016.193.1_MeetingAbstracts.A2564 (accessed on 12 July 2020).
  131. Mukhtar, H.; Das, M.; Khan, W.; Wang, Z.; Bik, D.; Bickers, D. Exceptional activity of tannic acid among naturally occurring plant phenols in protecting against 7,12-dimethylbenz(a)anthracene-, benzo(a)-pyrene, 3-methylcholanthrene-, and N-methyl-N-nitrosourea-induced skin tumorigenesis in mice. Cancer Res. 1988, 48, 2361–2365. Available online: https://cancerres.aacrjournals.org/content/48/9/2361.short (accessed on 12 July 2020). [PubMed]
  132. Chaudhry, P.; Cabrera, J.; Juliani, H.; Varma, S. Inhibition of human lens aldose reductase by flavonoids, sulindac and indomethacin. Biochem. Pharmcol. 1983, 32, 1995–1998. [Google Scholar] [CrossRef]
  133. Ong, K.K. Insulinomimetic effects of myricetin on lipogenesis and glucose transport in rat adipocytes but not glucose transporter translocation. Biochem. Pharmacol. 1996, 51, 423–429. [Google Scholar] [CrossRef]
  134. Robak, J.G. Flavonoids are scavengers of superoxide anions. Biochem. Pharmacol. 1988, 37, 837–841. [Google Scholar] [CrossRef]
  135. Ono, K.; Nakane, H. Mechanisms of inhibition of various cellular DNA and RNA polymerases by several flavonoids. J. Biochem. 1990, 108, 609–613. Available online: https://www.jstage.jst.go.jp/article/biochemistry1922/108/4/108_4_609/_article/-char/ja/ (accessed on 12 July 2020). [CrossRef]
  136. Brown, J.; Dietrich, P. Mutagenicity of plant flavonols in the Salmonella/mammalian microsome test: Activation of flavonol glycosides by mixed glycosidases from rat cecal bacteria and other sources. MRGTEM 1979, 66, 223–240. [Google Scholar] [CrossRef]
  137. Tundis, R.; Loizzo, M.; Menichini, F.; Bonesi, M.; Colica, C.; Menichini, F. In vitro cytotoxic activity of extracts and isolated constituents of Salvia leriifolia Benth. Against a panel of human cancer cell lines. Chem. Biodiv. 2011, 8, 1152–1162. [Google Scholar] [CrossRef]
  138. Sabarinathan, D.; Mahalakshmi, P.; Vanisree, A. Naringenin promote apoptosis in cerebrally implanted C6 glioma cells. Mol. Cell. Biochem. 2010, 345, 215–222. [Google Scholar] [CrossRef]
  139. Oršolić, N.; Gajski, G.; Garaj-Vrhovac, V.; Dikić, D.; Prskalo, Z.; Sirovina, D. DNA-protective effects of quercetin or naringenin in alloxan-induced diabetic mice. Eur. J. Pharmacol. 2011, 656, 110–118. [Google Scholar] [CrossRef]
  140. Yi, L.; Li, C.; Zhan, X.; Cui, C.; Xiao, F.; Zhou, L. Involvement of monoaminergic system in the antidepressant-like effect of the flavonoid naringenin in mice. Progr. Neuro Psychopharmacol. Biol. Psych. 2010, 34, 1223–1228. [Google Scholar] [CrossRef]
  141. Lakshmi, V.; Joseph, S.; Srivastava, S.; Verma, S.; Sahoo, M.; Dube, V. Antifilarial activity in vitro and in vivo of some flavonoids tested against Brugia malayi. Acta Tropica 2010, 116, 127–133. [Google Scholar] [CrossRef] [PubMed]
  142. Prabu, S.; Shagirtha, K.; Renugadevi, J. Naringenin in combination with vitamins C and E potentially protects oxidative stress-mediated hepatic injury in cadmium-intoxicated rats. J. Nutr. Sci. Vitaminol. 2011, 57, 177–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Sánchez-Salgado, J.; Castillo-España, P.; Ibarra-Barajas, M.; Villalobos-Molina, R.; Estrada-Soto, S. Cochlospermum vitifolium induces vasorelaxant and antihypertensive effects mainly by activation of NO/cGMP signaling pathway. J. Ethnopharmachol. 2010, 130, 477–484. [Google Scholar] [CrossRef] [PubMed]
  144. Chao, C.; Weng, C.; Chang, N.; Lin, J.; Kao, S.; Ho, F. Naringenin more effectively inhibits inducible nitric oxide synthase and cyclooxygenase-2 expression in macrophages than in microglia. Nutr. Res. 2010, 30, 858–864. [Google Scholar] [CrossRef]
  145. Yamamoto, T.; Yoshimura, M.; Yamaguchi, F.; Kouchi, T.; Tsuji, R.; Saito, M.; Obata, A.; Kikuchi, M. Anti-allergic Activity of Naringenin Chalcone from a Tomato Skin Extract. Biosci. Biotechnol. Biochem. 2014, 68, 1706–1711. [Google Scholar] [CrossRef] [Green Version]
  146. Yagura, T.; Motomiya, T.; Ito, M.; Honda, G.; Iida, A.; Kiuchi, F.; Tokuda, H.; Nishino, H. Anticarcinogenic compound in the Uzbek medicinal plant, Helichrysum maracandicum. J. Nat. Med. 2008, 62, 174–178. [Google Scholar] [CrossRef]
  147. Anto, R.; Sukumaran, K.; Kuttan, G.; Rao, M.; Subbaraju, V.; Kuttan, R. Anticancer and antioxidant activity of synthetic chalcones and related compounds. Cancer Lett. 1995, 97, 33–37. [Google Scholar] [CrossRef]
  148. Hirai, S.; Kim, Y.I.; Goto, T.; Kang, M.S.; Yoshimura, M.; Obata, A.; Yu, R.; Kawada, T. Inhibitory effect of naringenin chalcone on inflammatory changes in the interaction between adipocytes and macrophages. Life Sci. 2007, 81, 1272–1279. [Google Scholar] [CrossRef]
  149. Pereira, M.; Grubbs, C.; Barnes, L.; Li, H.; Olson, G.; Eto, I. Effects of the phytochemicals, curcumin and quercetin, upon azoxymethane-induced colon cancer and 7,12-dimethylbenz[a]anthracene-induced mammary cancer in rats. Carcinogenesis 1996, 17, 1305–1311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Hayek, T.; Fuhrman, B.; Vaya, J.; Rosenblat, M.; Belinky, P.; Coleman, R. Reduced progression of atherosclerosis in apolipoprotein E-deficient mice following consumption of red wine, or its polyphenols quercetin or catechin, is associated with reduced susceptibility of LDL to oxidation and aggregation. ATVB 1997, 17, 2744–2752. [Google Scholar] [CrossRef] [PubMed]
  151. Pignatelli, P.; Pulcinelli, F.; Celestini, A.; Lenti, L.; Ghiselli, A.; Gazzaniga, P. The flavonoids quercetin and catechin synergistically inhibit platelet function by antagonizing the intracellular production of hydrogen peroxide. Am. J. Clin. Nutr. 2000, 72, 1150–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Perez-Vizcaino, F.; Ibarra, M.; Cogolludo, A.; Duarte, J.; Zaragoza-Arnaez, F.; Moreno, L. Endothelium-independent vasodilator effects of the flavonoid quercetin and its methylated metabolites in rat conductance resistance arteries. J. Pharmacol. Exp. Theor. 2002, 302, 66–72. [Google Scholar] [CrossRef] [Green Version]
  153. Ferry, D.; Smith, A.; Malkhandi, J.; Fyfe, D.; de Takats, P.; Anderson, D. Phase I clinical trial of the flavonoid quercetin: Pharmacokinetics and evidence for in vivo tyrosine kinase inhibition. Clin. Cancer Res. 1996, 2, 659–668. Available online: https://clincancerres.aacrjournals.org/content/2/4/659.short (accessed on 15 July 2020).
  154. Saint-Cricq de Gaulejac, N.; Provost, C.; Vivas, N. Comparative study of polyphenol scavenging activities assessed by different methods. J. Food Agric. Chem. 1999, 47, 425–431. [Google Scholar] [CrossRef]
  155. Jung, H.; Ali, M.; Bhakta, H.; Min, B.; Choi, J. Prunin is a highly potent flavonoid from Prunus davidiana stems that inhibits protein tyrosine phosphatase 1B and stimulates glucose uptake in insulin-resistant HepG2 cells. Acta Pharm. Res. 2017, 40, 37–48. [Google Scholar] [CrossRef]
  156. Klaman, L.D.; Boss, O.; Peroni, O.D.; Kim, J.K.; Martino, J.L.; Zabolotny, J.M.; Moghal, N.; Lubkin, M.; Kim, Y.B.; Sharpe, A.H.; et al. Increased energy expenditure, decreased adiposity, and tissue specific insulin sensitivity in protein-tyrosine phosphatase 1B-deficient mice. Mol. Cell Biol. 2000, 20, 5479–5489. [Google Scholar] [CrossRef] [Green Version]
  157. Han, X.; Gao, S.; Cheng, Y.; Sun, Y.; Liu, W.; Tang, L.; Ren, D. Protective effect of naringenin-7-O-glucoside against oxidative stress induced by doxorubicin in H9c2 cardiomyocytes. Biosci. Trends 2012, 6, 19–25. [Google Scholar] [CrossRef] [Green Version]
  158. Han, X.; Ren, D.; Fan, P.; Shen, T.; Lou, H. Protective effects of naringenin-7-O-glucoside on doxorubicin-induced apoptosis in H9C2 cells. Eur. J. Pharmacol. 2007, 581, 47–53. [Google Scholar] [CrossRef] [Green Version]
  159. Donnini, S.; Finetti, F.; Lusini, L.; Morbidelli, L.; Cheynier, V.; Barron, D. Divergent effects of quercetin conjugates on angiogenesis. Br. J. Nutr. 2006, 95, 1016–1023. [Google Scholar] [CrossRef] [PubMed]
  160. Guo, X.; Zhang, D.; Gao, X.; Parry, J.; Liu, K.; Liu, B.; Wang, M. Quercetin and quercetin-3-O-glucuronide are equally effective in ameliorating endothelial insulin resistance through inhibition of reactive oxygen species-associated inflammation. Mol. Nutr. Food Res. 2013, 57, 1037–1045. [Google Scholar] [CrossRef]
  161. Moon, J.T.; Nakahara, K.; Terao, J. Identification of quercetin 3-O-beta-d-glucuronide as an antioxi- dative metabolite in rat plasma after oral administration of quercetin. Free Radic. Biol. Med. 2001, 30, 1274–1285. [Google Scholar] [CrossRef]
  162. Ho, L.; Ferruzzi, M.G.; Janle, E.M.; Wang, J.; Gong, B.; Chen, T.Y.; Lobo, J.; Cooper, B.; Wu, Q.L.; Talcott, S.T.; et al. Identification of brain-targeted bioactive dietary quercetin-3-O-glucuronide as a novel intervention for Alzheimer’s disease. FASEB J. 2013, 27, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Bylka, W.; Matlawska, I.; Pilewski, N. Natural flavonoids as antimicrobial agents. JANA 2004, 7, 24–31. Available online: https://www.researchgate.net/profile/Irena_Matlawska/publication/228800384_Natural_flavonoids_as_antimicrobial_agents/links/5452b81a0cf2bccc49094b17.pdf (accessed on 16 July 2020).
  164. Huber, G.; Rupasinghe, H.; Shahidi, F. Inhibition of oxidation of omega-3 polyunsaturated fatty acids and fish oil by quercetin glycosides. Food Chem. 2009, 117, 290–295. [Google Scholar] [CrossRef]
  165. Liu, H.; Yu, D.; Shin, S.; Park, H.; Park, J.; Bark, K. Spectroscopic Properties of Quercetin Derivatives, Quercetin-3-O-rhamnoside and Quercetin-3-O-rutinoside, in Hydro-organic Mixed Solvents. Photochem. Photobiol. 2009, 85, 934–942. [Google Scholar] [CrossRef]
  166. Wang, T.J.; Ngo, D.; Psychogios, N.; Dejam, A.; Larson, M.G.; Vasan, R.S.; Ghorbani, A.; O’Sullivan, J.; Cheng, S.; Rhee, E.P.; et al. 2-Aminoadipic acid is a biomarker for diabetes risk. J. Clin. Investig. 2013, 123, 10. [Google Scholar] [CrossRef]
  167. Sell, D.; Strauch, M.; Shen, W.; Monnier, V. 2-Aminoadipic acid is a marker of protein carbonyl oxidation in the aging human skin: Effects of diabetes, renal failure and sepsis. Biochem. J. 2007, 404, 269–277. [Google Scholar] [CrossRef]
  168. Da Silva, J.C.; Amaral, A.U.; Cecatto, C.; Wajner, A.; dos Santos Godoy, K.; Ribeiro, R.T.; de Mello Gonçalves, A.; Zanatta, Â.; da Rosa, M.S.; Loureiro, S.O.; et al. α-Ketoadipic Acid and α-Aminoadipic Acid Cause Disturbance of Glutamatergic Neurotransmission and Induction of Oxidative Stress In Vitro in Brain of Adolescent Rats. Neurotox. Res. 2017, 32, 276–290. [Google Scholar] [CrossRef] [PubMed]
  169. Pieper, G.; Peltier, B. Amelioration by L-arginine of a dysfunctional arginine/nitric oxide pathway in diabetic endothelium. J. Cardiovasc. Pharmacol. 1995, 25, 397–403. [Google Scholar] [CrossRef]
  170. Giroux, I.; Kurowska, E.; Caroll, K. Role of dietary lysine, methionine and arginine in the regulation of hypercholesterolemia in rabbits. J. Nutr. Biochem. 1999, 10, 166–171. [Google Scholar] [CrossRef]
  171. Maxwell, A.; Cooke, J. Cardiovascular effects of L-arginine. Curr. Opin. Nephrol. Hypertens. 1998, 7, 63–70. Available online: https://journals.lww.com/co-nephrolhypertens/Abstract/1998/01000/Cardiovascular_effect_of_L_arginine.11.aspx (accessed on 18 July 2020). [CrossRef] [PubMed]
  172. Lewis, B.; Langkamp-Henken, B. Arginine enhances in vivo immune responses in young, adult and aged mice. J. Nutr. 2000, 130, 1827–1830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Luo, M.; Brooks, M.; Wicha, M. Asparagine and Glutamine: Co-conspirators Fueling Metastasis. Cell Met. 2018, 27, 947–949. [Google Scholar] [CrossRef] [Green Version]
  174. Ruzzo, E.; Capo-Chichi, J.; Ben-Zeev, B.; Chitayat, D.; Mao, H.; Pappas, A.; Goldstein, D. Deficiency of Asparagine Synthetase Causes Congenital Microcephaly and a Progressive Form of Encephalopathy. Neuron 2013, 80, 429–441. [Google Scholar] [CrossRef] [Green Version]
  175. Berl, S.; Takagaki, G.; Clarke, D.; Waelsch, H. Metabolic compartmentsin vitro. Ammonia and glutamic acid metabolism in brain and liver. J. Biol. Chem. 1962, 237, 2562–2569. [Google Scholar]
  176. Marks, N.; Datta, R.; Lajtha, A. Distribution of amino acids and of exo- and endo-peptidases along vertebrate and invertebrate nerves. J. Neurochem. 1970, 17, 53–63. [Google Scholar] [CrossRef]
  177. Sheiner, J.; Morris, P.; Anderson, G. Food intake suppression by histidine. Pharmacol. Biochem. Behav. 1985, 23, 721–726. [Google Scholar] [CrossRef]
  178. Gerber, D. Low free serum histidine concentration in rheumatoid arthritis. A measure of disease activity. J. Clin. Investig. 1975, 55, 1164–1173. Available online: https://www.jci.org/articles/view/108033 (accessed on 18 July 2020). [CrossRef] [PubMed] [Green Version]
  179. Frestedt, J.; Zenk, J.; Kuskowski, M. A whey-protein supplement increases fat loss and spares lean muscle in obese subjects: A randomized human clinical study. Nutr. Met. 2008, 5, 8–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Rieu, I.; Sornet, C.; Bayle, G. Leucine-supplemented meal feeding for ten days beneficially affects postprandial muscle protein synthesis in old rats. J. Nutr. 2003, 133, 1198–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Rieu, I.; Balage, M.; Sornet, C. Leucine supplementation improves muscle protein synthesis in elderly men independently of hyperaminoacidaemia. J. Physiol. 2006, 575, 305–315. [Google Scholar] [CrossRef]
  182. Layman, D. The role of leucine in weight loss diets and glucose homeostasis. J. Nutr. 2003, 133, 261–267. [Google Scholar] [CrossRef]
  183. Papes, F.; Surpili, M.; Langone, F.; Trigo, J.; Arruda, P. The essential amino acid lysine acts as precursor of glutamate in the mammalian central nervous system. FEBS Lett. 2001, 488, 34–38. [Google Scholar] [CrossRef] [Green Version]
  184. Van Spronsen, F.; Hoeksma, M.; Reijngoud, D. Brain dysfunction in phenylketonuria: Is phenylalanine toxicity the only possible cause? J. Inherit. Met. Dis. 2009, 32, 46–51. [Google Scholar] [CrossRef]
  185. Griffiths, P.; Paterson, L.; Harvie, A. Neuropsychological effects of subsequent exposure to phenylalanine in adolescents and young adults with eariy-treated phenylketonuria. J. Intellect. Disabil. Res. 1995, 39, 365–372. [Google Scholar] [CrossRef]
  186. Rasheed, A.; Kumar, C. Tyrosine and glycine derivatives as potential prodrugs: Design, synthesis, and pharmacological evaluation of amide derivatives of mefenamic acid. J. Enzyme Inhib. Med. Chem. 2010, 25, 804–811. [Google Scholar] [CrossRef] [Green Version]
  187. Wurtman, R.; Hefti, F.; Melamed, E. Precursor control of neurotransmitter synthesis. Pharmacol. Rev. 1980, 32, 315–335. Available online: https://pharmrev.aspetjournals.org/content/32/4/315/tab-article-info (accessed on 19 July 2020).
  188. Deijen, J.; Wientjes, C.; Vullinghs, H.; Cloin, P.; Langefeld, J. Tyrosine improves cognitive performance and reduces blood pressure in cadets after one week of a combat training course. Brain Res. Bull. 1999, 48, 203–209. [Google Scholar] [CrossRef]
  189. Korol, S.V.; Jin, Z.; Jin, Y.; Bhandage, A.K.; Tengholm, A.; Gandasi, N.R.; Barg, S.; Espes, D.; Carlsson, P.O.; Laver, D.; et al. Functional Characterization of Native, High-Affinity GABA Receptors in Human Pancreatic β Cells. EBioMedicine 2018, 30, 273–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Bhandage, A.K.; Jin, Z.; Korol, S.V.; Shen, Q.; Pei, Y.; Deng, Q.; Espes, D.; Carlsson, P.O.; Kamali-Moghaddam, M.; Birnir, B. GABA Regulates Release of Inflammatory Cytokines From Peripheral Blood Mononuclear Cells and CD4+ T Cells and Is Immunosuppressive in Type 1 Diabetes. EBioMedicine 2018, 30, 283–294. [Google Scholar] [CrossRef] [Green Version]
  191. Scheel-Krüger, J. Dopamine-GABA interactions: Evidence that GABA transmits, modulates and mediates dopaminergic functions in the basal ganglia and the limbic system. Acta Neurol. Scand. 1986, 73 (Suppl. S107), 54–67. Available online: https://psycnet.apa.org/record/1987-27370-001 (accessed on 19 July 2020).
  192. Huang, K.; Liang, X.; Zhong, Y.; He, W.; Wang, Z. 5-Caffeoylquinic acid decreases diet-induced obesity in rats by modulating PPARα and LXRα transcription. J. Sci. Food Agric. 2015, 95, 1903–1910. [Google Scholar] [CrossRef]
  193. Cho, A.; Jeon, S.; Kim, M.; Yeo, J.; Seo, K.; Choi, M.; Lee, M. Chlorogenic acid exhibits anti-obesity property and improves lipid metabolism in high-fat diet-induced-obese mice. Food Chem. Toxicol. 2010, 48, 937–943. [Google Scholar] [CrossRef]
  194. Yagasaki, K.; Miura, Y.; Okauchi, R.; Furuse, T. Inhibitory effects of chlorogenic acid and its related compounds on the invasion of hepatoma cells in culture. Cytotechnology 2000, 33, 229–235. [Google Scholar] [CrossRef] [PubMed]
  195. Crozier, T.; Stalmach, A.; Lean, M.; Crozier, A. Espresso coffees, caffeine and chlorogenic acid intake: Potential health implications. Food Funct. 2012, 2, 30–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Ong, K.; Hsu, A.; Tan, B. Antidiabetic and anti-lipidemic effects of chlorogenic acid are mediated by ampk activation. Biochem. Pharmacol. 2013, 85, 1341–1351. [Google Scholar] [CrossRef] [PubMed]
  197. Mubarak, A.; Bondonno, C.P.; Liu, A.H.; Considine, M.J.; Rich, L.; Mas, E.; Croft, K.D.; Hodgson, J.M. Acute effects of chlorogenic acid on nitric oxide status, endothelial function, and blood pressure in healthy volunteers: A randomized trial. J. Agric. Food Chem. 2012, 60, 9130–9136. [Google Scholar] [CrossRef]
  198. Sticha, H.; Rosina, M.; Wub, C.; Powrieb, W. A comparative genotoxicity study of chlorogenic acid (3-O-caffeoylquinic acid). MRGTEM 1981, 90, 201–212. [Google Scholar] [CrossRef]
  199. Raina, K.; Rajamanickam, S.; Deep, G.; Singh, M.; Agarwal, R.; Agarwal, C. Chemopreventive effects of oral gallic acid feeding on tumor growth and progression in TRAMP mice. Mol. Cancer Ther. 2008, 7, 1258–1267. [Google Scholar] [CrossRef] [Green Version]
  200. Gichner, T.; Pospísil, F.; Velemínský, J.; Volkeová, V.; Volke, J. Two Types of Antimutagenic Effects of Gallic and Tannic Acids towards N-nitroso-compounds-Induced Mutagenicity in the Ames Salmonella Assay. Folia Microbiol. 1987, 32, 55–62. Available online: https://0-link-springer-com.brum.beds.ac.uk/content/pdf/10.1007/BF02877259.pdf (accessed on 19 July 2020). [CrossRef]
  201. Kim, D.; Lee, K.; Lee, H.; Lee, C. Vitamin C equivalent antioxidant capacity of phenolic phytochemicals. J. Agric. Food Chem. 2002, 50, 3713–3717. [Google Scholar] [CrossRef] [PubMed]
  202. Kroes, B.; van den Berg, A.; Quarles van Ufford, H.; van Dijk, H.; Labadie, R. Anti-inflammatory Activity of Gallic Acid. Planta Med. 1992, 58, 499–504. [Google Scholar] [CrossRef]
  203. Luae, Z.; Nieb, G.; Beltonc, P.; Tangd, H.; Zhao, B. Structure–Activity Relationship Analysis of Antioxidant Ability and Neuroprotective Effect of Gallic Acid Derivatives. Neurochem. Int. 2006, 48, 263–274. [Google Scholar] [CrossRef]
  204. Lodovici, M.; Caldini, S.; Morbidelli, L.; Akpan, V.; Ziche, M.; Dolara, P. Protection against ultraviolet B-induced oxidative DNA damage in rabbit corneal-derived cells (SIRC) by 4-coumaric acid. Toxicology 2003, 184, 141–147. [Google Scholar] [CrossRef]
  205. Luceri, C.; Guglielmi, F.; Lodovici, M.; Giannini, L.; Messerini, L.; Dolara, P. Plant phenolic 4-coumaric acid protects against intestinal inflammation in rats. Scand. J. Gastroenterol. 2004, 39, 1128–1133. [Google Scholar] [CrossRef] [PubMed]
  206. Chowdhury, M.; Kabir, S.; Pal, A.; Pakrashi, A. Modulation of luteinizing hormone receptors: Effect of an inhibitor of prolactin secretion, p-coumaric acid. J. Endocrinol. 1983, 98, 307–311. [Google Scholar] [CrossRef]
  207. Kim, H.; Lee, S.; Hwang, E.; Maeng, S.; Park, J. p-Coumaric Acid Enhances Long-term Potentiation and Recovers Scopolamine-Induced Learning and Memory Impairments. Biochem. Biophys. Res. Commun. 2017, 492, 493–499. [Google Scholar] [CrossRef]
  208. Symes, A.; Shavandi, A.; Zhang, H.; Mohamed Ahmed, I.; Al-Juhaimi, F.; Bekhit, A. Antioxidant Activities and Caffeic Acid Content in New Zealand Asparagus (Asparagus officinalis) Roots Extracts. Antioxidants 2018, 7, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Kırmızıbekmez, H.; İnan, Y.; Reis, R.; Sipahi, H.; Gören, A.; Yeşilada, E. Phenolic compounds from the aerial parts of Clematis viticella L. and their in vitro anti-inflammatory activities. Nat. Prod. Res. 2018, 33, 2541–2544. [Google Scholar] [CrossRef] [PubMed]
  210. Fan, G.J.; Jin, X.L.; Qian, Y.P.; Wang, Q.; Yang, R.T.; Dai, F.; Tang, J.J.; Shang, Y.J.; Cheng, L.X.; Yang, J.; et al. Hydroxycinnamic acids as DNA-cleaving agents in the presence of Cu(II) ions: Mechanism, structure-activity relationship, and biological implications. Chemistry 2009, 15, 12889–12899. [Google Scholar] [CrossRef]
  211. Yun, K.J.; Koh, D.J.; Kim, S.H.; Park, S.J.; Ryu, J.H.; Kim, D.G.; Lee, J.Y.; Lee, K.T. Anti-Inflammatory Effects of Sinapic Acid through the Suppression of Inducible Nitric Oxide Synthase, Cyclooxygase-2, and Proinflammatory Cytokines Expressions via Nuclear Factor-κB Inactivation. J. Agric. Food Chem. 2008, 56, 10265–10272. [Google Scholar] [CrossRef]
  212. Graf, E. Antioxidant Potential of Ferulic Acid. Free Radic. Biol. Med. 1992, 13, 435–448. [Google Scholar] [CrossRef]
  213. Stewart, E. Adenine Compounds, Their Chemical, Physiological, and Therapeutic Properties. JAPhA 1948, 1, 3–9. [Google Scholar] [CrossRef]
  214. Changyuan, W.; Zhendong, S.; Haiqing, Y.; Kexin, L.; Xiaodong, M. Adenine: An important drug scaffold for the design of antiviral agents. Acta Pharm. Sin. B 2015, 5, 431–441. [Google Scholar] [CrossRef] [Green Version]
  215. Wang, S.; Wan, T.; Ye, M.; Qiu, Y.; Pei, L.; Jiang, R.; Pang, N.; Huang, Y.; Liang, B.; Ling, W.; et al. Nicotinamide riboside attenuates alcohol induced liver injuries via activation of SirT1/PGC-1α/mitochondrial biosynthesis pathway. Redox Biol. 2018, 17, 89–98. [Google Scholar] [CrossRef] [PubMed]
  216. Ma, A.; Medenica, M. Response of Generalized Granuloma Annulare to High-Dose Niacinamide. Arch. Dermatol. 1983, 119, 836–839. [Google Scholar] [CrossRef]
  217. Handfield-Jones, S.; Jones, S.; Peachey, R. High-dose nicotinamide in the treatment of necrobiosis lipoidica. Br. J. Dermatol. 1988, 118, 693–696. [Google Scholar] [CrossRef]
  218. Reiche, L.; Wojnarowska, F.; Mallon, E. Combination therapy with nicotinamide and tetracyclines for cicatricial pemphigoid: Further support for its efficacy. Clin. Exp. Dermatol. 1998, 23, 254–257. [Google Scholar] [CrossRef] [PubMed]
  219. Neumann, R.; Rappold, E.; Pohl-Mark, L. Treatmentof polymorphous light eruption with nicotinamide: A pilot study. Br. J. Dermatol. 1986, 115, 77–80. [Google Scholar] [CrossRef]
  220. Lampeter, E.; Klinghammer, A.; Scherbaum, W.; Heinze, E.; Haastert, B.; Giani, G.; Kolb, H. The Deutsche Nicotinamide Intervention Study: An attempt to prevent type 1 diabetes. DENIS Group. Diabetes 1998, 47, 980–984. [Google Scholar] [CrossRef]
  221. Hoffer, A. Safety, Side Effects and Relative Lack of Toxicity of Nicotinic Acid and Nicotinamide. Schizophrenia 1969, 1, 78–87. [Google Scholar]
  222. Gianfrilli, D.; Lauretta, R.; Di Dato, C.; Graziadio, C.; Pozza, C.; De Larichaudy, J.; Giannetta, E.; Isidori, A.M.; Lenzi, A. Propionyl-L-carnitine, L-arginine and niacin in sexual medicine: A nutraceutical approach to erectile dysfunction. Andrologia 2012, 44 (Suppl. S1), 600–604. [Google Scholar] [CrossRef]
  223. Adams, G.G.; Imran, S.; Wang, S.; Mohammad, A.; Kok, M.S.; Gray, D.A.; Channell, G.A.; Harding, S.E. The hypoglycemic effect of pumpkin seeds, Trigonelline (TRG), Nicotinic acid (NA), and D-Chiro-inositol (DCI) in controlling glycemic levels in diabetes mellitus. Crit. Rev. Food Sci. Nutr. 2014, 54, 1322–1329. [Google Scholar] [CrossRef] [PubMed]
  224. Sanyal, S.; Karas, R.; Kuvin, J. Present-day uses of niacin: Effects on lipid and non-lipid parameters. Expert Opin. Pharmacother. 2007, 8, 1711–1717. [Google Scholar] [CrossRef]
  225. Morris, M.C.; Evans, D.A.; Bienias, J.L.; Scherr, P.A.; Tangney, C.C.; Hebert, L.E.; Bennett, D.A.; Wilson, R.S.; Aggarwal, N. Dietary niacin and the risk of incident Alzheimer’s disease and of cognitive decline. JNNP 2004, 75, 1093–1099. Available online: https://0-jnnp-bmj-com.brum.beds.ac.uk/content/75/8/1093 (accessed on 30 July 2020). [CrossRef] [Green Version]
  226. Shi, H.; Enriquez, A.; Rapadas, M.; Martin, E.M.; Wang, R.; Moreau, J.; Lim, C.K.; Szot, J.O.; Ip, E.; Hughes, J.N.; et al. NAD Deficiency, Congenital Malformations, and Niacin Supplementation. N. Engl. J. Med. 2017, 377, 544–552. [Google Scholar] [CrossRef] [PubMed]
  227. Fu, L.; Doreswamy, V.; Prakash, R. The biochemical pathways of central nervous system neural degeneration in niacin deficiency. NRR 2014, 9, 1509–1513. [Google Scholar] [CrossRef]
  228. Nemazannikova, N.; Mikkelsen, K.; Stojanovska, L.; Blatch, G.; Apostolopoulos, V. Is there a Link between Vitamin B and Multiple Sclerosis? Med. Chem. 2018, 14, 170–180. [Google Scholar] [CrossRef]
  229. Dumont, M. Clinical, pathogenic and therapeutic study of muscular cramps in pregnant women; peculiar action of panthenol. Gynecol. Obs. 1951, 50, 501–509. Available online: https://europepmc.org/article/med/14917312 (accessed on 30 July 2020).
  230. Naruta, E.; Egorov, A.; Omel’ianchik, C.; Buko, V. The influence of panthotenic acid mitochondrial oxidation and oxidative phosphorylation in liver of rats with alimentary obesity. Vopr. Pitan. 2004, 73, 3–7. Available online: https://europepmc.org/article/med/15460980 (accessed on 3 August 2020). [PubMed]
  231. Zhang, Y.M.; Chohnan, S.; Virga, K.G.; Stevens, R.D.; Ilkayeva, O.R.; Wenner, B.R.; Bain, J.R.; Newgard, C.B.; Lee, R.E.; Rock, C.O.; et al. Chemical knockout of pantothenate kinase reveals the metabolic and genetic program responsible for hepatic coenzyme A homeostasis. Chem. Biol. 2007, 14, 291–302. [Google Scholar] [CrossRef] [Green Version]
  232. Voelkel, A. Mechanism of action of panthothenic acid in insulin shock therapy. Arch. Int. Pharmacodyn. Ther. 1956, 107, 271–274. [Google Scholar]
  233. Schutte, A.; van Rooyen, J.; Huisman, H.; Kruger, H.; Malan, N.; De Ridder, J. Dietary risk markers that contribute to the aetiology of hypertension in black South African children: The THUSA BANA study. J. Hum. Hypertens. 2003, 17, 29–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Gominak, S. Vitamin D deficiency changes the intestinal microbiome reducing B vitamin production in the gut. The resulting lack of pantothenic acid adversely affects the immune system, producing a “pro-inflammatory” state associated with atherosclerosis and autoimmunity. Med. Hypotheses 2016, 94, 103–107. [Google Scholar] [CrossRef]
  235. Stettler, H.; Kurka, P.; Kandzora, J.; Pavel, V.; Breuer, M.; Macura-Biegun, A. A new topical panthenol-containing emollient for maintenance treatment of childhood atopic dermatitis: Results from a multicenter prospective study. J. Dermatol. Treatm. 2017, 28, 774–779. [Google Scholar] [CrossRef]
  236. Bassaganya-Riera, J.; Skoneczka, J.; Kingston, D.G.; Krishnan, A.; Misyak, S.A.; Guri, A.J.; Pereira, A.; Carter, A.B.; Minorsky, P.; Tumarkin, R.; et al. Mechanisms of Action and Medicinal Applications of Abscisic Acid. Curr. Med. Chem. 2010, 17, 467–478. [Google Scholar] [CrossRef] [PubMed]
  237. Sánchez-Sarasúa, S.; Moustafa, S.; García-Avilés, Á.; López-Climent, M.; Gómez-Cadenas, A.; Olucha-Bordonau, F.; Sánchez-Pérez, A. The effect of abscisic acid chronic treatment on neuroinflammatory markers and memory in a rat model of high-fat diet induced neuroinflammation. Nutr. Metab. 2016, 13, 73–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Pilar Herrero, M.; Shafer, W. Pharmaceutical and Nutraceutical Compositions of Abscisic Acid. U.S. Patent 8,536,224, 19 September 2011. Available online: https://patents.google.com/patent/US8536224B2/en (accessed on 3 August 2020).
  239. Fernández-Ponce, M.; López-Biedma, A.; Sánchez-Quesada, C.; Casas, L.; Mantell, C.; Gaforio, J.; Martínez de la Ossa, E. Selective antitumoural action of pressurized mango leaf extracts against minimally and highly invasive breast cancer. Food Funct. 2017, 8, 3610–3620. [Google Scholar] [CrossRef] [PubMed]
  240. Whang, W.K.; Park, H.S.; Ham, I.; Oh, M.; Namkoong, H.; Kim, H.K.; Hwang, D.W.; Hur, S.Y.; Kim, T.E.; Park, Y.G.; et al. Methyl gallate and chemicals structurally related to methyl gallate protect human umbilical vein endothelial cells from oxidative stress. Exp. Mol. Med. 2005, 37, 343–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Correa, L.B.; Pádua, T.A.; Seito, L.N.; Costa, T.E.; Silva, M.A.; Candéa, A.L.; Rosas, E.C.; Henriques, M.G. Anti-inflammatory Effect of Methyl Gallate on Experimental Arthritis: Inhibition of Neutrophil Recruitment, Production of Inflammatory Mediators, and Activation of Macrophages. J. Nat. Prod. 2016, 79, 1554–1566. [Google Scholar] [CrossRef] [PubMed]
  242. Zeisel, S. Dietary choline: Biochemistry, Physiology, and Pharmacology. Annu Rev. Nutr. 1981, 1, 95–121. Available online: https://0-www-annualreviews-org.brum.beds.ac.uk/doi/pdf/10.1146/annurev.nu.01.070181.000523 (accessed on 7 August 2020). [CrossRef]
  243. Dronkers, N.; Baldo, J. Encyclopedia of Neuroscience; Squire, L., Ed.; Academic Press: La Jolla, CA, USA, 2009; Volume 10. [Google Scholar]
  244. Porteiro, B.; Fondevila, M.F.; Buque, X.; Gonzalez-Rellan, M.J.; Fernandez, U.; Mora, A.; Beiroa, D.; Senra, A.; Gallego, R.; Fernø, J.; et al. Pharmacological stimulation of p53 with low-dose doxorubicin ameliorates diet-induced nonalcoholic steatosis and steatohepatitis. Mol. Metab. 2018, 8, 132–143. [Google Scholar] [CrossRef]
  245. Best, C.; Huntsman, M. Effect of choline on liver fat of rats in various states of nutrition. J. Physiol. 1935, 83, 255–274. [Google Scholar] [CrossRef] [PubMed]
  246. Shao, M.; Dai, W.; Yuan, S.; Lu, Y.; Chen, D.; Wang, Q. Iridoids from Pedicularis verticillata and their anti-complementary activity. Chem. Biodivers. 2018, 15, e1800033. [Google Scholar] [CrossRef]
  247. Wang, K.; Chang, J.; Chiang, L.; Lin, C. 4-Methoxycinnamaldehyde inhibited human respiratory syncytial virus in a human larynx carcinoma cell line. Phytomedicine 2009, 16, 882–886. [Google Scholar] [CrossRef]
  248. Li, X.Q.; Liu, X.X.; Wang, X.Y.; Xie, Y.H.; Yang, Q.; Liu, X.X.; Ding, Y.Y.; Cao, W.; Wang, S.W. Cinnamaldehyde Derivatives Inhibit Coxsackievirus B3-Induced Viral Myocarditis. Biomol. Ther. 2017, 25, 279–287. [Google Scholar] [CrossRef] [Green Version]
  249. Srisook, E.; Palachot, M.; Mankhong, S.; Srisook, K. Anti-inflammatory effect of Etlingera pavieana (Pierre ex Gagnep.) R.M.Sm. rhizomal extract and its phenolic compounds in lipopolysaccharide-stimulated macrophages. Pharmacogn. Mag. 2017, 13, 230–235. [Google Scholar] [CrossRef] [Green Version]
  250. Germond, J.; Philippossian, G.; Richli, U.; Bracco, I.; Arnaud, M. Rapid and complete urinary elimination of [14C]-5-hydroxymethyl-2-furaldehyde administered orally or intravenously to rats. J. Toxicol. Environ. Health 1987, 22, 79–89. [Google Scholar] [CrossRef] [PubMed]
  251. Kang, H.M.; Son, H.S.; Cui, Y.H.; Youn, B.; Son, B.; Kaushik, N.K.; Uddin, N.; Lee, J.S.; Song, J.Y.; Kaushik, N.; et al. Phytosphingosine exhibits an anti-epithelial-mesenchymal transition function by the inhibition of EGFR signaling in human breast cancer cells. Oncotarget 2017, 8, 77794–77808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Jang, E.J.; Shin, Y.; Park, H.J.; Kim, D.; Jung, C.; Hong, J.Y.; Kim, S.; Lee, S.K. Anti-melanogenic activity of phytosphingosine via the modulation of the microphthalmia-associated transcription factor signaling pathway. J. Dermatol. Sci. 2017, 87, 19–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Kim, B.; Lee, J.; Jung, Y.; Kim, S.; Kim, T. Phytosphingosine Derivatives Ameliorate Skin Inflammation by Inhibiting NF-κB and JAK/STAT Signaling in Keratincoytes and Mice. J. Investig. Dermatol. 2014, 134, 1023–1032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Gupta, S.; Sharma, B. Pharmacological benefits of agomelatine and vanillin in experimental model of Huntington’s disease. Pharmacol. Biochem. Behav. 2014, 122, 122–135. [Google Scholar] [CrossRef]
  255. Iannuzzi, C.; Borriello, M.; Irace, G.; Cammarota, M.; Di Maro, A.; Sirangelo, I. Vanillin Affects Amyloid Aggregation and Non-Enzymatic Glycation in Human Insulin. Sci. Rep. 2017, 7, 15086–15100. [Google Scholar] [CrossRef] [Green Version]
  256. Hannemann, A.; Cytlak, U.; Gbotosho, O.; Rees, D.; Tewari, S.; Gibson, J. Effects of o-vanillin on K+ transport of red blood cells from patients with sickle cell disease. Blood Cells Mol. Dis. 2014, 53, 21–26. [Google Scholar] [CrossRef] [Green Version]
  257. Abraham, D.; Mehanna, A.; Wireko, F.; Whitney, J.; Thomas, R.; Orringer, E. Vanillin, a potential agent for the treatment of sickle cell anemia. Blood 1991, 77, 1334–1341. Available online: https://pubmed.ncbi.nlm.nih.gov/2001455/ (accessed on 9 August 2020). [CrossRef] [Green Version]
  258. Lirdprapamongkol, K.; Kramb, J.; Suthiphongchai, T.; Surarit, R.; Srisomsap, C.; Dannhardt, G.; Svasti, J. Vanillin suppresses metastatic potential of human cancer cells through PI3K inhibition and decreases angiogenesis in vivo. J. Agric. Food Chem. 2009, 57, 3055–3063. [Google Scholar] [CrossRef]
  259. Tai, A.; Sawano, T.; Yazama, F.; Ito, H. Evaluation of antioxidant activity of vanillin by using multiple antioxidant assays. Biochim. Biophys. Acta 2011, 1810, 170–177. [Google Scholar] [CrossRef]
  260. Makni, M.; Chtourou, Y.; Fetoui, H.; Garoui el, M.; Boudawara, T.; Zeghal, N. Evaluation of the antioxidant, anti-inflammatory and hepatoprotective properties of vanillin in carbon tetrachloride-treated rats. Eur. J. Pharmacol. 2011, 668, 133–139. [Google Scholar] [CrossRef] [PubMed]
  261. Belagali, Y.; Ullal, S.; Shoeb, A.; Bhagwath, V.; Ramya, K.; Maskeri, R. Effect of vanillin on lipid profile in a model of hyperlipidemia, a preliminary study. Indian J. Exp. Biol. 2013, 51, 288–291. Available online: http://nopr.niscair.res.in/handle/123456789/16551 (accessed on 9 August 2020). [PubMed]
  262. Colombani, J.; Raisin, S.; Pantalacci, S.; Radimerski, T.; Montagne, J.; Leopold, P. A nutrient sensor mechanism controls Drosophila growth. Cell 2003, 114, 739–749. [Google Scholar] [CrossRef] [Green Version]
  263. Haselton, A.; Sharmin, E.; Schrader, J.; Sah, M.; Poon, P.; Fridell, Y.W. Partial ablation of adult Drosophila insulin-producing neurons modulates glucose homeostasis and extends life span without insulin resistance. Cell Cycle 2010, 9, 3063–3071. [Google Scholar] [CrossRef] [Green Version]
  264. Kim, S.K.; Rulifson, E.J. Conserved mechanisms of glucose sensing and regulation by Drosophila corpora cardiaca cells. Nature 2004, 431, 316–320. [Google Scholar] [CrossRef] [PubMed]
  265. Lee, G.; Park, J.H. Hemolymph sugar homeostasis and starvation-induced hyperactivity affected by genetic manipulations of the adipokinetic hormone-encoding gene in Drosophila melanogaster. Genetics 2004, 167, 311–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Bai, H.; Kang, P.; Tatar, M. Drosophila insulin-like peptide-6 (dilp6) expression from fat body extends lifespan and represses secretion of Drosophila insulin-like peptide-2 from the brain. Aging Cell 2012, 11, 978–985. [Google Scholar] [CrossRef]
  267. Chatterjee, D.; Katewa, S.D.; Qi, Y.; Jackson, S.A.; Kapahi, P.; Jasper, H. Control of metabolic adaptation to fasting by dILP6-induced insulin signaling in Drosophila oenocytes. PNAS 2014, 111, 17959–17964. [Google Scholar] [CrossRef] [Green Version]
  268. Doupé, D.P.; Marshall, O.J.; Dayton, H.; Brand, A.H.; Perrimon, N. Drosophila intestinal stem and progenitor cells are major sources and regulators of homeostatic niche signals. PNAS 2018, 115, 12218–12223. [Google Scholar] [CrossRef] [Green Version]
  269. Padmanabha, D.; Baker, K.D. Drosophila gains traction as a repurposed tool to investigate metabolism. Trends Endocrinol. Metab. 2014, 25, 518–527. [Google Scholar] [CrossRef]
  270. Alfa, R.W.; Kim, S.K. Using Drosophila to discover mechanisms underlying type 2 diabetes. Dis. Model. Mech. 2016, 9, 365–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Chen, J.; Seol-min, K.; Kwon, J.Y. A Systematic Analysis of Drosophila Regulatory Peptide Expression in Enteroendocrine Cells. Mol. Cell. 2016, 39, 358–366. [Google Scholar] [CrossRef]
  272. Lahr, E.C.; Dean, D.; Ewer, J. Genetic analysis of ecdysis behavior in Drosophila reveals partially overlapping functions of two unrelated neuropeptides. J. Neurosci. 2012, 32, 6819–6829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Clark, A.C.; del Campo, M.L.; Ewer, J. Neuroendocrine control of larval ecdysis behavior in Drosophila: Complex regulation by partially redundant neuropeptides. J. Neurosci. 2004, 24, 4283–4292. [Google Scholar] [CrossRef]
  274. Williams, M.J.; Akram, M.; Barkauskaite, D.; Patil, S.; Kotsidou, E.; Kheder, S.; Vitale, G.; Filaferro, M.; Blemings, S.W.; Maestri, G.; et al. CCAP regulates feeding behavior via the NPF pathway in Drosophila adults. PNAS 2020, 117, 7401–7408. [Google Scholar] [CrossRef]
  275. Sieber, M.H.; Thummel, C.S. The DHR96 Nuclear Receptor Controls Triacylglycerol Homeostasis in Drosophila. Cell Metab. 2009, 10, 481–490. [Google Scholar] [CrossRef] [Green Version]
  276. Horner, M.A.; Pardee, K.; Liu, S.; King-Jones, K.; Lajoie, G.; Edwards, A.; Krause, H.M.; Thummel, C.S. The Drosophila DHR96 nuclear receptor binds cholesterol and regulates cholesterol homeostasis. Genes Dev. 2009, 23, 2711–2716. [Google Scholar] [CrossRef] [Green Version]
  277. Martelli, C.; Pech, U.; Kobbenbring, S.; Pauls, D.; Bahl, B.; Sommer, M.V.; Pooryasin, A.; Barth, J.; Arias, C.W.P.; Vassiliou, C.; et al. SIFamide Translates Hunger Signals into Appetitive and Feeding Behavior in Drosophila. Cell Rep. 2017, 20, 464–478. [Google Scholar] [CrossRef] [Green Version]
  278. Hermann-Luibl, C.; Helfrich-Förster, C. Clock network in Drosophila. Curr. Opin. Insect Sci. 2015, 7, 65–70. [Google Scholar] [CrossRef]
  279. Xu, K.; Zheng, X.; Sehgal, A. Regulation of feeding and metabolism by neuronal and peripheral clocks in Drosophila. Cell Metab. 2008, 8, 289–300. [Google Scholar] [CrossRef] [Green Version]
  280. Parisky, K.M.; Agosto, J.; Pulver, S.R.; Shang, Y.; Kuklin, E.; Hodge, J.J.; Kang, K.; Liu, X.; Garrity, P.A.; Rosbash, M.; et al. PDF cells are a GABA-responsive wake-promoting component of the Drosophila sleep circuit. Neuron 2008, 60, 672–682. [Google Scholar] [CrossRef] [Green Version]
  281. Chen, J.; Reiher, W.; Hermann-Luibl, C.; Sellami, A.; Cognigni, P.; Kondo, S.; Helfrich-Förster, C.; Veenstra, J.A.; Wegener, C. Allatostatin A signalling in Drosophila regulates feeding and sleep and is modulated by PDF. PLoS Genet. 2016, 12, e1006346. [Google Scholar] [CrossRef] [Green Version]
  282. Jung, S.-H.; Lee, J.-H.; Chae, H.-S.; Seong, J.Y.; Park, Y.; Park, Z.-Y.; Kim, Y.-J. Identification of a novel insect neuropeptide, CNMa and its receptor. FEBS Lett. 2014, 588, 2037–2041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Gáliková, M.; Klepsatel, P.; Xu, Y.; Kühnlein, R.P. The obesity-related Adipokinetic hormone controls feeding and expression of neuropeptide regulators of Drosophila metabolism. Eur. J. Lipid Sci Technol. 2016, 119, 1600138. [Google Scholar] [CrossRef]
  284. Puig, O.; Marr, M.T.; Ruhf, M.L.; Tjian, R. Control of cell number by Drosophila FOXO: Downstream and feedback regulation of the insulin receptor pathway. Genes Dev. 2003, 17, 2006–2020. Available online: http://www.genesdev.org/cgi/doi/10.1101/gad.1098703 (accessed on 9 August 2020). [CrossRef] [PubMed] [Green Version]
  285. Jünger, M.A.; Rintelen, F.; Stocker, H.; Wasserman, J.D.; Végh, M.; Radimerski, T.; Greenberg, M.E.; Hafen, E. The Drosophila forkhead transcription factor FOXO mediates the reduction in cell number associated with reduced insulin signaling. J. Biol. 2003, 2, 20. [Google Scholar] [CrossRef] [Green Version]
  286. Wu, Y.; Baum, M.; Huang, C.-L.; Rodan, A.R. Two inwardly rectifying potassium channels, Irk1 and Irk2, play redundant roles in Drosophila renal tubule function. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 309, R747–R756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Duckworth, W.C.; Bennett, R.G.; Hamel, F.G. Insulin degradation: Progress and potential. Endocr. Rev. 1998, 19, 608–624. [Google Scholar] [CrossRef] [Green Version]
  288. Valera Mora, M.E.; Scarfone, A.; Calvani, M.; Greco, A.V.; Mingrone, G. Insulin clearance in obesity. J. Am. Coll. Nutr. 2003, 22, 487–493. [Google Scholar] [CrossRef]
  289. Rudovich, N.; Pivovarova, O.; Fisher, E.; Fischer-Rosinsky, A.; Spranger, J.; Möhlig, M.; Schulze, M.B.; Boeing, H.; Pfeiffer, A.F. Polymorphisms within insulin-degrading enzyme (IDE) gene determine insulin metabolism and risk of type 2 diabetes. J. Mol. Med. 2009, 87, 1145. [Google Scholar] [CrossRef]
  290. Maianti, J.P.; McFedries, A.; Foda, Z.H.; Kleiner, R.E.; Du, X.Q.; Leissring, M.A.; Tang, W.J.; Charron, M.J.; Seeliger, M.A.; Saghatelian, A.; et al. Anti-diabetic activity of insulin-degrading enzyme inhibitors mediated by multiple hormones. Nature 2014, 511, 94–98. [Google Scholar] [CrossRef] [Green Version]
  291. Tundo, G.R.; Sbardella, D.; Ciaccio, C.; Grasso, G.; Gioia, M.; Coletta, A.; Polticelli, F.; Di Pierro, D.; Milardi, D.; Van Endert, P.; et al. Multiple functions of insulin-degrading enzyme: A metabolic crosslight? Crit. Rev. Biochem. Mol. Biol. 2017, 52, 554–582. [Google Scholar] [CrossRef] [PubMed]
  292. Kumar, S.; Chen, D.; Jang, C.; Nall, A.; Zheng, X.; Sehgal, A. An ecdysone-responsive nuclear receptor regulates circadian rhythms in Drosophila. Nat. Commun. 2014, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
  293. Moraru, A.; Cakan-Akdogan, G.; Strassburger, K.; Males, M.; Mueller, S.; Jabs, M.; Muelleder, M.; Frejno, M.; Braeckman, B.P.; Ralser, M.; et al. THADA regulates the organismal balance between energy storage and heat production. Dev. Cell 2017, 41, 72–81. [Google Scholar] [CrossRef] [Green Version]
  294. Zeggini, E.; Scott, L.J.; Saxena, R.; Voight, B.F.; Marchini, J.L.; Hu, T.; de Bakker, P.I.; Abecasis, G.R.; Almgren, P.; Andersen, G.; et al. Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat. Genet. 2008, 40, 638–645. [Google Scholar] [CrossRef] [PubMed]
  295. Sasorith, S.; Billas, I.M.; Iwema, T.; Moras, D.; Wurtz, J.M. Structure-based analysis of the ultraspiracle protein and docking studies of putative ligands. J. Insect Sci. 2002, 2. [Google Scholar] [CrossRef]
  296. Oro, A.E.; McKeown, M.; Evans, R.M. Relationship between the product of the Drosophila ultraspiracle locus and the vertebrate retinoid X receptor. Nature 1990, 347, 298–301. [Google Scholar] [CrossRef]
  297. Riddiford, L.M.; Ajami, A.M. Juvenile hormone: Its assay and effects on pupae of Manduca sexta. J. Insect Physiol. 1973, 19, 749–762. [Google Scholar] [CrossRef]
  298. International Agency for Research on Cancer—World Health Organization. Colorectal Cancer—Source: Globocan 2018; The Global Cancer Observatory: Lyon, France, 2019. [Google Scholar]
  299. Chu, F.; Cui, Y.; Li, K.; Xiao, X.; Zhang, L.; Zhang, L.; Wang, L.; Gao, L.; Yin, N.; Wu, H. Long noncoding RNA THOR is highly expressed in colorectal cancer and predicts a poor prognosis. Future Oncol. 2020, 16, 1911–1920. [Google Scholar] [CrossRef]
Figure 1. Ion chromatograms in positive and negative mode obtained using UHPLC-ESI-MS/MS.
Figure 1. Ion chromatograms in positive and negative mode obtained using UHPLC-ESI-MS/MS.
Antioxidants 09 01067 g001
Figure 2. Composition based on the number of representatives of each class of compounds in the bilberry ripe fruit aqueous (E1), methanolic (E2), and hydro-methanolic (E3) extracts.
Figure 2. Composition based on the number of representatives of each class of compounds in the bilberry ripe fruit aqueous (E1), methanolic (E2), and hydro-methanolic (E3) extracts.
Antioxidants 09 01067 g002
Figure 3. Hemolymph trehalose levels for Drosophila melanogaster Canton Special exposed to high-sugar diet with and without supplementation of hydro-methanolic bilberry extract (E3). Data are reported as average of triplicate experiments, based on Table S8.
Figure 3. Hemolymph trehalose levels for Drosophila melanogaster Canton Special exposed to high-sugar diet with and without supplementation of hydro-methanolic bilberry extract (E3). Data are reported as average of triplicate experiments, based on Table S8.
Antioxidants 09 01067 g003
Figure 4. Foldchange in neuropeptide gene expression of wild-type Drosophila melanogaster reared for 12 days on a high-sugar diet, as compared to a normal diet control. Data reported as average of triplicate experiments, as ratio of gene expression values on high-sugar diet to gene expression levels on normal diet control.
Figure 4. Foldchange in neuropeptide gene expression of wild-type Drosophila melanogaster reared for 12 days on a high-sugar diet, as compared to a normal diet control. Data reported as average of triplicate experiments, as ratio of gene expression values on high-sugar diet to gene expression levels on normal diet control.
Antioxidants 09 01067 g004
Figure 5. Foldchange in neuropeptide gene expression of wild-type Drosophila melanogaster reared for 12 days on a high-sugar diet, supplemented or not with 3% hydro-methanolic extract (E3). Data reported as average of triplicate experiments, as ratio of gene expression values on high-sugar diet to gene expression levels on normal diet control.
Figure 5. Foldchange in neuropeptide gene expression of wild-type Drosophila melanogaster reared for 12 days on a high-sugar diet, supplemented or not with 3% hydro-methanolic extract (E3). Data reported as average of triplicate experiments, as ratio of gene expression values on high-sugar diet to gene expression levels on normal diet control.
Antioxidants 09 01067 g005
Figure 6. Health benefits of the compounds identified in the three extract formulations, aqueous, methanolic, and hydro-methanolic. Only health promoting effects shown by more than 20% of the phytochemicals are presented. This figure is based on the summarized information in Table S6 [45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254,255,256,257,258,259,260,261].
Figure 6. Health benefits of the compounds identified in the three extract formulations, aqueous, methanolic, and hydro-methanolic. Only health promoting effects shown by more than 20% of the phytochemicals are presented. This figure is based on the summarized information in Table S6 [45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254,255,256,257,258,259,260,261].
Antioxidants 09 01067 g006
Table 1. Summary of the putative identification of phytonutrients in the extracts, with compounds pertaining to each extract (E1, E2, and E3) marked ✓—if present and ✗—if absent. Compounds are organized by chemical classification.
Table 1. Summary of the putative identification of phytonutrients in the extracts, with compounds pertaining to each extract (E1, E2, and E3) marked ✓—if present and ✗—if absent. Compounds are organized by chemical classification.
Chemical ClassificationPutative IdentificationE1E2E3
FlavonoidsAmpelopsin (Ampeloptin, Dihydromyricetin)
Avicularin (Quercetin-3-O-arabinofuranoside, Fenicularin)
Catechin or Epicatechin-O-hexoside
Cinnamtannin B1
Cinnamtannin D1
Cyanidin-3-O-arabinoside
Cyanidin-3-O-glucoside (Kuromanin, Asterin, Chrysanthemin)
Cyanidin-3-O-sambubioside (Sambicyanin, Gossypicyanin)
Cyanidin-O-(coumaroyl)hexoside
Delphinidin-3-O-arabinoside
Delphinidin-3-O-galactoside (Empetrin)
Delphinidin-O-(pentosyl)hexoside
Epicatechin
Epigallocatechin
Gallocatechin
Hyperoside (Quercetin-3-O-galactoside, Hyperin)
Idaein (Idein, Cyanidin-3-O-galactoside)
Isoquercitrin (Hirsutrin, Quercetin-3-O-glucoside)
Isorhamnetin-O-glucuronide
Kaempferol-3-O-glucuronide
Laricitrin (Myricetin-3′-O-methyl ether)
Laricitrin-O-hexoside
Malvidin-3-O-arabinoside
Malvidin-O-(coumaroyl)hexoside
Malvidin-O-hexoside
Myricetin
Myricetin-3-O-arabinoside
Myricetin-O-hexoside
Myricetin-O-pentoside isomer
Naringenin
Naringenin chalcone
Pentahydroxyflavone (Hypolaetin, Quercetin, Tricetin)
Peonidin-3-O-arabinoside
Peonidin-O-(coumaroyl)hexoside
Peonidin-O-(pentosyl)hexoside
Peonidin-O-hexoside
Peonidin-O-pentoside isomer
Petunidin-3-O-arabinoside
Petunidin-3-O-galactoside
Petunidin-O-(pentosyl)hexoside
Procyanidin B1 or B3
Prunin (Naringenin 7-O-glucoside)
Quercetin-3-O-[3-Hydroxy-3-methylglutaroyl-(→4)-rhamnoside]
Quercetin-3-O-glucuronide
Quercetin-O-(coumaroyl)hexoside
Quercetin-O-rhamnoside-O-pentoside
Quercitrin (Quercetin-3-O-rhamnoside)
Syringetin-O-hexoside
Trihydroxyflavanone
Amino acids2-Aminoadipic acid
Arginine
Asparagine
Glutamic acid
Histidine
Isoleucine or Leucine
Lysine
Phenylalanine
Threonine
Tryptophan
Tyrosine
γ-Aminobutyric acid (GABA)
PolyphenolsCaffeoylshikimic acid
Chlorogenic acid (3-O-Caffeoylquinic acid)
Coumaroylquinic acid
Coumaroyl-shikimate
Feruloylquinic acid
Gallic acid (3,4,5-Trihydroxybenzoic acid)
Carboxylic acids4-Coumaric acid
Caffeic acid
Dihydroxy-methoxybenzoic acid
Dimethoxy-hidroxycinnamic acid (Sinapic acid)
Ferulic acid
VitaminsAdenine
Nicotinamide
Nicotinic acid (B3)
Pantothenic acid (B5)
TerpenoidsAbscisic acid (ABA)
SugarsSaccharic acid
LactonesGulonic acid γ-lactone or δ-Gluconic acid δ-lactone
EstersMethyl gallate
AlkaloidsCholine
Iridoids7-Deoxyloganic acid
Miscellaneous4-Methoxycinnamaldehyde
5-Hydroxymethyl-2-furaldehyde
5-Methyl-2-furaldehyde
N-(2-Phenylethyl)acetamide
Phytosphingosine
Vanillin
Table 2. Total polyphenolic and flavonoid content, along with antioxidant activity of the three formulations of the bilberry ripe fruit extracts.
Table 2. Total polyphenolic and flavonoid content, along with antioxidant activity of the three formulations of the bilberry ripe fruit extracts.
Extract FormulationTotal Polyphenols [mg GAE/g dw]Total Flavonoids
[mg QE/g dw]
Antioxidant Activity
[%]
Aqueous extract (E1)12.48 ± 0.133.88 ± 0.1150.87 ± 0.12
Methanolic extract (E2)20.42 ± 0.177.27 ± 0.1277.32 ± 0.09
Hydro-methanolic extract (E3)21.68 ± 0.198.41 ± 0.1178.03 ± 0.16
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Neamtu, A.-A.; Szoke-Kovacs, R.; Mihok, E.; Georgescu, C.; Turcus, V.; Olah, N.K.; Frum, A.; Tita, O.; Neamtu, C.; Szoke-Kovacs, Z.; et al. Bilberry (Vaccinium myrtillus L.) Extracts Comparative Analysis Regarding Their Phytonutrient Profiles, Antioxidant Capacity along with the In Vivo Rescue Effects Tested on a Drosophila melanogaster High-Sugar Diet Model. Antioxidants 2020, 9, 1067. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9111067

AMA Style

Neamtu A-A, Szoke-Kovacs R, Mihok E, Georgescu C, Turcus V, Olah NK, Frum A, Tita O, Neamtu C, Szoke-Kovacs Z, et al. Bilberry (Vaccinium myrtillus L.) Extracts Comparative Analysis Regarding Their Phytonutrient Profiles, Antioxidant Capacity along with the In Vivo Rescue Effects Tested on a Drosophila melanogaster High-Sugar Diet Model. Antioxidants. 2020; 9(11):1067. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9111067

Chicago/Turabian Style

Neamtu, Andreea-Adriana, Rita Szoke-Kovacs, Emoke Mihok, Cecilia Georgescu, Violeta Turcus, Neli Kinga Olah, Adina Frum, Ovidiu Tita, Carmen Neamtu, Zsombor Szoke-Kovacs, and et al. 2020. "Bilberry (Vaccinium myrtillus L.) Extracts Comparative Analysis Regarding Their Phytonutrient Profiles, Antioxidant Capacity along with the In Vivo Rescue Effects Tested on a Drosophila melanogaster High-Sugar Diet Model" Antioxidants 9, no. 11: 1067. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9111067

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