Next Article in Journal
On the Mechanical Properties of N-Functionalised Dipeptide Gels
Next Article in Special Issue
Antioxidants of Natural Plant Origins: From Sources to Food Industry Applications
Previous Article in Journal
Novel Hit Compounds as Putative Antifungals: The Case of Aspergillus fumigatus
Previous Article in Special Issue
Ultrasound and Microwave Assisted Extraction of Opuntia Fruit Peels Biocompounds: Optimization and Comparison Using RSM-CCD
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fruit Seeds as Sources of Bioactive Compounds: Sustainable Production of High Value-Added Ingredients from By-Products within Circular Economy

1
MSc in Food Science and Technology, Ponta Grossa 84035010, Brazil
2
Graduate Program in Chemistry, State University of Ponta Grossa, Avenida Carlos Cavalcanti, 4748, Ponta Grossa 84030900, Brazil
3
Food Processing and Quality, Innovative Food System, Production Systems Unit, Natural Resources Institute Finland (Luke), Tietotie 2, FI-02150 Espoo, Finland
4
Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia
5
Food Chemistry and Food Development Unit, Department of Biochemistry, University of Turku, FI-20014 Turku, Finland
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(21), 3854; https://doi.org/10.3390/molecules24213854
Submission received: 8 October 2019 / Revised: 14 October 2019 / Accepted: 18 October 2019 / Published: 25 October 2019

Abstract

:
The circular economy is an umbrella concept that applies different mechanisms aiming to minimize waste generation, thus decoupling economic growth from natural resources. Each year, an estimated one-third of all food produced is wasted; this is equivalent to 1.3 billion tons of food, which is worth around US$1 trillion or even $2.6 trillion when social and economic costs are included. In the fruit and vegetable sector, 45% of the total produced amount is lost in the production (post-harvest, processing, and distribution) and consumption chains. Therefore, it is necessary to find new technological and environmentally friendly solutions to utilize fruit wastes as new raw materials to develop and scale up the production of high value-added products and ingredients. Considering that the production and consumption of fruits has increased in the last years and following the need to find the sustainable use of different fruit side streams, this work aimed to describe the chemical composition and bioactivity of different fruit seeds consumed worldwide. A comprehensive focus is given on the extraction techniques of water-soluble and lipophilic compounds and in vitro/in vivo functionalities, and the link between chemical composition and observed activity is holistically explained.

Graphical Abstract

1. Introduction

According to Homrich et al. [1], the circular economy (CE) is an umbrella concept that applies different mechanisms aiming to minimize waste generation, thus decoupling economic growth from natural resources. European Union (EU) countries are leading this concept by promoting the responsive and cyclical use of resources and contributing to sustainability [2]. From the economic standpoint, CE is a model that should replace the conventional, linear material, and energy flow models by addressing the issues of environmental deterioration, social equity, and long, sustainable economic growth [3]. In fact, a holistic assessment based on a dashboard of qualitative and quantitative indicators is necessary to encompass the environmental, economic, social, and technical dimensions of CE [2,4].
Much discussion has been dedicated to the true definitions and scopes of CE, yet it is not fully comprehended in quantitative terms according to Moraga et al. [2]. According to the United Nations Sustainable Development Goals, CE can improve social equity, promote economic growth, and permanently reduce the rate of extraction of raw materials by closing material loops [5,6]. According to these goals, more specifically Goal 9 and Goal 12, the technological progress is the foundation of efforts to achieve environmental objectives, such as increased resource and energy efficiency. There needs to be more investment in high-tech manufacturing for the increased efficiency and decreased waste for a sustainable and responsible consumption of natural resources. Indeed, net welfare gains from economic activities can increase by reducing resource use, degradation, and pollution along the entire life cycle in a food supply chain, while increasing the quality of life [7].
According to Slorach et al. [8], each year, an estimated one-third of all food produced is wasted; this is equivalent to 1.3 billion tons of food, which is worth around US$1 trillion or even $2.6 trillion when social and economic costs are included. This amount of food ends up rotting in the bins of consumers and retailers or spoiling due to poor transportation and harvesting practices. Following the CE principles and directives, ideally, this should be fully avoided, or if not possible, recycled to recover resources [8]. Food and pharmaceutical companies should decisively work on identifying niches within the value chains where such interventions have the greatest potential to improve the environment and enhance the social impacts of production, while integrating the economic feasibility in the production and consumption chains [9].
From 2007 to 2017, the largest fruit producers were China, India, Brazil, the USA, Turkey, Mexico, Spain, Indonesia, Italy, and the Philippines [10], in total accounting for more than 810 million metric tons of produce. The most produced items were: watermelons (118 million tons), bananas (114 million tons), apples (83 million tons), grapes (74 million tons), oranges (73 million tons), mango (51 million tons), tangerines (33 million tons), pineapple (27 million tons), peaches and nectarines (25 million tons), pears (24 million tons), lemons and limes (17 million tons), papaya (13 million tons), and plums/sloes (12 million tons) [11]. In the fruit and vegetable sector, 45% of the total production is lost in post-harvest, processing, distribution, and consumptions chains [12]. Therefore, it is necessary to find new technological and environmentally friendly solutions to utilize fruit wastes as new raw materials and develop scale-ups for the production of high value-added products [13,14,15,16,17].
Searching for solutions based on CE requirements, Sagar et al. and Ayoub et al. [18,19] reviewed the extraction technologies, bioactive compounds, and possible utilization of fruit and vegetable waste. According to these authors, on average, 25–30% of wastes are generated in the processing of fruits and vegetables, but this number can even be in a higher range of 30–50% for mangos and oranges, and up to 70% for durian, jackfruit, and mangosteen.
So far, the literature has proposed the use of fruit wastes in the development of new products as functional extracts and flours from grape waste [20,21], yogurts enriched with grape skin flour [22], cookies added with grape skin and grape seed flours [23], fish oil added with apple peel phenolics [24], avocado oil added with phenolic compounds [25], frozen yogurt containing sea buckthorn berry pomace [26], bologna sausage added with citrus fiber [27], meat products added with berry extracts [28], corn starch extrudate containing fruit pomace (cranberry, grape, apple, and blueberry) [29], oil-in-water emulsion containing Ander berry [30], and Dijon-style mustard added with berry fruit pomace [31], among other technological applications. Surprisingly enough, the peels and seeds of the most conventionally grown fruits and vegetables are not yet used to develop new foods, natural extracts, and pharmaceutical products [32,33,34,35]. This is probably because of the remarkable presence of pesticide residues in the peels [36].
Not only technological applications containing fruit side streams have been studied, but also the optimization of extraction parameters aiming to increase the level of bioactive compounds, namely phenolic compounds and carotenoids, to produce bioactive-rich extracts and then further add into different foods [37]. For instance, Takii et al. [38] studied the effects of ethanol (EtOH), extraction time, and temperature on the extraction rate of phenolic compounds and antioxidants from banana peel, whereas Wong et al. [39] optimized the extraction conditions of total phenolic content and antioxidant activity of passion fruit peel using the same factors (e.g., EtOH concentration in the extracting solution, time, and temperature). In the same line of research, Dorta et al. [40] assessed the effects of EtOH, water, and acetone as solvents and different temperatures on the extraction of antioxidants from mango peel and seeds. Table 1 contains some studies [15,28,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69] regarding the extraction of bioactive compounds from fruit seeds and the bioactivity already studied. It is clear that new studies regarding fruit seeds should be focused on the optimization of extraction conditions and on the extraction techniques so that new value-added extracts can be potentially used as ingredients by pharmaceutical and food sectors. The extraction methods are highlighted to provide useful information about new techniques that can be explored industrially for scale-up process, and therefore enlarge the use of natural side streams as raw materials to obtain bioactive-rich extracts. Aside from that, conventional extracting techniques, such as maceration, are time consuming, which can limit its applicability in the industrial sector.
Considering that the production and consumption of fruits has increased over the last years and following the need to find sustainable uses of underutilized fruit byproducts, this work aimed to describe the chemical composition and bioactivity of different fruit seeds consumed worldwide. A comprehensive focus is given on the extraction techniques of water-soluble and lipophilic compounds and in vitro/in vivo functionalities, and the link between chemical composition and observed activity is holistically explained.

2. Fruit By-Products (Seeds): Chemical Characterization and Bioactivities

2.1. Apple (Pyrus malus L.)

Apples are mainly consumed fresh; however, 25–33% of fruits are processed into juice, cider, jams, and other products. Globally, the most used processed apple product is the juice. The recovery of juice in the industrial processing is around 70–75%, whereas 25–30% w/w of apple pomace is generated. Seeds account for 2–4% w/w of the pomace [11,74]. Considering the massive production of apples, seeds represent a significant side stream.
The oil content of apple seeds varies from 17% to 29% v/w, and the main fatty acids are oleic (27.0–46.5% w/w) and alpha linoleic acid (43.8–60.0% w/w). Other prominent fatty acids are palmitic, stearic, and arachidic acids [75,76,77,78,79]. A high percentage of unsaturated fatty acids makes apple seed oil nutritionally favorable, having positive effects on lowering low-density lipoproteins (LDL) cholesterol and preventing risks of cardiovascular diseases [79]. Apple seed oil is also a rich source of tocopherols, sterols, and phospholipids [76,77,80]. According to Tian et al. [78], apple seed oil is active against bacteria, mildews were less sensitive to apple seed oil than yeasts, and the minimum inhibitory concentration (MIC) of apple seed oil ranged from 0.3 to 0.6 mg/mL. The observed biological activities showed that the oil had a good potential for uses in the food industry and pharmacy.
The nutritional value of protein from defatted apple seed meal was relatively low in the murine study performed by Opyd et al. [81]. However, the protein content of apple seeds is quite high, varying from 34 to 50% w/w, and it is rich in sulfur-containing amino acids. The fiber content of seeds is 3.9–4.3% w/w. Seeds also contain significant amounts of minerals such as phosphorus, potassium, magnesium, calcium, and iron, in the order of 720, 650, 510, 210 and 110 mg/100 g, respectively [78,79].
Apple seeds are a good source of polyphenols. According to Xu et al., Lu and Yeap Foo et al., and Schieber et al. [50,71,72], the seeds of apples contain phenolic acids (protocatechuic, chlorogenic, coumaric, ferulic, and caffeic acids), quercetin derivatives, (+)-catechin, (-)-epicatechin, proanthocyanin B2, and phloridzin. Phloridzin is the predominant phenolic compound, and its content has been reported to be 192 mg/100 g dry weight (DW) in dried apple seeds [71], 240–864 mg/100 g DW in the seeds of seven apple cultivars [50], 326–2235 mg/100 g in dried defatted apple seeds of 12 dessert and cider cultivars [52], and 64 mg/100 g DW in the seeds of Ida red cultivar [57]. Phloridzin contents are higher in seeds than in peels and pomaces [50,57].
Apple phytochemicals and especially phloridzin may have many beneficial health effects. It has anti-inflammatory and antioxidant activities, and it reduces hyperglycemia by blocking renal glucose resorption and intestinal glucose absorption through inhibition of the sodium–glucose symporters [38,82,83,84,85]. According to the more recent murine study by Shin et al. [86], phloridzin may prevent diet-induced obesity, hepatic steatosis, inflammation, fibrosis, and insulin resistance. Phloridzin can be potentially used as treatment for type 2 diabetes, as a weight loss agent for obesity, and in the acute management of hyperglycemia [83].
Although apple seeds seem to be an interesting nutritional source, the presence of toxic cyanogenic glycosides critically limit their utilization as a food ingredient. The amygdalin content of seeds from 15 apple cultivars varied from 1 to 4 mg/g. These amygdalin contents could generate 0.06–0.2 mg cyanide equivalents per gram of apple seeds [87]. These values are relatively high, because acute cyanide toxicity can occur in humans at doses between 0.5 and 3.5 mg/kg body weight [88]. However, many processing techniques can reduce the cyanide content. Processing allows contact between cyanogenic glycosides and endogenous enzymes, which results in the hydrolytic breakdown of cyanogenic glycosides to hydrogen cyanide [87]. According to a murine study from Opyd et al. [81] a daily dose of 160 mg/kg body weight of apple seed meal for two weeks did not exert any observable adverse effects. The results showed increased protein digestibility as well as exerted beneficial effects on the intestinal tract, blood lipid profile, and antioxidant status of rats.
In conclusion, apple seeds are rich in nutrients, high-quality oil, and polyphenols, and can be a rich source of functional ingredients for health-promoting foods, feeds, nutraceuticals, and cosmetics. However, the contents of bioactivities vary significantly between various cultivars. In addition, for utilizing apple seeds, there is a risk concerning cyanogenic glucosides that must be considered, and the seeds must be processed in such a way that amygdalin is not physiologically toxic.

2.2. Grape (Vitis Labrusca and Vitis Vinifera)

Grapes are an important cash crop throughout the world. The berries stand out for their bright color, juicy flavor, and high nutrient content, and they are rich in vitamins, anthocyanins, carotenoids, and several antioxidant compounds, which are substances that can effectively remove free radicals and delay senility in the body [89,90].
As reported by The Plant List [91], the grape genus Vitis includes 68 species. Responsible for studying multiple genera, the Department of Agriculture (USDA) and the Natural Resources Conservation Service (NRCS) in the United States have classified the Vitis L., namely: Vitis acerifolia Raf. (mapleleaf grape), Vitis aestivalis Michx. (summer grape), Vitis amurensis Rupr. (Amur grape), Vitis arizonica Engelm. (canyon grape), Vitis californica Benth. (California wild grape), Vitis cinerea Engelm. ex Millardet (graybark grape), Vitis girdiana Munson (desert wild grape), Vitis labrusca L. (fox grape), Vitis monticola Buckley (sweet mountain grape), Vitis mustangensis Buckley (mustang grape), Vitis × novae-angliae Fernald (peregrine grape), Vitis palmata Vahl (catbird grape), Vitis riparia Michx (riverbank grape), Vitis rotundifolia Michx (muscadine), Vitis rupestris Scheele (sand grape), Vitis shuttleworthii House (calloose grape), Vitis tiliifolia Humb. and Bonpl. ex Schult. (West Indian Grape), Vitis vinifera L. (European or wine grape), V. vinifera L. ssp. sylvestris Hegi (wild grape), and V. vulpina L. (frost grape).
The basis of the phenolic composition of grapes and derived products is flavonoids, such as anthocyanins, flavones, and flavonols; stilbenes, such as trans-resveratrol; and phenolic acids, as gallic, vanillic, syringic, and caffeic [53]. Fiume et al. [92] suggested that amongst all the phenolic extractables in grapes, about 10% are present in the pulp, 28–35% are present in the peel, and 60–70% are present in the seeds. Several studies claim that there are several biological activities in grape phenolic compounds, both in vitro and in vivo, viz. anti-angiogenic, anti-proliferative, pro-apoptotic, antioxidant, and other functions [93,94,95,96,97]. Specific phenolic compounds extracted from grapevines studied by Hakimuddin et al., Singh et al., Schlachterman et al., and Singletary et al. [94,98,99,100] demonstrated anti-cancer and anti-metastatic activity, most especially when tested in vivo and in vitro against breast cancer cells. According to Yang et al. [101], the mechanism of action that could prevent tumor initiation may be related to the antioxidant, anti-inflammatory, and anti-proliferative activity of grape phenolic compounds.
Burin et al. [102] studied grape juice (Vitis vinifera cv. Cabernet Sauvignon), and determined using liquid chromatography the quantification of phenolic compounds as gallic acid: 82.6 mg/L; vanillic acid: 108.5 mg/L; ellagic acid: 543.4 mg/L; and p-coumaric 367.5 mg/L, with DPPH (1,1-diphenyl-2-picrylhydrazyl radical) and ABTS (2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt) antioxidant analyzes showing values of 191.1 and 213.5 µmol Trolox equivalents/100 mL, respectively. Granato et al. [103], also studying grape juices (Vitis labrusca cv. Bordeaux/Concord/Niagara/Isabel), determined by liquid chromatography the presence of 68 mg/L p-coumaric acid, 155 mg/L of chlorogenic acid, 108 mg/L of myricetin, 2.08 mg/L of trans-resveratrol, and 71 mg/L of malvidin 3,5-diglucoside, with 83.5% of transition metal-chelating ability (Fe2+) and 44.54% DPPH inhibition.
As stated in a review by Salehi et al. [104], V. vinifera wines contain a variety of phenolic compounds, such as hydroxybenzoic acids (e.g., gallate dimers), hydroxycinnamic acids (e.g., trans-caffeic acid), pyranoanthocyanins, flavonols (e.g., quercetin and myricetin), flavones (e.g., luteolin), and flavanols in their polymeric forms (e.g., procyanidins dimers). Tkacz et al. [105] document that the average amount of total polyphenolic compounds in white grape seeds (Vitis vinifera cv. Pinot Gris) exceeds by at least nine times the value found in the pulp and by more than five times what is found in the peel. The antioxidant activity was highest in seeds and lowest in pulp, as in a study by Yilmaz et al. [106].
Composed of skin, stems, and seeds, grape pomace represents the largest waste in the wine and juice industry [107,108]. The seeds represent about 5% of the fruit weight, generating approximately 40–50% of solid residues in the production process [109]. Grape seed oil has several applications, mainly in cosmetic formulations. It can contribute to nutraceutical effects due to its composition and antioxidant capacity [28]. Widely available in vegetables and fruits, flavan-3-ols represent the most common group of typical flavonoids of the human diet, and are present in higher concentrations in fruit seeds [110].
Consisting of high oil and polyphenols, grape seeds also contain oligosaccharides that display prebiotic activity [111,112]. According to Bordiga et al. [113], the oligosaccharides identified in grape seeds using chromatography can be considered, in some combinations, as a “functional ingredient” with potential prebiotic activity for L. acidophilus P18806, allowing improved growth during in vitro fermentation. Since grape seeds and skins are more valuable sources in compounds with high biological activity than fruit pulp, further studies to determine the biological properties with in vivo and in vitro testing of the morphological parts of the grape are very important for designing food products with high-sensory health-related quality. Moreover, seeds are an integral part of bagasse; thus, increasing their use in the food, pharmaceutical, and cosmetic industries would be economically justified.
Deolindo et al. [64] state that the hydroethanolic extracts of grape seed (Vitis labrusca cv. Bordeaux) have not only chemical antioxidant activity (based on DPPH analysis, where 3637 ± 4 mg AAE/100 g was obtained for the seeds and 1033 ± 10 mg AAE/100 g for the skin, extracted with a 60% v/v ethanolic solution), but also the potential to inhibit ACE-I (angiotensin-I-converting enzyme) activity in vitro. Salehi et al. [114] justify the inhibition of ACE-I activity to phenolic compounds in grape juice, peel, and seeds mainly due to the presence of (+)-catechin, (–)-epicatechin, (+)-gallocatechin and (+)-epigallocatechin, these substances being alternatives to the use of synthetic drugs to control or prevent hypertension.
Harbeoui et al. [65] confirmed the anti-inflammatory ability of the ethanol/water extract (80:20, v/v) of Vitis vinifera L. grape seeds by inhibiting the expression and production of different inflammatory markers. The authors also suggested that the different mechanisms involved in this anti-inflammatory activity could be due to the synergism between several phenolic compounds, as well as the specific structural elements of flavonoids (dominant fraction), constituting the primary determinants of anti-inflammatory activity.
Studies of the extract (ethanol/acidic water) of seed oils from cold-pressed fresh grapes, performed by Cecchi et al. [28], demonstrate in vitro inhibitory activity against PTP-1B (protein tyrosine phosphatase 1B enzyme), which is an overexpressed enzyme in type 2 diabetes, showing maximum inhibition values (98%) for Vitis vinifera cv. Sauvignon Blanc and minimal inhibition (40%) for Vitis vinifera cv. Cabernet Sauvignon. The authors assert an inhibitory activity exerted by the isolated phenolic fraction of these oils, and argue that these results urge for further studies, so that it is possible to confirm the action of grape seed oils and understand the mechanism behind them.

2.3. Pomegranate (Punica granatum)

The pomegranate (Punica granatum) is a fruit belonging to the Punicaceae family, which is native to Iran and is currently cultivated worldwide due to its great ability to adapt to various climatic conditions [43]. Pomegranates are mainly composed of bark, arils, and seeds, where residues can represent up to 50% of the total weight of the fruit [115]. Recent studies have sought to find methods to avoid wasting these residues by assessing the following: phenolic composition and antioxidant activity [42]; concentration of linoleic acid isomers [43]; in vitro and in vivo anthelmintic activity in relation to Ascaridia galli [116]; enrichment of fatty acid profile and reduced activity of desaturases in rat livers [117]; and decreased risk of non-communicable chronic diseases such as cancer, cardiovascular disease, diabetes, and obesity [44].
Ethanolic [41,42] and methanolic extracts [43] of pomegranate seeds were analysed on the presence of flavonoids, condensed tannins, tocochromanols, and carotenoids [44]. Ambigaipalan et al. [44] performed three different extractions of free phenolic compounds that were esterified and insoluble-bound. The authors demonstrated the abundance of phenolic compounds in pomegranate seeds and that different results can be reported when using different solvents. In the aforementioned study, 47 phenolic compounds were identified using HPLC-DAD-ESI-MSn, which were grouped into phenolic acids, flavonoids, proanthocyanidins, and anthocyanins. The following compounds were identified for the first time in pomegranate seeds: protocatechuic acid; vanillic acid; gallic acid; brevifolin carboxylic acid; p-hydroxybenzoic acid hexoside; cis and trans-caffeic acid hexoside; derivative of caffeic acid hexoside; vanillic acid hexoside; ferulic acid hexoside; catechin; quercetin hexoside; cis and trans-dihydrokaempferol hexoside; ellagic acid; ellagic acid pentoside; ellagic acid deoxyhexose; ellagic acid hexoside; valoneic acid bilactone; digalloyl hexoside; and galloyl-hexahydroxydiphenoyl hexoside.
Yoshime et al. [45] evaluated the conjugated α-linolenic acids (CLnAs) present in oil extracted by cold pressing pomegranate seeds, as well as bitter gourd seeds (Momordica charantia L.). These compounds were associated with anti-inflammatory and antioxidant properties. The fatty acid profile was characterized by gas chromatography and palmitic (16:0), stearic (18:0), oleic (18:1 ω-9), and linoleic acids (18:2 ω-6) were detected in both of the analysed oils. Punicic acid (PA; C18:3-9c11t13c) was the major fatty acid (>50%) present in pomegranate seed oil.
The antioxidant activity of pomegranate has been analysed in several studies. These included antioxidant activity by the DPPH method; antioxidant activity by the ABTS method; β-carotene by the bleaching method; antioxidant activity by electron paramagnetic resonance (EPR) spectrometry; metal-chelating ability; and oxygen radical absorbance capacity (ORAC) [41,43,44,45]. In these studies, pomegranate seeds showed antioxidant activity in all the methods that were used. Derakhshan et al. [42] evaluated the positive correlation between phenolic compound content and antioxidant activity using the β-carotene bleaching method. It was concluded that different methods should be employed to assess the types of antioxidant mechanism of action, such as single electron transfer, transition metal chelator capacity, and hydrogen atom transfer [118].
Biological assays with different protocols and experimental conditions are often used to evaluate natural products. In addition to identifying phenolic compounds, Ambigaipalan et al. [44] also studied the inhibitory effects of α-glucosidase and pancreatic lipase, the inhibition of peroxyl and hydroxyl radical-induced super stranded scission, and the inhibition of LDL cholesterol oxidation, confirming the effectiveness of the pomegranate seeds in the assays. Ferreira et al. [119] performed in vivo studies using mice and nanoemulsions of pomegranate seed oil for improving the photostability and antinociceptive effects of non-steroidal anti-inflammatory drugs. Zarepourfard et al. [120] performed in vivo assays on cloned goats to confirm that dietary supplementation with pomegranate seeds can improve sperm motility and viability after freeze–thawing, as well as maintaining developmental competence. Additionally, Białek et al. [117] performed dietary supplementation with pomegranate seed oil in Sprague–Dawley rats. The fatty acid content, lipid oxidation biomarker levels, and the activity of the main enzymes that catalyse lipid metabolism, confirmed that pomegranate seed oil is an alternative source of conjugated linoleic acids.
The use of pomegranate seeds for industrial purposes is already underway. A study by Van Nieuwenhove et al. [43] included pomegranate extracts as a component for the enrichment of conjugated linolenic acids in yoghurt. By using the DPPH method, there was a confirmed enrichment of the fatty acid profile, as well as an increase in antioxidant activity in the fermented products, while maintaining their nutritional, microbiological, and sensory characteristics. However, in vitro and in vivo evaluations of antimicrobial, cytotoxic, and antioxidant activity may be improved methods to study pomegranate seed extracts in greater detail.

2.4. Camu-Camu (Myrciaria Dubia H.B.K. McVaugh)

Camu-camu is a fruit species from the Myrtaceae family that naturally grows near rivers and lakes in the Amazon basin [121]. Pulp and peel extraction industries often use its fruits and seeds, which accounts for 50% of the total fruit weight and great loos if discarded [15].
The camu-camu seeds are a potentially rich source of phenolic compounds, while their functional and biological effects were previously reported. Studies such as those by Fidelis et al., Myoda et al., Azevêdo et al., and Maria et al. [15,46,47,48,49] have verified the high concentrations of phenolic compounds in the seeds, which are even higher than the concentrations from camu-camu pulp. Fidelis et al. [15] identified the presence of bioactive compounds by HPLC coupled to diode array detection. Fifteen phenolic compounds (phenolic acids, flavonoids, and trans-resveratrol) were identified in aqueous, ethanolic, and propanone extracts. There was a significant difference (p < 0.05) in the content of all the bioactive compounds among the three obtained extracts, except for 2-hydroxycinnamic acid. Differences in the solubility and polarity of the compounds may explain these differences, as varying polarity of solvents can result in a different compound being extracted.
Do Carmo et al. [49] performed five extractions with different proportions of water and EtOH for the in vitro evaluation of phenolic composition, antioxidant activity, cytotoxicity, and the inhibition of cisplatin-induced chromosomal aberrations. The extract with 50% of each solvent presented the highest concentrations of total phenolic compounds. The highest antioxidant capacity evaluated by three different methods (DPPH radical scavenging; ferric reducing–antioxidant power, FRAP; and Folin–Ciocalteu reducing capacity) also inhibited the growth of four cancer cell lines without cytotoxic effects on normal cells. In addition, the extract also presented a protective effect by decreasing the cisplatin-induced chromosomal break index by 37%.
Myoda et al. [46] analysed the antioxidant activity of camu-camu seeds through DPPH radical elimination and reduction power tests. They used five fractions of crude extract obtained with different proportions of water and methanol and verified the effectiveness of the extract on in vitro antioxidant activity. The aforementioned study evaluated the antimicrobial activity of the five fractions of the extract against Staphylococcus aureus, Escherichia coli, and Saccharomyces cerevisiae bacteria. Here, antibacterial action was only observed in relation to Staphylococcus aureus bacteria. A further study by Kaneshima et al. [122] isolated C-glycosidic ellagitannins in camu-camu seed extracts, which showed antioxidant action against DPPH and ABTS radicals.
The evaluation of antibacterial and cytotoxic effects was performed by Camere-Colarossi et al. [123], who compared methanolic extracts of camu-camu pulp and seeds. Both extracts showed antibacterial activities against Streptococcus mutans (ATCC 25175) and Streptococcus sanguinis (ATCC 10556) bacteria. However, the seed extract presented the largest inhibitory halos, while the extracts showed no cytotoxic effects at the concentrations used.
Regarding in vivo studies, Yazawa et al. [124] studied the methanolic extracts of camu-camu seeds, and found anti-inflammatory action in carrageenan-induced paw oedema in murine models. Studies of camu-camu pulp are more frequently found in the literature [125,126,127,128,129]. However, it was stressed to use the waste that is generated while obtaining the pulp. In vitro and in vivo research with camu-camu seeds should be further developed, because this residue is recognized as a source of high concentrations of bioactive compounds and antioxidants.

2.5. Plums (Prunus sp.)

Plums (Prunus sp.) are among the most popular processed fruits. The global production of plums has been increasing and reached 11.8 million tons in 2017 [12]. Most often, plums are processed into dry fruits, jams, and juices, which generate tons of seeds as by-products. Thus, utilization of the seeds is of growing interest due to environmental and economic aspects.
The oil content of plum seeds varies from 23% (w/w) to 53% (w/w) per dry weight basis, depending mainly on the variety [73]. The dominant fatty acids in plum seed oil are oleic and linolenic acids; oleic acid constitutes 46–71% of the oil, and linoleic acid constitutes 23–45% of the oil, depending on the variety [73]. Based on the high content of unsaturated fatty acids, plum seeds have potential to lower LDL cholesterol and prevent cardiovascular diseases [79]. The oil is also rich in bioactive compounds, such as carotenoids, tocopherols, tocotrienols, phytosterols, and squalene [73]. The content of tocochromanols in 100 g of the oil is reported to be up to 209 mg, the content of total carotenoids is reported to be up to 3 mg, the content of phytosterols is reported to be up to 1579 mg, and the content of squalene is reported to be up to 80 mg [73]. Although the content of the bioactives varies greatly between the varieties, β-sitosterol and γ-tocopherol remain the main lipophilic compounds in plum seed oils. The contents of β-sitosterol constitutes are 209–1259 mg/100 g of oil and γ-tocopherol constitutes are 61 and 182 mg/100 g of oil [73].
Phytosterols, such as plant stanols and sterols, are steroid compounds that have a significant impact on human health. For example, phytosterols can reduce the levels of cholesterol in blood serum [130]. For example, tocochromanols include tocopherol and tocotrienol, which are active forms of vitamin E. They have important functions in human nutrition, and they possess various physiological and biological activities, such as the prevention of cardiovascular diseases and diabetes (e.g., [131]). Carotenoids are natural isoprenoid pigments with various biological and physiological functions, such as antioxidant and antitumor activities (e.g., [132]). One of the most common carotenoids in the plants is β-carotene, which exhibits pro-vitamin A activity. Squalene (2,6,10,15,19,23-hexamethyl-2,6,10,14,18,20-tetracosahexane) is a biochemical precursor of vitamin D, cholesterol, and steroid hormones, and thus is of high importance for human health [133].
In addition to the healthy fatty acids, lipophilic bioactives, and polyphenols, plum seeds have been characterized as a source of functional proteins and bioactive peptides. The protein content of plum seeds is up to 40% on a dry matter basis [134]. High-intensity ultrasound treatment has been applied for extracting the proteins [134,135]. The produced protein concentrates have been shown to possess good solubility, emulsifying, and foaming properties, as well as gel and film formation capacity. These functional properties indicate that the plum seed protein concentrates could meet the complex requirements of manufactured food products. For example, excellent emulsifying and gel-forming properties are needed in sausage products and emulsifying and film-forming capacities enable the production of emulsion-based edible films. In addition, proteins can be used for the production of biologically active peptides. This far, plum seed protein-derived peptides have shown antioxidant and antihypertensive (angiotensin I converting enzyme inhibitory) properties in vitro [134,136]. Bioactive peptides have been produced by hydrolyzing the plum seed proteins with proteolytic enzymes. Among the tested enzymes, alcalase most efficiently released the antioxidant and potentially antihypertensive peptides. The peptide sequences IYSPH, IYTPH, IFSPR, and VAIP have been reported to relate to the potential antihypertensive activity [136].
In summary, plum seeds are rich sources of nutrients, especially oils and proteins, and a wide variety of natural bioactive compounds. Based on these compounds, plum seeds have potential for exploitation in health-enhancing functional foods, nutraceuticals, extending the shelf life and active ingredients in food and cosmetic products. However, it needs to be taken into account that the concentration of the bioactive compounds varies significantly between the different varieties, and also, the seeds contain toxic cyanogenic glycosides, which need to be diminished during processing.

2.6. Jabuticaba (Plinia jaboticaba and Plinia cauliflora Mart. Kausel)

Jabuticabeira belongs to the Myrtaceae family, and it is a native species of Brazil with great economic importance. It is produced mainly in the states of Minas Gerais, Espirito Santo, Rio de Janeiro, São Paulo, and Paraná [137]. Due to its high water and sugar content, jabuticaba is highly perishable, and is not widely consumed in its fresh form. On the other hand, its pulp is usually extracted to make jams, wines, liqueurs, and vinegars [54]. According to Jorge et al. [138], the residues generated in the extraction of this fruit are seeds and peel, which represent approximately 50% of the fruit. The purposeful processing of these residues would reduce the production of organic wastes and add value to the food products, because they are a rich source of bioactive compounds, e.g., dietary fiber, minerals, and phenolic compounds [139].
Extraction is fundamental to obtain accurate results in relation to the studied matrix. To evaluate the phenolic compound content in jabuticaba seeds, Paludo et al. [53] performed optimized extraction using three solvents (water, EtOH, and methanol) and compared combinations of these solvents. The EtOH:water composition (60:40 v/v) had the highest extraction efficiency and proved the presence of high levels of phenolic compounds in the jabuticaba seeds. Inada et al. [54] evaluated the technological potential of the fractions (peel, seeds, and pulp) of jabuticaba and determined the antioxidant capacity and chemical composition. In the identification of phenolic compounds, soluble and insoluble fractions were extracted, and gallic acid was found to be the main compound present in jabuticaba seeds. The fractions showed in vitro antioxidant capacity in the methods used (Folin–Ciocalteu; ferric-reducing ability, Trolox equivalent antioxidant capacity, and oxygen radical antioxidant capacity). A positive correlation was observed between total phenolic compounds and antioxidant capacity, regardless of the assay, indicating that these bioactive compounds are effective in antioxidant action.
Similarly, Hacke et al. [55] performed the extraction and chemical characterization of antioxidant and antimicrobial compounds from jabuticaba seeds. The effects of three solvents (water, ethyl alcohol, and propanone) were analysed, and the binary combination of water and propanone (60:40 v/v) obtained the best results regarding phenolic content and antioxidant activity using the DPPH method After optimization, the crude extract was purified, and ellagic acid and ellagitannin compounds were identified using electrospray ionization coupled with tandem mass spectrometry (ESI-MS/MS). Alezandro et al. [56] evaluated jabuticaba fractions and also reported the presence of ellagic acid derivatives in jabuticaba seeds. In this same study, the methanolic extract of jabuticaba seeds presented higher antioxidant capacity when compared to the skin and pulp extracts, regardless of the method used (Folin–Ciocalteu reducing capacity, DPPH scavenging ability, or ferric-reducing ability). This study demonstrates how important it is to consume the whole fruit, because each fraction has its own particular importance and helps to maintain health.
Jorge et al. and Lima et al. [138,140] evaluated the profile of fatty acids and organic acids in jabuticaba seeds, respectively. Regarding fatty acids, polyunsaturated fatty acid concentrations were obtained, with a predominance of linoleic acid and α-linolenic acid. The organic acids that were detected included citric acid, succinic acid, malic acid, oxalic acid, and acetic acid. Such compounds are essential in maintaining health, because they contain beneficial properties for the body such as antioxidant, antimicrobial, and anti-inflammatory capacity.
In a practical use of jabuticaba seeds, Baldin et al. [139] evaluated the antioxidant and antimicrobial activity of the microencapsulated aqueous extract of jabuticaba residues (bark and seeds) used as dyes in the manufacture of mortadella. The addition of the extract provided better sensory acceptance, and did not interfere with the chemical composition, lipid oxidation, and bacterial development. According to the authors, the addition of natural extracts is promising and represents great potential for application during the manufacture of products enriched with natural antioxidants.
Jabuticaba peel has been identified as a potential source of bioactive compounds with biological activities. In vivo studies were performed by Batista et al. [141], who obtained an improvement in the triglyceride excretion and hepatic lipid peroxidation in rats fed a high-fat diet enriched with lyophilized jabuticaba peel. Palozi et al. [142] showed that the oral use of ethanolic extracts of jabuticaba peel in rabbits did not cause significant changes in respiratory, cardiovascular, and central nervous system functions, ensuring pharmacological safety for the use of this residue. However, biological studies of jabuticaba seeds are scarce, and further research is required to assure the complete safety of these potential sources of bioactive compounds. Hacke et al. [55] evaluated the protective effects of jabuticaba seed extract using in vivo micromolecular assays on murine bone marrow cells combined with cyclophosphamide. Ellagitannins (castalagin, vescalagin, and pedunculagin) were the main compounds isolated from jabuticaba seed extracts, which protected DNA from damage and accelerated DNA reparation.
Future research regarding jabuticaba seeds, particularly in relation to in vitro antioxidant, antimicrobial, and antiproliferative activities, as well as in vivo analysis, may provide more specific information on how to best utilize these residues.

2.7. Avocado (Persea americana Mill.)

Persea americana (Lauraceae), commonly known as avocado, is native to Mexico and Central America, and can be widely found throughout tropical countries. Most of avocado’s chemicals and bioactivity studies are focused on pulp, but little is known about the seed. Extensive research on this part of the fruit may be of great interest due to its anti-inflammatory properties (decreasing the generation of interleukin-6 pro-inflammatory mediators, IL-6; and prostaglandin-E2, PGE2; anti-cancer, antioxidant, and antihypertensive factors). Numerous chemical characterizations have shown a large number of polyphenols, such as catechins, procyanidins and other tannins, flavonoids, triterpenes, and unsaturated fatty acids in avocado seeds [143,144].
Using gas chromatography (GC) for the lipid extraction of avocado seeds, Alkhalf et al. [51] found linoleic (39.43%), oleic (25.30%), palmitic (20.05%), palmitoleic (3.66%), stearic (3.36%), and α-linoleic acids (2.62%). According to Alkhalf et al. [51], the extraction (chloroform/methanol) showed antioxidant activity using two electron transfer methods (ABTS and DPPH) at a concentration of 2 × 10−4 g/mL (69.73% and 36.64% inhibition, respectively) as compared to Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid) and BHT (2,6-bis(1,1-dimethylethyl)-4-methylphenol). The lipid extract (chloroform/methanol) of the seeds recorded significantly higher activity than that of the fruit. Through an in vitro experiment on human red blood cells, it was also revealed that both the pulp and seeds have significant dose-dependent anti-inflammatory potential compared to a control standard (membrane stabilization method). Still, according to Alkhalf et al. [51], the results show the antiproliferative activity of avocado seed and pulp lipid extractions in hepatocellular carcinoma (HepG2) and colon cancer (HCT116) cell lines.
In vivo studies performed by Athaydes et al. [61] demonstrated that avocado seed extracts with ethyl acetate have gastroprotective activity and can be used as an adjunctive treatment for indomethacin-induced gastric mucosal injury. Uchenna et al. [145] have shown promising component values in avocado seeds as a source of energy, protein, and bioactive phytochemicals to stimulate growth and metabolism. Additionally, the authors reported that the inclusion of avocado seeds in diets has also lowered cholesterol levels and suppressed hyperglycemia in rats.
Studies on avocado seed pigmentation conducted by Hatzakis et al. [146] demonstrated that the seed has perseorangan as the most abundant component, which may contribute to its final coloration. The authors stated that further research is needed to characterize the usefulness of this compound as a food coloring additive and to identify its biosynthetic precursors and potential natural derivatives. Likewise, toxicological studies should be performed on cells and mice to ascertain its safety. In vitro and in vivo studies of the extract from avocado seeds could be an alternative to better understand its biological activities and encourage its use as an ingredient in the cosmetic, pharmaceutical, and food industries.

2.8. Passion Fruit (Passiflora edulis Sims)

Passiflora, originally from the tropical and warm climates of South America, is the largest genus in the Passifloraceae family, containing about 500 different species. Due to their pleasant natural and intense bittersweet flavor, Passiflora fruits, popularly known as passion fruit, are much appreciated in the world. In the food industry, its juice is the main source of nutritional properties, mainly due to the presence of many phytochemical constituents such as phenolic compounds. Although hundreds of Passiflora species can be found worldwide, only a few are identified as edible. In addition, these fruits contribute to some beneficial health properties, such as antioxidant, anti-inflammatory, antipyretic, analgesic, sedative, and hypotensive activities [67,147,148,149]. Passiflora fruits are also rich in minerals (calcium and phosphorus), retinol, ascorbic acid, thiamine, riboflavin, and niacin [150].
In vitro studies using enzymes as a substrate show that phenolic and flavonoid compounds can inactivate α-amylase, α-glucosidase, and lipase through non-specific enzyme binding [151]. Passiflora species, such as P. edulis (flavicarpa), P. alata, P. incarnata, P. mollissima, P. tripartita, P. ligularis, and P. quadrangularis are widely cultivated and hold several biologically active secondary metabolites, including phenolic compounds.
C-Glycosylated derivatives of apigenin and luteolin, vitexin, isovitexin, orientin, schaftoside, 2-O-rhamnoside, luteolin-7-O-(2-rhamnoside), quercetin 3-β-glucoside, and iso-scoparin-2-O-glucosides are some new functional compounds of the Passiflora species [67,152,153]. According to Shanmugam et al. [149], protocatechuic acid is the main component found in P. subpeltata fruit pulp, quantified as 115.43 ηg/g, followed by ferulic acid (55.76 ηg/g), vanillic acid (46, 01 ηg/g), (–)-epicatechin (22.45 ηg/g), and p-coumaric acid (20.54 ηg/g). In addition, three compounds were found: cinnamic acid (9.08 ηg/g), eriodictyol (8.02 ηg/g), and quercetin-3-glucoside (3.04 ηg/g). Through DPPH analysis, the value of half of the maximum inhibitory concentration (IC50) was (5667 μg/mL) of fresh pulp, which is very close to standard rutin (5670 μg/mL) and BHT (6380 μg/mL) values. The maximum values found in fresh pulp relate to its antioxidant capacity, which eliminates free radicals by hydrogen donation, as defined by a previously reported hypothesis [154]. Rotta et al. [70], studying Passiflora species using liquid chromatography and the QuEChERS extraction method (quick, easy, cheap, effective, rugged, and safe), state that fruit pulp is an important source of phenolic compounds. In their studies with Passiflora pulp extract (acetonitrile), they determined phenolic compounds (quercetin, rutin, 4-hydroxybenzoic, chlorogenic, ferulic, vanillic, caffeic, trans-cinnamic, and p-coumaric acids). They also state that P. edulis showed the highest abundance of vanillic acid (426 ± 29 40 µg/kg) and quercetin (416 ± 6 µg/kg), whereas in P. alata and P. ligularis species, rutin (289 ± 6 41 µg / kg) and caffeic acid (64 ± 2 µg/kg), respectively, were found at the highest levels. Through DPPH and ABTS analyses, the authors concluded that all the extracts studied present antioxidant activity and phenolic content.
Studying tropical fruit by-products water extracts (mix of shell and seed in powder form), Albuquerque et al. [66] assert their potential to act or be a source of antioxidant dietary fiber. The passion fruit extract evaluated in the study is capable of acting as a prebiotic with antioxidant activity when analyzed through ORAC (oxygen radical antioxidant capacity) and DPPH. Using various types of extractions including maceration, (MAC), ultrasound-assisted leaching (UE), supercritical fluid extraction (SFE), and solvents (hexane, ethanol, ethanol/water and ethyl acetate), Oliveira et al. [69] demonstrated the antimicrobial activity of passion fruit (P. edulis sp.) seed in E. coli and L. innocua, presenting minimum inhibitory concentration (MIC) values of 8.0 mg/mL in the combination of maceration extraction (ethanol/water) and UE (ethanol/water); and 8.0 mg/mL in the combination of supercritical fluid extraction–150/300 bar 40 °C and 4.0 mg/mL 150 bar 50 °C, respectively. Lam and Ng [155], studying a new dimeric protein from passion fruit seeds, P. edulis (passiflin), determined specific antifungal activity against Rhizoctonia solani with an IC50 of 16 µM and a potent inhibitory action on breast cancer cells (MCF-7) with an IC50 of 15 µM.
Lourith and Kanlayavattanakul [156], studying the ethanolic extract of P. edulis seeds, state that the processing residue of passion fruit juice (seed) is a source of functional compounds, and the antioxidant fraction, determined by DPPH, FRAP, and ABTS analyses, works as a sunscreen with an effective protection against UV rays, as well as a skin-whitening agent. The sun protection capability of the extracts was confirmed by the fact that it contains quercetin and rosmarinic acid in its composition, which have potential action against photooxidative damage. Thus, this eco-friendly antioxidant is considered safe and effective for health-beneficial uses, and therefore, passion fruit seed can be used as an ingredient in the development of cosmetics, especially anti-aging and anti-wrinkle products, which are effective in protecting against UV, combating photoaging.
Malacrida and Jorge [157] confirmed through the study of passion fruit seed methanolic extract that the antioxidant amount present in the seed can serve as a dietary source of natural antioxidants, helping to prevent diseases by combating free radicals in vivo or as a food additive, increasing the stability and quality of food products. This antioxidant activity is attributed to phenolic compounds (linoleic, oleic, palmitic, and stearic) and to the presence of δ-tocopherol and γ-tocopherol (natural antioxidants). The authors further state that oil extraction from passion fruit seeds could add value to products, which is generally discarded as waste. The seed can also be used as a raw material in different industries, including food, detergents, cosmetics, supplements, vitamins, and biodiesel.
The results obtained by Macagnan et al. [158] suggest that apple, orange, and passion fruit by-products positively influence lipids and glucose metabolism, without negative effects in vivo, not affecting the growth of mice studied during the experiment period. By-products promoted a significant reduction in triglyceride and hepatic cholesterol levels and exerted important effects on postprandial glucose control. Furthermore, the authors found that these fiber sources are important for intestinal health without affecting the regularity of intestinal transit, demonstrating that apple pomace, orange pomace, and passion fruit peel can be considered as fonts of dietary fiber with relevant functional properties to health promotion and protection.

2.9. Berry Seeds

Most of the research published so far has been focused on the berry seeds from different species of Rubus (including blackberry, R. ulmifolius, R. fruticosus; raspberry, R. idaeus, R. occidentalis; and cloudberry, R. chamaemorus) and Vaccinium (blueberry, V. corymbosum; bilberry, V. myrtillus; and cranberry, V. oxycoccus, V. macrocarpon, V. oxycoccos). The seeds of sea buckthorn (Hippophae rhamnoides), strawberry (Fragaria × ananassa), and blackcurrant (Ribes nigrum) have been investigated. The major bioactive compounds and bioactivities of berry seeds are summarized in Table 2 [19,159,160,161,162,163,164,165,166,167,168,169,170].
The bioactive compounds found in berry seeds mainly are phenolic compounds, fatty acids, and tocopherols. Previous studies shown in Table 2 showed a large deviation in the total content of phenolic compounds in each berry species. It is difficult to compare the abundance of phenolics among different berry seeds, since the values are influenced by many factors, such as cultivars of berries, cultivation condition, extraction solvents, and analytical methods. The primary phenolic compounds in seeds are dependent on the berry species. Ayoub et al. [19] compared the phenolic profiles of seed meals of blackberry, raspberry (black), and blueberry.
Blackberry seeds contained the highest amounts of total phenolics (13 mg gallic acid equivalents - GAE/g defatted seed meals), followed raspberry (7 mg GAE/g) and blueberry (2 mg GAE/g). Based on the content obtained by HPLC, the phenolic compounds in these three seeds mainly are phenolic acids, proanthocyanidins, flavan-3-ols, flavonols, and anthocyanins; representing as free, esterified, and insoluble-bound forms. Gallic acid and its derivatives were the main phenolic acids in all berry seed meals. For flavonoids, B-type procyanidin dimers were the dominant compounds in blueberry seeds. However, quercetin, together with its glycosides, accounted for over 50% of the total flavonoids in the seeds of both blackberry and raspberry. van Hoed and co-workers investigated phenolic compounds in the seed oils of raspberry (red), blackberry, cranberry, blueberry, and strawberry by using HPLC-DAD [167]. The total amount of phenolics (mainly as phenolic acids) in cold-pressed berry seed oils varied from 0.1 (blackberry) to 15.8 mg/g of oil (strawberry). p-Coumaric acid formed the major phenolic compounds in the seed oils of cranberry, while 4-(2-hydroxyethyl)phenol was dominant in both strawberry and raspberry seed oils. Other compounds, such as homovanillic acid, vanillic acid, vanillin, and protocatechuic acid were quantified in studied oil samples. Ellagitannins were abundant in the seeds of cloudberries and blackberries [171,172]. The primary isomers included pedunculagin, casuarictin/potentillin, castalagin/vescalagin, lambertianin A/sanguiin H-6, lambertianin C, and lambertianin D. The wealth of these isomers varied in the different cultivars of berries [166]. As a large group of phenolics, lignans were also detected form the seeds of cloudberry, blackberry, cranberry, bilberry, raspberry, sea buckthorn, and blackcurrant. The total content ranged from 0.02 (blackcurrant) to 0.4 mg/g of dry seeds (cloudberry) [173].
Previous research suggested that fatty acids in berry seeds were present mostly as polyunsaturated fatty acids. They accounted for 83% in raspberry seeds [160], 65–78% in strawberry seeds [159,160], 78–79% in blackcurrant seeds [159,170], 75% in cranberry seeds [159], and 64% in bilberry seeds [159]. Although the concentrations varied among berry species, the dominant fatty acids were linoleic acid (C18:2, n-6, 12–62%), α-linolenic acid (C18:3, n-3, 4–44%), and oleic acid (C18:1, n-9, 4–45%) [159,160,163,164,165,170,174,175]. The n-6/n-3 ratio was close to 1 in the seed oils of elderberry (Sambucus nigra), strawberry, cranberry, bilberry, and raspberry, while the value ranged from 2.7 to 3.6 in blackberry and blackcurrant seeds [159,160,163,164,165,170]. Both tocopherols and tocotrienols were detected in berry seeds. The good sources were the seeds of raspberry (44–420 mg/100 g oil, mainly as γ-tocopherol), cloudberry (270, α-tocopherol and γ-tocopherol), cranberry (114–195, γ-tocotrienol), blackberry (164, γ-tocopherol), sea buckthorn (100–200, α-tocopherol and γ-tocopherol), lingonberry (Vaccinium vitis-idaea, 130, γ-tocotrienol), elderberry (122, γ-tocotrienol), and blackcurrants (106–110, α-tocopherol and γ-tocopherol) [159,160,163,167,174].
The antioxidant activity of berry seeds has been studied extensively [19,159,160,161,162,163,164,165,167,168,169,170]. Most of the studied seed oils or seed extracts have shown strong capacities of directly scavenging free radicals, chelating metal cations, or inhibiting the oxidation (or DNA damage) caused by free radicals. Yet, the compounds responsible for anti-oxidative effects have not been determined, which is likely due to the large chemical diversity that was found in the extracts. The related conclusions have been drawn only based on the correlation between antioxidant results and the concentration of chemical components. Furthermore, it is difficult to rank berry seed extracts based on their antioxidant effects due to different extraction solvents applied. For example, Helbig and co-workers evaluated both the hexane and water extracts of several seed press residues using the trolox equivalent antioxidant capacity - TEAC method [159]. The results showed that the antioxidant capacity of both hexane and water extracts decreased in the order of elderberry > blackcurrant > cranberry > bilberry. Similarity might be caused by the presence of different bioactive compounds in extracts. Additionally, considering the nature of free radicals, the inhibitory effects of certain berry seeds might differ in various antioxidant assays. Yang et al. reported the peroxyl radical scavenging capacities of supercritical CO2 extracted berry seed oils [163]. The activity was positively correlated with the content of tocopherols and tocotrienols in the seed oils. Among the seed oils studied, raspberry seed oils exhibited the best activity against peroxyl radicals, but the lowest efficacy of inhibiting lipid oxidation compared to seed oils from the other species of the Rubus and Vaccinium families. In contrast, bilberry seed oil was a potent inhibitor against lipid oxidation, yet weakened with scavenging peroxyl radicals [163]. In the same research, the authors reported the protective effects of sea buckthorn seed oil against oxidative damages of DNA.
Regarding other bioactivities of berry seed extracts, Basu et al. [176] confirmed that that supercritical CO2-extracted sea buckthorn seed oil had a significant anti-atherogenic activity when administrated to normal or hypercholesterolemic rabbits. The ethanolic extracts of sea buckthorn seeds strongly inhibited the growth of Enterecoccus durans (68%), Candida albicans (68%), and Bacillus cereus (64%). This was attributed to the high levels of total phenolic compounds, which were represented primarily with condensed tannins [161]. Puupponen-Pimiä et al. [169] evaluated two fractions of acetone extracts of cloudberry seeds in liquid cultures of selected microbial strains. Both fine and seed coarse seed fractions showed strong antibacterial efficacy on the growth of Staphylococcus aureus and Escherichia coli, whereas inhibition was observed on C. albicans and Saccharomyces cerevisiae. It was speculated that high contents of casuarictin/potentillin and sanguiin H2 isomers in the fractions might be responsible for the strong antimicrobial activity [171]. In the same study, the anti-inflammatory abilities of cloudberry seeds were estimated in a dose-dependent manner and caused a significant decrease on pro-inflammatory factors (bacterial lipopolysaccharide induced nitric oxide, NO, IL-6, and inducible nitric oxide synthase, iNOS). As compared to the seed fine faction, the coarse counterpart had the stronger anti-inflammatory effects at the concentration of 1–3 µg/mL, which was associated with the presence of quercetin derivatives [171]. The methanolic extracts of seeds of blackberry and elderberry were tested against bacterial lipopolysaccharide-induced NO and CCL20 in macrophages. The results revealed that blackberry seed extracts had a strong anti-inflammatory property, inhibiting 60% of NO and over 90% of CCL20 production at the dose level of 50 µg/mL. Nevertheless, no inhibitory effect was detected in elderberry seed extracts [165].
The clinical evidence on the health-promoting effects of berry seeds has been summarized by Yang and Kortesniemi [177]. Dietary supplementations with berry seed oils have shown positive impacts on atopic dermatitis [178], plasma lipid profiles [178,179], and platelet aggregation [180,181]. The beneficial effects were due to the high content of polyunsaturated fatty acids with favorable ratios of ω6/ω3 that were close to the recommendation given by the global experts. However, other lipophilic components such as phytosterols, tocopherols, and tocotrienols may also play an important role. [101,163,181] reported that the supplementation with 2 g of sea buckthorn oil (extracted from both seed and pulp) for three months has been shown to have a positive effect on human subjects between 20 and 70 years old suffering from dry eye symptoms [179]. Moreover, Linnamaa et al. [182] suggested that the prevalence of atopic dermatitis in infants could be reduced by dietary intervention using blackcurrant seed oil, when administered to both women during pregnancy and lactation, and to the babies after weaning.

3. Extraction Technologies of Water-Soluble and Lipophilic Bioactive Compounds

The extraction of bioactive compounds has a long tradition using conventional solvent extraction techniques based on different organic solvents. However, the demand for improving the extraction yield of the target bioactive compounds from plant matrices and the pressure on the environmental friendliness of the production process indicated the need for the development of novel extraction techniques that are based on benign pre-extraction technology by the implementation of enzymes [183]. Marathe et al. [184] described the effectiveness of the enzymatic hydrolysis in the reduction of cellular wall thickness to ensure the better passage of the solvent toward compounds that may be bound in the wall. This way, the extraction will eventually lead to higher yields, more efficiency, a reduction in the processing time and the solvent used, and at the end, a prospective technological solution that is sustainability and commercially viable.
Different enzymes can be used to facilitate the extraction of bioactive compounds, e.g., phenolics from grape seed as an example. Fernández et al. [185] compared the effectiveness of pectinases, cellulases, and tannases, and the blend of these enzymes. All the studied enzymes increased the total phenolic content in the extracts (versus controls) with a 1.18–1.34-fold increased yield. Comprehensive collection on the findings for enzymatic aids in processing for the extraction of polyphenols was recently published by Gligor et al. [186]. This review gives information on the use of hemicellulases and lignanases for the hydrolysis and oxidizing of cellular walls. However, González-García et al. [136] describe that the use of lignanases is not viable for food industrial applications, due to the formation of low-molecular reactive compounds in the oxidation process. The authors reported that although there are positive outcomes regarding the efficiency and environmental friendliness of enzyme-assisted extractions, the high costs and low popularity slows down their prevalence for food industrial applications.
Recently, the combination of these enzymes with microwave, ultrasound, supercritical fluid extraction, or high-pressure extraction was described in order to overcome the drawbacks mentioned above. The target compounds that strongly bind to the cell walls usually will not be fully released with enzymatic digestion by polysaccharide-degrading enzymes or hemicellulases [187]. It was reported that approximately 24% of the total phenolics are bound in the food matrices with polysaccharides (e.g., hemicellulose, cellulose, and pectin), and with certain biomolecules, as proteins. From this point of view, enzymatic utilization of a different nature can significantly enhance the recovery of the phenolics. Pap et al. [188] showed that the enzymatic depectinization of the blackcurrant juice by pectinase significantly increased the amount of available flavonols, but a similar observation for anthocyanins were not observed. Fernandes and Carvalho [189] furthermore explained that the treatment with cellulases, hemicellulases, and pectines resulted in the formation of smaller oligosaccharide chains and the liberation of compounds, such as oils and proteins.
Ultrasound techniques are based on the acoustic cavitation in which small vapor-filled bubbles are generated. These bubbles will eventually explode and result in high pressure and temperature in the cell walls [190]. Ultrasound waves that are exploited for the treatments of different food materials are generally over 20 kHz [191]. The application of ultrasound extraction (US) for polyphenols from different fruit seeds has already been covered in the literature. Segovia et al. [62] studied the batch and continuous US for recovering polyphenols from avocado seeds. These authors also modeled the extraction process and described that the film theory and Fick’s law models could sufficiently describe the US process, for both batch and continuous phase. Such a mathematical modeling provides understanding on the proceeding of the extraction.
Da Porto et al. [192] investigated the extraction of oils and polyphenols in US from grape seeds and compared the results with conventional extraction. The results indicated that the removal of oil was almost as efficient with US as with Soxhlet when a power of 150 W for 30 min was used. Comparing the polyphenolic extraction from grape seed by maceration versus US, these authors reported that the preliminary treatment with US increased the yield of polyphenols in maceration. However, a significant decrease of polyphenolic content was observed with double US treatments, when both the oil removal and the polyphenolic extraction were done by the US. Since polyphenolic compounds are naturally heat sensitive, this observation can be attributed to the significantly higher temperatures and pressures in the US hot spots [193] and consequential breakdown of the polyphenols.
Opposite to ultrasounds, microwaves are non-ionizing radiations at frequencies between 300 MHz to 300 GHz [194]. Considering the mechanism of microwave action, they are different in nature in a way that heating, and the extent of the absorbed heat will depend on the ability of polar solvents to absorb the microwave energy. Ionic conduction and dipole rotation of the solvent will result with heating, which is selective in nature, and opposite to conventional extractions where generally, the vessel is heated, and the solvent absorbs the heat through it. Li et al. [195] reported the effectiveness of the microwave-assisted extraction (MAE) of polyphenols from grape seeds. The authors optimized the MAE of the grape seeds in a five-level, three independent variable central composite rotatable design, and concluded that the yield of polyphenols in MAE extracts were comparable to that of US extraction and conventional extraction. However, a significant benefit was the much shorter time that was required for the extraction. The length of the process is always a crucial factor, since some of the compounds, especially heat-sensitive ones, may start to degrade during the course of the processing. On the other hand, Bucić-Kojić et al. [196] developed a statistical model for the description of the kinetics of grape seed phenolics depending on the temperature. The understanding and the implementation of such modeling is an extremely useful tool in process scale-up and industrial application as well.
Pressurized hot water extraction also plays an important role in novel extraction techniques. This extraction technology is gaining more interest for the production of bioactive compounds. Here, the pressure is adjusted to keep the water in its liquid state, and the temperature is between the boiling and critical point [197]. This technology benefits from the changes of the dielectric constant of water, as increasing the temperature up to 200–350 °C will decrease the dielectric constant of the water and provide similar polarities to those that methanol, EtOH, or acetone have at room temperature [198]. The hot water is usually recirculated, or the process can be also multi-staged. The complexity of the technology increases when matrices with multiple compounds are used in real applications, since the operation parameters must be well manipulated in order to extract the desired compound without the other contaminants. However, it is important to avoid undesirable changes of the target compounds that could deteriorate during the extraction. Plaza and Marina [199] published a comprehensive review on the potential application of the pressurized hot water extraction. The application for the extraction of phenolic compounds, diterpenes, triperpenes, polysaccharides, and proteins from different plant materials, peels, leaves, seeds, barks, and wastes were covered, and a positive trend for their future importance was indicated by this technology. Beside, several authors reported the results of the study on the extraction of polyphenolic compounds [198,200] from grape peels and pomegranate seed residues [201] for anthocyanins from grape pomace [202].
In supercritical fluid extraction, the most commonly used solvent is carbon dioxide, which due to its low critical temperature allows the extraction of thermally sensitive compounds [184,203]. Still, the challenge is related to the high cost of processing as compared to a low yield of extracted compounds. Nonetheless, the nature of the CO2 as a solvent enables the extraction of compounds with non-polar character. However, when used alone in the extraction, it is inadequate for the extraction of highly polar compounds, e.g., polyphenols being one of them [204]. Due to this, for the most polyphenolic recovery from plant seeds, the CO2 is combined with other solvents, (e.g., ethanol) for potential applications in food production. Castro-Vargas et al. [129] extracted bioactives from guava seeds with supercritical CO2 extraction combined with EtOH, and found a correlation between the applied pressure and temperature on the antioxidant activity of the extracts.
Yilmaz et al. [203] pointed out that the challenge with the valorization of the grape seeds is the residual oil obtained along with the phenolic compounds. Hexane is commonly used for the conventional extraction of oil, but it needs additional evaporation step(s) to remove the residual solvent after the defatting. The authors reported that with the co-addition of ethanol in the supercritical extraction, with a suitable adjustment of pressure and temperature, proanthocyanidins were successfully recovered from the grape seeds. Da Porto and Natolino [205] used response surface optimization to estimate the optimum process parameters for the extraction of polyphenols from white grape seeds in a two-stage supercritical CO2 extraction. The optimization was carried out by studying the effects of pressure, the amount of ethanol as co-solvent, and the CO2 flow rate for the number of total polyphenols and proanthocyanidins in the extracts. The results indicated that the optimization was crucial in order to make a good compromise between the concentration of the valuable compounds of the extracts, and also the length of the extraction process to ensure the economic feasibility for industrial application.
The above summary describes the most common novel extraction techniques that are combined with the enzymatic treatments of the plant materials. However, Ran et al. [206] recently reported the application of the conventional extraction of proanthocyanidins from grape seeds with boosting the extraction with the addition of ionic liquids as adjuvants. The authors reported on the increase of the extraction efficiency when adding ionic liquids. However, they also concluded that further studies are required to benefit fully from the utilization of ionic liquids, including the recovery and reuse of the solvents. On the other hand, the widespread application of ionic liquids in food industrial processes are hindered due to the lack of proper information on their toxicity, and stability and bioactivity limits [207].
When the most promising technological concept is selected for the extraction of bioactive compounds from plant seeds, the extract of the target compounds can undergo downstream processing, such as (pre)concentration by the membrane technology and drying. Shi et al. [208] showed that the concentration of polyphenols with membranes was useful after the extraction with 50% ethanol and 50% water mixtures when a 0.22-µm membrane was applied. Pap et al. [209] also reported on the concentration of anthocyanins and flavonols of blackcurrants in juice by using a reverse osmosis process. An approximate 1.5-fold increase of anthocyanins and flavonols was observed when the process was combined with pectinase enzyme treatment. Conidi et al. [210] successfully implemented a multistep ultrafiltration (150 kDa and 2 kDa membranes) of pomegranate juice to separate anthocyanins from glucose and fructose, and to produce end-products as nutraceuticals and food additives. These findings emphasized the importance of the proper selection of membranes to separate and pre-concentrate the bioactive compounds before the final drying. Due to the heat sensitivity of the bioactives, drying is a challenging step in the downstream processing. For instance, although freeze-drying well preserves the compounds, its widespread industrial application for extract processing will be likely impeded because of the price.
As final remarks, enzyme-assisted extraction, and its combination with novel extraction techniques are gaining importance. There is an increased need to improve the performance of these processes so that they become more resource efficient. In the case of the enzymatic treatment, these should focus on selecting the proper enzyme(s) or blend, and process optimization for operating under the optimal pH, temperature, and enzyme-to-substrate ratio that are defined by their kinetics. The length of the enzymatic treatment is also an important factor to be optimized to reach economic yields. It is also important to consider that chosen enzymes have high stability against the effects that these novel techniques exhibit during the processing. In the case of the novel co-extraction techniques, the operational parameters usually involved are the microwave and ultrasound power, temperatures, pressures, solvent-to-solid ratio, pH, and the extraction time. A target compound is usually determined by the nature of the solvent or solvent mixtures.
As a summary of the extraction technologies, Table 3 [211,212,213,214,215,216,217,218] gathers some studies linking the chemical compounds of fruit seeds and their respective bioactivity with the extracting technology used.

4. Final Comments and Upcoming Research Prospects

The potential use of fruit seeds for the recovery of phytochemicals is of pivotal importance within circular economy premises of production and utilization of natural resources. Bioactive-rich extracts may be used for either pharmaceutical or food sectors, and in some cases for both. Future research should focus on the optimization of innovative extraction techniques and on the biological effects of the bioactive-rich extracts using different in vitro and in vivo protocols. In addition, after checking to ensure that the extracts are safe from the chemical and toxicological standpoints, applications in cosmetics and food models should be conducted to assess the effectiveness of such extracts regarding their antioxidant, antimicrobial, anti-proliferative, antilipidemic, hypoglycemic, and anti-inflammatory effects.

Author Contributions

The review was conceived and designed by D.G. We declare that the data were collected, interpreted and analyzed by all authors named in this article. All authors read, made significant changes and approved the manuscript for publication.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Homrich, A.S.; Galvão, G.; Abadia, L.G.; Carvalho, M.M. The circular economy umbrella: Trends and gaps on integrating pathways. J. Clean. Prod. 2018, 175, 525–543. [Google Scholar] [CrossRef]
  2. Moraga, G.; Huysveld, S.; Mathieux, F.; Blengini, G.A.; Alaerts, L.; Van Acker, K.; de Meester, S.; Dewulf, J. Circular economy indicators: What do they measure? Resour. Conserv. Recycl. 2019, 146, 452–461. [Google Scholar] [CrossRef] [PubMed]
  3. Millar, N.; McLaughlin, E.; Börger, T. The Circular Economy: Swings and Roundabouts? Ecol. Econ. 2019, 158, 11–19. [Google Scholar] [CrossRef]
  4. Pauliuk, S. Critical appraisal of the circular economy standard BS 8001:2017 and a dashboard of quantitative system indicators for its implementation in organizations. Resour. Conserv. Recycl. 2018, 129, 81–92. [Google Scholar] [CrossRef]
  5. Schroeder, P.; Anggraeni, K.; Weber, U. The Relevance of Circular Economy Practices to the Sustainable Development Goals. J. Ind. Ecol. 2019, 23, 77–95. [Google Scholar] [CrossRef]
  6. Save Food Infographics: Fruits and Vegetables Food Losses|SAVE FOOD: Global Initiative on Food Loss and Waste Reduction|Food and Agriculture Organization of the United Nations. Available online: http://www.fao.org/save-food/resources/keyfindings/infographics/fruit/en/ (accessed on 4 October 2019).
  7. About the Sustainable Development Goals - United Nations Sustainable Development. Available online: https://www.un.org/sustainabledevelopment/sustainable-development-goals/ (accessed on 4 October 2019).
  8. Slorach, P.C.; Jeswani, H.K.; Cuéllar-Franca, R.; Azapagic, A. Environmental and economic implications of recovering resources from food waste in a circular economy. Sci. Total Environ. 2019, 693, 133516. [Google Scholar] [CrossRef]
  9. Velenturf, A.P.M.; Archer, S.A.; Gomes, H.I.; Christgen, B.; Lag-Brotons, A.J.; Purnell, P. Circular economy and the matter of integrated resources. Sci. Total Environ. 2019, 689, 963–969. [Google Scholar] [CrossRef]
  10. Rijswick, C. Rabobank Food and Agrobusiness. World Fruit Map. Rabobank. 2018. Available online: https://research.rabobank.com/far/en/sectors/regional-food-agri/world_fruit_map_2018.html (accessed on 4 October 2019).
  11. Bhushan, S.; Kalia, K.; Sharma, M.; Singh, B.; Ahuja, P.S. Processing of Apple Pomace for Bioactive Molecules. Crit. Rev. Biotechnol. 2008, 28, 285–296. [Google Scholar] [CrossRef]
  12. FAOSTAT. Available online: http://www.fao.org/faostat/en/#data/QC (accessed on 4 October 2019).
  13. Deng, G.F.; Shen, C.; Xu, X.R.; Kuang, R.D.; Guo, Y.J.; Zeng, L.S.; Gao, L.L.; Lin, X.; Xie, J.F.; Xia, E.Q.; et al. Potential of fruit wastes as natural resources of bioactive compounds. Int. J. Mol. Sci. 2012, 13, 8308–8323. [Google Scholar] [CrossRef]
  14. Melgar, B.; Dias, M.I.; Ciric, A.; Sokovic, M.; Garcia-Castello, E.M.; Rodriguez-Lopez, A.D.; Barros, L.; Ferreira, I.C.R.F. Bioactive characterization of Persea americana Mill. by-products: A rich source of inherent antioxidants. Ind. Crops Prod. 2018, 111, 212–218. [Google Scholar] [CrossRef]
  15. Fidelis, M.; Santos, J.S.; Escher, G.B.; Vieira do Carmo, M.; Azevedo, L.; Cristina da Silva, M.; Putnik, P.; Granato, D. In vitro antioxidant and antihypertensive compounds from camu-camu (Myrciaria dubia McVaugh, Myrtaceae) seed coat: A multivariate structure-activity study. Food Chem. Toxicol. 2018, 120, 479–490. [Google Scholar] [CrossRef] [PubMed]
  16. Majerska, J.; Michalska, A.; Figiel, A. A review of new directions in managing fruit and vegetable processing by-products. Trends Food Sci. Technol. 2019, 88, 207–219. [Google Scholar] [CrossRef]
  17. Naeem, A.; Shabbir, M.A.; Khan, M.R.; Ahmad, N.; Roberts, T.H. Mango Seed Kernel Fat as a Cocoa Butter Substitute Suitable for the Tropics. J. Food Sci. 2019, 84, 1315–1321. [Google Scholar] [CrossRef] [PubMed]
  18. Sagar, N.A.; Pareek, S.; Sharma, S.; Yahia, E.M.; Lobo, M.G. Fruit and Vegetable Waste: Bioactive Compounds, Their Extraction, and Possible Utilization. Compr. Rev. Food Sci. Food Saf. 2018, 17, 512–531. [Google Scholar] [CrossRef] [Green Version]
  19. Ayoub, M.; de Camargo, A.C.; Shahidi, F. Antioxidants and bioactivities of free, esterified and insoluble-bound phenolics from berry seed meals. Food Chem. 2016, 197, 221–232. [Google Scholar] [CrossRef]
  20. Karnopp, A.R.; Margraf, T.; Maciel, L.G.; Santos, J.S.; Granato, D. Chemical composition, nutritional and in vitro functional properties of by-products from the Brazilian organic grape juice industry. Int. Food Res. J. 2017, 24, 207–214. [Google Scholar]
  21. Tang, G.Y.; Zhao, C.N.; Liu, Q.; Feng, X.L.; Xu, X.Y.; Cao, S.Y.; Meng, X.; Li, S.; Gan, R.Y.; Li, H.B. Potential of grape wastes as a natural source of bioactive compounds. Molecules 2018, 23, 2598. [Google Scholar] [CrossRef]
  22. Karnopp, A.R.; Oliveira, K.G.; de Andrade, E.F.; Postingher, B.M.; Granato, D. Optimization of an organic yogurt based on sensorial, nutritional, and functional perspectives. Food Chem. 2017, 233, 401–411. [Google Scholar] [CrossRef]
  23. Karnopp, A.R.; Figueroa, A.M.; Los, P.R.; Teles, J.C.; Simões, D.R.S.; Barana, A.C.; Kubiaki, F.T.; de Oliveira, J.G.B.; Granato, D. Effects of whole-wheat flour and bordeaux grape pomace (Vitis labrusca L.) on the sensory, physicochemical and functional properties of cookies. Food Sci. Technol. 2015, 35, 750–756. [Google Scholar] [CrossRef]
  24. Sekhon-Loodu, S.; Warnakulasuriya, S.N.; Rupasinghe, H.P.V.; Shahidi, F. Antioxidant ability of fractionated apple peel phenolics to inhibit fish oil oxidation. Food Chem. 2013, 140, 189–196. [Google Scholar] [CrossRef]
  25. Santana, I.; Castelo-Branco, V.N.; Guimarães, B.M.; Silva, L.d.O.; Peixoto, V.O.D.S.; Cabral, L.M.C.; Freitas, S.P.; Torres, A.G. Hass avocado (Persea americana Mill.) oil enriched in phenolic compounds and tocopherols by expeller-pressing the unpeeled microwave dried fruit. Food Chem. 2019, 286, 354–361. [Google Scholar] [CrossRef] [PubMed]
  26. Terpou, A.; Papadaki, A.; Bosnea, L.; Kanellaki, M.; Kopsahelis, N. Novel frozen yogurt production fortified with sea buckthorn berries and probiotics. LWT 2019, 105, 242–249. [Google Scholar] [CrossRef]
  27. Powell, M.J.; Sebranek, J.G.; Prusa, K.J.; Tarté, R. Evaluation of citrus fiber as a natural replacer of sodium phosphate in alternatively-cured all-pork Bologna sausage. Meat Sci. 2019, 157, 107883. [Google Scholar] [CrossRef] [PubMed]
  28. Cecchi, L.; Innocenti, M.; Urciuoli, S.; Arlorio, M.; Paoli, P.; Mulinacci, N. In depth study of phenolic profile and PTP-1B inhibitory power of cold-pressed grape seed oils of different varieties. Food Chem. 2019, 271, 380–387. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, S.; Gu, B.-J.; Ganjyal, G.M. Impacts of the inclusion of various fruit pomace types on the expansion of corn starch extrudates. LWT 2019, 110, 223–230. [Google Scholar] [CrossRef]
  30. Ospina, M.; Montaña-Oviedo, K.; Díaz-Duque, Á.; Toloza-Daza, H.; Narváez-Cuenca, C.-E. Utilization of fruit pomace, overripe fruit, and bush pruning residues from Andes berry (Rubus glaucus Benth) as antioxidants in an oil in water emulsion. Food Chem. 2019, 281, 114–123. [Google Scholar] [CrossRef] [PubMed]
  31. Davis, L.; Jung, J.; Colonna, A.; Hasenbeck, A.; Gouw, V.; Zhao, Y. Quality and Consumer Acceptance of Berry Fruit Pomace-Fortified Specialty Mustard. J. Food Sci. 2018, 83, 1921–1932. [Google Scholar] [CrossRef] [PubMed]
  32. Gouw, V.P.; Jung, J.; Zhao, Y. Functional properties, bioactive compounds, and in vitro gastrointestinal digestion study of dried fruit pomace powders as functional food ingredients. LWT 2017, 80, 136–144. [Google Scholar] [CrossRef]
  33. Rtibi, K.; Selmi, S.; Saidani, K.; Grami, D.; Amri, M.; Sebai, H.; Marzouki, L. Reverse Effect of Opuntia ficus-indica L. Juice and Seeds Aqueous Extract on Gastric Emptying and Small-Bowel Motility in Rat. J. Food Sci. 2018, 83, 205–211. [Google Scholar] [CrossRef]
  34. Marques, T.R.; Cesar, P.H.S.; Braga, M.A.; Marcussi, S.; Corrêa, A.D. Fruit Bagasse Phytochemicals from Malpighia Emarginata Rich in Enzymatic Inhibitor with Modulatory Action on Hemostatic Processes. J. Food Sci. 2018, 83, 2840–2849. [Google Scholar] [CrossRef]
  35. View of Phenolic Content, Antioxidant and Anti-Inflammatory Activities of Seeds and Leaves of Date Palm (Phoenix dactylifera L.). Available online: http://www.isnff-jfb.com/index.php/JFB/article/view/72/137 (accessed on 3 October 2019).
  36. Gong, X.; Tang, M.; Gong, Z.; Qiu, Z.; Wang, D.; Fan, M. Screening pesticide residues on fruit peels using portable Raman spectrometer combined with adhesive tape sampling. Food Chem. 2019, 295, 254–258. [Google Scholar] [CrossRef] [PubMed]
  37. Sirisena, S.; Zabaras, D.; Ng, K.; Ajlouni, S. Characterization of Date (Deglet Nour) Seed Free and Bound Polyphenols by High-Performance Liquid Chromatography-Mass Spectrometry. J. Food Sci. 2017, 82, 333–340. [Google Scholar] [CrossRef] [PubMed]
  38. Takii, H.; Matsumoto, K.; Kometani, T.; Okada, S.; Fushiki, T. Lowering Effect of Phenolic Glycosides on the Rise in Postprandial Glucose in Mice. Biosci. Biotechnol. Biochem. 1997, 61, 1531–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Wong, Y.S.; Sia, C.M.; Khoo, H.E.; Ang, Y.K.; Chang, S.K.; Chang, S.K.; Yim, H.S. Influence of extraction conditions on antioxidant properties of passion fruit (Passiflora edulis) peel. Acta Sci. Pol. Technol. Aliment. 2014, 13, 257–265. [Google Scholar] [CrossRef]
  40. Dorta, E.; Lobo, M.G.; Gonzalez, M. Reutilization of Mango Byproducts: Study of the Effect of Extraction Solvent and Temperature on Their Antioxidant Properties. J. Food Sci. 2012, 77, C80–C88. [Google Scholar] [CrossRef]
  41. Durante, M.; Montefusco, A.; Marrese, P.P.; Soccio, M.; Pastore, D.; Piro, G.; Mita, G.; Lenucci, M.S. Seeds of pomegranate, tomato and grapes: An underestimated source of natural bioactive molecules and antioxidants from agri-food by-products. J. Food Compos. Anal. 2017, 63, 65–72. [Google Scholar] [CrossRef]
  42. Derakhshan, Z.; Ferrante, M.; Tadi, M.; Ansari, F.; Heydari, A.; Hosseini, M.S.; Conti, G.O.; Sadrabad, E.K. Antioxidant activity and total phenolic content of ethanolic extract of pomegranate peels, juice and seeds. Food Chem. Toxicol. 2018, 114, 108–111. [Google Scholar] [CrossRef]
  43. Van Nieuwenhove, C.P.; Moyano, A.; Castro-Gómez, P.; Fontecha, J.; Sáez, G.; Zárate, G.; Pizarro, P.L. Comparative study of pomegranate and jacaranda seeds as functional components for the conjugated linolenic acid enrichment of yogurt. LWT 2019, 111, 401–407. [Google Scholar] [CrossRef]
  44. Ambigaipalan, P.; de Camargo, A.C.; Shahidi, F. Identification of phenolic antioxidants and bioactives of pomegranate seeds following juice extraction using HPLC-DAD-ESI-MS n. Food Chem. 2017, 221, 1883–1894. [Google Scholar] [CrossRef]
  45. Yoshime, L.T.; de Melo, I.L.P.; Sattler, J.A.G.; Torres, R.P.; Mancini-Filho, J. Bioactive compounds and the antioxidant capacities of seed oils from pomegranate (Punica granatum L.) and bitter gourd (Momordica charantia L.). Food Sci. Technol. 2018. Ahead of Print. [Google Scholar] [CrossRef]
  46. Myoda, T.; Fujimura, S.; Park, B.J.; Nagashima, T.; Nakagawa, J.; Nishizawa, M. Antioxidative and antimicrobial potential of residues of camu-camu juice production. J. Food, Agric. Environ. 2010, 8, 304–307. [Google Scholar]
  47. Azevêdo, J.C.S.; Fujita, A.; de Oliveira, E.L.; Genovese, M.I.; Correia, R.T.P. Dried camu-camu (Myrciaria dubia H.B.K. McVaugh) industrial residue: A bioactive-rich Amazonian powder with functional attributes. Food Res. Int. 2014, 62, 934–940. [Google Scholar] [CrossRef]
  48. Maria, L.G.; Edvan, A.C.; Bala, R.; Pollyana, C.C.; Antonia, R.V.d.S.; Sara, T.M.S.; Railin, R.d.O. Qualitative evaluation and biocompounds present in different parts of camu-camu (Myrciaria dubia) fruit. Afr. J. Food Sci. 2017, 11, 124–129. [Google Scholar] [CrossRef]
  49. Do Carmo, M.A.V.D.; Fidelis, M.; Pressete, C.G.; Marques, M.J.; Castro-Gamero, A.M.; Myoda, T.; Granato, D.; Azevedo, L. Hydroalcoholic Myrciaria dubia (camu-camu) seed extracts prevent chromosome damage and act as antioxidant and cytotoxic agents. Food Res. Int. 2019, 125, 108551. [Google Scholar] [CrossRef]
  50. Xu, Y.; Fan, M.; Ran, J.; Zhang, T.; Sun, H.; Dong, M.; Zhang, Z.; Zheng, H. Variation in phenolic compounds and antioxidant activity in apple seeds of seven cultivars. Saudi J. Biol. Sci. 2016, 23, 379–388. [Google Scholar] [CrossRef]
  51. Alkhalf, M.I.; Alansari, W.S.; Ibrahim, E.A.; ELhalwagy, M.E.A. Anti-oxidant, anti-inflammatory and anti-cancer activities of avocado (Persea americana) fruit and seed extract. J. King Saud Univ. - Sci. 2018. [Google Scholar] [CrossRef]
  52. Fromm, M.; Bayha, S.; Carle, R.; Kammerer, D.R. Characterization and Quantitation of Low and High Molecular Weight Phenolic Compounds in Apple Seeds. J. Agric. Food Chem. 2012, 60, 1232–1242. [Google Scholar] [CrossRef]
  53. Paludo, M.; Colombo, R.; Teixeira, J.; Hermosín-Gutiérrez, I.; Ballus, C.; Godoy, H.; Paludo, M.C.; Colombo, R.C.; Teixeira Filho, J.; Hermosín-Gutiérrez, I.; et al. Optimizing the Extraction of Anthocyanins from the Skin and Phenolic Compounds from the Seed of Jabuticaba Fruits (Myrciaria jabuticaba (Vell.) O. Berg) with Ternary Mixture Experimental Designs. J. Braz. Chem. Soc. 2019, 30, 1506–1515. [Google Scholar] [CrossRef]
  54. Inada, K.O.P.; Oliveira, A.A.; Revorêdo, T.B.; Martins, A.B.N.; Lacerda, E.C.Q.; Freire, A.S.; Braz, B.F.; Santelli, R.E.; Torres, A.G.; Perrone, D.; et al. Screening of the chemical composition and occurring antioxidants in jabuticaba (Myrciaria jaboticaba) and jussara (Euterpe edulis) fruits and their fractions. J. Funct. Foods 2015, 17, 422–433. [Google Scholar] [CrossRef]
  55. Hacke, A.C.M.; Granato, D.; Maciel, L.G.; Weinert, P.L.; do Prado-Silva, L.; Alvarenga, V.O.; de Souza Sant’Ana, A.; Bataglion, G.A.; Eberlin, M.N.; Rosso, N.D. Jabuticaba (Myrciaria cauliflora) Seeds: Chemical Characterization and Extraction of Antioxidant and Antimicrobial Compounds. J. Food Sci. 2016, 81, C2206–C2217. [Google Scholar] [CrossRef]
  56. Alezandro, M.R.; Dubé, P.; Desjardins, Y.; Lajolo, F.M.; Genovese, M.I. Comparative study of chemical and phenolic compositions of two species of jaboticaba: Myrciaria jaboticaba (Vell.) Berg and Myrciaria cauliflora (Mart.) O. Berg. Food Res. Int. 2013, 54, 468–477. [Google Scholar] [CrossRef]
  57. Rupasinghe, H.P.V.; Kean, C. Polyphenol concentrations in apple processing by-products determined using electrospray ionization mass spectrometry. Can. J. Plant Sci. 2008, 88, 759–762. [Google Scholar] [CrossRef]
  58. Jham, G.N. High-performance liquid chromatographic quantitation of phloridzin in apple seed, leaf and callus. J. Chromatogr. A 1996, 719, 444–449. [Google Scholar] [CrossRef]
  59. Rosero, J.C.; Cruz, S.; Osorio, C.; Hurtado, N. Analysis of Phenolic Composition of Byproducts (Seeds and Peels) of Avocado (Persea americana Mill.) Cultivated in Colombia. Molecules 2019, 24, 3209. [Google Scholar] [CrossRef]
  60. Figueroa, J.G.; Borrás-Linares, I.; Lozano-Sánchez, J.; Segura-Carretero, A. Comprehensive characterization of phenolic and other polar compounds in the seed and seed coat of avocado by HPLC-DAD-ESI-QTOF-MS. Food Res. Int. 2018, 105, 752–763. [Google Scholar] [CrossRef]
  61. Athaydes, B.R.; Alves, G.M.; de Assis, A.L.E.M.; Gomes, J.V.D.; Rodrigues, R.P.; Campagnaro, B.P.; Nogueira, B.V.; Silveira, D.; Kuster, R.M.; Pereira, T.M.C.; et al. Avocado seeds (Persea americana Mill.) prevents indomethacin-induced gastric ulcer in mice. Food Res. Int. 2019, 119, 751–760. [Google Scholar] [CrossRef]
  62. Segovia, F.J.; Corral-Pérez, J.J.; Almajano, M.P. Avocado seed: Modeling extraction of bioactive compounds. Ind. Crops Prod. 2016. [Google Scholar] [CrossRef]
  63. Amariz, A.; de Lima, M.A.C.; Alves, R.E. Quality and antioxidant potential of byproducts from refining of fruit pulp. Food Sci. Technol. 2018, 38, 203–209. [Google Scholar] [CrossRef] [Green Version]
  64. Deolindo, C.T. P.; Monteiro, P.I.; Santos, J.S.; Cruz, A.G.; Cristina da Silva, M.; Granato, D. Phenolic-rich Petit Suisse cheese manufactured with organic Bordeaux grape juice, skin, and seed extract: Technological, sensory, and functional properties. LWT 2019, 115, 108493. [Google Scholar] [CrossRef]
  65. Harbeoui, H.; Hichami, A.; Wannes, W.A.; Lemput, J.; Tounsi, M.S.; Khan, N.A. Anti-inflammatory effect of grape (Vitis vinifera L.) seed extract through the downregulation of NF-κB and MAPK pathways in LPS-induced RAW264.7 macrophages. S. Afr. J. Bot. 2019, 125, 1–8. [Google Scholar] [CrossRef]
  66. De Albuquerque, M.A.C.; Levit, R.; Beres, C.; Bedani, R.; de Moreno de LeBlanc, A.; Saad, S.M.I.; LeBlanc, J.G. Tropical fruit by-products water extracts as sources of soluble fibres and phenolic compounds with potential antioxidant, anti-inflammatory, and functional properties. J. Funct. Foods 2019, 52, 724–733. [Google Scholar] [CrossRef]
  67. Saravanan, S.; Parimelazhagan, T. Total phenolic content, Free radical scavenging and Antimicrobial activities of Passiflora subpeltata seeds. J. Appl. Pharm. Sci. 2013, 3, 67–72. [Google Scholar]
  68. Meini, M.-R.; Cabezudo, I.; Boschetti, C.E.; Romanini, D. Recovery of phenolic antioxidants from Syrah grape pomace through the optimization of an enzymatic extraction process. Food Chem. 2019, 283, 257–264. [Google Scholar] [CrossRef] [PubMed]
  69. Oliveira, D.A.; Angonese, M.; Gomes, C.; Ferreira, S.R.S. Valorization of passion fruit (Passiflora edulis sp.) by-products: Sustainable recovery and biological activities. J. Supercrit. Fluids 2016, 111, 55–62. [Google Scholar] [CrossRef]
  70. Rotta, E.M.; Rodrigues, C.A.; Jardim, I.C.S.F.; Maldaner, L.; Visentainer, J.V. Determination of phenolic compounds and antioxidant activity in passion fruit pulp (Passiflora spp.) using a modified QuEChERS method and UHPLC-MS/MS. LWT 2019, 100, 397–403. [Google Scholar] [CrossRef]
  71. Schieber, A.; Hilt, P.; Streker, P.; Endreß, H.-U.; Rentschler, C.; Carle, R. A new process for the combined recovery of pectin and phenolic compounds from apple pomace. Innov. Food Sci. Emerg. Technol. 2003, 4, 99–107. [Google Scholar] [CrossRef]
  72. Lu, Y.; Yeap Foo, L. Constitution of some chemical components of apple seed. Food Chem. 1998, 61, 29–33. [Google Scholar] [CrossRef]
  73. Górnaś, P.; Rudzińska, M.; Soliven, A. Industrial by-products of plum Prunus domestica L. and Prunus cerasifera Ehrh. as potential biodiesel feedstock: Impact of variety. Ind. Crops Prod. 2017, 100, 77–84. [Google Scholar] [CrossRef]
  74. Skinner, R.; Warren, D.; Lateef, S.; Benedito, V.; Tou, J. Apple Pomace Consumption Favorably Alters Hepatic Lipid Metabolism in Young Female Sprague-Dawley Rats Fed a Western Diet. Nutrients 2018, 10, 1882. [Google Scholar] [CrossRef]
  75. Walia, M.; Rawat, K.; Bhushan, S.; Padwad, Y.S.; Singh, B. Fatty acid composition, physicochemical properties, antioxidant and cytotoxic activity of apple seed oil obtained from apple pomace. J. Sci. Food Agric. 2014, 94, 929–934. [Google Scholar] [CrossRef]
  76. Bada, J.C.; León-Camacho, M.; Copovi, P.; Alonso, L. Characterization of apple seed oil with Denomination of Origin from Asturias, Spain. Grasas y Aceites 2014, 65, e027. [Google Scholar] [Green Version]
  77. Matthäus, B.; Musazcan Özcan, M. Oil Content, Fatty Acid Composition and Distributions of Vitamin-E-Active Compounds of Some Fruit Seed Oils. Antioxidants 2015, 4, 124–133. [Google Scholar]
  78. Tian, H.-L.; Zhan, P.; Li, K.-X. Analysis of components and study on antioxidant and antimicrobial activities of oil in apple seeds. Int. J. Food Sci. Nutr. 2010, 61, 395–403. [Google Scholar] [CrossRef] [PubMed]
  79. Yu, X.; van de Voort, F.R.; Li, Z.; Yue, T. Proximate Composition of the Apple Seed and Characterization of Its Oil. Int. J. Food Eng. 2007, 3. [Google Scholar] [CrossRef]
  80. Górnaś, P. Unique variability of tocopherol composition in various seed oils recovered from by-products of apple industry: Rapid and simple determination of all four homologues (α, β, γ and δ) by RP-HPLC/FLD. Food Chem. 2015, 172, 129–134. [Google Scholar] [CrossRef]
  81. Opyd, P.; Jurgoński, A.; Juśkiewicz, J.; Milala, J.; Zduńczyk, Z.; Król, B. Nutritional and Health-Related Effects of a Diet Containing Apple Seed Meal in Rats: The Case of Amygdalin. Nutrients 2017, 9, 1091. [Google Scholar] [CrossRef]
  82. Gosch, C.; Halbwirth, H.; Stich, K. Phloridzin: Biosynthesis, distribution and physiological relevance in plants. Phytochemistry 2010, 71, 838–843. [Google Scholar] [CrossRef]
  83. Ehrenkranz, J.R.L.; Lewis, N.G.; Ronald Kahn, C.; Roth, J. Phlorizin: A review. Diabetes. Metab. Res. Rev. 2005, 21, 31–38. [Google Scholar] [CrossRef]
  84. Rossetti, L.; Smith, D.; Shulman, G.I.; Papachristou, D.; DeFronzo, R.A. Correction of hyperglycemia with phlorizin normalizes tissue sensitivity to insulin in diabetic rats. J. Clin. Investig. 1987, 79, 1510–1515. [Google Scholar] [CrossRef]
  85. Rossetti, L.; Shulman, G.I.; Zawalich, W.; DeFronzo, R.A. Effect of chronic hyperglycemia on in vivo insulin secretion in partially pancreatectomized rats. J. Clin. Investig. 1987, 80, 1037–1044. [Google Scholar] [CrossRef]
  86. Shin, S.-K.; Cho, S.-J.; Jung, U.; Ryu, R.; Choi, M.-S. Phlorizin Supplementation Attenuates Obesity, Inflammation, and Hyperglycemia in Diet-Induced Obese Mice Fed a High-Fat Diet. Nutrients 2016, 8, 92. [Google Scholar] [CrossRef] [PubMed]
  87. Bolarinwa, I.F.; Orfila, C.; Morgan, M.R.A. Amygdalin content of seeds, kernels and food products commercially-available in the UK. Food Chem. 2014, 152, 133–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Speijers, G. Cyanogenic glycosides. WHO JECFA Food Add. Ser. 1993, 30, 299–337. Available online: http://www.inchem.org/documents/jecfa/jecmono/v30je18.htm (accessed on 3 October 2019).
  89. Zhou, K.; Raffoul, J.J. Potential Anticancer Properties of Grape Antioxidants. J. Oncol. 2012, 2012, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Ye, J.; Chen, Q.; Tao, T.; Wang, G.; Xu, F. Promotive effects of 5-Aminolevulinic acid on growth, photosynthetic gas exchange, chlorophyll, and antioxidative enzymes under salinity stress in Prunnus persica (L.) Batseh Seedling. Emirates J. Food Agric. 2016, 28, 786–795. [Google Scholar] [CrossRef]
  91. The Plant List. 2013. Available online: http://www.theplantlist.org/ (accessed on 3 October 2019).
  92. Fiume, M.M.; Bergfeld, W.F.; Belsito, D. V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Vitis vinifera (Grape)-Derived Ingredients as Used in Cosmetics. Int. J. Toxicol. 2014, 33, 48S–83S. [Google Scholar] [CrossRef] [PubMed]
  93. Asensi, M.; Medina, I.; Ortega, A.; Carretero, J.; Baño, M.C.; Obrador, E.; Estrela, J.M. Inhibition of cancer growth by resveratrol is related to its low bioavailability. Free Radic. Biol. Med. 2002, 33, 387–398. [Google Scholar] [CrossRef]
  94. Hakimuddin, F.; Tiwari, K.; Paliyath, G.; Meckling, K. Grape and wine polyphenols down-regulate the expression of signal transduction genes and inhibit the growth of estrogen receptor–negative MDA-MB231 tumors in nu/nu mouse xenografts. Nutr. Res. 2008, 28, 702–713. [Google Scholar] [CrossRef] [PubMed]
  95. Kaur, M.; Velmurugan, B.; Rajamanickam, S.; Agarwal, R.; Agarwal, C. Gallic Acid, an Active Constituent of Grape Seed Extract, Exhibits Anti-proliferative, Pro-apoptotic and Anti-tumorigenic Effects Against Prostate Carcinoma Xenograft Growth in Nude Mice. Pharm. Res. 2009, 26, 2133–2140. [Google Scholar] [CrossRef] [Green Version]
  96. Kim, H.; Hall, P.; Smith, M.; Kirk, M.; Prasain, J.K.; Barnes, S.; Grubbs, C. Chemoprevention by Grape Seed Extract and Genistein in Carcinogen-induced Mammary Cancer in Rats Is Diet Dependent. J. Nutr. 2004, 134, 3445S–3452S. [Google Scholar] [CrossRef]
  97. Morré, D.M.; Morré, D.J. Anticancer activity of grape and grape skin extracts alone and combined with green tea infusions. Cancer Lett. 2006, 238, 202–209. [Google Scholar] [CrossRef]
  98. Singh, T.; Sharma, S.D.; Katiyar, S.K. Grape Proanthocyanidins Induce Apoptosis by Loss of Mitochondrial Membrane Potential of Human Non-Small Cell Lung Cancer Cells In vitro and In vivo. PLoS ONE 2011, 6, e27444. [Google Scholar] [CrossRef] [PubMed]
  99. Schlachterman, A.; Valle, F.; Wall, K.M.; Azios, N.G.; Castillo, L.; Morell, L.; Washington, A.V.; Cubano, L.A.; Dharmawardhane, S.F. Combined Resveratrol, Quercetin, and Catechin Treatment Reduces Breast Tumor Growth in a Nude Mouse Model. Transl. Oncol. 2008, 1, 19–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Singletary, K.W.; Stansbury, M.J.; Giusti, M.; van Breemen, R.B.; Wallig, M.; Rimando, A. Inhibition of Rat Mammary Tumorigenesis by Concord Grape Juice Constituents. J. Agric. Food Chem. 2003, 51, 7280–7286. [Google Scholar] [CrossRef] [PubMed]
  101. Yang, B.; Karlsson, R.M.; Oksman, P.H.; Kallio, H.P. Phytosterols in Sea Buckthorn (Hippophaë rhamnoides L.) Berries:  Identification and Effects of Different Origins and Harvesting Times. J. Agric. Food Chem. 2001, 49, 5620–5629. [Google Scholar] [CrossRef]
  102. Burin, V.M.; Ferreira-Lima, N.E.; Panceri, C.P.; Bordignon-Luiz, M.T. Bioactive compounds and antioxidant activity of Vitis vinifera and Vitis labrusca grapes: Evaluation of different extraction methods. Microchem. J. 2014, 114, 155–163. [Google Scholar] [CrossRef]
  103. Granato, D.; Koot, A.; Schnitzler, E.; van Ruth, S.M. Authentication of geographical origin and crop system of grape juices by phenolic compounds and antioxidant activity using chemometrics. J. Food Sci. 2015, 80, C584–C593. [Google Scholar] [CrossRef]
  104. Salehi, B.; Vlaisavljevic, S.; Adetunji, C.O.; Adetunji, J.B.; Kregiel, D.; Antolak, H.; Pawlikowska, E.; Uprety, Y.; Mileski, K.S.; Devkota, H.P.; et al. Plants of the genus Vitis: Phenolic compounds, anticancer properties and clinical relevance. Trends Food Sci. Technol. 2019, 91, 362–379. [Google Scholar] [CrossRef]
  105. Tkacz, K.; Wojdyło, A.; Nowicka, P.; Turkiewicz, I.; Golis, T. Characterization in vitro potency of biological active fractions of seeds, skins and flesh from selected Vitis vinifera L. cultivars and interspecific hybrids. J. Funct. Foods 2019, 56, 353–363. [Google Scholar] [CrossRef]
  106. Yilmaz, Y.; Göksel, Z.; Erdoğan, S.S.; Öztürk, A.; Atak, A.; Özer, C. Antioxidant Activity and Phenolic Content of Seed, Skin and Pulp Parts of 22 Grape (Vitis vinifera  L.) Cultivars (4 Common and 18 Registered or Candidate for Registration). J. Food Process. Preserv. 2015, 39, 1682–1691. [Google Scholar] [CrossRef]
  107. Fontana, A.R.; Antoniolli, A.; Bottini, R. Grape Pomace as a Sustainable Source of Bioactive Compounds: Extraction, Characterization, and Biotechnological Applications of Phenolics. J. Agric. Food Chem. 2013, 61, 8987–9003. [Google Scholar] [CrossRef]
  108. Yu, J.; Ahmedna, M. Functional components of grape pomace: Their composition, biological properties and potential applications. Int. J. Food Sci. Technol. 2013, 48, 221–237. [Google Scholar] [CrossRef]
  109. Brenes, A.; Viveros, A.; Chamorro, S.; Arija, I. Use of polyphenol-rich grape by-products in monogastric nutrition. A review. Anim. Feed Sci. Technol. 2016, 211, 1–17. [Google Scholar] [CrossRef]
  110. Bordiga, M.; Travaglia, F.; Locatelli, M.; Coïsson, J.D.; Arlorio, M. Characterisation of polymeric skin and seed proanthocyanidins during ripening in six Vitis vinifera L. cv. Food Chem. 2011, 127, 180–187. [Google Scholar] [CrossRef]
  111. Qiang, X.; YongLie, C.; QianBing, W. Health benefit application of functional oligosaccharides. Carbohydr. Polym. 2009, 77, 435–441. [Google Scholar] [CrossRef]
  112. Bordiga, M. Valorization of Wine Making By-Products; CRC Press, Taylor & Francis Group: Novara, Italy, 2015. [Google Scholar]
  113. Bordiga, M.; Meudec, E.; Williams, P.; Montella, R.; Travaglia, F.; Arlorio, M.; Coïsson, J.D.; Doco, T. The impact of distillation process on the chemical composition and potential prebiotic activity of different oligosaccharidic fractions extracted from grape seeds. Food Chem. 2019, 285, 423–430. [Google Scholar] [CrossRef]
  114. Nunes, M.A.; Pimentel, F.B.; Costa, A.S.G.; Alves, R.C.; Oliveira, M.B.P.P. Olive by-products for functional and food applications: Challenging opportunities to face environmental constraints. Innov. Food Sci. Emerg. Technol. 2016, 35, 139–148. [Google Scholar] [CrossRef]
  115. Andrade, M.A.; Lima, V.; Sanches Silva, A.; Vilarinho, F.; Castilho, M.C.; Khwaldia, K.; Ramos, F. Pomegranate and grape by-products and their active compounds: Are they a valuable source for food applications? Trends Food Sci. Technol. 2019, 86, 68–84. [Google Scholar] [CrossRef]
  116. Aziz, A.R.A.; AbouLaila, M.R.; Aziz, M.; Omar, M.A.; Sultan, K. In vitro and in vivo anthelmintic activity of pumpkin seeds and pomegranate peels extracts against Ascaridia galli. Beni-Suef Univ. J. Basic Appl. Sci. 2018, 7, 231–234. [Google Scholar] [CrossRef]
  117. Białek, A.; Stawarska, A.; Bodecka, J.; Białek, M.; Tokarz, A. Pomegranate seed oil influences the fatty acids profile and reduces the activity of desaturases in livers of Sprague-Dawley rats. Prostaglandins Other Lipid Mediat. 2017, 131, 9–16. [Google Scholar] [CrossRef]
  118. Granato, D.; Shahidi, F.; Wrolstad, R.; Kilmartin, P.; Melton, L.D.; Hidalgo, F.J.; Miyashita, K.; van Camp, J.; Alasalvar, C.; Ismail, A.B.; et al. Antioxidant activity, total phenolics and flavonoids contents: Should we ban in vitro screening methods? Food Chem. 2018, 264, 471–475. [Google Scholar] [CrossRef]
  119. Ferreira, L.M.; Sari, M.H.M.; Cervi, V.F.; Gehrcke, M.; Barbieri, A.V.; Zborowski, V.A.; Beck, R.C.R.; Nogueira, C.W.; Cruz, L. Pomegranate seed oil nanoemulsions improve the photostability and in vivo antinociceptive effect of a non-steroidal anti-inflammatory drug. Colloids Surf. B Biointerfaces 2016, 144, 214–221. [Google Scholar] [CrossRef] [PubMed]
  120. Zarepourfard, H.; Riasi, A.; Frouzanfar, M.; Hajian, M.; Nasr Esfahani, M.H. Pomegranate seed in diet, affects sperm parameters of cloned goats following freezing-thawing. Theriogenology 2019, 125, 203–209. [Google Scholar] [CrossRef] [PubMed]
  121. Akter, M.S.; Oh, S.; Eun, J.B.; Ahmed, M. Nutritional compositions and health promoting phytochemicals of camu-camu (Myrciaria dubia) fruit: A review. Food Res. Int. 2011, 44, 1728–1732. [Google Scholar] [CrossRef]
  122. Kaneshima, T.; Myoda, T.; Nakata, M.; Fujimori, T.; Toeda, K.; Nishizawa, M. Antioxidant activity of C-Glycosidic ellagitannins from the seeds and peel of camu-camu (Myrciaria dubia). LWT - Food Sci. Technol. 2016, 69, 76–81. [Google Scholar] [CrossRef]
  123. Camere-Colarossi, R.; Ulloa-Urizar, G.; Medina-Flores, D.; Caballero-García, S.; Mayta-Tovalino, F.; del Valle-Mendoza, J. Antibacterial activity of Myrciaria dubia (Camu camu) against Streptococcus mutans and Streptococcus sanguinis. Asian Pac. J. Trop. Biomed. 2016, 6, 740–744. [Google Scholar] [CrossRef]
  124. Yazawa, K.; Suga, K.; Honma, A.; Shirosaki, M.; Koyama, T. Anti-inflammatory effects of seeds of the tropical fruit camu-camu (Myrciaria dubia). J. Nutr. Sci. Vitaminol. (Tokyo) 2011, 57, 104–107. [Google Scholar] [CrossRef]
  125. Inoue, T.; Komoda, H.; Uchida, T.; Node, K. Tropical fruit camu-camu (Myrciaria dubia) has anti-oxidative and anti-inflammatory properties. J. Cardiol. 2008, 52, 127–132. [Google Scholar] [CrossRef]
  126. Schwertz, M.C.; Maia, J.R.P.; de Sousa, R.F.S.; Aguiar, J.P.L.; Yuyama, L.K.O.; Lima, E.S. Efeito hipolipidêmico do suco de camu-camu em ratos. Rev. Nutr. 2012, 25, 35–44. [Google Scholar] [CrossRef]
  127. Nascimento, O.V.; Boleti, A.P.A.; Yuyama, L.K.O.; Lima, E.S. Effects of diet supplementation with Camu-camu (Myrciaria dubia HBK McVaugh) fruit in a rat model of diet-induced obesity. An. Acad. Bras. Cienc. 2013, 85, 355–363. [Google Scholar] [CrossRef]
  128. Gonçalves, A.E.S.S.; Lellis-Santos, C.; Curi, R.; Lajolo, F.M.; Genovese, M.I. Frozen pulp extracts of camu-camu (Myrciaria dubia McVaugh) attenuate the hyperlipidemia and lipid peroxidation of Type 1 diabetic rats. Food Res. Int. 2014, 64, 1–8. [Google Scholar] [CrossRef]
  129. Castro-Vargas, H.I.; Rodríguez-Varela, L.I.; Ferreira, S.R.S.; Parada-Alfonso, F. Extraction of phenolic fraction from guava seeds (Psidium guajava L.) using supercritical carbon dioxide and co-solvents. J. Supercrit. Fluids 2010, 51, 319–324. [Google Scholar] [CrossRef]
  130. Chen, Z.-Y.; Jiao, R.; Ma, K.Y. Cholesterol-Lowering Nutraceuticals and Functional Foods. J. Agric. Food Chem. 2008, 56, 8761–8773. [Google Scholar] [CrossRef] [PubMed]
  131. Aggarwal, B.B.; Sundaram, C.; Prasad, S.; Kannappan, R. Tocotrienols, the vitamin E of the 21st century: Its potential against cancer and other chronic diseases. Biochem. Pharmacol. 2010, 80, 1613–1631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Antwi, S.O.; Steck, S.E.; Zhang, H.; Stumm, L.; Zhang, J.; Hurley, T.G.; Hebert, J.R. Plasma carotenoids and tocopherols in relation to prostate-specific antigen (PSA) levels among men with biochemical recurrence of prostate cancer. Cancer Epidemiol. 2015, 39, 752–762. [Google Scholar] [CrossRef] [Green Version]
  133. Moreda, W.; Pérez-Camino, M.; Cert, A. Gas and liquid chromatography of hydrocarbons in edible vegetable oils. J. Chromatogr. A 2001, 936, 159–171. [Google Scholar] [CrossRef]
  134. González-García, E.; Marina, M.L.; García, M.C. Plum (Prunus Domestica L.) by-product as a new and cheap source of bioactive peptides: Extraction method and peptides characterization. J. Funct. Foods 2014, 11, 428–437. [Google Scholar] [CrossRef]
  135. Xue, F.; Zhu, C.; Liu, F.; Wang, S.; Liu, H.; Li, C. Effects of high-intensity ultrasound treatment on functional properties of plum (Pruni domesticae semen) seed protein isolate. J. Sci. Food Agric. 2018, 98, 5690–5699. [Google Scholar] [CrossRef]
  136. González-García, E.; García, M.C.; Marina, M.L. Capillary liquid chromatography-ion trap-mass spectrometry methodology for the simultaneous quantification of four angiotensin-converting enzyme-inhibitory peptides in Prunus seed hydrolysates. J. Chromatogr. A 2018, 1540, 47–54. [Google Scholar] [CrossRef]
  137. Citadim, I.; Danner, M.A.; Sasso, S.A.Z. Rasileira de. Rev. Bras. Frutic. 2010, 32, 656. [Google Scholar]
  138. Jorge, N.; Luzia, D.M.M.; Bertanha, B.J. Antioxidant activity and profile fatty acids of jabuticaba seeds (Myrciaria cauliflora Berg). Acta Biol. Colomb. 2011, 16, 75–82. [Google Scholar]
  139. Baldin, J.C.; Munekata, P.E.S.; Michelin, E.C.; Polizer, Y.J.; Silva, P.M.; Canan, T.M.; Pires, M.A.; Godoy, S.H.S.; Fávaro-Trindade, C.S.; Lima, C.G.; et al. Effect of microencapsulated Jabuticaba (Myrciaria cauliflora) extract on quality and storage stability of mortadella sausage. Food Res. Int. 2018, 108, 551–557. [Google Scholar] [CrossRef] [PubMed]
  140. Lima, A.d.J.B.; Corrêa, A.D.; Dantas-Barros, A.M.; Nelson, D.L.; Amorim, A.C.L. Sugars, organic acids, minerals and lipids in jabuticaba. Rev. Bras. Frutic. 2011, 33, 540–550. [Google Scholar] [CrossRef] [Green Version]
  141. Batista, Â.G.; Lenquiste, S.A.; Moldenhauer, C.; Godoy, J.T.; Reis, S.M.P.M.; Maróstica Júnior, M.R. Jaboticaba (Myrciaria jaboticaba (Vell.) Berg.) peel improved triglycerides excretion and hepatic lipid peroxidation in high-fat-fed rats. Rev. Nutr. 2013, 26, 571–581. [Google Scholar] [CrossRef]
  142. Palozi, R.A.C.; Guarnier, L.P.; Romão, P.V.M.; Nocchi, S.R.; Dos Santos, C.C.; Lourenço, E.L.B.; Silva, D.B.; Gasparotto, F.M.; Gasparotto Junior, A. Pharmacological safety of Plinia cauliflora (Mart.) Kausel in rabbits. Toxicol. Rep. 2019, 6, 616–624. [Google Scholar] [CrossRef] [PubMed]
  143. Dabas, D.; Shegog, R.; Ziegler, G.; Lambert, J. Avocado (Persea americana) Seed as a Source of Bioactive Phytochemicals. Curr. Pharm. Des. 2013, 19, 6133–6140. [Google Scholar] [CrossRef] [PubMed]
  144. Kosińska, A.; Karamać, M.; Estrella, I.; Hernández, T.; Bartolomé, B.; Dykes, G.A. Phenolic Compound Profiles and Antioxidant Capacity of Persea americana Mill. Peels and Seeds of Two Varieties. J. Agric. Food Chem. 2012, 60, 4613–4619. [Google Scholar] [CrossRef] [PubMed]
  145. Uchenna, U.E.; Shori, A.B.; Baba, A.S. Inclusion of avocado (Persea americana) seeds in the diet to improve carbohydrate and lipid metabolism in rats. Rev. Argent. Endocrinol. Metab. 2017, 54, 140–148. [Google Scholar] [CrossRef]
  146. Hatzakis, E.; Mazzola, E.P.; Shegog, R.M.; Ziegler, G.R.; Lambert, J.D. Perseorangin: A natural pigment from avocado (Persea americana) seed. Food Chem. 2019, 293, 15–22. [Google Scholar] [CrossRef]
  147. Dembitsky, V.M.; Poovarodom, S.; Leontowicz, H.; Leontowicz, M.; Vearasilp, S.; Trakhtenberg, S.; Gorinstein, S. The multiple nutrition properties of some exotic fruits: Biological activity and active metabolites. Food Res. Int. 2011, 44, 1671–1701. [Google Scholar] [CrossRef]
  148. Shanmugam, S.; Thangaraj, P.; Lima, B.d.S.; Chandran, R.; de Souza Araújo, A.A.; Narain, N.; Serafini, M.R.; Júnior, L.J.Q. Effects of luteolin and quercetin 3-β- d -glucoside identified from Passiflora subpeltata leaves against acetaminophen induced hepatotoxicity in rats. Biomed. Pharmacother. 2016, 83, 1278–1285. [Google Scholar] [CrossRef]
  149. Shanmugam, S.; Gomes, I.A.; Denadai, M.; dos Santos Lima, B.; de Souza Araújo, A.A.; Narain, N.; Neta, M.T.S.L.; Serafini, M.R.; Quintans-Júnior, L.J.; Thangaraj, P. UHPLC-QqQ-MS/MS identification, quantification of polyphenols from Passiflora subpeltata fruit pulp and determination of nutritional, antioxidant, α-amylase and α-glucosidase key enzymes inhibition properties. Food Res. Int. 2018, 108, 611–620. [Google Scholar] [CrossRef] [PubMed]
  150. Jiménez, A.M.; Sierra, C.A.; Rodríguez-Pulido, F.J.; González-Miret, M.L.; Heredia, F.J.; Osorio, C. Physicochemical characterisation of gulupa (Passiflora edulis Sims. fo edulis) fruit from Colombia during the ripening. Food Res. Int. 2011, 44, 1912–1918. [Google Scholar] [CrossRef]
  151. Zhang, B.; Deng, Z.; Ramdath, D.D.; Tang, Y.; Chen, P.X.; Liu, R.; Liu, Q.; Tsao, R. Phenolic profiles of 20 Canadian lentil cultivars and their contribution to antioxidant activity and inhibitory effects on α-glucosidase and pancreatic lipase. Food Chem. 2015, 172, 862–872. [Google Scholar] [CrossRef]
  152. Gazola, A.C.; Costa, G.M.; Castellanos, L.; Ramos, F.A.; Reginatto, F.H.; de Lima, T.C.M.; Schenkel, E.P.; Gazola, A.C.; Costa, G.M.; Castellanos, L.; et al. Involvement of GABAergic pathway in the sedative activity of apigenin, the main flavonoid from Passiflora quadrangularis pericarp. Rev. Bras. Farmacogn. 2015, 25, 158–163. [Google Scholar] [CrossRef]
  153. Shuayprom, A.; Sanguansermsri, D.; Sanguansermsri, P.; Fraser, I.H.; Wongkattiya, N. Quantitative determination of vitexin in Passiflora foetida Linn. leaves using HPTLC. Asian Pac. J. Trop. Biomed. 2016, 6, 216–220. [Google Scholar] [CrossRef] [Green Version]
  154. Saito, S.; Kawabata, J. Effects of electron-withdrawing substituents on DPPH radical scavenging reactions of protocatechuic acid and its analogues in alcoholic solvents. Tetrahedron 2005, 61, 8101–8108. [Google Scholar] [CrossRef]
  155. Lam, S.K.; Ng, T.B. Passiflin, a novel dimeric antifungal protein from seeds of the passion fruit. Phytomedicine 2009, 16, 172–180. [Google Scholar] [CrossRef]
  156. Lourith, N.; Kanlayavattanakul, M. Antioxidant Activities and Phenolics of Passiflora edulis Seed Recovered from Juice Production Residue. J. Oleo Sci. 2013, 62, 235–240. [Google Scholar] [CrossRef]
  157. Malacrida, C.R.; Jorge, N. Yellow passion fruit seed oil (Passiflora edulis f. flavicarpa): Physical and chemical characteristics. Braz. Arch. Biol. Technol. 2012, 55, 127–134. [Google Scholar] [CrossRef]
  158. Macagnan, F.T.; dos Santos, L.R.; Roberto, B.S.; de Moura, F.A.; Bizzani, M.; da Silva, L.P. Biological properties of apple pomace, orange bagasse and passion fruit peel as alternative sources of dietary fibre. Bioact. Carbohydr. Diet. Fibre 2015, 6, 1–6. [Google Scholar] [CrossRef]
  159. Helbig, D.; Böhm, V.; Wagner, A.; Schubert, R.; Jahreis, G. Berry seed press residues and their valuable ingredients with special regard to black currant seed press residues. Food Chem. 2008, 111, 1043–1049. [Google Scholar] [CrossRef]
  160. Pieszka, M.; Tombarkiewicz, B.; Roman, A.; Migdał, W.; Niedziółka, J. Effect of bioactive substances found in rapeseed, raspberry and strawberry seed oils on blood lipid profile and selected parameters of oxidative status in rats. Environ. Toxicol. Pharmacol. 2013, 36, 1055–1062. [Google Scholar] [CrossRef] [PubMed]
  161. Michel, T.; Destandau, E.; Le Floch, G.; Lucchesi, M.E.; Elfakir, C. Antimicrobial, antioxidant and phytochemical investigations of sea buckthorn (Hippophaë rhamnoides L.) leaf, stem, root and seed. Food Chem. 2012, 131, 754–760. [Google Scholar] [CrossRef]
  162. Sharma, U.K.; Sharma, K.; Sharma, N.; Sharma, A.; Singh, H.P.; Sinha, A.K. Microwave-Assisted Efficient Extraction of Different Parts of Hippophae rhamnoides for the Comparative Evaluation of Antioxidant Activity and Quantification of Its Phenolic Constituents by Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) . J. Agric. Food Chem. 2008, 56, 374–379. [Google Scholar] [CrossRef] [PubMed]
  163. Yang, B.; Ahotupa, M.; Määttä, P.; Kallio, H. Composition and antioxidative activities of supercritical CO2-extracted oils from seeds and soft parts of northern berries. Food Res. Int. 2011, 44, 2009–2017. [Google Scholar] [CrossRef]
  164. Gustinelli, G.; Eliasson, L.; Svelander, C.; Andlid, T.; Lundin, L.; Ahrné, L.; Alminger, M. Supercritical Fluid Extraction of Berry Seeds: Chemical Composition and Antioxidant Activity. J. Food Qual. 2018, 2018, 1–10. [Google Scholar] [CrossRef]
  165. Fazio, A.; Plastina, P.; Meijerink, J.; Witkamp, R.F.; Gabriele, B. Comparative analyses of seeds of wild fruits of Rubus and Sambucus species from Southern Italy: Fatty acid composition of the oil, total phenolic content, antioxidant and anti-inflammatory properties of the methanolic extracts. Food Chem. 2013, 140, 817–824. [Google Scholar] [CrossRef]
  166. Gođevac, D.; Tešević, V.; Vajs, V.; Milosavljević, S.; Stanković, M. Blackberry Seed Extracts and Isolated Polyphenolic Compounds Showing Protective Effect on Human Lymphocytes DNA. J. Food Sci. 2011, 76, C1039–C1043. [Google Scholar] [CrossRef]
  167. Van Hoed, V.; Barbouche, I.; De Clercq, N.; Dewettinck, K.; Slah, M.; Leber, E.; Verhé, R. Influence of filtering of cold pressed berry seed oils on their antioxidant profile and quality characteristics. Food Chem. 2011, 127, 1848–1855. [Google Scholar] [CrossRef]
  168. Parry, J.; Su, L.; Moore, J.; Cheng, Z.; Luther, M.; Rao, J.N.; Wang, J.-Y.; Yu, L.L. Chemical Compositions, Antioxidant Capacities, and Antiproliferative Activities of Selected Fruit Seed Flours. J. Agric. Food Chem. 2006, 54, 3773–3778. [Google Scholar] [CrossRef]
  169. Puupponen-Pimiä, R.; Nohynek, L.; Hartmann-Schmidlin, S.; Kähkönen, M.; Heinonen, M.; Määttä-Riihinen, K.; Oksman-Caldentey, K.M. Berry phenolics selectively inhibit the growth of intestinal pathogens. J. Appl. Microbiol. 2005, 98, 991–1000. [Google Scholar] [CrossRef] [PubMed]
  170. Bakowska-Barczak, A.M.; Schieber, A.; Kolodziejczyk, P. Characterization of Canadian Black Currant (Ribes nigrum L.) Seed Oils and Residues. J. Agric. Food Chem. 2009, 57, 11528–11536. [Google Scholar] [CrossRef] [PubMed]
  171. Puupponen-Pimiä, R.; Nohynek, L.; Juvonen, R.; Kössö, T.; Truchado, P.; Westerlund-Wikström, B.; Leppänen, T.; Moilanen, E.; Oksman-Caldentey, K.-M. Fermentation and dry fractionation increase bioactivity of cloudberry (Rubus chamaemorus). Food Chem. 2016, 197, 950–958. [Google Scholar] [CrossRef] [PubMed]
  172. Hager, T.J.; Howard, L.R.; Liyanage, R.; Lay, J.O.; Prior, R.L. Ellagitannin Composition of Blackberry As Determined by HPLC-ESI-MS and MALDI-TOF-MS. J. Agric. Food Chem. 2008, 56, 661–669. [Google Scholar] [CrossRef] [PubMed]
  173. Smeds, A.I.; Eklund, P.C.; Willför, S.M. Content, composition, and stereochemical characterisation of lignans in berries and seeds. Food Chem. 2012, 134, 1991–1998. [Google Scholar] [CrossRef] [PubMed]
  174. Olas, B. Sea buckthorn as a source of important bioactive compounds in cardiovascular diseases. Food Chem. Toxicol. 2016, 97, 199–204. [Google Scholar] [CrossRef]
  175. Bal, L.M.; Meda, V.; Naik, S.N.; Satya, S. Sea buckthorn berries: A potential source of valuable nutrients for nutraceuticals and cosmoceuticals. Food Res. Int. 2011, 44, 1718–1727. [Google Scholar] [CrossRef]
  176. Basu, M.; Prasad, R.; Jayamurthy, P.; Pal, K.; Arumughan, C.; Sawhney, R.C. Anti-atherogenic effects of seabuckthorn (Hippophaea rhamnoides) seed oil. Phytomedicine 2007, 14, 770–777. [Google Scholar] [CrossRef]
  177. Yang, B.; Kortesniemi, M. Clinical evidence on potential health benefits of berries. Curr. Opin. Food Sci. 2015, 2, 36–42. [Google Scholar] [CrossRef]
  178. Yang, B.; Kalimo, K.O.; Mattila, L.M.; Kallio, S.E.; Katajisto, J.K.; Peltola, O.J.; Kallio, H.P. Effects of dietary supplementation with sea buckthorn (Hippophaë rhamnoides) seed and pulp oils on atopic dermatitis. J. Nutr. Biochem. 1999, 10, 622–630. [Google Scholar] [CrossRef]
  179. Larmo, P.S.; Kangas, A.J.; Soininen, P.; Lehtonen, H.-M.; Suomela, J.-P.; Yang, B.; Viikari, J.; Ala-Korpela, M.; Kallio, H.P. Effects of sea buckthorn and bilberry on serum metabolites differ according to baseline metabolic profiles in overweight women: A randomized crossover trial. Am. J. Clin. Nutr. 2013, 98, 941–951. [Google Scholar] [CrossRef] [PubMed]
  180. Johansson, A.K.; Korte, H.; Yang, B.; Stanley, J.C.; Kallio, H.P. Sea buckthorn berry oil inhibits platelet aggregation. J. Nutr. Biochem. 2000, 11, 491–495. [Google Scholar] [CrossRef]
  181. Yang, B.; Kallio, H. Composition and physiological effects of sea buckthorn (Hippophaë) lipids. Trends Food Sci. Technol. 2002, 13, 160–167. [Google Scholar] [CrossRef]
  182. Linnamaa, P.; Nieminen, K.; Koulu, L.; Tuomasjukka, S.; Kallio, H.; Yang, B.; Tahvonen, R.; Savolainen, J. Black currant seed oil supplementation of mothers enhances IFN-γ and suppresses IL-4 production in breast milk. Pediatr. Allergy Immunol. 2013, 24, 562–566. [Google Scholar] [CrossRef] [PubMed]
  183. Nayak, A.; Bhushan, B. An overview of the recent trends on the waste valorization techniques for food wastes. J. Environ. Manag. 2019, 233, 352–370. [Google Scholar] [CrossRef]
  184. Marathe, S.J.; Jadhav, S.B.; Bankar, S.B.; Kumari Dubey, K.; Singhal, R.S. Improvements in the extraction of bioactive compounds by enzymes. Curr. Opin. Food Sci. 2019, 25, 62–72. [Google Scholar] [CrossRef] [Green Version]
  185. Fernández, K.; Vega, M.; Aspé, E. An enzymatic extraction of proanthocyanidins from País grape seeds and skins. Food Chem. 2015, 168, 7–13. [Google Scholar] [CrossRef]
  186. Gligor, O.; Mocan, A.; Moldovan, C.; Locatelli, M.; Crișan, G.; Ferreira, I.C.F.R. Enzyme-assisted extractions of polyphenols—A comprehensive review. Trends Food Sci. Technol. 2019, 88, 302–315. [Google Scholar] [CrossRef]
  187. Nadar, S.S.; Rao, P.; Rathod, V.K. Enzyme assisted extraction of biomolecules as an approach to novel extraction technology: A review. Food Res. Int. 2018, 108, 309–330. [Google Scholar] [CrossRef]
  188. Pap, N.; Mahosenaho, M.; Pongrácz, E.; Mikkonen, H.; Jaakkola, M.; Virtanen, V.; Myllykoski, L.; Horváth-Hovorka, Z.; Hodúr, C.; Vatai, G.; et al. Effect of Ultrafiltration on Anthocyanin and Flavonol Content of Black Currant Juice (Ribes nigrum L.). Food Bioprocess Technol. 2012, 5, 921–928. [Google Scholar] [CrossRef]
  189. Fernandes, P.; Carvalho, F. Microbial enzymes for the food industry. In Biotechnology of Microbial Enzymes: Production, Biocatalysis and Industrial Applications, 1st ed.; Brahmachari, G., Ed.; Academic Press: Cambridge, MA, USA, 2017; Volume 19, pp. 513–544. [Google Scholar]
  190. Nadar, S.S.; Rathod, V.K. Sonochemical Effect on Activity and Conformation of Commercial Lipases. Appl. Biochem. Biotechnol. 2017, 181, 1435–1453. [Google Scholar] [CrossRef] [PubMed]
  191. Roselló-Soto, E.; Galanakis, C.M.; Brnčić, M.; Orlien, V.; Trujillo, F.J.; Mawson, R.; Knoerzer, K.; Tiwari, B.K.; Barba, F.J. Clean recovery of antioxidant compounds from plant foods, by-products and algae assisted by ultrasounds processing. Modeling approaches to optimize processing conditions. Trends Food Sci. Technol. 2015, 42, 134–149. [Google Scholar] [CrossRef]
  192. Da Porto, C.; Porretto, E.; Decorti, D. Comparison of ultrasound-assisted extraction with conventional extraction methods of oil and polyphenols from grape (Vitis vinifera L.) seeds. Ultrason. Sonochem. 2013, 20, 1076–1080. [Google Scholar] [CrossRef] [PubMed]
  193. Romdhane, M. Investigation in solid–liquid extraction: Influence of ultrasound. Chem. Eng. J. 2002, 87, 11–19. [Google Scholar] [CrossRef]
  194. Barba, F.J.; Zhu, Z.; Koubaa, M.; Sant’Ana, A.S.; Orlien, V. Green alternative methods for the extraction of antioxidant bioactive compounds from winery wastes and by-products: A review. Trends Food Sci. Technol. 2016, 49, 96–109. [Google Scholar] [CrossRef]
  195. Li, Y.; Skouroumounis, G.K.; Elsey, G.M.; Taylor, D.K. Microwave-assistance provides very rapid and efficient extraction of grape seed polyphenols. Food Chem. 2011, 129, 570–576. [Google Scholar] [CrossRef]
  196. Bucić-Kojić, A.; Sovová, H.; Planinić, M.; Tomas, S. Temperature-dependent kinetics of grape seed phenolic compounds extraction: Experiment and model. Food Chem. 2013, 136, 1136–1140. [Google Scholar] [CrossRef]
  197. Herrero, M.; Castro-Puyana, M.; Rocamora-Reverte, L.; Ferragut, J.A.; Cifuentes, A.; Ibáñez, E. Formation and relevance of 5-hydroxymethylfurfural in bioactive subcritical water extracts from olive leaves. Food Res. Int. 2012, 47, 31–37. [Google Scholar] [CrossRef] [Green Version]
  198. Duba, K.S.; Casazza, A.A.; Mohamed, H.B.; Perego, P.; Fiori, L. Extraction of polyphenols from grape skins and defatted grape seeds using subcritical water: Experiments and modeling. Food Bioprod. Process. 2015, 94, 29–38. [Google Scholar] [CrossRef]
  199. Plaza, M.; Marina, M.L. Pressurized hot water extraction of bioactives. TrAC Trends Anal. Chem. 2019, 116, 236–247. [Google Scholar] [CrossRef]
  200. Aliakbarian, B.; Fathi, A.; Perego, P.; Dehghani, F. Extraction of antioxidants from winery wastes using subcritical water. J. Supercrit. Fluids 2012, 65, 18–24. [Google Scholar] [CrossRef]
  201. He, L.; Zhang, X.; Xu, H.; Xu, C.; Yuan, F.; Knez, Ž.; Novak, Z.; Gao, Y. Subcritical water extraction of phenolic compounds from pomegranate (Punica granatum L.) seed residues and investigation into their antioxidant activities with HPLC–ABTS+ assay. Food Bioprod. Process. 2012, 90, 215–223. [Google Scholar] [CrossRef]
  202. Monrad, J.K.; Suárez, M.; Motilva, M.J.; King, J.W.; Srinivas, K.; Howard, L.R. Extraction of anthocyanins and flavan-3-ols from red grape pomace continuously by coupling hot water extraction with a modified expeller. Food Res. Int. 2014, 65, 77–87. [Google Scholar] [CrossRef]
  203. Yilmaz, E.E.; Özvural, E.B.; Vural, H. Extraction and identification of proanthocyanidins from grape seed (Vitis Vinifera) using supercritical carbon dioxide. J. Supercrit. Fluids 2011, 55, 924–928. [Google Scholar] [CrossRef]
  204. Berna, A.; Cháfer, A.; Montón, J.; Subirats, S. High-pressure solubility data of system ethanol (1)+catechin (2)+CO2 (3). J. Supercrit. Fluids 2001, 20, 157–162. [Google Scholar] [CrossRef]
  205. Da Porto, C.; Natolino, A. Supercritical fluid extraction of polyphenols from grape seed (Vitis vinifera): Study on process variables and kinetics. J. Supercrit. Fluids 2017, 130, 239–245. [Google Scholar] [CrossRef]
  206. Ran, L.; Yang, C.; Xu, M.; Yi, Z.; Ren, D.; Yi, L. Enhanced aqueous two-phase extraction of proanthocyanidins from grape seeds by using ionic liquids as adjuvants. Sep. Purif. Technol. 2019, 226, 154–161. [Google Scholar] [CrossRef]
  207. Martins, P.L.G.; Braga, A.R.; de Rosso, V.V. Can ionic liquid solvents be applied in the food industry? Trends Food Sci. Technol. 2017, 66, 117–124. [Google Scholar] [CrossRef]
  208. Shi, J.; Nawaz, H.; Pohorly, J.; Mittal, G.; Kakuda, Y.; Jiang, Y. Extraction of Polyphenolics from Plant Material for Functional Foods—Engineering and Technology. Food Rev. Int. 2005, 21, 139–166. [Google Scholar] [CrossRef]
  209. Pap, N.; Pongrácz, E.; Jaakkola, M.; Tolonen, T.; Virtanen, V.; Turkki, A.; Horváth-Hovorka, Z.; Vatai, G.; Keiski, R.L. The effect of pre-treatment on the anthocyanin and flavonol content of black currant juice (Ribes nigrum L.) in concentration by reverse osmosis. J. Food Eng. 2010, 98, 429–436. [Google Scholar] [CrossRef]
  210. Conidi, C.; Cassano, A.; Caiazzo, F.; Drioli, E. Separation and purification of phenolic compounds from pomegranate juice by ultrafiltration and nanofiltration membranes. J. Food Eng. 2017, 195, 1–13. [Google Scholar] [CrossRef]
  211. Du, G.; Zhu, Y.; Wang, X.; Zhang, J.; Tian, C.; Liu, L.; Meng, Y.; Guo, Y. Phenolic composition of apple products and by-products based on cold pressing technology. J. Food Sci. Technol. 2019, 56, 1389–1397. [Google Scholar] [CrossRef] [PubMed]
  212. Nile, S.H.; Nile, A.; Liu, J.; Kim, D.H.; Kai, G. Exploitation of apple pomace towards extraction of triterpenic acids, antioxidant potential, cytotoxic effects, and inhibition of clinically important enzymes. Food Chem. Toxicol. 2019, 131, 110563. [Google Scholar] [CrossRef] [PubMed]
  213. Gunes, R.; Palabiyik, I.; Toker, O.S.; Konar, N.; Kurultay, S. Incorporation of defatted apple seeds in chewing gum system and phloridzin dissolution kinetics. J. Food Eng. 2019, 255, 9–14. [Google Scholar] [CrossRef]
  214. Fernandes, P.A.R.; Ferreira, S.S.; Bastos, R.; Ferreira, I.; Cruz, M.T.; Pinto, A.; Coelho, E.; Passos, C.P.; Coimbra, M.A.; Cardoso, S.M.; et al. Apple Pomace Extract as a Sustainable Food Ingredient. Antioxidants 2019, 8, 189. [Google Scholar] [CrossRef]
  215. Rodríguez Madrera, R.; Suárez Valles, B. Characterization of apple seeds and their oils from the cider-making industry. Eur. Food Res. Technol. 2018, 244, 1821–1827. [Google Scholar] [CrossRef]
  216. Chandrasekar, V.; Martín-González, M.F.S.; Hirst, P.; Ballard, T.S. Optimizing Microwave-Assisted Extraction of Phenolic Antioxidants from Red Delicious and Jonathan Apple Pomace. J. Food Process Eng. 2015, 38, 571–582. [Google Scholar] [CrossRef]
  217. Zhang, Z.; Poojary, M.M.; Choudhary, A.; Rai, D.K.; Tiwari, B.K. Comparison of selected clean and green extraction technologies for biomolecules from apple pomace. Electrophoresis 2018, 39, 1934–1945. [Google Scholar] [CrossRef]
  218. Lohani, U.C.; Muthukumarappan, K. Application of the pulsed electric field to release bound phenolics in sorghum flour and apple pomace. Innov. Food Sci. Emerg. Technol. 2016, 35, 29–35. [Google Scholar] [CrossRef]
Table 1. Chemical compounds and bioactivity of different fruit seeds.
Table 1. Chemical compounds and bioactivity of different fruit seeds.
Fruit SeedExtracting SolventTotal Phenolic ContentIndividual CompoundsBioactivityReference
Avocado
(Persea americana)
Methanol/chloroformnot determinedMyristic acid
Myristoleic
Palmitic acid
Palmitoleic acid
Stearic acid
Oleic acid
Linoleic acid
α-Linolenic acid
Eicosadienoic acid
Eicosatrienoic acid
DPPH radical scavenging activity

ABTS scavenging assay

Anti-inflammatory activity


Anticancer activity
(HepG2 and HCT116)
[51]
Hydroalcoholic 70%
(ethanol)
366.79 ± 5.05 mg GAE/gPalmitic acid
Linoleic acid
Oleic acid
Stearic acid
p-Hydroxycoumaroyl quinic acid
Caffeoylquinic acid
Feruloylquinic acid
Gastroprotective activity[61]
Ethanol-water
(1:1, v/v)
not determinedVanillic acid
3-Feruloylquinic acid
5-O-caffeoylquinic acid
Procyanidin dimer B7
Procyanidin pentamer B1
Procyanidin trimer A4
4-O-caffeoylquinic acid
Procyanidin trimer B2
Procyanidin tetramer B1
Procyanidin dimer B8
Procyanidin pentamer B2
Procyanidin trimer A5
Benzoic acid
Procyanidin dimer B9
p-Coumaric acid
Procyanidin dimer A1
(−)-Epicatechin
(Epi)catechin gallate
5-p-Coumaroylquinic acid
Vanillin
4-p-Coumaroylquinic acid Pyrocatechol
Procyanidin trimer A1
Procyanidin dimer B3
Syringic acid
Procyanidin dimer B4
Procyanidin trimer B1
Tyrosol-glucoside
Ferulic acid
Procyanidin trimer A2
(Epi)catechin glucopyranoside (isomer 2)
4-Hydroxybenzoic acid
Procyanidin tetramer A4
Procyanidin dimer B5
(+)-Catechin
Hydroxyferulic acid
3-p-Coumaroylquinic acid
Gentisic acid
Procyanidin trimer A3
Tyrosol-glucosyl-pentoside
Procyanidin tetramer A5
Penstemide
Procyanidin dimer B Procyanidin dimer B10
Procyanidin trimer A6
Dihydrocaffeic acid
Procyanidin dimer A2
Cinchonain (isomer 1)
Caffeic acid
4-Feruloylquinic acid
Procyanidin dimer A3
Procyanidin trimer A7
Procyanidin dimer A4
Procyanidin dimer B11
Quercetin-diglucoside (isomer 1)
Quercetin-diglucoside (isomer 2)
Procyanidin dimer A5
Hydroxyabscisic acid glucoside
(Epi)gallocatechin
Quercetin-3-β-glucoside
Ethyl Protocatechuate
Cinchonain (isomer 2)
Quercetin
(±)-Naringenin
Sakuranetin
Trolox equivalent
antioxidant capacity (TEAC)
ABTS scavenging assay
DPPH radical scavenging activity
[60]
Aqueous methanol 80%
and
aqueous acetone 70%
328.8 ± 13.5 mg GAE/gCaffeic acid
(−)-Epicatechin
Vanillin
p-Coumaric acid
Ferulic acid
Sinapic acid
Procyanidin B4
Quercetin diglucoside
Quercetin-3-O-arabinosyl-glucoside
Quercetin-3-O-glucoside
Quercetin-3-O-rutinoside
Quinic acid
Procyanidin dimer A
Procyanidin trimer B1
Procyanidin dimer B1
Procyanidin trimer B2 Syringic acid
Procyanidin dimer B2
(+)-Catechin
Procyanidin trimer A
Procyanidin dimer B2
Procyanidin trimer B3
5-O-Caffeoyl-quinic acid
Quercetin
Phloridzin
Quercetin 3-O-rhamnoside
Quercetin
Apigenin
Kaempferol
ABTS scavenging assay
DPPH radical scavenging activity
[59]
Water45 mg GAE/L-ORAC (oxygen radical
antioxidant capacity)
[62]
Grape (Vitis sp.)Hydroalcoholic 60%
(ethylic alcohol)
13643±690 mg GAE/gDelphinidin-3-glucoside
Malvidin-3-glucoside
Cyanidin-3-glucoside
Malvidin-3,5-diglucoside
2,5 Dihydroxybenzoic acid
2,4 Dihydroxybenzoic acid
Syringic acid
5-O-Caffeoylquinic acid
Gallic acid
2-Hydroxycynnamic acid
Ellagic acid
Quercetin
Quercetin-3-rutinoside
(+)-Catechin
(-)-Epicatechin
trans-Resveratrol
Ferric-reducing
antioxidant power (FRAP)
Folin-Ciocalteu reducing capacity
DPPH radical scavenging activity
[64]
Ethanol/acidic water
(pH 3.2)
7:3 v/v
not determinedVanillic acid
p-Coumaric acid
E-resveratrol
Quercetin
Kaempferol
Pinoresinol
PTP-1B (Protein Tyrosine
Phosphatase 1B enzyme)
inhibitory power
[28]
Ethanol/water
(80:20, v/v)
161.66 mg GAE/gPhenolic acid
Gallic acid
Epicatechin
Epigallocatechin gallate
Epicatechin gallate
Epigallocatechin
Procyanidin B1
Procyanidin B4
Procyanidin B2
Flavonol
Kaempferol
Myricetin
Quercetin
Cytotoxicity
in murine macrophage
RAW 264.7

Anti-inflammatory activity
[65]
50 mM
acetate buffer at 1:10 w/v
0.81 g GAE/100gGallic acid
p-Coumaric acid
Syringic acid
(+)-Catechin
Resveratrol
Malvidin-3-O-glucoside
TEAC[68]
Passion fruit
(Passiflora sp)
Acetonitrile~40 mg GAE/100g4-hydroxybenzoic acid
Chlorogenic acid
Vanillic acid
Caffeic acid
p-Coumaric acid
Ferulic acid
Rutin
Quercetin
trans-Cinnamic acid
ABTS scavenging assay
DPPH radical scavenging activity
Gastroprotective activity
[70]
Water0.14 mg GAE/mLVanillic acid
Syringic acid
Gallic acid
Rutin
Quercetin
ORAC and DPPH assays
Folin-Ciocalteu assay
[66]
Hexane
Ethyl acetate
Ethanol
Ethanol/water
Maceration (EtOH–H2O 142.4 ± 0.4 mg GAE/g, EtOH 75 ± 2 mg GAE/g, EtOAc 24.6 ± 0.8 mg, GAE/g, Hx 30 ± 1 mg GAE/g)

Ultrasound-assisted
(EtOH–H2O 61.3 ± 0.4 mg GAE/g, EtOH 21.4 ± 0.4 mg GAE/g, EtOAc 19 ± 1 mg GAE/g, Hx 22.3 ± 0.1 mg GAE/g)

Supercritical fluid extraction (40 °C 150 bar 33.0 ± 0.8 mg GAE/g, 250 bar 30 ± 1 mg GAE/g, 300 bar 18 ± 2 mg GAE/g, 50 °C 150 bar 26.4 ± 0.8 mg GAE/g, 250 bar 24.3 ± 0.4 mg GAE/g, 300 bar 19.3 ± 0.8 mg GAE/g)
not determinedABTS scavenging assay
DPPH radical scavenging activity
Antimicrobial activity
[69]
Petroleum ether
Chloroform
Acetone
Methanol
115.13 ± 8.42 mg GAE/g,
113.45 ± 6.19 mg GAE/g,
417.65 ± 7.33 mg GAE/g,
227.17 ± 10.97 mg GAE/g
Quercetin
Gallic acid
Apigenin
Catechin
DPPH
FRAP
Metal chelating activity
Superoxide radical scavenging activity
Analgesic activity
Anti-inflammatory activity
Antipyretic effect
[67]
Pomegranate (Punica granatum)Ethanolic extracts - soluble and insoluble-bound phenolic compounds3.1 ± 0.3 (mg GAE/g)not determinedORAC and TEAC assays[41]
Ethanolic extracts73 ± 13.35 (mg GAE/g)not determinedFolin-Ciocalteu reducing capacity and β-Carotene oxidation method[42]
Methanolic extracts (oil)88.45 ± 3.89 mg GAE/kg oilnot determinedDPPH[43]
Isolation of free, esterified and insoluble-bound phenolic compoundsFree 1.38 ± 0.01 Esterified 1.39 ± 0.01 and Insoluble-bound 0.62 ± 0.01 (mg GAE/g)Gallic acid (major phenolic acid present)TEAC, electron paramagnetic resonance (EPR) spectrometry, Metal chelating ability[44]
Ethanolic extracts (oil)not determinedPalmitic (16:0)
stearic (18:0)
oleic (18:1 ω-9)
linoleic (18:2 ω-6) acids
β-carotene bleaching assays, DPPH, ORAC, and ABTS assays[45]
Camu-camu (Myrciaria dubia)aqueous acetone (50:50 v/v)369.4±9.6 (mg GAE/g)not determinedABTS scavenging assay
DPPH radical scavenging activity
[46]
Ethanolic extracts3738.0 ± 20.8 mg GAE/100 gEllagic acid
Syringic acid
Quercetin
Myricetin
Catechin
DPPH, Folin–Ciocalteau reducing capacity[47]
Aqueous extractsnot detectednot determinedFRAP and DPPH assays[48]
Ethanolic extracts128 mg GAE/100 gRosmarinic acid
2,4-dihydroxybenzoic acid
Ellagic acid
Cyanidin-3-glucoside
Methylvescalagin
trans-Resveratrol
Quercetin
DPPH, FRAP and Folin-Ciocalteu reducing capacity[49]
EtOH: ethyl alcohol, H2O: water, (CH3)2CO: propanone)2400 (EtOH), 4000 (H2O) and 1300 ((CH3)2CO) mg GAE/100 gGallic and chlorogenic acid (H2O)
Ellagic acid (EtOH)
Ferulic acid ((CH3)2CO)
DPPH, FRAP and Folin-Ciocalteu reducing capacity assays[15]
Jabuticaba (Plinia jaboticaba and Plinia cauliflora)Ethanolic extracts116.17 ± 7.10
mg of GAE/ g
not determinednot determined[53]
Soluble phenolics, Alkaline hydrolysis and Acid hydrolysisnot determinedCyanidin-3-O-glucoside
Gallic acid
Delphinidin-3-O-glucoside
Rutin
Folin–Ciocalteu, FRAP, TEAC and ORAC assays[54]
water:propanone (60:40 v/v)8.65 g GAE/100 gEllagic acid and ellagitanninsDPPH scavenging activity and total reducing capacity[55]
Methanolic extractsnot determinedEllagic acid derivativesFolin–Ciocalteu reducing capacity, DPPH and FRAP assays[56]
Apple (Malus domestica)Methanol5.74–17.44 GAE/gProtocatechuic acid
(+)-Catechin
Proanthocyanin B2
Chlorogenic acid
Caffeic acid
(-)-Epicatechin
Quercetin
Hyperin
Phloridzin
DPPH, FRAP and ABTS assays[50]
Aqueous acetone (30:70; v/v) and ethyl acetate extraction after hexane extractionnot determined3-p-Coumaroylquinic acid
5-Caffeoylquinic acid
4-Caffeoylquinic acid
Caffeic acid
3-Caffeoylquinic acid
5-p-Coumaroylquinic acid
4-p-Coumaroylquinic acid
Proanthocyanin B2
Epicatechin
3-hydroxyphloridzin
Phloretin-xyloglucoside
Phloridzin
Phloretin
Procyanidins
not determined[52]
Ethanol-water (30:70)not determinedPhloridzinnot determined[58]
Methanolnot determinedQuercetin
Quercetin-3-O-galactoside
Quercetin-3-O-glucoside
Quercetin-3-O-rhamnoside
Phloridzin
Phloretin
(+)-Catechin
(-)-Epicatechin
Cyanidin-3-O-galactoside
Luteolin-7-O-glucoside
Caffeic acid
Chlorogenic acid
Ferulic acid
Isoferulic acid
not determined[57]
Methanolnot determinedEpicatechin
Procyanidin B2
Catechin
Chlorogenic acid
p-Coumaroylquinic acid
Quercetin-3-O-galactoside
Quercetin-3-O-rhamnoside
Quercetin-3-O-glucoside
Phloridzin
Phloretin-xyloglucoside
Phloretin
not determined[71]
Hexane and 70% aqueous acetone after hexane extractionnot determinedLinoleic acid
Palmitic acid
Linolenic acid
Stearic acid
Oleic acid
Chlorogenic acid
p-Coumaroylquinic acid
Quercetin-3-galactoside
Quercetin-3-rutinoside
Quercetin-3-glucoside
3-hydroxyphloridzin
Quercetin-3-xyloside
Phloretin-2′-xyloglucoside
Quercetin-3-arabinoside
Quercetin-3-rhamnoside
Phloridzin
not determined[72]
Plum (Prunus sp.)Aqueous methyl alcohol (50%) and aqueous acetone (70%)Total extractable polyphenols 86 mg gallic acid/100 g ABTS assay[63]
Total carotenoids 3.9 μg/g
n-hexane and ultrasonication α-Tocopherol [73]
β-Tocopherol
γ-Tocopherol
δ-Tocopherol
α-Tocotrienol
γ-Tocotrienol
Cholesterol
Campesterol
Δ5-Stigmasterol
β-Sitosterol
Sitostanol
Δ5-Avenasterol
Δ7-Stigmasterol
Cycloartenol
Table 2. Previous research on phenolic compounds and bioactivity of berry seeds.
Table 2. Previous research on phenolic compounds and bioactivity of berry seeds.
Fruit SeedsExtraction SolventsTotal Phenolics aIndividual PhenolicsBioactivity bReference
Bilberry
Vaccinium myrtilluswater2.3 mg GAE/g dry extractnot determinedAnti-oxidative activity
TEAC (0.1 and 82 μmol TE/g dry extract in n-hexane and distilled water, respectively)
[159]
seed oil, Vaccinium myrtillus not determinednot determinedAnti-oxidative activity
OOR• scavenging (519 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (1188 μmol trolox equivalent/100 g oil)
[163]
seed oil, Vaccinium myrtillus not determinednot determinedAntioxidant activity
DPPH (EC50 = 5.5–9.5 mg oil/mL)
[164]
Blackberry
Rubus spp.methanol:acetone:water (7:7:6, v/v/v), acetone:water (8:2, v/v), methanol:water (7:3, v/v), water, diethyl ether:ethyl acetate (1:1, v/v)13 mg GAE/g dry extractprotocatechuic acid, p-coumaric acid, gallic acid, gallic hexoside, caffeic acid, syringic acid, catechin, epicatechin, epigallocatechin, epicatechin gallate, B-type procyanidin dimmers, quercetin, quercetin 3-O-glucoronide, quercetin pentose, myricetin, peonidin 3-O-glucosideAntioxidant activity
OH• scavenging (125 μmol TE/g dry extract), ORAC (60 μmol TE/g dry extract), Reducing power (88 μmol TE/g dry extract), Inhibition against human LDL-cholesterol oxidation (21–60% from 0 to 22 h), Iron(II) chelating (199 μmol TE/g dry extract), Inhibition of OH• and OOR• induced supercoiled DNA strand scission (85–95% for OH•, and 90–95% for OOR•)
[19]
Rubus ulmifoliusmethanol171 mg GAE/g dry seedscyanidin 3-O-glucoside, ellagic acid, galloyl-HHDP-glucose, galloyl-bis-HHDP-glucoseAntioxidant activity
DPPH (93, 96, and 98% radicals scavenged at 42, 83, 167 μg/mL of extracts, respectively; EC50 = 1 μg/mL)
Anti-inflammatory activity
Strong inhibitory effects on the production of LPS-induced inflammatory mediators (NO, CCL-20)
[165]
Rubus fruticosus
3 cultivars
methanol:water (1:1, v/v)not determinedEllagitannins (41 compounds), ellagic acid derivatives (10), gallic acid derivatives (4), protocatechuic acid, chlorogenic acid, salicylic acidProtective effect on chromosome aberrations in peripheral human lymphocytes[166]
seed oil, Rubus fruticosusmethanol:water (1:1, v/v)5.6–9.1 mg CAE/g oilp-coumaric acid, vanillic acid, vanillinAntioxidant activity
FRAP (0.3 FeSO4 equivalent (μmol/L)/g oil)
[167]
Blueberry
Vaccinium spp.methanol:acetone:water (7:7:6, v/v/v), acetone:water (8:2, v/v), methanol:water (7:3, v/v), water, diethyl ether:ethyl acetate (1:1, v/v)2 mg GAE/g dry extractprotocatechuic acid, p-coumaric acid, gallic acid, gallic hexoside, caffeic acid, syringic acid, catechin, epicatechin, epigallocatechin, epicatechin gallate, B-type procyanidin dimmers, quercetin, quercetin pentose, myricetin, kaempferol hexoside, delphinidin 3-O-hexoside, cyanidin 3-O-(6-O-acetyl)galactoside, peonidin 3-O-glucoside, peonidin 3-O-arabinoside, petunidin 3-O-galactoside, petunidin 3-O-arabinosideAntioxidant activity
OH• scavenging (37 μmol TE/g dry extract), ORAC (5 μmol TE/g dry extract), Reducing power (60 μmol TE/g dry extract), Inhibition against human LDL-cholesterol oxidation (6–49% from 0 to 22 h), Iron(II) chelating (12 μmol TE/g dry extract), Inhibition of OH• and OOR• induced supercoiled DNA strand scission (50–70% for OH•, and 55–75% for OOR•)
[19]
seed oil, Vaccinium corymbosummethanol:water (1:1, v/v)8.8–9.5 mg CAE/g oilhomovanillic acid, vanillinAntioxidant activity
FRAP (0.26–0.41 FeSO4 equivalent (μmol/L)/g oil)
[167]
seed flour, Vaccinium corymbosumacetone:water (1:1, v/v)16 mg GAE/g seed flournot determinedAntioxidant activity
ORAC (153 μmol TE/g seed flour), DPPH (ED50 = 670 μg flour equivalents/mL), Iron(II) chelating (1.9 mg EDTA equivalents/g seed flour)
[168]
Cloudberry
Rubus chamaemorus, after fermentationacetone:water (7:3, v/v)457 mg/g dry extracts (measured by HPLC)ellagic acid, ellagic acid glycosides, sanguiin H10, casuarictin/potentillin, lambertianin C, sanguiin H6, sanguiin H2, ferulic acid, quercetin 3-O-[6″-(3-hydroxy-
3-methylglutaroyl)-β-glucoside], quercetin-3-O-glucuronide
Anti-bacterial activity
Staphylococcus aureus (very strong inhibition), Escherichia coli (strong inhibition), Pseudomonas aeruginosa (clear inhibition), Candida albicans (no inhibitory effects), Saccharomyces cerevisiae (no inhibitory effects)
Anti-inflammatory activity
significantly reduced NO and IL-6 production and iNOS expression in activated macrophages
[171]
seed oil, Rubus chamaemorus not determinednot determinedAntioxidant activity
OOR• scavenging (1157 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (650 μmol trolox equivalent/100 g oil)
[163]
Cranberry
Vaccinium oxycoccuswaternot determinednot determinedAntioxidant activity
TEAC (0.1 and 50 μmol TE/g dry extract in n-hexane and distilled water, respectively)
[159]
seed oil, Vaccinium macrocarponmethanol:water (1:1, v/v)11.0–11.3 mg CAE/g oil4-(2-hydroxyethyl)phenol, p-coumaric acid, homovanillic acid, vanillic acid, protocatechuic acidAntioxidant activity
FRAP (0.27–0.29 FeSO4 equivalent (μmol/L)/g oil)
[167]
seed flour, Vaccinium macrocarponacetone:water (1:1, v/v)15 mg GAE/g seed flournot determinedAntioxidant activity
ORAC (111 μmol TE/g seed flour), DPPH (ED50 = 1260 μg flour equivalents/mL), Iron(II) chelating (2.1 mg EDTA equivalents/g seed flour)
Anti-proliferative activity
significant inhibited HT-29 cell proliferation
[168]
seed oil, Vaccinium oxycoccos not determinednot determinedAntioxidant activity
OOR• scavenging (1543 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (399 μmol trolox equivalent/100 g oil)
[163]
seed oil, Vaccinium macrocarpon not determinednot determinedAntioxidant activity
OOR• scavenging (975 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (338 μmol trolox equivalent/100 g oil)
[163]
Currant
black, Ribes nigrum
5 cultivars
acetone:water (1:1, v/v)1.6–2.3 mg GAE/g dry seed residuesdelphinidin 3-O-glucoside, delphinidin 3-O-rutinoside, cyanidin 3-O-glucoside, cyanidin 3-O-rutinoside, myricetin 3-O-glucoside, myricetin 3-O-rutinoside, quercetin 3-O-glucoside, quercetin 3-O-rutinoside, kaempferol 3-O-glucoside, kaempferol 3-O-rutinoside, p-coumaric acid, p-coumaroyl glycosideAntioxidant activity
ABTS (14–17 μmol TE/g dry seed residues), DPPH (11–13 μmol TE/g dry seed residues)
[170]
black, Ribes nigrumwater0.9–1.8 mg GAE/g dry extractnot determinedAntioxidant activity
TEAC (0.6–0.7 and 26–60 μmol TE/g dry extract in n-hexane and distilled water, respectively)
[159]
black, seed oil, Ribes nigrum not determinednot determinedAntioxidant activity
OOR• scavenging (1068 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (478 μmol trolox equivalent/100 g oil)
[163]
Elderberry
Sambucus nigramethanol54 mg GAE/g dry seedscyaniding 3-O-sambubioside
-5-O-glucoside, pelargonidin 3-O-rutinoside, quercetin 3-O-rutinoside, quercetin 3-O-glucoside
Antioxidant activity
DPPH (42, 55, 82% radicals scavenged at 42, 83, 167 μg/mL of extracts, respectively; EC50 = 82 μg/mL)
Anti-inflammatory activity
No inhibitory effects on the production of LPS-induced inflammatory mediators (NO, CCL-20)
[165]
Sambucus nigrawater1.5 mg GAE/g dry extractnot determinedAntioxidant activity
TEAC (0.8 and 30 μmol TE/g dry extract in n-hexane and distilled water, respectively)
[159]
Lingonberry
seed oil, Vaccinium vitis-idaea not determinednot determinedAntioxidant activity
OOR• scavenging (299 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal
lipid peroxidation (329 μmol trolox equivalent/100 g oil)
[163]
Raspberry
black, Rubus spp.methanol:acetone:water (7:7:6, v/v/v), acetone:water (8:2, v/v), methanol:water (7:3, v/v), water, diethyl ether:ethyl acetate (1:1, v/v)7 mg GAE/g dry extractprotocatechuic acid, p-coumaric acid, gallic acid, gallic hexoside, caffeic acid, syringic acid, catechin, epicatechin, epigallocatechin, epicatechin gallate, B-type procyanidin dimmers, quercetin, quercetin 3-O-glucoronide, quercetin pentose, myricetin, peonidin 3-O-glucosideAntioxidant activity
OH• scavenging (67 μmol TE/g dry extract), ORAC (47 μmol TE/g dry extract), Reducing power (60 μmol TE/g dry extract), Inhibition against human LDL-cholesterol oxidation (18–57% from 0 to 22 h), Iron(II) chelating (69 μmol TE/g dry extract), Inhibition of OH• and OOR• induced supercoiled DNA strand scission (60-80% for OH•, and 80-85% for OOR•)
[19]
seed oil, Rubus spp.methanol2.7 mg CAE/100 g oilnot determinedAntioxidant activity
Inhibitory effects on the activities of superoxide dismutase and glutathione peroxidase
[160]
red, seed oil, Rubus idaeusmethanol:water (1:1, v/v)8.4 mg CAE/g oil4-(2-hydroxyethyl)phenolAntioxidant activity
FRAP (0.34 FeSO4 equivalent (μmol/L)/g oil)
[167]
black, seed flour, Rubus occidentalisacetone:water (1:1, v/v)41 mg GAE/g seed flournot determinedAntioxidant activity
ORAC (296 μmol TE/g seed flour), DPPH (ED50 = 200 μg flour equivalents/mL), Iron(II) chelating (3.6 mg EDTA equivalents/g seed flour)
Anti-proliferative activity
significant inhibited HT-29 cell proliferation
[168]
red, seed flour, Rubus ideausacetone:water (1:1, v/v)25 mg GAE/g seed flournot determinedAntioxidant activity
ORAC (276 μmol TE/g seed flour), DPPH (ED50 = 510 μg flour equivalents/mL), Iron(II) chelating (3.9 mg EDTA equivalents/g seed flour)
[168]
seed oil, Rubus idaeus not determinednot determinedAntioxidant activity
OOR• scavenging (2315 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (424 μmol trolox equivalent/100 g oil)
[163]
Sea buckthorn
Hippophaë rhamnoidesethanol:water (1:9, v/v)120 mg GAE/g dry extracttannins, gallic acid, quercetin 3-O-galactoside, kaempferol, isorhamnetin, isorhamnetin 3-O-glucoside, isorhamnetin 3-O-rutinosideAntioxidant activity
DPPH (529 mg TE/g dry extract), FRAP (454 mg TE/g dry extract)
Anti-bacterial activity
Enterecoccus durans (68% inhibition percentage), Candida albicans (68%), Bacillus cereus (64%), Staphylococcus aureus (41%), Escherichia coli (38%), Pseudomonas aeruginosa (28%)
[161]
Hippophaë rhamnoidesethanol9.4–23.5 mg GAE/g dry seedsquercetin-3-O-galactoside, quercetin, myricetin, isorhamnetinAntioxidant activity
ABTS (44–182 μmol TE/g dry seeds), DPPH (128–283 μmol TE/g dry seeds)
[162]
seed oil, Hippophaë rhamnoides not determinednot determinedAntioxidant activity
OOR• scavenging (1323 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (245 μmol trolox equivalent/100 g oil), Inhibition of LDL oxidation (IC50 = 2.0 μL oil/mg LDL), O2•- scavenging (IC50 = 2 g oil/L), Inhibition against DNA oxidation (protective effects on purified DNA and rat liver homogenate from UV-induced DNA oxidation in vitro)
[163]
Strawberry
seed oil, Fragaria × ananassamethanol1.8 mg CAE/100 g oilsnot determinedAntioxidant activity
Inhibitory effects on the activities of superoxide dismutase and glutathione peroxidase
[160]
Fragaria × ananassawater1.7 mg GAE/g dry extractnot determinednot determined[159]
seed oil, Fragaria × ananassamethanol:water (1:1, v/v)9.3–10.4 mg CAE/g oilp-coumaric acid, ferulic acid, 4-(2-hydroxyethyl)phenol, homovanillic acid, vanillic acid, vanillin.Antioxidant activity
FRAP (0.24–0.30 FeSO4 equivalent (μmol/L)/g oil)
[167]
seed oil, Fragaria × ananassa not determinednot determinedAntioxidant activity
OOR• scavenging (296 μmol α-tocopherol equivalent/100 g oil), Inhibition of microsomal lipid peroxidation (1015 μmol trolox equivalent/100 g oil)
[163]
Note: a GAE means gallic acid equivalents, CAE means caffeic acid equivalents; b TE means trolox equivalents.
Table 3. Examples of extractions of bioactive compound from apple pomace.
Table 3. Examples of extractions of bioactive compound from apple pomace.
ExtractionAgro-Industrial WasteTotal Bioactive CompoundsIndividual Bioactive CompoundsAntioxidant Activity (AA)Conclusion RemarksReference
Conventional extraction:

30 °C for 30 min in a shaker
70% with methanol containing 2% formic acid
Apple pomace (peel and seed)Total phenols (mg GAE/L)
Peel: 2228.49 Pomace: 208.75

Total flavanols (mg CTE/L)
Peel: 170.33 Pomace: 40.91
Phenolic compounds (mg/L)
Peel:
Gallic acid: 40.42
Vanillic acid: 58.70
Caffeic acid: 0.21
Chlorogenic acid: 7.05 Catechin: 37.67
Epicatechin gallate: 12.85
Phlorizin: 60.28

Pomace:
Gallic acid: 29.57
Vanillic acid: 56.02
Chlorogenic acid: 5.29 Catechin: 35.40
Epicatechin gallate: 21.68
Phlorizin: 12.81
DPPH (µmol TE/L)
Peels: 4203.59
Pomace: 3830.72
Peel pomace contained significantly higher amounts of bioactive compounds in comparison to pulp pomace.

Due to high levels of
antioxidant and bioactive compounds, apple pomaces
could be a
better material to make apple cider vinegar.
[211]
Conventional extraction:

Magnetic stirring with methanol during 30 min.
After centrifugation and filtration, methanol was evaporated through Soxhlet extraction (60 ± 0.5 °C).

Crude extract was further dissolved and extracted with
ethanol, chloroform, ethyl acetate and n-hexane to obtain their fractions.
Apple pomace
freeze-dried and milled
Total phenols (mg GAE/g DW)

Methanol: 3.48
Ethanol: 3.04
Ethyl acetate: 2.85
Chloroform: 1.64
n-hexane: 1.50
The average amounts of 6 triterpenoic acids detected (mg/100 g DW):
maslinic acid: 0.96 oleanolic acid: 3.18
ursolic acid: 6.14 betulinic acid: 1.78
erythrodiol: 0.78
uvaol: 0.84
DPPH (%)
Methanol: 72.6
Ethanol: 67.5
Ethyl acetate: 64.2
Chloroform: 59.6
n-hexane: 56.2

FRAP (%)
Methanol: 65.8
Ethanol: 60.3
Ethyl acetate: 58.3
Chloroform: 55.2
n-hexane: 50.1

ABTS (%)
Methanol: 84.3
Ethanol: 79.2
Ethyl acetate: 72.9
Chloroform: 65.3
n-hexane: 60.8
The antioxidant activity results showed that the methanol and ethanol fractions were more effective radical scavengers than chloroform, ethyl acetate and n-hexane.

Both alcoholic fractions showed potential toward tyrosinase, xanthine oxidase and urease inhibition.

Among the TAAs, ursolic acid, betulinic acid and maslinic acid showed effective radical scavenging activity.

All TAAs showed lower inhibitory activity towards free radicals in vitro than the apple pomace extracts.
Apple pomace methanol extract and ursolic acid revealed prominent anticancer activity on Hela, Skov-3, Caski, and NCL cancer cell lines, respectively.
[212]
Ultrasound Assisted Extraction (UAE):

Defatted seed flours were mixed with 80% methanol and
held in an ultrasonic bath for 30 min at room temperature.
Apple seeds
(defatted seed flours)
Total phenols (mg GAE/kg DS)
Fuji Zhen Aztec 2861
Granny Smith 3581
Jeromine 4096
Pink Lady 3616 Super Chief 5141
Phenolic compounds (mg/kg DM):

Fuji Zhen Aztec
Phloridzin: 1748.7
Ellagic acid: 189.5
(-)-Epicatechin: 76.3
Caffeic acid: 114.2
(+)-Catechin: 90.1
Ferulic acid: 57.8
Protocatechuic acid: 46.8
Gallic acid: 4.2
Granny Smith
Phloridzin: 2106.6
Ellagic acid: 275.3
(-)-Epicatechin: 77.2
Caffeic acid: 9.1
(+)-Catechin: 5.0
Ferulic acid: 142.2
Protocatechuic acid: 48.7
Gallic acid: 7.5

Jeromine
Phloridzin: 2623.5
Ellagic acid: 286.7
(-)-Epicatechin: 164.6
Caffeic acid: 11.8
(+)-Catechin: N.D.
Ferulic acid: 21.2
Protocatechuic acid: 153.8
Gallic acid: 5.6
Pink Lady
Phloridzin: 1888.4
Ellagic acid: 216.5
(-)-Epicatechin: 73.9
Caffeic acid: 10.4
(+)-Catechin: 21.0
Ferulic acid: 8.2
Protocatechuic acid: 39.6
Gallic acid: 4.2

Super Chief
Phloridzin: 3462.2
Ellagic acid: 230.7
(-)-Epicatechin: 69.0
Caffeic acid: 14.2
(+)-Catechin: 191.0
Ferulic acid: 36.4
Protocatechuic acid: 161.3
Gallic acid: 7.9
ABTS (mg GAE/kg DS)
Fuji Zhen Aztec 363
Granny Smith 292
Jeromine 368
Pink Lady 392
Super Chief 343 mg

DPPH µmol TE/g DS)

Fuji Zhen Aztec 24.28
Granny Smith 33.12
Jeromine 32.04
Pink Lady 21.45
Super Chief 43.56
Phloridzin represented 52–67% and 75–83% of the total phenolics measured by the Folin-Ciocalteu assay and HPLC method, respectively.

Chewing gum could be a suitable delivering material for phloridzin uptake originated from apple seeds.

5 min was enough for the dissolution of almost all added phloridzin (88.43–96%).
[213]
Hot water extraction (HWE):

Boiling water with 1% acetic acid at S/L=1/60 g/mL during 10 min.
Apple pomace obtained after processing of aTotal phenols (µg GAE of extract/mL)

HWE: 10.7
pHWE: 149
pHWE was mainly composed of flavonols: quercetin-3-O-galactoside (27%), quercetin-3-O-
rhamnoside (23%), quercetin- 3-O-arabinofuranoside (13%), and
DPPH (EC50, µg of extract/mL)

HWE: 1339
pHWE: 82.4

ABTS (EC50, µg of extract/mL)
HWE represented 29% of dry apple pomace and presented 11 g/kg of polyphenols.

3.26 g GAE of polyphenols per kg of apple pomace were extracted.
Fractionation processes of HWE extracts:
ultrafiltration to obtain LMWM (low molecular weight material) and HMWM (high molecular weight material);
SPE to obtain polyphenol-isolated fraction (pHWE) and non-retained material (NrFr).
mixture of apples (Royal
Gala variety), employing milling, enzymatic digestion and pressing processes, afterwards it was freeze-dried.
Ultrafiltration: 11% of the polyphenols from the HWE remained in the HMWM. This fraction accounted for 6.9% of the apple pomace.

SPE: The hydrophobic fraction pHWE corresponded to 1.6% of the
dry apple pomace and accounted for 63% of those in the HWE.
the dihydrochalcone phloretin-2-O-glucoside
(14%).
HWE: 532
pHWE: 35.2
When apple pomace HWE were added to yogurt formulations, a final product with improved fiber content and antioxidant properties was achieved in comparison to control sample (plain yogurt).[214]
Ultrasound Assisted Extraction (UAE):

Extraction with
aqueous acetone (70/30 vol) for 5 min in an ultrasonic bath at 20°C.
Apple seeds oil and defatted seed flour from the cider-
making industry
In defatted seeds flour:

Total phenols
2.7-6.7 mg GAE/g defatted matter

Condensed tannins
2.4-4.0 mg cyanidin/g defatted matter

Hydrolysable tannins
34.5-47.2 mg GAE/g defatted matter
In apple seeds oil:

DPPH
IC50 = 4.8-5.8 mg of oil/mL
Apple seeds oil exhibited an significant antioxidant activity due to high levels of tocopherols
(1280 ± 104.8 mg/kg oil) with β-tocopherol (794.5 ± 62.2 mg/kg oil) being most abundant, followed
by α-tocopherol (439.2 ± 34.5 mg/kg oil).

Defatted seeds flour contained considerable amounts of phenolic compounds.
[215]
Microwave-assisted extraction (MAE):

MAE parameters:
solvent type:
70% acetone (Ac),
60% ethanol (Et);
microwave power: 100–900W;
solvent volume to sample ratio:
4–12 mL/g dry pomace;
extraction time:
30–180 s.
Freeze-dried apple pomace
of Red Delicious (RD) and Jonathan (J) apple varieties
Total phenols (mg GAE/g DW) under the optimum MAE parameters:


RD-Ac: 6.66
RD-Et: 15.81
J-Ac: 5.79
J-Et: 7.65
Phenolic compounds (µg/g dry pomace)

Phloridzin
RD-Ac: 82.59
RD-Et: 25.25
J-Ac: 102.9
J-Et: 79.80

Gallic acid:
RD-Ac: nd
RD-Et: 189.09
J-Ac: nd
J-Et: 23.08
Caffeic acid:
RD-Ac: 10.00
RD-Et: 27.63
J-Ac: 9.77
J-Et: 34.67
Chlorogenic acid:
RD-Ac: nd
RD-Et: nd
J-Ac: nd
J-Et: 22.56

Quercetin:
RD-Ac: 46.33
RD-Et: 59.20
J-Ac: 115.15
J-Et: 211.34
DPPH (%)

60% ethanol: 77.1
70% acetone: 93.7
The optimum conditions for MAE: 735 W power and 149 s extraction
time with 10.3 mL of ethanol per gram dry sample.

Higher total phenols and DPPH radical scavenging activity was found in Red Delicious apple pomace extracts.

Extraction solvent was not found as significant extraction parameter.
[216]
Conventional extraction (CE):
agitation (1000 rpm) of samples (2.5 g) in water (50 mL) at room temperature.

Ultrasound-assisted extraction (UAE):
10 g of sample in 200 mL of water
ultrasonic (US) probe diameter: 1.2 cm, height: 13.7 cm at a depth of ca. 20 mm.
US power: 7.8 or 49.5 W
The aliquots were sampled every 2, 5,
10, 20 and 30 min and stored until further analysis.

Microwave-assisted extraction (MAE):
10 g of sample in 200 mL of water. Power: 400, 600, 1000 W (which corresponds to the actual power of 302, 384, and 435 W) for 2 or 4 irradiation cycles, each consisted of 1 or 1.5 min.
High speed
homogenization (HH)+MAE:
Samples were pre-treated with a high-speed homogenizer
prior to MAE.
Homogenization at 22,000 rpm for 1 min + MAE: 1000 W for 4 cycles of 1.5 min each.

Ultrasound-assisted enzymatic extraction (UAEE):
the same as UAE + the pectinase solution
Apple pomace consisted of stalk, skin and the fruit kernel were mixed thoroughly and ground into powder.Total phenols (mg GAE/g raw sample)

CE: 2.62-2.68
UAE: 0.7-2.11
MAE: 2.23-2.81
HH: 2.32-2.44
HH+MAE: 2.65-2.78
UAEE: 2.50-4.62
LC-MS analysis of fractions obtained from UAEE revealed the presence of 26
phenolic compounds.
The DPPH scavenging ability of the extracts ranged from 27.1 to 54.6 ± 6.9 mg
trolox/L, depending on the extraction parameters.
In most cases, extraction time and the particle size of the sample had significant effect on the extraction yield and longer extraction times lead to increased TPC in the extract.

UAEE found to be superior techniques to extract TPC from apple pomace due to the synergistic effects of US and
enzymes.

The antioxidant activity decreased with
the extraction time and enzyme dosage.
[217]
Mild intensity pulsed electric field (PEF)

PEF treatment:
1, 2 and 3 kV/cm
500, 875 and 1250 μs
The specific energy intake: 0.44-17.0 kJ/kg

Extraction:
Fluor to water ratio (FWR): 5, 8.75, 12.5% (w/v)
Centrifugation with methanol.

Free phenolic acid extraction: shaking with 80% aqueous methanol, centrifugation, evaporation and
Fermented apple pomace powder (12.5% w/v).Total phenols

402.7 mg GAE/100g DW
(at 12.5% FWR)
Phenolic compounds (μg/g DW)

Control samples:
Protocatechuic acid: 392.6
Chlorogenic acid: 72.5
Caffeic acid: 130.8
p-Coumaric acid: 24.5
Ferulic acid: 64.4
Salicylic acid: 131.9

PEF treated samples:
Protocatechuic acid: 98.9
Caffeic acid: 141.1
p-Coumaric acid: 34.3
Ferulic acid: 70.1
Salicylic acid: 170.2
DPPH

799.3 μmol TE/100g DW (at 12.5% FWR)
Total phenols and AA were significantly (p < 0.05) influenced by the FWR and its
interaction with the electric filed intensity and time.

With the increase in electric filed intensity from 1 kV/cm to 3 kV/cm, a significant (p < 0.05) increase of 4.8% TPC and 4.4% AA was observed.

A significant (p < 0.05) decrease of 3.8% and 2.6% was observed in total phenols and AA, respectively with the increase in treatment time from 500 μs to 875 μs followed by no
significant (p > 0.05) change in both total phenols and AA.
The optimum condition of PEF treatment was found as 12.5% (w/v) FWR, 2 kV/cm electric field intensity and 500 μs treatment time with a specific energy input of 3.0 kJ/kg.

Obtained results indicated that the PEF treatment could be useful tool for the processing of food with enhanced levels of
phenolic antioxidants.
[218]

Share and Cite

MDPI and ACS Style

Fidelis, M.; de Moura, C.; Kabbas Junior, T.; Pap, N.; Mattila, P.; Mäkinen, S.; Putnik, P.; Bursać Kovačević, D.; Tian, Y.; Yang, B.; et al. Fruit Seeds as Sources of Bioactive Compounds: Sustainable Production of High Value-Added Ingredients from By-Products within Circular Economy. Molecules 2019, 24, 3854. https://doi.org/10.3390/molecules24213854

AMA Style

Fidelis M, de Moura C, Kabbas Junior T, Pap N, Mattila P, Mäkinen S, Putnik P, Bursać Kovačević D, Tian Y, Yang B, et al. Fruit Seeds as Sources of Bioactive Compounds: Sustainable Production of High Value-Added Ingredients from By-Products within Circular Economy. Molecules. 2019; 24(21):3854. https://doi.org/10.3390/molecules24213854

Chicago/Turabian Style

Fidelis, Marina, Cristiane de Moura, Tufy Kabbas Junior, Nora Pap, Pirjo Mattila, Sari Mäkinen, Predrag Putnik, Danijela Bursać Kovačević, Ye Tian, Baoru Yang, and et al. 2019. "Fruit Seeds as Sources of Bioactive Compounds: Sustainable Production of High Value-Added Ingredients from By-Products within Circular Economy" Molecules 24, no. 21: 3854. https://doi.org/10.3390/molecules24213854

Article Metrics

Back to TopTop