Next Article in Journal
Improved Tetanic Force and Mitochondrial Calcium Homeostasis by Astaxanthin Treatment in Mouse Skeletal Muscle
Next Article in Special Issue
Medicinal Profile, Phytochemistry, and Pharmacological Activities of Murraya koenigii and its Primary Bioactive Compounds
Previous Article in Journal
(–)-Epicatechin Reduces the Blood Pressure of Young Borderline Hypertensive Rats During the Post-Treatment Period
Previous Article in Special Issue
Phytochemical Profile and Antioxidant Capacity of Coffee Plant Organs Compared to Green and Roasted Coffee Beans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Grown to Be Blue—Antioxidant Properties and Health Effects of Colored Vegetables. Part II: Leafy, Fruit, and Other Vegetables

by
Francesco Di Gioia
1,
Nikolaos Tzortzakis
2,
Youssef Rouphael
3,
Marios C. Kyriacou
4,
Shirley L. Sampaio
5,
Isabel C.F.R. Ferreira
5 and
Spyridon A. Petropoulos
6,*
1
Department of Plant Science, The Pennsylvania State University, University Park, PA 16802, USA
2
Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, 3603 Lemesos, Cyprus
3
Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
4
Department of Vegetable Crops, Agricultural Research Institute, 1516 Nicosia, Cyprus
5
Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
6
Department of Agriculture, Crop Production and Rural Environment, University of Thessaly, 38446 N. Ionia, Magnissia, Greece
*
Author to whom correspondence should be addressed.
Submission received: 30 December 2019 / Revised: 16 January 2020 / Accepted: 20 January 2020 / Published: 23 January 2020

Abstract

:
The current trend for substituting synthetic compounds with natural ones in the design and production of functional and healthy foods has increased the research interest about natural colorants. Although coloring agents from plant origin are already used in the food and beverage industry, the market and consumer demands for novel and diverse food products are increasing and new plant sources are explored. Fresh vegetables are considered a good source of such compounds, especially when considering the great color diversity that exists among the various species or even the cultivars within the same species. In the present review we aim to present the most common species of colored vegetables, focusing on leafy and fruit vegetables, as well as on vegetables where other plant parts are commercially used, with special attention to blue color. The compounds that are responsible for the uncommon colors will be also presented and their beneficial health effects and antioxidant properties will be unraveled.

1. Introduction

Vegetables are considered an invaluable ingredient of human diet, since they diversify color of various food products and they also possess beneficial health effects due to their content in various phytochemicals such as flavonoids, betalains and carotenoids and the overall high antioxidant capacity [1,2,3]. Consumption of purple/blue fresh produce is associated with increased nutrient intake and reduced risk for metabolic syndrome [4]. Based on food intake data from NHANES 2001–2002, the daily intake of anthocyanins was estimated to be 12.5 mg/day/person in the United States [5]. The predominant dietary anthocyanins are malvidin, delphinidin, and peonidin glycosides [6], which can be found in many plant foods, including berries, purple sweet potatoes, grapes, and wine [7]. In comparison with other flavonoids, anthocyanins possess a positive charge on their C-ring, which leads to different colors in response to various pH [8].
Anthocyanins not only have aesthetic importance by generating characteristic purple, bluish, orange, and reddish pigments in various plant tissues [9], but also have biological functions including protective effects against radiation, reactive oxygen species scavenging, defense against pathogens and stress conditions, and attracting seed and pollen dispersers [10,11,12]. These compounds also have nutritional value and potential health benefits [13].
Leafy vegetables are widely consumed throughout the world and they significantly contribute to the overall recommended daily intake for several nutrients essential for human body [14]. They include several species among which lettuce is considered the most important salad vegetable and several reports highlighted its significance in human nutrition [15,16]. On the other hand, the Solanaceae family has approximately 2700 species and 99 genera and includes some of the most important fruit vegetables consumed globally. Solanum, the largest and most complex genus in this family, is of great economic importance with several species used as foods, medicines, and ornamental plants [17]. In other popular vegetables consumed worldwide such as broccoli, cauliflower, and artichoke, the immature inflorescence constitutes the edible portion and represent a rich source of flavonoids, anthocyanins, and other bioactive compounds that are also responsible for their pigmentation [18,19,20,21]. In the case of sweet corn, another popular vegetable used for fresh consumption as well as for canned and freezing processing, differing from the regular corn only for a higher accumulation of sugar in the kernels, the ear including cob and kernels constitute a rich source of natural colorants including carotenoids and flavonoids [22]. Yet, in other cases like asparagus the edible portion and source of anthocyanins is constituted by the young stems [23].
Recently we published a review paper regarding colored root vegetable species focusing on the most important coloring compounds and their antioxidant effects. With the present review we aim to continue this work and present the rest of colored vegetable crops, focusing on leafy and fruit vegetables and relevant species where other plant parts are consumed. Having in mind the same context, the main coloring compounds are highlighted, while a special section for each species is allocated to their health effects and antioxidant properties. The information presented in this review was systematically gathered from scientific databases such as Scopus, ScienceDirect, PubMed, Google Scholar, and ResearchGate by using various keywords and key phrases, e.g., the common and Latin of the main species and/or the terms “health effects”, “antioxidant compounds”, “colored leafy vegetables”, “colored fruit vegetables”, “blue vegetables”, “purple vegetables”, and “anthocyanins”.

2. Leafy Vegetables

2.1. Lettuce

Lettuce (Lactuca sativa L.) belongs to the Asteraceae (Compositae) family and is a very popular vegetable crop used for fresh consumption and as salad ingredient owing to its sensory and health-promoting properties [14,24,25,26]. Lettuce is widely cultivated throughout the globe and it is rightly considered the most important of leafy greens as it ranks highest in production (27 million tons in 2017; [27]). Lettuce comes in a wide variety of head formations, textures, sizes, leaf shapes, and colors and it is conventionally classified according to Mou [28] into six major groups (i.e., types): (i) Butterhead, (ii) Cos or Romaine, (iii) Crisphead, (iv) Leaf or Cutting, v) Stalk or Stem, and vi) Latin. Compared to several other leafy vegetables, lettuce is an excellent source of vitamin B9 and total flavonoids. Wang et al. [29] and Gan and Azrina [30] reported that total vitamin B9 and flavonoid contents in lettuce were higher by 16% and 220%, respectively, compared to spinach, which is another important and widely consumed leafy green. Quali-quantitative variation in lettuce vitamins and secondary metabolites (i.e., phytochemicals) depends on many pre-harvest factors such as genotype, environmental conditions, harvest maturity, and agricultural practices [31]. However, the genetic material is the predominant pre-harvest factor and the major determinant of the biosynthesis and accumulation of lipophilic (i.e., carotenoids, chlorophylls, and vitamin E) and hydrophilic (i.e., phenolic compounds and vitamin C) antioxidant molecules [32]. Vitamins are essential micronutrients required for human metabolism and functionalities implicated in the reduction of cardiovascular and degenerative diseases [33]. Folate (vitamin B9) and vitamin C (as ascorbic and dehydroascorbic acids) are eminently present in lettuce [14]. In their review article Kim and co-workers reported that folate and vitamin C contents vary with leaf type and particularly leaf coloration, with red leaf, butterhead, and romaine lettuces being particularly good sources of folate [15,29,34], while leaf green lettuce had the highest vitamin C concentration [24,25,34,35].
Carotenoids, which constitute an important group of lipophilic pigments frequently present in yellow-orange vegetables and in dark green leafy vegetables, vary in concentration among lettuce types and colors. Mou [36] assessed the genetic variability in β-carotene and lutein content, the most abundant carotenoids in lettuce, across 52 genotypes (including butterhead, crisphead, Latin, leaf, primitive, romaine, stem lettuce and wild species) that were categorized by type in the following order: Romaine and green leaf > red leaf > butterhead > crisphead. The author also reported that the two target carotenoids were significantly and positively correlated with chlorophyll a and b as well as with total chlorophyll content. Contrarily to the findings of Mou [36], Baslam et al. [37], and Nicolle et al. [25] demonstrated that the content of β-carotene and lutein may not entirely correlate with leaf green pigmentation, since the carotenoids content appeared to be lower in green compared to red-pigmented lettuce plants. The contradiction between these results may indicate that the content in carotenoids may not be consistently related to leaf pigmentation [14]. Nevertheless, the frequent consumption of carotenoids-rich lettuce could be of high importance since several epidemiological studies demonstrated that the onset of chronic diseases such as heart disease, vision impairment, and certain types of cancer (lung, prostate, and colon) could be reduced [38,39,40,41].
Several authors reported that the contents of secondary metabolites in lettuce differed greatly among genotypes and depended particularly on leaf color (dark red, red, green/red, and green) [14,42]. According to Mulabagal et al. [43] red lettuce contains a single anthocyanin, namely cyanidin-3-O-(6’’-malonyl-β-glucopyranoside) which is further converted in two cyanidin derivatives (cyanidin-3-O-(6’’-malonyl-β-glucopyranoside methyl ester) and cyanidin-3-O-β-glucopyranoside), all presenting significant antioxidant activities against lipid peroxidation and cyclooxygenase activity. Kim et al. [42] explored the genetic material of 23 lettuce cultivars belonging to three major lettuce groups (crisphead, oak-leaf, and romaine) in respect to their phytochemical content and antioxidant potential. The authors reported that most phytochemicals varied significantly with genetic material and were associated mainly with leaf color. The red-leaf and to a lesser extent the green/red cultivars exhibited the highest concentration of the following antioxidant molecules: Cyanidin, carotenoids (lutein, violaxanthin and lactucaxanthin), fatty acids such as α-linolenic and linoleic acid, total polyphenols, and antioxidant potential [42]. In the same study, the authors were also able to demonstrate that the methanolic extract of red-pigmented lettuce contained potent scavengers of ABTS (scavenging assay of 2,2-azino-bis(3-ethylbenzthiazoline-6-sulphonic acid) diammonium salt radical) and DPPH (scavenging assay of 2,2-diphenyl-1-picrylhydrazyl radical) radicals. The higher radical-quenching activity of red-pigmented lettuce cultivars irrespective of type renders them more bioactive thus their systematic inclusion in the human diet could be an efficient tool to minimize the impact of oxidative stress-related diseases [25]. Similarly, Hao et al. [44], reported the differences in nutritive quality (e.g., cellulose, protein, starch, sugar, and vitamin C contents) among 74 red/purple and green varieties of leaf lettuce using grey correlation analysis (a methodology of treating and analyzing qualitative data according to grey system procedure). The authors concluded that purple-leaf lettuce “P-S23’’ exhibited significantly higher grey comprehensive evaluation value (0.8) in comparison to the green counterparts (values < 0.5). In addition, leaf pigmentation significantly correlates to the constitution and concentration of phenolic compounds belonging mainly in the subgroups of phenolic acids, flavonoids, and anthocyanins [14]. Chicoric, caffeic, and chlorogenic acids and their derivatives are the most abundant phenolic acids present in lettuce, whereas the most outstanding flavonoids include anthocyanins, quercetin, kaempferol, and flavone luteolin [35,45,46,47]. Several authors reported a higher total phenolic content in red butterhead, red leaf and red romaine lettuces compared to their green counterparts [16,35,48,49,50,51]. The red color of lettuce has been associated with a higher total phenolics content, known to impart a greater antioxidant activity than vitamins C and E [52], and has been attributed primarily to anthocyanins, an important group of flavonoids responsible for the red/purple coloration [53].
Regarding the health benefits attributed to lettuce, according to an in vivo study carried out by Lee et al. [54] on mice fed with a high-fat diet, supplementation of the diet with 8% red lettuce on a body weight (bw) basis decreased the total cholesterol and the low density lipoprotein (LDL) by 9% and 123%, respectively, thus highlighting the potential effects of red-pigmented lettuce consumption against cardiovascular disease. A putative mechanism behind the cholesterol reduction could be the synergistic effects of lipophilic and hydrophilic antioxidant molecules such as α-tocopherol, anthocyanins, β-carotene, and phenolic compounds. Similarly, Nicolle et al. [24] observed that feeding male rats with 20% of red oak-leaf lettuce decreased significantly the LDL (low density lipoproteins)/HDL (high-density lipoproteins) cholesterol ratio and the liver cholesterol content.
In addition to preclinical trials, clinical studies demonstrated that weekly consumption of lettuce was able to reduce the incidence of colorectal cancer [55]. The protective effect of frequent lettuce consumption against colorectal cancer has been attributed to the presence of β-carotene and vitamin C and not to Ca and vitamins B9 and E contents [55]. Recently, Qin [56] reported that the new cultivar B-2 of red-pigmented lettuce, characterized by high concentration of anthocyanins, flavones, and phenolic acids, can minimize oxidative stress-related diseases, leading to anti-tumor effects against human lung adenocarcinoma, hepatoma, and human cancer colorectal adenoma cell lines. Based on the above considerations, clinical and preclinical studies demonstrated that frequent consumption of fresh lettuce, in particular the red-pigmented varieties, carries potential anti-diabetic, cholesterol lowering, and anti-tumor properties.

2.2. Basil

Basil (Ocimum basilicum L.) belongs to the Lamiaceae family and it is one of the most important aromatic herbs cultivated worldwide as it flourishes under a wide range of climatic conditions. Basil cultivated both in open-field and under greenhouse conditions is an essential ingredient of renowned pesto sauce (widely consumed in Italy), while it is also used for fresh consumption and as a culinary spice [57]. In addition, the extraction of essential oils from basil is of high interest to both cosmetic and pharmaceutical industries. Basil comes in a wide variety of types, conventionally classified into seven morphotypes: (i) Large-leafed “Italian” basil, (ii) tall, slender basil, (iii) dwarf “Bush” basil, (iv) compact “Thai” basil, (v) purple basil (with clove-like aroma), (vi) citriodorum basil (flavored types), and (vii) purpurascens basil (sweet purple-colored basil) [58]. The herbs of the Lamiaceae family, such as basil, are characterized by strong antioxidant capacity. Basil in particular is a rich source of phenolic compounds, including phenolic acids such as rosmarinic, caffeic, chicoric and caftaric acids [59,60,61], vitamin C, and carotenoids such as lutein and β-carotene [62]. Furthermore, certain purple/red cultivars also have important concentrations of the hydrophilic anthocyanins, especially the “Dark Opal”, “Purple Ruffles”, and “Rubin” cultivars” [61,63].
The main carotenoids detected in basil are mostly lutein and β-carotene; but Calucci et al. [64] ranks basil first among aromatic herbs with respect to the concentrations of xanthophyll carotenoids. In the study of Kopsell et al. [62], the main detected carotenoids in sweet basil were identified as lutein, β-carotene, and zeaxanthin, and significant differences in carotenoid profiles were observed between different growing conditions (open-field versus greenhouse) and cultivars (“Cinnamon”, “Genovese”, “Italian large leaf”, “Nufar”, “Osmin purple”, “Red Rubin”, “Spicy bush”, and “Sweet Tai”). According to Marchand et al. [38] and Johnson et al. [40], the frequent consumption of vegetables and herbs was more strongly correlated with reduced risk of certain types of cancer and degenerative ophthalmic diseases, in comparison to the ingestion of monomolecular carotenoid supplements. In addition to carotenoids, basil is considered also an important source of vitamin C. It is well established that vitamin C is crucial for immune and antioxidant functions, and according to the WHO, 80–90 mg of vitamin C should be ingested daily [65]. However, due to the water solubility of ascorbic acid, regular dietary intake is essential to normal metabolic functioning [66]. According to Murarikova and Neugebauerova [65] the ascorbic acid content varied from 34.3 to 220.0 mg/kg fresh weight among the tested varieties (“Dark Green”, “Lettuce Leaf”, “Mammolo Genovese”, “Manes”, “Ohre”, “Purple Opal”, and “Red Rubin”) and the different growing seasons.
Rosmarinic acid is noted in the scientific literature as the most abundant phenolic constituent of basil [59,60]. On the other hand, a study carried out by Kwee and Niemeyer [61] showed that 9 over 15 basil cultivars contained other caffeic acid derivatives, such as chicoric acid, in higher concentrations than rosmarinic acid. Moreover, the antioxidant and antimicrobial properties of “Napoletano” green and purple basil were analyzed by Tenore et al. [67], who reported that the main phenolic acids in purple basil were rosmarinic, ferulic and gallic acid; while in green basil, the most abundant phenolic constituents were gallic acid, followed by rosmarinic and ferulic acids. Interestingly, the functional molecule rosmarinic acid detected in “Napoletano” type basil was by far higher than what on average has been reported for other common varieties such as “Sweet basil”, “Thai basil”, “Genovese Italiano”, and “Purple Petra” (112, 128, 117, and 352 per 100 g fresh weight, respectively) [59]. The main anthocyanins detected in purple basil extract were cyanidin-based p-coumaril and malonil acids, acting as powerful antioxidants with potential use as medicinal agents [68]. Tenore and co-workers also demonstrated in the same study that extracts of “Napoletano” green and purple basil both had a broad antimicrobial spectrum able to reduce the growth of all human pathogenic and food spoilage bacteria and molds tested [67].
Regarding the health-promoting effects of basil, several preclinical and clinical studies showed that extracts from basil, particularly the purple one, may alleviate hyperglycemia associated with type 2 diabetes [69,70]. The anti-diabetic beneficial effects of basil extract may be due in part to catechin and especially to rosmarinic acid, which has been found to inhibit key enzymes such as α-amylase, α-glucosidase, and aldose reductase [71,72,73]. In addition to being anti-diabetic, basil extract may also be an efficient tool against hyperlipidemia by effecting lower cholesterol and triglyceride levels in the blood [74,75]. Reduced uptake of lipids and lower values of total cholesterol and low density lipoprotein may reduce the risk of cardiovascular diseases.

2.3. Perilla

Perilla (Perilla frutescens L. Briit) belongs to the Lamiaceae family (formely Labiateae) which consists of 235 genera and more than 700 species [76]. Perilla is an edible herb widely consumed in Asian countries such as China, Korea, Japan, and India. Similar to spinach, perilla leaves are also characterized by a high concentration of carotenoids. In fact, according to Müller-Waldeck et al. [77], perilla may contain high contents of carotenoids, especially up to five-fold higher lutein than other carotenoid-rich leafy vegetables. The leaves of perilla contain a range of bioactive phenolic molecules such as caffeic acid, catechin, chrysoeriol, ferrulic acid, luteolin, quercetin, and rosmarinic acid [78,79]. In particular, secondary metabolites such as rosmarinic acid and perillaldehyde (an essential oil constituent) have demonstrated potential to prevent a wide range of diseases particularly owing to their anti-diabetic, anti-depressant, anti-bacterial, anti-cancer, and antimicrobial properties [76]. Thus the concentrations of these two phytochemicals are crucial for their clinical and culinary applications. It is worth noting that these two secondary metabolites are produced in perilla by two different biosynthetic pathways, namely the monoterpene and phenylpropanoid pathways, and may increase independently in relation to the perilla chemotype and abiotic environmental stress conditions [80]. Perilla is present in nature in two main chemical-varietal phenotypes: (i) The red-pigmented cultivar P. frutescens var. crispa, known as “Zi-So” and widely grown in China where it is used as a spicy herb, leafy vegetable, and medicinal plant, and (ii) the non-pigmented green cultivar P. frutescens var. frutescens known in Japan as “Shisoyo” or “Shiso” and mainly used as an oil crop but also as ingredient of skin creams and food products [81,82]. According to Meng et al. [78,83], three cinnamic derivates (caffeic acid, coumaroyl tartaric acid, and rosmarinic acid) ranged from 0.1 to 11 mg/g; six flavonoids (apigenin 7-O-caffeoylglucoside, apigenin 7-O-diglucuronide, luteolin 7-O-diglucuronide, luteolin 7-O-glucuronide, scutellarein 7-O-diglucuronide, and scutellarein 7-O-glucuronide) ranged from 3.5 to 18.5 mg/g; and six anthocyanins (0.7–2 mg/g) including cis-shisonin, cyanidin 3-O-(E)-caffeoylglucoside-5-O-malonylglucoside malonylshisonin and shisonin were detected on eight tested cultivars of perilla. Concerning the health effects of perilla extract, Narisawa et al. [84] reported that perilla leaves carry anti-tumor properties. In their work, the authors showed that treatment of female rats with a 12% fat diet based on perilla extract and safflower oil in 1:3 or 1:1 ratio effected better protection against colon cancers as compared to safflower oil alone [84].
Comparing green (Korean cultivar) and red-pigmented (Japanese cultivar) perilla, Rouphael et al. [82] observed that green perilla produced exclusively perilla ketone (PK), whereas the red perilla contained perillaldehyde (PA). Similar results were reported by Martinetti et al. [81] in a study profiling two red-leaf (“Aka Shiso” and “Purple Zi Su”) and three green-leaf cultivars (“Ao Shiso”, “Qing Su”, and “Korean perilla”) with the later containing PK instead of PA. The terpenoid component present in green-pigmented perilla has been demonstrated to be toxic for cattle and horses, since PK is considered a potent lung toxin [77,85], but the health-effect as well as the toxic dose/concentration to humans is still controversial, therefore Müller-Waldeck and co-workers [77] concluded that some Korean genotypes (green-pigmented cultivars) are not suitable/recommended for fresh consumption. Interestingly, several authors showed that PA and PK present in red and green perilla can stimulate the TRPA1 (Transient Receptor Potential) cation channels which are actively involved in multiple biological mechanisms such as pain perception and their functional role in the prevention of certain types of tumor has been proved [86,87].

2.4. Swiss Chard

Green chard also known as Swiss chard (Beta vulgaris var. cicla L.) belongs to the Amaranthaceae-Chenopodiaceae family and is considered an important leafy vegetable grown for its green or reddish leaves and the white, yellow, or red leaf stalk. Green beet belongs to the same family of the root vegetable red beet (Beta vulgaris var. rubra L.). Traditionally, Swiss chard has been employed for its health-promoting properties as folk remedy for liver/kidney diseases, for triggering the hematopoietic and immune systems and also as a target diet in some tumors treatment [88]. As they grow, Swiss chard leaves accumulate a wide range of macro and micro minerals such as P, K, Ca, Mg, and Fe and several lipophilic vitamins (such as A and E and also carotenoids), as well as hydrophilic vitamins (such as B3, B5, B9, and C) [89]. According to Mzoughi et al. [90] Swiss chard leaves have a nutritional and functional profile catering to modern human diets. In the latter study, Swiss chard leaves were characterized by high concentrations of secondary metabolites such as (myricitrin, p-coumaric, and rosmarinic acid), flavonoids, carotenoids (β-carotene, chlorophyll, and lycopene) and some target volatile compounds (decanal, E-anethole, and octanoic acid). Mzoughi and co-workers demonstrated that the high antioxidant capacity on ABTS and DPPH of Swiss chard ethanol extract was accompanied with significant inhibitory effects on α-amylase and α-glucosidase; thus the Swiss chard extract could be explored in the near future as potential functional food with antioxidant and anti-diabetic properties [90].
In a recent review paper, Ninfali et al. [91] reported that green beet extract may regulate the hematic concentration of glucose, decrease lipid peroxidation, lower triglycerides and cholesterol levels, and improve glutathione levels. The health protective secondary metabolites found in B. vulgaris cicla have been identified as a class of G-Glycosyl flavonoids including (i) isovitexin, (ii) vitexin, (iii) vitexin-2-O-xyloside and iv) vitexin-2-O-rhamnoside, which are characterized by high biological activity [91]. According to Lee et al. [92], vitexin is able to reduce drastically the mitochondrial membrane potential in leukemia cell. Similarly, Nifali et al. [93] and Gennari et al. [94] reported that vitexin-2-O-xyloside and vitexin-2-O-rhamnoside were able to reduce the proliferation rate of MCF-7 breast and RKO cancer cells. Concerning the anti-inflammatory properties of Swiss chard, Borghi et al. [95] demonstrated that the administration of 10 mg/kg of vitexin is able to decrease the levels of pro-inflammatory cytokines. Overall, in vitro and in vivo experiments carried out on animals and humans demonstrated that the biological activity of vitexin, vitexin-2-O-xyloside, and vitexin-2-O-rhamnoside can trigger the expression of a wide range of genes associated with inhibition of cancer cell proliferation and anti-inflammation activities.

2.5. Brassica Leafy Vegetables

The group of brassicaceous leafy vegetables, formerly referred to as cruciferous vegetables, includes a wide range of species with potential health-promoting properties such as kale (Brassica oleracea var. sabellica), pack choi (Brassica rapa var. chinensis), mizuna (Brassica rapa var. japonica), watercress (Nasturtium officinale R.Br.), wild and salad rocket (Diplotaxis tenuifolia [L.] DC and Eruca cesicaria [L.] Cav., respectively). According to FAOSTAT [27] database, roughly 12% of the vegetables grown worldwide are members of the Brassicaceae family. Recent reports [96,97] have suggested that brassicaceous leafy vegetables constitute valuable sources of phytochemicals. They contain high levels of vitamins (C, E [as α- and γ-tocopherols] and K [phylloquinone]), carotenoids, and phenolic compounds. In addition to the latter phytochemicals, Brassica leafy vegetables are characterized by sulfur-containing glucosinolates and methylcysteinsulfoxide compounds. The genetic factor is the most important and influential one in terms of modulating the biosynthesis and accumulation of phytochemicals in Brassica leafy vegetables [97]. In a comparative study of antioxidant molecules in four Brassica leafy vegetables (mizuna, salad rocket, watercress, and wild rocket), the authors observed a large variability in phytochemical concentrations [98]. For instance, watercress showed the highest polyphenol and vitamin C content, while salad and wild rocket were characterized by high concentrations of kaempferol and quercetin derivatives and finally mizuna exhibited significant concentrations of isorhamnetin and sinapic acid [98]. The authors highlighted the potential value of salad Brassica leafy vegetables as dietary sources of antioxidants conferring a wide range of positive health effects against type 2 diabetes and cardiovascular diseases.
Kale is a leafy Brassica species considered a potent source of glucosinolates and isothiocyanates, their main breakdown products. A study from the CDC (center of disease control and prevention) reported kale being ranked 15th among “powerhouse” vegetables and fruits [99]. The most abundant glucosinolates in kale were: 3-(methylsulphinyl)propyl, 2-propenyl and also 4-(methylsulphinyl)butyl glucosinolates. Genotypic variation within eight cultivars of kale (“Starbor”, “Beira, “Scarlet”, “Premier”, “Olympic Red”, “Toscano”, “Dwarf Siberian”, and “Red Russian”) revealed that “Beira” and “Olympic Red” were characterized by the highest total concentration of glucosinolates and were proposed as functional foods [100]. In another experiment carried out by Hahn et al. [101] on 25 kale cultivars, the authors observed a great variation in glucosinolate profiles. Similarly, Ferioli et al. [102] reported higher variation in the aliphatic compared to the indole glucosinolates (9- and 5-fold, respectively) across 25 kale cultivars harvested from different European countries (Italy, Portugal, and Turkey). The presence of isothiocyanates in Brassica leafy vegetables has been reported to confer anti-diabetic, anti-inflammatory and anti-cancer properties [103,104,105,106,107,108]. Although less popular, even “ornamental cabbage” or kale (Brassica oleracea L. var. acephala DC.) have genotypes characterized by the accumulation of different pigments [109,110,111]. Brassica leafy vegetables, in particular kale, are considered additionally as a rich source of carotenoids (lutein and β-carotene), as well as chlorophylls (a and b). Carotenoid concentrations of 33 kale cultivars were analyzed and quantified [112]. Zeaxanthin was the most abundant carotenoid in 21 cultivars. Moreover, American and hybrid cultivars and accessions were characterized by high concentrations of zeaxanthin, whereas, German landraces, German commercial varieties, Italian, and red-colored kale varieties exhibited high concentrations of chlorophyll a and b [112].
Emerging market trends catering to shifting consumer perceptions of quality [32], have resulted in colored Brassica leafy vegetables (e.g., violet kale or pack choi) containing anthocyanins garnering the attention of nutritionists and horticultural scientists. Recently, Mageney et al. [112] proposed that the anthocyanin content could be used as a marker to differentiate between varieties/cultivars. Testing green and red-pigmented pack choi, Zheng et al. [113] observed that red pack choi produced higher concentrations of carotenoids, total phenolic compounds, total flavonoids, glucosinolates, and anthocyanins compared to its green counterpart. Importantly, the regular intake of anthocyanins from such colored leafy vegetables has been positively correlated with the prevention of various liver diseases, and also with the reduction of colon cancer, hepatic inflammation and oxidative stress [114].
The main pigments isolated in the studied leafy vegetables are presented in Table 1.

3. Fruit Vegetables

3.1. Tomato

Tomato (Solanum lycopersicum L.) is an important fruit vegetable, widely consumed all over the world due to its rich nutrient content, special taste, and diverse ways of consumption (fresh, soups, juices, purees, dried, and sauces) [124]. Fruit color and pigments content are two important traits that largely reflect tomato fruit quality, as well as the antioxidant activity which is mainly correlated to the hydrophilic (e.g., soluble phenolic compounds and vitamin C) than to the lipophilic compounds (e.g., carotenoids, vitamin E, and lipophilic phenols) [125]. Fruit color is mainly related to pigments content, such as chlorophyll, carotene, lycopene, phytoene and anthocyanin, and their relative proportions at different maturity stage [126]. The most abundant carotenoid is lycopene, followed by phytoene, phytofluene, ζ-carotene, γ-carotene, ß-carotene, neurosporene, and lutein [127]. Color development is due to the chlorophyll degradation and the synthesis of carotenoids as fruit is developed and ripen. Therefore, the genetic development of tomato fruit color and pigments content is an interesting research area to improve fruit quality and satisfy the diverse consumers’ demands [128].
The consumption of tomato and related food products is associated with the decrease of various diseases incidence such as chronic degenerative diseases, cardiovascular disease, and age-related macular degeneration (AMD) in human health [129]. Raiola et al. [124] reported the nutritional importance of tomato phytochemicals against inflammation processes and prevention of chronic non-communicable diseases (e.g., obesity, diabetes, coronary heart disease, and hypertension). Anthocyanins normally are not produced in tomato fruit, however, some wild tomato species, such as S. chilense, S. cheesmaniae, S. lycopersicoides, and S. habrochaites biosynthesize anthocyanins in the sub-epidermal tissue of the fruit, and some alleles from those genotypes have been introgressed into cultivated genotypes [130]. Therefore, combining the dominant Anthocyanin fruit (Aft) gene from S. chilense and the recessive atroviolacea (atv) gene from S. cheesmaniae into a cultivated tomato background, anthocyanins biosynthesis has been achieved [131,132].
Purple tomatoes have antioxidants and phytochemical properties in both flesh and peel, often in superior levels than those found in conventional red tomatoes [133]. A genetically modified (GM) purple tomato was found to have additional health-promoting effects by prolonging the life of cancer-susceptible mice compared to tomatoes with conventional (red) color [134]. Extracts from fruit of purple tomato (breeding line V118) showed significant and dose dependent anti-inflammatory effect against paw edema in an in vivo study with rat models (edema inhibition: 7.48%–13.8%), suggesting that anthocyanins may play a role in the anti-inflammatory effect [135]. Interestingly, during the last 20 years there has been an increasing interest in developing highly consumed food, such as flavonoids-rich tomato fruit. To that direction, transgenic approaches have been applied to modify the biosynthesis of phenylpropanoids, in order to alter the tomato flavonoid biosynthesis [130].
Phenolic content is varied at different developmental stages of tomatoes, as sun black (SB) tomato had 5.8 and 8.6 mg GAE/g dw phenolic content at mature green and red ripening stage, respectively, contents that were 152% and 134% higher than wild type (WT) [130]. Li et al. [136] reported similar total phenolic content (659.11 mg GAE/100 g dw) for purple tomato as has been reported for other tomato varieties (from 290 to 500 mg GAE/100 g dw) [137]. Individual components of phenolics may also vary among purple and red varieties, as the main phenolic compounds content (chlorogenic acid, naringenin, and rutin) was higher (65.56, 12.82, and 52.39 mg/100 g dw, respectively) [136] in purple tomatoes compared with red tomatoes were chlorogenic acid, naringenin, and rutin content was 2.67, 1.84, and 6.61 mg/100 g dw [138] and 16.7, 2.2, and 16.9 mg/100 g dw, respectively [139]. Apart from chlorogenic acid, naringenin, and rutin Li et al. [136] reported other phenolic compounds in purple tomatoes such as p-coumaric acid (15.68±0.74 mg/100 g dw), gentistic acid (15.25 ± 0.76 mg/100 g dw), ferulic acid (14.51 ± 0.99 mg/100 g dw), caffeic acid (13.65 ± 0.83 mg/100 g dw), and protocatechuic acid (8.95 ± 0.16 mg/100 g dw). Indeed, chlorogenic acid content depends on the developmental stage of fruit and varied from 0.5 to 1.3 mg/g dw in sun black (SB) tomato extracts [130]. Moreover, composition of flesh and peel in tomato mutants differed as flesh contained chlorogenic acid (7.96 ± 0.75 mg/100 g fw), quercetin (5.03 ± 1.02 mg/100 g fw), luteolin (0.45 ± 0.01 mg/100 g fw), and total phenolics (1.18 ± 0.06 mg/g fw), while in peel the respective compounds content was: chlorogenic acid (8.43 ± 0.15 mg/100 g fw), quercetin (5276.15 ± 15.10 mg/100 g fw), luteolin (21.28 ± 1.07 mg/100 g fw), and total phenolics (5.95 ± 0.27 mg/g fw) [133].
Among the several phytochemicals identified in plants, cis- and trans-resveratrol (3,4,5-trihydroxystilbene) are polyphenols that belong to the stilbene class; however, only the trans form is biologically active in the human body [140]. Numerous biocidal activities exerted by resveratrol have been reported such as antioxidant, antidiabetic and estrogenic activity, anticancer effects through the preservation of the regular cell cycle, the inhibition of tumor invasion and angiogenesis, and cardiovascular effects through the reduction of the expression of endothelial adhesion cells and the inhibition of cell apoptosis and platelet aggregation [141,142,143,144,145]. However, resveratrol’s daily intake by humans has still to be established [143]. Vagula et al. [146] quantified trans-resveratrol in S. americanum Mill. fruit, which ranged between 1.07 and 0.796 μg/g for fruit pulp and peel, respectively, and these levels were significantly higher when compared to freeze-stored fruit (0.1353 μg of trans-resveratrol/g of sample) and to other berries [146].
Vasco et al. [147] reported the higher antioxidant capacity of purple (purple-red variety) tamarillo or tree tomato (Solanum betaceum Cav.), compared to the golden-yellow variety and reported 9.3 μmol trolox/g fw, 3.0 μmol trolox/g fw and 40 μmol trolox/g fw for seed-jelly, pulp, and peel tissues of purple fruits compared to 3.8 μmol trolox/g fw, 2.3 μmol trolox/g fw and 22 μmol trolox/g fw for seed-jelly, pulp and peel tissues of yellow fruits, respectively. Similarly, Sestari et al. [133] reported increased antioxidant capacity (DPPH) in peel (38.12 ± 4.27 μmol trolox/g fw) compared to flesh (8.64 ± 0.45 μmol trolox/g fw) in tomato mutants. Interestingly, the oxygen radical absorption capacity assay (ORAC) value for the hydrophilic extracts in purple tomato reported by Li et al. [136] was 323.23 μmol trolox/g dw, which was 2-fold higher than the ORAC value of the traditional tomato cultivar San Marzano (140 μmol trolox/g dw) reported by Ninfali et al. [148]. Similar observations were made by Blando et al. [130] who stated 3-fold higher trolox equivalent antioxidant capacity (TEAC) value (31.6 μmol trolox/g dw) in sun black tomatoes compared with the wild type (10.3 μmol trolox/g dw) at the red-ripening stage. Moreover, antioxidant capacity of purple fruit was higher at the ripe (red ripe stage-RR) fruit compared to the unripe (mature green stage-MG) ones, probably explained by the great increase in polyphenols accumulation during ripening (from 5.8 to 8.6 mg GAE/g dw, in MG and RR, respectively) [130]. In the same study, total ascorbic acid content was higher in sun black than wild type fruits (37.3 ± 1.4 vs. 27.1 ± 1.1 mg 100/g fw, respectively) [130].
The antioxidant capacity of fruit is not only related to phenolics and ascorbic acid but also to the carotenoids, flavonoids, and anthocyanins content. Lycopene, the main phytochemical of tomatoes, is known for its important role in human health related functionalities [149]. Lycopene supplementation in an in vivo study with iodoacetamide-induced colitis rats showed reduced tissue malondialdehyde (MDA) levels, the histological signs of colon injury, and increased superoxide dismutase levels in the red blood cells [150]. In the study Li et al. [136], lycopene was the dominant carotenoid (185.01 μg/g dw) in breeding line V118, followed by β-carotene (47.11 μg/g dw) and lutein (2.66 μg/g dw). About 8.1% of the total carotenoids in V118 were cis-carotenoids, a lower value compared to that of most of the tomato varieties studied [136]. Moreover, Li et al. [128] reported that purple fruit (cv “Zi Ying”) had increased antioxidant capacity compared to green fruit (cv “Lv Ying”), with lycopene content of 36.51 ± 2.86 mg/kg fw and β-carotene 13.38 ± 1.31 mg/kg fw in purple fruit versus lycopene content of 1.35 ± 0.05 mg/kg fw and β-carotene 6.80 ± 0.32 mg/kg fw in green fruit. In a comparative study, Hazra et al. [151] pointed out the dietary role of purple tomato (AftAft dgdg genotype) due to the increased values in ascorbic acid (31.56 ± 2.41 mg/100 g fw), lycopene (6.13 ± 0.39 mg/100 g fw), β-carotene (0.65 ± 0.14 mg/100 g fw), and anthocyanin (20.73 ± 2.86 mg/100 g fw), compared to the overall mean value of 31 hybrids.
Li et al. [136] reported a total carotenoid content of breeding line V118 of 234.78 μg/g dw, being within the range of the average amounts reported for red tomatoes (132–583 μg/g dw) [152]. At the red ripe stage, total carotenoids did not differ between the sun black (SB) and wild type (WT) tomatoes; however, the β-carotene content was significant higher in the SB sample, whereas the lycopene content was lower [130]. Similarly, Vasco et al. [147] reported higher β-carotene levels in purple tamarillo than in yellow tomato variety. Generated double and triple mutants (Anthocyanin fruit/high pigment 2(Aft/hp2) and Anthocyanin fruit/atroviolacium/high pigment 2 (Aft/atv/hp2)) of purple tomatoes had higher lycopene and β-carotene levels and up to 63% of vitamin C compared to tomato cultivar Micro-Tom, suggesting accumulating trends of relevant phytochemicals in near-isogenic lines [133].
Anthocyanins, the most abundant flavonoid constituents in pigmented fruit and vegetables, posse potential health beneficial effects, such as antioxidant, anti-inflammatory, anticancer, and antidiabetic activities [153,154]. Anthocyanins also had notable effects against inflammation by inhibiting cyclooxygenase-2 (COX-2) expression, inducible nitric oxide protein and mRNA expression [155]. The “Giant” and “New Zealand” purple cultivars, had total anthocyanins content of 102.35 ± 1.46 mg/100 g dw and 168.88 ± 2.65 mg/100 g dw, but also revealed high antioxidant activity which might be related to their overall phenolic composition [156]. Zhang et al. [157] reported the role of anthocyanins in postharvest storage of tomatoes as in purple tomatoes, anthocyanins doubled the self-life of fruit by delaying over-ripening and reducing susceptibility to Botrytis cinerea.
Li et al. [136] through an LC-MS study reported three major anthocyanins, which were mainly acylglycosides of petunidin and malvidin. Among these anthocyanidins, petunidin was the predominant aglycone (91.9%), and the rest of the minor aglycones accounted for only 9.1% of the total anthocyanidins [136]. Moreover, petunidin is not usually synthesized in vegetables and fruit, and little is known about its health benefits, however in tomato mutants petunidin revealed considerable amounts (>60 mg/100 mg fw) in fruit peels of the lines combining Aft and hp2 genes [133]. The total anthocyanin content in breeding line V118 was 72.31 mg/100 g dw, including 9.04, 50.18, and 13.09 mg/100 g dw of petunidin-3-O-caffeoyl-rutinoside-5-O-glucoside, petunidin-3-O-(p-coumaryl)-rutinoside-5-O-glucoside, and malvidin-3-O-(p-coumaryl)-rutinoside-5-O-glucoside, respectively [136]. Moreover, Blando et al. [130] reported that petanin (Petunidin 3-(6-(4-(E-p-coumaroyl)rhamnosyl)glucoside)-5-glucoside (petanin)) and negretein (Malvidin 3-(6-(4-(E-p-coumaroyl)rhamnosyl)glucoside)-5-glucoside) represented 56.6% and 21.4% of the total anthocyanins content in sun black (SB) fruit peel, respectively, whereas no anthocyanins were detected in wild type (WT) tomato fruit.
The content of anthocyanins in the Del/Ros1 transgenic tomato is equally distributed within fruit, with 5.1 ± 0.5 g/kg dw being detected in the peel and 5.8 ± 0.3 g/kg dw in the flesh, but not detected in seeds [7]. These values are higher than those reported for well-known anthocyanin-rich foods such as red raspberry (3.9 g/kg dw; [158]), strawberry (3.2 g/kg dw; [158]), and mulberry (2.1 g/kg dw; [159]). In a study with transgenic plants, the predominant anthocyanins in the Del/Ros1 transgenic tomato were delphinidin-3-(trans-coumaroyl)-rutinoside-5-glucoside and petunidin-3-(trans-coumaroyl)-rutino side-5-glucoside, which contributed to nearly 86% of the total anthocyanins content, while two new anthocyanins, malvidin-3-(p-coumaroyl)-rutinoside-5-glucoside and malvidin-3-(feruloyl)-rutinoside-5-glucoside making up to 6% of the total anthocyanins content, were also reported [7]. Three mutant genes have been identified that can lead to the production of anthocyanins in the peel of the fruit, namely Anthocyanin fruit (Aft), Aubergine (abg), and atroviolacea (atv), while the Aft gene was also identified in crosses with Solanum chilense Dunal [160]. This gene is located in chromosome 10 and its presence in tomato leads to the production of anthocyanin pigments, mainly delphinidin, malvidin and petunidin, as well as to higher levels of the flavonols quercetin (3.6-fold), and kaempferol (2.7-fold), in tomato fruit [160,161].
Tamarillo crop is attracting research interest lately due to the high content in antioxidants and phytochemicals. The tamarillo, a non-climacteric edible fruit, is quite popular in local markets, especially in South America, consumed in juices or fresh and being highly appreciated due to high polyphenols levels [156], β-carotene (provitamin A), vitamin B6, vitamin C (ascorbic acid), vitamin E, and iron contents [162]. Among phenols, the presence of anthocyanins (delphinidin, cyanidin, and pelargonidin glycosides) and hydroxycinnamoyl derivatives (e.g., 3-O-caffeoylquinic acids, caffeoyl glucose and feruloyl glucose) have been described in several reports [147,163,164], while recently rosmarinic acid has been also identified [156]. The hydroxycinnamoyl derivatives show antioxidant properties and have been related to protective effects on human health [165]. In particular, the caffeoyl ester of rosmarinic acid has various biocidal activities, such as antiviral, antibacterial, anti-inflammatory, and antioxidant effects [166]. Other compounds were tentatively identified as different rosmarinic acid glucosides, caffeoyl glucoside, feruloyl glucoside, and ferulic acid dehydrodimers. Pelargonidin 3-O-rutinoside and delphinidin 3-O-rutinoside were the main anthocyanins in purple cultivars of tomato fruit [167]. Vasco et al. [147] reported anthocyanins content of 38 mg/100 g fw in purple tamarillo which was higher than previous reports in yellow fruit (8.5 mg/100 g fw) [167].
Both Solanum americanum Mill. and S. villosum Mill. are important medicinal plants of the Solanaceae family, however the blackish-purple (S. americanum) and reddish-orange (S. villosum) colored fruit are mostly consumed in India, Ethiopia, Ghana, China, and Brazil [168]. Mohy-Ud-Din et al. [169] reported the different important steroidal glycoalkaloids like β-Solamargine, α-Solamargine, Solasonine, α-Solanine, solasodine, and Solanidine, with latter being well-recognized for its anticancer activities [170].

3.2. Eggplant

Eggplant (Solanum melongena L.) fruit are very popular vegetables grown worldwide in subtropical and tropical regions [171]. They considered as one of the top 10 vegetable in terms of antioxidant capacity [172] and contain a variety of antioxidants and phytochemicals such as, ascorbic acid, phenolics, and flavonoids that provide health benefits [173]. The most abundant phenolic compound is 5-O-caffeoylquinic acid, known as chlorogenic acid (ChA), which is considered as the main contributor to the overall antioxidant capacity [174,175]. However, eggplant fruit are poor sources of provitamin A and vitamin E, with average values of 27 IU 100/g fw and 0.30 mg/100 g fw, respectively [173]. American purple fruit are the most commonly marketed type, though white cultivars have gained consumers acceptance in recent years.
Eggplants are quite versatile vegetables and could be subjected to a number of different processing and cooking methods which may further affect fruit antioxidant capacity [176]. Akanitapichat et al. [173] reported that the antioxidant activities of eggplant were correlated (r = 0.531–0.796) with the total amounts of phenolics and flavonoids. Significant correlation was found between hepatoprotective activities and total phenolics/flavonoids content (r = 0.637–0.884) and antioxidant activities (r = 0.585–0.958), indicating the contribution of the polyphenols present in eggplant to its hepatoprotective effect (human hepatoma cell line HepG2) against tert-Butylhydroperoxide (t-BuOOH)-induced toxicity [173]. Akanitapichat et al. [173] reported that total phenolics content in purple fruit was of 1002.67 ± 8.33 mg GAE/100 g extract. Nisha et al. [176] suggested a higher content of total phenolics and anthocyanins in a purple small-sized fruit variety (106.98 mg/100 g fw and 0.756 mg/100 g fw, respectively) than the three other examined varieties (purple moderate-sized fruit (80.31 mg/100 g fw and 0.525 mg 100/g fw, for total phenolics and anthocyanins, respectively), green long-sized (50.79 mg/100 g fw and 0.0475 mg/100 g fw, for total phenolics and anthocyanins, respectively), and purple big-sized (49.02 mg/100 g fw and 0.53 mg/100 g fw, for total phenolics and anthocyanins, respectively)). In the same study, it was reported that purple small-sized variety revealed the greatest antioxidant activity.
Flavonoids represent only about 10–15% of total phenolics [177] and hydroxycinnamic acid derivatives, particularly free chlorogenic acid is the major phenolic antioxidant regardless of the genotype [177,178]. However, the chlorogenic acid content in eggplant is influenced by both genetic and environmental factors, including the fruit developmental stage, cultivar, and crop and postharvest management [179]. For example, the chlorogenic acid content was 16% higher in purple eggplants compared to white eggplant slices [180]. According to Akanitapichat et al. [173] who compared five eggplant varieties, the purple fruit had higher total flavonoids content (3954.2 ± 6.06 mg catechin equivalents-CE/100 g extract) content compared with green and white ones. Sadilova et al. [181] reported that flavonoids isolated from S. melongena showed potent antioxidant activity against chromosomal aberrations induced by Doxorubicin. The purple eggplant had high antioxidant activity of DPPH and ABTS with EC50 of 66.74 ± 4.60 μg/mL and 53.18 ± 0.71 μg/mL, compared with green and white varieties [173].
In purple-fruited genotypes of pepper and eggplant the abundance of anthocyanin levels is superior in unripe fruits and decrease upon ripening, often to complete disappearance [182]. It is noteworthy that eggplant fruit reaches its commercial maturity long before its physiological ripeness, as practically it is harvested at the immature fruit stage [183]. The anthocyanin concentration in the purple fruit eggplant cultivars is higher in comparison to other deeply colored fruits and vegetables, e.g., 2.34-fold that of grapes, and 7.08-fold that of red onions [5]. In purple pigmented eggplants, the antioxidant anthocyanins (delphinidin derivatives) is limited as found at the peel tissue which represents less than 5% of the total fruit weight [178,184]. Examining the anthocyanins content of wild type (WT), purple-black (S9-1), green (L6-4), and white (U36-1) eggplants, Xi-Ou et al. [185] found that the anthocyanin content of purple-black (S9-1) was higher than that in WT eggplant, while green eggplant (L6-4) had the lowest levels of anthocyanins. In another study, Zhang et al. [186] reported the total anthocyanins content from purple eggplant (cv Zi Chang) skin to be 1.24 mg/g dw. Moreover, nasunin (delphinidin-3-(p-coumaroylrutinoside)-5-glucoside), an anthocyanin isolated from the skin of purple eggplant fruit, is associated with both inhibition of hydroxyl radical generation and superoxide scavenging activity [173,187].
Extracts from eggplant fruit skin were demonstrated to possess high capacity in scavenging of superoxide free radicals and inhibiting hydroxyl radical generation by chelating ferrous iron [187]. Additionally, eggplant extract resulted in hypolipidemic activity in rats fed normal as well as high fat diets [188], suppressed tumor growth and metastasis [189], and inhibited inflammation that can lead to atherosclerosis [190]. Not only fruit but various parts of the plant are useful in the treatment of inflammatory conditions, cardiac debility, neuralgia, ulcers of nose, cholera, bronchitis, and asthma, while they possess analgesic and hypolipidemic properties [191]. S. melongena is also a natural source of vitamin A affecting the eye health in children [192].

3.3. Pepper

Pepper (Capsicum spp.) is one of the oldest domesticated and utilized crops and the genus Capsicum consists of approximately 31 species of which the five domesticated species are C. annuum, C. baccatum, C. chinense, C. frutescens, and C. pubescen [193]. Average world production and cultivated area of dry and green peppers are estimated at 3.9 and 34.5 million tons respectively, harvested from 1.8 and 1.9 million hectares respectively [27]. Pepper fruit have high nutritive value, as they are rich in vitamin C (ascorbic acid), provitamin A (β-carotene), vitamin E (tocopherols), flavonoids and capsaicinoids, and other carotenoid pigments such as lycopene and zeaxanthin [194]. The noticeable level of phenolic compounds and carotenoid pigments also contributes to the antioxidant properties of sweet pepper [195].
Immature fruits are usually colored in white, green, purple, and black shades and gradually color changes to yellow, orange, red and brown as fruit maturity advances [196,197]. The differences in fruit color is mainly due to the differential accumulation of flavonoids and carotenoids [198]. Anthocyanin accumulation in the outer epidermis of immature pepper fruit is responsible for the purple or black color at 30 d after anthesis [199] and turns to red color at 50 d after anthesis [182].
Liu et al. [182] compared the flavonoids biosynthesis at 30 d after anthesis for green, white, and purple varieties, and reported that anthocyanins, flavones, and flavonols content was significantly higher in purple variety than in the other varieties, with delphinidin, luteolin, chrysoeriol and quercetin derivatives being the most abundant polyphenols. Delphinidin, cyanidin, and malvidin derivatives were the major anthocyanins in colored peppers among the 16 anthocyanins detected of which delphinidin 3,5-diglucoside and delphinidin 3-O-rutinoside were specifically accumulated in purple peppers [182]. In the same study, it was reported that the purple color of fruit is related to the high accumulation of cyanidin and delphinidin derivatives at 30 d after anthesis.
In chili peppers, anthocyanins’ presence and pigmentation of purple or black and magenta is also possible, and is usually found in flowers, fruit, and foliage [200]. Several reports highlighted the presence of anthocyanins in chili pepper fruit, but so far delphinidin is the only anthocyanidin identified [200,201]. Similarly, Sadilova et al. [181] reported delphinidin-3-trans-coumaroylrutinoside-5-glucoside (nasunin 89%) and delphinidin-3-cis coumaroylrutinoside-5-glucoside (4.6%) as the main anthocyanins (averaged at 320 μg/g fw) in German chili pepper (C. annuum L.), while similar results were found in two Mexican chili peppers [202]. Moreover, hydroponically grown dark violet pepper (cv. “Zorro”) had the highest concentration of quercetin and catechin when compared to orange, red and yellow fruit cultivars [203].
Capsanthin-capsorubin synthase (CCS), as a unique enzyme in pepper and tiger lily, converts antheraxanthin and violaxanthin into capsanthin and capsorubin, respectively [204]. Although the exact mechanism is under investigation, Liu et al. [182] reported that the highly active CCS drives antheraxanthin to be converted into capsanthin in purple fruit, which reduces the flux to violaxanthin, eventually resulting in significantly lower levels of antheraxanthin and violaxanthin in purple than in green and white fruit varieties.

3.4. Lablab and Common Bean

Lablab (Lablab purpureus L.), an ancient legume species, serves as a vegetable and is widely cultivated throughout the tropics, subtropics and temperate zones [205]. Fruit are green pods, 6 cm long by 2 cm wide, flattened, contain 4–5 seeds and turn light brown-purple when mature [206]. Al-Snafi [207] and Momim et al. [208] reviewed the phytochemical properties of lablab and its medicinal importance, exhibiting antidiabetic, anti-inflammatory, analgesic, antioxidant, cytotoxic, hypolipidemic, antimicrobial, insecticidal, hepatoprotective, antilithiatic, antispasmodic effects. Moreover, Momim et al. [208] and Deoda et al. [209] reported that the juice derived from the fruit pods was used as astringent, digestive, stomachic, to expel worms and for the treatment of inflamed ears and throats. Soetan [210] studied the pharmacological potentials of three varieties (“Rongai brown”, “Rongai white”, and “Highworth black”) of L. purpureus seeds and showed that raw and aqueous extracts contained various phytochemicals including trypsin inhibitors, hemagglutinin, cyanogenic glycosides, oxalates, phytates, tannins, and saponins, with greater contents in raw material compered to aqueous extracts. Other biocidal effects have been reported including antilithianic activity [209], hepatopreotective effects [211], and inhibited trypsin and plasmin activity [212].
Momim et al. [208] reported significant antioxidant capacity (DPPH) with the lowest IC50 found in purple lablab compared to the white one (430.00 μg/mL vs 853.13 μg/mL). In the same study, total flavonoids content in purple fruit was 32.09 ± 0.36 mg quercetin equivalent/g fw while in green lablab it was 42.55 ± 5.77 mg quercetin equivalent/g fw. Bhaisare et al. [206] reported vitamin C content of 81.00 ± 0.16 mg/g and vitamin E content of 73.66 ± 0.08 mg/g in fresh bean seeds of L. purpureus.
Total content of anthocyanins in purple (cv. “Hong Fu”) pods was about 1.58 mg/g, while low amounts were detected in green (cv. “Qing Feng”) ones. Compared to green pods, five kinds of anthocyanins (malvidin, delphinidin, and petunidin derivatives) were found in purple pods by HPLC-ESI-MS/MS and the major compounds were identified as delphinidin derivatives [213]. Besides, nine kinds of polyphenol derivatives, namely quercetin, myricetin, kaempferol, and apigenin derivatives were detected by UPLC-ESI-MS/MS and the major components were quercetin and myricetin derivatives [213].
Common bean (Phaseolus vulgaris L.) is another legumes species with pods of varied colors, including black, red, blue, and violet [214,215]. Anthocyanins content may vary significantly depending on the genotype, while polyphenols content is highly associated with the antioxidant activities of pods [216]. Tsuda et al. [217] reported that pelargonidin-3-glucoside, cyanidin-3-glucoside, and delphinidin-3-glucoside isolated from P. vulgaris (black bean) seed coat, as well as their standard aglycones, have strong antioxidative activity in a liposomal system and reduced formation of malondialdehyde by UVB irradiation [217]. According to Mazewski et al. [218], purple beans contain mostly condensed tannins which are responsible for the antiproliferative activities against human colon cancer cell lines (HCT-116 and HT-29).

3.5. Pepino

Pepino (Solanum muricatum Aiton), a close relative to tomato and potato, is an herbaceous Andean domesticated species grown for its juicy, sweet, and aromatic fruit, with increasing commercial and export interest in South America from exotic fruit markets [219]. Fruit color may be white, cream, yellow, maroon, or purplish [220]. Unripe pepino fruit is green while 51 days from fruit set, newly-acquired purple stripes appear, resulting in fruit softening along with decreases in total pectin and hemicellulose content [221].
Various health benefits were revealed for pepino, including treatment of diabetes, stroke, high blood pressure, heartburn (indigestion), cancer, kidney, constipation, and hemorrhoids [220], activities mostly attributed to the significant amounts of vitamin C, carotenoids, and phenolics [222]. Moreover, Hsu et al. [222] reported the antioxidative, anti-inflammatory, and antiglycative effects of pepino extract. In the same study, aqueous and ethanol extracts had similar content of total phenolic acids (averaged at 1145 mg/100 g dw) but aqueous extracts were richer than ethanol extracts in terms of ascorbic acid (43.8 vs. 6.6 mg/100 g dw), total flavonoids (875 vs. 461 mg/100 g dw), cinnamic acid (75.7 vs. 23.0 mg/100 g dw), ferulic acid (82.3 vs. 11.8 mg/100 g dw), rosmarinic acid (47.2 vs. 8.4 mg/100 g dw), quercetin (126.5 vs. 90.3 mg/100 g dw), and naringenin (57.2 vs. 14.7 mg/100 g dw) [222].
The main pigments isolated in the fruit vegetables are presented in Table 2.

4. Other Vegetables

4.1. Broccoli and Cauliflower

Broccoli (Brassica oleracea L., var. italica Plenk) and cauliflower (B. oleracea L., var. botrytis) are the two most popular vegetable crops belonging to the Brassicaceae family. Native of the Mediterranean Basin, both species are adapted to a wide range of environmental conditions and are cultivated in all five continents, with an annual production that reached about 26 million tons in 2017, from an estimated harvested area of over 1.39 million hectares worldwide [27]. Primarily known and appreciated for their typical organosulfur compounds [224,225,226], broccoli, cauliflower, and other Brassica species are also a rich source of anthocyanins which are responsible of the purple pigmentation of some varieties [18,227]. Typically green, some cultivars and populations of broccoli are characterized by a purple pigmentation of the sepals of the inflorescence [227,228]. Branca et al. [228] found high levels of anthocyanins in a Sicilian broccoli landrace called “Broccolo nero” (Black broccoli) grown around Mount Etna and characterized by a dark violet pigmentation of the inflorescence, stem, and leaf midribs. In a recent work, Yu et al. [227] identified and mapped a major locus and two minor loci associated with the purple sepal trait in broccoli, while the authors hypothesized that the development of purple color may be induced by cold temperatures. In another recent study, Rahim et al. [229] working on the hypocotyl of young green and purple broccoli seedlings identified seven putative candidate genes (BoPAL, BoDFR, BoMYB114, BoTT8, BoMYC1.1, BoMYC1.2, and BoTTG1) responsible for the biosynthesis of anthocyanins. Among those, BoTT8 was expressed considerably more in purple hypocotyl compared to the green ones. Testing the in vitro cytotoxic effect of Sicilian black broccoli stem and leaf extracts at different concentrations (0.05%, 0.1%, 0.5%, 1%, and 5%) against HT29 (colon cancer) and A2058 (melanoma cancer) cells after 24 h treatment in presence or not of the myrosinase enzyme (responsible for the hydrolyzation of glucosinolates), Terzo et al. [230] found that the juice was less toxic in presence of myrosinase especially at higher concentration of the extract (1%–5%), suggesting that factors other than the glucosinolate content, such as polyphenols (including anthocyanins) could be responsible for the cytotoxic effects against HT29 and A2058 cancer cells. Examining the anthocyanin profile of three cultivars of heat-tolerant purple sprouting broccoli, Rodríguez-Hernández et al. [231] found that cyanidin 3-O-diglucoside-5-O-glucoside derivatives were the major acylated anthocyanins, and each cultivar and plant portion (leaves, inflorescence) had a particular prominent acylated anthocyanin. The same study revealed that compared to green broccoli cv. “Marathon”, purple sprouting broccoli was characterized also by higher levels of glucosinolates. Similarly, Verkerk et al. [232] observed exceptionally high glucoiberin content (396.5 µmol/100 g FW) in purple sprouting broccoli cv “Bordeaux” compared to other green broccoli genotypes, which suggests that there is some sort of interaction between purple pigmentation and glucosinolate profile of broccoli. In another study, analyzing the acylated anthocyanin profile of purple sprouting broccoli and three other green broccoli varieties at the sprouting stage, Moreno et al. [233] observed a significantly higher content of anthocyanins in the purple genotype compared to the green ones and observed that the quantity and quality of anthocyanin pigments were highly variable among the tested genotypes. Out of seventeen anthocyanins identified in the four genotypes only three isomers were predominant in all the genotypes examined: Cyanidin 3-O-(acyl)diglucoside-5-O-glucoside, cyanidin 3-O-(acyl1)(acyl2)diglucoside-5-O-glucoside, and cyanidin 3-O-(acyl1)(acyl2)diglucoside-5-O-(malonyl)glucoside. The purple sprouting genotype was characterized by a higher content of cyanidin 3-O-(sinapoyl)(sinapoyl)diglucoside-5-O-glucoside, cyanidin 3-O-(sinapoyl)diglucoside-5-O-glucoside, cyanidin 3-O-(feruloyl)diglucoside-5-O-glucoside, cyanidin 3-O-(sinapoyl)(feruloyl)diglucoside-5-O-(malonyl)glucoside, and cyanidin 3-O-(sinapoyl)(sinapoyl)diglucoside-5-O-(malonyl)glucoside (Table 3). In agreement with previous studies, Moreno et al. [233] concluded that broccoli sprouts could be an excellent source of bioactive compounds rich of flavonoids, including acylated anthocyanins, along with glucosinolates, vitamins, and minerals [234,235,236], and that future studies should evaluate the potential of further enhancing the content of bioactive compounds in broccoli sprouts.
In the case of cauliflower, while most of the cultivars have been traditionally selected for their white curds [237], many local landraces and commercial cultivars are characterized by colored heads with characteristic pigmentation ranging from green to dark violet. In Italy, green cauliflowers are traditionally grown in Lazio and Marche, while dark violet selections are typically grown in Sicily, Puglia, and other Southern regions characterized by high levels of solar radiation which make it more challenging to produce white curds as traditionally required by the European market [228,238]. Lately, the interest for colored cauliflower varieties substantially increased due to the potential health-beneficial properties of the phenolic compounds that provide the pigmentation of plant tissues [239]. Anthocyanins are in fact responsible for the purple–violet pigmentation also in the case of cauliflower and are considered highly beneficial for human health [8]. The biosynthesis of anthocyanins in cauliflowers is regulated mainly at transcriptional level, and in a particular purple cauliflower mutant it has been demonstrated that the tissue-specific activation of the gene BoMYB2 up-regulated the expression of both BobHLH1 and BobHLH2, leading to the formation of a complex regulation network MYB–bHLH–WD40 (MBW), consisting of MYB, basic Helix-Loop-Helix (bHLH), and WD40 proteins, which in turn activates the structural genes responsible for the biosynthesis of anthocyanins [18,240]. β-carotene accumulation has been also observed in cauliflower curds due to a rare carotenoid gene (Or orange) mutation that activates the biosynthesis of carotenoids in tissues that otherwise would be white [241,242]. Nevertheless, such mutation received limited attention at commercial level and is more relevant to advance our understanding of the carotenoid biosynthesis regulation [243,244].
Analyzing by LC–MS/MS nine Sicilian landraces of violet cauliflower, Scalzo et al. [19] identified cyanidin-3-(6-p-coumaryl)-sophoroside-5-glucoside as the main anthocyanin along with p-coumaryl and feruloyl esterified forms of cyanidin-3-sophoroside-5-glucoside. Scalzo et al. [19] and Kapusta et al. [245] examined also the stability of anthocyanins after processing (blanching, microwave-heating, convection steaming and freezing, and conventional water cooking) and observed substantial changes with the formation of isomers from cyanidin-3-sophoroside-5-glucoside rather than the hydrolysis of anthocyanins, suggesting good stability especially after microwave-heating which could be interesting for food processing applications.

4.2. Cabbage and Kale

Among the Brassica species, cabbage (Brassica oleracea L. var. capitata) and Savoy cabbage (B. oleracea L. var. sabauda L.) are other two popular cole crops grown all over the world for their “heavy” heads constituted by leaves surrounding the terminal buds and that can be green or red-purple [246]. As for broccoli and cauliflower, the pigmentation of red cabbage genotypes is due to the accumulation of anthocyanins. Comparing four green and four red cabbage genotypes, Yuan et al. [247] observed that the structural genes involved in the biosynthesis of anthocyanins (CHS, F3H, F30H, DFR, LDOX, and GST), were steadily up-regulated in red genotypes for the entire growing period. The same authors observed that the expression of the structural genes responsible for the biosynthesis of anthocyanins was up-regulated in correspondence of nitrogen and phosphorous deficiency. Consistently with the mechanism of transcriptional regulation observed in purple cauliflower, in correspondence of the structural gene up-regulation it was observed a simultaneous increase of the transcript levels of the bHLH gene BoTT8, and of the MYB transcription factor BoMYB2. In a recent study analyzing the gene associated with the purple pigmentation of ornamental cabbage characterized by green external leaves and inner purple leaves, Jin et al. [109] found that phytoregulators such as abscisic acid (ABA) and ethylene (ET) play a key role in promoting the biosynthesis of anthocyanins. The same study identified 14 and 19 putative candidate genes involved in the biosynthesis of ABA and ET, respectively, and among those two ABA-biosynthesis related genes (BoNCED2.1, BoNCED2.2) and two ET-biosynthesis related genes (BoACS11, BoACO4) were expressed significantly more in purple leaves than in green leaves and were strongly correlated with the total anthocyanin content of the purple inner leaves.
Analyzing the anthocyanin profile of red cabbage using HPLC/DAD-ESI/Qtrap MS, Arapitsas et al. [248] separated and identified up to 24 anthocyanins all characterized by cyanidin as aglycon, mono- and/or di-glycoside, non-acylated, or acylated with aromatic and aliphatic acids. Similarly, using HPLC-DAD-MS/MS, Wiczkowski et al. [249] identified twenty cyanidin derivatives, with cyanidin-3-diglucoside-5-glucoside as the base structure, and cyanidin-3-diglucoside-5-glucoside, cyanidin-3-(sinapoyl)(sinapoyl)-diglucoside-5-glucoside, and cyanidin-3-(p-coumaroyl)-diglucoside-5-glucoside were the most abundant non-acylated anthocyanins. Moreover, Koss-Mikołajczyk et al. [250] identified nineteen different cyanidin derivatives, with cyanidin-3-(feruloyl)-diglucoside-5-glucoside and cyanidin-3-(sinapoyl)(sinapoyl)-diglucoside-5-glucoside having been the most predominant. Similar results were obtained by other authors, who however using different analytical procedures and equipment identified a lower number of anthocyanins [19,251].
Analyzing raw and pickled red cabbage, consistently with other studies McDougall et al. [252] identified eighteen anthocyanin structures, most of which had cyanidin-3-diglucoside-5-glucoside as the core structure non-acylated, mono-acylated or di-acylated with p-coumaric, caffeic, ferulic and sinapic acids, but pelargonidin-3-glucoside and new cyanidin-3-O-triglucoside-5-O-glucoside di-acylated with hydroxycinnamic acids were also identified. The same authors examining the stability of anthocyanins after simulated gastrointestinal digestion found that anthocyanin structures were quite stable, and acylated structures were markedly more stable than non-acylated anthocyanins, nevertheless the after-digestion total recovery of anthocyanins was about 25%.
Feeding twelve volunteers with increasing doses (100, 200, and 300 g) of steamed red cabbage containing 1.38 μmol of anthocyanins/g (containing 30 acylated and 6 non-acylated anthocyanins), Charron et al. [253] evaluated the red cabbage anthocyanin bioavailability analyzing the excretion of intact and metabolized anthocyanin compounds in the urine. After 24 h, from the excreted urine were recovered 3 non-acylated and 8 acylated intact anthocyanins and 4 glucuronidated and methylated anthocyanin metabolites. Overall, the recovery of anthocyanins in excreted urine was four times higher for non-acylated compared to the acylated anthocyanins.
Comparing the biological activities (antioxidant, cytotoxic, anti-genotoxic, and influence on enzymatic activities) of the extract of green and red cabbage Koss-Mikołajczyk et al. [250] found that the anthocyanin content and profile was highly correlated with the antioxidant capacity of tested plant extracts measured through different spectrophotometric assays (ABTS, FC, DPPH, and FRAP), and by testing the cellular antioxidant activity. Instead, all the other biological activities tested were not correlated with the content of neither anthocyanins nor glucosinolate derivatives, suggesting that the food matrix effect may be more relevant than the biological activity of the single compound. This aspect should be further examined considering that other cabbage-like vegetables may be subject to different processing and anthocyanins and glucosinolates may have different levels of stability depending on the type of thermal or non-thermal processing and even the effect of the food matrix may change [19,254].

4.3. Artichoke

Native of the Mediterranean Basin and domesticated in Southern Italy during the Roman Empire [255,256], artichoke [Cynara cardunculus L. var. scòlymus (L.) Fiori] or globe artichoke constitutes a rich source of bioactive compounds and an important component of the Mediterranean diet to which are attributed a number of medicinal properties [20,21]. Artichoke is a major vegetable crop gaining popularity as a natural functional food. Grown on about 122,390 ha worldwide, over 1.5 million tons of artichoke heads were produced in 2017 [27]. As traditional producer and consumer of artichokes, Italy (33.1% of globe artichoke harvested area), Spain (13.4%), France (6.2%), and a few other European countries in the Mediterranean area continue to dominate the production of artichoke at global level. Nevertheless, their share is decreasing as the cultivation of this crop and its consumption are gradually expanding in other regions and reached important land investment in Egypt (10,159 ha), Algeria (5532 ha), Tunisia (3687 ha), Turkey (2994 ha), and Morocco (2923 ha) within the Mediterranean Basin, westward in Perú (8646 ha), Argentina (4472 ha), United States (2914 ha), and Chile (1464 ha), as well as eastward, especially in China (11,803 ha) that has the third largest investment on artichoke crops after Italy and Spain [27]. As a perennial crop, artichoke produces a robust stalk and large leaf biomass (about 75–80% of the above-ground plant biomass) from which during the reproductive phase the floral stems emerge sustaining the primary edible portion constituted by the immature inflorescences called heads or capitula. The capitula is constituted by a fleshy receptacle (the base of the inflorescence) with the inner heart made of tender bracts protected by more fibrous external bracts. The immature inflorescences are a rich source of macro- and microminerals, dietary fibers, inulin, vitamins, sesquiterpene lactones, and phenolic compounds including a series of flavonoids compounds and anthocyanins that are responsible for the pigmentation of the bracts characterizing the heads of different genotypes [20,21,257,258]. The pigmentation of the heads is in fact along with other morphology parameters (shape, presence of spines on bracts) and agronomic characteristics (earliness), one of the main factors used to classify artichoke varietal types. Based on the pigmentation of the heads, artichoke landraces and new hybrids are distinguished in two groups, namely green and pigmented artichokes. The pigmentation of the heads is considered an important quality parameter affecting consumer acceptance in different regions [20,259], and often it is anticipated in the name of the varietal type especially for the pigmented varietal type like in the case of the French “Violet de Provence”, “Violet de Hyères”, and “Violet du Gapeau”, or the Italian “Violetto di provenza”, “Violetto di Sicilia”, “Violetto Toscano”, “Violetto di S. Erasmo”, and “Violetto di Chioggia” [260]. In a recent study on the inheritance of artichoke bract pigmentation, Portis et al. [261] found a good level of heritability for bract pigmentation. However, previous studies show that there is large variations of the pigmentation of the bracts within the same landrace, and in the case of artichoke the biosynthesis and accumulation of anthocyanins is highly influenced by environmental conditions and especially by temperatures [261,262]. Artichoke pigmentation is considered a complex trait in which several metabolic pathways could be involved. In 1969, Pochard et al. [263] proposed that the anthocyanin pigmentation in artichoke could be genetically determined by one or two major genes with the involvement of several modifiers. Later, based on the segregation pattern observed in populations obtained crossing inbreed lines and clones and self-pollinated clones of different genetic origins, Cravero et al. [264] proposed that the pigmentation of artichoke heads may be controlled genetically by two independent genes P and U with a simple recessive epistasis, where plants of genotype PP or Pp allow the biosynthesis of anthocyanins and result in purple bracts whereas pp genotype inhibits anthocyanin synthesis resulting in green bracts; at the same time genotypes UU or Uu are characterized by a non-uniform pigmentation encoded by the allele P, and uu genotypes develop uniformly pigmented bracts in presence of the allele P. While further studies should confirm this simple model, the variability of the pigmentation observed within the same genotype suggests that the genetic basis for anthocyanin pigmentation of artichoke bracts is more complex and likely the two loci model proposed is regulated by several modifier genes or multiple alleles that determine the expression of the intensity of the pigmentation in artichoke heads [261,262,264]. More recently, De Palma et al. [265] isolated and functionally characterized CcF3’H as the first structural gene of the flavonoid biosynthesis (encoding for flavonoid 3’-hydroxylase) from C. cardunculus. While Blanco et al. [266] isolated and functionally characterized CcMYB12, the artichoke putative homologue of the transcription factor R2R3-MYBs expressing proteins that regulate the biosynthesis of flavonoids and anthocyanins in many species.
Analyzing the polyphenolic profile of artichokes in different genotypes, it is possible to identify two main classes of phenolic compounds, namely hydroxicinnamic acids (C3-C6 skeleton) and flavonoids (C6-C3-C6 skeleton). Within the first class chlorogenic, 3,5-O-dicaffeoylquinic, and 1,5-O-dicaffeoylquinic acids are the predominant compounds followed by other minor mono- and di-caffeoylquinic acids [20,21,267]. Among the flavonoids that quantitatively represent about 10% of the total phenolic compound in artichoke [20], the flavones apigenin-7-O-glucuronide, apigenin-7-O-rutinoside, apigenin-7-O-glucoside, luteolin-7-O-glucuronide, luteolin-7-O-rutinoside, luteolin-7-O-glucoside, naringenin-7-O-rutinoside, and naringenin-7-O-glucoside are the predominant compounds, followed by the anthocyanins including cyanidin, peonidin, and delphinidin derivatives [20,21,257,267,268]. Other studies have revealed that specific phenolic compounds are accumulated only in certain genotypes and certain plant portions even within the same capitula [269,270].
The first attempts to identify the anthocyanin profile of green and purple artichokes were conducted in the late 1970s [20]. However, only later Schütz et al. [271] analyzing by HPLC the pigmented bracts of seven different varietal types of artichoke isolated and identified the main anthocyanins as cyanidin 3,5-diglucoside, cyanidin 3-glucoside, cyanidin 3,5-malonyldiglucoside, cyanidin 3-(3′′-malonyl)glucoside, and cyanidin 3-(6′′-malonyl)glucoside along with some minor compounds such as peonidin 3-glucoside, peonidin 3-(6′′-malonyl)glucoside, and delphinidin glycoside. The same authors observed that the anthocyanin profile varies between different genotypes, nevertheless limited information is available on the genotypic variation of these compounds in artichoke. Analyzing new hybrids and local landraces characterized by bracts with different levels of pigmentation, Bonasia et al. [272] observed that “Violetto di Provenza” and “Tempo” characterized by higher pigmentation had also the highest total phenolic content in the heart and external bracts and in the external bracts, respectively. Examining the content of total anthocyanins in bracts, leaves, and floral stems of two genotypes grown in Tunisia, namely “Violet d’Hyéres” and “Blanc d’Oran”, Dabbou et al. [273] found that leaves of “Blanc d’Oran” had the highest concentration of anthocyanins (20.5 µg/g DW) while bracts and floral stems had the lowest concentration (8.3 and 5.9 µg/g DW, respectively). Instead, lower variability between different plant portions was observed in the case of “Violet d’Hyéres” that on average had a total anthocyanin concentration of 14.2 µg/g DW). These results suggest that even the leaves of artichoke plants which represent a big portion of plant biomass may constitute a good source of anthocyanins besides being a source of other phenolic compounds [274]. A very limited number of studies have been conducted to evaluate specifically the biological activity of artichoke anthocyanins [257]. The main bioactive effect attributed to anthocyanins, flavonoids, and other phenolic compounds extracted from artichoke plant tissues is their antioxidant activity demonstrated by several in vitro and in vivo studies [20,21,275,276]. Anthocyanins have also shown lipid lowering effects in a placebo-control double-blind study by reducing serum LDL cholesterol by 7.9%, triglycerides by 23.0%, apolipoprotein by 16.5%, and apolipoprotein C-III by 11.0%, and increasing HDL cholesterol by 19.4% compared with placebo after administration of 160 mg of anthocyanins for 24 weeks twice daily [277]. Intake of anthocyanins seems to have positive effects on the cardiovascular system by also reducing arterial stiffness [278]. In another study, it was observed that artichoke leaf extracts and artichoke flavonoids up-regulate the gene expression of endothelial-type nitric oxide synthase (eNOS, a vasoprotective molecule) in human endothelial cells [279]. While in a follow-up study Xia et al. [280] observed that treatment of human coronary artery smooth muscle cells (HCASMC) with artichoke leaf extracts and particularly with cynarin and cyanidin induced a down-regulation of inducible nitrous oxide synthase (iNOS, a pro-inflammatory molecule that can cause vascular dysfunctions), suggesting that artichoke flavonoid compounds may have great therapeutic potential. In recent years, a number of studies contributed to demonstrate that artichoke heads and leaf extracts and, in some cases, specific phenolic compounds have health-beneficial properties including anti-inflammatory activity, anti-bacterial activity, hepatoprotective activity, hypocholesterolemic and low density lipoproteins (LDL) oxidation inhibition effect, hypoglycemic effect, as well as anticancer activity [20,21,257]. Nevertheless, in most of the cases these biological activities cannot be attributed to a single compound, but are determined by the combined synergistic effect of different compounds [20,21]. In this perspective, the matrix effect is fundamental in determining the bioaccessibility, bioavailability, and the effect of polyphenols, especially considering that polyphenols and anthocyanins in particular have a relatively low bioavailability being quickly transformed into derivatives of phenolic acids [21,281,282,283].

4.4. Asparagus

Used since ancient time for the diuretic and medicinal properties of its spears [284], cultivated asparagus (Asparagus officinalis L.) is now a popular vegetable grown worldwide on over 1.5 million hectares producing over 8.9 million tons of spears [27]. Native of Eastern Europe, cultivated asparagus is grown at commercial level mainly in China, followed far behind by Perú, Mexico, Germany, Spain, USA, Italy, Japan, France, and the Netherlands. The edible portion of this herbaceous perennial is constituted by its spears or young stems produced by the underground crown, which may be green, green-purple, or purple when harvested above ground or white when purposely harvested before exposed to sunlight. Asparagus spears are considered a rich source of minerals [285], amino acids and dietary fibers [286], saponins [287,288], vitamins and volatile sulfur organic compounds [289], and especially of phenolic compounds and flavonoids [290], including anthocyanins that are responsible for the purple color of the bracts of green spears or of the whole spears in purple asparagus genotypes [23,154,291]. Being rich of all these bioactive compounds it is difficult to isolate the effect of anthocyanins and specific pigments, nevertheless several studies have demonstrated the antioxidant properties of anthocyanins in the species [284]. Comparing green, white, and purple spears, Maeda et al. [291] found that purple spears had significantly higher levels of rutin compared to green spears, while rutin was not detected in white asparagus spears. The same authors observed a positive correlation between spears total polyphenol content, rutin, and DPPH radical scavenging activities, suggesting that purple asparagus may provide higher levels of antioxidants compared to green and white asparagus [291]. In this perspective, while most of the cultivars have been selected to produce green spears and have a relatively small content of anthocyanins, and even less pigments when etiolated to produce white spears, more recently, as for other crops, specific functional breeding programs have developed purple cultivars characterized by high levels of anthocyanins to satisfy the need of consumers attracted by new colors and seeking richer sources of natural antioxidants [291]. Most of the modern commercial cultivars of green and white asparagus are diploid (2n = 20) and were developed from an old population called “Purple Dutch” from which French growers selected two stocks called “Precoce d’Argenteuil” and “Tardive d’Argenteuil” which were subsequently used to develop modern cultivars and hybrids in The Netherlands, France, Germany, United Kingdom, and in the USA [292,293]. Although any green asparagus genotype may be used to produce etiolated white spears, Dutch, French, and German breeding programs have selected cultivars suitable specifically for white spears production to satisfy the European market requirements by reducing as much possible the content of anthocyanins that may develop and accumulate in the tips of white-harvested spears even during storage [292,294]. On the other hand, using “Violetto d’Albenga” a tetraploid local population traditionally grown in Northern Italy, likely derived from interspecific crossing between A. officinalis and A. maritimus [295] and characterized by the production of larger, sweeter, and less fibrous spears ranging in color from green to dark purple, breeding programs in the US and New Zealand have developed in the early 1990s the first cultivars producing purple spears such as “Purple Passion” [296] and “Purple Pacific” and “Stewarts Purple” [297].
Using HPLC and NMR to analyze the fresh peel of “Purple Passion” asparagus, Sakaguchi et al. [23] isolated two major anthocyanins identified as: cyanidin 3-[3’’-(O-β-d-glucopyranosyl)-6’’-(O-α-l-rhamnopyranosyl)-O-β-d-glucopyranoside], and cyanidin 3-rutinoside characterized by high antioxidant activity as determined through ORAC (Oxygen Radical Absorbance Capacity) assay. Recently, Dong et al. [298] comparing the anthocyanin profile of three purple asparagus cultivars (“Jing Zi-2”, “Purple Passion”, and “Pacific Purple”) with a green control (“Jing Lv-1”), identified sixteen anthocyanins, with peonidin, cyanidin and their glycoside derivatives being the predominant compounds. In the same study, through transcriptomics and quantitative real-time polymerase chain reaction (qRT-PCR) analysis, several anthocyanin synthesizing genes (PAL, C4h, 4CL, CHS, CHI, F3H, F3’H, DFR, ANS, and 3GT) and transcription factors genes (bHLH137-like, TT2-like, WD40-like, bZIP61-like, and MADS18-like) that regulates the biosynthesis of anthocyanins like reported for other species were identified and showed to be differentially expressed between purple and green cultivars [298]. The expression of the same genes and the content of anthocyanins were examined also comparing spears grown in the presence of light and in the dark. In accordance with previous studies [299,300], results demonstrated that anthocyanin biosynthetic and regulatory genes are considerably down-regulated in absence of light and pigments are not synthesized in dark conditions, suggesting that light conditions are a key factor for anthocyanin biosynthesis and accumulation. This is further corroborated by the findings of Huyskens-Keil et al. [301] who showed the effects of light quality (white, red, blue, UV-C) on the activity of phenylalanine ammonia-lyase (PAL) and peroxidase (POD) and the synthesis and accumulation of anthocyanins in basal and apical segments of white asparagus spears during postharvest storage. Postharvest conditions and handling may in fact considerably affect the stability of anthocyanins. In another recent study, Barberis et al. [302] reported that dipping the spears of “Purple Passion” for 5 min in a 3 mM solution of oxalic acid (pH 2.9) reduced the lightness of the spears after 12 days only by 13% compared to spears treated with water (control pH 8) or with 1 mM oxalic acid (pH 6) solution, suggesting that the low pH treatment enhanced the stability of the anthocyanin pigments. Analyzing the phenolic profile of green and purple asparagus at harvest and during storage, in accordance with Sakaguchi et al. [23] the same authors [302] found that cyanidin glucosyl rutinoside, cyanidin rutinoside and peonidin rutinoside (at harvest 774.2, 125.5, and 84.8 mg/kg, respectively) were the main anthocyanins identified in “Purple Passion” spears and determining the typical purple color. During storage, the first anthocyanin decreased overtime and was negatively affected by oxalic acid treatment, cyanidin rutinoside increased over time, while peonidin rutinoside remained stable.

4.5. Sweet Corn

Native to America, corn (Zea mays L.) has been cultivated in Central America by indigenous populations at least since 3500 BC [303]. When the European explorers arrived in North America, Iroquois Indians living in the region corresponding to the current state of Pennsylvania and New York grew a variety of sweet corn (Zea mays L. saccharata Sturt.) that turned blue upon ripening, along with other multi-colored varieties [304]. The first samples of maize seeds that arrived in Spain were described as characterized by different colors ranging from white to black [305,306]. Initially grown for curiosity, with the arrival of seeds from North America adapted to higher latitudes, and thus more suitable to the European photoperiod and climate conditions, the new cereal crop started to spread from Spain to the rest of Europe. The rapid diffusion of maize in Europe is also testified by the painter Giuseppe Arcimboldo, in his famous painting “Summer” made in 1573 in which a maize ear is visible [305]. With the process of selection initially conducted in Europe the new crop lost its multi-color pigmentation with the exceptions of a few local populations and assumed the typical white or yellow color of the varieties currently cultivated worldwide.
With over 1.134 million metric tons of corn produced at global level in 2017 [27], corn is by far the first cereal produced in the world, second in terms of acreage only to wheat. Among the different types of maize, sweet corn is particularly important as it is used for human consumption worldwide. In the US, first producer of sweet corn, in 2017 were harvested 464,600 acres (about 188.000 ha) of sweet corn generating a crop value of over $892 million [307]. About 75% of the sweet corn is produced for the fresh market and the remaining portion is used for canned and frozen food processing, making sweet corn the second most important processing vegetable after processing tomato.
Sweet corn is derived from a natural genetic mutation of field corn (dent corn) that was first reported in Pennsylvania in 1770, although it was probably cultivated before by native Americans [308]. The mutant genotype sugary (su) accumulates more sugar in the endosperm (10.2%) than the standard starchy maize (3.5%) resulting in sweeter taste. Years later, several sugary sweet corn varieties have been selected. These traditional varieties characterized mainly by white or yellow kernels, are harvested before complete physiological ripening when the level of sugar is maximum and have short shelf-life because after harvest the sugar is rapidly converted to starch [309]. Subsequently, sweeter hybrid varieties have been developed by selecting sugar-enhanced (se) mutants characterized by double content of sugar (20–35%), tender kernels, creamy texture and good corn flavor. Yet, more recently supersweet varieties have been developed selecting shrunken-2 (sh2) mutants characterized by even higher sugar content at the immature milky stage (29.9%) and extended shelf life, with lighter shrunken kernels. Lately high sugar hybrids are developed combining su, se, and sh2 genes to obtain optimal combinations of sugar, texture, taste, and long shelf life [309,310]. Most of the sweet corn hybrids grown and consumed today at commercial level are characterized by yellow (60%), white (20%), or bicolor (20%, yellow and white) kernels. Nevertheless, in recent years, the interest in reviving ancient colored sweet corn varieties or developing new pigmented varieties characterized by high content of carotenoids [311] and especially anthocyanins is increasing [305,312,313,314,315,316] due to the potential functional properties of anthocyanin-rich genotypes [317,318], as well as to the increasing demand of natural colorants [22,319,320]. The biosynthesis of anthocyanins in maize aleurone (external part of the endosperm) and pericarp (external part of the kernel) of the kernels or in plant tissues involve over twenty structural and regulatory genes that have been identified and functionally characterized as reviewed by Petroni et al. [305,321,322,323]. The wide range of colors observed in maize kernels is mainly due to the biosynthesis and accumulation of carotenoids and flavonoids. Carotenoids such as β-carotene, zeaxanthin and lutein which are lipid-soluble pigments and are responsible for the color of kernels ranging from yellow to deep orange [311,324]. Maize flavonoids include two main classes of pigments: (i) phlobaphenes which are water-insoluble 3-deoxyflavanoid pigments that accumulate in the pericarp of the kernels and the cob and are responsible for the development of kernel colors ranging from orange to brick red [315]; and (ii) anthocyanins which are water-soluble pigments responsible for the development of pink, red, purple, and blue color in the aleurone and pericarp of the kernels as well as in other plant tissues. Within the anthocyanins, the main class of pigments identified are cyanidin, peonidin, and pelargonidin derivatives with the first two providing bluish-red color and the latter being responsible for more orange-red color [22]. The combination of carotenoids and flavonoids in pericarp and aleurone generates an incredible variety of shades [22] and may be influenced also by the condensation with flavanols [319,320,325,326] or other modifications of the anthocyanin compounds. Analyzing the anthocyanin profile of the kernels of six different purple maize genotypes, González-Manzano et al. [326] identified the dimer catechin-(4a-8)-cyanidin-3,5-O-diglucoside and other flavanol-anthocyanin condensed pigments along with several related anthocyanin pigments such as cyanidin-3-glucoside, cyanidin-3-malonylglucoside, peonidin-3-glucoside, peonidin-3-malonylglucoside, pelargonidin-3-dimalonylglucoside, and other derivatives. Malonyl- and dimalonyl acylated anthocyanins are particularly interesting for their higher stability as natural colorants [22]. Analyzing the extract of corn cobs and kernels of a Chinese purple corn, Yang and Zhai [327] identified cyanidin-3-glucoside, pelargonidin-3-glucoside and peonidin-3-glucoside including their malonated derivatives. Besides the great potential of maize pigments as natural food colorants [22,319], there is great interest in the development of anthocyanin-rich maize functional food products [305,311,312,321,328]. Several studies have reported the potential health-beneficial properties of anthocyanin-rich maize [305,317,329]. Examining the phenolic and anthocyanin content of eighteen Mexican maize genotypes, Lopez-Martinez et al. found that total anthocyanins ranged from 1.54 to 850.9 mg of cyanidin-glucoside equivalents/100 g of whole grain flour. Purple genotypes rich in anthocyanins exhibited also the greatest antioxidant activity [330]. Andean purple corn had higher antioxidant activity and antiradical kinetics than blueberries and higher or similar level of anthocyanins [331]. Similarly, analyzing 49 lines of waxy corn (Zea mays L. var. ceratina) characterized by different colors Harakotr et al. [332] found a large variability of phenolic and anthocyanin compounds and a positive correlation coefficient between anthocyanins and 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging ability (r = 0.94) and between anthocyanins and Trolox equivalent antioxidant capacity (TEAC, r = 0.88). Introduction in the diet of anthocyanin-rich corn may contribute to prevent also obesity and diabetes. Tsuda et al. [317] found that feeding high fat (HF) mice for 12 weeks with cyanidin 3-glucoside-rich purple corn significantly reduced HF body weight and white and brown adipose tissues compared to HF mice not receiving purple corn supplement. The HF diet induced also hyperglycemia, hyperinsulinemia, and hyperleptinemia, whereas the same physiological conditions were not observed in HF mice receiving the purple corn supplement. Moreover, an increase of tumor necrosis factor (TNF)-α mRNA level was observed in the HF-diet while it was not observed in the HF group receiving the purple corn supplement which suppressed the mRNA of enzymes involved in fatty acid and triacylglycerol synthesis and reduced the level of sterol regulatory element binding protein-1 mRNA in white adipose tissue. Using near-isogenic maize lines which differed only in the presence or not of anthocyanins, and feeding rats with anthocyanin-rich and anthocyanin-deprived maize for 8 weeks it was observed that in the group fed with anthocyanin-rich corn cardiac tissue damaged following ischemic conditions was reduced by about 30% compared to the group fed with anthocyanin-deprived corn [333]. An increase of myocardial glutathione and omega-3 fatty acids levels in blood indicated that diet supplementation with anthocyanins regulated the cardiac antioxidant defense and the conversion of α-linolenic acid to omega-3 fatty acids [333,334]. In an in vivo study, using db/db mice fed with 10 mg/kg of purple corn extract for 8 weeks, Kang et al. [335] found that anthocyanin-rich purple corn extracts reduced glomerular angiogenesis of diabetic kidneys by inhibiting the induction of vascular endothelial growth factor (VEGF) and hypoxia inducible factor (HIF)-1a induced by hyperglycemic condition. Kang et al. results demonstrated that purple corn extract inhibited glomerular angiogenesis caused by chronic hyperglycemia and diabetes by disturbing the Angpt-Tie-2 ligand-receptor system linked to the renal VEGF receptor 2 (VEGFR2), suggesting that purple corn extract could be used to target abnormal angiogenesis in diabetic nephropathy leading to kidney failure [335]. Yet, in a recent study Mazewski et al. [336] found that anthocyanin-rich purple and red corn may potentially contribute to inhibiting human colon cancer cell proliferation by promoting apoptosis and suppressing angiogenesis.
The main pigments isolated in vegetables consumed for various plant parts are presented in Table 3.

5. Conclusions

Vegetable products are pivotal in human health and their daily consumption is highly recommended due to their high content in phytochemicals with diverse beneficial health effects. The vegetable crops presented in this review, namely leafy and fruit vegetables and species where other plant parts are consumed, are well known and widely consumed throughout the world, although their colored variants are not always accepted by consumers and proper marketing is needed to introduce them to the public. Currently, the food and beverage industry is seeking for natural alternatives of synthetic compounds to satiate market demands for “non-chemical additives” food products. Therefore, the broad color palette the presented vegetables exhibit not only could diversify human diet and make food products more attractive to consumers and increase their appeal, but also could be proved as valuable sources of natural coloring agents. Under the circumstances, future research should focus on the identification of those promising coloring agents and the establishment of efficient and sustainable techniques for their isolation. Other future requirements, include breeding and agronomic protocols that could improve pigments content in the final produce, as well as post-harvest and processing conditions to increase extraction efficiency of natural coloring agents. Finally, the mechanisms of actions related with the health effects and antioxidant activities of coloring compounds have to be revealed and valorized in the pharmaceutical and food industry for the design of novel functional foods and drugs.

Funding

This work was supported by the USDA National Institute of Food and Agriculture and Hatch Appropriations under Project #PEN04723 and Accession #1020664; The authors are grateful to Foundation for Science and Technology (FCT, Portugal) and FEDER under Programme PT2020 for financial support to CIMO (UID/AGR/00690/2019); to FEDER-Interreg España-Portugal programme for financial support through the project 0377_Iberphenol_6_E and TRANSCoLAB 0612_TRANS_CO_LAB_2_P; the European Regional Development Fund (ERDF) through the Regional Operational Program North 2020, within the scope of project Mobilizador Norte-01-0247-FEDER-024479: ValorNatural®.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sasaki, N.; Nishizaki, Y.; Ozeki, Y.; Miyahara, T. The role of acyl-glucose in anthocyanin modifications. Molecules 2014, 19, 18747–18766. [Google Scholar] [CrossRef]
  2. Luo, W.P.; Fang, Y.J.; Lu, M.S.; Zhong, X.; Chen, Y.M.; Zhang, C.X. High consumption of vegetable and fruit colour groups is inversely associated with the risk of colorectal cancer: A case-control study. Br. J. Nutr. 2015, 113, 1129–1138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Carlsen, M.H.; Halvorsen, B.L.; Holte, K.; Bøhn, S.K.; Dragland, S.; Sampson, L.; Willey, C.; Senoo, H.; Umezono, Y.; Sanada, C.; et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr. J. 2010, 9, 3. [Google Scholar] [CrossRef] [PubMed]
  4. Mcgill, C.R.; Wightman, J.D.; Fulgoni, S.A.; Fulgoni, V.L. Consumption of Purple/Blue Produce Is Associated with Increased Nutrient Intake and Reduced Risk for Metabolic Syndrome: Results From the National Health and Nutrition Examination Survey 1999–2002. Am. J. Lifestyle Med. 2011, 5, 279–290. [Google Scholar] [CrossRef]
  5. Wu, X.; Beecher, G.R.; Holden, J.M.; Haytowitz, D.B.; Gebhardt, S.E.; Prior, R.L. Concentrations of anthocyanins in common foods in the United States and estimation of normal consumption. J. Agric. Food Chem. 2006, 54, 4069–4075. [Google Scholar] [CrossRef]
  6. Bognar, E.; Sarszegi, Z.; Szabo, A.; Debreceni, B.; Kalman, N.; Tucsek, Z.; Sumegi, B.; Gallyas, F. Antioxidant and Anti-Inflammatory Effects in RAW264.7 Macrophages of Malvidin, a Major Red Wine Polyphenol. PLoS ONE 2013, 8, e65355. [Google Scholar] [CrossRef]
  7. Su, X.; Xu, J.; Rhodes, D.; Shen, Y.; Song, W.; Katz, B.; Tomich, J.; Wang, W. Identification and quantification of anthocyanins in transgenic purple tomato. Food Chem. 2016, 202, 184–188. [Google Scholar] [CrossRef] [Green Version]
  8. Wang, L.S.; Stoner, G.D. Anthocyanins and their role in cancer prevention. Cancer Lett. 2008, 269, 281–290. [Google Scholar] [CrossRef] [Green Version]
  9. Grotewold, E. The genetic and biochemistry of floral pigments. Annu. Rev. Plant Biol. 2006, 57, 761–780. [Google Scholar] [CrossRef]
  10. Schaefer, H.M.; Schaefer, V.; Levey, D.J. How plant-animal interactions signal new insights in communication. Trends Ecol. Evol. 2004, 19, 577–584. [Google Scholar] [CrossRef] [Green Version]
  11. Chaves-Silva, S.; dos Santos, A.L.; Chalfun-Júnior, A.; Zhao, J.; Peres, L.E.P.; Benedito, V.A. Understanding the genetic regulation of anthocyanin biosynthesis in plants—Tools for breeding purple varieties of fruits and vegetables. Phytochemistry 2018, 153, 11–27. [Google Scholar] [CrossRef] [PubMed]
  12. Menzies, I.J.; Youard, L.W.; Lord, J.M.; Carpenter, K.L.; van Klink, J.W.; Perry, N.B.; Schaefer, H.M.; Gould, K.S. Leaf colour polymorphisms: A balance between plant defence and photosynthesis. J. Ecol. 2016, 104, 104–113. [Google Scholar] [CrossRef]
  13. He, J.; Giusti, M.M. Anthocyanins: Natural Colorants with Health-Promoting Properties. Annu. Rev. Food Sci. Technol. 2010, 1, 163–187. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, M.J.; Moon, Y.; Tou, J.C.; Mou, B.; Waterland, N.L. Nutritional value, bioactive compounds and health benefits of lettuce (Lactuca sativa L.). J. Food Compos. Anal. 2016, 49, 19–34. [Google Scholar] [CrossRef]
  15. Johansson, M.; Jägerstad, M.; Frølich, W. Folates in lettuce: A pilot study. Scand. J. Food Nutr. 2007, 51, 22–30. [Google Scholar] [CrossRef]
  16. Giordano, M.; El-Nakhel, C.; Pannico, A.; Kyriacou, M.C.; Stazi, S.R.; De Pascale, S.; Rouphael, Y. Iron biofortification of red and green pigmented lettuce in closed soilless cultivation impacts crop performance and modulates mineral and bioactive composition. Agronomy 2019, 9, 290. [Google Scholar] [CrossRef] [Green Version]
  17. Morais, M.G.; Da Costa, G.A.F.; Aleixo, Á.A.; de Oliveira, G.T.; Alves, L.F.; Duarte-Almeida, J.M.; Ferreira, J.M.S.; Dos Santos Lima, L.A.R. Antioxidant, antibacterial and cytotoxic potential of the ripe fruits of Solanum lycocarpum A. St. Hil. (Solanaceae). Nat. Prod. Res. 2015, 29, 480–483. [Google Scholar] [CrossRef]
  18. Chiu, L.W.; Zhou, X.; Burke, S.; Wu, X.; Prior, R.L.; Li, L. The purple cauliflower arises from activation of a MYB transcription factor. Plant Physiol. 2010, 154, 1470–1480. [Google Scholar] [CrossRef] [Green Version]
  19. Lo Scalzo, R.; Genna, A.; Branca, F.; Chedin, M.; Chassaigne, H. Anthocyanin composition of cauliflower (Brassica oleracea L. var. botrytis) and cabbage (B. oleracea L. var. capitata) and its stability in relation to thermal treatments. Food Chem. 2008, 107, 136–144. [Google Scholar] [CrossRef]
  20. Lattanzio, V.; Kroon, P.A.; Linsalata, V.; Cardinali, A. Globe artichoke: A functional food and source of nutraceutical ingredients. J. Funct. Foods 2009, 1, 131–144. [Google Scholar] [CrossRef]
  21. D’Antuono, I.; Di Gioia, F.; Linsalata, V.; Rosskopf, E.N.; Cardinali, A. Impact on health of artichoke and cardoon bioactive compounds: Content, bioaccessibility, bioavailability, and bioactivity. In Phytochemicals in Vegetables: A Valuable Source of Bioactive Compounds; Petropoulos, S.A., Ferreira, I.C.F.R., Barros, L., Eds.; Bentham Science Publishers Ltd.: Sharjah, UAE, 2018; p. 373. [Google Scholar]
  22. Chatham, L.A.; Paulsmeyer, M.; Juvik, J.A. Prospects for economical natural colorants: Insights from maize. Theor. Appl. Genet. 2019, 132, 2927–2946. [Google Scholar] [CrossRef] [PubMed]
  23. Sakaguchi, Y.; Ozaki, Y.; Miyajima, I.; Yamaguchi, M.; Fukui, Y.; Iwasa, K.; Motoki, S.; Suzuki, T.; Okubo, H. Major anthocyanins from purple asparagus (Asparagus officinalis). Phytochemistry 2008, 69, 1763–1766. [Google Scholar] [CrossRef] [PubMed]
  24. Nicolle, C.; Cardinault, N.; Gueux, E.; Jaffrelo, L.; Rock, E.; Mazur, A.; Amouroux, P.; Rémésy, C. Health effect of vegetable-based diet: Lettuce consumption improves cholesterol metabolism and antioxidant status in the rat. Clin. Nutr. 2004, 23, 605–614. [Google Scholar] [CrossRef] [PubMed]
  25. Nicolle, C.; Carnat, A.; Fraisse, D.; Lamaison, J.L.; Rock, E.; Michel, H.; Amoureux, P.; Remesy, C. Characterisation and variation of antioxidant micronutrients in lettuce (Lactuca sativa folium). J. Sci. Food Agric. 2004, 84, 2061–2069. [Google Scholar] [CrossRef]
  26. Rouphael, Y.; Kyriacou, M.C.; Vitaglione, P.; Giordano, M.; Pannico, A.; Colantuono, A.; De Pascale, S. Genotypic variation in nutritional and antioxidant profile among iceberg lettuce cultivars. Acta Sci. Pol. Hortorum Cultus 2017, 16, 37–45. [Google Scholar] [CrossRef]
  27. FAO FAOSTAT, Crops. FAO Statistics Division. Food and Agriculture Organization of the United Nations 2017. Available online: http://www.fao.org/faostat/en/#data/QC (accessed on 30 December 2019).
  28. Mou, B. Lettuce. In Vegetables, I “Asteraceae, Brassicaceae, Chenopodiaceae, and Cucurbitaceae; Prohens, J., Nuez, F., Eds.; Springer: New York, NY, USA, 2008; pp. 75–116. [Google Scholar]
  29. Wang, C.; Riedl, K.M.; Schwartz, S.J. Fate of folates during vegetable juice processing—Deglutamylation and interconversion. Food Res. Int. 2013, 53, 440–448. [Google Scholar] [CrossRef]
  30. Gan, Y.Z.; Azrina, A. Antioxidant properties of selected varieties of lettuce (Lactuca sativa L.) commercially available in Malaysia. Int. Food Res. J. 2016, 23, 2357–2362. [Google Scholar]
  31. Rouphael, Y.; Kyriacou, M.C.; Petropoulos, S.A.; De Pascale, S.; Colla, G. Improving vegetable quality in controlled environments. Sci. Hortic. 2018, 234, 275–289. [Google Scholar] [CrossRef]
  32. Kyriacou, M.C.; Rouphael, Y. Towards a new definition of quality for fresh fruits and vegetables. Sci. Hortic. 2018, 234, 463–469. [Google Scholar] [CrossRef]
  33. Survase, S.A.; Bajaj, I.B.; Singhal, R.S. Biotechnological production of vitamins. Food Technol. Biotechnol. 2006, 44, 381–396. [Google Scholar]
  34. López, A.; Javier, G.A.; Fenoll, J.; Hellín, P.; Flores, P. Chemical composition and antioxidant capacity of lettuce: Comparative study of regular-sized (Romaine) and baby-sized (Little Gem and Mini Romaine) types. J. Food Compos. Anal. 2014, 33, 39–48. [Google Scholar] [CrossRef]
  35. Llorach, R.; Martínez-Sánchez, A.; Tomás-Barberán, F.A.; Gil, M.I.; Ferreres, F. Characterisation of polyphenols and antioxidant properties of five lettuce varieties and escarole. Food Chem. 2008, 108, 1028–1038. [Google Scholar] [CrossRef] [PubMed]
  36. Mou, B. Genetic variation of beta-carotene and lutein contents in lettuce. J. Am. Soc. Hortic. Sci. 2005, 130, 870–876. [Google Scholar] [CrossRef] [Green Version]
  37. Baslam, M.; Morales, F.; Garmendia, I.; Goicoechea, N. Nutritional quality of outer and inner leaves of green and red pigmented lettuces (Lactuca sativa L.) consumed as salads. Sci. Hortic. 2013, 151, 103–111. [Google Scholar] [CrossRef]
  38. Le Marchand, L.; Hankin, J.H.; Kolonel, L.N.; Beecher, G.R.; Wilkens, L.R.; Zhao, L.P. Intake of Specific Carotenoids and Lung Cancer Risk. Cancer Epidemiol. Biomark. Prev. 1993, 2, 183–187. [Google Scholar]
  39. Giovannucci, E. Tomatoes, Tomato-Based Products, Lycopene, and Cancer: Review of the Epidemiologic Literature. J. Natl. Cancer Inst. 1999, 91, 317–331. [Google Scholar] [CrossRef]
  40. Johnson, E.J.; Hammond, B.R.; Yeum, K.J.; Qin, J.; Wang, X.D.; Castaneda, C.; Snodderly, D.M.; Russell, R.M. Relation among serum and tissue concentrations of lutein and zeaxanthin and macular pigment density. Am. J. Clin. Nutr. 2000, 71, 1555–1562. [Google Scholar] [CrossRef] [Green Version]
  41. Slattery, M.L.; Benson, J.; Curtin, K.; Ma, K.N.; Schaeffer, D.; Potter, J.D. Carotenoids and colon cancer. Am. J. Clin. Nutr. 2000, 71, 575–582. [Google Scholar] [CrossRef] [Green Version]
  42. Kim, D.E.; Shang, X.; Assefa, A.D.; Keum, Y.S.; Saini, R.K. Metabolite profiling of green, green/red, and red lettuce cultivars: Variation in health beneficial compounds and antioxidant potential. Food Res. Int. 2018, 105, 361–370. [Google Scholar] [CrossRef]
  43. Mulabagal, V.; Ngouajio, M.; Nair, A.; Zhang, Y.; Gottumukkala, A.L.; Nair, M.G. In vitro evaluation of red and green lettuce (Lactuca sativa) for functional food properties. Food Chem. 2010, 118, 300–306. [Google Scholar] [CrossRef]
  44. Hao, J.H.; Wang, L.; Liu, C.J.; Zhang, X.D.; Qi, Z.Y.; Li, B.Y.; Xiao, S.; Fan, S.X. Evaluation of nutritional quality in different varieties of green and purple leaf lettuces. In IOP Conference Series: Earth and Environmental Science; IOP Publishing: Bristol, UK, 2018; Volume 185. [Google Scholar]
  45. El-Nakhel, C.; Petropoulos, S.A.; Pannico, A.; Kyriacou, M.C.; Giordano, M.; Colla, G.; Dario Troise, A.; Vitaglione, P.; De Pascale, S.; Rouphael, Y. The bioactive profile of lettuce produced in a closed soilless system as configured by combinatorial effects of genotype and macrocation supply composition. Food Chem. 2020, 309, 125713. [Google Scholar] [CrossRef] [PubMed]
  46. El-Nakhel, C.; Pannico, A.; Kyriacou, M.C.; Giordano, M.; De Pascale, S.; Rouphael, Y. Macronutrient deprivation eustress elicits differential secondary metabolites in red and green-pigmented butterhead lettuce grown in a closed soilless system. J. Sci. Food Agric. 2019, 99, 6962–6972. [Google Scholar] [CrossRef] [PubMed]
  47. El-Nakhel, C.; Giordano, M.; Pannico, A.; Carillo, P.; Fusco, G.M.; De Pascale, S.; Rouphael, Y. Cultivar-specific performance and qualitative descriptors for butterhead salanova lettuce produced in closed soilless cultivation as a candidate salad crop for human life support in space. Life 2019, 9, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Mampholo, B.M.; Maboko, M.M.; Soundy, P.; Sivakumar, D. Phytochemicals and overall quality of leafy lettuce (Lactuca sativa L.) varieties grown in closed hydroponic system. J. Food Qual. 2016, 39, 805–815. [Google Scholar] [CrossRef]
  49. Luna, M.C.; Martínez-Sánchez, A.; Selma, M.V.; Tudela, J.A.; Baixauli, C.; Gil, M.I. Influence of nutrient solutions in an open-field soilless system on the quality characteristics and shelf life of fresh-cut red and green lettuces (Lactuca sativa L.) in different seasons. J. Sci. Food Agric. 2013, 93, 415–421. [Google Scholar] [CrossRef]
  50. Rouphael, Y.; Petropoulos, S.A.; El-Nakhel, C.; Pannico, A.; Kyriacou, M.C.; Giordano, M.; Troise, A.D.; Vitaglione, P.; De Pascale, S. Reducing Energy Requirements in Future Bioregenerative Life Support Systems (BLSSs): Performance and Bioactive Composition of Diverse Lettuce Genotypes Grown Under Optimal and Suboptimal Light Conditions. Front. Plant Sci. 2019, 10, 1305. [Google Scholar] [CrossRef]
  51. Pannico, A.; El-Nakhel, C.; Kyriacou, M.C.; Giordano, M.; Stazi, S.R.; De Pascale, S.; Rouphael, Y. Combating Micronutrient Deficiency and Enhancing Food Functional Quality Through Selenium Fortification of Select Lettuce Genotypes Grown in a Closed Soilless System. Front. Plant Sci. 2019, 10, 1495. [Google Scholar] [CrossRef] [Green Version]
  52. Rice-Evans, C.A.; Miller, N.J.; Paganga, G. Antioxidant properties of phenolic compounds. Trends Plant Sci. 1997, 2, 152–159. [Google Scholar] [CrossRef]
  53. Pérez-López, U.; Pinzino, C.; Quartacci, M.F.; Ranieri, A.; Sgherri, C. Phenolic composition and related antioxidant properties in differently colored lettuces: A study by electron paramagnetic resonance (EPR) kinetics. J. Agric. Food Chem. 2014, 62, 12001–12007. [Google Scholar] [CrossRef]
  54. Lee, J.H.; Felipe, P.; Yang, Y.H.; Kim, M.Y.; Oh, Y.K.; Sok, D.E.; Kim, H.C.; Kim, M.R. Effects of dietary supplementation with red-pigmented leafy lettuce (Lactuca sativa) on lipid profiles and antioxidant status in C57BL/6J mice fed a high-fat high-cholesterol diet. Br. J. Nutr. 2009, 101, 1246–1254. [Google Scholar] [CrossRef] [Green Version]
  55. Fernandez, E.; La Vecchia, C.; D’Avanzo, B.; Negri, E.; Franceschi, S. Risk factors for colorectal cancer in subjects with family history of the disease. Br. J. Cancer 1997, 75, 1381–1384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Qin, X.X.; Zhang, M.Y.; Han, Y.Y.; Hao, J.H.; Liu, C.J.; Fan, S.X. Beneficial phytochemicals with anti-tumor potential revealed through metabolic profiling of new red pigmented lettuces (Lactuca sativa L.). Int. J. Mol. Sci. 2018, 19, 1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Sgherri, C.; Cecconami, S.; Pinzino, C.; Navari-Izzo, F.; Izzo, R. Levels of antioxidants and nutraceuticals in basil grown in hydroponics and soil. Food Chem. 2010, 123, 416–422. [Google Scholar] [CrossRef]
  58. Simon, J.; Morales, M.; Phippen, W.; Vieira, R.; Hao, Z. Basil: A source of aroma compounds and a popular culinary and ornamental herb. Perspect. New Crop. New Uses 1999, 16, 499–505. [Google Scholar]
  59. Lee, J.; Scagel, C.F. Chicoric acid found in basil (Ocimum basilicum L.) leaves. Food Chem. 2009, 115, 650–656. [Google Scholar] [CrossRef]
  60. Javanmardi, J.; Khalighi, A.; Kashi, A.; Bais, H.P.; Vivanco, J.M. Chemical characterization of basil (Ocimum basilicum L.) found in local accessions and used in traditional medicines in Iran. J. Agric. Food Chem. 2002, 50, 5878–5883. [Google Scholar] [CrossRef]
  61. Kwee, E.M.; Niemeyer, E.D. Variations in phenolic composition and antioxidant properties among 15 basil (Ocimum basilicum L.) cultivars. Food Chem. 2011, 128, 1044–1050. [Google Scholar] [CrossRef]
  62. Kopsell, D.A.; Kopsell, D.E.; Curran-Celentano, J. Carotenoid and chlorophyll pigments in sweet basil grown in the field and greenhouse. HortScience 2005, 40, 1230–1233. [Google Scholar] [CrossRef]
  63. Flanigan, P.M.; Niemeyer, E.D. Effect of cultivar on phenolic levels, anthocyanin composition, and antioxidant properties in purple basil (Ocimum basilicum L.). Food Chem. 2014, 164, 518–526. [Google Scholar] [CrossRef]
  64. Calucci, L.; Pinzino, C.; Zandomeneghi, M.; Capocchi, A.; Ghiringhelli, S.; Saviozzi, F.; Tozzi, S.; Galleschi, L. Effects of γ-irradiation on the free radical and antioxidant contents in nine aromatic herbs and spices. J. Agric. Food Chem. 2003, 51, 927–934. [Google Scholar] [CrossRef]
  65. Muráriková, A.; Neugebauerová, J. Seasonal variation of ascorbic acid and nitrate levels in selected basil (Ocimum basilicum L.) varieties. Hortic. Sci. 2018, 45, 47–52. [Google Scholar] [CrossRef] [Green Version]
  66. Gallie, D.R. Increasing vitamin C content in plant foods to improve their nutritional value-successes and challenges. Nutrients 2013, 5, 3424–3446. [Google Scholar] [CrossRef] [Green Version]
  67. Tenore, G.C.; Campiglia, P.; Ciampaglia, R.; Izzo, L.; Novellino, E. Antioxidant and antimicrobial properties of traditional green and purple “Napoletano” basil cultivars (Ocimum basilicum L.) from Campania region (Italy). Nat. Prod. Res. 2017, 31, 2067–2071. [Google Scholar] [CrossRef] [PubMed]
  68. Phippen, W.B.; Simon, J.E. Anthocyanins in Basil (Ocimum basilicum L.). J. Agric. Food Chem. 1998, 46, 1734–1738. [Google Scholar] [CrossRef]
  69. Cazzola, R.; Camerotto, C.; Cestaro, B. Anti-oxidant, anti-glycant, and inhibitory activity against α-amylase and α-glucosidase of selected spices and culinary herbs. Int. J. Food Sci. Nutr. 2011, 62, 175–184. [Google Scholar] [CrossRef]
  70. El-Beshbishy, H.A.; Bahashwan, S.A. Hypoglycemic effect of basil (Ocimum basilicum) aqueous extract is mediated through inhibition of α-glucosidase and α-amylase activities: An in vitro study. Toxicol. Ind. Health 2011, 28, 42–50. [Google Scholar] [CrossRef]
  71. Kwon, Y.I.I.; Vattem, D.A.; Shetty, K. Evaluation of clonal herbs of Lamiaceae species for management of diabetes and hypertension. Asia Pac. J. Clin. Nutr. 2006, 15, 107–118. [Google Scholar]
  72. Ha, T.J.; Lee, J.H.; Lee, M.H.; Lee, B.W.; Kwon, H.S.; Park, C.H.; Shim, K.B.; Kim, H.T.; Baek, I.Y.; Jang, D.S. Isolation and identification of phenolic compounds from the seeds of Perilla frutescens (L.) and their inhibitory activities against α-glucosidase and aldose reductase. Food Chem. 2012, 135, 1397–1403. [Google Scholar] [CrossRef]
  73. Yilmazer-Musa, M.; Griffith, A.M.; Michels, A.J.; Schneider, E.; Frei, B. Grape seed and tea extracts and catechin 3-gallates are potent inhibitors of α-amylase and α-glucosidase activity. J. Agric. Food Chem. 2012, 60, 8924–8929. [Google Scholar] [CrossRef] [Green Version]
  74. Harnafi, H.; Serghini Caid, H.; el Houda Bouanani, N.; Aziz, M.; Amrani, S. Hypolipemic activity of polyphenol-rich extracts from Ocimum basilicum in Triton WR-1339-induced hyperlipidemic mice. Food Chem. 2008, 108, 205–212. [Google Scholar] [CrossRef]
  75. Amrani, S.; Harnafi, H.; Bouanani, N.E.H.; Aziz, M.; Caid, H.S.; Manfredini, S.; Besco, E.; Napolitano, M.; Bravo, E. Hypolipidaemic Activity of Aqueous Ocimum basilicum Extract in Acute Hyperlipidaemia Induced by Triton WR-1339 in Rats and its Antioxidant Property. Phyther. Res. 2006, 20, 1040–1045. [Google Scholar] [CrossRef] [PubMed]
  76. Dhyani, A.; Chopra, R.; Garg, M. A review on nutritional value, functional properties and pharmacological application of perilla (Perilla frutescens L.). Biomed. Pharmacol. J. 2019, 12, 649–660. [Google Scholar] [CrossRef]
  77. Müller-Waldeck, F.; Sitzmann, J.; Schnitzler, W.H.; Graßmann, J. Determination of toxic perilla ketone, secondary plant metabolites and antioxidative capacity in five Perilla frutescens L. varieties. Food Chem. Toxicol. 2010, 48, 264–270. [Google Scholar] [CrossRef] [PubMed]
  78. Meng, L.; Lozano, Y.; Bombarda, I.; Gaydou, E.M.; Li, B. Polyphenol extraction from eight Perilla frutescens cultivars. Comptes Rendus Chim. 2009, 12, 602–611. [Google Scholar] [CrossRef]
  79. Peng, Y.; Ye, J.; Kong, J. Determination of phenolic compounds in Perilla frutescens L. by capillary electrophoresis with electrochemical detection. J. Agric. Food Chem. 2005, 53, 8141–8147. [Google Scholar] [CrossRef]
  80. Lu, N.; Bernardo, E.L.; Tippayadarapanich, C.; Takagaki, M.; Kagawa, N.; Yamori, W. Growth and accumulation of secondary metabolites in perilla as affected by photosynthetic photon flux density and electrical conductivity of the nutrient solution. Front. Plant Sci. 2017, 8, 708. [Google Scholar] [CrossRef] [Green Version]
  81. Martinetti, L.; Ferrante, A.; Bassoli, A.; Borgonovo, G.; Tosca, A.; Spoleto, P. Characterization of some qualitative traits in different perilla cultivars. Acta Hortic. 2012, 939, 301–308. [Google Scholar] [CrossRef]
  82. Rouphael, Y.; Kyriacou, M.C.; Carillo, P.; Pizzolongo, F.; Romano, R.; Sifola, M.I. Chemical Eustress Elicits Tailored Responses and Enhances the Functional Quality of Novel Food Perilla frutescens. Molecules 2019, 24, 185. [Google Scholar] [CrossRef] [Green Version]
  83. Meng, L.; Lozano, Y.F.; Gaydou, E.M.; Li, B. Antioxidant activities of polyphenols extracted from Perilla frutescens varieties. Molecules 2009, 14, 133–140. [Google Scholar] [CrossRef] [Green Version]
  84. Narisawa, T.; Fukaura, Y.; Yazawa, K.; Ishikawa, C.; Isoda, Y.; Nishizawa, Y. Colon cancer prevention with a small amount of dietary perilla oil high in alpha-linolenic acid in an animal model. Cancer 1994, 73, 2069–2075. [Google Scholar] [CrossRef]
  85. Banno, N.; Akihisa, T.; Tokuda, H.; Yasukawa, K.; Higashihara, H.; Ukiya, M.; Watanabe, K.; Kimura, Y.; Hasegawa, J.I.; Nishino, H. Triterpene acids from the leaves of Perilla frutescens and their anti-inflammatory and antitumor-promoting effects. Biosci. Biotechnol. Biochem. 2004, 68, 85–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Bassoli, A.; Borgonovo, G.; Caimi, S.; Scaglioni, L.; Morini, G.; Moriello, A.S.; di Marzo, V.; de Petrocellis, L. Taste-guided identification of high potency TRPA1 agonists from Perilla frutescens. Bioorganic Med. Chem. 2009, 17, 1636–1639. [Google Scholar] [CrossRef] [PubMed]
  87. Laureati, M.; Buratti, S.; Bassoli, A.; Borgonovo, G.; Pagliarini, E. Discrimination and characterisation of three cultivars of Perilla frutescens by means of sensory descriptors and electronic nose and tongue analysis. Food Res. Int. 2010, 43, 959–964. [Google Scholar] [CrossRef]
  88. Kanner, J.; Harel, S.; Granit, R. Betalains - A new class of dietary cationized antioxidants. J. Agric. Food Chem. 2001, 49, 5178–5185. [Google Scholar] [CrossRef]
  89. Daiss, N.; Lobo, M.G.; Gonzalez, M. Changes in postharvest quality of swiss chard grown using 3 organic preharvest treatments. J. Food Sci. 2008, 73, S314–S320. [Google Scholar] [CrossRef]
  90. Mzoughi, Z.; Chahdoura, H.; Chakroun, Y.; Cámara, M.; Fernández-Ruiz, V.; Morales, P.; Mosbah, H.; Flamini, G.; Snoussi, M.; Majdoub, H. Wild edible Swiss chard leaves (Beta vulgaris L. var. cicla): Nutritional, phytochemical composition and biological activities. Food Res. Int. 2019, 119, 612–621. [Google Scholar] [CrossRef]
  91. Ninfali, P.; Antonini, E.; Frati, A.; Scarpa, E.S. C-Glycosyl Flavonoids from Beta vulgaris Cicla and Betalains from Beta vulgaris rubra: Antioxidant, Anticancer and Antiinflammatory Activities—A Review. Phyther. Res. 2017, 31, 871–884. [Google Scholar] [CrossRef]
  92. Lee, C.Y.; Chien, Y.S.; Chiu, T.H.; Huang, W.W.; Lu, C.C.; Chiang, J.H.; Yang, J.S. Apoptosis triggered by vitexin in U937 human leukemia cells via a mitochondrial signaling pathway. Oncol. Rep. 2012, 28, 1883–1888. [Google Scholar] [CrossRef] [Green Version]
  93. Ninfali, P.; Bacchiocca, M.; Antonelli, A.; Biagiotti, E.; Di Gioacchino, A.M.; Piccoli, G.; Stocchi, V.; Brandi, G. Characterization and biological activity of the main flavonoids from Swiss Chard (Beta vulgaris subspecies cycla). Phytomedicine 2007, 14, 216–221. [Google Scholar] [CrossRef]
  94. Gennari, L.; Felletti, M.; Blasa, M.; Angelino, D.; Celeghini, C.; Corallini, A.; Ninfali, P. Total extract of Beta vulgaris var. cicla seeds versus its purified phenolic components: Antioxidant activities and antiproliferative effects against colon cancer cells. Phytochem. Anal. 2011, 22, 272–279. [Google Scholar] [CrossRef]
  95. Borghi, S.M.; Carvalho, T.T.; Staurengo-Ferrari, L.; Hohmann, M.S.N.; Pinge-Filho, P.; Casagrande, R.; Verri, W.A. Vitexin Inhibits Inflammatory Pain in Mice by Targeting TRPV1, Oxidative Stress, and Cytokines. J. Nat. Prod. 2013, 76, 1141–1149. [Google Scholar] [CrossRef] [PubMed]
  96. Khanam, U.K.S.; Oba, S.; Yanase, E.; Murakami, Y. Phenolic acids, flavonoids and total antioxidant capacity of selected leafy vegetables. J. Funct. Foods 2012, 4, 979–987. [Google Scholar] [CrossRef]
  97. Neugart, S.; Baldermann, S.; Hanschen, F.S.; Klopsch, R.; Wiesner-Reinhold, M.; Schreiner, M. The intrinsic quality of brassicaceous vegetables: How secondary plant metabolites are affected by genetic, environmental, and agronomic factors. Sci. Hortic. 2018, 233, 460–478. [Google Scholar] [CrossRef]
  98. Martínez-Sánchez, A.; Gil-Izquierdo, A.; Gil, M.I.; Ferreres, F. A comparative study of flavonoid compounds, vitamin C, and antioxidant properties of baby leaf Brassicaceae species. J. Agric. Food Chem. 2008, 56, 2330–2340. [Google Scholar] [CrossRef]
  99. Di Noia, J. Defining powerhouse fruits and vegetables: A nutrient density approach. Prev. Chronic Dis. 2014, 11, 3–7. [Google Scholar] [CrossRef] [Green Version]
  100. Kim, M.J.; Chiu, Y.C.; Ku, K.M. Glucosinolates, carotenoids, and vitamins E and K Variation from selected kale and collard cultivars. J. Food Qual. 2017, 2017, 5123572. [Google Scholar] [CrossRef] [Green Version]
  101. Hahn, C.; Müller, A.; Kuhnert, N.; Albach, D. Diversity of Kale (Brassica oleracea var. sabellica): Glucosinolate Content and Phylogenetic Relationships. J. Agric. Food Chem. 2016, 64, 3215–3225. [Google Scholar] [CrossRef]
  102. Ferioli, F.; Giambanelli, E.; D’Antuono, L.F.; Costa, H.S.; Albuquerque, T.G.; Silva, A.S.; Hayran, O.; Koçaoglu, B. Comparison of leafy kale populations from italy, portugal, and turkey for their bioactive compound content: Phenolics, glucosinolates, carotenoids, and chlorophylls. J. Sci. Food Agric. 2013, 93, 3478–3489. [Google Scholar] [CrossRef]
  103. Bentley-Hewitt, K.L.; Chen, R.K.Y.; Lill, R.E.; Hedderley, D.I.; Herath, T.D.; Matich, A.J.; Mckenzie, M.J. Consumption of selenium-enriched broccoli increases cytokine production in human peripheral blood mononuclear cells stimulated ex vivo, a preliminary human intervention study. Mol. Nutr. Food Res. 2014, 58, 2350–2357. [Google Scholar] [CrossRef]
  104. Lippmann, D.; Lehmann, C.; Florian, S.; Barknowitz, G.; Haack, M.; Mewis, I.; Wiesner, M.; Schreiner, M.; Glatt, H.; Brigelius-Flohé, R.; et al. Glucosinolates from pak choi and broccoli induce enzymes and inhibit inflammation and colon cancer differently. Food Funct. 2014, 5, 1073–1081. [Google Scholar] [CrossRef]
  105. Herz, C.; Márton, M.R.; Tran, H.T.T.; Gründemann, C.; Schell, J.; Lamy, E. Benzyl isothiocyanate but not benzyl nitrile from Brassicales plants dually blocks the COX and LOX pathway in primary human immune cells. J. Funct. Foods 2016, 23, 135–143. [Google Scholar] [CrossRef]
  106. Guzmán-Pérez, V.; Bumke-Vogt, C.; Schreiner, M.; Mewis, I.; Borchert, A.; Pfeiffer, A.F.H. Benzylglucosinolate Derived Isothiocyanate from Tropaeolum majus Reduces Gluconeogenic Gene and Protein Expression in Human Cells. PLoS ONE 2016, 11, e0162379. [Google Scholar] [CrossRef] [PubMed]
  107. Abbaoui, B.; Lucas, C.R.; Riedl, K.M.; Clinton, S.K.; Mortazavi, A. Cruciferous Vegetables, Isothiocyanates, and Bladder Cancer Prevention. Mol. Nutr. Food Res. 2018, 62, 1800079. [Google Scholar] [CrossRef] [PubMed]
  108. Waterman, C.; Rojas-Silva, P.; Tumer, T.B.; Kuhn, P.; Richard, A.J.; Wicks, S.; Stephens, J.M.; Wang, Z.; Mynatt, R.; Cefalu, W.; et al. Isothiocyanate-rich Moringa oleifera extract reduces weight gain, insulin resistance, and hepatic gluconeogenesis in mice. Mol. Nutr. Food Res. 2015, 59, 1013–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Jin, S.W.; Rahim, M.A.; Kim, H.T.; Park, J.I.; Kang, J.G.; Nou, I.S.; Cheng, Z.M. Molecular analysis of anthocyanin-related genes in ornamental cabbage. Genome 2018, 61, 111–120. [Google Scholar] [CrossRef] [Green Version]
  110. Jin, S.W.; Rahim, M.A.; Jung, H.J.; Afrin, K.S.; Kim, H.T.; Park, J.I.; Kang, J.G.; Nou, I.S. Abscisic acid and ethylene biosynthesis-related genes are associated with anthocyanin accumulation in purple ornamental cabbage (Brassica oleracea var. acephala). Genome 2019, 62, 513–526. [Google Scholar] [CrossRef] [Green Version]
  111. Zhang, B.; Hu, Z.; Zhang, Y.; Li, Y.; Zhou, S.; Chen, G. A putative functional MYB transcription factor induced by low temperature regulates anthocyanin biosynthesis in purple kale (Brassica oleracea var. acephala f. tricolor). Plant Cell Rep. 2012, 31, 281–289. [Google Scholar] [CrossRef]
  112. Mageney, V.; Baldermann, S.; Albach, D.C. Intraspecific Variation in Carotenoids of Brassica oleracea var. sabellica. J. Agric. Food Chem. 2016, 64, 3251–3257. [Google Scholar] [CrossRef]
  113. Zheng, Y.J.; Zhang, Y.T.; Liu, H.C.; Li, Y.M.; Liu, Y.L.; Hao, Y.W.; Lei, B.F. Supplemental blue light increases growth and quality of greenhouse pak choi depending on cultivar and supplemental light intensity. J. Integr. Agric. 2018, 17, 2245–2256. [Google Scholar] [CrossRef] [Green Version]
  114. Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef] [Green Version]
  115. McCance, K.R.; Flanigan, P.M.; Quick, M.M.; Niemeyer, E.D. Influence of plant maturity on anthocyanin concentrations, phenolic composition, and antioxidant properties of 3 purple basil (Ocimum basilicum L.) cultivars. J. Food Compos. Anal. 2016, 53, 30–39. [Google Scholar] [CrossRef] [Green Version]
  116. Fernandes, F.; Pereira, E.; Círić, A.; Soković, M.; Calhelha, R.C.; Barros, L.; Ferreira, I.C.F.R. Ocimum basilicum var. purpurascens leaves (red rubin basil): A source of bioactive compounds and natural pigments for the food industry. Food Funct. 2019, 10, 3161–3171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Fujiwara, Y.; Kono, M.; Ito, A.; Ito, M. Anthocyanins in perilla plants and dried leaves. Phytochemistry 2018, 147, 158–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Lu, N.; Takagaki, M.; Yamori, W.; Kagawa, N. Flavonoid Productivity Optimized for Green and Red Forms of Perilla frutescens via Environmental Control Technologies in Plant Factory. J. Food Qual. 2018, 2018, 4270279. [Google Scholar] [CrossRef] [Green Version]
  119. He, Y.K.; Yao, Y.Y.; Chang, Y.N. Characterization of anthocyanins in Perilla frutescens var. acuta Extract by Advanced UPLC-ESI-IT-TOF-MSn Method and their anticancer bioactivity. Molecules 2015, 20, 9155–9169. [Google Scholar] [CrossRef] [Green Version]
  120. Cui, L.; Zhang, Z.; Li, H.; Li, N.; Li, X.; Chen, T. Optimization of ultrasound assisted extraction of phenolic compounds and anthocyanins from perilla leaves using response surface methodology. Food Sci. Technol. Res. 2017, 23, 535–543. [Google Scholar] [CrossRef] [Green Version]
  121. Stintzing, F.C.; Carle, R. Betalains - emerging prospects for food scientists. Trends Food Sci. Technol. 2007, 18, 514–525. [Google Scholar] [CrossRef]
  122. Khan, M.I.; Giridhar, P. Plant betalains: Chemistry and biochemistry. Phytochemistry 2015, 117, 267–295. [Google Scholar] [CrossRef]
  123. Kugler, F.; Stintzing, F.C.; Carle, R. Identification of Betalains from Petioles of Differently Colored Swiss Chard (Beta vulgaris L. ssp. cicla [L.] Alef. Cv. Bright Lights) by High-Performance Liquid Chromatography-Electrospray Ionization Mass Spectrometry. J. Agric. Food Chem. 2004, 52, 2975–2981. [Google Scholar] [CrossRef]
  124. Raiola, A.; Rigano, M.M.; Calafiore, R.; Frusciante, L.; Barone, A. Enhancing the health-promoting effects of tomato fruit for biofortified food. Mediat. Inflamm. 2014, 2014, 139873. [Google Scholar] [CrossRef]
  125. Kotíková, Z.; Lachman, J.; Hejtmánková, A.; Hejtmánková, K. Determination of antioxidant activity and antioxidant content in tomato varieties and evaluation of mutual interactions between antioxidants. LWT Food Sci. Technol. 2011, 44, 1703–1710. [Google Scholar] [CrossRef]
  126. Borghesi, E.; Ferrante, A.; Gordillo, B.; Rodríguez-Pulido, F.J.; Cocetta, G.; Trivellini, A.; Mensuali-Sodi, A.; Malorgio, F.; Heredia, F.J. Comparative physiology during ripening in tomato rich-anthocyanins fruits. Plant Growth Regul. 2016, 80, 207–214. [Google Scholar] [CrossRef]
  127. Khachik, F.; Carvalho, L.; Bernstein, P.S.; Muir, G.J.; Zhao, D.Y.; Katz, N.B. Chemistry, distribution, and metabolism of tomato carotenoids and their impact on human health. Exp. Biol. Med. 2002, 227, 845–851. [Google Scholar] [CrossRef] [PubMed]
  128. Li, F.; Song, X.; Wu, L.; Chen, H.; Liang, Y.; Zhang, Y. Heredities on fruit color and pigment content between green and purple fruits in tomato. Sci. Hortic. 2018, 235, 391–396. [Google Scholar] [CrossRef]
  129. Rao, A.V.; Rao, L.G. Carotenoids and human health. Pharmacol. Res. 2007, 55, 207–216. [Google Scholar] [CrossRef]
  130. Blando, F.; Berland, H.; Maiorano, G.; Durante, M.; Mazzucato, A.; Picarella, M.E.; Nicoletti, I.; Gerardi, C.; Mita, G.; Andersen, Ø.M. Nutraceutical Characterization of Anthocyanin-Rich Fruits Produced by “Sun Black” Tomato Line. Front. Nutr. 2019, 6, 133. [Google Scholar] [CrossRef]
  131. Povero, G.; Gonzali, S.; Bassolino, L.; Mazzucato, A.; Perata, P. Transcriptional analysis in high-anthocyanin tomatoes reveals synergistic effect of Aft and atv genes. J. Plant Physiol. 2011, 168, 270–279. [Google Scholar] [CrossRef]
  132. Maligeppagol, M.; Sharath Chandra, G.; Navale, P.M.; Deepa, H.; Rajeev, P.R.; Asokan, R.; Prasad Babu, K.; Bujji Babu, C.S.; Keshava Rao, V.; Krishna Kumar, N.K. Anthocyanin enrichment of tomato (Solanum lycopersicum L.) fruit by metabolic engineering. Curr. Sci. 2013, 105, 72–80. [Google Scholar]
  133. Sestari, I.; Zsögön, A.; Rehder, G.G.; de Lira Teixeira, L.; Hassimotto, N.M.A.; Purgatto, E.; Benedito, V.A.; Peres, L.E.P. Near-isogenic lines enhancing ascorbic acid, anthocyanin and carotenoid content in tomato (Solanum lycopersicum L. cv Micro-Tom) as a tool to produce nutrient-rich fruits. Sci. Hortic. 2014, 175, 111–120. [Google Scholar] [CrossRef]
  134. Gonzali, S.; Mazzucato, A.; Perata, P. Purple as a tomato: Towards high anthocyanin tomatoes. Trends Plant Sci. 2009, 14, 237–241. [Google Scholar] [CrossRef] [Green Version]
  135. Li, H.; Deng, Z.; Liu, R.; Loewen, S.; Tsao, R. Bioaccessibility, in vitro antioxidant activities and in vivo anti-inflammatory activities of a purple tomato (Solanum lycopersicum L.). Food Chem. 2014, 159, 353–360. [Google Scholar] [CrossRef] [PubMed]
  136. Li, H.; Deng, Z.; Liu, R.; Young, J.C.; Zhu, H.; Loewen, S.; Tsao, R. Characterization of phytochemicals and antioxidant activities of a purple tomato (Solanum lycopersicum L.). J. Agric. Food Chem. 2011, 59, 11803–11811. [Google Scholar] [CrossRef] [PubMed]
  137. Zhou, K.; Yu, L. Total phenolic contents and antioxidant properties of commonly consumed vegetables grown in Colorado. LWT Food Sci. Technol. 2006, 39, 1155–1162. [Google Scholar] [CrossRef]
  138. Raffo, A.; La Malfa, G.; Fogliano, V.; Maiani, G.; Quaglia, G. Seasonal variations in antioxidant components of cherry tomatoes (Lycopersicon esculentum cv. Naomi F1). J. Food Compos. Anal. 2006, 19, 11–19. [Google Scholar] [CrossRef]
  139. Capanoglu, E.; Beekwilder, J.; Boyacioglu, D.; Hall, R.; De Vos, R. Changes in antioxidant and metabolite profiles during production of tomato paste. J. Agric. Food Chem. 2008, 56, 964–973. [Google Scholar] [CrossRef]
  140. Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef]
  141. Stewart, Z.A.; Westfall, M.D.; Pietenpol, J.A. Cell-cycle dysregulation and anticancer therapy. Trends Pharmacol. Sci. 2003, 24, 139–145. [Google Scholar] [CrossRef]
  142. Gupta, S.C.; Kannappan, R.; Reuter, S.; Kim, J.H.; Aggarwal, B.B. Chemosensitization of tumors by resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 150–160. [Google Scholar] [CrossRef] [Green Version]
  143. Lin, H.Y.; Tang, H.Y.; Davis, F.B.; Davis, P.J. Resveratrol and apoptosis. Ann. N. Y. Acad. Sci. 2011, 1215, 79–88. [Google Scholar] [CrossRef]
  144. Szkudelski, T.; Szkudelska, K. Anti-diabetic effects of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 34–39. [Google Scholar] [CrossRef] [PubMed]
  145. Fuentes, E.; Palomo, I. Antiplatelet effects of natural bioactive compounds by multiple targets: Food and drug interactions. J. Funct. Foods 2014, 6, 73–81. [Google Scholar] [CrossRef]
  146. Vagula, J.M.; Bertozzi, J.; Castro, J.C.; de Oliveira, C.C.; Clemente, E.; de Oliveira Santos Júnior, O.; Visentainer, J.V. Determination of trans-resveratrol in Solanum americanum Mill. by HPLC. Nat. Prod. Res. 2016, 30, 2230–2234. [Google Scholar] [CrossRef] [PubMed]
  147. Vasco, C.; Avila, J.; Ruales, J.; Svanberg, U.; Kamal-Eldin, A. Physical and chemical characteristics of golden-yellow and purple-red varieties of tamarillo fruit (Solanum betaceum Cav.). Int. J. Food Sci. Nutr. 2009, 60, 278–288. [Google Scholar] [CrossRef] [PubMed]
  148. Ninfali, P.; Mea, G.; Giorgini, S.; Rocchi, M.; Bacchiocca, M. Antioxidant capacity of vegetables, spices and dressings relevant to nutrition. Br. J. Nutr. 2005, 93, 257–266. [Google Scholar] [CrossRef] [Green Version]
  149. Riso, P.; Brusamolino, A.; Contino, D.; Martini, D.; Vendrame, S.; Del Bo’, C.; Porrini, M. Lycopene absorption in humans after the intake of two different single-dose lycopene formulations. Pharmacol. Res. 2010, 62, 318–321. [Google Scholar] [CrossRef]
  150. Reifen, R.; Nissenkorn, A.; Matas, Z.; Bujanover, Y. 5-ASA and lycopene decrease the oxidative stress and inflammation induced by iron in rats with colitis. J. Gastroenterol. 2004, 39, 514–519. [Google Scholar] [CrossRef]
  151. Hazra, P.; Longjam, M.; Chattopadhyay, A. Stacking of mutant genes in the development of “purple tomato” rich in both lycopene and anthocyanin contents. Sci. Hortic. 2018, 239, 253–258. [Google Scholar] [CrossRef]
  152. Guil-Guerrero, J.L.; Rebolloso-Fuentes, M.M. Nutrient composition and antioxidant activity of eight tomato (Lycopersicon esculentum) varieties. J. Food Compos. Anal. 2009, 22, 123–129. [Google Scholar] [CrossRef]
  153. Castañeda-Ovando, A.; de Louedes Pacheco-Hernández, M.; Páez-Hernández, M.E.; Rodríguez, J.A.; Galán-Vidal, C.A. Chemical studies of anthocyanins: A review. Food Chem. 2009, 113, 859–871. [Google Scholar] [CrossRef]
  154. Li, H.; Deng, Z.; Zhu, H.; Hu, C.; Liu, R.; Young, J.C.; Tsao, R. Highly pigmented vegetables: Anthocyanin compositions and their role in antioxidant activities. Food Res. Int. 2012, 46, 250–259. [Google Scholar] [CrossRef]
  155. Gomes, A.; Fernandes, E.; Lima, J.; Mira, L.; Corvo, M. Molecular Mechanisms of Anti-Inflammatory Activity Mediated by Flavonoids. Curr. Med. Chem. 2008, 15, 1586–1605. [Google Scholar] [CrossRef] [PubMed]
  156. Espin, S.; Gonzalez-Manzano, S.; Taco, V.; Poveda, C.; Ayuda-Durán, B.; Gonzalez-Paramas, A.M.; Santos-Buelga, C. Phenolic composition and antioxidant capacity of yellow and purple-red Ecuadorian cultivars of tree tomato (Solanum betaceum Cav.). Food Chem. 2016, 194, 1073–1080. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, Y.; Butelli, E.; De Stefano, R.; Schoonbeek, H.J.; Magusin, A.; Pagliarani, C.; Wellner, N.; Hill, L.; Orzaez, D.; Granell, A.; et al. Anthocyanins double the shelf life of tomatoes by delaying overripening and reducing susceptibility to gray mold. Curr. Biol. 2013, 23, 1094–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Wang, S.Y.; Lin, H.S. Antioxidant activity in fruits and leaves of blackberry, raspberry, and strawberry varies with cultivar and developmental stage. J. Agric. Food Chem. 2000, 48, 140–146. [Google Scholar] [CrossRef] [PubMed]
  159. Bae, S.H.; Suh, H.J. Antioxidant activities of five different mulberry cultivars in Korea. LWT Food Sci. Technol. 2007, 40, 955–962. [Google Scholar] [CrossRef]
  160. Jones, C.M.; Mes, P.; Myers, J.R. Characterization and Inheritance of the Anthocyanin fruit (Aft) Tomato. J. Hered. 2003, 94, 449–456. [Google Scholar] [CrossRef] [Green Version]
  161. Sapir, M.; Oren-Shamir, M.; Ovadia, R.; Reuveni, M.; Evenor, D.; Tadmor, Y.; Nahon, S.; Shlomo, H.; Chen, L.; Meir, A.; et al. Molecular aspects of Anthocyanin fruit tomato in relation to high pigment-1. J. Hered. 2008, 99, 292–303. [Google Scholar] [CrossRef] [Green Version]
  162. Lister, C.E.; Morrison, S.C.; Kerkhofs, N.S.; Wright, K.M. The Nutritional Composition and Health Benefits of New Zealand Tamarillos; Crop & Food Research: Wellington, New Zealand, 2005. [Google Scholar]
  163. Mertz, C.; Gancel, A.L.; Gunata, Z.; Alter, P.; Dhuique-Mayer, C.; Vaillant, F.; Perez, A.M.; Ruales, J.; Brat, P. Phenolic compounds, carotenoids and antioxidant activity of three tropical fruits. J. Food Compos. Anal. 2009, 22, 381–387. [Google Scholar] [CrossRef]
  164. Osorio, C.; Hurtado, N.; Dawid, C.; Hofmann, T.; Heredia-Mira, F.J.; Morales, A.L. Chemical characterisation of anthocyanins in tamarillo (Solanum betaceum Cav.) and Andes berry (Rubus glaucus Benth.) fruits. Food Chem. 2012, 132, 1915–1921. [Google Scholar] [CrossRef]
  165. Crozier, A.; Jaganath, I.B.; Clifford, M.N. Dietary phenolics: Chemistry, bioavailability and effects on health. Nat. Prod. Rep. 2009, 26, 1001–1043. [Google Scholar] [CrossRef]
  166. Petersen, M. Rosmarinic acid: New aspects. Phytochem. Rev. 2013, 12, 207–227. [Google Scholar] [CrossRef]
  167. De Rosso, V.V.; Mercadante, A.Z. HPLC-PDA-MS/MS of anthocyanins and carotenoids from dovyalis and tamarillo fruits. J. Agric. Food Chem. 2007, 55, 9135–9141. [Google Scholar] [CrossRef]
  168. Samuels, J. Biodiversity of food species of the solanaceae family: A preliminary taxonomic inventory of subfamily Solanoideae. Resources 2015, 4, 277–322. [Google Scholar] [CrossRef] [Green Version]
  169. Mohy-Ud-Din, A.; Khan, Z.U.D.; Ahmad, M.; Kashmiri, M.A. Chemotaxonomic value of alkaloids in Solanum nigrum complex. Pak. J. Bot. 2010, 42, 653–660. [Google Scholar]
  170. Xu, X.; Conrad, C.; Doktor, D. Optimising phenological metrics extraction for different crop types in Germany using the moderate resolution imaging Spectrometer (MODIS). Remote Sens. 2017, 9, 254. [Google Scholar] [CrossRef] [Green Version]
  171. Gramazio, P.; Prohens, J.; Plazas, M.; Andjar, I.; Herraiz, F.J.; Castillo, E.; Knapp, S.; Meyer, R.S.; Vilanova, S. Location of chlorogenic acid biosynthesis pathway and polyphenol oxidase genes in a new interspecific anchored linkage map of eggplant. BMC Plant Biol. 2014, 14, 350. [Google Scholar] [CrossRef] [Green Version]
  172. Cao, G.; Sofic, E.; Prior, R.L. Antioxidant Capacity of Tea and Common Vegetables. J. Agric. Food Chem. 1996, 44, 3426–3431. [Google Scholar] [CrossRef]
  173. Akanitapichat, P.; Phraibung, K.; Nuchklang, K.; Prompitakkul, S. Antioxidant and hepatoprotective activities of five eggplant varieties. Food Chem. Toxicol. 2010, 48, 3017–3021. [Google Scholar] [CrossRef]
  174. Whitaker, B.D.; Stommel, J.R. Distribution of hydroxycinnamic acid conjugates in fruit of commercial eggplant (Solanum melongena L.) cultivars. J. Agric. Food Chem. 2003, 51, 3448–3454. [Google Scholar] [CrossRef]
  175. Gajewski, M.; Katarzyna, K.; Bajer, M. The influence of postharvest storage on quality characteristics of fruit of eggplant cultivars. Not. Bot. Horti Agrobot. Cluj-Napoca 2009, 37, 200–205. [Google Scholar]
  176. Nisha, P.; Abdul Nazar, P.; Jayamurthy, P. A comparative study on antioxidant activities of different varieties of Solanum melongena. Food Chem. Toxicol. 2009, 47, 2640–2644. [Google Scholar] [CrossRef]
  177. Zaro, M.J.; Keunchkarian, S.; Chaves, A.R.; Vicente, A.R.; Concellón, A. Changes in bioactive compounds and response to postharvest storage conditions in purple eggplants as affected by fruit developmental stage. Postharvest Biol. Technol. 2014, 96, 110–117. [Google Scholar] [CrossRef]
  178. Plazas, M.; Andújar, I.; Vilanova, S.; Hurtado, M.; Gramazio, P.; Herraiz, F.J.; Prohens, J. Breeding for Chlorogenic Acid Content in Eggplant: Interest and Prospects. Not. Bot. Horti Agrobot. Cluj-Napoca 2013, 41, 26–35. [Google Scholar] [CrossRef] [Green Version]
  179. Concellón, A.; Zaro, M.J.; Chaves, A.R.; Vicente, A.R. Changes in quality and phenolic antioxidants in dark purple American eggplant (Solanum melongena L. cv. Lucía) as affected by storage at 0 °C and 10 °C. Postharvest Biol. Technol. 2012, 66, 35–41. [Google Scholar] [CrossRef]
  180. Zaro, M.J.; Ortiz, L.C.; Keunchkarian, S.; Chaves, A.R.; Vicente, A.R.; Concellón, A. Chlorogenic acid retention in white and purple eggplant after processing and cooking. LWT Food Sci. Technol. 2015, 64, 802–808. [Google Scholar] [CrossRef]
  181. Sadilova, E.; Stintzing, F.C.; Carle, R. Anthocyanins, colour and antioxidant properties of eggplant (Solanum melongena L.) and violet pepper (Capsicum annuum L.) peel extracts. Z. Nat. Sect. C J. Biosci. 2006, 61, 527–535. [Google Scholar] [CrossRef]
  182. Liu, Y.; Lv, J.; Liu, Z.; Wang, J.; Yang, B.; Chen, W.; Ou, L.; Dai, X.; Zhang, Z.; Zou, X. Integrative analysis of metabolome and transcriptome reveals the mechanism of color formation in pepper fruit (Capsicum annuum L.). Food Chem. 2020, 306, 125629. [Google Scholar] [CrossRef]
  183. Mennella, G.; Lo Scalzo, R.; Fibiani, M.; DAlessandro, A.; Francese, G.; Toppino, L.; Acciarri, N.; De Almeida, A.E.; Rotino, G.L. Chemical and bioactive quality traits during fruit ripening in eggplant (S. melongena L.) and allied species. J. Agric. Food Chem. 2012, 60, 11821–11831. [Google Scholar] [CrossRef]
  184. Prohens, J.; Whitaker, B.D.; Plazas, M.; Vilanova, S.; Hurtado, M.; Blasco, M.; Gramazio, P.; Stommel, J.R. Genetic diversity in morphological characters and phenolic acids content resulting from an interspecific cross between eggplant, Solanum melongena, and its wild ancestor (S. incanum). Ann. Appl. Biol. 2013, 162, 242–257. [Google Scholar] [CrossRef]
  185. Xi-Ou, X.; Wenqiu, L.; Wei, L.; Xiaoming, G.; Lingling, L.; Feiyue, M.; Yuge, L. The analysis of physiological variations in M2 generation of Solanum melongena L. mutagenized by ethyl methane sulfonate. Front. Plant Sci. 2017, 8, 17. [Google Scholar] [CrossRef] [Green Version]
  186. Zhang, Y.; Hu, Z.; Chu, G.; Huang, C.; Tian, S.; Zhao, Z.; Chen, G. Anthocyanin accumulation and molecular analysis of anthocyanin biosynthesis-associated genes in eggplant (Solanum melongena L.). J. Agric. Food Chem. 2014, 62, 2906–2912. [Google Scholar] [CrossRef]
  187. Noda, Y.; Kneyuki, T.; Igarashi, K.; Mori, A.; Packer, L. Antioxidant activity of nasunin, an anthocyanin in eggplant peels. Toxicology 2000, 148, 119–123. [Google Scholar] [CrossRef]
  188. Sudheesh, S.; Presannakumar, G.; Vijayakumar, S.; Vijayalakshmi, N.R. Hypolipidemic effect of flavonoids from Solanum melongena. Plant Foods Hum. Nutr. 1997, 51, 321–330. [Google Scholar] [CrossRef]
  189. Matsubara, K.; Kaneyuki, T.; Miyake, T.; Mori, M. Antiangiogenic activity of nasunin, an antioxidant anthocyanin, in eggplant peels. J. Agric. Food Chem. 2005, 53, 6272–6275. [Google Scholar] [CrossRef]
  190. Han, S.W.; Tae, J.; Kim, J.A.; Kim, D.K.; Seo, G.S.; Yun, K.J.; Choi, S.C.; Kim, T.H.; Nah, Y.H.; Lee, Y.M. The aqueous extract of Solanum melongena inhibits PAR2 agonist-induced inflammation. Clin. Chim. Acta 2003, 328, 39–44. [Google Scholar] [CrossRef]
  191. Mutalik, S.; Paridhavi, K.; Rao, C.M.; Udupa, N. Antipyretic and analgesic effect of leaves of Solanum melongena Linn. in Rodents. Indian J. Pharmacol. 2003, 35, 312–315. [Google Scholar]
  192. Igwe, S.A.; Akunyili, D.N.; Ogbogu, C. Effects of Solanum melongena (garden egg) on some visual functions of visually active Igbos of Nigeria. J. Ethnopharmacol. 2003, 86, 135–138. [Google Scholar] [CrossRef]
  193. Nadeem, M.; Anjum, F.M.; Khan, M.R.; Saeed, M.; Riaz, A. Antioxidant Potential of Bell Pepper (Capsicum annum L.)—A Review. Pak. J. Food Sci. 2011, 21, 45–51. [Google Scholar]
  194. Howard, L.R.; Wildman, R.E.C. Antioxidant vitamin and phytochemical content of fresh and processed pepper fruit (Capsicum annuum). In Handbook of Nutraceuticals and Functional Foods, 2nd ed.; CRC Press: Boca Raton, FL, USA, 2016; pp. 165–191. ISBN 9781420006186. [Google Scholar]
  195. Topuz, A.; Ozdemir, F. Assessment of carotenoids, capsaicinoids and ascorbic acid composition of some selected pepper cultivars (Capsicum annuum L.) grown in Turkey. J. Food Compos. Anal. 2007, 20, 596–602. [Google Scholar] [CrossRef]
  196. Matsufuji, H.; Ishikawa, K.; Nunomura, O.; Chino, M.; Takeda, M. Anti-oxidant content of different coloured sweet peppers, white, green, yellow, orange and red (Capsicum annuum L.). Int. J. Food Sci. Technol. 2007, 42, 1482–1488. [Google Scholar] [CrossRef]
  197. Wahyuni, Y.; Ballester, A.R.; Sudarmonowati, E.; Bino, R.J.; Bovy, A.G. Metabolite biodiversity in pepper (Capsicum) fruits of thirty-two diverse accessions: Variation in health-related compounds and implications for breeding. Phytochemistry 2011, 72, 1358–1370. [Google Scholar] [CrossRef]
  198. Delgado-Vargas, F.; Paredes-Lopez, O. Natural Colorants for Food and Nutraceutical Uses; CRC Press: Boca Raton, FL, USA, 2003; ISBN 1587160765. [Google Scholar]
  199. Lightbourn, G.J.; Griesbach, R.J.; Novotny, J.A.; Clevidence, B.A.; Rao, D.D.; Stommel, J.R. Effects of anthocyanin and carotenoid combinations on foliage and immature fruit color of Capsicum annuum L. J. Hered. 2008, 99, 105–111. [Google Scholar] [CrossRef] [Green Version]
  200. Aza-González, C.; Núñez-Palenius, H.G.; Ochoa-Alejo, N. Molecular biology of chili pepper anthocyanin biosynthesis. J. Mex. Chem. Soc. 2012, 56, 93–98. [Google Scholar] [CrossRef]
  201. Borovsky, Y.; Oren-Shamir, M.; Ovadia, R.; De Jong, W.; Paran, I. The A locus that controls anthocyanin accumulation in pepper encodes a MYB transcription factor homologous to Anthocyanin2 of Petunia. Theor. Appl. Genet. 2004, 109, 23–29. [Google Scholar] [CrossRef]
  202. Aza-González, C.; Ochoa-Alejo, N. Characterization of anthocyanins from fruits of two Mexican chili peppers (Capsicum annuum L.). J. Mex. Chem. Soc. 2012, 56, 149–151. [Google Scholar] [CrossRef]
  203. Ghasemnezhad, M.; Sherafati, M.; Payvast, G.A. Variation in phenolic compounds, ascorbic acid and antioxidant activity of five coloured bell pepper (Capsicum annum) fruits at two different harvest times. J. Funct. Foods 2011, 3, 44–49. [Google Scholar] [CrossRef]
  204. Yuan, H.; Zhang, J.; Nageswaran, D.; Li, L. Carotenoid metabolism and regulation in horticultural crops. Hortic. Res. 2015, 2, 1–11. [Google Scholar] [CrossRef] [Green Version]
  205. Subagio, A.; Morita, N. Effects of protein isolate from hyacinth beans (Lablab purpureus (L.) Sweet) seeds on cake characteristics. Food Sci. Technol. Res. 2008, 14, 12–17. [Google Scholar] [CrossRef] [Green Version]
  206. Bhaisare, M. Antioxidant potential in Dolichose lablab L. (Lablab purpureus). Indian J. Appl. Res. 2019, 9, 35–36. [Google Scholar]
  207. Al-Snafi, A.E. The pharmacology and medical importance of Dolichos lablab (Lablab purpureus)—A review. IOSR J. Pharm. 2017, 7, 22–30. [Google Scholar] [CrossRef]
  208. Momim, M.; Habib, M.R.; Hasan, M.R.; Nayeem, J.; Uddon, N.; Rana, M.S. Anti-inflammatory, antioxidant and cytotoxicity potential of methanolic extract of two Bangladeshi mean Lablab purpureus (L.) sweet white and purple. Int. J. Pharm. Sci. Res. 2012, 3, 779–781. [Google Scholar]
  209. Deoda, R.; Pandya, H.; Patel, M.; Yadav, K.; Kadam, P.; Patil, M. Antilithiatic Activity of Leaves, Bulb and Stem Of Nymphea odorata and Dolichos Lablab beans. Res. J. Pharm. Biol. Chem. Sci. 2011, 3, 814–819. [Google Scholar]
  210. Soetan, K. Comparative evaluation of phytochemicals in the raw and aqueous crude extracts from seeds of three Lablab purpureus varieties. Afr. J. Plant Sci. 2012, 6, 410–415. [Google Scholar]
  211. Im, A.R.; Kim, Y.H.; Lee, H.W.; Song, K.H. Water Extract of Dolichos lablab Attenuates Hepatic Lipid Accumulation in a Cellular Nonalcoholic Fatty Liver Disease Model. J. Med. Food 2016, 19, 495–503. [Google Scholar] [CrossRef] [PubMed]
  212. Koo, S.H.; Choi, Y.; Choi, S.K.; Shin, Y.; Kim, B.G.; Lee, B.L. Purification and Characterization of Serine Protease Inhibitors from Dolichos lablab Seeds; Prevention Effects on Pseudomonal Elastase-Induced Septic Hypotension. BMB Rep. 2000, 33, 112–119. [Google Scholar]
  213. Cui, B.; Hu, Z.; Zhang, Y.; Hu, J.; Yin, W.; Feng, Y.; Xie, Q.; Chen, G. Anthocyanins and flavonols are responsible for purple color of Lablab purpureus (L.) sweet pods. Plant Physiol. Biochem. 2016, 103, 183–190. [Google Scholar] [CrossRef]
  214. Díaz-Batalla, L.; Widholm, J.M.; Fahey, G.C.; Castaño-Tostado, E.; Paredes-López, O. Chemical components with health implications in wild and cultivated Mexican common bean seeds (Phaseolus vulgaris L.). J. Agric. Food Chem. 2006, 54, 2045–2052. [Google Scholar] [CrossRef]
  215. Ombra, M.N.; D’Acierno, A.; Nazzaro, F.; Riccardi, R.; Spigno, P.; Zaccardelli, M.; Pane, C.; Maione, M.; Fratianni, F. Phenolic Composition and Antioxidant and Antiproliferative Activities of the Extracts of Twelve Common Bean (Phaseolus vulgaris L.) Endemic Ecotypes of Southern Italy before and after Cooking. Oxid. Med. Cell. Longev. 2016, 2016, 1398298. [Google Scholar] [CrossRef] [Green Version]
  216. Aquino-Bolaños, E.N.; García-Díaz, Y.D.; Chavez-Servia, J.L.; Carrillo-Rodríguez, J.C.; Vera-Guzmán, A.M.; Heredia-García, E. Anthocyanin, polyphenol, and flavonoid contents and antioxidant activity in Mexican common bean (Phaseolus vulgaris L.) landraces. Emirates J. Food Agric. 2016, 28, 581–588. [Google Scholar] [CrossRef] [Green Version]
  217. Tsuda, T.; Ohshima, K.; Kawakishi, S.; Osawa, T. Inhibition of Lipid Peroxidation and Radical Scavenging Effect of Anthocyanin Pigments Isolated from the Seeds of Phaseolus vulgaris L. In Food Factors for Cancer Prevention; Springer: Tokyo, Japan, 1997; pp. 318–322. [Google Scholar]
  218. Mazewski, C.; Liang, K.; Gonzalez de Mejia, E. Comparison of the effect of chemical composition of anthocyanin-rich plant extracts on colon cancer cell proliferation and their potential mechanism of action using in vitro, in silico, and biochemical assays. Food Chem. 2018, 242, 378–388. [Google Scholar] [CrossRef]
  219. Blanca, J.M.; Prohens, J.; Anderson, G.J.; Zuriaga, E.; Cañizares, J.; Nuez, F. AFLP and DNA sequence variation in an Andean domesticate, pepino (Solanum muricatum, Solanaceae): Implications for evolution and domestication. Am. J. Bot. 2007, 94, 1219–1229. [Google Scholar] [CrossRef] [PubMed]
  220. Mahato, S.; Gurung, S.; Chakravarty, S.; Chhetri, B.; Khawas, T. An introduction to Pepino (Solanum muricatum Aiton): Review. Int. J. Environ. Agric. Biotechnol. 2016, 1, 143–148. [Google Scholar]
  221. O’Donoghue, E.M.; Somerfield, S.D.; De Vré, L.A.; Heyes, J.A. Developmental and ripening-related effects on the cell wall of pepino (Solanum muricatum) fruit. J. Sci. Food Agric. 1997, 73, 455–463. [Google Scholar] [CrossRef]
  222. Hsu, C.C.; Guo, Y.R.; Wang, Z.H.; Yin, M.C. Protective effects of an aqueous extract from pepino (Solanum muricatum Ait.) in diabetic mice. J. Sci. Food Agric. 2011, 91, 1517–1522. [Google Scholar] [CrossRef] [PubMed]
  223. Mauro, R.P.; Agnello, M.; Rizzo, V.; Graziani, G.; Fogliano, V.; Leonardi, C.; Giuffrida, F. Recovery of eggplant field waste as a source of phytochemicals. Sci. Hortic. 2019, 261, 109023. [Google Scholar] [CrossRef]
  224. Petropoulos, S.A.; Di Gioia, F.; Ntatsi, G. Vegetable organosulfur compounds and their health promoting effects. Curr. Pharm. Des. 2017, 23, 2850–2875. [Google Scholar] [CrossRef] [PubMed]
  225. Di Gioia, F.; Avato, P.; Serio, F.; Argentieri, M.P. Glucosinolate profile of Eruca sativa, Diplotaxis tenuifolia and Diplotaxis erucoides grown in soil and soilless systems. J. Food Compos. Anal. 2018, 69, 197–204. [Google Scholar] [CrossRef]
  226. Di Gioia, F.; Rosskopf, E.N.; Leonardi, C.; Giuffrida, F. Effects of application timing of saline irrigation water on broccoli production and quality. Agric. Water Manag. 2018, 203, 97–104. [Google Scholar] [CrossRef]
  227. Yu, H.; Wang, J.; Sheng, X.; Zhao, Z.; Shen, Y.; Branca, F.; Gu, H. Construction of a high-density genetic map and identification of loci controlling purple sepal trait of flower head in Brassica oleracea L. italica. BMC Plant Biol. 2019, 19, 228. [Google Scholar] [CrossRef] [Green Version]
  228. Branca, F.; Chiarenza, G.L.; Cavallaro, C.; Gu, H.; Zhao, Z.; Tribulato, A. Diversity of Sicilian broccoli (Brassica oleracea var. italica) and cauliflower (Brassica oleracea var. botrytis) landraces and their distinctive bio-morphological, antioxidant, and genetic traits. Genet. Resour. Crop Evol. 2018, 65, 485–502. [Google Scholar] [CrossRef]
  229. Rahim, M.A.; Afrin, K.S.; Jung, H.J.; Kim, H.T.; Park, J.I.; Hur, Y.; Nou, I.S.; Deynze, A. Van Molecular analysis of anthocyanin biosynthesis-related genes reveal BoTT8 associated with purple hypocotyl of broccoli (Brassica oleracea var. italica L.). Genome 2019, 62, 253–266. [Google Scholar] [CrossRef] [PubMed]
  230. Terzo, M.N.; Pezzino, F.; Amodeo, L.; Catalano, D.; Viola, M.; Tribulato, A.; Travali, S.; Branca, F. Evaluation of a sicilian black broccoli extract on in vitro cell models. Acta Hortic. 2018, 1202, 135–142. [Google Scholar] [CrossRef]
  231. Rodríguez-Hernández, M.D.C.; Moreno, D.A.; Carvajal, M.; García-Viguera, C.; Martínez-Ballesta, M.D.C. Natural Antioxidants in Purple Sprouting Broccoli under Mediterranean Climate. J. Food Sci. 2012, 77, C1058–C1063. [Google Scholar] [CrossRef] [PubMed]
  232. Verkerk, R.; Tebbenhoff, S.; Dekker, M. Variation and distribution of glucosinolates in 42 cultivars of Brassica oleracea vegetable crops. Acta Hortic. 2010, 856, 63–69. [Google Scholar] [CrossRef] [Green Version]
  233. Moreno, D.A.; Pérez-Balibrea, S.; Ferreres, F.; Gil-Izquierdo, Á.; García-Viguera, C. Acylated anthocyanins in broccoli sprouts. Food Chem. 2010, 123, 358–363. [Google Scholar] [CrossRef]
  234. Kyriacou, M.C.; Rouphael, Y.; Di Gioia, F.; Kyratzis, A.; Serio, F.; Renna, M.; De Pascale, S.; Santamaria, P. Micro-scale vegetable production and the rise of microgreens. Trends Food Sci. Technol. 2016, 57, 103–115. [Google Scholar] [CrossRef]
  235. Di Gioia, F.; Renna, M.; Santamaria, P. Sprouts, Microgreens and “Baby Leaf” Vegetables; Springer: Boston, MA, USA, 2017; pp. 403–432. [Google Scholar]
  236. Di Gioia, F.; Petropoulos, S.A.; Ozores-Hampton, M.; Morgan, K.; Rosskopf, E.N. Zinc and Iron Agronomic Biofortification of Brassicaceae Microgreens. Agronomy 2019, 9, 677. [Google Scholar] [CrossRef] [Green Version]
  237. Crisp, P.; Jewell, P.A.; Gray, A.R. Improved selection against the purple colour defect of cauliflower curds. Euphytica 1975, 24, 177–180. [Google Scholar] [CrossRef]
  238. Branca, F. Cauliflower and Broccoli. In Vegetables I; Springer: New York, NY, USA, 2007; pp. 151–186. [Google Scholar]
  239. Lo Piero, A.R.; Lo Cicero, L.; Ragusa, L.; Branca, F. Change in the expression of anthocyanin pathway genes in developing inflorescences of sicilian landraces of pigmented broccoli and cauliflower. Acta Hortic. 2013, 1005, 253–260. [Google Scholar] [CrossRef]
  240. Chiu, L.W.; Li, L. Characterization of the regulatory network of BoMYB2 in controlling anthocyanin biosynthesis in purple cauliflower. Planta 2012, 236, 1153–1164. [Google Scholar] [CrossRef]
  241. Li, L.; Lu, S.; Cosman, K.M.; Earle, E.D.; Garvin, D.F.; O’Neill, J. β-Carotene accumulation induced by the cauliflower Or gene is not due to an increased capacity of biosynthesis. Phytochemistry 2006, 67, 1177–1184. [Google Scholar] [CrossRef] [PubMed]
  242. Li, L.; Paolillo, D.J.; Parthasarathy, M.V.; DiMuzio, E.M.; Garvin, D.F. A novel gene mutation that confers abnormal patterns of β-carotene accumulation in cauliflower (Brassica oleracea var. botrytis). Plant J. 2001, 26, 59–67. [Google Scholar] [CrossRef] [PubMed]
  243. Zhou, X.; Sun, T.H.; Wang, N.; Ling, H.Q.; Lu, S.; Li, L. The cauliflower Orange gene enhances petiole elongation by suppressing expression of eukaryotic release factor 1. New Phytol. 2011, 190, 89–100. [Google Scholar] [CrossRef] [PubMed]
  244. Lu, S.; Van Eck, J.; Zhou, X.; Lopez, A.B.; O’Halloran, D.M.; Cosman, K.M.; Conlin, B.J.; Paolillo, D.J.; Garvin, D.F.; Vrebalov, J.; et al. The cauliflower Or gene encodes a DnaJ cysteine-rich domain-containing protein that mediates high levels of β-carotene accumulation. Plant Cell 2006, 18, 3594–3605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Kapusta-Duch, J.; Szeląg-Sikora, A.; Sikora, J.; Niemiec, M.; Gródek-Szostak, Z.; Kuboń, M.; Leszczyńska, T.; Borczak, B. Health-Promoting Properties of Fresh and Processed Purple Cauliflower. Sustainability 2019, 11, 4008. [Google Scholar] [CrossRef] [Green Version]
  246. Ordás, A.; Cartea, M.E. Cabbage and Kale. In Vegetables I; Springer: New York, NY, USA, 2007; pp. 119–149. [Google Scholar]
  247. Yuan, Y.; Chiu, L.W.; Li, L. Transcriptional regulation of anthocyanin biosynthesis in red cabbage. Planta 2009, 230, 1141–1153. [Google Scholar] [CrossRef]
  248. Arapitsas, P.; Sjöberg, P.J.R.; Turner, C. Characterisation of anthocyanins in red cabbage using high resolution liquid chromatography coupled with photodiode array detection and electrospray ionization-linear ion trap mass spectrometry. Food Chem. 2008, 109, 219–226. [Google Scholar] [CrossRef]
  249. Wiczkowski, W.; Szawara-Nowak, D.; Topolska, J. Red cabbage anthocyanins: Profile, isolation, identification, and antioxidant activity. Food Res. Int. 2013, 51, 303–309. [Google Scholar] [CrossRef]
  250. Koss-Mikołajczyk, I.; Kusznierewicz, B.; Wiczkowski, W.; Płatosz, N.; Bartoszek, A. Phytochemical composition and biological activities of differently pigmented cabbage (Brassica oleracea var. capitata) and cauliflower (Brassica oleracea var. botrytis) varieties. J. Sci. Food Agric. 2019, 99, 5499–5507. [Google Scholar]
  251. Pliszka, B.; Huszcza-Ciołkowska, G.; Mieleszko, E.; Czaplicki, S. Stability and antioxidative properties of acylated anthocyanins in three cultivars of red cabbage (Brassica oleracea L. var. capitata L. f. rubra). J. Sci. Food Agric. 2009, 89, 1154–1158. [Google Scholar] [CrossRef]
  252. McDougall, G.J.; Fyffe, S.; Dobson, P.; Stewart, D. Anthocyanins from red cabbage—Stability to simulated gastrointestinal digestion. Phytochemistry 2007, 68, 1285–1294. [Google Scholar] [CrossRef]
  253. Charron, C.S.; Clevidence, B.A.; Britz, S.J.; Novotny, J.A. Effect of dose size on bioavailability of acylated and nonacylated anthocyanins from red cabbage (Brassica oleracea L. Var. capitata). J. Agric. Food Chem. 2007, 55, 5354–5362. [Google Scholar] [CrossRef] [PubMed]
  254. Volden, J.; Borge, G.I.A.; Bengtsson, G.B.; Hansen, M.; Thygesen, I.E.; Wicklund, T. Effect of thermal treatment on glucosinolates and antioxidant-related parameters in red cabbage (Brassica oleracea L. ssp. capitata f. rubra). Food Chem. 2008, 109, 595–605. [Google Scholar] [CrossRef]
  255. Rottenberg, A.; Zohary, D. Wild genetic resources of cultivated artichoke. Acta Hortic. 2005, 681, 307–314. [Google Scholar] [CrossRef]
  256. Sonnante, G.; Pignone, D.; Hammer, K. The domestication of artichoke and cardoon: From Roman times to the genomic age. Ann. Bot. 2007, 100, 1095–1100. [Google Scholar] [CrossRef] [Green Version]
  257. de Falco, B.; Incerti, G.; Amato, M.; Lanzotti, V. Artichoke: Botanical, agronomical, phytochemical, and pharmacological overview. Phytochem. Rev. 2015, 14, 993–1018. [Google Scholar] [CrossRef]
  258. Petropoulos, S.A.; Pereira, C.; Ntatsi, G.; Danalatos, N.; Barros, L.; Ferreira, I.C.F.R. Nutritional value and chemical composition of Greek artichoke genotypes. Food Chem. 2018, 267, 296–302. [Google Scholar] [CrossRef]
  259. Segovia, M.S.; Palma, M.A.; Leskovar, D.I. Factors affecting consumer preferences and willingness to pay for artichoke products. Acta Hortic. 2016, 1147, 271–280. [Google Scholar] [CrossRef]
  260. Lanteri, S.; Portis, E. Globe Artichoke and Cardoon. In Vegetables I; Springer: New York, NY, USA, 2007; pp. 49–74. [Google Scholar]
  261. Portis, E.; Mauro, R.P.; Acquadro, A.; Moglia, A.; Mauromicale, G.; Lanteri, S. The inheritance of bract pigmentation and fleshy thorns on the globe artichoke capitulum. Euphytica 2015, 206, 523–531. [Google Scholar] [CrossRef]
  262. Basnizki, J.; Zohary, D. Breeding of seed planted artichoke. Plant Breed. Rev. 1994, 12, 253–269. [Google Scholar]
  263. Pochard, E.; Foury, C.; Chambonet, D. Il miglioramento genetico del carciofo. In Proceedings of the I International Congress on Artichoke, Minerva Medica, Bari, Italy, 23–24 November 1969; pp. 117–143. [Google Scholar]
  264. Cravero, V.P.; Picardi, L.A.; Cointry, E.L. An approach for understanding the heredity of two quality traits (head color and tightness) in globe artichoke (Cynara scolymus L.). Genet. Mol. Biol. 2005, 28, 431–434. [Google Scholar] [CrossRef] [Green Version]
  265. De Palma, M.; Fratianni, F.; Nazzaro, F.; Tucci, M. Isolation and functional characterization of a novel gene coding for flavonoid 3′-hydroxylase from globe artichoke. Biol. Plant. 2014, 58, 445–455. [Google Scholar] [CrossRef]
  266. Blanco, E.; Sabetta, W.; Danzi, D.; Negro, D.; Passeri, V.; de Lisi, A.; Paolocci, F.; Sonnante, G. Isolation and Characterization of the Flavonol Regulator CcMYB12 From the Globe Artichoke [Cynara cardunculus var. scolymus (L.) Fiori]. Front. Plant Sci. 2018, 9, 941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Schütz, K.; Kammerer, D.; Carle, R.; Schieber, A. Identification and quantification of caffeoylquinic acids and flavonoids from artichoke (Cynara scolymus L.) heads, juice, and pomace by HPLC-DAD-ESI/MSn. J. Agric. Food Chem. 2004, 52, 4090–4096. [Google Scholar] [CrossRef]
  268. Pandino, G.; Lombardo, S.; Mauromicale, G.; Williamson, G. Profile of polyphenols and phenolic acids in bracts and receptacles of globe artichoke (Cynara cardunculus var. scolymus) germplasm. J. Food Compos. Anal. 2011, 24, 148–153. [Google Scholar] [CrossRef]
  269. Fratianni, F.; Tucci, M.; de Palma, M.; Pepe, R.; Nazzaro, F. Polyphenolic composition in different parts of some cultivars of globe artichoke (Cynara cardunculus L. var. scolymus (L.) Fiori). Food Chem. 2007, 104, 1282–1286. [Google Scholar] [CrossRef]
  270. Negro, D.; Montesano, V.; Grieco, S.; Crupi, P.; Sarli, G.; De Lisi, A.; Sonnante, G. Polyphenol Compounds in Artichoke Plant Tissues and Varieties. J. Food Sci. 2012, 77, C244–C252. [Google Scholar] [CrossRef]
  271. Schütz, K.; Persike, M.; Carle, R.; Schieber, A. Characterization and quantification of anthocyanins in selected artichoke (Cynara scolymus L.) cultivars by HPLC-DAD-ESI-MS n. Anal. Bioanal. Chem. 2006, 384, 1511–1517. [Google Scholar] [CrossRef]
  272. Bonasia, A.; Conversa, G.; Lazzizera, C.; Gambacorta, G.; Elia, A. Morphological and qualitative characterisation of globe artichoke head from new seed-propagated cultivars. J. Sci. Food Agric. 2010, 90, 2689–2693. [Google Scholar] [CrossRef]
  273. Dabbou, S.; Dabbou, S.; Flamini, G.; Pandino, G.; Gasco, L.; Helal, A.N. Phytochemical Compounds from the Crop Byproducts of Tunisian Globe Artichoke Cultivars. Chem. Biodivers. 2016, 13, 1475–1483. [Google Scholar] [CrossRef]
  274. Pandino, G.; Lombardo, S.; Mauromicale, G. Globe artichoke leaves and floral stems as a source of bioactive compounds. Ind. Crops Prod. 2013, 44, 44–49. [Google Scholar] [CrossRef]
  275. Garbetta, A.; Capotorto, I.; Cardinali, A.; D’Antuono, I.; Linsalata, V.; Pizzi, F.; Minervini, F. Antioxidant activity induced by main polyphenols present in edible artichoke heads: Influence of in vitro gastro-intestinal digestion. J. Funct. Foods 2014, 10, 456–464. [Google Scholar] [CrossRef]
  276. Gebhardt, R. Antioxidative and protective properties of extracts from leaves of the artichoke (Cynara scolymus L.) against hydroperoxide-induced oxidative stress in cultured rat hepatocytes. Toxicol. Appl. Pharmacol. 1997, 144, 279–286. [Google Scholar] [CrossRef] [PubMed]
  277. Li, D.; Zhang, Y.; Liu, Y.; Sun, R.; Xia, M. Purified Anthocyanin Supplementation Reduces Dyslipidemia, Enhances Antioxidant Capacity, and Prevents Insulin Resistance in Diabetic Patients. J. Nutr. 2015, 145, 742–748. [Google Scholar] [CrossRef]
  278. Jennings, A.; Welch, A.A.; Fairweather-Tait, S.J.; Kay, C.; Minihane, A.M.; Chowienczyk, P.; Jiang, B.; Cecelja, M.; Spector, T.; Macgregor, A.; et al. Higher anthocyanin intake is associated with lower arterial stiffness and central blood pressure in women. Am. J. Clin. Nutr. 2012, 96, 781–788. [Google Scholar] [CrossRef] [Green Version]
  279. Li, H.; Xia, N.; Brausch, I.; Yao, Y.; Förstermann, U. Flavonoids from artichoke (Cynara scolymus L.) up-regulate endothelial-type nitric-oxide synthase gene expression in human endothelial cells. J. Pharmacol. Exp. Ther. 2004, 310, 926–932. [Google Scholar] [CrossRef] [Green Version]
  280. Xia, N.; Pautz, A.; Wollscheid, U.; Reifenberg, G.; Förstermann, U.; Li, H. Artichoke, cynarin and cyanidin downregulate the expression of inducible nitric oxide synthase in human coronary smooth muscle cells. Molecules 2014, 19, 3654–3668. [Google Scholar] [CrossRef] [Green Version]
  281. Dufour, C.; Loonis, M.; Delosière, M.; Buffière, C.; Hafnaoui, N.; Santé-Lhoutellier, V.; Rémond, D. The matrix of fruit & vegetables modulates the gastrointestinal bioaccessibility of polyphenols and their impact on dietary protein digestibility. Food Chem. 2018, 240, 314–322. [Google Scholar]
  282. Cicero, A.F.G.; Colletti, A.; Bajraktari, G.; Descamps, O.; Djuric, D.M.; Ezhov, M.; Fras, Z.; Katsiki, N.; Langlois, M.; Latkovskis, G.; et al. Lipid lowering nutraceuticals in clinical practice: Position paper from an International Lipid Expert Panel. Arch. Med. Sci. 2017, 13, 965–1005. [Google Scholar] [CrossRef]
  283. D’Antuono, I.; Garbetta, A.; Linsalata, V.; Minervini, F.; Cardinali, A. Polyphenols from artichoke heads (Cynara cardunculus (L.) subsp. scolymus Hayek): In vitro bio-accessibility, intestinal uptake and bioavailability. Food Funct. 2015, 6, 1268–1277. [Google Scholar]
  284. Guo, Q.; Wang, N.; Liu, H.; Li, Z.; Lu, L.; Wang, C. The bioactive compounds and biological functions of Asparagus officinalis L.—A review. J. Funct. Foods 2019, 103727. [Google Scholar] [CrossRef]
  285. Conversa, G.; Miedico, O.; Chiaravalle, A.E.; Elia, A. Heavy metal contents in green spears of asparagus (Asparagus officinalis L.) grown in Southern Italy: Variability among farms, genotypes and effect of soil mycorrhizal inoculation. Sci. Hortic. 2019, 256, 108559. [Google Scholar] [CrossRef]
  286. Slatnar, A.; Mikulic-Petkovsek, M.; Stampar, F.; Veberic, R.; Horvat, J.; Jakse, M.; Sircelj, H. Game of tones: Sugars, organic acids, and phenolics in green and purple asparagus (Asparagus officinalis L.) cultivars. Turk. J. Agric. For. 2018, 42, 55–66. [Google Scholar] [CrossRef]
  287. Di Gioia, F.; Petropoulos, S.A. Phytoestrogens, Phytosteroids and Saponins in Vegetables: Biosynthesis, Functions, Health Effects And Practical Applications. In Functional Food Ingredients from Plants, 1st ed.; Ferreira, I.C.F.R., Barros, L., Eds.; Elsevier Inc.: London, UK, 2019; Volume 90. [Google Scholar]
  288. Dawid, C.; Hofmann, T. Structural and sensory characterization of bitter tasting steroidal saponins from asparagus spears (Asparagus officinalis L.). J. Agric. Food Chem. 2012, 60, 11889–11900. [Google Scholar] [CrossRef] [PubMed]
  289. Pegiou, E.; Mumm, R.; Acharya, P.; de Vos, R.C.H.; Hall, R.D. Green and White Asparagus (Asparagus officinalis): A Source of Developmental, Chemical and Urinary Intrigue. Metabolites 2020, 10, 17. [Google Scholar] [CrossRef] [PubMed]
  290. Sun, T.; Powers, J.R.; Tang, J. Evaluation of the antioxidant activity of asparagus, broccoli and their juices. Food Chem. 2007, 105, 101–106. [Google Scholar] [CrossRef]
  291. Maeda, T.; Kakuta, H.; Sonoda, T.; Motoki, S.; Ueno, R.; Suzuki, T.; Oosawa, K. Antioxidation capacities of extracts from green, purple, and white asparagus spears related to polyphenol concentration. HortScience 2005, 40, 1221–1224. [Google Scholar] [CrossRef] [Green Version]
  292. Geoffriau, E.; Denoue, D.; Rameau, C. Assessment of genetic variation among asparagus (Asparagus officinalis L.) populations and cultivars: Agromorphological and isozymic data. Euphytica 1991, 61, 169–179. [Google Scholar] [CrossRef]
  293. López Anido, F.; Cointry, E. Asparagus. In Handbook of Plant Breeding. Vegetable II: Fabaceae, Liliaceae, Solanaceae and Umbelliferae; Prohens, J., Nuez, F., Eds.; Springer: New York, NY, USA, 2008; p. 258. ISBN 9780387741086. [Google Scholar]
  294. Siomos, A.S.; Dogras, C.C.; Sfakiotakis, E.M. Color development in harvested white asparagus spears in relation to carbon dioxide and oxygen concentration. Postharvest Biol. Technol. 2001, 23, 209–214. [Google Scholar] [CrossRef]
  295. Moreno, R.; Espejo, J.A.; Cabrera, A.; Gil, J. Origin of tetraploid cultivated asparagus landraces inferred from nuclear ribosomal DNA internal transcribed spacers’ polymorphisms. Ann. Appl. Biol. 2008, 153, 233–241. [Google Scholar] [CrossRef]
  296. Benson, B.L.; Mullen, R.J.; Dean, B.B. Three new green asparagus cultivars; Apollo, atlas and grande and one purple cultivar, purple passion. Acta Hortic. 1996, 415, 59–65. [Google Scholar] [CrossRef]
  297. Falloon, P.G.; Andersen, A.M. Breeding purple asparagus from tetraploid “violetto D’Albenga.”. Acta Hortic. 1999, 479, 109–113. [Google Scholar] [CrossRef]
  298. Dong, T.; Han, R.; Yu, J.; Zhu, M.; Zhang, Y.; Gong, Y.; Li, Z. Anthocyanins accumulation and molecular analysis of correlated genes by metabolome and transcriptome in green and purple asparaguses (Asparagus officinalis, L.). Food Chem. 2019, 271, 18–28. [Google Scholar] [CrossRef] [PubMed]
  299. Flores, F.B.; Oosterhaven, J.; Martínez-Madrid, M.C.; Romojaro, F. Possible regulatory role of phenylalanine ammonia-lyase in the production of anthocyanins in asparagus (Asparagus officinalis L). J. Sci. Food Agric. 2005, 85, 925–930. [Google Scholar] [CrossRef]
  300. Mastropasqua, L.; Tanzarella, P.; Paciolla, C. Effects of postharvest light spectra on quality and health-related parameters in green Asparagus officinalis L. Postharvest Biol. Technol. 2016, 112, 143–151. [Google Scholar] [CrossRef]
  301. Huyskens-Keil, S.; Eichholz-Dündar, I.; Hassenberg, K.; Herppich, W.B. Impact of light quality (white, red, blue light and UV-C irradiation) on changes in anthocyanin content and dynamics of PAL and POD activities in apical and basal spear sections of white asparagus after harvest. Postharvest Biol. Technol. 2020, 161, 111069. [Google Scholar] [CrossRef]
  302. Barberis, A.; Cefola, M.; Pace, B.; Azara, E.; Spissu, Y.; Serra, P.A.; Logrieco, A.F.; D’hallewin, G.; Fadda, A. Postharvest application of oxalic acid to preserve overall appearance and nutritional quality of fresh-cut green and purple asparagus during cold storage: A combined electrochemical and mass-spectrometry analysis approach. Postharvest Biol. Technol. 2019, 148, 158–167. [Google Scholar] [CrossRef]
  303. Long, A.; Benz, B.F.; Donahue, D.J.; Jull, A.J.T. First direct AMS dates on early maize from Tehuacán Mexico. Radiocarbon 1989, 31, 1035–1040. [Google Scholar] [CrossRef] [Green Version]
  304. Pleasant, J.M. Traditional Iroquois Corn: Its History, Cultivation, and Use; Plant and Life Sciences Publishing: Ithaca, NY, USA, 2010; ISBN 9781933395142 1933395141. [Google Scholar]
  305. Petroni, K.; Pilu, R.; Tonelli, C. Anthocyanins in corn: A wealth of genes for human health. Planta 2014, 240, 901–911. [Google Scholar] [CrossRef]
  306. Brandolini, A.; Brandolini, A. Maize introduction, evolution and diffusion in Italy. Maydica 2009, 54, 233–242. [Google Scholar]
  307. United States Department of Agriculture; National Agricultural Statistics Service USDA-NASS, Data and Statistics. 2019. Available online: https://www.nass.usda.gov/Data_and_Statistics/index.php (accessed on 30 December 2019).
  308. Erwin, A.T. Sweet Corn—Mutant or historic species? Econ. Bot. 1951, 5, 302–306. [Google Scholar] [CrossRef]
  309. Lertrat, K.; Pulam, T. Breeding for Increased Sweetness in Sweet Corn. Int. J. Plant Breed. 2007, 1, 27–30. [Google Scholar]
  310. Juvik, J.A.; Yousef, G.G.; Han, T.H.; Tadmor, Y.; Azanza, F.; Tracy, W.F.; Barzur, A.; Rocheford, T.R. QTL Influencing Kernel Chemical Composition and Seedling Stand Establishment in Sweet Corn with the shrunken2 and sugary enhancerl Endosperm Mutations. J. Am. Soc. Hortic. Sci. 2003, 128, 864–875. [Google Scholar] [CrossRef] [Green Version]
  311. O’Hare, T.J.; Fanning, K.J.; Martin, I.F. Zeaxanthin biofortification of sweet-corn and factors affecting zeaxanthin accumulation and colour change. Arch. Biochem. Biophys. 2015, 572, 184–187. [Google Scholar] [CrossRef] [PubMed]
  312. Lago, C.; Landoni, M.; Cassani, E.; Atanassiu, S.; Canta-Luppi, E.; Pilu, R. Development and characterization of a coloured sweet corn line as a new functional food. Maydica 2014, 59, 191–200. [Google Scholar]
  313. Casas, M.I.; Duarte, S.; Doseff, A.I.; Grotewold, E. Flavone-rich maize: An opportunity to improve the nutritional value of an important commodity crop. Front. Plant Sci. 2014, 5, 440. [Google Scholar] [CrossRef] [Green Version]
  314. Lago, C.; Landoni, M.; Cassani, E.; Cantaluppi, E.; Doria, E.; Nielsen, E.; Giorgi, A.; Pilu, R. Study and characterization of an ancient European flint white maize rich in anthocyanins: Millo Corvo from Galicia. PLoS ONE 2015, 10, e0126521. [Google Scholar]
  315. Cassani, E.; Puglisi, D.; Cantaluppi, E.; Landoni, M.; Giupponi, L.; Giorgi, A.; Pilu, R. Genetic studies regarding the control of seed pigmentation of an ancient European pointed maize (Zea mays L.) rich in phlobaphenes: The “Nero Spinoso” from the Camonica valley. Genet. Resour. Crop Evol. 2017, 64, 761–773. [Google Scholar] [CrossRef]
  316. Lago, C.; Landoni, M.; Cassani, E.; Doria, E.; Nielsen, E.; Pilu, R. Study and characterization of a novel functional food: Purple popcorn. Mol. Breed. 2013, 31, 575–585. [Google Scholar] [CrossRef]
  317. Tsuda, T.; Horio, F.; Uchida, K.; Aoki, H.; Osawa, T. Dietary Cyanidin 3-O-β-D-Glucoside-Rich Purple Corn Color Prevents Obesity and Ameliorates Hyperglycemia in Mice. J. Nutr. 2003, 133, 2125–2130. [Google Scholar] [CrossRef]
  318. Tsuda, T. Dietary anthocyanin-rich plants: Biochemical basis and recent progress in health benefits studies. Mol. Nutr. Food Res. 2012, 56, 159–170. [Google Scholar] [CrossRef] [PubMed]
  319. Chatham, L.A.; Howard, J.E.; Juvik, J.A. A natural colorant system from corn: Flavone-anthocyanin copigmentation for altered hues and improved shelf life. Food Chem. 2019, 310, 125734. [Google Scholar] [CrossRef] [PubMed]
  320. Luna-Vital, D.; Li, Q.; West, L.; West, M.; Gonzalez de Mejia, E. Anthocyanin condensed forms do not affect color or chemical stability of purple corn pericarp extracts stored under different pHs. Food Chem. 2017, 232, 639–647. [Google Scholar] [CrossRef] [PubMed]
  321. Petroni, K.; Tonelli, C. Recent advances on the regulation of anthocyanin synthesis in reproductive organs. Plant Sci. 2011, 181, 219–229. [Google Scholar] [CrossRef]
  322. Sharma, M.; Chai, C.; Morohashi, K.; Grotewold, E.; Snook, M.E.; Chopra, S. Expression of flavonoid 3′-hydroxylase is controlled by P1, the regulator of 3-deoxyflavonoid biosynthesis in maize. BMC Plant Biol. 2012, 12, 196. [Google Scholar] [CrossRef] [Green Version]
  323. Wittmeyer, K.; Cui, J.; Chatterjee, D.; Lee, T.F.; Tan, Q.; Xue, W.; Jiao, Y.; Wang, P.H.; Gaffoor, I.; Ware, D.; et al. The dominant and poorly penetrant phenotypes of maize Unstable factor for orange1 are caused by DNA methylation changes at a linked transposon. Plant Cell 2018, 30, 3006–3023. [Google Scholar] [CrossRef] [Green Version]
  324. Calvo-Brenes, P.; O’Hare, T. Effect of freezing and cool storage on carotenoid content and quality of zeaxanthin-biofortified and standard yellow sweet-corn (Zea mays L.). J. Food Compos. Anal. 2020, 86, 103353. [Google Scholar] [CrossRef]
  325. Janovská, D.; Štočková, L.; Stehno, Z. Prehranske lastnosti mladih rastlin ajde. Acta Agric. Slov. 2010, 95, 157–162. [Google Scholar]
  326. González-Manzano, S.; Pérez-Alonso, J.J.; Salinas-Moreno, Y.; Mateus, N.; Silva, A.M.S.; de Freitas, V.; Santos-Buelga, C. Flavanol-anthocyanin pigments in corn: NMR characterisation and presence in different purple corn varieties. J. Food Compos. Anal. 2008, 21, 521–526. [Google Scholar] [CrossRef]
  327. Yang, Z.; Zhai, W. Identification and antioxidant activity of anthocyanins extracted from the seed and cob of purple corn (Zea mays L.). Innov. Food Sci. Emerg. Technol. 2010, 11, 169–176. [Google Scholar] [CrossRef]
  328. Lago, C.; Cassani, E.; Zanzi, C.; Landoni, M.; Trovato, R.; Pilu, R. Development and study of a maize cultivar rich in anthocyanins: Coloured polenta, a new functional food. Plant Breed. 2014, 133, 210–217. [Google Scholar] [CrossRef]
  329. Tsuda, T. Anthocyanins as functional food factors—Chemistry, nutrition and health promotion. Food Sci. Technol. Res. 2012, 18, 315–324. [Google Scholar] [CrossRef] [Green Version]
  330. Lopez-Martinez, L.X.; Oliart-Ros, R.M.; Valerio-Alfaro, G.; Lee, C.H.; Parkin, K.L.; Garcia, H.S. Antioxidant activity, phenolic compounds and anthocyanins content of eighteen strains of Mexican maize. LWT Food Sci. Technol. 2009, 42, 1187–1192. [Google Scholar] [CrossRef]
  331. Cevallos-Casals, B.A.; Cisneros-Zevallos, L. Stoichiometric and Kinetic Studies of Phenolic Antioxidants from Andean Purple Corn and Red-Fleshed Sweetpotato. J. Agric. Food Chem. 2003, 51, 3313–3319. [Google Scholar] [CrossRef] [PubMed]
  332. Harakotr, B.; Suriharn, B.; Scott, M.P.; Lertrat, K. Genotypic variability in anthocyanins, total phenolics, and antioxidant activity among diverse waxy corn germplasm. Euphytica 2015, 203, 237–248. [Google Scholar] [CrossRef] [Green Version]
  333. Toufektsian, M.C.; de Lorgeril, M.; Nagy, N.; Salen, P.; Donati, M.B.; Giordano, L.; Mock, H.P.; Peterek, S.; Matros, A.; Petroni, K.; et al. Chronic Dietary Intake of Plant-Derived Anthocyanins Protects the Rat Heart against Ischemia-Reperfusion Injury. J. Nutr. 2008, 138, 747–752. [Google Scholar] [CrossRef] [Green Version]
  334. Toufektsian, M.C.; Salen, P.; Laporte, F.; Tonelli, C.; de Lorgeril, M. Dietary Flavonoids Increase Plasma Very Long-Chain (n-3) Fatty Acids in Rats. J. Nutr. 2011, 141, 37–41. [Google Scholar] [CrossRef] [Green Version]
  335. Kang, M.K.; Lim, S.S.; Lee, J.Y.; Yeo, K.M.; Kang, Y.H. Anthocyanin-rich purple corn extract inhibit diabetes-associated glomerular angiogenesis. PLoS ONE 2013, 8, e79823. [Google Scholar] [CrossRef] [Green Version]
  336. Mazewski, C.; Liang, K.; Gonzalez de Mejia, E. Inhibitory potential of anthocyanin-rich purple and red corn extracts on human colorectal cancer cell proliferation in vitro. J. Funct. Foods 2017, 34, 254–265. [Google Scholar] [CrossRef]
Table 1. The main pigments isolated in various leafy vegetables.
Table 1. The main pigments isolated in various leafy vegetables.
SpeciesColorClass of CompoundsCompounds (Content on a Fresh Weight (fw) or Dry Weight (dw) Basis)References
Lettuce (Lactuca sativa L.)RedAnthocyanins cyanidin (1558.0–3656.9 μγ/g dw), cyanidin-3-O-(6’’-malonyl-β-glucopyranoside), cyanidin-3-O-(6’’-malonyl-β-glucopyranoside methyl ester), cyanidin-3-O-β-glucopyranoside, cyanidin-3-glucoside (1.40–3.07 g/100 g fw) [42,43,46,48]
Carotenoidsall-E-violaxanthin (23.9–33.4 μg/g fw), 9′-Z-neoxanthin (11.3–14.6 μg/g fw), all-E-Luteoxanthin, all-E-lactucaxanthin (19.7–23.0 μg/g fw), all-E-lutein (31.3–38.2 μg/g fw), and all-E-β-carotene (9.0–13.3 μg/g fw)[42]
GreenCarotenoidsall-E-violaxanthin (15.9–37.1 μg/g fw), 9′-Z-neoxanthin (5.0–11.4 μg/g fw), all-E-Luteoxanthin, all-E-lactucaxanthin (7.5–17.4 μg/g fw), all-E-lutein (16.4–36.3 μg/g fw), and all-E-β-carotene (4.2–12.9 μg/g fw)[42]
ChlorophyllsChlorophyll a and b (6.95–26.92 mg/100 g fw and 4.60–10.30 mg/100 g fw, respectively)[43,48]
AnthocyaninsCyanidin-3-glucoside (0.192–0.260 g/100 g fw)[48]
Basil (Ocimum basilicum L.).Purple/redAnthocyaninsAnthocyanin A (0.325–0.423 mg/g dw),-anthocyanin B (0.057–0.641 mg/g dw), anthocyanin C (0.362–0.877 mg/g dw), anthocyanin D (0.063–0.662 mg/g dw), cyanidin-based (1.78–3.18 mg/g dw) pigments- and peonidin-based (19.8% fw) pigments, cyanidin-3-(6,6′-di-p-coumaroyl)-sophoroside-5-glcucoside (7.5 mg/g extract)[63,68,115,116]
Green CarotenoidsLutein (4.99–6.64 mg/100 g fw or 22.1–24.0 mg/100 g dw), β-carotene 4.42–6.01 mg/100 g fw, zeaxanthin (0.30–0.45 mg/100 g fw or 1.8 mg/100 g dw)[62,64]
Perilla (Perilla frutescens L. Briit)Red AnthocyaninsCyanidin and cyanidin derivatives (6.44 mg/g dw), shisonin (0.126–0.416 mg/g dw), malonylshishonin (0.462–1.116 mg/g dw)[83,117,118,119,120]
Swiss chard (Beta vulgaris var. cicla L.)YellowBetaxanthinsVulgaxanthin I, miraxanthin V[121,122]
Red/purpleBetacyaninsBetanin, isobetanin, betanidin, and isobetanidin[123]
Table 2. The main pigments isolated in various fruit vegetables.
Table 2. The main pigments isolated in various fruit vegetables.
SpeciesColorClass of CompoundsCompoundsReferences
Tomato
Solanum lycopersicum L.
Red tomatoCarotenoidsLycopene, phytoene, phytofluene, ζ-carotene, γ-carotene, β-carotene, neurosporene, lutein[127,149]
Purple tomatoCarotenoidsLycopene (36.51–61.30 mg/kg fw), β-carotene (6.5–6.8 mg/kg fw), lutein[128,136]
Purple tomatoPetunidin and malvidin acylglycosidespetunidin-3-O-caffeoyl-rutinoside-5-O-glucoside (1.88–15.36 mg/100 g dw),
petunidin-3-O-(p-coumaryl)-rutinoside-5-O-glucoside (16.97–50.18 mg/100 g dw)1,
malvidin-3-O-(p-coumaryl)-rutinoside-5-O-glucoside (6.17-27.06 mg/100 g dw),
delphinidin-3-(trans-coumaroyl)-rutinoside-5-glucoside (114.53–162.43 mg/100 g dw)
[7,136,156]
Purple/black tomatoPetunidin and malvidin acylglycosidespetunidin 3-(6-(4-(E-p-coumaroyl)rhamnosyl)glucoside)-5-glucoside (petanin) (2.77 mg/g dw),
Malvidin 3-(6-(4-(E-p-coumaroyl)rhamnosyl)glucoside)-5-glucoside (1.05 mg/g dw)
[130]
Eggplant
Solanum melongena L.
Purple eggplantdelphinidin derivativesdelphinidin-3 glucoside-5-(coumaryl) dirhamnoside (1.10 mg/g dw),
delphinidin-3-(p-coumaroylrutinoside)-5-glucoside (1357–3200 mg/kg dw)
[173,186,187,223]
Pepper
Capsicum spp. L.
Green/red peppersCarotenoidsβ-carotene, lycopene, zeaxanthin[194]
Purple/black pepperDelphinidin, cyanidin and malvidin derivativesdelphinidin 3,5-diglucoside,
delphinidin 3-O-rutinoside,
delphinidin-3-trans-coumaroylrutinoside-5-glucoside (284.8 μg/g fw),
delphinidin-3-cis coumaroylrutinoside-5-glucoside (14.72 μg/g fw)
[181,182,200]
Lablab
Lablab purpureus L.
purple lablabMalvidin and petunidin derivativesmalvidin 3-sambubiose,
malvidin 3-glucoside,
delphinidin 3-glucoside-5-rutinoside,
delphinidin 3-glucose-5-rhamnose and petunidin 3-rutinose
[213]
Common bean
Phaseolus vulgaris L.
Black beanPelargonidin,
Cyaniding and delphinidin glucosides
pelargonidin-3-glucoside,
cyanidin-3-glucoside,
delphinidin-3-glucoside
[217]
Table 3. The main pigments isolated in vegetables consumed for plant parts other than fruit and leaves.
Table 3. The main pigments isolated in vegetables consumed for plant parts other than fruit and leaves.
SpeciesEdible PartColorClass of CompoundsCompoundsReferences
Broccoli (Brassica oleracea L., var. italica Plenk)InflorescencePurpleAcylated anthocyaninscyanidin 3-O-(sinapoyl)(sinapoyl)diglucoside-5-O-glucoside (0.0653–0.3716 mg/100 g fw), cyanidin 3-O-(sinapoyl)diglucoside-5-O-glucoside (0.0119–0.0158 mg/100 g fw), cyanidin 3-O-(feruloyl)diglucoside-5-O-glucoside (0.0201–0.0765 mg/100 g fw), cyanidin 3-O-(sinapoyl)(feruloyl)diglucoside-5-O-(malonyl)glucoside (0.013–0.048 mg/100 g fw), and cyanidin 3-O-(sinapoyl)(sinapoyl)diglucoside-5-O-(malonyl)glucoside (0.0159–0.1035 mg/100 g fw)[231,233]
Cauliflower (Brassica oleracea L., var. botrytis)InflorescencePurple or dark violet Cyanidin derivativescyanidin-3-(6-p-coumaryl)-sophoroside-5-glucoside (9.0–29.9 mg/kg fw), cyanidin-3-(6-p-coumaryl)-sophoroside-5-(6-sinapyl)-glucoside (27.8–37.8 mg/kg fw)[19]
Cabbage (Brassica oleracea L. var. capitata)LeavesPurpleCyanidin derivativescyanidin-3-diglucoside-5-glucoside (0.64 mg/g dw), cyanidin-3-(sinapoyl)(sinapoyl)-diglucoside-5-glucoside (0.26-0.58 mg/g dw), cyanidin-3-(sinapoyl)-diglucoside-5-glucoside (0.85 mg/g dw)and cyanidin-3-(p-coumaroyl)-diglucoside-5-glucoside (0.19-0.92 mg/g dw),
cyanidin-3-(feruloyl)-diglucoside-5-glucoside (0.14–0.55 mg/g dw)
[249,250,252]
Artichoke [Cynara cardunculus L. var. scòlymus (L.) Fiori]Immature inflorescencePurpleCyanidin, peonidin, and delphinidin derivativescyanidin 3,5-diglucoside (0–11.7 mg/kg dw), cyanidin 3-glucoside (0.3–194.1 mg/kg dw), cyanidin 3,5-malonyldiglucoside (0.5-218.0 mg/kg dw), cyanidin 3-(3′′-malonyl)glucoside (0–47.3 mg/kg dw), and cyanidin 3-(6′′-malonyl)glucoside (7.6-1234.3 mg/kg dw) along with some minor compounds such as peonidin 3-glucoside, peonidin 3-(6′′-malonyl)glucoside, and delphinidin glycoside.[271]
Asparagus (Asparagus officinalis L.)Spears (young stems)PurpleCyanidin and peonidin derivativesCyanidin 3-[3’’-(O-β-d-glucopyranosyl)-6’’-(O-α-l-rhamnopyranosyl)-O-β-d-glucopyranoside], cyanidin 3-rutinoside (774.2 mg/kg fw), cyanidin glucosyl rutinoside (125.5 mg/kg fw), peonidin rutinoside (84.8 mg/kg fw)[23,298,302]
Sweet corn (Zea mays saccharata Sturt.)Maize kernelsYellow – deep orangeCarotenoidsβ-carotene (2.62 μg/g dw),
β-cryptoxanthin (3.96 μg/g dw), zeaxanthin (23.74 μg/g fw),
zeinoxanthin (0.75 μg/g dw),
lutein (8.2 μg/g dw),
antheraxanthin (2.5 μg/g dw))
[311,324]
Maize kernel pericarp and cobsOrange – red brikPhlobaphenesPhlobaphenes (320.24 A510/100 g)[315]
Maize cobs and kernel aleurone and pericarp Pink, red, purple, blueAnthocyanins (cyanidin, penodin, pelargonidin, cyanidin-3-glucoside,
pelargonidin-3- glucoside,
penodin-3-glucoside, cyanidin-3-(6-malonylglucoside), pelargonidin-3-(6-malonylglucoside),
peonidin-3-(6-malonylglucoside),
peonidin-3-glucoside, peonidin-3-malonylglucoside, pelargonidin-3-dimalonylglucoside
[326,327]

Share and Cite

MDPI and ACS Style

Di Gioia, F.; Tzortzakis, N.; Rouphael, Y.; Kyriacou, M.C.; Sampaio, S.L.; C.F.R. Ferreira, I.; Petropoulos, S.A. Grown to Be Blue—Antioxidant Properties and Health Effects of Colored Vegetables. Part II: Leafy, Fruit, and Other Vegetables. Antioxidants 2020, 9, 97. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9020097

AMA Style

Di Gioia F, Tzortzakis N, Rouphael Y, Kyriacou MC, Sampaio SL, C.F.R. Ferreira I, Petropoulos SA. Grown to Be Blue—Antioxidant Properties and Health Effects of Colored Vegetables. Part II: Leafy, Fruit, and Other Vegetables. Antioxidants. 2020; 9(2):97. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9020097

Chicago/Turabian Style

Di Gioia, Francesco, Nikolaos Tzortzakis, Youssef Rouphael, Marios C. Kyriacou, Shirley L. Sampaio, Isabel C.F.R. Ferreira, and Spyridon A. Petropoulos. 2020. "Grown to Be Blue—Antioxidant Properties and Health Effects of Colored Vegetables. Part II: Leafy, Fruit, and Other Vegetables" Antioxidants 9, no. 2: 97. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox9020097

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop