Next Article in Journal
Evaluation of the Effect of Light Color on Albumins and Globulins Content During Bean Germination
Previous Article in Journal
Power Ultrasound- and Organic Acid-Based Hurdle Technology to Reduce Listeria monocytogenes and Salmonella enterica on Whole Apples and Peaches
Previous Article in Special Issue
Bioaccessibility and Functional Food Potential of Equisetum telmateia Ehrh. Against Diabetes-Induced Kidney Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic and Preventive Potential of Plant-Derived Antioxidant Nutraceuticals

1
Universidade de Vigo, Nutrition and Food Group (NuFoG), Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)—CITEXVI, 36310 Vigo, Spain
2
Investigaciones Agroalimentarias Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
3
Centro de Investigação de Montanha (CIMO), LA SusTEC, Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
4
REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
*
Authors to whom correspondence should be addressed.
Foods 2025, 14(10), 1749; https://doi.org/10.3390/foods14101749
Submission received: 12 April 2025 / Revised: 9 May 2025 / Accepted: 10 May 2025 / Published: 14 May 2025

Abstract

:
Oxidative stress and its relation to the onset of several chronic diseases have been increasingly highlighted in recent years. In parallel, there has been an increasing interest in the antioxidant properties of phytochemicals. Phytochemicals are products of plant secondary metabolism, including structural polysaccharides, unsaturated fatty acids, pigments (chlorophylls, carotenoids, and anthocyanins), or phenolic compounds. Phytochemicals can be obtained from lower and higher plants, their fruits, and even from macro- or microalgae. Their diverse structural features are linked to different beneficial effects through various molecular mechanisms, contributing to disease prevention. Beyond antioxidant activity, many phytochemicals also display anti-inflammatory, antidiabetic, anti-obesity, and neuroprotective effects, which can be intertwined. Beyond these, other natural antioxidants can also be obtained from animal, fungal, and bacterial sources. Thus, a wide range of antioxidants have the potential to be used as nutraceuticals with chemopreventive effects on the onset of various diseases related to antioxidant stress. Given their enormous structural and sourcing diversity, the present work provides an updated insight into the therapeutic and preventive potential of plant-derived antioxidants and nutraceuticals.

1. Introduction

Phytochemicals are secondary metabolites synthesized by plants, encompassing a wide variety of chemical groups. These include phenolic compounds (PCs), fatty acids (FAs), pigments, polysaccharides (PSs), terpenoids, alkaloids, phytosterols, and saponins, among others. Their broad functional potential has positioned them as essential components in various industrial and health-related processes, including medicine, agriculture, food, textile, and cosmetic industries [1]. In particular, dietary phytochemicals have garnered increasing scientific attention in recent decades, largely due to their associated health benefits, such as cardioprotective, neuroprotective, anti-inflammatory, and gut-health-promoting effects, among others [2]. Yet, the major capacity of phytochemicals is to act as ameliorators of oxidative stress. Oxidative stress is the result of excessive levels of reactive oxygen species (ROS), which are produced by normal oxidative metabolism and exposure to the environment. The term ROS refers to molecules, such as superoxide radical (O22−), hydroxyl radicals (OH), and hydrogen peroxide (H2O2), among others. These are produced by the sequential reduction of molecular oxygen in metabolic reactions. Oxidative stress may ultimately cause damage to biomolecules and can deregulate and even disrupt physiological redox signaling, causing changes in the rate of metabolic reduction reactions, an increased rate of DNA mutations, and deregulation of cell proliferation [3]. They are also one of the major markers of cell death, which, in turn, triggers inflammation and intertwines both processes. The impact that oxidative stress has on altering normal physiological performance is known to be a factor leading to the development of inflammatory, cardiovascular, neurodegenerative, and metabolic disorders, which, in turn, may result in cancer development [4,5]. Thus, despite the presence of endogenous antioxidant systems, the intake and use of phytochemicals as significant antioxidants has been proven as a suitable alternative to improve oxidative stress status [2].
Key contributors to this activity include PCs, unsaturated fatty acids (UFAs), carotenoids, and polysaccharides, which are commonly isolated from plant-based matrices [6]. The growing recognition of these bioactivities has not only influenced dietary recommendations and favored increased consumption of fruits and vegetables, but it has also catalyzed the expansion of the nutraceutical sector. This industry now focuses on formulating products enriched with specific phytochemicals, such as purified phenolics, aimed at delivering targeted health benefits [7]. However, in recent decades, the interest in dietary phytochemicals has risen because they are thought to provide many health benefits, such as cardioprotection, neuroprotection, anti-inflammatory effects, and improved gut health, among others [2]. These effects are mainly attributed to the antioxidant properties of various groups of phytochemicals, with phenolic compounds, UFAs, and polysaccharides being chief among the key contributors [6]. Thus, diets rich in vegetables and fruits that may act as sources of phytochemicals have attracted more interest from researchers, industry, and society, with a focus on more conscious and healthy diets, as extensive research in the last decades has suggested that they are an overall healthy alternative [8,9].
To support the formulation of effective phytochemical-enriched products, it is essential to optimize extraction methodologies that maximize recovery while preserving compound integrity. Among the wide array of phytochemicals, UFAs are a particularly valuable group because of their well-documented health benefits. However, despite advances in extraction technologies, most industrial-scale studies and applications continue to rely on conventional techniques, such as solvent extraction or mechanical pressing. These approaches, while widely implemented, often present limitations in terms of selectivity, efficiency, and environmental impact [10]. UFAs, for example, can be feasibly obtained from oily crops, such as olive, sunflower, rapeseed, sesame, palm, and peanut [11], although their composition and concentration vary depending on the plant source and extraction technique. Beyond these conventional crops, several underutilized fruit seeds, including those of pepper, grape, and avocado, have also been identified as promising UFA sources [12,13]. Additionally, microalgae have gained increasing attention in recent years as sustainable and efficient producers of UFAs with high nutritional value [14].
In addition to UFAs, pigments represent another important class of phytochemicals with significant health-promoting properties and broad industrial applicability, primarily due to their antioxidant activity and their function as natural colorants [15]. Among these, carotenoids stand out as the principal group of pigments used in industrial sectors. Carotenoids can be distinguished between carotenes, which include carotene and lycopene, and xanthophylls, with some main examples being lutein, zeaxanthin, and lycopene [15]. Moreover, they can be further distinguished between provitamin A carotenoids, which include carotenes and β-cryptoxanthin, and non-provitamin A carotenoids, including mainly xanthophylls, like lycopene, lutein, and zeaxanthin [16]. Intuitively, carotenoids can be obtained from yellow, orange, and red-colored fruits and vegetables, with higher concentrations in their peels. In recent years, both seaweeds and microalgae have become feasible and cost-effective sources of carotenoids, with some major examples being fucoxanthin from brown seaweeds and astaxanthin from red microalgae [17]. In this sense, carotenoids have been highlighted as relevant phytochemicals with various potential health benefits, ranging from their nutritional value as precursors of vitamin A to health-related properties, such as antioxidant, metabolic modulation, anti-inflammatory, and neuroprotective [18].
Another important class of phytochemicals is PSs, which play a critical role in both plant biology and human health. Among them, cellulose, hemicellulose, and pectin are the major PSs present in plants, which constitute dietary fiber [19]. These structural PSs may be found more abundantly in the “harder” parts of plants, such as seeds, leaves, and peels, and are of high interest as promoters of healthy gut microbiota [20]. Conversely, storage polysaccharides (that act as stored energy), which include starches and other PSs (i.e., fructans), are more abundant in bulbs, tubers, and pulps [21].
Lastly, PCs are antioxidant phytochemicals of paramount importance that have been exhaustively studied in the last two decades and have a ubiquitous distribution in the plant kingdom. PCs are mainly synthesized in plants in response to biotic (predators, infections) or abiotic (drought, salinity) factors [3]. Their diverse structural complexity and degree of polymerization also determine their bioavailability, activities, and occurrence. Some well-known examples of PCs and sources include flavanols in grapes or tea, flavonols in citrus, and anthocyanins in berries [3,22].
Given their vast variety, sources, and beneficial effects related to health, natural antioxidants have been increasingly used as nutraceuticals. Although these diverse molecules have been generally observed to display null or usually lower toxicity than synthetic or conventional chemicals, they may also display some undesired effects. For instance, it is known that PCs establish synergies in their antioxidant effects when various diverse molecules are present; however, when a single compound is present at high concentrations, PCs may act as prooxidant molecules, a fact that has also been observed with other antioxidant molecules [2]. Moreover, as with every other nutrient and chemical, an excessive intake can lead to adverse effects on health. For example, excessive intake of β-carotene has been associated with an increase in serum triglycerides, cholesterol, and bone loss [23]. On the other hand, ROS and other reactive species that are produced as a result of physical activity are needed for endogenous responses that enable muscle growth, which antioxidant supplementation may partially hurdle [24]. Nonetheless, beyond potential hepatotoxicity if the daily consumption is consistently very high, negative side effects of these types of natural compounds are limited, and mainly attributed to other elements, especially metals [25,26].
As research continues to uncover the full extent of their health benefits, these compounds are increasingly recognized for their potential to act as antioxidants, support disease prevention, enhance immune function, and improve overall well-being. As interest in these compounds continues to grow, there is a shift towards more economically viable production methods, including chemical and microbial synthesis, which offer greater consistency and scalability [27]. Additionally, the extraction of phytochemicals from natural sources, particularly through the use of agri-food waste from fruits and vegetables, is emerging as a sustainable and cost-effective approach [28]. Although these methods may be cost-effective, chemical extraction of phytochemicals from their natural sources is becoming increasingly widespread, especially using fruit and vegetable agri-food waste as a source of phytochemicals [28]. The aim of the present work is to discuss the role of antioxidants in plants, their significance in the mitigation of oxidative damage, and their implications for plant resilience and human overall health through other beneficial effects on health, such as anti-inflammatory, antidiabetic, anti-obesity, and neuroprotection. Although this work focuses on UFAs, PCs, PSs, and carotenoids, various other natural compounds with antioxidant properties, as well as significant benefits to health and potential use as nutraceuticals, are also explored.

2. Chemical Classes of Antioxidant Phytochemicals: Structural Significance

2.1. Unsaturated Fatty Acids

FAs are hydrophobic aliphatic carbon chains of variable length that contain a carboxyl (-COOH) group at the end of the chain [29]. FAs are classified by the number of carbons in the chain, but they mainly depend on the presence of double bonds between the carbons (Figure 1). FAs with no double bonds present are called saturated FAs (SMFAs), while those with double bonds are called UFAs, which are further classified based on whether they have a single bond (monounsaturated FAs, MFAs) or two or more bonds (polyunsaturated FAs, PUFAs) [30]. In addition, fatty acids can also be distinguished depending on their stereochemistry, presenting cis or trans isomers, with cis being more prevalent in plants and trans more prevalent in animal fats [31]. Depending on the position of the double bonds in the chain, UFAs can be further distinguished as omega (ω). In the case of plant organisms, fatty acids may usually present a length in the range of 12 to 22 carbons, with 16 and 18 carbons being generally the most abundant [32]. Among MFAs, plant organisms generally accumulate palmitic (C16:0) and stearic (C18:0) acids. Moreover, plants are the main source of UFAs in nature and, thus, in the human diet, with some major representatives being oleic acid (C18:1) and linoleic acid (C18:2) [11].

2.2. Phenolic Compounds

As mentioned, PCs are produced by plants primarily in response to environmental stress factors, such as drought, extreme temperatures, UV exposure, pollution, and pathogen attacks [33], and their biosynthesis follows pathways like the pentose phosphate, shikimate, and phenylpropanoid routes [34]. Chemically, PCs are defined by the presence of one or more hydroxyl groups attached to an aromatic ring, though their complexity varies across different subclasses. These compounds encompass a broad range of molecules, including phenolic acids, flavonoids, stilbenes, lignans, and related structures, like chalcones, humulones, and alcohols [33,35]. These compounds are classified according to their structure, with more than 8000 variations currently identified. The classification is typically based on the number of phenolic rings and the nature of the linkages between them. The major groups include phenolic acids, flavonoids, stilbenes, and lignans [36].
Phenolic acids are generally the most ubiquitous and the smallest PCs in terms of molecular size and weight, and they are divided into two main categories: hydroxybenzoic acids (e.g., gallic acid) and hydroxycinnamic acids (e.g., caffeic, ferulic, coumaric acids) [36,37]. Flavonoids, another key category, have a C6–C3–C6 backbone and are further categorized into subgroups (Figure 1), such as flavonols (e.g., quercetin, kaempferol, morin, myricetin), flavones (e.g., luteolin, acacetin, chrysin, baicalein, wogonin), isoflavones (e.g., genistein), anthocyanins, flavanones (e.g., naringenin, hesperetin), flavanols, and chalcones (e.g., xanthohumol isoxanthohumol, isobavachalcone, licochalcone A) [37]. In plant cells, flavonoids are typically stored as glycosides, and their antioxidant potential is influenced by the degree of glycosylation. Aglycones, like quercetin and myricetin, are generally more potent antioxidants than their glycoside forms, but with hampered bioavailability [38].
Tannins, another class of phenolics, are mainly categorized into hydrolysable tannins (gallotannins, ellagitannins) and condensed tannins and phlorotannins (based on phloroglucinol, which is only found in brown seaweeds). These compounds are known for their protein-binding capabilities, contributing to the astringent taste of certain fruits and their antimicrobial activities [36,39].
Additionally, some PCs, like curcumin (diferuloylmethane), display a broad spectrum of biological functions, including antioxidant, anti-inflammatory, antimicrobial, antitumor, and liver-protective effects [40]. The structure of PCs is closely linked to their bioactivities, with different chemical arrangements providing specific biological functions [41,42].
Given their broad spectrum of bioactivities, PCs are increasingly being incorporated into nutraceuticals and functional foods aimed at improving human health and preventing chronic diseases [43]. Furthermore, these compounds contribute to the sensory qualities of many plant-based foods and beverages, particularly in terms of flavor and color [44]. In the context of human nutrition, PCs are known for their strong antioxidant properties and represent a significant portion of the antioxidant compounds in plants [45]. In terms of dietary applications, phenolic acids are commonly found in functional foods because of their beneficial health effects, such as reducing inflammation, preventing allergic reactions, and offering cardiovascular protection [46]. As research continues to elucidate the specific mechanisms underlying their health benefits, phenolic compounds hold promise as key ingredients in the development of novel therapeutic agents and functional food products [46,47]. Additionally, phenolic compounds have extensive applications in the food, cosmetic, and pharmaceutical industries, where they are used for their antioxidant and preservative properties, as well as their ability to improve the stability and shelf life of products [2]. They also represent a significant portion of the antioxidant content in plant-based foods [45].

2.3. Carotenoids

Given their nature and metabolic function as reducing agents, plant pigments are used as colorants and antioxidants. Carotenoids represent a class of pigments naturally produced by plants, fungi, algae, and specific bacteria [48]. These compounds are responsible for the vibrant red, yellow, and orange colors of various fruits and vegetables [49]. Structurally, carotenoids are classified as tetraterpenoids, composed of a 40-carbon polyene chain that provides their antioxidant properties (Figure 2) [50]. The conjugated double bond system present in the structure of carotenoids facilitates electron delocalization, thereby increasing their ability to neutralize free radicals and singlet oxygen [51]. Additionally, their antioxidant efficiency is modulated by the degree of unsaturation and the presence of functional groups, like hydroxyl (-OH) or keto oxo (C=O) groups [52].
Based on their structure, carotenoids can be divided into two primary categories: carotenes, which are purely hydrocarbons, and xanthophylls, which contain oxygenated groups [53]. The functional groups in xanthophylls generally improve their solubility and interaction with cellular membranes, potentially influencing their antioxidant effectiveness [54]. In fact, the presence of additional oxygen-containing functional groups often makes xanthophylls more potent antioxidants compared to carotenes [55].
Plants, algae, and fungi can produce different isomers of carotenes, with α-carotene and β-carotene being the most prevalent in plants (Table 1). Other isomeric forms include γ-carotene, δ-carotene, ε-carotene, and ζ-carotene. Both α-carotene and β-carotene serve as precursors to vitamin A [56], although β-carotene exhibits greater antioxidant activity compared to α-carotene [57]. Among xanthophylls, the key molecules include fucoxanthin, astaxanthin, lutein, zeaxanthin, and β-cryptoxanthin [53]. These compounds are recognized for their strong antioxidant properties, which play a role in their anticancer, anti-inflammatory, and neuroprotective effects, as well as their ability to enhance immune responses [53,58]. Owing to these attributes, carotenoids hold considerable potential for therapeutic use in managing OS-related diseases [59]. The variation in their efficacy stems from differences in their structural characteristics, such as terminal functional groups and chain length [53]. For instance, lutein and zeaxanthin, commonly found in leafy greens, are known to accumulate in the retina’s macular region, where they filter harmful blue light and serve as powerful antioxidants [58].
Additionally, several studies support the inclusion of carotenoids in functional foods and dietary supplements, linking their intake to a reduced risk of chronic diseases, including cardiovascular disorders, specific cancers, and age-related macular degeneration [49]. Their role as precursors to vitamin A further enhances their significance in promoting visual health and supporting the immune system [60]. The numerous benefits of these compounds have favored the inclusion of these molecules as ingredients in the development of nutraceuticals aimed at improving health and preventing diseases.

2.4. Polysaccharides

PSs are complex polymers composed of monosaccharide units connected by glycosidic bonds, which are widely distributed in plants, fungi, and algae [61]. These natural polymers have attracted significant attention due to their potential as nutraceuticals, particularly for their antioxidant, immunomodulatory, and anti-inflammatory activities [62,63]. Additionally, PSs prevent ROS-induced tissue damage through both free radical scavenging (Table 1) and modulation of the immune response [64,65]. However, under specific conditions, certain PSs may also exhibit prooxidant behavior, adding complexity to their antioxidant profile [66,67].
Overall, the antioxidant capacity of PSs is multifactorial, owing not only to their size but also to their structure and functional groups [68]. Structural features, such as monosaccharide composition, glycosidic linkages, molecular weight, and branching degree, contribute to their antioxidant activity [69]. Moreover, PSs with higher molecular weights, increased branching, and specific functional groups, such as sulfate or carboxyl groups, often demonstrate stronger antioxidant potential [70,71]. Moreover, the method of extraction plays a crucial role in determining their functional properties [72,73]. However, despite the established antioxidant potential of PSs, there is limited research on highly purified forms; thus, the possible influence of other bioactive components, which may be present in combination with PSs, must be taken into account [68].
PSs are classified based on their structure, origin, and function. Structurally, they can be divided into hetero-PSs (e.g., hyaluronic acid, chondroitin sulfate, heparin) and homo-PSs (e.g., fructans, galactan, glycogen) [74]. Based on their source, they are categorized into several groups: plant-based (e.g., pectin, cellulose, starch), microbial-based (e.g., curdlan, dextran, bacterial cellulose), fungi-derived (e.g., β-glucans, chitin derivatives), animal-derived (e.g., chitosan, chitin, heparin), and marine-derived (e.g., agar, alginate, fucoidans) [75]. PSs like pectin, which is rich in galacturonic acid, exhibit significant antioxidant properties. This bioactivity is influenced by their uronic acid content and degree of polymerization [76]. Moreover, sulfated PSs from marine algae are particularly effective in neutralizing free radicals and chelating metal ions, enhancing their antioxidant performance [77]. Ongoing research continues to shed light on the mechanisms by which these PSs exert their health-promoting effects, opening new avenues for their use in preventive and therapeutic nutrition [78]. Moreover, beyond their role as prebiotics, PSs are also widely utilized in industry for various functions, including as thickeners, emulsifiers, stabilizers, gelling agents, and controlled-release agents, broadening their applications [74,78].
Table 1. Concentration and antioxidant activity of antioxidant phytochemicals in principal sources: data from chemical assays.
Table 1. Concentration and antioxidant activity of antioxidant phytochemicals in principal sources: data from chemical assays.
ClassCompoundMain SourceConcentrationAssayAARef.
UFAs *
ω-3ALAOlive, sunflower, linseed, rapeseed, fruit and vegetable seeds, other oily crops5.5–61.5%ROS16.86 mM[79,80,81,82]
EPASeaweed, microalgae, fish oil6.6–22.5%ROS150 µM[83,84]
DHASeaweed, microalgae, fish oil1–6.6%ROS100 µM[83,85,86]
ω-6LAOlive, sunflower, linseed, rapeseed, nuts, fruit and vegetable seeds, other oily crops16.5–62.5%ROS39.5 mM[79,82,87,88,89]
ω-7PAOlive, nuts, macadamia nuts, microalgae0.6–50.1[90,91,92,93,94]
ω-9OAMicroalgae, linseed, rapeseed, nuts, fruit and vegetable seeds, other oily crops1.4–79.6%SOD53.1 mM[82,83,91,95,96,97]
Carotenoids
Carotenesα-caroteneCarrots, pumpkins13.44–30.11 mg/kg fwROS40.6 µmol TE/g dw[98,99]
β-caroteneCarrots, red peppers, oranges, potatoes, green vegetables41.60–71.2 mg/kg fwROS7.2 µmol TE/g dw[98,99]
XanthophyllsFucoxanthinBrown algae0.02–18.60 mg/g dwROS201 μg/mL[53,100]
AstaxanthinHaematococcus pluvialis3.8%ROS1.33 mM[101]
LuteinMicroalgae, algae, vegetables (i.e., kale, spinach)0.7–5% dwROS1.8–22 μg/mL[102]
ZeaxanthinRed and brown seaweed, red/orange vegetables/fruits0.49–1230 µg/g dwROS2.2 μg/mL[53,103]
β-cryptoxanthinAlgae, red/orange vegetables/fruits409–1103 µg/g dwROS38.30 μg/mL[104,105]
Polysaccharides
HEHyaluronic acidStreptococcus spp., Tremella fuciformis1300 µg/mLROS69.2–78.4%[106,107]
Chondroitin sulfateBacteria and cartilage-MCC3.33 mg/mL[108,109]
HeparinMarine organism, Asteraceae plants-EA2.20 mg/mL[110,111]
HOFructanProkaryotes, lower and higher plants0.9–1.8 g/100 g in different wheat cultivarsEA, MCC0.12 mg/mL[21,112]
GalactanSeaweeds, seeds of some plants-SOD, GSH-Px9 μM[113]
PlantPectinCell walls of terrestrial plantsCitrus peels 30% fw, oranges 0.5–3.5% fw, carrots 1.4% fwROS161.94 ppm[114,115]
CelluloseCell walls of terrestrial plants40–50% fwROS80.9 ppm[116,117]
StarchCereals, pseudocereals, legumes, root tubers60–75% fwROS97 µg/mL[118,119]
MicrobialCurdlanAgrobacterium sp., Rhizobium sp.34.04 mg/gROS82% DPPH, 72% ABTS[120,121]
DextranLactic acid bacteria580 mg/100 mL dwROS97 μg/mL[122,123]
CelluloseAcetobacter spp., Sarcina spp., Agrobacterium spp.60.7% dwROS80.9 ppm[116,124]
Fungiβ-glucansFungal cell walls31% dwROS, EA161–4019 μg/mL[125,126]
ChitosanCell wall of filamentous fungi20–45% dwROS0.022 mg/mL[127,128]
MarineFucoidanBrown seaweed20% dwROS0.058 mg/mL[129,130]
AlginateBrown seaweed20–60% dwROS121.4–346.3 mol/g[131,132]
CelluloseGreen algae1.5–34%dwROS0.15–0.39 mg/mL[124,133,134]
Phenolic compounds
Total PCs-Phoenix dactylifera var Bunarinja34.90 mg/100g fwROS0.875 μg/mL[135]
Phenolic acidsCaffeic acidGreen coffee6.56 μg/mLROS6.31 μg/mL[136]
Chicoric acidEchinacea purpurea56.03 mg/g dwROS15 μg/mL[137]
Ferulic acidRice bran8.71 mg/gROS9.9 μg/mL[138,139]
FlavonoidsMyricetinGreen tea0.40–0.79 mg/gROS4.68 µg/mL[140]
ApigeninGentiana veitchiorum37.50 mg/LROS8.26 mg/mL[141]
Total tannin-Ginger35.08 mg/gROS1 mg/mL[142]
-Garlic7.44 mg/gROS3.7 mg/mL[142]
-Myristica fragrans14.03% w/wROS89.98 μg/mL[143]
Abbreviations: UFAs: unsaturated fatty acids; EFAs: Essential Fatty Acids; OA: oleic acid; DHA: docosahexaenoic acid; EPA: Eicosapentaenoic Acid; HEs: Heteropolysaccharides; HOs: Homopolysaccharides; PCs: phenolic compounds; dw: Dry Weight; fw: Fresh Weight; MCC: metal cation chelation; EAs: enzymatic antioxidants; ROS: reactive oxygen species; AA: antioxidant activity (IC₅₀); ns: Not Specified. * UFA concentrations are presented as % of total fatty acids.

3. Chemopreventive and Therapeutic Properties of Antioxidant Phytochemicals

Diets rich in plant-based foods are generally associated with reduced cancer risk and all-cause mortality due to their related health benefits, including antioxidant, anti-inflammatory, antidiabetic, anti-obesity, and, potentially, anticancer effects (Figure 3). This suggests that dietary phytochemicals from various plant species hold potential for cancer chemoprevention [144]. This section will explore in vivo and in vitro studies on the biological properties of phytochemicals and other antioxidants from common and uncommon sources, highlighting their effects and mechanisms described (Table 2) [145].

3.1. Antioxidant and Anticancer Properties

Antioxidants and phytochemicals act through two main mechanisms: chain breaking by donating electrons to free radicals and quenching ROS and other free radicals. As mentioned, there is a preeminent intertwining between oxidative stress and various diseases, including being considered a major contributor to the onset of cancer [186]. As such, various antioxidants from common and uncommon sources have been studied, considering both properties, by direct and indirect pathways (Table 2). The authors of a recent study examined the antioxidant properties of allicin, a major and fragrant sulfur organic compound from garlic, in in vitro and in vivo models of cholangiocarcinoma (CCA) [146]. Allicin was found to reduce oxidative stress and inhibit the STAT3 signaling pathway, which promotes tumor growth. The study demonstrated that allicin suppressed CCA cell proliferation, migration, and invasion while inducing apoptosis. Its dual action as an antioxidant and inhibitor of tumor-promoting signals suggests allicin could be a promising therapeutic option for CCA [146].
An in vivo study evaluated the phytochemicals in green tea (Camellia sinensis), including catechins and curcuma (Curcuma longa) curcumin, for chemopreventive effects in a hamster buccal pouch carcinoma model [144]. Green tea PCs, which mainly comprise flavan-3-ols and, mainly, epicatechin gallate, inhibited angiogenesis, while curcumin induced apoptosis through cytochrome c release and caspase activation. Both phytochemicals reduced cell proliferative and angiogenic activity, and when combined, they were more effective at inducing apoptosis and cell cycle arrest [144]. The PC-rich aqueous extract of virgin’s rose (Fagonia cretica) was tested for its effects on breast cancer cell lines. The results proved that the extract induced cell cycle arrest and apoptosis through both p53-dependent and -independent mechanisms. Specifically, it led to a significant increase in tumor protein p53 and FOXO3a expression [151]. On the other hand, plant-derived quercetin exhibited chemopreventive effects in HepG2 (human liver cancer) cells by reducing ROS, downregulating PI3K, PKC, and COX-2, and increasing pro-apoptotic p53 and BAX expression, highlighting its antioxidant and anticarcinogenic action [157]. One in silico and in vitro study attributed Nile tulip’s (Markhamia lutea) strong antioxidant properties primarily to its rich PC content, mainly including flavonoids and phenolic acids, which effectively scavenged free radicals and chelated metal ions [155].
A study on tobacco weed (Elephantopus mollis) highlighted 3,4-di-O-caffeoyl quinic acid for its significant antioxidant activity and chemopreventive properties, including apoptosis induction in NCI-H23 (human lung adenocarcinoma) cells and β-glucosidase inhibition [153]. Additionally, the leaf extract of Onobrychis argyrea has demonstrated significant antioxidant, antidiabetic, anti-Alzheimer’s disease, and anticancer in vitro properties. It induced apoptosis in human colorectal adenocarcinoma HT-29 cells by disrupting mitochondrial membranes and activating caspases, and it showed high antioxidant activity, enzyme inhibitory activities, and strong anticancer capacity [152].

3.2. Anti-Inflammatory Properties

Anti-inflammatory phytochemicals may help prevent cancer by targeting inflammation-related mechanisms. They induce cell cycle arrest by inhibiting cell cycle regulators, leading to halted cancer cell proliferation [187]. Moreover, they promote apoptosis by upregulating pro-apoptotic proteins and caspases while downregulating anti-apoptotic factors, thus facilitating cancer cell death [188]. They also promote autophagy by upregulating autophagy-related proteins and inhibit mTOR signaling, leading to the elimination of damaged cells. These combined effects decrease inflammation and suppress tumor progression [189].
It has been reported that mango (Mangifera indica) polyphenols (mainly gallic acid, hydroxybenzoic acid hexoside, and hydrolysable tannins, such as monogalloyl-glucoside) reduced NF-κB expression and phosphorylation in tumor necrosis factor-alpha TNF-α-treated MCF-12A cells, indicating anti-inflammatory effects. Phytochemicals identified in mango modulated the phosphoinositide 3-kinase/protein kinase B/mechanistic target of the rapamycin (PI3K/AKT/mTOR) signaling pathway [159]. Commiphora leptophloeos extract, rich in phenolic acids and flavonoids, showed anti-inflammatory effects by downregulating NF-κB/COX-2 and reducing pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). This suggests its potential in the treatment of inflammatory bowel disease (IBD) [162]. Studies on the anti-inflammatory activity of Helicteres isora fruit extracts have been focused on its effects on pro-inflammatory mediators such as prostaglandin E2, COX-2, and TNF-α. Compared to celecoxib, a known COX-2 inhibitor, a hexane extract showed the strongest inhibition of PGE-2 (69.68%), followed by an ethanol extract (57.17%). All extracts were less effective than dexamethasone, a standard anti-inflammatory drug, but the hexane extract showed 51.61% inhibition of TNF-α [160]. The extract of Euphorbia hirta has been shown to have anti-inflammatory activity in vitro, which is relevant to its potential chemoprotective effects. By reducing nitric oxide (NO) production through the inhibition of inducible NO synthase (iNOS), E. hirta can help to lower inflammation, a known contributor to cancer advancement. Chronic inflammation can facilitate tumor growth and spread, so the extract’s ability to suppress NO and inflammatory cytokines may play a role in its anticancer properties [167]. Chamomile phytochemicals, including quercetin and the terpenoids β-amyrin and lupeol, act as NF-κB inhibitors and induce G2/M cell cycle arrest. These compounds lower pro-inflammatory cytokines IL-1β and IL-6 and interact with microtubules, which supports their potential as anticancer modulators of inflammatory pathways [163].

3.3. Antidiabetic Properties

Diabetes mellitus (DM) is a metabolic syndrome that increases the levels of glucose in the blood [173,190]. The main chronic complications of this disease are linked to the level of exposure of patients with hyperglycemia [190]. Despite the availability of chemical drugs that act as therapeutic agents, there is a need to find natural alternatives to control the complications derived from the disease and improve glycemic control and glucose intolerance [173]. In this sense, traditional medicine revealed several molecules present in plants with antidiabetic activity, as well as their mechanisms of action [190].
For instance, leaf extracts have been widely studied to determine their antidiabetic potential. An in vivo and in vitro study investigated the volatile and phenolic compounds present in basil (Ocimun basilicum) essential oil to determine its potential antidiabetic activity. Streptozotocin (50 mg/kg of body weight) was used as a model to induce DM in Wistar Albino rats [191]. After 28 days of feeding basil essential oil (70 mg/kg of body weight) rich in PCs, the rats showed a protective effect in pancreatic β-cells and a decrease in blood glucose levels. Regarding in vitro assays, the inhibitory effects on the digestive enzymes α-amylase and α-glucosidase inhibitory activity were studied, showing a strong inhibition potential. The authors linked the antidiabetic effect of basil essential oil to the phenolic and flavonoid compound content. However, the mechanism of action was not elucidated [191]. In another in vivo study, Derris elliptica methanolic leaf extract’s phytochemical profile and antidiabetic potential were determined [173]. Streptozotocin was used to induce DM in Sprague Dawley rats. After 14 days of feeding with the plant’s extract at two different concentrations (200 and 400 mg/kg body weight), the results showed a hypoglycemic effect and reduced cholesterol levels in a dose-dependent manner. Moreover, the highest dose increased serum insulin levels, and an antihyperglycemic effect was shown. Although the authors did not explain the mechanism of action, quercetin was the compound linked to these antidiabetic properties [173]. Eugenia sonderiana leaf extracts were also studied to determine their antidiabetic potential and to develop a structure–activity correlation [190]. Both in vivo and in vitro studies were assessed, which showed great α-amylase and α-glucosidase inhibitory activity and reduced glucose, pancreatic enzymes, and triglycerides, while HDL cholesterol levels were maintained within normal ranges. To explain the antidiabetic ability of E. sonderiana leaf extracts, the authors suggested a synergistic effect between PCs by increasing glycogen synthesis, α-amylase, and saponins by a complex formation with cholesterol [190].

3.4. Anti-Obesity Properties

Obesity contributes to the increase in the prevalence of chronic diseases, including type II DM, hypertension, dyslipidemia, cardiovascular diseases, and some types of cancer [177,192]. Common strategies to decrease the incidence of obesity are physical activity, surgery, drugs, and diet modifications [192]. In this regard, the natural compounds present in plants have been suggested as potential chemicals to be used for both the prevention and treatment of obesity [192]. Several studies have assessed the anti-obesity potential of phytochemicals. For instance, green tea has been traditionally used as a medical plant, and scientific studies have revealed its potential for obesity prevention, linked to its high content of PCs. In vitro results regarding green tea leaves suggest the anti-obesity potential of this plant by inhibiting adipocyte differentiation and proliferation [193].
In a study, C57BL/6J mice fed a high-fat diet supplemented with 0.25% Provence rose (Rosa centifolia) polyphenol extract for 35 days showed decreased body weight and adipose tissue, as well as reduced serum cholesterol and hepatic triglyceride levels. The treatment increased fecal triglycerides and increased lipolysis-related proteins while suppressing lipid synthesis enzymes. Ellagic acid, a key component of the Providence rose, is likely responsible for these anti-obesity effects, emphasizing its capability for treating obesity [177].
Mustard (Brassica juncea) attenuated lipid accumulation in 3T3-L1 adipocytes and lowered epididymal white adipose tissue mass in obese mice fed a high-fat diet [179]. Finally, acetone extract fractions from Rumex rothschildianus, which is rich in flavonoids and phenolics, strongly inhibited lipase activity, comparable to orlistat, and also affected α-amylase and α-glucosidase [182].

3.5. Neuroprotective Properties

In the past decades, there has been a rise in the incidence of neurodegenerative disorders, which has been linked to the increase in longevity that has occurred. Neurodegenerative diseases encompass a wide range of conditions that affect the central nervous system, including alterations in the neuronal structure and cellular dysfunction that lead to progressive degeneration [194]. The most prevalent are Alzheimer’s disease, Parkinson’s disease, and lateral amyotrophic sclerosis. Although there are some drugs available to ameliorate these conditions, their efficacy is limited. In the past years, natural compounds have been suggested as potential alternatives with the ability to delay disease onset, reduce disease progression, and regenerate damage via their anti-amyloid, antioxidant, and anti-inflammatory properties [195].
Paeonia ostia has traditionally been used as a medical and ornamental plant in China. The neuroprotective effect of the stamens of this plant has been studied by the NO inhibition assay in lipopolysaccharide-induced BV-2 cells. The (+)-3′′-methoxy-oxylactiflorin present in the plant has excellent inhibitory properties in inhibiting NO production. Moreover, molecular docking results showed that the anti-inflammatory mechanism of this molecule was a binding action with COX-2 [183]. In another study, Indian gooseberry (Phyllantys emblica) fruit extract, which is rich in PCs, was tested in rats with sodium valproate-induced postnatal autism. After treatment with the fruit’s extract at 100 mg/kg of body weight, the amelioration of social interactions, social affiliation, anxiety, and motor coordination was achieved. Moreover, the results showed the restoring effect of the extract in glutathione-S-transferase and glutathione reductase, which are oxidative enzymes, and a reduction in malondialdehyde (MDA) and NO levels [184]. These activities showcase its ability to induce neuroprotection by anti-inflammatory and antioxidant effects, as well.

4. Development of Phytochemicals as Nutraceuticals

4.1. Extraction, Purification, and Encapsulation

The direct ingestion of phytochemicals within their native plant matrices presents several challenges [196]. The bioavailability of these compounds is often restricted by factors such as low concentrations, poor solubility, instability under gastrointestinal conditions, and rapid metabolic degradation and excretion [197]. These obstacles highlight the need for developing methods to concentrate, purify, and encapsulate phytochemicals, thereby enhancing their effectiveness as nutraceuticals (Figure 4) [198].
The initial phase in the utilization of phytochemicals for nutraceutical purposes involves their extraction from plant sources [199]. This extraction process is complex and must be carefully optimized to preserve the structure and bioactivity of the target compounds [200]. Traditional extraction techniques, such as solvent extraction, have been improved by emerging methods, like microwave-assisted extraction and ultrasound-assisted extraction, since they offer increased efficiency and selectivity [201]. A major challenge in phytochemical extraction and recovery is to incorporate environmental sustainability into the design and optimization of production processes, with the aim of developing more eco-friendly methodologies [202] and enhancing economic outcomes within the food industry [203]. The choice of appropriate extraction techniques is crucial, as it directly impacts the quality and characteristics of the final product [204]. The key factors influencing the efficacy of both conventional and emerging extraction techniques include the solid-to-solvent ratio, solvent concentration, particle size, and the mode of extraction (flow or batch) [205]. These parameters critically affect the efficiency of extraction and the phytochemical composition of the resultant extract [206].
Following extraction, the subsequent step involves the purification of the bioactive compounds from the crude extract. This is necessary because the initial phytochemical mixture typically contains a combination of active and inactive substances, along with impurities that could affect the safety and efficacy of the final product [207]. The degree of purification achieved is linked to the pharmacokinetic properties of the compounds, which influence their bioavailability, distribution, metabolism, and excretion [208]. Recent advancements in the purification and isolation techniques for bioactive compounds from plant sources have addressed some of the challenges posed by the complexity and diversity of phytochemicals [209,210]. These advancements focus on optimizing methods to concentrate the desired compounds, thereby enhancing bioactivity—such as antioxidant, antibacterial, or cytotoxic effects—while maintaining simplicity, specificity, and efficiency in processing [211,212]. Techniques such as high-performance liquid chromatography (HPLC), solid-phase extraction (SPE), and preparative thin-layer chromatography (TLC) are commonly used for the isolation and purification of bioactive molecules because of their practicality, cost-effectiveness, and the availability of various stationary phases [204,211]. Notably, silica, alumina, cellulose, and polyamide have proven effective in separating phytochemicals [211]. To achieve the separation of complex mixtures, the use of multiple mobile phases with varying polarities is often required [204,211].
Despite the successful purification of phytochemicals, the formulation of these compounds into nutraceuticals remains challenged by issues of stability, solubility, and bioavailability. Encapsulation technologies have emerged as vital strategies to overcome these challenges [213,214]. Additionally, encapsulation can improve the quality of food products by masking undesirable odors and flavors and extending shelf life [203]. This process involves enclosing phytochemicals within a protective matrix or carrier system, which shields them from environmental degradation, enhances their solubility in biological fluids, and facilitates controlled release in the gastrointestinal tract [215,216]. Various encapsulation methods have been developed, each offering unique advantages depending on the physicochemical properties of the phytochemical and its intended application [217]. Microencapsulation, for instance, is widely used to protect sensitive compounds and improve their stability during processing and storage [218,219]. Conversely, nanoencapsulation provides enhanced bioavailability and targeted delivery due to small particle sizes and large surface areas [220,221], which improve interactions with biological membranes and allow for more precise control over release kinetics [222,223].
In conclusion, the effective application of phytochemicals as nutraceuticals is closely linked to advances in extraction, purification, and encapsulation techniques. These processes not only enhance the pharmacological potential of phytochemicals but also ensure their stability, bioavailability, and functional integration into various nutraceutical products.

4.2. Considerations of Bioavailability

Phytochemicals offer a wide range of health benefits, yet their effectiveness is often compromised by challenges related to bioavailability, including issues with water dispersibility, chemical stability, and gastrointestinal absorption [224]. For phytochemicals to be effective, they must be released from their food matrices, solubilized in gastrointestinal fluids, and absorbed by enterocytes in the gastrointestinal tract. However, these compounds may precipitate, interact with other dietary molecules, or be chemically altered by digestive enzymes, metabolic pathways, or gut microbiota, which can significantly reduce their bioactive forms by the time they enter systemic circulation [225].
Therefore, the bioavailability of phytochemicals is affected by several factors, including the complexity of the food matrix, the chemical form of the compound, and individual physiological differences, such as gut microbiota composition, mucosal health, and metabolic activity. These factors contribute to considerable inter- and intra-individual variability in bioavailability, complicating the prediction of their efficacy [226]. Additionally, the impact of food processing on phytochemical stability and bioavailability is critical, as processing techniques can either enhance or diminish their bioactive potential [227].
To address these issues, in vitro digestion models and cellular uptake assays are employed to assess factors affecting both bioaccessibility and bioavailability. These models are essential for screening and optimizing formulations prior to more expensive in vivo studies. Despite the availability of various assessment methods, a universally accepted standard for bioaccessibility remains elusive, and the current methods must account for the diverse structural characteristics of phytochemicals [208]. For lipophilic compounds, such as carotenoids and fat-soluble vitamins, formulation techniques like encapsulation and emulsification have shown potential in enhancing bioaccessibility and protecting these compounds from degradation [208].
In silico modeling, commonly used in pharmaceutical research, offers an interesting approach as it allows for predicting phytochemical bioavailability based on physicochemical properties. However, these models are still being developed for dietary phytochemicals, indicating a need for further research to establish effective predictive tools [228]. Moreover, technological processes, such as thermal processing and biotechnological interventions, including enzymatic hydrolysis and fermentation, have demonstrated their influence in improving the bioavailability of phenolic compounds. However, while these strategies have shown promise in animal studies, further human trials are necessary to validate their effectiveness [229]. Thus, both research and innovation in processing and formulation techniques are essential for advancing nutraceuticals and optimizing the bioavailability of phytochemicals.

5. Current Applications of Nutraceuticals

Nutraceuticals, which combine nutritional and medicinal properties, are increasingly being incorporated into food products to promote health and prevent disease. These bioactive compounds, derived from a variety of matrices, offer a wide range of benefits, such as improving cardiovascular and immune system function and managing chronic diseases. In addition, advances in encapsulation and nanotechnology have further improved their efficacy in food applications, expanding the potential of health-promoting foods [204].

5.1. Ingredients in Functional Foods

The incorporation of antioxidants into functional foods has been widely researched because of their health benefits and potential to enhance food quality and shelf life. This incorporation can be performed in several ways, including through the addition of crude extracts, fibers, color additives, and other food enrichment (Figure 5) [230].
Crude extracts are raw, unrefined substances obtained from plants, animals, and other natural sources that contain bioactive compounds. The most widely utilized are plant polyphenol extracts, such as green tea extract, grape seed extract, and rosemary extract, but also mushroom, spice, and herbal extracts (garlic extract, ginger extract, turmeric extract), vanilla extract, and, more recently, microalgae extract (Spirulina, Chlorella extracts) [231]. These extracts also exhibit antimicrobial activity, with green tea extract having major antimicrobial activity against major foodborne pathogens, such as Listeria monocytogenes, Salmonella typhimurium, Escherichia coli O157:H7, and Campylobacter jejuni, contributing to food safety [232,233]. Some crude extracts also contribute to flavoring, such as mushroom extracts, garlic extracts, rosemary extracts, and spice and herbal extracts, in general. These can also provide distinctive aromas and colors, enhancing their sensory attributes. For example, Spirulina extracts that are naturally greenish blue can modify the color of a food [234].
Fiber is added to foods because of its functional and health benefits. Fiber can be roughly divided into soluble and insoluble fibers. Soluble fiber includes pectin and inulin, while insoluble fiber includes cellulose, psyllium, and wheat bran. Functionally, adding fiber to foods can impact their sensory properties, such as appearance, texture, taste, and flavor [235]. Pectin can be found in fruit jams and jellies; it acts as a gelling agent, giving jams and jellies their thick, spreadable consistency. Additionally, it allows for a lower amount of sugar to be used while improving product shelf life and stability [236]. Inulin, usually extracted from chicory root, is used as a fat replacer and provides a creamy texture in low-fat or reduced-calorie foods [237]. It also serves as a prebiotic, promoting the growth of beneficial gut bacteria [238]. Cellulose and its derivatives, such as microcrystalline cellulose and bacterial cellulose, are commonly used as stabilizers and thickeners in food products, including meat products, emulsions, beverages, dairy products, and bakery items [239]. Furthermore, microcrystalline cellulose and other cellulose derivatives are used in functional and nutraceutical foods for their positive effects on gastrointestinal health and lipid metabolism [239]. Cellulose gels are also widely used in the food industry for their high-water absorption capacity and biocompatibility. They are applied in food packaging, functional foods, and food safety because of their structural flexibility and stimuli-responsive properties [240]. Other more unrefined fibers, such as psyllium husk and wheat bran, are also often used in the food industry. Unmodified wheat bran and psyllium husk are used as nutritional enhancements as they are a source of dietary fiber, minerals, vitamins, and bioactive compounds, such as phenolic acids, which contribute to improved bowel health, the prevention of diseases, like colon cancer, and cardiovascular health [241]. These fibers, when modified through processes such as mechanical milling, enzymatic hydrolysis, and thermal treatments, can be utilized as functional ingredients. For example, modified wheat bran can be incorporated into cereal foods, baked products, and fried snacks to reduce their oil content and increase their fiber content [241].
To specifically modify the color of a food, color additives can be used. The main natural color additives used in the food industry are anthocyanins, beta-carotene, betanin, and curcumin. Unlike synthetic dyes, these are usually less resistant to degradation via sunlight or heat exposure, but they do not carry risks, like causing hyperactivity in children [242]. Anthocyanins are widely used as natural food colorants because of their vibrant colors, which range from red to blue depending on pH levels [243,244]. Often, these anthocyanins are chemically modified, including acylation and glycosylation, which offers enhanced color stability, making them more suitable for industrial applications as natural colorants [245]. These also provide additional health benefits, improving carbohydrate metabolism and decreasing the risk factors of metabolic disorders [245]. Anthocyanins are used in beverages, such as fruit juices, smoothies, teas, and energy drinks, dairy products, such as flavored milk and ice creams, candies, jams and jellies, bakery products, and many others [246]. β-carotene is a pro-vitamin A carotenoid that contributes to the recommended intake of this essential nutrient in foods where it is added [60]. β-carotene is used to provide an orange hue to various food products, such as margarine and butter substitutes, fruit juices and smoothies, dairy products, such as cheese and yogurt, snacks, baked goods, and infant formula, among many others [247]. Betanin, a red-violet pigment found in beetroots, is also widely used in the food industry. Betanin exhibits high antioxidant activity, which helps to scavenge reactive oxygen species and prevent lipid oxidation in foods, thereby preserving food quality [248,249]. Betanin shows significant stability at low temperatures, which is beneficial for its use in frozen and refrigerated foods [249]. Encapsulation techniques, such as liposomal nanocarriers and microencapsulation, improve the stability and bioavailability of betanin [250,251]. Furthermore, betanin can delay the retrogradation of starches, which is beneficial for maintaining the quality and texture of starchy foods, like bread and pastries [252].
Curcumin, whether as a turmeric extract or an unprocessed form, is commonly used to impart a yellowish-orange hue to foods [253]. It has also demonstrated potential in the prevention and management of various health conditions, including cardiovascular diseases, diabetes, metabolic syndrome, arthritis, and mental disorders [254,255].
In conclusion, the incorporation of antioxidants, fibers, crude extracts, and natural color additives into functional foods has been shown to enhance not only the nutritional value but also the sensory quality, shelf life, and safety of various products. Both soluble and insoluble fibers, such as pectin, inulin, cellulose, psyllium husk, and wheat bran, play crucial roles in improving texture and stability while providing health benefits, like enhanced digestive health and disease prevention. Some examples of the mentioned nutraceuticals include Curcumin Elite™ or Mega Green Tea Extract™ from Life Extension® Ltd. [256], Inülin from Töufood® Ltd. [257], and Blue Spirulina from Bluetec Colorants® Ltd. [258]

5.2. Isolated Phytochemicals as Nutraceuticals

Isolated phytochemicals are bioactive compounds derived from plants that have gained significant attention as nutraceuticals because of their potent health-promoting properties. These compounds, which include flavonoids, alkaloids, terpenoids, and polyphenols, are being increasingly isolated and studied for their potential to prevent and manage various chronic diseases, such as cancer, cardiovascular disorders, and neurodegenerative conditions (Table 3). Phytochemicals are often extracted from agricultural and food waste streams, promoting a circular economy by converting waste into value-added functional ingredients [259]. By concentrating the active ingredients, isolated phytochemicals offer more targeted therapeutic effects compared to whole plant extracts, making them valuable in the development of functional foods and dietary supplements [200].
Polyphenol extracts, which are concentrated from fruits, vegetables, herbs, and seeds, are well known for their antioxidant properties. Green tea catechins, particularly epigallocatechin-3-gallate, are effective in delaying lipid oxidation and extending the shelf life of lipid-containing foods [231,254]. These polyphenols are also incorporated into convenient products, such as chewing gum and gelatin gummies, enhancing consumer accessibility and palatability [277]. Furthermore, green tea extracts demonstrate antimicrobial properties against common foodborne pathogens, enhancing both food safety and quality [232]. Similarly, resveratrol, often sourced from grape industry waste, exhibits strong antioxidant and antibacterial activity, making it suitable for use in oil-in-water emulsions, bulk oils, and ground meat and as a potential natural preservative in food packaging and processing [278,279].
Omega-3 FAs, particularly eicosatetraenoic acid (EPA) and docosahexaenoic acid (DHA), are widely recognized for their roles in supporting brain development and cardiovascular health [280]. When sourced from marine species, such as some microalgae like Schizochytrium and Nannochloropsis, these oils are extracted through solvent methods [281]. Because of the growing consumer demand for plant-based omega-3s, microalgae supplements have become popular as they contain lower levels of heavy metals compared to traditional fish oils [282]. However, the high cost and instability of algae oils limit their use in food products, although nano-emulsification techniques have shown promise in stabilizing omega-3s for functional food applications [283]. Additionally, algae-derived omega-3s are used in livestock feed, which enriches meat, milk, and eggs with bioactive compounds that benefit human health [284], including increasing omega-3 blood indices and reducing diastolic blood pressure [285].
Fucoidans are polysaccharides extracted from brown algae that have gained recognition for their multifunctional bioactive properties. These include anticoagulant, antiviral, antitumor, antibacterial, and immunomodulatory effects [286]. Fucoidans are also valuable as gelling and thickening agents in the food industry, offering both functional and health benefits [287]. Their solubility and ease of incorporation into food formulations make them suitable for various applications, including the development of UVB-protective cosmetics and functional foods [288,289].
Agar and carrageenan, two polysaccharides derived from red algae, are widely used as gelling, thickening, and stabilizing agents in food products. Agar’s ability to form gels at low concentrations makes it ideal for candies, soups, snacks, and plant-based formulations [273,290]. Agar is also often used in plant-based and halal formulations of products that otherwise require animal gelatin [291]. Additionally, agar and its derivatives exhibit multiple bioactive functions, such as antioxidant, antiviral, antibacterial, prebiotic, antitumor, anticoagulant, and antidiabetic activities. These bioactive properties make agar a valuable ingredient in functional foods and nutraceuticals [273,290]. Carrageenan is similarly utilized as a thickening, gelling, and protein-suspending agent, enhancing texture and water retention in food products while also providing various health benefits [292]. In low-fat meat formulations, the incorporation of carrageenan promotes gel formation and water retention, thereby improving textural properties [292]. Similar to agar, carrageenan exhibits anti-inflammatory, anticoagulant, antitumor, and antithrombotic effects [293].
Quercetin, a flavonoid present in a variety of plants, is predominantly extracted from onions and apple peels because of their commercial availability [294]. In food products like bread, quercetin demonstrates antioxidant and antiglycation effects in a dose-dependent manner. Furthermore, it inhibits the formation of advanced glycation end products (AGEs), thereby enhancing the shelf life of these products [295]. Quercetin has also been shown to offer protective benefits against cardiovascular and neurodegenerative diseases, underscoring its potential as a valuable bioactive compound for the development of functional foods aimed at promoting health [296].
In summary, phytochemicals, such as polyphenols, omega-3-rich oils, fucoidans, agar, carrageenan, and quercetin, are increasingly recognized for their dual roles in improving both the quality and health benefits of food products. These compounds, sourced from plants and algae, contribute to enhanced shelf life, nutritional value, and functional properties while offering a wide range of health benefits, including antioxidant, antimicrobial, and cardiovascular protective effects. Some commercialized available examples of these ingredients include Ceamsea ™ from Ceamsa Ltd. [297], Fucoidan Extract from Swanson Ltd. [298], Agar-Agar from Algamar Ltd. [299], and Omega-3 formula from Kohbo Labs Ltd. [300].

6. Conclusions

This review uncovers the versatile functions of antioxidants in plant organisms and their involvement in protecting against OS. Several antioxidant compounds, such as flavonoids, carotenoids, and phenolic acids, promote plant resistance and are also beneficial for human health. A better understanding of these mechanisms may allow the optimization of agricultural practices and food supplies. Further research should be directed towards the more efficient use of plant antioxidants to improve their productive capacity and nutraceutical value. In this way, the integration of advanced omics technologies, such as metabolomics and transcriptomics, may improve the understanding of the biosynthetic pathways and regulatory networks of plant antioxidants. Furthermore, the convergence of plant biotechnology with sustainable agricultural practices offers promising avenues not only for increasing crop yields but also for enriching the nutritional and functional qualities of food products.

Author Contributions

Conceptualization: A.G.P., J.E., A.O.S.J., R.N.-M., E.N.Y., P.B., A.P.-V., F.C., M.B.P.P.O., M.C. and M.A.P.; methodology: A.G.P., J.E., A.O.S.J., R.N.-M., E.N.Y., P.B., A.P.-V., F.C. and M.C.; validation, A.G.P., M.B.P.P.O., M.C. and M.A.P.; formal analysis: A.G.P., J.E., A.O.S.J., R.N.-M., E.N.Y., P.B., A.P.-V., F.C. and M.C.; investigation: A.G.P., J.E., A.O.S.J., R.N.-M., E.N.Y., P.B., A.P.-V., F.C. and M.C.; resources: A.G.P., J.E., A.O.S.J., R.N.-M., E.N.Y., P.B., A.P.-V., F.C. and M.C.; data curation: A.G.P., A.O.S.J., P.B., A.P.-V. and M.C.; writing—original draft preparation: A.G.P., J.E., A.O.S.J., R.N.-M., E.N.Y., P.B., A.P.-V., F.C. and M.C.; writing—review and editing: A.G.P., A.O.S.J., M.C., M.B.P.P.O. and M.A.P.; visualization: A.G.P., A.O.S.J., P.B., A.P.-V. and F.C.; supervision: A.G.P., M.C., M.B.P.P.O. and M.A.P.; project administration: M.B.P.P.O. and M.A.P.; funding acquisition: M.B.P.P.O. and M.A.P. All authors have read and agreed to the published version of the manuscript.

Funding

The research leading to these results was supported by MICIU/AEI/10.13039/501100011033, in support of the predoctoral industrial grant for A. Perez-Vazquez (DIN2024-013416) in collaboration with Mercantia Desarrollos Alimentarios S.L.; Xunta de Galicia in support of the post-doctoral grant of A.G. Pereira (IN606B-2024/011); and the pre-doctoral grant of P. Barciela (ED481A-2024-230). The authors thank the EU-FORA Fellowship Program (EUBA-EFSA-2023-ENREL-01) that supports the work of F. Chamorro (INNOV2SAFETY-GA13). The authors are grateful for the national funding from FCT, Foundation for Science and Technology, through the individual research grants of J. Echave (2023.04987.BD) and A.O.S. Jorge (2023.00981.BD).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Carpena, M.; Garcia-Oliveira, P.; Pereira, A.G.; Soria-Lopez, A.; Chamorro, F.; Collazo, N.; Jarboui, A.; Simal-Gandara, J.; Prieto, M.A. Plant Antioxidants from Agricultural Waste: Synergistic Potential with Other Biological Properties and Possible Applications. In Plant Antioxidants and Health; Ekiert, H.M., Ramawat, K.G., Arora, J., Eds.; Springer: Cham, Switzerland, 2021; pp. 1–38. ISBN 9783030452995. [Google Scholar]
  2. Shahidi, F.; Ambigaipalan, P. Phenolics and Polyphenolics in Foods, Beverages and Spices: Antioxidant Activity and Health Effects–A Review. J. Funct. Foods 2015, 18, 820–897. [Google Scholar] [CrossRef]
  3. Jimenez-Lopez, C.; Fraga-Corral, M.; Carpena, M.; García-Oliveira, P.; Echave, J.; Pereira, A.G.; Lourenço-Lopes, C.; Prieto, M.A.; Simal-Gandara, J. Agriculture Waste Valorisation as a Source of Antioxidant Phenolic Compounds within a Circular and Sustainable Bioeconomy. Food Funct. 2020, 11, 4853–4877. [Google Scholar] [CrossRef]
  4. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative Stress, Inflammation, and Cancer: How Are They Linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef]
  5. Pistollato, F.; Battino, M. Role of Plant-Based Diets in the Prevention and Regression of Metabolic Syndrome and Neurodegenerative Diseases. Trends Food Sci. Technol. 2014, 40, 62–81. [Google Scholar] [CrossRef]
  6. Zhang, Y.-J.; Gan, R.-Y.; Li, S.; Zhou, Y.; Li, A.-N.; Xu, D.-P.; Li, H.-B. Antioxidant Phytochemicals for the Prevention and Treatment of Chronic Diseases. Molecules 2015, 20, 21138–21156. [Google Scholar] [CrossRef]
  7. Allaqaband, S.; Dar, A.H.; Patel, U.; Kumar, N.; Nayik, G.A.; Khan, S.A.; Ansari, M.J.; Alabdallah, N.M.; Kumar, P.; Pandey, V.K.; et al. Utilization of Fruit Seed-Based Bioactive Compounds for Formulating the Nutraceuticals and Functional Food: A Review. Front. Nutr. 2022, 9, 902554. [Google Scholar] [CrossRef]
  8. Jabri, A.; Kumar, A.; Verghese, E.; Alameh, A.; Kumar, A.; Khan, M.S.; Khan, S.U.; Michos, E.D.; Kapadia, S.R.; Reed, G.W.; et al. Meta-Analysis of Effect of Vegetarian Diet on Ischemic Heart Disease and All-Cause Mortality. Am. J. Prev. Cardiol. 2021, 7, 100182. [Google Scholar] [CrossRef]
  9. Selinger, E.; Neuenschwander, M.; Koller, A.; Gojda, J.; Kühn, T.; Schwingshackl, L.; Barbaresko, J.; Schlesinger, S. Evidence of a Vegan Diet for Health Benefits and Risks–an Umbrella Review of Meta-Analyses of Observational and Clinical Studies. Crit. Rev. Food Sci. Nutr. 2023, 63, 9926–9936. [Google Scholar] [CrossRef]
  10. Jablonský, M.; Škulcová, A.; Malvis, A.; Šima, J. Extraction of Value-Added Components from Food Industry Based and Agro-Forest Biowastes by Deep Eutectic Solvents. J. Biotechnol. 2018, 282, 46–66. [Google Scholar] [CrossRef]
  11. Oswell, N.J.; Gunstone, F.D.; Pegg, R.B. Vegetable Oils. In Bailey’s Industrial Oil and Fat Products; Wiley: Hoboken, NJ, USA, 2020; pp. 1–30. ISBN 9781439816851. [Google Scholar]
  12. Chouaibi, M.; Rezig, L.; Hamdi, S.; Ferrari, G. Chemical Characteristics and Compositions of Red Pepper Seed Oils Extracted by Different Methods. Ind. Crops Prod. 2019, 128, 363–370. [Google Scholar] [CrossRef]
  13. García-Lomillo, J.; González-SanJosé, M.L. Applications of Wine Pomace in the Food Industry: Approaches and Functions. Compr. Rev. Food Sci. Food Saf. 2017, 16, 3–22. [Google Scholar] [CrossRef]
  14. Lu, Q.; Li, H.; Xiao, Y.; Liu, H. A State-of-the-Art Review on the Synthetic Mechanisms, Production Technologies, and Practical Application of Polyunsaturated Fatty Acids from Microalgae. Algal Res. 2021, 55, 102281. [Google Scholar] [CrossRef]
  15. Russo, G.L.; Moccia, S.; Russo, M.; Spagnuolo, C. Redox Regulation by Carotenoids: Evidence and Conflicts for Their Application in Cancer. Biochem. Pharmacol. 2021, 194, 114838. [Google Scholar] [CrossRef]
  16. Bohn, T. Provitamin a Carotenoids: Occurrence, Intake and Bioavailability. Food Nutr. Components Focus 2012, 1, 142–161. [Google Scholar]
  17. Poojary, M.M.; Barba, F.J.; Aliakbarian, B.; Donsì, F.; Pataro, G.; Dias, D.A.; Juliano, P. Innovative Alternative Technologies to Extract Carotenoids from Microalgae and Seaweeds. Mar. Drugs 2016, 14, 214. [Google Scholar] [CrossRef]
  18. Rodriguez-Concepcion, M.; Avalos, J.; Bonet, M.L.; Boronat, A.; Gomez-Gomez, L.; Hornero-Mendez, D.; Limon, M.C.; Meléndez-Martínez, A.J.; Olmedilla-Alonso, B.; Palou, A.; et al. A Global Perspective on Carotenoids: Metabolism, Biotechnology, and Benefits for Nutrition and Health. Prog. Lipid Res. 2018, 70, 62–93. [Google Scholar] [CrossRef]
  19. Nie, Q.; Hu, J.; Gao, H.; Li, M.; Sun, Y.; Chen, H.; Zuo, S.; Fang, Q.; Huang, X.; Yin, J.; et al. Bioactive Dietary Fibers Selectively Promote Gut Microbiota to Exert Antidiabetic Effects. J. Agric. Food Chem. 2021, 69, 7000–7015. [Google Scholar] [CrossRef]
  20. Baiano, A. Recovery of Biomolecules from Food Wastes—A Review. Molecules 2014, 19, 14821–14842. [Google Scholar] [CrossRef]
  21. Saeed, F.; Pasha, I.; Arshad, M.U.; Anjum, F.M.; Hussain, S.; Rasheed, R.; Nasir, M.A.; Shafique, B. Physiological and Nutraceutical Perspectives of Fructan. Int. J. Food Prop. 2015, 18, 1895–1904. [Google Scholar] [CrossRef]
  22. Pereira, A.G.; Garcia-Perez, P.; Cassani, L.; Chamorro, F.; Cao, H.; Barba, F.J.; Simal-Gandara, J.; Prieto, M.A. Camellia Japonica: A Phytochemical Perspective and Current Applications Facing Its Industrial Exploitation. Food Chem. X 2022, 13, 100258. [Google Scholar] [CrossRef]
  23. Salehi, B.; Martorell, M.; Arbiser, J.L.; Sureda, A.; Martins, N.; Maurya, P.K.; Sharifi-Rad, M.; Kumar, P.; Sharifi-Rad, J. Antioxidants: Positive or Negative Actors? Biomolecules 2018, 8, 124. [Google Scholar] [CrossRef]
  24. Li, S.; Fasipe, B.; Laher, I. Potential Harms of Supplementation with High Doses of Antioxidants in Athletes. J. Exerc. Sci. Fit. 2022, 20, 269–275. [Google Scholar] [CrossRef]
  25. García-Cortés, M.; Robles-Díaz, M.; Ortega-Alonso, A.; Medina-Caliz, I.; Andrade, R. Hepatotoxicity by Dietary Supplements: A Tabular Listing and Clinical Characteristics. Int. J. Mol. Sci. 2016, 17, 537. [Google Scholar] [CrossRef]
  26. Stickel, F.; Kessebohm, K.; Weimann, R.; Seitz, H.K. Review of Liver Injury Associated with Dietary Supplements. Liver Int. 2011, 31, 595–605. [Google Scholar] [CrossRef]
  27. Li, S.; Li, Y.; Smolke, C.D. Strategies for Microbial Synthesis of High-Value Phytochemicals. Nat. Chem. 2018, 10, 395–404. [Google Scholar] [CrossRef]
  28. Reguengo, L.M.; Salgaço, M.K.; Sivieri, K.; Júnior, M.R.M. Agro-Industrial by-Products: Valuable Sources of Bioactive Compounds. Food Res. Int. 2022, 152, 110871. [Google Scholar] [CrossRef]
  29. Watkins, P.A. Fatty Acids: Metabolism. In Encyclopedia of Human Nutrition; Elsevier: Amsterdam, The Netherlands, 2013; Volume 2–4, pp. 220–230. ISBN 9780123848857. [Google Scholar]
  30. Ristic-Medic, D.; Vucic, V.; Takic, M.; Karadžic, I.; Glibetic, M. Polyunsaturated Fatty Acids in Health and Disease. J. Serbian Chem. Soc. 2013, 78, 1269–1289. [Google Scholar] [CrossRef]
  31. Moghadasian, M.H.; Shahidi, F. Fatty Acids. In International Encyclopedia of Public Health; Elsevier: Amsterdam, The Netherlands, 2017; Volume 3, pp. 114–122. ISBN 9780128037089. [Google Scholar]
  32. He, M.; Ding, N.Z. Plant Unsaturated Fatty Acids: Multiple Roles in Stress Response. Front. Plant Sci. 2020, 11, 562785. [Google Scholar] [CrossRef]
  33. Tsao, R. Chemistry and Biochemistry of Dietary Polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef]
  34. Jimenez-Lopez, C.; Pereira, A.G.; Lourenço-Lopes, C.; Garcia-Oliveira, P.; Cassani, L.; Fraga-Corral, M.; Prieto, M.A.; Simal-Gandara, J. Main Bioactive Phenolic Compounds in Marine Algae and Their Mechanisms of Action Supporting Potential Health Benefits. Food Chem. 2021, 341, 128262. [Google Scholar] [CrossRef]
  35. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and Human Health: The Role of Bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef]
  36. Han, X.; Shen, T.; Lou, H. Dietary Polyphenols and Their Biological Significance. Int. J. Mol. Sci. 2007, 8, 950–988. [Google Scholar] [CrossRef]
  37. Zhang, Z.; Li, X.; Sang, S.; McClements, D.J.; Chen, L.; Long, J.; Jiao, A.; Jin, Z.; Qiu, C. Polyphenols as Plant-Based Nutraceuticals: Health Effects, Encapsulation, Nano-Delivery, and Application. Foods 2022, 11, 2189. [Google Scholar] [CrossRef]
  38. Haminiuk, C.W.I.; Maciel, G.M.; Plata-Oviedo, M.S.V.; Peralta, R.M. Phenolic Compounds in Fruits-an Overview. Int. J. Food Sci. Technol. 2012, 47, 2023–2044. [Google Scholar] [CrossRef]
  39. Smeriglio, A.; Barreca, D.; Bellocco, E.; Trombetta, D. Proanthocyanidins and Hydrolysable Tannins: Occurrence, Dietary Intake and Pharmacological Effects. Br. J. Pharmacol. 2017, 174, 1244–1262. [Google Scholar] [CrossRef]
  40. Rashmi, H.B.; Negi, P.S. Phenolic Acids from Vegetables: A Review on Processing Stability and Health Benefits. Food Res. Int. 2020, 136, 109298. [Google Scholar] [CrossRef]
  41. Caleja, C.; Ribeiro, A.; Barreiro, M.F.; Ferreira, I.C.F.R. Phenolic Compounds as Nutraceuticals or Functional Food Ingredients. Curr. Pharm. Des. 2017, 23, 2787–2806. [Google Scholar] [CrossRef]
  42. Vuolo, M.M.; Lima, V.S.; Junior, M.R.M. Phenolic Compounds. In Bioactive Compounds: Health Benefits and Potential Applications; Campos, M.R.S., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 33–50. ISBN 9780128147740. [Google Scholar]
  43. Sorrenti, V.; Burò, I.; Consoli, V.; Vanella, L. Recent Advances in Health Benefits of Bioactive Compounds from Food Wastes and By-Products: Biochemical Aspects. Int. J. Mol. Sci. 2023, 24, 2019. [Google Scholar] [CrossRef]
  44. Lin, J.K.; Weng, M.S. Flavonoids as Nutraceuticals. Sci. Flavonoids 2006, 7, 213–238. [Google Scholar] [CrossRef]
  45. Dias, M.; Romaní-Pérez, M.; Romaní, A.; de la Cruz, A.; Pastrana, L.; Fuciños, P.; Amado, I.R. Recent Technological Advances in Phenolic Compounds Recovery and Applications: Source of Nutraceuticals for the Management of Diabetes. Appl. Sci. 2022, 12, 9271. [Google Scholar] [CrossRef]
  46. Rahman, M.M.; Rahaman, M.S.; Islam, M.R.; Rahman, F.; Mithi, F.M.; Alqahtani, T.; Almikhlafi, M.A.; Alghamdi, S.Q.; Alruwaili, A.S.; Hossain, M.S.; et al. Role of Phenolic Compounds in Human Disease: Current Knowledge and Future Prospects. Molecules 2022, 27, 233. [Google Scholar] [CrossRef]
  47. Vignesh, A.; Amal, T.C.; Sarvalingam, A.; Vasanth, K. A Review on the Influence of Nutraceuticals and Functional Foods on Health. Food Chem. Adv. 2024, 5, 100749. [Google Scholar] [CrossRef]
  48. Maoka, T. Carotenoids as Natural Functional Pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef]
  49. Crupi, P.; Faienza, M.F.; Naeem, M.Y.; Corbo, F.; Clodoveo, M.L.; Muraglia, M. Overview of the Potential Beneficial Effects of Carotenoids on Consumer Health and Well-Being. Antioxidants 2023, 12, 1069. [Google Scholar] [CrossRef]
  50. Lakshminarayana, R.; Paul, B. Free Radical Chemistry of Carotenoids and Oxidative Stress Physiology of Cancer. In Handbook of Oxidative Stress in Cancer: Therapeutic Aspects: Volume 1; Chakraborti, S., Ed.; Springer: Singapore, 2022; Volume 1, pp. 3077–3097. ISBN 9789811654220. [Google Scholar]
  51. Srivastava, R. Physicochemical, Antioxidant Properties of Carotenoids and Its Optoelectronic and Interaction Studies with Chlorophyll Pigments. Sci. Rep. 2021, 11, 18365. [Google Scholar] [CrossRef]
  52. Saini, R.K.; Prasad, P.; Lokesh, V.; Shang, X.; Shin, J.; Keum, Y.S.; Lee, J.H. Carotenoids: Dietary Sources, Extraction, Encapsulation, Bioavailability, and Health Benefits—A Review of Recent Advancements. Antioxidants 2022, 11, 795. [Google Scholar] [CrossRef]
  53. Pereira, A.G.; Otero, P.; Echave, J.; Carreira-Casais, A.; Chamorro, F.; Collazo, N.; Jaboui, A.; Lourenço-Lopes, C.; Simal-Gandara, J.; Prieto, M.A. Xanthophylls from the Sea: Algae as Source of Bioactive Carotenoids. Mar. Drugs 2021, 19, 188. [Google Scholar] [CrossRef]
  54. Widomska, J.; Zareba, M.; Subczynski, W.K. Can Xanthophyll-Membrane Interactions Explain Their Selective Presence in the Retina and Brain? Foods 2016, 5, 7. [Google Scholar] [CrossRef]
  55. Ojulari, O.V.; Lee, S.G.; Nam, J.O. Therapeutic Effect of Seaweed Derived Xanthophyl Carotenoid on Obesity Management; Overview of the Last Decade. Int. J. Mol. Sci. 2020, 21, 2502. [Google Scholar] [CrossRef]
  56. Gürbüz, M.; Aktaç, Ş. Understanding the Role of Vitamin A and Its Precursors in the Immune System. Nutr. Clin. Metab. 2022, 36, 89–98. [Google Scholar] [CrossRef]
  57. Ebadi, M.; Mohammadi, M.; Pezeshki, A.; Jafari, S.M. Health Benefits of Beta-Carotene. In Handbook of Food Bioactive Ingredients; Jafari, S.M., Rashidinejad, A., Simal-Gandara, J., Eds.; Springer International Publishing: Cham, Switzerland, 2023; pp. 1–26. ISBN 978-3-030-81404-5. [Google Scholar]
  58. Aziz, E.; Batool, R.; Akhtar, W.; Rehman, S.; Shahzad, T.; Malik, A.; Shariati, M.A.; Laishevtcev, A.; Plygun, S.; Heydari, M.; et al. Xanthophyll: Health Benefits and Therapeutic Insights. Life Sci. 2020, 240, 117104. [Google Scholar] [CrossRef] [PubMed]
  59. Singh, A.; Omer, K. Recent Advancement in Therapeutic Activity of Carotenoids. In Dietary Carotenoids; Rao, A.V., Rao, L., Eds.; IntechOpen: Rijeka, Croatia, 2024. [Google Scholar]
  60. Grune, T.; Lietz, G.; Palou, A.; Ross, A.C.; Stahl, W.; Tang, G.; Thurnham, D.; Yin, S.A.; Biesalski, H.K. β-Carotene Is an Important Vitamin A Source for Humans. J. Nutr. 2010, 140, 2268S–2285S. [Google Scholar] [CrossRef]
  61. Murphy, E.J.; Fehrenbach, G.W.; Abidin, I.Z.; Buckley, C.; Montgomery, T.; Pogue, R.; Murray, P.; Major, I.; Rezoagli, E. Polysaccharides—Naturally Occurring Immune Modulators. Polymers 2023, 15, 2373. [Google Scholar] [CrossRef]
  62. Pedrosa, L.d.F.; de Vos, P.; Fabi, J.P. Nature’s Soothing Solution: Harnessing the Potential of Food-Derived Polysaccharides to Control Inflammation. Curr. Res. Struct. Biol. 2023, 6, 100112. [Google Scholar] [CrossRef]
  63. Baghel, R.S.; Choudhary, B.; Pandey, S.; Pathak, P.K.; Patel, M.K.; Mishra, A. Rehashing Our Insight of Seaweeds as a Potential Source of Foods, Nutraceuticals, and Pharmaceuticals. Foods 2023, 12, 3642. [Google Scholar] [CrossRef]
  64. Lin, G.p.; Wu, D.s.; Xiao, X.w.; Huang, Q.y.; Chen, H.b.; Liu, D.; Fu, H.q.; Chen, X.h.; Zhao, C. Structural Characterization and Antioxidant Effect of Green Alga Enteromorpha Prolifera Polysaccharide in Caenorhabditis Elegans via Modulation of MicroRNAs. Int. J. Biol. Macromol. 2020, 150, 1084–1092. [Google Scholar] [CrossRef]
  65. Zhang, Y.; Xu, M.; Hu, C.; Liu, A.; Chen, J.; Gu, C.; Zhang, X.; You, C.; Tong, H.; Wu, M.; et al. Sargassum Fusiforme Fucoidan SP2 Extends the Lifespan of Drosophila Melanogaster by Upregulating the Nrf2-Mediated Antioxidant Signaling Pathway. Oxidative Med. Cell. Longev. 2019, 2019, 8918914. [Google Scholar] [CrossRef]
  66. Cui, H.; Wu, S.; Shang, Y.; Li, Z.; Chen, M.; Li, F.; Wang, C. Pleurotus Nebrodensis Polysaccharide(PN50G) Evokes A549 Cell Apoptosis by the ROS/AMPK/PI3K/AKT/MTOR Pathway to Suppress Tumor Growth. Food Funct. 2016, 7, 1616–1627. [Google Scholar] [CrossRef]
  67. Zeng, F.S.; Yao, Y.F.; Wang, L.F.; Li, W.J. Polysaccharides as Antioxidants and Prooxidants in Managing the Double-Edged Sword of Reactive Oxygen Species. Biomed. Pharmacother. 2023, 159, 114221. [Google Scholar] [CrossRef]
  68. Wang, J.; Hu, S.; Nie, S.; Yu, Q.; Xie, M. Reviews on Mechanisms of in Vitro Antioxidant Activity of Polysaccharides. Oxidative Med. Cell. Longev. 2016, 2016, 5692852. [Google Scholar] [CrossRef]
  69. Devi, N.; Sarmah, M.; Khatun, B.; Maji, T.K. Encapsulation of Active Ingredients in Polysaccharide–Protein Complex Coacervates. Adv. Colloid Interface Sci. 2017, 239, 136–145. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, Z.; Sun, X. A Critical Review of the Abilities, Determinants, and Possible Molecular Mechanisms of Seaweed Polysaccharides Antioxidants. Int. J. Mol. Sci. 2020, 21, 7774. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, N.; Jiang, T.; Xu, J.; Xi, W.; Shang, E.; Xiao, P.; Duan, J.a. The Relationship between Polysaccharide Structure and Its Antioxidant Activity Needs to Be Systematically Elucidated. Int. J. Biol. Macromol. 2024, 270, 132391. [Google Scholar] [CrossRef] [PubMed]
  72. Fernandes, P.A.R.; Coimbra, M.A. The Antioxidant Activity of Polysaccharides: A Structure-Function Relationship Overview. Carbohydr. Polym. 2023, 314, 120965. [Google Scholar] [CrossRef]
  73. Bai, L.; Xu, D.; Zhou, Y.M.; Zhang, Y.B.; Zhang, H.; Chen, Y.B.; Cui, Y.L. Antioxidant Activities of Natural Polysaccharides and Their Derivatives for Biomedical and Medicinal Applications. Antioxidants 2022, 11, 2491. [Google Scholar] [CrossRef]
  74. Niyigaba, T.; Liu, D.; Habimana, J.d.D. The Extraction, Functionalities and Applications of Plant Polysaccharides in Fermented Foods: A Review. Foods 2021, 10, 3004. [Google Scholar] [CrossRef]
  75. Sheibani, E.; Hosseini, A.; Nasab, A.S.; Adib, K.; Ganjali, M.R.; Pourmortazavi, S.M.; Ahmadi, F.; Khosrowshahi, E.M.; Mirsadeghi, S.; Rahimi-Nasrabadi, M.; et al. Application of Polysaccharide Biopolymers as Natural Adsorbent in Sample Preparation. Crit. Rev. Food Sci. Nutr. 2023, 63, 2626–2653. [Google Scholar] [CrossRef]
  76. Teng, C.; Qin, P.; Shi, Z.; Zhang, W.; Yang, X.; Yao, Y.; Ren, G. Structural Characterization and Antioxidant Activity of Alkali-Extracted Polysaccharides from Quinoa. Food Hydrocoll. 2021, 113, 106392. [Google Scholar] [CrossRef]
  77. Bhuyan, P.P.; Nayak, R.; Patra, S.; Abdulabbas, H.S.; Jena, M.; Pradhan, B. Seaweed-Derived Sulfated Polysaccharides; The New Age Chemopreventives: A Comprehensive Review. Cancers 2023, 15, 715. [Google Scholar] [CrossRef]
  78. Livingstone, K.M.; Ramos-Lopez, O.; Pérusse, L.; Kato, H.; Ordovas, J.M.; Martínez, J.A. Precision Nutrition: A Review of Current Approaches and Future Endeavors. Trends Food Sci. Technol. 2022, 128, 253–264. [Google Scholar] [CrossRef]
  79. Rhazi, L.; Depeint, F.; Gotor, A.A. Loss in the Intrinsic Quality and the Antioxidant Activity of Sunflower (Helianthus annuus L.) Oil during an Industrial Refining Process. Molecules 2022, 27, 916. [Google Scholar] [CrossRef] [PubMed]
  80. Ambra, R.; Natella, F.; Lucchetti, S.; Forte, V.; Pastore, G. α-Tocopherol, β-Carotene, Lutein, Squalene and Secoiridoids in Seven Monocultivar Italian Extra-Virgin Olive Oils. Int. J. Food Sci. Nutr. 2017, 68, 538–545. [Google Scholar] [CrossRef] [PubMed]
  81. Fagali, N.; Catalá, A. Antioxidant Activity of Conjugated Linoleic Acid Isomers, Linoleic Acid and Its Methyl Ester Determined by Photoemission and DPPH Techniques. Biophys. Chem. 2008, 137, 56–62. [Google Scholar] [CrossRef] [PubMed]
  82. Lewinska, A.; Zebrowski, J.; Duda, M.; Gorka, A.; Wnuk, M. Fatty Acid Profile and Biological Activities of Linseed and Rapeseed Oils. Molecules 2015, 20, 22872–22880. [Google Scholar] [CrossRef]
  83. Hulatt, C.J.; Wijffels, R.H.; Bolla, S.; Kiron, V. Production of Fatty Acids and Protein by Nannochloropsis in Flat-Plate Photobioreactors. PLoS ONE 2017, 12, e0170440. [Google Scholar] [CrossRef]
  84. Xiao, B.; Li, Y.; Lin, Y.; Lin, J.; Zhang, L.; Wu, D.; Zeng, J.; Li, J.; Liu, J.w.; Li, G. Eicosapentaenoic Acid (EPA) Exhibits Antioxidant Activity via Mitochondrial Modulation. Food Chem. 2022, 373, 131389. [Google Scholar] [CrossRef]
  85. Li, G.; Li, Y.; Xiao, B.; Cui, D.; Lin, Y.; Zeng, J.; Li, J.; Cao, M.-J.; Liu, J. Antioxidant Activity of Docosahexaenoic Acid (DHA) and Its Regulatory Roles in Mitochondria. J. Agric. Food Chem. 2021, 69, 1647–1655. [Google Scholar] [CrossRef]
  86. Li, Z.; Chen, X.; Li, J.; Meng, T.; Wang, L.; Chen, Z.; Shi, Y.; Ling, X.; Luo, W.; Liang, D.; et al. Functions of PKS Genes in Lipid Synthesis of Schizochytrium Sp. by Gene Disruption and Metabolomics Analysis. Mar. Biotechnol. 2018, 20, 792–802. [Google Scholar] [CrossRef]
  87. Rodrigues, N.; Casal, S.; Pinho, T.; Cruz, R.; Peres, A.M.; Baptista, P.; Pereira, J.A. Fatty Acid Composition from Olive Oils of Portuguese Centenarian Trees Is Highly Dependent on Olive Cultivar and Crop Year. Foods 2021, 10, 496. [Google Scholar] [CrossRef]
  88. Petraru, A.; Ursachi, F.; Amariei, S. Nutritional Characteristics Assessment of Sunflower Seeds, Oil and Cake. Perspective of Using Sunflower Oilcakes as a Functional Ingredient. Plants 2021, 10, 2487. [Google Scholar] [CrossRef]
  89. Ali, Y.M.; Kadir, A.A.; Ahmad, Z.; Yaakub, H.; Zakaria, Z.A.; Abdullah, M.N.H. Free Radical Scavenging Activity of Conjugated Linoleic Acid as Single or Mixed Isomers. Pharm. Biol. 2012, 50, 712–719. [Google Scholar] [CrossRef] [PubMed]
  90. Zhou, W.; Wang, H.; Chen, L.; Cheng, W.; Liu, T. Bioresource Technology Heterotrophy of Filamentous Oleaginous Microalgae Tribonema Minus for Potential Production of Lipid and Palmitoleic Acid. Bioresour. Technol. 2017, 239, 250–257. [Google Scholar] [CrossRef]
  91. Borges, T.H.; Pereira, J.A.; Cabrera-Vique, C.; Lara, L.; Oliveira, A.F.; Seiquer, I. Characterization of Arbequina Virgin Olive Oils Produced in Different Regions of Brazil and Spain: Physicochemical Properties, Oxidative Stability and Fatty Acid Profile. Food Chem. 2017, 215, 454–462. [Google Scholar] [CrossRef]
  92. Hu, W.; Fitzgerald, M.; Topp, B.; Alam, M.; O’Hare, T.J. A Review of Biological Functions, Health Benefits, and Possible de Novo Biosynthetic Pathway of Palmitoleic Acid in Macadamia Nuts. J. Funct. Foods 2019, 62, 103520. [Google Scholar] [CrossRef]
  93. Mohammed, D.; Freije, A.; Abdulhussain, H.; Khonji, A.; Hasan, M.; Ferraris, C.; Gasparri, C.; Aljar, M.A.A.; Redha, A.A.; Giacosa, A.; et al. Analysis of the Antioxidant Activity, Lipid Profile, and Minerals of the Skin and Seed of Hazelnuts (Corylus avellana L.), Pistachios (Pistacia vera) and Almonds (Prunus dulcis)—A Comparative Analysis. AppliedChem 2023, 3, 110–118. [Google Scholar] [CrossRef]
  94. Shuai, X.; Dai, T.; Chen, M.; Liang, R.; Du, L.; Chen, J.; Liu, C. Comparative Study of Chemical Compositions and Antioxidant Capacities of Oils Obtained from 15 Macadamia (Macadamia integrifolia) Cultivars in China. Foods 2021, 10, 1031. [Google Scholar] [CrossRef]
  95. Górnaś, P.; Rudzińska, M.; Grygier, A.; Lācis, G. Diversity of Oil Yield, Fatty Acids, Tocopherols, Tocotrienols, and Sterols in the Seeds of 19 Interspecific Grapes Crosses. J. Sci. Food Agric. 2019, 99, 2078–2087. [Google Scholar] [CrossRef]
  96. Henry, G.E.; Momin, R.A.; Nair, M.G.; Dewitt, D.L. Antioxidant and Cyclooxygenase Activities of Fatty Acids Found in Food. J. Agric. Food Chem. 2002, 50, 2231–2234. [Google Scholar] [CrossRef]
  97. Wang, J.; Zhang, Y.; Fang, Z.; Sun, L.; Wang, Y.; Liu, Y.; Xu, D.; Nie, F.; Gooneratne, R. Oleic Acid Alleviates Cadmium-Induced Oxidative Damage in Rat by Its Radicals Scavenging Activity. Biol. Trace Elem. Res. 2019, 190, 95–100. [Google Scholar] [CrossRef]
  98. Xavier, A.A.O.; Pérez-Gálvez, A. Carotenoids as a Source of Antioxidants in the Diet. In Sub-Cellular Biochemistry; Stange, C., Ed.; Springer International Publishing: Cham, Switzerland, 2016; Volume 79, pp. 359–375. ISBN 978-3-319-39126-7. [Google Scholar]
  99. Bozalan, N.K.; Karadeniz, F. Carotenoid Profile, Total Phenolic Content, and Antioxidant Activity of Carrots. Int. J. Food Prop. 2011, 14, 1060–1068. [Google Scholar] [CrossRef]
  100. Lourenço-Lopes, C.; Fraga-Corral, M.; Jimenez-Lopez, C.; Carpena, M.; Pereira, A.G.; Garcia-Oliveira, P.; Prieto, M.A.; Simal-Gandara, J. Biological Action Mechanisms of Fucoxanthin Extracted from Algae for Application in Food and Cosmetic Industries. Trends Food Sci. Technol. 2021, 117, 163–181. [Google Scholar] [CrossRef]
  101. Ambati, R.R.; Moi, P.S.; Ravi, S.; Aswathanarayana, R.G. Astaxanthin: Sources, Extraction, Stability, Biological Activities and Its Commercial Applications—A Review. Mar. Drugs 2014, 12, 128–152. [Google Scholar] [CrossRef] [PubMed]
  102. Iwamoto, H.; Soccol, C.R.; Molina-Aulestia, D.T.; Cardoso, J.; de Melo Pereira, G.V.; de Souza Vandenberghe, L.P.; Manzoki, M.C.; Ambati, R.R.; Ravishankar, G.A.; de Carvalho, J.C. Lutein from Microalgae: An Industrial Perspective of Its Production, Downstream Processing, and Market. Fermentation 2024, 10, 106. [Google Scholar] [CrossRef]
  103. Tudor, C.; Pintea, A. A Brief Overview of Dietary Zeaxanthin Occurrence and Bioaccessibility. Molecules 2020, 25, 4067. [Google Scholar] [CrossRef]
  104. Burri, B.J.; La Frano, M.R.; Zhu, C. Absorption, Metabolism, and Functions of β-Cryptoxanthin. Nutr. Rev. 2016, 74, 69–82. [Google Scholar] [CrossRef]
  105. Brahma, D.; Dutta, D. Evaluating β-Cryptoxanthin Antioxidant Properties against ROS-Induced Macromolecular Damages and Determining Its Photo-Stability and in-Vitro SPF. World J. Microbiol. Biotechnol. 2023, 39, 310. [Google Scholar] [CrossRef]
  106. Galla, R.; Ruga, S.; Aprile, S.; Ferrari, S.; Brovero, A.; Grosa, G.; Molinari, C.; Uberti, F. New Hyaluronic Acid from Plant Origin to Improve Joint Protection—An In Vitro Study. Int. J. Mol. Sci. 2022, 23, 8114. [Google Scholar] [CrossRef]
  107. Mohammed, A.A.; Niamah, A.K. Identification and Antioxidant Activity of Hyaluronic Acid Extracted from Local Isolates of Streptococcus Thermophilus. Mater. Today Proc. 2022, 60, 1523–1529. [Google Scholar] [CrossRef]
  108. Schiraldi, C.; Cimini, D.; De Rosa, M. Production of Chondroitin Sulfate and Chondroitin. Appl. Microbiol. Biotechnol. 2010, 87, 1209–1220. [Google Scholar] [CrossRef]
  109. Campo, G.M.; Avenoso, A.; Campo, S.; Ferlazzo, A.M.; Calatroni, A. Antioxidant Activity of Chondroitin Sulfate. In Advances in Pharmacology; Academic Press: Cambridge, MA, USA, 2006; Volume 53, pp. 417–431. ISBN 0120329557. [Google Scholar]
  110. Pavão, M.S.G.; Mourão, P.A.S. Challenges for Heparin Production: Artificial Synthesis or Alternative Natural Sources? Glycobiol. Insights 2012, 3, 1–6. [Google Scholar] [CrossRef]
  111. Bijak, M.; Saluk, J.; Szelenberger, R.; Nowak, P. Popular Naturally Occurring Antioxidants as Potential Anticoagulant Drugs. Chem. Biol. Interact. 2016, 257, 35–45. [Google Scholar] [CrossRef] [PubMed]
  112. Medlej, M.K.; Batoul, C.; Olleik, H.; Li, S.; Hijazi, A.; Nasser, G.; Maresca, M.; Pochat-Bohatier, C. Antioxidant Activity and Biocompatibility of Fructo-Polysaccharides Extracted from a Wild Species of Ornithogalum from Lebanon. Antioxidants 2021, 10, 68. [Google Scholar] [CrossRef] [PubMed]
  113. Delattre, C.; Fenoradosoa, T.A.; Michaud, P. Galactans: An Overview of Their Most Important Sourcing and Applications as Natural Polysaccharides. Brazilian Arch. Biol. Technol. 2011, 54, 1075–1092. [Google Scholar] [CrossRef]
  114. Srivastava, P.; Malviya, R. Sources of Pectin, Extraction and Its Applications in Pharmaceutical Industry—an Overview. Indian J. Nat. Prod. Resour. 2011, 2, 10–18. [Google Scholar]
  115. Wathoni, N.; Shan, C.Y.; Shan, W.Y.; Rostinawati, T.; Indradi, R.B.; Pratiwi, R.; Muchtaridi, M. Characterization and Antioxidant Activity of Pectin from Indonesian Mangosteen (Garcinia mangostana L.) Rind. Heliyon 2019, 5, e02299. [Google Scholar] [CrossRef]
  116. Indrianingsih, A.W.; Rosyida, V.T.; Apriyana, W.; Hayati, S.N.; Darsih, C.; Nisa, K.; Ratih, D. Antioxidant and Antibacterial Properties of Bacterial Cellulose-Indonesian Plant Extract Composites for Mask Sheet. J. Appl. Pharm. Sci. 2020, 10, 37–42. [Google Scholar] [CrossRef]
  117. Cabañas-Romero, L.V.; Valls, C.; Valenzuela, S.V.; Roncero, M.B.; Pastor, F.I.J.; Diaz, P.; Martínez, J. Bacterial Cellulose-Chitosan Paper with Antimicrobial and Antioxidant Activities. Biomacromolecules 2020, 21, 1568–1577. [Google Scholar] [CrossRef]
  118. Hashem, A.H.; Al Abboud, M.A.; Alawlaqi, M.M.; Abdelghany, T.M.; Hasanin, M. Synthesis of Nanocapsules Based on Biosynthesized Nickel Nanoparticles and Potato Starch: Antimicrobial, Antioxidant, and Anticancer Activity. Starch/Staerke 2022, 74, 2100165. [Google Scholar] [CrossRef]
  119. Mikulíková, D.; Kraic, J.Á.N. Natural Sources of Health-Promoting Starch Natural Sources of Health-Promoting Starch. J. Food Nutr. Res. 2018, 45, 69–76. [Google Scholar]
  120. Zhang, R.; Edgar, K.J. Properties, Chemistry, and Applications of the Bioactive Polysaccharide Curdlan. Biomacromolecules 2014, 15, 1079–1096. [Google Scholar] [CrossRef]
  121. Li, H.; Xu, S.; Xie, Y.; Zhang, Q.; Ding, S.; Wang, R.; Fu, F.; Zhan, X. Curdlan-Polyphenol Complexes Prepared by PH-Driven Effectively Enhanced Their Physicochemical Stability, Antioxidant and Prebiotic Activities. Int. J. Biol. Macromol. 2024, 267, 131579. [Google Scholar] [CrossRef] [PubMed]
  122. Kareem, A.J.; Salman, J.A.S. Production of Dextran from Locally Lactobacillus Spp. Isolates. Reports Biochem. Mol. Biol. 2019, 8, 278–286. [Google Scholar]
  123. Rosca, I.; Turin-Moleavin, I.A.; Sarghi, A.; Lungoci, A.L.; Varganici, C.D.; Petrovici, A.R.; Fifere, A.; Pinteala, M. Dextran Coated Iron Oxide Nanoparticles Loaded with Protocatechuic Acid as Multifunctional Therapeutic Agents. Int. J. Biol. Macromol. 2024, 256, 128314. [Google Scholar] [CrossRef] [PubMed]
  124. Jonas, R.; Farah, L.F. Production and Application of Microbial Cellulose. Polym. Degrad. Stab. 1998, 59, 101–106. [Google Scholar] [CrossRef]
  125. Rop, O.; Mlcek, J.; Jurikova, T. Beta-Glucans in Higher Fungi and Their Health Effects. Nutr. Rev. 2009, 67, 624–631. [Google Scholar] [CrossRef]
  126. Mebrek, S.; Djeghim, H.; Mehdi, Y.; Meghezzi, A.; Anwar, S.; Awadh, N.A.A.; Benali, M. Antioxidant, Anti-Cholinesterase, Anti-α-Glucosidase and Prebiotic Properties of Beta-Glucan Extracted from Algerian Barley. Int. J. Phytomedicine 2018, 10, 58. [Google Scholar] [CrossRef]
  127. Huq, T.; Khan, A.; Brown, D.; Dhayagude, N.; He, Z.; Ni, Y. Sources, Production and Commercial Applications of Fungal Chitosan: A Review. J. Bioresour. Bioprod. 2022, 7, 85–98. [Google Scholar] [CrossRef]
  128. Xing, R.; Yu, H.; Liu, S.; Zhang, W.; Zhang, Q.; Li, Z.; Li, P. Antioxidant Activity of Differently Regioselective Chitosan Sulfates in Vitro. Bioorganic Med. Chem. 2005, 13, 1387–1392. [Google Scholar] [CrossRef]
  129. De Souza, M.C.R.; Marques, C.T.; Dore, C.M.G.; Da Silva, F.R.F.; Rocha, H.A.O.; Leite, E.L. Antioxidant Activities of Sulfated Polysaccharides from Brown and Red Seaweeds. J. Appl. Phycol. 2007, 19, 153–160. [Google Scholar] [CrossRef]
  130. Rhein-Knudsen, N.; Reyes-Weiss, D.; Horn, S.J. Extraction of High Purity Fucoidans from Brown Seaweeds Using Cellulases and Alginate Lyases. Int. J. Biol. Macromol. 2023, 229, 199–209. [Google Scholar] [CrossRef]
  131. Pereira, L.; COTAS, J. Alginates-Recent Uses of This Natural Polymer; IntechOpen: Rijeka, Croatia, 2019; ISBN 978-1-78985-642-2. [Google Scholar]
  132. El-Sheekh, M.; Kassem, W.M.A.; Alwaleed, E.A.; Saber, H. Optimization and Characterization of Brown Seaweed Alginate for Antioxidant, Anticancer, Antimicrobial, and Antiviral Properties. Int. J. Biol. Macromol. 2024, 278, 134715. [Google Scholar] [CrossRef] [PubMed]
  133. Machado, B.; Costa, S.M.; Costa, I.; Fangueiro, R.; Ferreira, D.P. The Potential of Algae as a Source of Cellulose and Its Derivatives for Biomedical Applications. Cellulose 2024, 31, 3353–3376. [Google Scholar] [CrossRef]
  134. Petchsomrit, A.; Chanthathamrongsiri, N.; Jiangseubchatveera, N.; Manmuan, S.; Leelakanok, N.; Plianwong, S.; Siranonthana, N.; Sirirak, T. Extraction, Antioxidant Activity, and Hydrogel Formulation of Marine Cladophora Glomerata. Algal Res. 2023, 71, 103011. [Google Scholar] [CrossRef]
  135. Al Harthi, S.S.; Mavazhe, A.; Al Mahroqi, H.; Khan, S.A. Quantification of Phenolic Compounds, Evaluation of Physicochemical Properties and Antioxidant Activity of Four Date (Phoenix dactylifera L.) Varieties of Oman. J. Taibah Univ. Med. Sci. 2015, 10, 346–352. [Google Scholar] [CrossRef]
  136. Palmieri, M.G.S.; Cruz, L.T.; Bertges, F.S.; Húngaro, H.M.; Batista, L.R.; da Silva, S.S.; Fonseca, M.J.V.; Rodarte, M.P.; Vilela, F.M.P.; Amaral, M.d.P.H.d. Enhancement of Antioxidant Properties from Green Coffee as Promising Ingredient for Food and Cosmetic Industries. Biocatal. Agric. Biotechnol. 2018, 16, 43–48. [Google Scholar] [CrossRef]
  137. Chiou, S.Y.; Sung, J.M.; Huang, P.W.; Lin, S.D. Antioxidant, Antidiabetic, and Antihypertensive Properties of Echinacea Purpurea Flower Extract and Caffeic Acid Derivatives Using in Vitro Models. J. Med. Food 2017, 20, 171–179. [Google Scholar] [CrossRef]
  138. Gharat, N.N.; Rathod, V.K. Extraction of Ferulic Acid from Rice Bran Using NADES-Ultrasound-Assisted Extraction: Kinetics and Optimization. J. Food Process Eng. 2023, 46, e14158. [Google Scholar] [CrossRef]
  139. Rivero-Cruz, J.F.; Granados-Pineda, J.; Pedraza-Chaverri, J.; Pérez-Rojas, J.M.; Kumar-Passari, A.; Diaz-Ruiz, G.; Rivero-Cruz, B.E. Phytochemical Constituents, Antioxidant, Cytotoxic, and Antimicrobial Activities of the Ethanolic Extract of Mexican Brown Propolis. Antioxidants 2020, 9, 70. [Google Scholar] [CrossRef]
  140. Priyandoko, D.; Widowati, W.; Kusuma, H.S.W.; Afifah, E.; Wijayanti, C.R.; Wahyuni, C.D.; Idris, A.M.; Putdayani, R.A.; Rizal, R. Antioxidant Activity of Green Tea Extract and Myricetin. In Proceedings of the InHeNce 2021—2021 IEEE International Conference on Health, Instrumentation and Measurement, and Natural Sciences, Medan, Indonesia, 14–16 July 2021; pp. 1–5. [Google Scholar]
  141. Dou, X.; Zhou, Z.; Ren, R.; Xu, M. Apigenin, Flavonoid Component Isolated from Gentiana Veitchiorum Flower Suppresses the Oxidative Stress through LDLR-LCAT Signaling Pathway. Biomed. Pharmacother. 2020, 128, 110298. [Google Scholar] [CrossRef]
  142. Akullo, J.O.; Kiage-Mokua, B.N.; Nakimbugwe, D.; Ng’ang’a, J.; Kinyuru, J. Phytochemical Profile and Antioxidant Activity of Various Solvent Extracts of Two Varieties of Ginger and Garlic. Heliyon 2023, 9, e18806. [Google Scholar] [CrossRef]
  143. Antasionasti, I.; Datu, O.S.; Lestari, U.S.; Abdullah, S.S.; Jayanto, I. Correlation Analysis of Antioxidant Activities with Tannin, Total Flavonoid, and Total Phenolic Contents of Nutmeg (Myristica Fragrans Houtt) Fruit Precipitated by Egg White. Borneo J. Pharm. 2021, 4, 301–310. [Google Scholar] [CrossRef]
  144. Saleh, M.M.; Darwish, Z.E.; El Nouaem, M.I.; Fayed, N.A.; Mourad, G.M.; Ramadan, O.R. The Potential Preventive Effect of Dietary Phytochemicals In Vivo. BDJ Open 2023, 9, 30. [Google Scholar] [CrossRef] [PubMed]
  145. Molina-Montes, E.; Salamanca-Fernández, E.; Garcia-Villanova, B.; Sánchez, M.J. The Impact of Plant-based Dietary Patterns on Cancer-related Outcomes: A Rapid Review and Meta-analysis. Nutrients 2020, 12, 2010. [Google Scholar] [CrossRef]
  146. Chen, H.; Zhu, B.; Zhao, L.; Liu, Y.; Zhao, F.; Feng, J.; Jin, Y.; Sun, J.; Geng, R.; Wei, Y. Allicin Inhibits Proliferation and Invasion in Vitro and in Vivo via SHP-1-Mediated STAT3 Signaling in Cholangiocarcinoma. Cell. Physiol. Biochem. 2018, 47, 641–653. [Google Scholar] [CrossRef]
  147. Kim, J.S.; Jeong, S.K.; Oh, S.J.; Lee, C.G.; Kang, Y.R.; Jo, W.S.; Jeong, M.H. The Resveratrol Analogue, HS-1793, Enhances the Effects of Radiation Therapy through the Induction of Anti-Tumor Immunity in Mammary Tumor Growth. Int. J. Oncol. 2020, 56, 1405–1416. [Google Scholar] [CrossRef]
  148. Bone, S.M.K. (Ed.) Principles of Herbal Pharmacology. In Principles and Practice of Phytotherapy; Elsevier: Amsterdam, The Netherlands, 2013; pp. 17–82. ISBN 9780443069925. [Google Scholar]
  149. Aras, D.; Cinar, O.; Cakar, Z.; Ozkavukcu, S.; Can, A. Can Dicoumarol Be Used as a Gonad-Safe Anticancer Agent: An in Vitro and in Vivo Experimental Study. Mol. Hum. Reprod. 2015, 22, 57–67. [Google Scholar] [CrossRef]
  150. Chen, Y.C.; Chia, Y.C.; Huang, B.M. Phytochemicals from Polyalthia Species: Potential and Implication on Anti-Oxidant, Anti-Inflammatory, Anti-Cancer, and Chemoprevention Activities. Molecules 2021, 26, 5369. [Google Scholar] [CrossRef]
  151. Lam, M.; Carmichael, A.R.; Griffiths, H.R. An Aqueous Extract of Fagonia Cretica Induces DNA Damage, Cell Cycle Arrest and Apoptosis in Breast Cancer Cells via FOXO3a and P53 Expression. PLoS ONE 2012, 7, e102655. [Google Scholar] [CrossRef]
  152. Yeniçeri, E.; Altay, A.; Koksal, E.; Altın, S.; Taslimi, P.; Yılmaz, M.A.; Cakir, O.; Tarhan, A.; Kandemir, A. Phytochemical Profile by LC-MS/MS Analysis and Evaluation of Antioxidant, Antidiabetic, Anti-Alzheimer, and Anticancer Activity of Onobrychis Argyrea Leaf Extracts. Eur. J. Integr. Med. 2024, 66, 102337. [Google Scholar] [CrossRef]
  153. Ooi, K.L.; Muhammad, T.S.T.; Tan, M.L.; Sulaiman, S.F. Cytotoxic, Apoptotic and Anti-α-Glucosidase Activities of 3,4-Di-O-Caffeoyl Quinic Acid, an Antioxidant Isolated from the Polyphenolic-Rich Extract of Elephantopus Mollis Kunth. J. Ethnopharmacol. 2011, 135, 685–695. [Google Scholar] [CrossRef]
  154. Amamra, S.; Cartea, M.E.; Belhaddad, O.E.; Soengas, P.; Baghiani, A.; Kaabi, I.; Arrar, L. Determination of Total Phenolics Contents, Antioxidant Capacity of Thymus Vulgaris Extracts Using Electrochemical and Spectrophotometric Methods. Int. J. Electrochem. Sci. 2018, 13, 7882–7893. [Google Scholar] [CrossRef]
  155. Magdy, M.; Elosaily, A.H.; Mohsen, E.; EL Hefnawy, H.M. Chemical Profile, Antioxidant and Anti-Alzheimer Activity of Leaves and Flowers of Markhamia Lutea Cultivated in Egypt: In Vitro and in Silico Studies. Futur. J. Pharm. Sci. 2024, 10, 103. [Google Scholar] [CrossRef]
  156. Abdelouhab, K.; Guemmaz, T.; Karamać, M.; Kati, D.E.; Amarowicz, R.; Arrar, L. Phenolic Composition and Correlation with Antioxidant Properties of Various Organic Fractions from Hertia Cheirifolia Extracts. J. Pharm. Biomed. Anal. 2023, 235, 115673. [Google Scholar] [CrossRef] [PubMed]
  157. Maurya, A.K.; Vinayak, M. Anticarcinogenic Action of Quercetin by Downregulation of Phosphatidylinositol 3-Kinase (PI3K) and Protein Kinase C (PKC) via Induction of P53 in Hepatocellular Carcinoma (HepG2) Cell Line. Mol. Biol. Rep. 2015, 42, 1419–1429. [Google Scholar] [CrossRef]
  158. Reimche, I.; Yu, H.; Ariantari, N.P.; Liu, Z.; Merkens, K.; Rotfuß, S.; Peter, K.; Jungwirth, U.; Bauer, N.; Kiefer, F.; et al. Phenanthroindolizidine Alkaloids Isolated from Tylophora Ovata as Potent Inhibitors of Inflammation, Spheroid Growth, and Invasion of Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2022, 23, 10319. [Google Scholar] [CrossRef]
  159. Arbizu-Berrocal, S.H.; Kim, H.; Fang, C.; Krenek, K.A.; Talcott, S.T.; Mertens-Talcott, S.U. Polyphenols from Mango (Mangifera indica L.) Modulate PI3K/AKT/MTOR-Associated Micro-RNAs and Reduce Inflammation in Non-Cancer and Induce Cell Death in Breast Cancer Cells. J. Funct. Foods 2019, 55, 9–16. [Google Scholar] [CrossRef]
  160. Rattanamaneerusmee, A.; Thirapanmethee, K.; Nakamura, Y.; Bongcheewin, B.; Chomnawang, M.T. Chemopreventive and Biological Activities of Helicteres Isora L. Fruit Extracts. Res. Pharm. Sci. 2018, 13, 476–483. [Google Scholar] [CrossRef]
  161. Monteillier, A.; Cretton, S.; Ciclet, O.; Marcourt, L.; Ebrahimi, S.N.; Christen, P.; Cuendet, M. Cancer Chemopreventive Activity of Compounds Isolated from Waltheria Indica. J. Ethnopharmacol. 2017, 203, 214–225. [Google Scholar] [CrossRef]
  162. Da Silva, V.C.; Guerra, G.C.B.; Araújo, D.F.D.S.; De Araújo, E.R.; De Araújo, A.A.; Dantas-Medeiros, R.; Zanatta, A.C.; Da Silva, I.L.G.; De Araújo Júnior, R.F.; Esposito, D.; et al. Chemopreventive and Immunomodulatory Effects of Phenolic-Rich Extract of Commiphora Leptophloeos against Inflammatory Bowel Disease: Preclinical Evidence. J. Ethnopharmacol. 2024, 328, 118025. [Google Scholar] [CrossRef]
  163. Drif, A.I.; Yücer, R.; Damiescu, R.; Ali, N.T.; Abu Hagar, T.H.; Avula, B.; Khan, I.A.; Efferth, T. Anti-Inflammatory and Cancer-Preventive Potential of Chamomile (Matricaria chamomilla L.): A Comprehensive In Silico and In Vitro Study. Biomedicines 2024, 12, 1484. [Google Scholar] [CrossRef]
  164. Sobhy, Y.S.; Abo-zeid, Y.S.; Mahgoub, S.S.; Mina, S.A.; Mady, M.S. In-vitro Cytotoxic and Anti-inflammatory Potential of Asparagus Densiflorus Meyeri and Its Phytochemical Investigation. Chem. Biodivers. 2024, 21, e202400959. [Google Scholar] [CrossRef] [PubMed]
  165. Thamer, F.H.; Al-opari, A.M.; Al-Gani, A.M.S.; Al-jaberi, E.A.; Allugam, F.A.; Almahboub, H.H.; Mosik, H.M.; Khalil, H.H.; Abduljalil, M.M.; Alpogosh, M.Y.; et al. Capparis Cartilaginea Decne. as a Natural Source of Antioxidant, Anti-Inflammatory, and Anti-Cancer Herbal Drug. Phytomedicine Plus 2024, 4, 100502. [Google Scholar] [CrossRef]
  166. Dania, O.E.; Dokunmu, T.M.; Adegboye, B.E.; Adeyemi, A.O.; Chibuzor, F.C.; Iweala, E.E.J. Pro-Estrogenic and Anti-Inflammatory Effects of Corchorus Olitorius and Amaranthus Hybridus Leaves in DMBA-Induced Breast Cancer. Phytomedicine Plus 2024, 4, 100567. [Google Scholar] [CrossRef]
  167. Sharma, N.; Samarakoon, K.W.; Gyawali, R.; Park, Y.H.; Lee, S.J.; Oh, S.J.; Lee, T.H.; Jeong, D.K. Evaluation of the Antioxidant, Anti-Inflammatory, and Anticancer Activities of Euphorbia Hirta Ethanolic Extract. Molecules 2014, 19, 14567–14581. [Google Scholar] [CrossRef]
  168. Imtiaz, F.; Islam, M.; Saeed, H.; Ishaq, M.; Shareef, U.; Qaisar, M.N.; Ullah, K.; Mansoor Rana, S.; Yasmeen, A.; Saleem, A. HPLC Profiling for the Simultaneous Estimation of Antidiabetic Compounds from Tradescantia Pallida. Arab. J. Chem. 2024, 17, 105703. [Google Scholar] [CrossRef]
  169. Latolla, N.; Reddy, S.; van de Venter, M.; Hlangothi, B. Phytochemical Composition and Antidiabetic Potential of the Leaf, Stem, and Rhizome Extracts of Cissampelos Capensis L.F. S. Afr. J. Bot. 2023, 163, 468–477. [Google Scholar] [CrossRef]
  170. Sharma, P.; Joshi, T.; Joshi, T.; Chandra, S.; Tamta, S. In Silico Screening of Potential Antidiabetic Phytochemicals from Phyllanthus Emblica against Therapeutic Targets of Type 2 Diabetes. J. Ethnopharmacol. 2020, 248, 112268. [Google Scholar] [CrossRef]
  171. Hannan, J.M.A.; Marenah, L.; Ali, L.; Rokeya, B.; Flatt, P.R.; Abdel-Wahab, Y.H.A. Ocimum Sanctum Leaf Extracts Stimulate Insulin Secretion from Perfused Pancreas, Isolated Islets and Clonal Pancreatic β-Cells. J. Endocrinol. 2006, 189, 127–136. [Google Scholar] [CrossRef]
  172. 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 2012, 28, 42–50. [Google Scholar] [CrossRef]
  173. Rahman, R.A.; Chellammal, H.S.J.; Shah, S.A.A.; Zohdi, R.M.; Ramachandran, D.; Mohsin, H.F. Exploring the Therapeutic Potential of Derris elliptica (Wall.) Benth in Streptozotocin-Induced Diabetic Rats: Phytochemical Characterization and Antidiabetic Evaluation: Exploring the Therapeutic Potential of Derris elliptica (Wall.). Saudi Pharm. J. 2024, 32, 102016. [Google Scholar] [CrossRef]
  174. Agada, R.; Thagriki, D.; Lydia, D.E.; Khusro, A.; Alkahtani, J.; Al Shaqha, M.M.; Alwahibi, M.S.; Elshikh, M.S. Antioxidant and Anti-Diabetic Activities of Bioactive Fractions of Carica Papaya Seeds Extract. J. King Saud Univ.-Sci. 2021, 33, 101342. [Google Scholar] [CrossRef]
  175. Mahmood, R.; Kayani, W.K.; Ahmed, T.; Malik, F.; Hussain, S.; Ashfaq, M.; Ali, H.; Rubnawaz, S.; Green, B.D.; Calderwood, D.; et al. Assessment of Antidiabetic Potential and Phytochemical Profiling of Rhazya Stricta Root Extracts. BMC Complement. Med. Ther. 2020, 20, 293. [Google Scholar] [CrossRef] [PubMed]
  176. Rizvi, S.N.R.; Afzal, S.; Khan, K.U.R.; Aati, H.Y.; Rao, H.; Ghalloo, B.A.; Shahzad, M.N.; Khan, D.A.; Esatbeyoglu, T.; Korma, S.A. Chemical Characterisation, Antidiabetic, Antibacterial, and In Silico Studies for Different Extracts of Haloxylon stocksii (Boiss.) Benth: A Promising Halophyte. Molecules 2023, 28, 3847. [Google Scholar] [CrossRef]
  177. Ye, Y.; Kawaguchi, Y.; Takeuchi, A.; Zhang, N.; Mori, R.; Mijiti, M.; Banno, A.; Okada, T.; Hiramatsu, N.; Nagaoka, S. Rose Polyphenols Exert Antiobesity Effect in High-Fat–Induced Obese Mice by Regulating Lipogenic Gene Expression. Nutr. Res. 2023, 119, 76–89. [Google Scholar] [CrossRef]
  178. Fang, J.Y.; Huang, T.H.; Chen, W.J.; Aljuffali, I.A.; Hsu, C.Y. Rhubarb Hydroxyanthraquinones Act as Antiobesity Agents to Inhibit Adipogenesis and Enhance Lipolysis. Biomed. Pharmacother. 2022, 146, 112497. [Google Scholar] [CrossRef]
  179. Im, J.H.; Lim, J.s.; Han, X.; Men, X.; Oh, G.; Fu, X.; Cho, G.h.; Hwang, W.s.; Choi, S.I.; Lee, O.H. Anti-Obesogenic Effect of Standardized Brassica Juncea Extract on Bisphenol A-Induced 3T3-L1 Preadipocytes and C57BL/6J Obese Mice. Food Sci. Biotechnol. 2024, 16, 13. [Google Scholar] [CrossRef]
  180. Magwaza, S.N.; Erukainure, O.L.; Olofinsan, K.; Meriga, B.; Islam, M.S. Evaluation of the Antidiabetic, Antiobesity and Antioxidant Potential of Anthophycus Longifolius ((Turner) Kützing). Sci. African 2024, 23, e02051. [Google Scholar] [CrossRef]
  181. Brattiya, K.; Jagdish, R.; Ahamed, A.; Anuradha, C.R.; Jeyasudha, J. Anti-Obesity and Anticancer Activity of Solanum Xanthocarpum Leaf Extract: An in Vitro Study. Natl. J. Physiol. Pharm. Pharmacol. 2023, 13, 1. [Google Scholar] [CrossRef]
  182. Jaradat, N.; Hawash, M. Anti-Obesity Activities of Rumex Rothschildianus Aarons. Extracts. BMC Complement. Med. Ther. 2021, 21, 107. [Google Scholar] [CrossRef]
  183. Peng, X.N.; Zhou, Y.; Liu, Y.X.; Huo, Y.L.; Ren, J.Y.; Bai, Z.Z.; Zhang, Y.L.; Tang, J.J. Neuroprotective Potential of Phytochemicals Isolated from Paeonia Ostii ‘Feng Dan’ Stamen. Ind. Crops Prod. 2023, 200, 116808. [Google Scholar] [CrossRef]
  184. Gouda, B.; Sinha, S.N.; Sangaraju, R.; Huynh, T.; Patangay, S.; Mullapudi, S.V.; Mungamuri, S.K.; Patil, P.B.; Periketi, M.C. Extraction, Phytochemical Profile, and Neuroprotective Activity of Phyllanthus Emblica Fruit Extract against Sodium Valproate-Induced Postnatal Autism in BALB/c Mice. Heliyon 2024, 10, e34992. [Google Scholar] [CrossRef] [PubMed]
  185. Khaled, N.; Ibrahim, N.; Ali, A.E.; Youssef, F.S.; El-Ahmady, S.H. LC-QTOF-MS/MS Phytochemical Profiling of Tabebuia Impetiginosa (Mart. Ex DC.) Standl. Leaf and Assessment of Its Neuroprotective Potential in Rats. J. Ethnopharmacol. 2024, 331, 118292. [Google Scholar] [CrossRef]
  186. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  187. Almilaibary, A. Phyto-Therapeutics as Anti-Cancer Agents in Breast Cancer: Pathway Targeting and Mechanistic Elucidation. Saudi J. Biol. Sci. 2024, 31, 103935. [Google Scholar] [CrossRef]
  188. Yun, C.W.; Jeon, J.; Go, G.; Lee, J.H.; Lee, S.H. The Dual Role of Autophagy in Cancer Development and a Therapeutic Strategy for Cancer by Targeting Autophagy. Int. J. Mol. Sci. 2021, 22, 179. [Google Scholar] [CrossRef]
  189. Koh, Y.-C.; Ho, C.-T.; Pan, M.-H. Recent Advances in Cancer Chemoprevention with Phytochemicals. J. Food Drug Anal. 2020, 28, 14–37. [Google Scholar] [CrossRef] [PubMed]
  190. Bastos, R.G.; de, O. Rodrigues, S.; Marques, L.A.; Oliveira, C.M. d.; Salles, B.C.C.; Zanatta, A.C.; Rocha, F.D.; Vilegas, W.; Pagnossa, J.P.; Fernanda, F.B.; et al. Eugenia Sonderiana O. Berg Leaves: Phytochemical Characterization, Evaluation of in Vitro and in Vivo Antidiabetic Effects, and Structure-Activity Correlation. Biomed. Pharmacother. 2023, 165, 115126. [Google Scholar] [CrossRef]
  191. Kanmaz, H.; Gokce, Y.; Hayaloglu, A.A. Volatiles, Phenolic Compounds and Bioactive Properties of Essential Oil and Aqueous Extract of Purple Basil (Ocimum basilicum L.) and Antidiabetic Activity in Streptozotocin-Induced Diabetic Wistar Rats. Food Chem. Adv. 2023, 3, 100429. [Google Scholar] [CrossRef]
  192. Tung, Y.C.; Hsieh, P.H.; Pan, M.H.; Ho, C.T. Cellular Models for the Evaluation of the Antiobesity Effect of Selected Phytochemicals from Food and Herbs. J. Food Drug Anal. 2017, 25, 100–110. [Google Scholar] [CrossRef]
  193. Wolfram, S.; Wang, Y.; Thielecke, F. Anti-Obesity Effects of Green Tea: From Bedside to Bench. Mol. Nutr. Food Res. 2006, 50, 176–187. [Google Scholar] [CrossRef]
  194. Suescun, J.; Chandra, S.; Schiess, M.C. The Role of Neuroinflammation in Neurodegenerative Disorders. In Translational Cell Biology; Elsevier: Amsterdam, The Netherlands, 2019; pp. 241–267. [Google Scholar] [CrossRef]
  195. Ayaz, M.; Mosa, O.F.; Nawaz, A.; Hamdoon, A.A.E.; Elkhalifa, M.E.M.; Sadiq, A.; Ullah, F.; Ahmed, A.; Kabra, A.; Khan, H.; et al. Neuroprotective Potentials of Lead Phytochemicals against Alzheimer’s Disease with Focus on Oxidative Stress-Mediated Signaling Pathways: Pharmacokinetic Challenges, Target Specificity, Clinical Trials and Future Perspectives. Phytomedicine 2024, 124, 155272. [Google Scholar] [CrossRef] [PubMed]
  196. Gil-Martín, E.; Forbes-Hernández, T.; Romero, A.; Cianciosi, D.; Giampieri, F.; Battino, M. Influence of the Extraction Method on the Recovery of Bioactive Phenolic Compounds from Food Industry By-Products. Food Chem. 2022, 378, 131918. [Google Scholar] [CrossRef] [PubMed]
  197. Aqil, F.; Munagala, R.; Jeyabalan, J.; Vadhanam, M.V. Bioavailability of Phytochemicals and Its Enhancement by Drug Delivery Systems. Cancer Lett. 2013, 334, 133–141. [Google Scholar] [CrossRef]
  198. Siddiqui, R.A.; Moghadasian, M.H. Nutraceuticals and Nutrition Supplements: Challenges and Opportunities. Nutrients 2020, 12, 1593. [Google Scholar] [CrossRef] [PubMed]
  199. Xu, J. Harnessing the Power of Plants: A Green Factory for Bioactive Compounds. Life 2023, 13, 2041. [Google Scholar] [CrossRef]
  200. Kumar, A.; Nirmal, P.; Kumar, M.; Jose, A.; Tomer, V.; Oz, E.; Proestos, C.; Zeng, M.; Elobeid, T.; Sneha, V.; et al. Major Phytochemicals: Recent Advances in Health Benefits and Extraction Method. Molecules 2023, 28, 887. [Google Scholar] [CrossRef]
  201. El Maaiden, E.; Bouzroud, S.; Nasser, B.; Moustaid, K.; El Mouttaqi, A.; Ibourki, M.; Boukcim, H.; Hirich, A.; Kouisni, L.; El Kharrassi, Y. A Comparative Study between Conventional and Advanced Extraction Techniques: Pharmaceutical and Cosmetic Properties of Plant Extracts. Molecules 2022, 27, 2074. [Google Scholar] [CrossRef]
  202. Cannavacciuolo, C.; Pagliari, S.; Celano, R.; Campone, L.; Rastrelli, L. Critical Analysis of Green Extraction Techniques Used for Botanicals: Trends, Priorities, and Optimization Strategies-A Review. TrAC-Trends Anal. Chem. 2024, 173, 117627. [Google Scholar] [CrossRef]
  203. Domínguez, R.; Pateiro, M.; Munekata, P.E.S.; McClements, D.J.; Lorenzo, J.M. Encapsulation of Bioactive Phytochemicals in Plant-Based Matrices and Application as Additives in Meat and Meat Products. Molecules 2021, 26, 3984. [Google Scholar] [CrossRef]
  204. Bommakanti, V.; Ajikumar, A.P.; Sivi, C.M.; Prakash, G.; Mundanat, A.S.; Ahmad, F.; Haque, S.; Prieto, M.A.; Rana, S.S. An Overview of Herbal Nutraceuticals, Their Extraction, Formulation, Therapeutic Effects and Potential Toxicity. Separations 2023, 10, 177. [Google Scholar] [CrossRef]
  205. Belwal, T.; Chemat, F.; Venskutonis, P.R.; Cravotto, G.; Jaiswal, D.K.; Bhatt, I.D.; Devkota, H.P.; Luo, Z. Recent Advances in Scaling-up of Non-Conventional Extraction Techniques: Learning from Successes and Failures. TrAC-Trends Anal. Chem. 2020, 127, 115895. [Google Scholar] [CrossRef]
  206. Pateiro, M.; Barba, F.J.; Domínguez, R.; Sant’Ana, A.S.; Khaneghah, A.M.; Gavahian, M.; Gómez, B.; Lorenzo, J.M. Essential Oils as Natural Additives to Prevent Oxidation Reactions in Meat and Meat Products: A Review. Food Res. Int. 2018, 113, 156–166. [Google Scholar] [CrossRef] [PubMed]
  207. Bitwell, C.; Indra, S.S.; Luke, C.; Kakoma, M.K. A Review of Modern and Conventional Extraction Techniques and Their Applications for Extracting Phytochemicals from Plants. Sci. Afr. 2023, 19, e01585. [Google Scholar] [CrossRef]
  208. Nicolescu, A.; Babotă, M.; Barros, L.; Rocchetti, G.; Lucini, L.; Tanase, C.; Mocan, A.; Bunea, C.I.; Crișan, G. Bioaccessibility and Bioactive Potential of Different Phytochemical Classes from Nutraceuticals and Functional Foods. Front. Nutr. 2023, 10, 1184535. [Google Scholar] [CrossRef] [PubMed]
  209. Altemimi, A.; Lightfoot, D.A.; Kinsel, M.; Watson, D.G. Employing Response Surface Methodology for the Optimization of Ultrasound Assisted Extraction of Lutein and β-Carotene from Spinach. Molecules 2015, 20, 6611–6625. [Google Scholar] [CrossRef]
  210. Altemimi, A.; Lakhssassi, N.; Baharlouei, A.; Watson, D.G.; Lightfoot, D.A. Phytochemicals: Extraction, Isolation, and Identification of Bioactive Compounds from Plant Extracts. Plants 2017, 6, 42. [Google Scholar] [CrossRef]
  211. Zhang, Z.; Pang, X.; Xuewu, D.; Ji, Z.; Jiang, Y. Role of Peroxidase in Anthocyanin Degradation in Litchi Fruit Pericarp. Food Chem. 2005, 90, 47–52. [Google Scholar] [CrossRef]
  212. Mulinacci, N.; Prucher, D.; Peruzzi, M.; Romani, A.; Pinelli, P.; Giaccherini, C.; Vincieri, F.F. Commercial and Laboratory Extracts from Artichoke Leaves: Estimation of Caffeoyl Esters and Flavonoidic Compounds Content. J. Pharm. Biomed. Anal. 2004, 34, 349–357. [Google Scholar] [CrossRef]
  213. Manocha, S.; Dhiman, S.; Grewal, A.S.; Guarve, K. Nanotechnology: An Approach to Overcome Bioavailability Challenges of Nutraceuticals. J. Drug Deliv. Sci. Technol. 2022, 72, 103418. [Google Scholar] [CrossRef]
  214. Zheng, B.; McClements, D.J. Formulation of More Efficacious Curcumin Delivery Systems Using Colloid Science: Enhanced Solubility, Stability, and Bioavailability. Molecules 2020, 25, 2791. [Google Scholar] [CrossRef]
  215. Agriopoulou, S.; Tarapoulouzi, M.; Varzakas, T.; Jafari, S.M. Application of Encapsulation Strategies for Probiotics: From Individual Loading to Co-Encapsulation. Microorganisms 2023, 11, 2896. [Google Scholar] [CrossRef] [PubMed]
  216. Kato, L.S.; Lelis, C.A.; da Silva, B.D.; Galvan, D.; Conte-Junior, C.A. Micro- and Nanoencapsulation of Natural Phytochemicals: Challenges and Recent Perspectives for the Food and Nutraceuticals Industry Applications. In Advances in Food and Nutrition Research; Toldrá, F., Ed.; Academic Press: Cambridge, MA, USA, 2023; Volume 104, pp. 77–137. ISBN 9780443193026. [Google Scholar]
  217. Alu’datt, M.H.; Alrosan, M.; Gammoh, S.; Tranchant, C.C.; Alhamad, M.N.; Rababah, T.; Zghoul, R.; Alzoubi, H.; Ghatasheh, S.; Ghozlan, K.; et al. Encapsulation-Based Technologies for Bioactive Compounds and Their Application in the Food Industry: A Roadmap for Food-Derived Functional and Health-Promoting Ingredients. Food Biosci. 2022, 50, 101971. [Google Scholar] [CrossRef]
  218. Choudhury, N.; Meghwal, M.; Das, K. Microencapsulation: An Overview on Concepts, Methods, Properties and Applications in Foods. Food Front. 2021, 2, 426–442. [Google Scholar] [CrossRef]
  219. Ozkan, G.; Franco, P.; De Marco, I.; Xiao, J.; Capanoglu, E. A Review of Microencapsulation Methods for Food Antioxidants: Principles, Advantages, Drawbacks and Applications. Food Chem. 2019, 272, 494–506. [Google Scholar] [CrossRef]
  220. Pateiro, M.; Munekata, P.E.S.; Barba, F.J.; Putnik, P. Nanoencapsulation of Promising Bioactive Compounds to Improve Their Absorption, Stability, Functionality and the Appearance of the Final Food Products. Molecules 2021, 26, 1547. [Google Scholar] [CrossRef]
  221. Altemimi, A.B.; Farag, H.A.M.; Salih, T.H.; Awlqadr, F.H.; Al-Manhel, A.J.A.; Vieira, I.R.S.; Conte-Junior, C.A. Application of Nanoparticles in Human Nutrition: A Review. Nutrients 2024, 16, 636. [Google Scholar] [CrossRef]
  222. Singh, A.K.; Pal, P.; Pandey, B.; Goksen, G.; Sahoo, U.K.; Lorenzo, J.M.; Sarangi, P.K. Development of “Smart Foods” for Health by Nanoencapsulation: Novel Technologies and Challenges. Food Chem. X 2023, 20, 100910. [Google Scholar] [CrossRef]
  223. Puttasiddaiah, R.; Lakshminarayana, R.; Somashekar, N.L.; Gupta, V.K.; Inbaraj, B.S.; Usmani, Z.; Raghavendra, V.B.; Sridhar, K.; Sharma, M.; Rennes-angers, L.I.A.; et al. Advances in Nanofabrication Technology for Nutraceuticals: New Insights and Future Trends. Bioengineered 2022, 9, 478. [Google Scholar] [CrossRef]
  224. Hu, Y.; Lin, Q.; Zhao, H.; Li, X.; Sang, S.; McClements, D.J.; Long, J.; Jin, Z.; Wang, J.; Qiu, C. Bioaccessibility and Bioavailability of Phytochemicals: Influencing Factors, Improvements, and Evaluations. Food Hydrocoll. 2023, 135, 108165. [Google Scholar] [CrossRef]
  225. Holst, B.; Williamson, G. Nutrients and Phytochemicals: From Bioavailability to Bioefficacy beyond Antioxidants. Curr. Opin. Biotechnol. 2008, 19, 73–82. [Google Scholar] [CrossRef]
  226. Epriliati, I.; Ginjom, I.R. Bioavailability of Phytochemicals. In Phytochemicals-A Global Perspective of Their Role in Nutrition and Health; Rao, V., Ed.; IntechOpen: Rijeka, Croatia, 2012. [Google Scholar]
  227. Shahidi, F.; Pan, Y. Influence of Food Matrix and Food Processing on the Chemical Interaction and Bioaccessibility of Dietary Phytochemicals: A Review. Crit. Rev. Food Sci. Nutr. 2022, 62, 6421–6445. [Google Scholar] [CrossRef] [PubMed]
  228. Selby-Pham, S.N.B.; Miller, R.B.; Howell, K.; Dunshea, F.; Bennett, L.E. Physicochemical Properties of Dietary Phytochemicals Can Predict Their Passive Absorption in the Human Small Intestine. Sci. Rep. 2017, 7, 1931. [Google Scholar] [CrossRef] [PubMed]
  229. Polia, F.; Pastor-Belda, M.; Martínez-Blázquez, A.; Horcajada, M.N.; Tomás-Barberán, F.A.; García-Villalba, R. Technological and Biotechnological Processes To Enhance the Bioavailability of Dietary (Poly)Phenols in Humans. J. Agric. Food Chem. 2022, 70, 2092–2107. [Google Scholar] [CrossRef] [PubMed]
  230. Helal, N.A.; Eassa, H.A.; Amer, A.M.; Eltokhy, M.A.; Edafiogho, I.; Nounou, M.I. Nutraceuticals’ Novel Formulations: The Good, the Bad, the Unknown and Patents Involved. Recent Pat. Drug Deliv. Formul. 2019, 13, 105–156. [Google Scholar] [CrossRef]
  231. Senanayake, S.P.J.N. Green Tea Extract: Chemistry, Antioxidant Properties and Food Applications—A Review. J. Funct. Foods 2013, 5, 1529–1541. [Google Scholar] [CrossRef]
  232. Perumalla, A.V.S.; Hettiarachchy, N.S. Green Tea and Grape Seed Extracts—Potential Applications in Food Safety and Quality. Food Res. Int. 2011, 44, 827–839. [Google Scholar] [CrossRef]
  233. Bora, A.F.M.; Ma, S.; Li, X.; Liu, L. Application of Microencapsulation for the Safe Delivery of Green Tea Polyphenols in Food Systems: Review and Recent Advances. Food Res. Int. 2018, 105, 241–249. [Google Scholar] [CrossRef]
  234. Nieto, G.; Martínez-Zamora, L.; Peñalver, R.; Marín-Iniesta, F.; Taboada-Rodríguez, A.; López-Gómez, A.; Martínez-Hernández, G.B. Applications of Plant Bioactive Compounds as Replacers of Synthetic Additives in the Food Industry. Foods 2023, 13, 47. [Google Scholar] [CrossRef]
  235. Duizer, L.M.; West, R.; Campanella, O.H. Fiber Addition to Cereal Based Foods: Effects on Sensory Properties. In Food Engineering Series; Springer: Cham, Switzerland, 2020; pp. 419–435. [Google Scholar]
  236. Muñoz-Almagro, N.; Garrido-Galand, S.; Taladrid, D.; Moreno-Arribas, M.V.; Villamiel, M.; Montilla, A. Use of Natural Low-Methoxyl Pectin from Sunflower by-Products for the Formulation of Low-Sucrose Strawberry Jams. J. Sci. Food Agric. 2022, 102, 5957–5964. [Google Scholar] [CrossRef]
  237. Tiwari, A.; Sharma, H.K.; Kumar, N.; Kaur, M. The Effect of Inulin as a Fat Replacer on the Quality of Low-Fat Ice Cream. Int. J. Dairy Technol. 2015, 68, 374–380. [Google Scholar] [CrossRef]
  238. Le Bastard, Q.; Chapelet, G.; Javaudin, F.; Lepelletier, D.; Batard, E.; Montassier, E. The Effects of Inulin on Gut Microbial Composition: A Systematic Review of Evidence from Human Studies. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 403–413. [Google Scholar] [CrossRef] [PubMed]
  239. Nsor-Atindana, J.; Chen, M.; Goff, H.D.; Zhong, F.; Sharif, H.R.; Li, Y. Functionality and Nutritional Aspects of Microcrystalline Cellulose in Food. Carbohydr. Polym. 2017, 172, 159–174. [Google Scholar] [CrossRef] [PubMed]
  240. Nath, P.C.; Debnath, S.; Sharma, M.; Sridhar, K.; Nayak, P.K.; Inbaraj, B.S. Recent Advances in Cellulose-Based Hydrogels: Food Applications. Foods 2023, 12, 350. [Google Scholar] [CrossRef]
  241. Onipe, O.O.; Ramashia, S.E.; Jideani, A.I.O. Wheat Bran Modifications for Enhanced Nutrition and Functionality in Selected Food Products. Molecules 2021, 26, 3918. [Google Scholar] [CrossRef]
  242. Maiani, G.; Castón, M.J.P.; Catasta, G.; Toti, E.; Cambrodón, I.G.; Bysted, A.; Granado-Lorencio, F.; Olmedilla-Alonso, B.; Knuthsen, P.; Valoti, M.; et al. Carotenoids: Actual Knowledge on Food Sources, Intakes, Stability and Bioavailability and Their Protective Role in Humans. Mol. Nutr. Food Res. 2009, 53, 194–218. [Google Scholar] [CrossRef]
  243. Wang, Y.; McClements, D.J.; Chen, L.; Peng, X.; Xu, Z.; Meng, M.; Ji, H.; Zhi, C.; Ye, L.; Zhao, J.; et al. Progress on Molecular Modification and Functional Applications of Anthocyanins. Crit. Rev. Food Sci. Nutr. 2023, 64, 11409–11427. [Google Scholar] [CrossRef]
  244. Pereira, A.G.; Fraga, M.; Oliveira, P.G.; Jimenez-Lopez, C.; Lourenço-Lopes, C.; Barros, L.; Ferreira, I.C.F.R.; Prieto, M.A.; Simal-Gandara, J. Identification, Quantification, and Method Validation of Anthocyanins. In Anthocyanins: Antioxidant Properties, Sources and Health Benefits; Nova Science Publishers: Hauppauge, NY, USA, 2020; pp. 51–83. ISBN 9781536178173. [Google Scholar]
  245. Jokioja, J.; Yang, B.; Linderborg, K.M. Acylated Anthocyanins: A Review on Their Bioavailability and Effects on Postprandial Carbohydrate Metabolism and Inflammation. Compr. Rev. Food Sci. Food Saf. 2021, 20, 5570–5615. [Google Scholar] [CrossRef]
  246. Mattioli, R.; Francioso, A.; Mosca, L.; Silva, P. Anthocyanins: A Comprehensive Review of Their Chemical Properties and Health Effects on Cardiovascular and Neurodegenerative Diseases. Molecules 2020, 25, 3809. [Google Scholar] [CrossRef]
  247. Niu, B.; Shao, P.; Feng, S.; Qiu, D.; Sun, P. Rheological Aspects in Fabricating Pullulan-Whey Protein Isolate Emulsion Suitable for Electrospraying: Application in Improving β-Carotene Stability. LWT 2020, 129, 109581. [Google Scholar] [CrossRef]
  248. Esatbeyoglu, T.; Wagner, A.E.; Schini-Kerth, V.B.; Rimbach, G. Betanin-A Food Colorant with Biological Activity. Mol. Nutr. Food Res. 2015, 59, 36–47. [Google Scholar] [CrossRef]
  249. da Silva, D.V.T.; dos Santos Baião, D.; de Oliveira Silva, F.; Alves, G.; Perrone, D.; Del Aguila, E.M.; Paschoalin, V.M.F. Betanin, a Natural Food Additive: Stability, Bioavailability, Antioxidant and Preservative Ability Assessments. Molecules 2019, 24, 458. [Google Scholar] [CrossRef] [PubMed]
  250. Pagano, A.P.E.; Khalid, N.; Kobayashi, I.; Nakajima, M.; Neves, M.A.; Bastos, E.L. Microencapsulation of Betanin in Monodisperse W/O/W Emulsions. Food Res. Int. 2018, 109, 489–496. [Google Scholar] [CrossRef] [PubMed]
  251. Amjadi, S.; Ghorbani, M.; Hamishehkar, H.; Roufegarinejad, L. Improvement in the Stability of Betanin by Liposomal Nanocarriers: Its Application in Gummy Candy as a Food Model. Food Chem. 2018, 256, 156–162. [Google Scholar] [CrossRef]
  252. Dai, T.; He, X.; Xu, J.; Geng, Q.; Li, C.; Sun, J.; Liu, C.; Chen, J.; He, X. Effects of Betanin on Pasting, Rheology and Retrogradation Properties of Different Starches. Foods 2022, 11, 1600. [Google Scholar] [CrossRef]
  253. Tsuda, T. Curcumin as a Functional Food-Derived Factor: Degradation Products, Metabolites, Bioactivity, and Future Perspectives. Food Funct. 2018, 9, 705–714. [Google Scholar] [CrossRef]
  254. Munekata, P.E.S.; Pateiro, M.; Zhang, W.; Dominguez, R.; Xing, L.; Fierro, E.M.; Lorenzo, J.M. Health Benefits, Extraction and Development of Functional Foods with Curcuminoids. J. Funct. Foods 2021, 79, 104392. [Google Scholar] [CrossRef]
  255. Xu, X.Y.; Meng, X.; Li, S.; Gan, R.Y.; Li, Y.; Li, H.B. Bioactivity, Health Benefits, and Related Molecular Mechanisms of Curcumin: Current Progress, Challenges, and Perspectives. Nutrients 2018, 10, 1553. [Google Scholar] [CrossRef]
  256. Life Extension. Available online: https://www.lifeextensioneurope.com/ (accessed on 6 May 2025).
  257. Toüfood. Available online: https://www.toufood.com/ (accessed on 7 May 2025).
  258. Bluetec Colorants Ltd. Available online: https://www.bestphycocyanin.com/ (accessed on 7 May 2025).
  259. McClements, D.J.; Öztürk, B. Utilization of Nanotechnology to Improve the Application and Bioavailability of Phytochemicals Derived from Waste Streams. J. Agric. Food Chem. 2022, 70, 6884–6900. [Google Scholar] [CrossRef]
  260. Zhang, S.; Waterhouse, G.I.N.; Xu, F.; He, Z.; Du, Y.; Lian, Y.; Wu, P.; Sun-Waterhouse, D. Recent Advances in Utilization of Pectins in Biomedical Applications: A Review Focusing on Molecular Structure-Directing Health-Promoting Properties. Crit. Rev. Food Sci. Nutr. 2023, 63, 3386–3419. [Google Scholar] [CrossRef]
  261. Qin, Y.Q.; Wang, L.Y.; Yang, X.Y.; Xu, Y.J.; Fan, G.; Fan, Y.G.; Ren, J.N.; An, Q.; Li, X. Inulin: Properties and Health Benefits. Food Funct. 2023, 14, 2948–2968. [Google Scholar] [CrossRef]
  262. Deroover, L.; Tie, Y.; Verspreet, J.; Courtin, C.M.; Verbeke, K. Modifying Wheat Bran to Improve Its Health Benefits. Crit. Rev. Food Sci. Nutr. 2020, 60, 1104–1122. [Google Scholar] [CrossRef] [PubMed]
  263. Ziai, S.A.; Larijani, B.; Akhoondzadeh, S.; Fakhrzadeh, H.; Dastpak, A.; Bandarian, F.; Rezai, A.; Badi, H.N.; Emami, T. Psyllium Decreased Serum Glucose and Glycosylated Hemoglobin Significantly in Diabetic Outpatients. J. Ethnopharmacol. 2005, 102, 202–207. [Google Scholar] [CrossRef] [PubMed]
  264. Chen, C.; Shang, C.; Xin, L.; Xiang, M.; Wang, Y.; Shen, Z.; Jiao, L.; Ding, F.; Cui, X. Beneficial Effects of Psyllium on the Prevention and Treatment of Cardiometabolic Diseases. Food Funct. 2022, 13, 7473–7486. [Google Scholar] [CrossRef] [PubMed]
  265. Saeed, M.; Naveed, M.; Arif, M.; Kakar, M.U.; Manzoor, R.; El-Hack, M.E.A.; Alagawany, M.; Tiwari, R.; Khandia, R.; Munjal, A.; et al. Green Tea (Camellia sinensis) and L-Theanine: Medicinal Values and Beneficial Applications in Humans—A Comprehensive Review. Biomed. Pharmacother. 2017, 95, 1260–1275. [Google Scholar] [CrossRef]
  266. Bogdanski, P.; Suliburska, J.; Szulinska, M.; Stepien, M.; Pupek-Musialik, D.; Jablecka, A. Green Tea Extract Reduces Blood Pressure, Inflammatory Biomarkers, and Oxidative Stress and Improves Parameters Associated with Insulin Resistance in Obese, Hypertensive Patients. Nutr. Res. 2012, 32, 421–427. [Google Scholar] [CrossRef]
  267. Gupta, M.; Dey, S.; Marbaniang, D.; Pal, P.; Ray, S.; Mazumder, B. Grape Seed Extract: Having a Potential Health Benefits. J. Food Sci. Technol. 2020, 57, 1205–1215. [Google Scholar] [CrossRef]
  268. Bendich, A. From 1989 to 2001: What Have We Learned about the “Biological Actions of Beta-Carotene”? J. Nutr. 2004, 134, 225S–230S. [Google Scholar] [CrossRef]
  269. Li, D.; Wang, P.; Luo, Y.; Zhao, M.; Chen, F. Health Benefits of Anthocyanins and Molecular Mechanisms: Update from Recent Decade. Crit. Rev. Food Sci. Nutr. 2017, 57, 1729–1741. [Google Scholar] [CrossRef]
  270. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
  271. Barber, T.M.; Kabisch, S.; Randeva, H.S.; Pfeiffer, A.F.H.; Weickert, M.O. Implications of Resveratrol in Obesity and Insulin Resistance: A State-of-the-Art Review. Nutrients 2022, 14, 2870. [Google Scholar] [CrossRef]
  272. Wang, Y.; Xing, M.; Cao, Q.; Ji, A.; Liang, H.; Song, S. Biological Activities of Fucoidan and the Factors Mediating Its Therapeutic Effects: A Review of Recent Studies. Mar. Drugs 2019, 17, 183. [Google Scholar] [CrossRef] [PubMed]
  273. Li, F.; Liu, K. Research Progress in the Preparation, Structural Characterization, Bioactivities, and Potential Applications of Sulfated Agarans from the Genus Gracilaria. J. Food Biochem. 2022, 46, e14401. [Google Scholar] [CrossRef] [PubMed]
  274. du Preez, R.; Paul, N.; Mouatt, P.; Majzoub, M.E.; Thomas, T.; Panchal, S.K.; Brown, L. Carrageenans from the Red Seaweed Sarconema Filiforme Attenuate Symptoms of Diet-Induced Metabolic Syndrome in Rats. Mar. Drugs 2020, 18, 97. [Google Scholar] [CrossRef] [PubMed]
  275. Deepika; Maurya, P.K. Health Benefits of Quercetin in Age-Related Diseases. Molecules 2022, 27, 2498. [Google Scholar] [CrossRef]
  276. Aghababaei, F.; Hadidi, M. Recent Advances in Potential Health Benefits of Quercetin. Pharmaceuticals 2023, 16, 1020. [Google Scholar] [CrossRef]
  277. Naware, N.; Ambatkar, S.; Kamble, T.; Bangar, S.; Uppar, K.; Shirke, K.; Patil, M.; Jain, A. A Review Focusing on the Benefits of Green Tea Catechins as Nutraceuticals. Sci. Phytochem. 2023, 2, 138–146. [Google Scholar] [CrossRef]
  278. Ma, D.S.L.; Tan, L.T.H.; Chan, K.G.; Yap, W.H.; Pusparajah, P.; Chuah, L.H.; Ming, L.C.; Khan, T.M.; Lee, L.H.; Goh, B.H. Resveratrol-Potential Antibacterial Agent against Foodborne Pathogens. Front. Pharmacol. 2018, 9, 102. [Google Scholar] [CrossRef]
  279. Ko, S.K.; Chung, S.J.; Park, Y.H.; Park, A.S.; Kim, H.J.; Park, G.Y.; Oh, Y.H. Determination of Hazardous Metals in Nail Enamel Containing Glitter. Korean J. Environ. Health Sci. 2017, 43, 103–110. [Google Scholar] [CrossRef]
  280. Cohen, S.; Emery, P.; Greenwald, M.; Yin, D.; Becker, J.C.; Melia, L.A.; Li, R.; Gumbiner, B.; Thomas, D.; Spencer-Green, G.; et al. A Phase I Pharmacokinetics Trial Comparing PF-05280586 (a Potential Biosimilar) and Rituximab in Patients with Active Rheumatoid Arthritis. Br. J. Clin. Pharmacol. 2016, 82, 129–138. [Google Scholar] [CrossRef]
  281. Khan, M.I.; Shin, J.H.; Kim, J.D. The Promising Future of Microalgae: Current Status, Challenges, and Optimization of a Sustainable and Renewable Industry for Biofuels, Feed, and Other Products. Microb. Cell Fact. 2018, 17, 36. [Google Scholar] [CrossRef]
  282. Hajipour, H.; Ghorbani, M.; Kahroba, H.; Mahmoodzadeh, F.; Emameh, R.Z.; Taheri, R.A. Arginyl-Glycyl-Aspartic Acid (RGD) Containing Nanostructured Lipid Carrier Co-Loaded with Doxorubicin and Sildenafil Citrate Enhanced Anti-Cancer Effects and Overcomes Drug Resistance. Process Biochem. 2019, 84, 172–179. [Google Scholar] [CrossRef]
  283. Bush, L.; Stevenson, L.; Lane, K.E. The Oxidative Stability of Omega-3 Oil-in-Water Nanoemulsion Systems Suitable for Functional Food Enrichment: A Systematic Review of the Literature. Crit. Rev. Food Sci. Nutr. 2019, 59, 1154–1168. [Google Scholar] [CrossRef]
  284. Kusmayadi, A.; Leong, Y.K.; Yen, H.W.; Huang, C.Y.; Chang, J.S. Microalgae as Sustainable Food and Feed Sources for Animals and Humans–Biotechnological and Environmental Aspects. Chemosphere 2021, 271, 129800. [Google Scholar] [CrossRef]
  285. Stanton, A.V.; James, K.; Brennan, M.M.; O’Donovan, F.; Buskandar, F.; Shortall, K.; El-Sayed, T.; Kennedy, J.; Hayes, H.; Fahey, A.G.; et al. Omega-3 Index and Blood Pressure Responses to Eating Foods Naturally Enriched with Omega-3 Polyunsaturated Fatty Acids: A Randomized Controlled Trial. Sci. Rep. 2020, 10, 15444. [Google Scholar] [CrossRef] [PubMed]
  286. Vo, T.S.; Kim, S.K. Fucoidans as a Natural Bioactive Ingredient for Functional Foods. J. Funct. Foods 2013, 5, 16–27. [Google Scholar] [CrossRef]
  287. Vickers, C.; Liu, F.; Abe, K.; Salama-Alber, O.; Jenkins, M.; Springate, C.M.K.; Burke, J.E.; Withers, S.G.; Boraston, A.B. Endo-Fucoidan Hydrolases from Glycoside Hydrolase Family 107 (GH107) Display Structural and Mechanistic Similarities to α-L-Fucosidases from GH29. J. Biol. Chem. 2018, 293, 18296–18308. [Google Scholar] [CrossRef]
  288. Wang, M.; Veeraperumal, S.; Zhong, S.; Cheong, K.L. Fucoidan-Derived Functional Oligosaccharides: Recent Developments, Preparation, and Potential Applications. Foods 2023, 12, 878. [Google Scholar] [CrossRef]
  289. Fernando, I.P.S.; Dias, M.K.H.M.; Madusanka, D.M.D.; Han, E.J.; Kim, M.J.; Heo, S.J.; Ahn, G. Fucoidan Fractionated from Sargassum Coreanum via Step-Gradient Ethanol Precipitation Indicate Promising Uvb-Protective Effects in Human Keratinocytes. Antioxidants 2021, 10, 347. [Google Scholar] [CrossRef]
  290. Chen, X.; Fu, X.; Huang, L.; Xu, J.; Gao, X. Agar Oligosaccharides: A Review of Preparation, Structures, Bioactivities and Application. Carbohydr. Polym. 2021, 265, 118076. [Google Scholar] [CrossRef]
  291. Karim, A.A.; Bhat, R. Gelatin Alternatives for the Food Industry: Recent Developments, Challenges and Prospects. Trends Food Sci. Technol. 2008, 19, 644–656. [Google Scholar] [CrossRef]
  292. Trius, A.; Sebranek, J.G. Carrageenans and Their Use in Meat Products. Crit. Rev. Food Sci. Nutr. 1996, 36, 69–85. [Google Scholar] [CrossRef] [PubMed]
  293. Zhu, B.; Ni, F.; Sun, Y.; Zhu, X.; Yin, H.; Yao, Z.; Du, Y. Insight into Carrageenases: Major Review of Sources, Category, Property, Purification Method, Structure, and Applications. Crit. Rev. Biotechnol. 2018, 38, 1261–1276. [Google Scholar] [CrossRef] [PubMed]
  294. Dai, Y.; Row, K.H. Application of Natural Deep Eutectic Solvents in the Extraction of Quercetin from Vegetables. Molecules 2019, 24, 2300. [Google Scholar] [CrossRef] [PubMed]
  295. Lin, J.; Zhou, W. Role of Quercetin in the Physicochemical Properties, Antioxidant and Antiglycation Activities of Bread. J. Funct. Foods 2018, 40, 299–306. [Google Scholar] [CrossRef]
  296. Bhat, I.U.H.; Bhat, R. Quercetin: A Bioactive Compound Imparting Cardiovascular and Neuroprotective Benefits: Scope for Exploring Fresh Produce, Their Wastes, and by-Products. Biology 2021, 10, 586. [Google Scholar] [CrossRef]
  297. Ceamsa. Available online: https://www.ceamsa.com/ (accessed on 7 May 2025).
  298. Swanson. Available online: https://swansoneurope.com/ (accessed on 7 May 2025).
  299. Algamar Ltd. Available online: https://algamar.com/ (accessed on 7 May 2025).
  300. Kohbo Labs. Available online: https://kobholabs.com/ (accessed on 7 May 2025).
Figure 1. Chemical structure and examples of phenolic compounds and fatty acids.
Figure 1. Chemical structure and examples of phenolic compounds and fatty acids.
Foods 14 01749 g001
Figure 2. Chemical structure of some major examples of polysaccharides and carotenoids with beneficial health effects.
Figure 2. Chemical structure of some major examples of polysaccharides and carotenoids with beneficial health effects.
Foods 14 01749 g002
Figure 3. Overall view of the therapeutic properties of antioxidant phytochemicals, as illustrated with some of the examples mentioned.
Figure 3. Overall view of the therapeutic properties of antioxidant phytochemicals, as illustrated with some of the examples mentioned.
Foods 14 01749 g003
Figure 4. Process of development of phytochemicals as nutraceuticals.
Figure 4. Process of development of phytochemicals as nutraceuticals.
Foods 14 01749 g004
Figure 5. Ingredient types, uses, and benefits in food applications.
Figure 5. Ingredient types, uses, and benefits in food applications.
Foods 14 01749 g005
Table 2. Chemopreventive activities of plant extracts and their phytochemicals: in silico, in vivo, and in vitro examples.
Table 2. Chemopreventive activities of plant extracts and their phytochemicals: in silico, in vivo, and in vitro examples.
Plant SpeciesExtractPhytochemicalsActivityAnalysis MethodAssayMechanism of ActionResultsRef.
Antioxidant activity
Garlic (Allium sativum L.)nsAllicin (purity > 90%)Antitumor (cholangiocarcinoma)CCK-8, FC, WBIn vitro/in vivoSTAT3 inhibition via SHP-1 upregulationSuppressed proliferation, invasion, EMT, and tumor growth[146]
nsHS-1793Resveratrol analogueAntitumor (murine breast cancer)LYM assays, DNA damage, Treg/TAMIn vivoInhibits immune suppression by Treg/TAMEnhanced LYM proliferation, reduced Tregs, and decreased IL-10/TGF-β[147]
Green tea Curcuma longa L.nsCatechins, curcuminAntitumor (OSCC)Histology, immunofluorescence, FCIn vivo AP induction and anti-angiogenesisReduced tumor growth, increased apoptosis[144]
Melilotus officinalis L.ns DC (coumarin derivative)Anti-proliferative, gonad-safeDC injection in BALB/c mice ovarian apoptosis, meiotic spindleIn vitro/In vivoCell cycle alteration without tubulin disruptionDC suppressed cell proliferation and increased AP in Vero and MCF-7 cells[148,149]
Polyalthia longifolia L.METetranorditerpeneAnticancer (prostate, leukemia cells)Proteomic analysisIn vitroER stress activation, apoptosisInhibited tumor cell growth[150]
Fagonia cretica L.AqEnsCytotoxic, induces cell cycle arrestsiRNA knockdown, MTT and FC, comet assay, WBIn vitroInduction of DNA damage, activation of p53 and FOXO3aInduced cell cycle arrest and AP in two phenotypic breast cancer cell lines[151]
Onobrychis argyrea L.ME (leaves)Quinic acid, isoquercitrin, epicatechin, routineAntioxidant, antidiabetic, anticancerLC-MS/MS, DPPH, iron reduction, enzyme inhibition, XTT, FCIn vitroME induces apoptosis in HT-29 cells by disrupting mitochondrial membranes and activating caspasesStrong antioxidant and cytotoxic effect[152]
Elephantopus mollis Kunth.ME3,4-di-O-caffeoyl quinic acidCytotoxicity, glucosidase inhibitionDPPH, FRAP, metal chelation, β-carotene, cytotoxicityIn vitroInduces cell death in NCI-H23 cells by triggering apoptosisHigh antioxidant capacity, induced apoptosis[153]
Thymus vulgaris L.MetE, EAE, ChE, BolE, AqE, PEEPolyphenols, tannins, flavonoids, sterols/triterpenesAntioxidantDPPH, ABTS, ferrous ion chelation, CVTIn vitroRadical scavenging, metal chelationStrong antioxidant capacity correlated with phenol and flavonoid content[154]
Markhamia lutea L.Leaf extractFlavonoids (O-glycosides)Antioxidant, anti-AChE, /BChE, Aβ-42 DPPH, ORAC, iron reduction, FRAPIn silicoin vitroInhibits AChE, BuChE, and Aβ-amyloid-42DPPH: 35.69 µg/mL, ORAC: 16,694.4 μM TE/mg, and iron chelation: 70.7 μM EDTA eq/mg[155]
Hertia cheirifolia L.Organic and EtOAc fraction Total phenolics (100–250 mg GAE/g)AntioxidantDPPH, ABTS, FRAP, β-caroteneIn vitroSynergistic compound interactionDPPH: 38.83 µg/mL, ABTS: 23.76 µg/mL, FRAP: 2628.87 µmol Fe2+ Eq/mL, and β-carotene: 58.91%[156]
nsAqEQUEAnticarcinogenicWB, RT-PCRIn vitroDownregulation of ROS, PKC, PI3K, and COX-2; upregulation of p53 and BAXQUE modulated OS and apoptotic pathways in HepG2 cells[157]
Anti-inflammatory activity
Tylophora ovata L.Natural and synthetic PAsO-methyltylophorinidine (1, 1s)Antitumor against TNBCNFκB inhibition, 3D co-cultureIn vitroStabilizes IκBα, blocks NFκBInhibited spheroid growth, surpassed paclitaxel[158]
Mangifera indica L.nsPolyphenolsAnti-inflammatory, anticancerReal-time PCR analysis and protein expressionIn vitroModulates PI3K/AKT/mTOR, NF-κB, PARP-1, and Bcl-2Reduced cancer cell growth by 90%[159]
Helicteres isora L.DCM-E/HeERosmarinic acidAnti-inflammatory, antioxidantELISAsIn vitroDifferentiation in cancer cells, shows no cytotoxic effect at high levelsReduced TNF-α, PGE-2, and NO levels; highest COX-2 inhibition[160]
Waltheria indica L.Roots and aerial parts/CH2Cl2 extractFlavonoids Anti-inflammatory, chemopreventiveNF-κB inhibition, luciferase reporter assay, QR-inducing assayIn vitroInduces phase 2 enzyme activity via QR induction assayA total of 7/29 compounds showed inhibitory activity on the NF-κB pathway[161]
Commiphora leptophloeos L.Hydroalcoholic leaf extractPhenolic acids and flavonoidsAnti-inflammatoryNO radical inhibition analysis, qPCR, physicochemical testsIn vitro/in vivoDownregulates NF-κB and COX-2, reduces cytokinesReduced inflammatory markers, promising for inflammatory bowel disease[162]
Matricaria chamomilla L.nsβ-Amyrin, β-eudesmol, β-sitosterol, apigenin, lupeol, quercetin, myricetinAnti-inflammatory, anticancerProteome analysis, WB, qRT-PCR, thermophoresisIn silicoin vitroInhibition of NF-κB, reduces IL-1β and IL6 mRNA expression and G2/M cell cycle arrestCancer prevention, reduced pro-inflammatory cytokine expression[163]
Asparagus densiflorus meyeri LRoot and aerial parts/DCM-ESaponins, glycosides, sterols, triterpenesCytotoxic, anti-inflammatoryMTT assay, MCF-7 cell stimulation using TNF-α, RT-PRCIn vitroReduces NO release and NF-κB gene expressionSignificant cytotoxicity (IC50 26.13 μg/mL)[164]
Capparis cartilaginea L.Ethanolic leaf extractAlkaloids, flavonoids, phenols, fatty acids, carotenesAntioxidant, cytotoxic, anti-inflammatoryFBRC, FRAP, MTT assay, COX-1 inhibitionIn vitro Dose-dependent inhibition of thermally induced protein denaturationAnti-inflammatory effects (IC50 60.23 and 17.67 µg/mL) were better than standards[165]
Corchorus olitorius L., Amaranthus hybridus L.Hydroethanolic leaf extractTannins, flavonoids, phenolics, terpenoids, cardiac glycosides, coumarinsPro-estrogenic, anti-inflammatoryPhytochemical and ELISA analyses In vivoLowers IL-6 and inhibits proliferation by binding phytoestrogens to ER-βAntioxidant because of its high tannin content/reduction in tumor size and incidence[166]
Euphorbia hirta L.Whole extract Phytol, fatty acids, 5-HMFAnti-inflammatoryNO productionIn vitroSuppression of PG generationInhibition of iNOS directly involved in inflammation[167]
Antidiabetic activity
Tradescantia pallida L.Leaf extractSyringic acid, p-coumaric acid, morin, catechinGlycosylation and hemoglobin activityα-Amylase assayIn vitroGlycosylation inhibition non-enzymaticallyBoosted insulin production, revitalized β-cells, inhibited AGEs, stimulated glucose transporters and AMPK[168]
Cissampelos capensis L.Leaf, stem, and rhizomeGlaziovine, pronuciferine, cissamanineAntihyperglycemicα-Amylase assayIn vitroEnzyme inhibition pathwayReduced glucose levels[169]
Phyllanthus emblica L.nsFlavonoidsAntihyperglycemicMolecular docking assayIn silicoHypoglycemic action, reduces relative risk of T2D, PPAR inhibition of T2DHigh binding affinity and selectivity for T2D therapeutic targets[170]
Ocinum sanctum L.LeavesEugenolAntihyperglycemic ELISA, RIA, and Neutral Red assayIn vitroPhysiological pathwayDecreased plasma glucose levels in T2D, increased islet insulin secretion, perfused pancreas[171]
Ocinum basilicum L.Leaves TPC and FCAntihyperglycemic Enzyme inhibitory activity assayIn vitroEnzyme inhibition pathway (α-glucosidase, α-amylase, DPP-IV, PTP1B, and SGLT2)Inhibition of intestinal sucrase, maltase, and porcine pancreatic α-amylase[172]
Derris elliptica L.LeavesQUE and ceramideAntihyperglycemic Biochemical analysis and histopathology studyIn vivoEnzyme inhibition pathwayIncreased insulin secretion, protected pancreatic β-cells from oxidative stress[173]
Carica papaya L.SeedsHexadecanoic acid methyl ester, 11-ODA oleic acidAntihyperglycemic α-Amylase and α-glucosidase inhibition assayIn vitroEnzyme inhibition pathwayReduced glucose levels[174]
Rhazya stricta L.RootsHexadecanoic acid, methyl esterAntihyperlipidemic, hepatoprotective DPP-IV, α-amylase, α-secretase inhibition assay, GLP-1 measurementIn vitro/in vivoEnzyme inhibition pathwayReduced blood glucose and HbA1c, reduced cholesterol and triglyceride levels, reduced liver enzyme activity[175]
Halooxylon stocksii L.Root and aerial parts8-ODA methyl esterAntidiabeticα-Amylase and α-glucosidase assayIn vitroEnzyme inhibition pathwayReduced glucose levels[176]
Anti-obesity activity
Rosa centifolia L.PetalsEllagic acid (polyphenols)Lipid metabolism improvementPCRIn vivoSuppression of lipid synthesis, inhibition of intestinal absorption, downregulation of Scd1 and Hmcgr mRNAs in the liverReduced body weight and adipose tissue, increased fecal triglycerides, lipid, and cholesterol metabolism[177]
Rheum rhabarbarum L.nsEmodin, rhein (anthraquinones)Lipid-lowering ELISA and histological evaluationIn vitro/in vivoFAS-ACC production prevention through decreased PPARγ and C/EBPα expression, reduced lipid accumulationBody weight and adipose tissue reduction[178]
Brassica juncea L.nsSinigrin (glucosinolate)Anti-obesityCell culture and XTT assay, WB, histological analysisIn vitro/in vivoReduces expression of adipogenic and lipid synthesis proteinsInhibited lipid accumulation in 3T3-L1 and decreased eWAT mass in obese mice fed a high-fat diet[179]
Anthophycus longifolius L.nsRhodomycinone, salsolinol, 5-HCO, 2-COS, demethylalangisideAnti-obesity, anti-hyperglycemiaα-Amylase, α-glucosidase, pancreatic lipase assayIn vitroEnzyme inhibition pathwayDelayed lipid, CH digestion, and absorption[180]
Solanum xanthocarpum L.Fresh and dry leavesSolasodine, carpesterol, β-sitosterol, diosgeninHypoglycemic, hepatoprotective, hypotensivePancreatic lipase inhibition assay, MTTIn vitronsAt 62.5 µg/mL, the fresh leaf extract reduced cancer cell viability by 50%[181]
Rumex rothschildianus L.Acetone fractionFlavonoids, phenolicsAnti-α-amylase, anti-α-glucosidase, anti-lipaseLipase inhibition activityIn vitro Inhibits OS, α-amylase, α-glucosidase, and lipaseStrong lipase inhibition (acetone fraction IC50 26.3 μg/mL), close to orlistat (IC50 12.3 μg/mL)[182]
Neuroprotective activity
Paeonia ostiiStamen(+)-3′′-methoxy-oxylactiflorinAnti-inflammatoryMolecular docking, NO inhibition assayIn vitro/in silicoInhibition of NO production by binding with protein COX-2Reduced NO production to values of EC50 3.02 μM[183]
Phyllanthus emblicaFruit extractnsAnti-inflammatoryNO inhibition assayIn vivoReduces IL-1β and TNF-α/increases expression of 5-HT1D, 5-HT2A, and D2 receptorsAmelioration of social interaction, social affiliation, anxiety, and motor coordination[184]
Tabebuia impetiginosaLeavesIridoids and organic acidsAnti-inflammatoryAChE inhibitory activity, LA detection, Y-maze test, PA assayIn vitro/in vivoCP attenuates cognitive impairment, effects rat performance in Y-maze and PA testsReduced CP-induced chemo-brain, restored hippocampal function[185]
Abbreviations: CCK-8: Cell Counting Kit-8; STAT3: Signal Transducer and Activator of Transcription 3; WB: Western Blotting; FC: Flow Cytometry; SHP-1: Src Homology 2 Domain-Containing Phosphatase-1; EMT: Epithelial–Mesenchymal Transition; Treg: Regulatory T Cell; TAM: Tumor-Associated Macrophage; LYM: Lymphocyte; IL-10: Interleukin-10; TGF-β: Transforming Growth Factor-Beta; ME: Methanol Extract; OSCC: Oral Squamous Cell Carcinoma; AP: apoptosis; DC: Dicoumarol; BALB/c: Bagg Albino Laboratory Strain/c; Vero: African Green Monkey Kidney Epithelial Cells; MCF-7: Human Breast Cancer Cells; siRNA: Small Interfering RNA; MTT assay: 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide Assay; p53: tumor protein p53; FOXO3a: Forkhead Box O3; QUE: quercetin; ROS: reactive oxygen species; OS: oxidative stress; PKC: Protein Kinase C; PI3K: Phosphatidylinositol 3-Kinase; COX-2: Cyclooxygenase-2; RT-PCR: Reverse Transcription Polymerase Chain Reaction; HepG2 cells: Human Liver Cancer Cell Line; AMPK: Adenosine Monophosphate-Activated Protein Kinase; AGES: advanced glycation end products; T2D: Type 2 Diabetes; PPAR: Peroxisome Proliferator-Activated Receptor; NFκB: Nuclear Factor Kappa B; IκBα: Inhibitor of Nuclear Factor Kappa B Alpha; HT-29 cells: Human Colorectal Adenocarcinoma Cell Line; DPPH: 2,2-Diphenyl-1-picrylhydrazyl; FRAP: Ferric-Reducing Antioxidant Power; NCI-H23 cells: Human Lung Adenocarcinoma Cell Line; ORAC: Oxygen Radical Absorbance Capacity; ER: Endoplasmic Reticulum; HeE: Hexane Extract; DCM-E: Dichloromethane Extract; MetE: Methanolic Extract; EAE: Ethyl Acetate Extract; ChE: Chloroform Extract; BolE: Butanol Extract; AqE: aqueous extract; PEE: Petroleum Ether Extract; CVT: Cyclic Voltammetry Technique; PI3K/AKT/mTOR: Phosphoinositide 3-Kinase/Protein Kinase B/Mechanistic Target of Rapamycin; PARP-1: Poly (ADP-ribose) Polymerase 1; Bcl-2: B-cell Lymphoma 2; TNF-α: tumor necrosis factor-alpha; THP-1 cells: Human Monocyte Cell Line; ELISA: Enzyme-Linked Immunosorbent Assay; QR: Quinone Reductase; NO: nitric oxide; 5-HMF: Hydroxymethyl-2-furancarboxaldehyde; iNOS: Inducible Nitric Oxide Synthase; PG: prostaglandin; RIA: Radioimmune Assay; TPCs: Total Polyphenol Compounds; FC: flavonoid content; 11-ODA: 11-Octadecanoic Acid; HbA1c: Glycosylated Hemoglobin; PCR: Polymerase Chain Reaction; FAS: Fatty Acid Synthase; ACC: Acetyl-CoA Carboxylase; 5-HCA: 5-Hydroxyconiferyl; 2-COS: 2-Caffeoylisocitrate; DPP-IV: Dipeptidyl Peptidase-IV; PTP1B: Protein Tyrosine Phosphatase 1B; SGLT2: Sodium–Glucose Co-Transporter 2; qPCR: Quantitative Polymerase Chain Reaction; FBRC: Ferric-Bipyridine-Reducing Capacity of Total Antioxidant; ER-β: Estrogen Receptor Beta; eWAT: epididymal white adipose tissue; IL-1β: Interleukin 1 Beta; TNF-α: tumor necrosis factor-alpha; 5-HT1D: Serotonin 1D Receptor; 5-HT2A: Serotonin 2A Receptor; D2: Dopamine 2 Receptor; AChE: Acetylcholinesterase; LA: Locomotor Activity; PA: Passive Avoidance; ns: Not Specified.
Table 3. Nutraceutical applications and functionality.
Table 3. Nutraceutical applications and functionality.
NutraceuticalSourceApplicationsFunctionalityHealth BenefitsRef.
PectinFruits (apple, citrus…)Jams, jellies, dairy productsGelling agent, thickenerAnticancer, immunomodulatory, anti-inflammatory, cholesterol-lowering[260]
InulinChicory rootLow-fat foods, fiber supplementsPrebiotic, fat replacerGut microbiota regulation, lipid metabolism regulation, mineral absorption enhancement, anti-inflammatory [261]
CellulosePlantsLow-fat foods, plant-based meats, bakery productsStabilizer, thickenerGut microbiota regulation, cholesterol reduction, blood glucose level regulation, anti-inflammatory [239]
Wheat branWheatCereals, bread, bakery productsTexture enhancer, fiber sourceGut microbiota regulation, cancer risk reduction, cardioprotective [262]
Psyllium huskPlantago ovata seedsFiber supplement, cerealsFiber source, thickenerAntidiabetic, reduces cholesterol levels, aids in gastrointestinal health [263,264]
CatechinsGreen tea leavesBeverages, supplements, snacksAntioxidant, antimicrobialAntioxidant, anti-inflammatory, antiviral, anti-obesity [265,266]
Grape seed extractGrape seedsBeverages, supplementsAntioxidant, antimicrobialAnti-inflammatory, antioxidant, cardioprotective, antimicrobial, anticancer [267]
β-caroteneCarrots, sweet potatoesSupplements, snacks, beverages, candiesColorantAntioxidant, supports immune function [268]
AnthocyaninsBerries, red cabbageSupplements, snacks, beverages, candiesColorantAntioxidant, anti-inflammatory, antidiabetic, anti-obesity [269]
BetaninsBeetrootSupplements, snacks, beverages, candiesColorantAntioxidative, anti-inflammatory, antidiabetic, potential anticancer benefits [248]
CurcuminTurmeric rootSupplements, snacks, beverages, candies ColorantAntioxidant, anti-inflammatory, anticancer, and immune-regulatory properties[270]
ResveratrolGrapesBeverages, supplementsAntioxidant, antimicrobialAntioxidant, anti-inflammatory, anticancer, cardioprotective [271]
FucoidansBrown algaeSupplements, fortified foodsGelling agent, thickenerAntioxidant, anti-inflammatory, anticoagulant, antitumor, antiviral [272]
AgarRed algaeJellies, jams, candy, plant-based gelatinGelling agent, texture enhancerAntioxidant, antiviral, antibacterial, prebiotic, antitumor [273]
CarrageenanRed algaeJellies, jams, candy, plant-based gelatinThickener, gelling agentCardioprotective, anticancer, antiviral, anticoagulant, antioxidant [274]
QuercetinOnions, apple peelsSupplements, fortified foodsAntioxidant, preservativeAnti-inflammatory, antimicrobial, anticancer, cardioprotective[275,276]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pereira, A.G.; Echave, J.; Jorge, A.O.S.; Nogueira-Marques, R.; Nur Yuksek, E.; Barciela, P.; Perez-Vazquez, A.; Chamorro, F.; P. P. Oliveira, M.B.; Carpena, M.; et al. Therapeutic and Preventive Potential of Plant-Derived Antioxidant Nutraceuticals. Foods 2025, 14, 1749. https://doi.org/10.3390/foods14101749

AMA Style

Pereira AG, Echave J, Jorge AOS, Nogueira-Marques R, Nur Yuksek E, Barciela P, Perez-Vazquez A, Chamorro F, P. P. Oliveira MB, Carpena M, et al. Therapeutic and Preventive Potential of Plant-Derived Antioxidant Nutraceuticals. Foods. 2025; 14(10):1749. https://doi.org/10.3390/foods14101749

Chicago/Turabian Style

Pereira, Antia G., Javier Echave, Ana O. S. Jorge, Rafael Nogueira-Marques, Ezgi Nur Yuksek, Paula Barciela, Ana Perez-Vazquez, Franklin Chamorro, Maria B. P. P. Oliveira, Maria Carpena, and et al. 2025. "Therapeutic and Preventive Potential of Plant-Derived Antioxidant Nutraceuticals" Foods 14, no. 10: 1749. https://doi.org/10.3390/foods14101749

APA Style

Pereira, A. G., Echave, J., Jorge, A. O. S., Nogueira-Marques, R., Nur Yuksek, E., Barciela, P., Perez-Vazquez, A., Chamorro, F., P. P. Oliveira, M. B., Carpena, M., & Prieto, M. A. (2025). Therapeutic and Preventive Potential of Plant-Derived Antioxidant Nutraceuticals. Foods, 14(10), 1749. https://doi.org/10.3390/foods14101749

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