Next Article in Journal
Design and Study of Nanoceria Modified by 5-Fluorouracil for Gel and Polymer Dermal Film Preparation
Previous Article in Journal
Cannabis-Based Medicine for Neuropathic Pain and Spasticity—A Multicenter, Randomized, Double-Blinded, Placebo-Controlled Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Citrus By-Products as a Valuable Source of Biologically Active Compounds with Promising Pharmaceutical, Biological and Biomedical Potential

1
Faculty of Food Technology Osijek, University of Osijek, Franje Kuhača 18, 31000 Osijek, Croatia
2
Department of Organic Chemistry, Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000 Split, Croatia
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(8), 1081; https://doi.org/10.3390/ph16081081
Submission received: 13 June 2023 / Revised: 26 July 2023 / Accepted: 27 July 2023 / Published: 29 July 2023
(This article belongs to the Section Natural Products)

Abstract

:
Citrus fruits processing results in the generation of huge amounts of citrus by-products, mainly peels, pulp, membranes, and seeds. Although they represent a major concern from both economical and environmental aspects, it is very important to emphasize that these by-products contain a rich source of value-added bioactive compounds with a wide spectrum of applications in the food, cosmetic, and pharmaceutical industries. The primary aim of this review is to highlight the great potential of isolated phytochemicals and extracts of individual citrus by-products with bioactive properties (e.g., antitumor, antimicrobial, antiviral, antidiabetic, antioxidant, and other beneficial activities with health-promoting abilities) and their potential in pharmaceutical, biomedical, and biological applications. This review on citrus by-products contains the following parts: structural and chemical characteristics; the utilization of citrus by-products; bioactivities of the present waxes and carotenoids, essential oils, pectins, and phenolic compounds; and citrus by-product formulations with enhanced biocactivities. A summary of the recent developments in applying citrus by-products for the treatment of different diseases and the protection of human health is also provided, emphasizing innovative methods for bioaccessibility enhancements (e.g., extract/component encapsulation, synthesis of biomass-derived nanoparticles, nanocarriers, or biofilm preparation). Based on the representative phytochemical groups, an evaluation of the recent studies of the past six years (from 2018 to 2023) reporting specific biological and health-promoting activities of citrus-based by-products is also provided. Finally, this review discusses advanced and modern approaches in pharmaceutical/biological formulations and drug delivery (e.g., carbon precursors for the preparation of nanoparticles with promising antimicrobial activity, the production of fluorescent nanoparticles with potential application as antitumor agents, and in cellular imaging). The recent studies implementing nanotechnology in food science and biotechnology could bring about new insights into providing innovative solutions for new pharmaceutical and medical discoveries.

Graphical Abstract

1. Introduction

Citrus fruits—more precisely, the genus Citrus L., which belongs to the subfamily Aurantioideae in the family Rutaceae—represent the major fruit crops commercially cultivated worldwide [1,2]. These fruits are widely known for their high health-benefiting properties, which is of great importance, since citrus fruits are the most widely consumed fruits globally [3,4,5,6]. The cultivation of Citrus genus includes the species such as lemon (C. limon (L.) Osbeck), sweet orange (C. sinensis (L.) Osbeck), mandarin (C. reticulata Blanco), grapefruit (C. paradisi Macfad.), pomelo (C. maxima (Burm.) Merr.), citron (C. medica L.), lime (C. aurantiifolia (Christm.) Swingle), and bergamot (C. × bergamia Risso & Poiteau) [4,5,7,8,9]. Although a significant part of the industrially processed citrus fruits is used to produce essential oils and juice, citrus-based candies, jellies, and extracts production also represent key factors for the food industry [5]. A large quantity of waste and by-products yearly produced during citrus processing has become a fundamental concern from both economical and environmental aspects. Citrus processing generates over 15 million tons of residue, mostly in the form of peels, seeds, and membranes [10,11]. Therefore, citrus waste valorization and proper industrial waste management are highly encouraged, being also a high-research priority topic for the scientific community recently. There have been many scientific reports related to the potential and beneficial utilization of industrial by-products, mainly dealing with innovative extraction methods for obtaining extracts rich in bioactive compounds [1,2,3,4,10,11,12]. These extracts have shown versatile health beneficial activities, due to the high content of biologically active compounds naturally present in the extracts [13,14]. Numerous studies have considered citrus extracts as a natural source of bioactive components exhibiting beneficial activities, including antioxidant [15,16,17,18,19], antibacterial [16,18,19,20,21], antidiabetic [17,22,23,24], neuroprotective [22,25,26,27], and anti-inflammatory [28,29,30,31] activities, as well as antitumor [32,33,34,35] potential. Also, citrus by-products are considered a valuable source of phytochemicals (such as d-limonene, essential oils, phenolic acids, carotenoids, vitamins, minerals, and flavonoids), which, isolated or in the form of mixtures/extracts, could exhibit versatile biological activities especially beneficial for the food industry [36,37,38]. Citrus-based essential oils exhibit significant antimicrobial activity against foodborne bacteria and also antioxidant activity to prevent the effects of oxidation; hence, citrus-based essential oils could act as natural preservatives [39,40,41]. Furthermore, several studies have reported the high repellent activity [42,43,44] and fumigant toxicity [45,46] of citrus-based essential oils against different insects.
It is well known that the presence of phenolic compounds is crucial for the bioactivity of the extracts; however, the lack of scientific evidence focusing on the challenges regarding their poor water solubility and the dependence on the temperature and pH environment, as well as poor bioaccessibility, are still limiting factors for the extract implementation in the in vivo studies [47,48]. Therefore, new solutions and technologies have emerged rapidly to promote and improve their bioaccessibility, including extract/component encapsulation [49,50,51,52], the synthesis of biomass-derived nanoparticles [53,54,55,56], nanocarriers, and biofilm preparation [57,58,59]. The identification and structural characterization of each chemical component present in the plant extract is surely beneficial to fully understand the mechanism for the formulation preparation, as well as to predict the underlying potential mechanism of action for in vitro and in vivo systems [60,61].
This review article gathers all the relevant evidence on the beneficial effects and promising health-promoting potential of the pure extracts and isolated phytochemicals derived from citrus by-products and aims to collect and provide the most recent literature on the developments and innovations regarding citrus-based by-products’ incorporation into the pharmaceutical, biomedical, and biological scientific areas. The major focus of this review will be directed toward providing a comprehensive view of the compositions of citrus by-products, such as essential oils, carotenoids, pectins, and phenolic compounds, based on their positions within the citrus fruit tissue, and also highlighting a wide diversity of their possible bioactivities and functionalities. Furthermore, the most recent improvements and developments in citrus by-product utilization, especially regarding the innovations in the pharmacological and biomedical fields, will be also reported, while the recent literature is indicative that nanotechnology could play a crucial role toward the specific drug delivery and bioaccessibility enhancements of versatile plant metabolites and plant-based formulations.

2. Structural and Chemical Characteristics of Citrus Fruits By-Products

Citrus fruits, like many other agricultural products, are characterized by their agricultural biodiversity [62]. Their physicochemical characteristics, as well as a diversity of chemical compounds, depend on a variety of factors and environmental conditions, such as soil, fertilization, age, position on the tree, maturity, and others [62,63,64,65]. It is interesting that all varieties of citrus fruits, by means of microscopic and macroscopic views, have similar structural and anatomical characteristics. A schematic view of the anatomical characteristics and structural compositions of citrus fruits is presented in Figure 1.
Citrus fruits are widely consumed due to their nutritional qualities and appealing taste and fragrance, and the products of citrus processing mostly include the production of food-grade (jellies, jams, candies, flavoring agents, etc.) and aromatic/cosmetic (essential oils) products [10,66,67]. The generation of significant amounts of waste during citrus fruit processing is a major concerning issue, as the waste represents almost 50% of the fresh fruit mass [66]. The generated waste includes by-products such as peels (the highest percentage of almost 50%), seeds (20–40%), pomace, and wastewater (the residue of spoiled parts of the fruits) [66,67,68].
The outer layer of the citrus fruit consists of the peel, which can be roughly divided into two regions: flavedo (lat. flavus means yellow) and albedo (lat. albus means white) [69,70]. The flavedo region comprises characteristic peel oils and pigments, and the white spongy part of the peel is referred to as the albedo [71]. Although, it is not uncommon that some literature reports refer to flavedo as epicarp and albedo as mesocarp [72]. The flavedo region is covered with a thin layer of cuticle, consisting of natural waxes and continuous polymerized materials [60,69]. The role of the cuticle is mainly protective against microorganism attacks, limit vapors, and water loss, regulating also the exchange of oxygen and carbon dioxide [73,74]. From a chemical point of view, natural waxes are characterized by the presence of long-chain alkanes, fatty acids, aldehydes, and alcohols [68,72], while the polymerized material originates from hydroxylated fatty acids [75]. Below the cuticle and within the flavedo region, pigments and essential oils are present. The citrus pigments are located within the chloroplasts (if green) and in chromoplasts (if yellow, orange, or red color) [62]. The composition and differences in the carotenoid content determine the color of citrus fruits [76]. The green or yellowish-green color of immature citrus fruits originates from the accumulation of lutein and a certain content of chloroplastic carotenoids, such as β- and α-carotene, neoxanthin, and zeaxanthin [76]. However, the orange color formed during the natural ripening of citrus fruits is caused by the increase in the content of colored carotenoids (β,β-xanthophylls) and by a decrease in the lutein concentration [77,78]. The essential oils are found in the oil glands located in the citrus flavedo layers and are defined as fragrant compounds present in the peel. The citrus essential oils consist of volatile compounds in the majority (85–99%) and in lower fractions as non-volatile compounds (1–15%) [79]. Interestingly, it has been reported that essential oils, although referred to as oils, are not typically oils in the chemical sense due to the absence of triglycerides, making this group of components the mixture of terpenes and terpenoids, which give rise to their hydrophobic character [62]. The volatile composition of citrus essential oils includes the highest content of monoterpenes (70–95%); sesquiterpenes; and their oxygenated derivatives (aldehydes, alcohols, esters of carboxylic acids, and ketones) [62,80], with d-limonene monoterpene as the major constituent of citrus essential oils [79,80,81]. The non-volatile fraction consists of carotenoids; fatty acids (oleic, linoleic, linolenic, stearic, palmitic, etc.); waxes; flavonoids; and sterols [82].
The albedo layer is considered to be a white and relatively porous layer of the citrus peel, consisting of pectic substances, cellulose, starch, and phenolic compounds [62,83]. The pectins are a complex group of compounds defined as non-starch polysaccharides, mostly consisting of conjugates of D-galacturonic acid; acid groups (methoxy esters); and some neutral sugars (rhamnose, glucose, xylose, arabinose, etc.) formed through α-(1-4)-glycosidic bonds [84,85]. Based on the degree of methylation and acetylation—more precisely, by the number of methoxyl and acetyl groups substituted by the carboxylic acid on the D-galacturonic acid chain—high-methoxyl or low-methoxyl pectins can be formed [84]. Although it is well known that low-methoxyl pectins are used in the food industry due to their gelation characteristics [86], pectins are also studied for their utilization in pharmacy and medicine (cholesterol reduction, drug delivery, immune modulation, etc.) [85]. Organic acids are classified as low molecular weight compounds that play a crucial role in plant metabolism; however, organic acids also exhibit protective and health-promoting activities [65,87]. The most commonly detected organic acids in citrus by-products are citric and malic, while benzoic, oxalic, tartaric, and succinic acids are also present in citrus by-products [88]. The most abundant vitamin in citrus fruits is ascorbic acid, most commonly known as vitamin C, while citrus fruits can also be a good source of folate (vitamin B9) [11,64,89]. Those vitamins have an important role in regulating immune functions with immune-enhancing potential [89]. Both organic acids and vitamins are predominantly found in the juice vesicles, located in the endocarp of citrus fruits [65].
Furthermore, phenolic compounds and flavonoids provide several benefits associated with health-promoting effects [65,90]. The major flavonoids present in citrus peels are hesperidin, narirutin, naringin, and rutin, while the content of each flavonoid depends on the physicochemical characteristics of the cultivated fruit and analyzed citrus variety [91]. The abundant chemical components present in Citrus fruits are shown in Figure 2. The citrus endocarp consists of juice vesicles and seeds, and interestingly, few scientific studies have reported the superior activity of citrus seed extracts compared to peel extracts [92,93]. The juice mostly consists of water (85–90%), and the soluble solids include carbohydrates; pigments; vitamins (vitamin C and vitamin B complex); minerals (calcium, potassium, magnesium, copper, and iron); and organic acids, while the pulp consists of the particles insoluble in the suspension of the juice [11,62,88].
The full potential of citrus seed utilization is still an area of interest for the scientific community, as there have been fewer studies dealing with citrus seed valorization and utilization [94]. One study reported the utilization of citrus seeds for fixed oil production, which were enriched with tocopherols, phytosterols, sugars, carotenoids, and minerals [95]. These oils were found to be adequate for soap making.

3. Converting Waste into Treasure—Utilization of Citrus By-Products

Recently, waste management has become one of the great concerns globally, and its valorization has created more sustainable and smart waste management solutions. Primarily, waste valorization includes employing different technologies toward obtaining value-added products with a wide spectrum of potential applications. Citrus by-products have been extensively studied due to their rich-bioactive properties, and their valorization enables beneficial gains from both economical and environmental points of view [96,97]. The scientific focus has been placed on innovative extraction methods for obtaining high-quality citrus essential oils [98,99,100,101] and enriched extracts in general [11,94,102,103]. Interestingly, the authors Tunç and Odabaş [99] reported a single-step ohmic heating-assisted extraction/hydrodistillation (OHAE/H) procedure to obtain the simultaneous recovery of essential oils and pectins from lemon waste. The process was optimized to obtain the maximum recovery of both components by response surface methodology. The liquid-to-solid ratio (w:v) and extraction/hydrodistillation time (min and voltage gradient (V/cm) were determined as independent variables, while the maximum pectin and essential oil yields were maintained as the dependent variables. The results obtained by the OHAE/H method were compared to the conventional extraction methods, and it was concluded that OHAE/H showed superior performance compared to the conventional methods. Moreover, in the study by Hwang et al. [104], the efficiency of hesperidin and narirutin extraction was investigated by combining pulsed electric field and subcritical water extraction techniques. Firstly, the samples were subjected to pulsed electric field treatments at the strength of 3 kV/cm for the times of 60 and 120 s. Subsequently, the subcritical water extraction was applied under the conditions of temperatures of 110–190 °C for 3–15 min. It was shown that the pulsed electric fields method enhanced the extraction process for obtaining both narirutin and hesperidin, increasing the yields by 22.1% and 33.6%.
Solid citrus waste can be also utilized for the production of animal food. Due to its good nutritional composition containing dietary fibers, lipids, flavonoids, enzymes, vitamins, and carotenoids, citrus waste represents a promising by-product for the production of livestock feeds [105]. The literature reports that citrus pulp (the main residue after juice extraction), citrus molasses (produced by concentrating on the press liquor of citrus peel residue with a high content of sugars), citrus peel liquor (similar to molasses but not as concentrated), and citrus-activated sludge (produced from liquid waste) could be considered as by-product feedstuffs [106]. The nutrient content of citrus by-products mainly depends on the source and variety of citrus fruits, as well as on the type of processing [88]. The main issues in the utilization of citrus by-products for the production of feedstuffs are the low nitrogen content and poor storage, which can lead to the development of mycotoxins [88]. Another valuable utilization of citrus waste is the production of packaging films that meets all the standards of sustainable and biodegradable forms of biopolymers [10,107]. Conventional packaging films are considered an environmental concern due to their poor biodegradable properties, and therefore, new innovative and sustainable solutions are welcomed [107]. An important advantage of using biopolymers derived from plant materials is that those raw materials naturally contain significant amounts of bioactive components that exhibit antioxidant and antimicrobial properties. The matrix of the citrus-based package is pectin, which enables solid support for the production of active packaging films [10]. In the study by Meydanju et al. [108], biodegradable film was prepared from lemon peel waste. Firstly, a composite of lemon waste powder, xanthan gum, and TiO2-Ag nanoparticles was produced. The additives enhanced the physicochemical properties of the prepared packaging; more precisely, the xanthan gum addition increased the thickness of the film, as well as the moisture content, due to the presence of -OH functional groups and the possibility of the hydrogen bonds forming. Also, both additives improved the thermal stability of the packaging films, and the antioxidant and antimicrobial properties were enhanced as well.
The application of plant-based and phenolic extracts in the food industry is highly limited due to their poor bioaccessibility, low water, and liquid solubility, while it is well known that bioactive phenolic compounds are extremely sensitive to light, oxidants, and changes in pH conditions and temperatures [47,97,109]. The main challenge presents as overcoming the limiting incorporation of low water-soluble compounds into aqueous-based foods, which directly limits the proper gastrointestinal bioaccessibility. In order to overcome these issues, encapsulation has been employed more frequently to protect bioactive compounds [47,110,111]. The spray-drying and freeze-drying techniques are commonly used methods for obtaining stable encapsulated functional substances, while extrusion methods, coacervation, and emulsification methods have been also applied [110]. In the study by Papoutsis et al. [112], different formulations of lemon by-product extracts combined with maltodextrin-coating agents (maltodextrin, maltodextrin and soybean proteins, maltodextrin, and ι-carrageenan) were prepared by both the spray- and freeze-drying methods. As expected, the formulations exhibited different morphological characteristics and showed an amorphous nature. The highest antioxidant activity was demonstrated with the sample containing lemon waste extract, maltodextrin, and soybean protein prepared by the freeze-drying process. In this case, the problem presented was polyphenol degradation due to freezing and dehydration and grinding the sample after the lyophilization, which could cause polyphenol oxidation. Moreover, polyphenol degradation occurred in the samples encapsulated by spray-drying, and exposing them to high inlet temperatures led to a significant decrease in the contents of the phenolic compounds.
Another interesting case of citrus by-product utilization is bioconversion into biofuels. It is known that the citrus residue contains significant amounts of carbohydrates and fermentable sugars; however, also, high contents of bioactive compounds inhibit the possible fermentation processes [113]. This issue could be overcome by extracting those bioactive molecules to be further used for biosorbents, biogas, and ethanol production by biotransformation. For example, the authors Oberoi et al. [114] used Kinnow mandarin waste to produce and optimize the process of bioethanol production. It has been reported that mandarin peel contains a high content of sugar, cellulose, and pectins and low content of lignin, making their citrus waste a promising substrate for bioethanol production. The process includes enzymatic hydrolysis, where the biomass is digested by enzymes into pentose and hexose sugars, which are used by microbes in the fermentation process.

4. Bioactivities of the Individual Groups of Compounds Present in Citrus By-Products

There are numerous published overviews dealing with the valorization of citrus by-products and their potential utilization in the food and cosmetic industries, emphasizing also their health-promoting properties due to the presence of bioactive compounds [1,2,3,4,11,13,97,105]. The present study uses the recent available scientific literature linking citrus by-products to their potential biological, pharmacological, and biomedical applications and includes both the utilization of plant extracts and pure compounds that can be separated from the citrus by-products. Furthermore, a brief overview of the application of nanotechnology in waste management and food science will also be provided.

4.1. Waxes and Carotenoids

Cuticular wax plays an important role in fruit preservation and proper storage, and it is well known that it acts as a natural barrier that protects plants from biological and non-biological stress [73,74]. Also, the structural characteristics, content, and composition of cuticular wax have been found to affect the postharvest storage quality against fruit water loss and softening and could be responsible for the resistance to fruit diseases, as summarized in Figure 3. Waxes are comprised of long-chain fatty acids and their derivates, esters, aldehydes, ketones, primary and secondary alcohols, and triterpenoids [115]. Most of the studies related to the topic of citrus cuticular waxes focused on the synthesis and transcriptional regulation of cuticular wax in citrus fruits. However, the authors Zhu et al. [116] carried out an investigation of the influence of C. reticulata cuticular wax on the colony expansion of the fungal pathogen Penicillium (P.) digitatum (green mold). The investigation included in vivo and Formvar®-based in vitro systems. Finally, it was concluded that the cuticular wax of mandarin fruit impairs P. digitatum colony expansion, acting as a physical barrier exhibiting antifungal activity. Furthermore, the content of carotenoids and the phytochemical profile of citrus fruits in general depend on the citrus variety, ripening stage, and the tissue [117]. It has also been reported that citrus fruits contain approximately 120 different carotenoids, classified as xanthophylls and carotenes [117].
The investigation of the carotenoid content separated from C. reticulata by-products and its influence on the immuneoxidative status of broiler chickens was carried out by Mavrommatis et al. [118]. The carotenoid-rich extract was prepared, and the chickens were fed a supplemented diet consisting of a freeze-dried formulation containing carotenoid extract and soluble starch. It was demonstrated that carotenoid-supplemented feed exerted inhibitory activity against Gram-positive (Staphylococcus (S.) aureus), as well as Gram-negative (Klebsiella (K.) oxytoca, Escherichia (E.) coli, and Salmonella (S.) typhimurium), bacteria. The implementation of the carotenoid content in the supplementation led to alanine aminotransferase and breast muscle malondialdehyde, and the activity of superoxide dismutase increased. Also, several parameters were downregulated, such as catalase, NADPH oxidase 2, interleukin 1β, and tumor necrosis factor. In the study by Barman et al. [119], β-carotene-loaded nanoemulsion was prepared from C. reticulata peels with the primary aim of carotenoid bioaccessibility improvement in fruit juice. Firstly, β-carotene was extracted using a mixture of hexane/acetone/ethanol solvents, and the sample was centrifuged, dried, and filtrated for nanoemulsion preparation. The nanoemulsions were prepared by suspending C. reticulata extract in hexane, and afterward, surfactants were added (caprylocaproyl polyoxyl-8-glycerides, polyoxyethylene, sorbitan monolaurate, and polyoxyethylene). The hexane was removed by rotary vacuum evaporation. The nanoemulsions were characterized by means of a physicochemical analysis, while the in vitro studies included gastrointestinal and gastric digestion. The results demonstrated that prepared formulations significantly increased the bioaccessibility of β-carotene and retinol activity equivalent in fruit juice. This study offered an alternative to synthetic color as a natural food colorant and, at the same time, provided health-promoting benefits to customers.

4.2. Aromatic Compounds—Essential Oils

Citrus essential oils are known as a fragrant mixture of chemical compounds exhibiting versatile activities used in the food, cosmetical, and pharmaceutical industries, as well as in aromatherapy [79]. The involvement of nanotechnology has provided new solutions for developing essential oil-based nanosystems with the aim of bioaccessibility enhancement. Interestingly, the formulation of C. lemon essential oil in nanohexosomes was prepared by the group of authors Sedeek et al. [120] for the purpose of antifungal activity investigation. Firstly, C. lemon, C. aurantifolia, C. maxima, and C. sinensis essential oils were extracted using hydrodistillation in a Clevenger’s apparatus from powdered peels. The hexosomal dispersions loaded with oils were prepared by the hot emulsification method reported by Abdel-Bar et al. [121]. In addition, the different obtained essential oils were assessed against phytopathogenic fungi (Rhizoctonia (R.) solani, Sclerotium (S.) rolfsii, Fusarium (F.) solani, Fusarium (F.) oxysporum, Fusarium (F.) semtectium, Botrytis (B.) cinerea, and Alternaria (A.) alternata), and it was concluded that all the tested essential oils exhibited strong antifungal activity, showing dose-dependent behavior. The C. lemon and C. aurantifolia essential oils exerted superior antifungal activity compared to the other essential oils, demonstrating the complete inhibition of F. solani, S. rolfsii, and F. oxysporum growth, while the C. lemon essential oil showed exclusive antifungal activity against A. alternata mycelial growth. Furthermore, the nanohexosomal formulation was prepared by using the best-performing sample of C. lemon essential oil, and it was shown that the nanohexosomal formulation completely inhibited mycelial growth of F. solani fungi at the applied concentration of 600 µL/mL, while the complete inhibition of S. rolfsii, A. alternata, and F. oxysporum was achieved at the concentration of 800 µL/mL. A moderate inhibitory effect was observed against R. solani, B. cinerea, and F. semtectium, with determined IC50 values of 416, 549.4, and 534 µL/mL, respectively. The authors Feng et al. [122] reported the potential hypercholesterolemia and hepatic steatosis preventive effects in male Sprague–Dawley rats on a high-fat diet. The essential oil of C. reticulata peel was obtained by the subcritical fluid extraction method, and limonene was determined to be the dominant component present in the essential oil formulation, followed by γ-terpinene and β-myrcene. The study combined biochemical analysis, lipidomics, and genes to investigate the hepatic steatosis and cholesterol improvements in high-fat diet rats. The high-fat diet in rats induced an increase in fat mass, liver weight, and hepatic lipid deposition with high serum and hepatic triacylglycerol levels. By introducing citrus essential oil as a food supplement, the total levels of the fatty acids, triacylglycerol, and cholesteryl ester classes in liver tissue significantly decreased, while the downregulation of lipogenesis-related genes and upregulation of bile acid-related genes was observed. The potential physiological stress amelioration and anti-inflammatory effects of C. depressa Hayata essential oil were reported by Asikin et al. [123]. The essential oil of C. depressa Hayata was extracted from the citrus pulp by applying hydrodistillation with a Clevenger-type apparatus. The GC–MS phytochemical profile analysis confirmed the presence of two dominant aromatic compounds, limonene and γ-terpinene. The influences of citrus essential oil on the neurological stress levels in nine healthy female panelists were monitored through electrocardiography (ECG) and electroencephalography (EEG) measurements, while the anti-inflammatory activity was assessed by nitric oxide (NO) and interleukin-1β inhibitory assays. By suppressing proinflammatory markers, the citrus essential oils showed promising anti-inflammatory potential, while the results of the EEG and ECG showed improvements in mental focus and stress reduction activity upon citrus essential oil inhalation.
A summary of the literature reporting the bioactivities of citrus-based essential oils is shown in Table 1.
Furthermore, the antimicrobial efficacy of citrus-based essential oils against foodborne pathogen Listeria monocytogenes (L. monocytogenes) was demonstrated in the study by Guo et al. [126]. Gram-positive bacteria L. monocytogenes is a highly adaptable pathogen that causes listeriosis, a life-threatening infection, and it is especially dangerous if the central nervous system is affected [141]. The essential oil from the C. Changshan-huyou Y.B. Chang (Huyou) species was extracted from peels by the steam distillation procedure using water as the solvent. The antimicrobial and antibiofilm ability of citrus essential oil was investigated against the L. monocytogenes pathogen, while the antilisterial mechanism was studied at the microscopic (SEM and TEM) (Figure 4) and molecular levels (RNA-seq analysis).
The results of the antimicrobial activity of Huyou essential oil against L. monocytogenes showed dose-dependant antimicrobial activity when comparing treatments of pathogens with the 1xMIC (minimum inhibitory concentration), 0.25xMIC, and 0.125 MIC. The study also discussed the changes in the physical morphology of L. monocytogenes biofilms when treated with 1xMIC for 8, 16, and 24 h by using scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM) analyses. As shown in Figure 4, the control sample demonstrated intact and complex structures, and upon the addition of Huyou essential oil, the destruction of the biofilms was observed. The most significant differences were observed after 16 and 24 h of treatment, where lysis and the death of the cells in the biofilms of L. monocytogenes were observed. When comparing the results of three different methods, such as SEM, CLSM, and COMSTAT, the authors concluded that, in the early stage of treatment (8 h), the predominant effect of rapid detachment of the biofilm was more likely to occur, while, in late stages (16 and 24 h), cell death might be the major effect to eradicate the rest of the biofilm. This study showed the great potential of citrus-based essential oils to be used as a natural food preservative for shelf life extension.

4.3. Pectins

The importance of pectins in the food industry is widely known; however, this group of polysaccharides has found their place in a variety of human applications, such as in the pharmaceuticals, cosmetics, drug delivery, and biomedical fields [86,141,142]. The versatile application of pectic biopolymers is enabled due to their structural diversity and chemical complexities, as well as the possibility of structural modifications [143]. The recent literature reports dealing with the bioactivities of citrus-based pectins are listed in Table 2. Recently, there has been an increase in interest in pectic biopolymers, mainly for their wide spectrum of bioactivities, and recently, pectic oligosaccharides have been evaluated for their promising prebiotic activity [144]. In addition, Zhang et al. [145] obtained pectin oligosaccharide fractions by the controlled degradation of citrus peel pectin. Three different oligosaccharides were prepared by adjusting the concentration of trifluoroacetic acid or H2O2 at the appropriate pH value, producing pectin oligosaccharides of variable molecular weight ranging from <2000 Da, 2000 to 3000 Da, and 3000 to 4000 Da. The results demonstrated a high prebiotic activity (pectic oligosaccharides obtained by H2O2 oxidation; 3543 Da) for Bifidobacterium (B.) bifidum and moderate activity against the Lactobacillus (L.) paracasei bacterial species. This study showed the enormous prebiotic potential of citrus-based pectic oligosaccharides; however, the greatest challenge remains to be overcome, as the human gastrointestinal tract includes complex pH-dependent processes and the presence of different enzymes that could affect in vivo digestion and bioaccessibility. Another interesting application of pectic biopolymers is their utilization as a carrier for drug delivery systems. The authors Lee and Chang [146] prepared quercetin-loaded hydrogel beads for the colon target, produced by deesterified pectin from yuzu (C. junos) peel and oligochitosan. A schematic illustration presenting the quercetin-loaded hydrogel beads preparation procedure and potential application in targeted therapy for colon cancer is shown in Figure 5.
For the purpose of the study, low-methoxyl pectin (DEYPP) was produced by deesterification with pectin methylesterase treatment, which is used for quercetin-DEYPP preparation. The hydrogel beads were prepared by dropping quercetin-DEYPP solution into a calcium chloride solution (1% w/w; pH = 6) containing oligosaccharide (1% w/w). Previously, cumulative quercetin release exposed to the simulated gastric fluid and intestinal fluid was very low (below 1%), and quercetin-loaded hydrogel beads significantly improved the bioaccessibility of quercetin in simulated colonic fluid (65.37–99.54%), which demonstrated the great efficiency of the developed quercetin drug delivery system for colon targeting. Furthermore, an example of citrus pectin-based drug delivery was reported by Jacob et al. [147], introducing pectin nanoparticles fabricated by ionotropic gelation using Mg2+ as a divalent cross-linker with the affinity of linking to the reactive carboxyl groups. Three different samples of nanoparticles were prepared as follows: high-methoxyl, low-methoxyl, and amidated low-methoxyl pectins. The cell viability on THP-1 (human leukemia monocytic cell lines) confirmed their excellent biocompatibility and potential application as a nanocarrier for oral drug delivery.
Table 2. Summary of the literature reporting bioactivities of pectins extracted from citrus by-products of the past six years (2018–2023).
Table 2. Summary of the literature reporting bioactivities of pectins extracted from citrus by-products of the past six years (2018–2023).
SourceFormulation/Chemical AnalyteBioactivityTesting SubjectsReferences
C. unshiu peelExtracted pectin (pH = 3; precipitation using 95% ethanol)Antioxidant activityTotal phenolic content (TPC), DPPH, ABTS•+, FRAP assay, ferrous ion chelating activity[148]
Citrus peel 1Commercially purchased pectins; pectin-capped copper
sulfide nanoparticles (pCuS NPs)
Antifungal activityIn vitro on Candida albicans[149]
Citrus peel 1Pectin oligosaccharide fraction obtained by controlled chemical degradation of citrus peel pectin (commercial)Prebiotic activityIn vitro on probiotic strains Bifidobacterium spp. and Lactobacillus spp./[145]
C. unshiu Marc. waste (remains from the canning processes)Depolymerized RG-I-enriched pectinPrebiotic activityIn vivo on male mice; Total serum cholesterol and triacylglycerol concentrations; Bacteroide thetaiotaomicron,
Bifidobacterium Longum
[150]
Citrus (lime/lemon) peel 1High methoxylated citrus pectin nanoparticles (HMP-NPs), low methoxylated citrus pectin nanoparticles (LMP-NPs), and low methoxyamidated citrus pectin
nanoparticles (AMP-NPs)
Oral drug deliveryIn vitro cell viability tests on THP-1 (human leukemia monocytic cell line) cell line[147]
Yuzu (C. junos) peelExtracted pectin (pH = 3.5; precipitation using 95% ethanol)/de-esterification of pectin/oligochitosan/quercetin hydrogel beads preparationDrug delivery/quercetin delivery system for the colon targetIn vitro release study using simulated gastric, intestinal,
and colonic fluids
[146]
C. reticulata peelsExtracted pectin (UAE 2; ammonium oxalate-oxalic acid—pH = 3.4; precipitation using 96% ethanol)Potential antitumor activityIn vitro on the normal human embryonic kidney (HEK293) cells and colon
cancer (HT29) cells
[151]
Lemon and lime peel 1Commercially purchased pectinsAnti-colitis activity/anti-inflammatory effectIn vivo on male C57BL/6 mice[152]
C. sinensis peel (IntegroPectin)Commercially purchased pectins/hesperidin-rich citrus pectinPrevention and therapy of COVID-19Computational studies: molecular model of the 3-chymotrypsin-like protease (3CLpro) structure of the SARSCoV-2[153]
Citrus peel 1Citrus pectin oligosaccharides obtained by H2O2 degradationHypocholesterolemic effectsIn vivo on male C57BL/6 mice[154]
Grapefruit peel (IntegroPectin) 1IntegroPectin isolated by freeze-drying of water-based extractCardioprotective effectsIn vivo on male Wistar rats[155]
1 Species not specified; 2 UAE—ultrasound-assisted extraction.

4.4. Phenolic Compounds

Natural phenolic compounds have been studied extensively for their essential role in plant protection, as well as for their beneficial effects on human health. It is well known that citrus by-products contain substantial contents of different phenolic compounds in the forms of acids and flavonoids, which have recently become the great subject of studies as natural antioxidants [156,157]. The representative bioactive compounds for the citrus family are flavanone aglycones (hesperetin, naringenin, and eriodictyol); flavone and flavonol aglycones (kaempferol, quercetin, apigenin, and diosmetin); flavanone-7-O-glycosides (eriocitrin, hesperidin, naringin, narirutin, poncirin, and didymin); and polymethoxyflavones (PMFs; nobiletin, tangeretin, and sinensetin) [62,91,158]. A summary of their bioactivities is listed in Table 3.
The great potential of citrus-based extracts lies in their health-promoting ability, exhibiting a wide spectrum of bioactivities, such as antioxidant, anti-inflammatory, and antiproliferative activity, against cancer. Interestingly, Shimamura et al. [159] studied the protective effects of hesperidin-rich extract obtained from C. unshiu (Chenpi) peel and commercially supplied hesperidin on aspirin-induced oxidative damage in rats. One of the major possible side effects of aspirin prescription and consumption is the possibility of peptic ulcer formation, which represents a serious gastrointestinal disease [160]. The citrus extract was obtained by reflux extraction, and a HPLC analysis confirmed the abundant presence of hesperidin in the extract sample. In order to evaluate the inhibitory effects of citrus extract and hesperidin on DNA oxidative damage in the stomach, kidney, and liver, the formamidopyrimidine DNA glycosylase (Fpg)-modified comet assay was applied. Also, the in vivo studies included five-week-old male ddY mice for evaluating analgesic activity and nine-week-old male Wistar rats for assessing oxidative damage. As demonstrated in Figure 6, the inhibitory effects of citrus extract and hesperidin were obvious by studying the aspirin-induced oxidative gastric mucosal injuries and by the reduction of the 8-oxoG content (content increases by oxidative stress) when the combined drug was administered.
Finally, the study indicated the protective effects of citrus extract and hesperidin in aspirin-induced damage, while the pharmacological action of aspirin did not change significantly. The important role of hesperidin in gastrointestinal health was also reported by Sharaf et al. [161]. Hesperidin was extracted from C. uranium peel by Soxhlet extraction with petroleum ether and methanol, while the crystallization of pure hesperidin was done with 6% glacial acetic acid (pH = 3–4). The isolated hesperidin was investigated for anti-Helicobacter (H.) pylori activity, which is also the main contributor to the occurrence of chronic gastritis and peptic ulcers, also increasing the risk of gastric adenocarcinoma [162]. The progressive reduction in urease activity by hesperidin and urease inhibition kinetic analyses indicated the anti-Helicobacter pylori activity of hesperidin by competitive mode in a time-dependent manner. Also, the in situ visualization of antimicrobial activity by laser scanning confocal microscopy (LSCM) demonstrated that hesperidin administration led to amino acid leakage from bacterial cells, while scanning electron microscopy (SEM) demonstrated the interaction of hesperidin and bacterial cells causing cell disruption and leakage of the cytoplasmic content. Furthermore, the molecular docking and simulation of the inhibitory effect of hesperidin (urease–ligand) on HpUre enzyme through slow-binding inhibition indicated the possible formation of hydrogen bonding, van der Waals, and alkyl interactions with important residues on enzyme HpUre-active sites. The bioavailability assays indicated the high potential of hesperidin for oral usage. The beneficial effects on the gastrointestinal system were also reported by applying the Ougan peel extract enriched with nobiletin, tangeretin, and 5-demethylnobiletin compounds exhibiting antitumor activity against gastric cancer cell lines [163], while protective and enhanced anticancer effects of orange peel extract and naringin in the doxorubicin treatment of esophageal cancer cells in a mice model were reported by Tajaldini et al. [164]. The anti-inflammatory effects of citrus-based phenolic compounds are also reported in Table 3. The combination of naringin obtained from C. maxima (Burm.) extract and sericin from Bombyx mori was investigated for the potential treatment of psoriasis by Deenonpoe et al. [165]. With the assumption that skin inflammation via proinflammatory cytokines is associated with the pathogenesis and clinical manifestation of psoriasis, the inhibitory effect of naringin/sericin drugs on the production of proinflammatory cytokines (TNF-α, IL-6, IL-23, and IL-12p40) and the expression of mRNA of the human peripheral blood mononuclear cells from psoriasis patients were investigated. The study demonstrated the successful dose-dependent formulation of naringin/sericin for downregulating the proinflammatory cytokines related to the inflammation mechanism in psoriasis pathogenesis.
Table 3. Summary of the literature reporting bioactivities of different (poly)phenolic compounds extractable from citrus by-products (2018–2023).
Table 3. Summary of the literature reporting bioactivities of different (poly)phenolic compounds extractable from citrus by-products (2018–2023).
SourceChemical AnalytesBioactivityTesting SubjectsReferences
Citrus by-products
Finger lime peels 1Dominant phenolic acids: malic, citric, and quinic acid/phenolic compounds: neohesperidin, α-glucosyl hesperidin, (7S,8S)-4,7,9,9′-tetrahydroxy-3,3′-dimethoxy-
8-4′-oxyneolignan-9′-O-D-glucopyranoside, lyoniresinol
9′-O-glucoside and poncirin
Antioxidant, anti-inflammatory effect, neuronal cell protectionAntioxidant: DPPH, ABTS•+, FRAP, ORAC/anti-inflammatory: in vitro on BV-2 (mouse microglial) cells and NO release analysis[166]
Citrus
(C. lumia Risso) albedo extract (peel and pulp)
Dominant phenolic acids: chlorogenic and ferulic acids/flavonoids: hesperidin and eriocitrinAntioxidant and cytoprotective activityAntioxidant: FRAP, TEAC, DPPH, ORAC, β-Carotene bleaching/cell viability on lymphocytes (lactate
dehydrogenase (LDH) activity)
[167]
C. unshiu (Chenpi) peelDominant flavonoid: hesperidin/Hesperidin (commercial product)Analgesic activity and gastroprotective effectIn vitro on gastric tissue/in vivo on male ddY mice[159]
C. amblycarpa peels and leavesPhenolics: quercetin, rutin, and ɣ-aminobutyric acid (GABA)Antihypertensive effectsACE Inhibitory Activity Assay[168]
Ougan peel extracts 1Flavonoids: nobiletin, tangeretin, and 5-demethylnobiletinAntitumor activityIn vitro on gastric cancer cell line AGS, BGC-823 and SGC-7901/in vivo BALB/c nude mice[163]
C. reticulata Cv. Suavissima peel extractFlavonoids: nobiletin, tangeretin, and 5-demethylnobiletinAnti-inflammatory effectIn vitro on BV-2 (mouse microglial) cells and NO release analysis, JAK2 inhibitor Ruxolitinib and the STAT3 inhibitor Stattic[169]
C. reticulata Blanco, C. grandis, C. reticulata c.v. Kinnow, C. limetta, and C. sinensis peel extractsDominant flavonoids: hesperidin, naringin, quercetin, rutin, apigenin, nobiletin, tangeretinAntioxidant activity, anti-inflammatory effect, neuroprotective effect Antioxidant: DPPH and ABTS•+ assay/Anti-inflammatory: protein denaturation assay (bovine serum albumin protein denaturation)/neuroprotective: Acetylcholinesterase inhibition assay[96]
C. japonica var. Margarita peelDetected phenolic acids: p-hydroxybenzoic acid, vanillic acid, protocatechuic acid, chlorogenic acid, sinapic acid, gallic acid, ferulic acid, caffeic acidAntioxidant and antimicrobial activityAntioxidant: DPPH/Antimicrobial: E. coli, Salmonella (S.) typhimurium, S. aureus
and Pseudomonas (P.) aeruginosa
[170]
C. sinensis (navel orange)Hydroethanolic extract, naringin, naringeninHepatopreventive activityIn vivo on male Wistar rats; histopathological investigation and immunohistochemical detection of p53, Bax, Caspase-3, and Bcl-2[171]
C. reticulata peelExtract (major components): hesperidin, nobiletin, narirutin, tangeretin, and sinensetinAntiobesity-
related effects.
In vitro on 3T3-L1 mouse preadipocytes[172]
Ten citrus samplesDetected components: nobiletin, quercetin, diosmin, naringenin, hesperidin, hesperetin, rutinAnti-estrogenic and anti-aromatase activity/antitumor activityIn vivo on immature female Swiss albino mice/in vitro on MCF-7 and T47D (breast cancer lines), as well as the normal human HFB4 cells[33]
C. unshiu peelDetected components: rutin, naringin, hesperidin, poncirinAnti-inflammatory and antioxidant activityIn vitro on RAW 264.7 macrophages (originating from Abelson leukemia virus)[173]
14 Chinese cultivars (mandarins, oranges, pummelos, hybrids, citrons, kumquats)Detected components: eriocitrin, naringin, hesperidin, didymin, poncirin, naringenin, hesperetin, sinensetin, nobiletin, tangeretin, and 5-O-demethylnobiletinAntioxidant activity, and effects on intestinal
microbiota
Antioxidant: DPPH, ABTS•+, FRAP, CUPRAC/a-Glucosidase inhibition assay/bile salt binding capacity determination assay/investigation on fecal samples/in vitro on simulated intestinal fermentation[13]
Sour orange, sweet orange, and lemon peels 1Dominant phenolic acids: o-coumaric acid, benzoic acid, ellagic acid, p-Hydroxybenzoic acid/flavonoids: myricetin, naringin, quercetinProbiotic and symbiotic activity (Acidophilus-bifidus-thermophilus (ABT)-Type Synbiotic
Yoghurt)
Antioxidant: DPPH/antibacterial: S. aureus, Bacillus (B.) subtilis, and E. coli[174]
C. limetta peelHesperidin-rich ethanol extractManagement of the rheumatoid arthritisIn vivo on Charles foster rats and Swiss albino mice[175]
Individual components
C. sinensis L. Osbeck peel and pulpHesperidin-rich extractAntioxidant and antidiabetic activityAntioxidant: DPPH, ABTS•+, iron chelating activity/in vitro α-Amylase inhibition assay[176]
Commercial productHesperetin and quercetinDrug deliveryIn vitro on MDCK II (Madin-Darby canine kidney cells) cell viability[177]
C. uranium
peel
HesperidinAnti-Helicobacter pylori
activity
In vitro on human H. pylori strains/urease inhibition assay/molecular docking[161]
C. reticulata peelHesperidinAntihyperglycemic, antihyperlipidemic, and antioxidant activityIn vivo on male Wistar rats/biochemical assay and histological investigation [17]
Commercial productHesperetinTreatment and prevention of cardiovascular diseasesEx vivo on porcine coronary arteries and human coronary artery smooth muscle cells [178]
Commercial productHesperidinAntitumor activityIn vivo on male-specified pathogen-free C57BL/6N mice/in vitro on Lewis lung carcinoma (LLC) cells[179]
Commercial productsA mixture of hesperidin and naringeninTreatment and prevention of cardiovascular diseasesIn vivo and ex vivo on male Wistar rats and aortic rings[180]
Commercial productHesperidinAntitumor activityIn vitro on PC3 and DU145 (human prostate cancer) cell lines[181]
Commercial productHesperetin and naringeninAntitumor activityIn vitro on MIA PaCa-2, PANC-1, SNU-213 (pancreatic cancer cell lines), Detroit 551 (skin fibroblast), and human umbilical vein endothelial cells (HUVECs)[182]
C. sinensis var. Valencia peelNaringeninHepato- and renoprotective effectsIn vivo on male Wistar rats/histological investigation of the liver and kidney tissues[183]
Commercial productNaringeninAnti-proliferative effect., wound healingIn vitro on human A549 lung cancer cells[184]
Commercial productNaringenin, nobiletin, and hesperidinTreatment of optic nerve injury, neuroprotectiveIn vivo on 6-weeks-old C57BL/6J mice/in vitro on HEK293T (human embryonic kidney cells) cells[185]
Combination of commercial products (naringin and doxorubicin), orange peel 1Combination of naringin, doxorubicin, and orange peel extractAntitumor activityIn vivo on mice models/in vitro on YM1 (human esophageal squamous cancer cell line)/[164]
C. junos Tanaka peelNaringinPreventive effect on pulmonary damageIn vivo on male 7-week-old BALB/c mice/in vitro on NCI-H460 (the human lung carcinoma cell lines)[186]
C. maxima (Burm.) Merr peelNaringin crystals + sericinTreatment of psoriasisIn vitro on isolated human peripheral blood mononuclear cells, investigation on proinflammatory cytokines (TNF-α, IL-6, IL-12p40, and IL-23)[165]
Commercial productNarirutinAntitumor activityIn vitro on PC-3 (prostate carcinoma and HEK-293 (embryonic kidney) cell lines[187]
Grapefruit 1 peelNarirutin-rich fractionsNeuroprotective effect (cerebral ischemia/reperfusion injury)In vivo on male Wistar rats[188]
Commercial productPoncirinAntidiabetic activityPTP1B inhibitory assay, α-Glucosidase inhibitory assay, HRAR inhibition assay/in vitro on C2C12 cell (skeletal muscle cells) line[189]
Commercial productPoncirin and isosakuranetinBeneficial effects on gut microbiotaIn vivo on thirty C57Bl/6J male mice/fecal microbiota [190]
C. sinensis peelRutinAntimicrobial activityIn vitro on Aeromonas (A.) hydrophila strains[191]
Citrus peel 1TangeretinAntitumor activityIn vitro on MCF-7 and MDA-MB-231 (breast cancer) cell lines[34]
Commercial productDiosmetinAntihypertensive effectsIn vivo on adult Sprague–Dawley rats/in vitro: vascular pathway inhibitors[192]
Commercial productDiosmetin and diosminAnti-inflammatory effect on atopic dermatitisIn vivo on six-week-old female SKH-1 hairless mice/in vitro: RBL-2H3 (basophilic leukemia) cell line[193]
1 Species not specified.

5. Citrus By-Products Formulations with Enhanced Bioactivities

The biomass-derived compounds are known for their health-promoting properties, and there is a rising trend in waste and by-product valorization to obtain value-added products with a wide spectrum of applications [2,11,194,195]. As it was already demonstrated in Table 1, Table 2 and Table 3, the beneficial effects of different citrus by-products on human health are not disputable; however, poor bioaccessibility is a crucial and limiting factor for successful in vivo applications. Therefore, new and innovative ideas with the implementation of nanotechnology brought about some new solutions in bioaccessibility and bioactivity enhancements (Table 4). The preparation of silver nanoparticles (AgNPs) from citrus (C. tangerina, C. sinensis, and C. limon) peel extract was reported by Niluxsshun et al. [196]. Firstly, citrus peel extracts were prepared by boiling peels in hot water, and afterward, a solution of AgNO3 was added to the flask when a golden colloidal suspension was formed. The structural and morphological analyses confirmed the presence of AgNPs in sizes of 10–70 nm, containing different morphological characteristics of nanoparticles. The presence of natural antioxidants, flavonoids, phenolic acids, and other phenolic compounds could act as a reducing agent, leading to the formation of silver nanoparticles. The AgNPs were investigated for antimicrobial activity against the Gram-negative bacteria E. coli and the Gram-positive bacteria S. aureus, and the results showed the superior antimicrobial activity of orange-based AgNPs on both bacteria strains. Also, it is expected that the bioactivity of nanoparticles is dose- and size-dependent [197], and it is assumed that silver potentially interacts with thiol groups of proteins on cell membranes, causing respiration blocking, which leads to cell death. Another example of AgNP synthesis by using citrus by-products for the purposes of antimicrobial investigation was reported by Alkhulaifi et al. [198]. In this study, C. limon peels were used for the synthesis of AgNPs, which were formed by the addition of a AgNO3 solution. Again, a possible explanation for the AgNPs formation was the reduction of Ag+ ions to silver nanoparticles in the presence of phenolic compounds, and the AgNPs demonstrated spherical- and rod-like-shaped morphologies. The antimicrobial activity investigation was carried out on Acinetobacter (A.) baumannii, S. typhimurium, E. coli, Pseudomonas (P.) aeruginosa, S. aureus, and Proteus (P.) vulgaris human pathogenic bacteria. The results indicated the good performance of AgNPs against the Gram-negative (E. coli, S. typhimurium, and P. aeruginosa) and Gram-positive (S. aureus) bacteria. Also, the cell viability on the MCF-7 (human breast cancer) and HCT-116 (human colon carcinoma) cell lines was evaluated, showing dose-dependent behavior. The good antimicrobial performance was potentially explained by four possible mechanisms: (1) the interactions of AgNPs with the cell membrane, altering the membrane permeability and perturbation of respiratory chain enzymes; (2) the gradual diffusion of AgNPs into the cells, leading to the conjugation of nanoparticles to DNA and causing adverse effects on the enzyme activity and the transcription processes; (3) the leakage of subcellular components as the interaction of AgNPs and plasma membrane were formed; and (4) the generation of free radicals [199,200]. Recently, biomass-derived carbon quantum dots (CQDs) are gaining attention due to their biocompatibility and versatile physicochemical and optical properties. By definition, CQDs are fluorescent carbon (zero-dimensional) nanoparticles possessing small size, low toxicity, controllable solubility, and tunable light-emitting properties. Therefore, all of these properties allow CQDs a wide spectrum of applications in bioimaging, biosensing, catalysis, and theranostics [201,202]. The authors Gudimella et al. [203] reported a green synthetic procedure for obtaining CQDs from citrus peel and the conjugation of CQDs with folic acid. A structural analysis confirmed the presence of nanoparticles of sizes 4.6 ± 0.28 nm, while fluorescence spectroscopy indicated that CQDs exhibited multiple colors at different excitation wavelengths. The biocompatibility of the CQDs was assessed on the L929 (mice fibroblasts) cell lines, and the CQDs conjugated with folic acid exhibited low cytotoxicity, showing good biocompatibility. The bioimaging of cancer cell lines was successfully studied on breast cancer (MCF-7) cell lines, as shown in Figure 7. The CQDs were introduced to MCF-7 cell lines and were illuminated by bright light, UV light (330–385 nm), blue (450–480 nm) light, and green (510–550 nm) light. It was demonstrated that MCF-7 cell lines treated with CQDs conjugated with folic acid exhibited brighter fluorescence emission compared to pure CQDs. It was reported that folic acid conjugation could produce a stronger fluorescent signal, which was probably caused by the enhanced cellular uptake of CQDs conjugated with folic acid in the cancer cell lines.
Similar results were obtained in the study by Šafranko et al. [204], where cellular imaging was investigated for the MCF-7 cell lines; however, specific antitumor activity against CFPAC-1 (ductal pancreatic adenocarcinoma) was determined. The CQD nanoparticles were prepared by a hydrothermal procedure from C. clementina peels and amino acids (Gly and Arg), and their application in Fe3+ ion sensing and bioimaging was determined, also exhibiting specific antitumor and antioxidant activities. A literature overview of innovative synthetic approaches for obtaining citrus by-product-based formulations is listed in Table 4.

6. Conclusions and Final Remarks

The enormous amount of waste and by-products generated during citrus processing is of great concern from both economical and environmental points of view. There are many valuable contributions dealing with the valorization of these by-products, converting them into value-added products with potential applications in the food, cosmetic, and pharmaceutical industries. The major focus has been on the promising bioactivities of different citrus by-products and their beneficial effects on human health, and this discovery offers new alternatives for safer, healthier, and sustainable product development. Although citrus by-products can be considered a valuable and natural source of bioactive compounds, the limiting factors for in vivo applicability are the poor bioaccessibility and solubility of different phenolic compounds and antioxidants, as well as sensitivity to light, pH, humidity, and heat. This challenge can be overcome by forming stable nanoemulsions and different formulations on a nanoscale that enhances the bioactivity, as well as the bioaccessibility, of the active substances. Furthermore, one of the major concerns is the lack of evidence for efficient citrus-by-product utilization/extraction at a larger industrial scale, as, currently, the available literature reports the extraction procedures on a laboratory scale and, in limited cases, on a pilot scale. Also, the extraction of bioactive compounds has limitations by means of low extraction yields of individual compounds or groups of bioactive compounds, and it is certainly questionable that these amounts can satisfy the demands of the different industries.
As was discussed in this review article, different citrus-based by-products show enormous potential in pharmaceutical and biomedical fields. The individual citrus-based compounds, mixtures such as essential oils, and extracts exhibit a diversity of bioactivities, including antitumor, antimicrobial, antiviral, antidiabetic, antioxidant, and other beneficial activities with health-promoting abilities. The investigation of the antitumor ability of different citrus-based by-products is an indisputably emerging trend in the pharmacological and biomedical domains; however, the major limiting factor for applying these formulations is often a lack of knowledge regarding the antitumor mechanisms of these by-products. In order for these by-products to be applied as an alternative to synthetic drugs in chemotherapy or in cancer target therapy, future studies should be more focused on the mechanisms of action to fully understand their antitumor activity, as well as on increasing their specific targeting properties for tumor cells. Furthermore, citrus-based by-products have enormous potential as antimicrobial agents in the food, agriculture, and pharmaceutical industries. Due to the serious threat to human health, the antimicrobial resistance topic has been widely discussed within the scientific community. In this review article, an overview of the recent literature related to the application of citrus-based by-products as antimicrobial agents has been provided. Although it is well known that plant-based by-products exhibit antimicrobial properties under laboratory conditions, more extensive investigations regarding the isolation of specific bioactive compounds, mechanisms of action, in vivo studies, and structure–activity relationship (SAR) analysis are welcomed in the future.
As shown in this review article, citrus by-products can be used as carbon precursors for the preparation of nanoparticles with promising antimicrobial activity, as well as for the production of fluorescent nanoparticles with potential applications as antitumor agents and in cellular imaging. Nowadays, nanoparticles are successfully overcoming the limitations of nonspecific drug delivery and offer multiple benefits in treating human diseases. However, more advanced solutions are needed regarding optimized drug delivery, improvements in their accumulation at the sites of interest, and minimizing unwanted toxicity to ovaerall improve patient outcomes. As plant-based products are generally less toxic in normal cells compared to synthetic compounds, those products show a promising future in medicine, especially with a multidisciplinary collaborative approach of plant science and nanotechnology. With the technological advances, and by applying an interdisciplinary approach, citrus by-products can be a valuable source of compounds with effective antitumor, antioxidant, and protective effects used as health-promoting agents.

Author Contributions

Conceptualization, S.Š., D.Š. and S.J.; writing—original draft preparation, S.Š.; writing—review and editing, S.Š., D.Š., I.J. and S.J.; supervision, D.Š. and S.J.; project administration, S.J.; and funding acquisition, I.J., S.J. and S.Š. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Croatian Science Foundation under the project “Application of innovative techniques of the extraction of bioactive compounds from by-products of plant origin” (UIP-2017-05-9909).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khan, U.M.; Sameen, A.; Aadil, R.M.; Shahid, M.; Sezen, S.; Zarrabi, A.; Ozdemir, B.; Sevindik, M.; Kaplan, D.N.; Selamoglu, Z.; et al. Citrus Genus and Its Waste Utilization: A Review on Health-Promoting Activities and Industrial Application. Evid.-Based Complement. Altern. Med. 2021, 2021, 2488804. [Google Scholar] [CrossRef] [PubMed]
  2. Lv, X.; Zhao, S.; Ning, Z.; Zeng, H.; Shu, Y.; Tao, O.; Xiao, C.; Lu, C.; Liu, Y. Citrus Fruits as a Treasure Trove of Active Natural Metabolites That Potentially Provide Benefits for Human Health. Chem. Cent. J. 2015, 9, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Liu, S.; Lou, Y.; Li, Y.; Zhang, J.; Li, P.; Yang, B.; Gu, Q. Review of Phytochemical and Nutritional Characteristics and Food Applications of Citrus L. Fruits. Front. Nutr. 2022, 9, 968604. [Google Scholar] [CrossRef] [PubMed]
  4. Russo, C.; Maugeri, A.; Lombardo, G.E.; Musumeci, L.; Barreca, D.; Rapisarda, A.; Cirmi, S.; Navarra, M. The Second Life of Citrus Fruit Waste: A Valuable Source of Bioactive Compounds. Molecules 2021, 26, 5991. [Google Scholar] [CrossRef] [PubMed]
  5. Ben Hsouna, A.; Sadaka, C.; Generalić Mekinić, I.; Garzoli, S.; Švarc-Gajić, J.; Rodrigues, F.; Morais, S.; Moreira, M.M.; Ferreira, E.; Spigno, G.; et al. The Chemical Variability, Nutraceutical Value, and Food-Industry and Cosmetic Applications of Citrus Plants: A Critical Review. Antioxidants 2023, 12, 481. [Google Scholar] [CrossRef]
  6. Rao, M.J.; Zuo, H.; Xu, Q. Genomic Insights into Citrus Domestication and Its Important Agronomic Traits. Plant Commun. 2021, 2, 100138. [Google Scholar] [CrossRef]
  7. Goh, R.M.V.; Pua, A.; Luro, F.; Ee, K.H.; Huang, Y.; Marchi, E.; Liu, S.Q.; Lassabliere, B.; Yu, B. Distinguishing Citrus Varieties Based on Genetic and Compositional Analyses. PLoS ONE 2022, 17, e0267007. [Google Scholar] [CrossRef]
  8. Conti, G.; Xoconostle-Cázares, B.; Marcelino-Pérez, G.; Hopp, H.E.; Reyes, C.A. Citrus Genetic Transformation: An Overview of the Current Strategies and Insights on the New Emerging Technologies. Front. Plant Sci. 2021, 12, 768197. [Google Scholar] [CrossRef]
  9. Wu, G.A.; Terol, J.; Ibanez, V.; López-García, A.; Pérez-Román, E.; Borredá, C.; Domingo, C.; Tadeo, F.R.; Carbonell-Caballero, J.; Alonso, R.; et al. Genomics of the Origin and Evolution of Citrus. Nature 2018, 554, 311–316. [Google Scholar] [CrossRef] [Green Version]
  10. Suri, S.; Singh, A.; Nema, P.K. Current Applications of Citrus Fruit Processing Waste: A Scientific Outlook. Appl. Food Res. 2022, 2, 100050. [Google Scholar] [CrossRef]
  11. Maqbool, Z.; Khalid, W.; Atiq, H.T.; Koraqi, H.; Javaid, Z.; Alhag, S.K.; Al-Shuraym, L.A.; Bader, D.M.D.; Almarzuq, M.; Afifi, M.; et al. Citrus Waste as Source of Bioactive Compounds: Extraction and Utilization in Health and Food Industry. Molecules 2023, 28, 1636. [Google Scholar] [CrossRef] [PubMed]
  12. Hussien Abou Baker, D.; Ahmed Ibrahim, E.; Abd El-Rhaman Salama, Z. Citrus Peels as a Source of Bioactive Compounds with Industrial and Therapeutic Applications. In Biochemistry; Badria, F.A., Ed.; IntechOpen: London, UK, 2022; Volume 26, ISBN 978-1-83969-346-5. [Google Scholar]
  13. Li, P.; Yao, X.; Zhou, Q.; Meng, X.; Zhou, T.; Gu, Q. Citrus Peel Flavonoid Extracts: Health-Beneficial Bioactivities and Regulation of Intestinal Microecology in Vitro. Front. Nutr. 2022, 9, 888745. [Google Scholar] [CrossRef] [PubMed]
  14. Matsuzaki, K.; Nakajima, A.; Guo, Y.; Ohizumi, Y. A Narrative Review of the Effects of Citrus Peels and Extracts on Human Brain Health and Metabolism. Nutrients 2022, 14, 1847. [Google Scholar] [CrossRef] [PubMed]
  15. Singh, B.; Singh, J.P.; Kaur, A.; Singh, N. Phenolic Composition, Antioxidant Potential and Health Benefits of Citrus Peel. Food Res. Int. 2020, 132, 109114. [Google Scholar] [CrossRef]
  16. Guo, C.; Shan, Y.; Yang, Z.; Zhang, L.; Ling, W.; Liang, Y.; Ouyang, Z.; Zhong, B.; Zhang, J. Chemical Composition, Antioxidant, Antibacterial, and Tyrosinase Inhibition Activity of Extracts from Newhall Navel Orange (Citrus sinensis Osbeck Cv. Newhall) Peel. J. Sci. Food Agric. 2020, 100, 2664–2674. [Google Scholar] [CrossRef]
  17. Ali, A.M.; Gabbar, M.A.; Abdel-Twab, S.M.; Fahmy, E.M.; Ebaid, H.; Alhazza, I.M.; Ahmed, O.M. Antidiabetic Potency, Antioxidant Effects, and Mode of Actions of Citrus reticulata Fruit Peel Hydroethanolic Extract, Hesperidin, and Quercetin in Nicotinamide/Streptozotocin-Induced Wistar Diabetic Rats. Oxidative Med. Cell. Longev. 2020, 2020, 1730492. [Google Scholar] [CrossRef]
  18. Lin, X.; Cao, S.; Sun, J.; Lu, D.; Zhong, B.; Chun, J. The Chemical Compositions, and Antibacterial and Antioxidant Activities of Four Types of Citrus Essential Oils. Molecules 2021, 26, 3412. [Google Scholar] [CrossRef]
  19. Shehata, M.G.; Awad, T.S.; Asker, D.; El Sohaimy, S.A.; Abd El- Aziz, N.M.; Youssef, M.M. Antioxidant and Antimicrobial Activities and UPLC-ESI-MS/MS Polyphenolic Profile of Sweet Orange Peel Extracts. Curr. Res. Food Sci. 2021, 4, 326–335. [Google Scholar] [CrossRef]
  20. Hou, H.-S.; Bonku, E.M.; Zhai, R.; Zeng, R.; Hou, Y.-L.; Yang, Z.-H.; Quan, C. Extraction of Essential Oil from Citrus Reticulate Blanco Peel and Its Antibacterial Activity against Cutibacterium acnes (Formerly Propionibacterium acnes). Heliyon 2019, 5, e02947. [Google Scholar] [CrossRef] [Green Version]
  21. Degirmenci, H.; Erkurt, H. Chemical Profile and Antioxidant Potency of Citrus aurantium L. Flower Extracts with Antibacterial Effect against Foodborne Pathogens in Rice Pudding. LWT 2020, 126, 109273. [Google Scholar] [CrossRef]
  22. Duletić-Laušević, S.; Oalđe, M.; Alimpić-Aradski, A. In vitro Evaluation of Antioxidant, Antineurodegenerative and Antidiabetic Activities of Ocimum basilicum L., Laurus nobilis L. Leaves and Citrus reticulata Blanco Peel Extracts. Lek. Sirovine 2019, 39, 60–68. [Google Scholar] [CrossRef] [Green Version]
  23. Ani, P.N.; Ochu, K.E. Anti-Diabetic, Anti-Hyperlipidemic and Hepatoprotective Potential of Shaddock (Citrus maxima) Peel Extract. Acta Sci. Pol. Technol. Aliment. 2020, 19, 271–278. [Google Scholar] [CrossRef] [PubMed]
  24. Benayad, O.; Bouhrim, M.; Tiji, S.; Kharchoufa, L.; Addi, M.; Drouet, S.; Hano, C.; Lorenzo, J.M.; Bendaha, H.; Bnouham, M.; et al. Phytochemical Profile, α-Glucosidase, and α-Amylase Inhibition Potential and Toxicity Evaluation of Extracts from Citrus aurantium (L) Peel, a Valuable By-Product from Northeastern Morocco. Biomolecules 2021, 11, 1555. [Google Scholar] [CrossRef] [PubMed]
  25. Jang, A.; Choi, G.; Kim, Y.; Lee, G.; Hyun, K. Neuroprotective properties of ethanolic extract of Citrus unshiu Markovich peel through NADPH oxidase 2 inhibition in chemotherapy-induced neuropathic pain animal model. Phytother. Res. 2021, 35, 6918–6931. [Google Scholar] [CrossRef]
  26. Furukawa, Y.; Okuyama, S.; Amakura, Y.; Sawamoto, A.; Nakajima, M.; Yoshimura, M.; Igase, M.; Fukuda, N.; Tamai, T.; Yoshida, T. Isolation and Characterization of Neuroprotective Components from Citrus Peel and Their Application as Functional Food. Chem. Pharm. Bull. 2021, 69, 2–10. [Google Scholar] [CrossRef]
  27. Eddin, L.B.; Jha, N.K.; Meeran, M.F.N.; Kesari, K.K.; Beiram, R.; Ojha, S. Neuroprotective Potential of Limonene and Limonene Containing Natural Products. Molecules 2021, 26, 4535. [Google Scholar] [CrossRef]
  28. Malik, A.; Najda, A.; Bains, A.; Nurzyńska-Wierdak, R.; Chawla, P. Characterization of Citrus nobilis Peel Methanolic Extract for Antioxidant, Antimicrobial, and Anti-Inflammatory Activity. Molecules 2021, 26, 4310. [Google Scholar] [CrossRef]
  29. Lala, M.; Modak, D.; Paul, S.; Sarkar, I.; Dutta, A.; Kumar, A.; Bhattacharjee, S.; Sen, A. Potent Bioactive Methanolic Extract of Wild Orange (Citrus macroptera Mont.) Shows Antioxidative, Anti-Inflammatory, and Antimicrobial Properties in in Vitro, in Vivo, and in Silico Studies. Bull. Natl. Res. Cent. 2020, 44, 81. [Google Scholar] [CrossRef]
  30. Roko, O.G.; Dougnon, V.; Hounkpatin, A.; Klotoé, J.R.; Baba-Moussa, L. Anti-Inflammatory, Analgesic and Antipyretic Properties of Ethanolic Extracts of Three Plants of Beninese’s Pharmacopoeia: Euphorbia hirta, Citrus aurantifolia and Heterotis rotundifolia. AJOB 2020, 8, 1–8. [Google Scholar] [CrossRef] [Green Version]
  31. Matuka, T.; Oyedeji, O.; Gondwe, M.; Oyedeji, A. Chemical Composition and In Vivo Anti-Inflammatory Activity of Essential Oils from Citrus sinensis (L.) Osbeck Growing in South Africa. J. Essent. Oil Bear. Plants 2020, 23, 638–647. [Google Scholar] [CrossRef]
  32. Ho, Y.; Suphrom, N.; Daowtak, K.; Potup, P.; Thongsri, Y.; Usuwanthim, K. Anticancer Effect of Citrus hystrix DC. Leaf Extract and Its Bioactive Constituents Citronellol and, Citronellal on the Triple Negative Breast Cancer MDA-MB-231 Cell Line. Pharmaceuticals 2020, 13, 476. [Google Scholar] [CrossRef]
  33. El-Kersh, D.M.; Ezzat, S.M.; Salama, M.M.; Mahrous, E.A.; Attia, Y.M.; Ahmed, M.S.; Elmazar, M.M. Anti-Estrogenic and Anti-Aromatase Activities of Citrus Peels Major Compounds in Breast Cancer. Sci. Rep. 2021, 11, 7121. [Google Scholar] [CrossRef]
  34. Ko, Y.-C.; Choi, H.S.; Liu, R.; Kim, J.-H.; Kim, S.-L.; Yun, B.-S.; Lee, D.-S. Inhibitory Effects of Tangeretin, a Citrus Peel-Derived Flavonoid, on Breast Cancer Stem Cell Formation through Suppression of Stat3 Signaling. Molecules 2020, 25, 2599. [Google Scholar] [CrossRef] [PubMed]
  35. Wen, S.; Sun, L.; An, R.; Zhang, W.; Xiang, L.; Li, Q.; Lai, X.; Huo, M.; Li, D.; Sun, S. A Combination of Citrus reticulata Peel and Black Tea Inhibits Migration and Invasion of Liver Cancer via PI3K/AKT and MMPs Signaling Pathway. Mol. Biol. Rep. 2020, 47, 507–519. [Google Scholar] [CrossRef] [PubMed]
  36. Caggia, C.; Palmeri, R.; Russo, N.; Timpone, R.; Randazzo, C.L.; Todaro, A.; Barbagallo, S. Employ of Citrus By-Product as Fat Replacer Ingredient for Bakery Confectionery Products. Front. Nutr. 2020, 7, 46. [Google Scholar] [CrossRef]
  37. Magalhães, D.; Vilas-Boas, A.A.; Teixeira, P.; Pintado, M. Functional Ingredients and Additives from Lemon By-Products and Their Applications in Food Preservation: A Review. Foods 2023, 12, 1095. [Google Scholar] [CrossRef]
  38. Taghavi Kevij, H.; Salami, M.; Mohammadian, M.; Khodadadi, M.; Emam-Djomeh, Z. Mechanical, Physical, and Bio-Functional Properties of Biopolymer Films Based on Gelatin as Affected by Enriching with Orange Peel Powder. Polym. Bull. 2021, 78, 4387–4402. [Google Scholar] [CrossRef]
  39. Bhandari, D.P.; Poudel, D.K.; Satyal, P.; Khadayat, K.; Dhami, S.; Aryal, D.; Chaudhary, P.; Ghimire, A.; Parajuli, N. Volatile Compounds and Antioxidant and Antimicrobial Activities of Selected Citrus Essential Oils Originated from Nepal. Molecules 2021, 26, 6683. [Google Scholar] [CrossRef]
  40. Manzur, M.; Luciardi, M.C.; Blázquez, M.A.; Alberto, M.R.; Cartagena, E.; Arena, M.E. Citrus sinensis Essential Oils an Innovative Antioxidant and Antipathogenic Dual Strategy in Food Preservation against Spoliage Bacteria. Antioxidants 2023, 12, 246. [Google Scholar] [CrossRef] [PubMed]
  41. Randazzo, W.; Jiménez-Belenguer, A.; Settanni, L.; Perdones, A.; Moschetti, M.; Palazzolo, E.; Guarrasi, V.; Vargas, M.; Germanà, M.A.; Moschetti, G. Antilisterial Effect of Citrus Essential Oils and Their Performance in Edible Film Formulations. Food Control 2016, 59, 750–758. [Google Scholar] [CrossRef] [Green Version]
  42. Sarma, R.; Adhikari, K.; Mahanta, S.; Khanikor, B. Insecticidal Activities of Citrus aurantifolia Essential Oil against Aedes aegypti (Diptera: Culicidae). Toxicol. Rep. 2019, 6, 1091–1096. [Google Scholar] [CrossRef]
  43. Asadollahi, A.; Khoobdel, M.; Zahraei-Ramazani, A.; Azarmi, S.; Mosawi, S.H. Effectiveness of Plant-Based Repellents against Different Anopheles Species: A Systematic Review. Malar. J. 2019, 18, 436. [Google Scholar] [CrossRef] [PubMed]
  44. Misni, N.; Mohamed Nor, Z.; Ahmad, R.; Ithnin, N.R.; Zasmy Unyah, N. Microencapsulation Preservation of the Stability and Efficacy of Citrus Grandis Oil-Based Repellent Formulation against Aedes aegypti during Storage. Molecules 2021, 26, 3599. [Google Scholar] [CrossRef] [PubMed]
  45. Changbunjong, T.; Boonmasawai, S.; Sungpradit, S.; Weluwanarak, T.; Leesombun, A. Contact and Fumigant Activities of Citrus aurantium Essential Oil against the Stable Fly Stomoxys calcitrans (Diptera: Muscidae). Plants 2022, 11, 1122. [Google Scholar] [CrossRef] [PubMed]
  46. Ostovar, E.; Khodayari, S.; Aramideh, S. Fumigant toxicity of essential oils of Citrus limon L., Citrus sinensis L. and Citrus aurantium L. peels on three important stored products pests. IJMAPR 2021, 37, 859–871. [Google Scholar] [CrossRef]
  47. Caballero, S.; Li, Y.O.; McClements, D.J.; Davidov-Pardo, G. Encapsulation and Delivery of Bioactive Citrus Pomace Polyphenols: A Review. Crit. Rev. Food Sci. Nutr. 2022, 62, 8028–8044. [Google Scholar] [CrossRef]
  48. Lippolis, T.; Cofano, M.; Caponio, G.R.; De Nunzio, V.; Notarnicola, M. Bioaccessibility and Bioavailability of Diet Polyphenols and Their Modulation of Gut Microbiota. IJMS 2023, 24, 3813. [Google Scholar] [CrossRef]
  49. Dadwal, V.; Gupta, M. Recent Developments in Citrus Bioflavonoid Encapsulation to Reinforce Controlled Antioxidant Delivery and Generate Therapeutic Uses: Review. Crit. Rev. Food Sci. Nutr. 2023, 63, 1187–1207. [Google Scholar] [CrossRef]
  50. Lu, X.; Zhang, H.; Zheng, T.; Liu, Q.; Zhu, J.; Huang, Q. Evaluation of Oral Bioaccessibility of Aged Citrus Peel Extracts Encapsulated in Different Lipid-Based Systems: A Comparison Study Using Different in Vitro Digestion Models. J. Agric. Food Chem. 2020, 68, 97–105. [Google Scholar] [CrossRef]
  51. Mahdi, A.A.; Al-Maqtari, Q.A.; Mohammed, J.K.; Al-Ansi, W.; Cui, H.; Lin, L. Enhancement of Antioxidant Activity, Antifungal Activity, and Oxidation Stability of Citrus reticulata Essential Oil Nanocapsules by Clove and Cinnamon Essential Oils. Food Biosci. 2021, 43, 101226. [Google Scholar] [CrossRef]
  52. Savic, I.M.; Savic Gajic, I.M.; Milovanovic, M.G.; Zerajic, S.; Gajic, D.G. Optimization of Ultrasound-Assisted Extraction and Encapsulation of Antioxidants from Orange Peels in Alginate-Chitosan Microparticles. Antioxidants 2022, 11, 297. [Google Scholar] [CrossRef] [PubMed]
  53. Dutta, T.; Ghosh, N.N.; Das, M.; Adhikary, R.; Mandal, V.; Chattopadhyay, A.P. Green Synthesis of Antibacterial and Antifungal Silver Nanoparticles Using Citrus limetta Peel Extract: Experimental and Theoretical Studies. J. Environ. Chem. Eng. 2020, 8, 104019. [Google Scholar] [CrossRef]
  54. Khane, Y.; Benouis, K.; Albukhaty, S.; Sulaiman, G.M.; Abomughaid, M.M.; Al Ali, A.; Aouf, D.; Fenniche, F.; Khane, S.; Chaibi, W.; et al. Green Synthesis of Silver Nanoparticles Using Aqueous Citrus limon Zest Extract: Characterization and Evaluation of Their Antioxidant and Antimicrobial Properties. Nanomaterials 2022, 12, 2013. [Google Scholar] [CrossRef]
  55. Jahan, I.; Erci, F.; Isildak, I. Facile Microwave-Mediated Green Synthesis of Non-Toxic Copper Nanoparticles Using Citrus sinensis Aqueous Fruit Extract and Their Antibacterial Potentials. J. Drug Deliv. Sci. Technol. 2021, 61, 102172. [Google Scholar] [CrossRef]
  56. Purushothaman, B.K.; Uma Maheswari, P.; Meera Sheriffa Begum, K.M. Magnetic Casein-CaFe2O4 Nanohybrid Carrier Conjugated with Progesterone for Enhanced Cytotoxicity of Citrus Peel Derived Hesperidin Drug towards Breast and Ovarian Cancer. Int. J. Biol. Macromol. 2020, 151, 293–304. [Google Scholar] [CrossRef] [PubMed]
  57. Hajizadeh, M.; Maleki, H.; Barani, M.; Fahmidehkar, M.; Mahmoodi, M.; Torkzadeh-Mahani, M. In Vitro Cytotoxicity Assay of D-Limonene Niosomes: An Efficient Nano-Carrier for Enhancing Solubility of Plant-Extracted Agents. Res. Pharma Sci. 2019, 14, 448. [Google Scholar] [CrossRef]
  58. Medina-Rodríguez, A.C.; Ávila-Sierra, A.; Ariza, J.J.; Guillamón, E.; Baños-Arjona, A.; Vicaria, J.M.; Jurado, E. Clean-in-Place Disinfection of Dual-Species Biofilm (Listeria and Pseudomonas) by a Green Antibacterial Product Made from Citrus Extract. Food Control 2020, 118, 107422. [Google Scholar] [CrossRef]
  59. Majumdar, M.; Khan, S.A.; Biswas, S.C.; Roy, D.N.; Panja, A.S.; Misra, T.K. In vitro and in silico Investigation of Anti-Biofilm Activity of Citrus macroptera Fruit Extract Mediated Silver Nanoparticles. J. Mol. Liq. 2020, 302, 112586. [Google Scholar] [CrossRef]
  60. Pradeep, M.; Kruszka, D.; Kachlicki, P.; Mondal, D.; Franklin, G. Uncovering the Phytochemical Basis and the Mechanism of Plant Extract-Mediated Eco-Friendly Synthesis of Silver Nanoparticles Using Ultra-Performance Liquid Chromatography Coupled with a Photodiode Array and High-Resolution Mass Spectrometry. ACS Sustain. Chem. Eng. 2022, 10, 562–571. [Google Scholar] [CrossRef]
  61. Singh, P.K.; Singh, J.; Medhi, T.; Kumar, A. Phytochemical Screening, Quantification, FT-IR Analysis, and In Silico Characterization of Potential Bio-Active Compounds Identified in HR-LC/MS Analysis of the Polyherbal Formulation from Northeast India. ACS Omega 2022, 7, 33067–33078. [Google Scholar] [CrossRef]
  62. Berk, Z. Citrus Fruit Processing; Academic Press: London, UK, 2016; ISBN 978-0-12-803133-9. [Google Scholar]
  63. Martínez-Nicolas, J.J.; Núñez-Gómez, D.; Lidón, V.; Martínez-Font, R.; Melgarejo, P.; Hernández, F.; Legua, P. Physico-Chemical Attributes of Lemon Fruits as Affected by Growing Substrate and Rootstock. Foods 2022, 11, 2487. [Google Scholar] [CrossRef]
  64. Diering, N.L.; Ulrich, A.; Scapini, T.; Müller, C.; Gasparetto, I.G.; Júnior, F.W.R.; Treichel, H.; Mossi, A.J. Microbial Natural Bioactive Formulations in Citrus Development. Biotechnol. Rep. 2022, 34, e00718. [Google Scholar] [CrossRef]
  65. Nieto, G.; Fernández-López, J.; Pérez-Álvarez, J.A.; Peñalver, R.; Ros-Berruezo, G.; Viuda-Martos, M. Valorization of Citrus Co-Products: Recovery of Bioactive Compounds and Application in Meat and Meat Products. Plants 2021, 10, 1069. [Google Scholar] [CrossRef] [PubMed]
  66. Andrade, M.A.; Barbosa, C.H.; Shah, M.A.; Ahmad, N.; Vilarinho, F.; Khwaldia, K.; Silva, A.S.; Ramos, F. Citrus By-Products: Valuable Source of Bioactive Compounds for Food Applications. Antioxidants 2022, 12, 38. [Google Scholar] [CrossRef]
  67. Casas Cardoso, L.; Cejudo Bastante, C.; Mantell Serrano, C.; Martínez de la Ossa, E.J. Application of Citrus By-Products in the Production of Active Food Packaging. Antioxidants 2022, 11, 738. [Google Scholar] [CrossRef] [PubMed]
  68. Petracek, P.D. Peel Morphology and Fruit Blemishes. In Citrus Flowering & Fruiting Short Course; Citrus Research and Education Center: Lake Alfred, FL, USA, 1997. [Google Scholar]
  69. Inthachat, W.; Temviriyanukul, P.; On-Nom, N.; Kanoongon, P.; Thangsiri, S.; Chupeerach, C.; Suttisansanee, U. Optimization of Phytochemical-Rich Citrus Maxima Albedo Extract Using Response Surface Methodology. Molecules 2023, 28, 4121. [Google Scholar] [CrossRef]
  70. Nomura, R.; Ohata, J.; Otsugu, M.; Okawa, R.; Naka, S.; Matsumoto-Nakano, M.; Nakano, K. Inhibitory Effects of Flavedo, Albedo, Fruits, and Leaves of Citrus unshiu Extracts on Streptococcus mutans. Arch. Oral. Biol. 2021, 124, 105056. [Google Scholar] [CrossRef]
  71. Ahmed, M.; Saeid, A. Citrus Fruits: Nutritive Value and Value-Added Products. In Citrus—Research, Development and Biotechnology; Sarwar Khan, M., Ahmad Khan, I., Eds.; IntechOpen: London, UK, 2021; ISBN 978-1-83968-723-5. [Google Scholar]
  72. Wang, J.; Hao, H.; Liu, R.; Ma, Q.; Xu, J.; Chen, F.; Cheng, Y.; Deng, X. Comparative Analysis of Surface Wax in Mature Fruits between Satsuma Mandarin (Citrus unshiu) and ‘Newhall’ Navel Orange (Citrus sinensis) from the Perspective of Crystal Morphology, Chemical Composition and Key Gene Expression. Food Chem. 2014, 153, 177–185. [Google Scholar] [CrossRef]
  73. Romero, P.; Lafuente, M.T. Abscisic Acid Deficiency Alters Epicuticular Wax Metabolism and Morphology That Leads to Increased Cuticle Permeability during Sweet Orange (Citrus sinensis) Fruit Ripening. Front. Plant Sci. 2020, 11, 594184. [Google Scholar] [CrossRef] [PubMed]
  74. Pashova, S. Application of Plant Waxes in Edible Coatings. Coatings 2023, 13, 911. [Google Scholar] [CrossRef]
  75. García-Coronado, H.; Tafolla-Arellano, J.C.; Hernández-Oñate, M.Á.; Burgara-Estrella, A.J.; Robles-Parra, J.M.; Tiznado-Hernández, M.E. Molecular Biology, Composition and Physiological Functions of Cuticle Lipids in Fleshy Fruits. Plants 2022, 11, 1133. [Google Scholar] [CrossRef] [PubMed]
  76. Lado, J.; Alós, E.; Manzi, M.; Cronje, P.J.R.; Gómez-Cadenas, A.; Rodrigo, M.J.; Zacarías, L. Light Regulation of Carotenoid Biosynthesis in the Peel of Mandarin and Sweet Orange Fruits. Front. Plant Sci. 2019, 10, 1288. [Google Scholar] [CrossRef] [Green Version]
  77. Kato, M. Mechanism of Carotenoid Accumulation in Citrus Fruit. J. Jpn. Soc. Hort. Sci. 2012, 81, 219–233. [Google Scholar] [CrossRef] [Green Version]
  78. Rodrigo, M.J.; Alquézar, B.; Alós, E.; Lado, J.; Zacarías, L. Biochemical Bases and Molecular Regulation of Pigmentation in the Peel of Citrus Fruit. Sci. Hortic. 2013, 163, 46–62. [Google Scholar] [CrossRef]
  79. Agarwal, P.; Sebghatollahi, Z.; Kamal, M.; Dhyani, A.; Shrivastava, A.; Singh, K.K.; Sinha, M.; Mahato, N.; Mishra, A.K.; Baek, K.-H. Citrus Essential Oils in Aromatherapy: Therapeutic Effects and Mechanisms. Antioxidants 2022, 11, 2374. [Google Scholar] [CrossRef]
  80. Bora, H.; Kamle, M.; Mahato, D.K.; Tiwari, P.; Kumar, P. Citrus Essential Oils (CEOs) and Their Applications in Food: An Overview. Plants 2020, 9, 357. [Google Scholar] [CrossRef] [Green Version]
  81. Ambrosio, C.M.S.; Diaz-Arenas, G.L.; Agudelo, L.P.A.; Stashenko, E.; Contreras-Castillo, C.J.; da Gloria, E.M. Chemical Composition and Antibacterial and Antioxidant Activity of a Citrus Essential Oil and Its Fractions. Molecules 2021, 26, 2888. [Google Scholar] [CrossRef]
  82. Mehl, F.; Marti, G.; Boccard, J.; Debrus, B.; Merle, P.; Delort, E.; Baroux, L.; Raymo, V.; Velazco, M.I.; Sommer, H.; et al. Differentiation of Lemon Essential Oil Based on Volatile and Non-Volatile Fractions with Various Analytical Techniques: A Metabolomic Approach. Food Chem. 2014, 143, 325–335. [Google Scholar] [CrossRef]
  83. Van den Bruinhorst, A.; Kouris, P.; Timmer, J.; De Croon, M.; Kroon, M. Exploring Orange Peel Treatment with Deep Eutectic Solvents and Diluted Organic Acids. Nat. Prod. Chem. Res. 2016, 4, 1–5. [Google Scholar] [CrossRef]
  84. Singhal, S.; Swami Hulle, N.R. Citrus Pectins: Structural Properties, Extraction Methods, Modifications and Applications in Food Systems—A Review. Appl. Food Res. 2022, 2, 100215. [Google Scholar] [CrossRef]
  85. Sharma, P.; Dadwal, V.; Rahmatkar, S.N.; Gupta, M.; Singh, D. Flavonoid Composition and Antioxidant Efficacy of Citrus Peels: An Integrated in Vitro and in Silico Approach toward Potential Neuroprotective Agents. J. Sci. Ind. Res. 2022, 81, 445–454. [Google Scholar]
  86. Chandel, V.; Biswas, D.; Roy, S.; Vaidya, D.; Verma, A.; Gupta, A. Current Advancements in Pectin: Extraction, Properties and Multifunctional Applications. Foods 2022, 11, 2683. [Google Scholar] [CrossRef] [PubMed]
  87. Drincovich, M.F.; Voll, L.M.; Maurino, V.G. Editorial: On the Diversity of Roles of Organic Acids. Front. Plant Sci. 2016, 7, 1592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Bampidis, V.A.; Robinson, P.H. Citrus By-Products as Ruminant Feeds: A Review. Anim. Feed. Sci. Technol. 2006, 128, 175–217. [Google Scholar] [CrossRef]
  89. Miles, E.A.; Calder, P.C. Effects of Citrus Fruit Juices and Their Bioactive Components on Inflammation and Immunity: A Narrative Review. Front. Immunol. 2021, 12, 712608. [Google Scholar] [CrossRef] [PubMed]
  90. Addi, M.; Elbouzidi, A.; Abid, M.; Tungmunnithum, D.; Elamrani, A.; Hano, C. An Overview of Bioactive Flavonoids from Citrus Fruits. Appl. Sci. 2021, 12, 29. [Google Scholar] [CrossRef]
  91. Saini, R.K.; Ranjit, A.; Sharma, K.; Prasad, P.; Shang, X.; Gowda, K.G.M.; Keum, Y.-S. Bioactive Compounds of Citrus Fruits: A Review of Composition and Health Benefits of Carotenoids, Flavonoids, Limonoids, and Terpenes. Antioxidants 2022, 11, 239. [Google Scholar] [CrossRef]
  92. Costanzo, G.; Iesce, M.R.; Naviglio, D.; Ciaravolo, M.; Vitale, E.; Arena, C. Comparative Studies on Different Citrus Cultivars: A Revaluation of Waste Mandarin Components. Antioxidants 2020, 9, 517. [Google Scholar] [CrossRef]
  93. Bocco, A.; Cuvelier, M.-E.; Richard, H.; Berset, C. Antioxidant Activity and Phenolic Composition of Citrus Peel and Seed Extracts. J. Agric. Food Chem. 1998, 46, 2123–2129. [Google Scholar] [CrossRef]
  94. Zayed, A.; Badawy, M.T.; Farag, M.A. Valorization and Extraction Optimization of Citrus Seeds for Food and Functional Food Applications. Food Chem. 2021, 355, 129609. [Google Scholar] [CrossRef]
  95. Ahmed, R.H.A.; Mariod, A.A. Citrus Seeds Fixed Oil, Composition and Its Biological Activities. In Multiple Biological Activities of Unconventional Seed Oils; Elsevier: Amsterdam, The Netherlands, 2022; pp. 229–235. ISBN 978-0-12-824135-6. [Google Scholar]
  96. Sharma, P.; Vishvakarma, R.; Gautam, K.; Vimal, A.; Kumar Gaur, V.; Farooqui, A.; Varjani, S.; Younis, K. Valorization of Citrus Peel Waste for the Sustainable Production of Value-Added Products. Bioresour. Technol. 2022, 351, 127064. [Google Scholar] [CrossRef] [PubMed]
  97. Chavan, P.; Singh, A.K.; Kaur, G. Recent Progress in the Utilization of Industrial Waste and By-products of Citrus Fruits: A Review. J. Food Process Eng. 2018, 41, e12895. [Google Scholar] [CrossRef]
  98. Martínez-Abad, A.; Ramos, M.; Hamzaoui, M.; Kohnen, S.; Jiménez, A.; Garrigós, M.C. Optimisation of Sequential Microwave-Assisted Extraction of Essential Oil and Pigment from Lemon Peels Waste. Foods 2020, 9, 1493. [Google Scholar] [CrossRef]
  99. Tunç, M.T.; Odabaş, H.İ. Single-Step Recovery of Pectin and Essential Oil from Lemon Waste by Ohmic Heating Assisted Extraction/Hydrodistillation: A Multi-Response Optimization Study. Innov. Food Sci. Emerg. Technol. 2021, 74, 102850. [Google Scholar] [CrossRef]
  100. Sandhu, H.K.; Sinha, P.; Emanuel, N.; Kumar, N.; Sami, R.; Khojah, E.; Al-Mushhin, A.A.M. Effect of Ultrasound-Assisted Pretreatment on Extraction Efficiency of Essential Oil and Bioactive Compounds from Citrus Waste By-Products. Separations 2021, 8, 244. [Google Scholar] [CrossRef]
  101. Teigiserova, D.A.; Tiruta-Barna, L.; Ahmadi, A.; Hamelin, L.; Thomsen, M. A Step Closer to Circular Bioeconomy for Citrus Peel Waste: A Review of Yields and Technologies for Sustainable Management of Essential Oils. J. Environ. Manag. 2021, 280, 111832. [Google Scholar] [CrossRef]
  102. Mahato, N.; Sinha, M.; Sharma, K.; Koteswararao, R.; Cho, M.H. Modern Extraction and Purification Techniques for Obtaining High Purity Food-Grade Bioactive Compounds and Value-Added Co-Products from Citrus Wastes. Foods 2019, 8, 523. [Google Scholar] [CrossRef] [Green Version]
  103. Panwar, D.; Panesar, P.S.; Chopra, H.K. Recent Trends on the Valorization Strategies for the Management of Citrus By-Products. Food Rev. Int. 2021, 37, 91–120. [Google Scholar] [CrossRef]
  104. Hwang, H.-J.; Kim, H.-J.; Ko, M.-J.; Chung, M.-S. Recovery of Hesperidin and Narirutin from Waste Citrus unshiu Peel Using Subcritical Water Extraction Aided by Pulsed Electric Field Treatment. Food Sci. Biotechnol. 2021, 30, 217–226. [Google Scholar] [CrossRef]
  105. Idamokoro, E.M.; Hosu, Y.S. Out-Look on Worldwide Trends of Related Studies on Citrus Waste as Feed for Livestock Production: A Scientometric Analysis. Front. Res. Metr. Anal. 2022, 7, 869974. [Google Scholar] [CrossRef]
  106. Mamma, D.; Christakopoulos, P. Biotransformation of Citrus By-Products into Value Added Products. Waste Biomass Valor. 2014, 5, 529–549. [Google Scholar] [CrossRef]
  107. Yun, D.; Liu, J. Recent Advances on the Development of Food Packaging Films Based on Citrus Processing Wastes: A Review. J. Agric. Food Res. 2022, 9, 100316. [Google Scholar] [CrossRef]
  108. Meydanju, N.; Pirsa, S.; Farzi, J. Biodegradable Film Based on Lemon Peel Powder Containing Xanthan Gum and TiO2–Ag Nanoparticles: Investigation of Physicochemical and Antibacterial Properties. Polym. Test. 2022, 106, 107445. [Google Scholar] [CrossRef]
  109. Spigno, G.; Donsì, F.; Amendola, D.; Sessa, M.; Ferrari, G.; De Faveri, D.M. Nanoencapsulation Systems to Improve Solubility and Antioxidant Efficiency of a Grape Marc Extract into Hazelnut Paste. J. Food Eng. 2013, 114, 207–214. [Google Scholar] [CrossRef]
  110. Jones, D.; Caballero, S.; Davidov-Pardo, G. Bioavailability of Nanotechnology-Based Bioactives and Nutraceuticals. In Advances in Food and Nutrition Research; Elsevier: Amsterdam, The Netherlands, 2019; Volume 88, pp. 235–273. ISBN 978-0-12-816073-2. [Google Scholar]
  111. Todorović, A.; Šturm, L.; Salević-Jelić, A.; Lević, S.; Osojnik Črnivec, I.G.; Prislan, I.; Skrt, M.; Bjeković, A.; Poklar Ulrih, N.; Nedović, V. Encapsulation of Bilberry Extract with Maltodextrin and Gum Arabic by Freeze-Drying: Formulation, Characterisation, and Storage Stability. Processes 2022, 10, 1991. [Google Scholar] [CrossRef]
  112. Papoutsis, K.; Golding, J.; Vuong, Q.; Pristijono, P.; Stathopoulos, C.; Scarlett, C.; Bowyer, M. Encapsulation of Citrus By-Product Extracts by Spray-Drying and Freeze-Drying Using Combinations of Maltodextrin with Soybean Protein and ι-Carrageenan. Foods 2018, 7, 115. [Google Scholar] [CrossRef] [Green Version]
  113. Mahato, N.; Sharma, K.; Sinha, M.; Dhyani, A.; Pathak, B.; Jang, H.; Park, S.; Pashikanti, S.; Cho, S. Biotransformation of Citrus Waste-I: Production of Biofuel and Valuable Compounds by Fermentation. Processes 2021, 9, 220. [Google Scholar] [CrossRef]
  114. Oberoi, H.S.; Vadlani, P.V.; Nanjundaswamy, A.; Bansal, S.; Singh, S.; Kaur, S.; Babbar, N. Enhanced Ethanol Production from Kinnow Mandarin (Citrus reticulata) Waste via a Statistically Optimized Simultaneous Saccharification and Fermentation Process. Bioresour. Technol. 2011, 102, 1593–1601. [Google Scholar] [CrossRef]
  115. Wu, W.; Jiang, B.; Liu, R.; Han, Y.; Fang, X.; Mu, H.; Farag, M.A.; Simal-Gandara, J.; Prieto, M.A.; Chen, H.; et al. Structures and Functions of Cuticular Wax in Postharvest Fruit and Its Regulation: A Comprehensive Review with Future Perspectives. Engineering 2023, 23, 118–129. [Google Scholar] [CrossRef]
  116. Zhu, M.; Ji, J.; Wang, M.; Zhao, M.; Yin, Y.; Kong, J.; Liu, M.; Li, Y.-F. Cuticular Wax of Mandarin Fruit Promotes Conidial Germination and Germ Tube Elongation, and Impairs Colony Expansion of the Green Mold Pathogen, Penicillium Digitatum. Postharvest Biol. Technol. 2020, 169, 111296. [Google Scholar] [CrossRef]
  117. Multari, S.; Licciardello, C.; Caruso, M.; Martens, S. Monitoring the Changes in Phenolic Compounds and Carotenoids Occurring during Fruit Development in the Tissues of Four Citrus Fruits. Food Res. Int. 2020, 134, 109228. [Google Scholar] [CrossRef] [PubMed]
  118. Mavrommatis, A.; Zografaki, M.-E.; Marka, S.; Myrtsi, E.D.; Giamouri, E.; Christodoulou, C.; Evergetis, E.; Iliopoulos, V.; Koulocheri, S.D.; Moschopoulou, G.; et al. Effect of a Carotenoid Extract from Citrus reticulata By-Products on the Immune-Oxidative Status of Broilers. Antioxidants 2022, 11, 144. [Google Scholar] [CrossRef] [PubMed]
  119. Barman, K.; Chowdhury, D.; Baruah, P.K. Development of Β-carotene Loaded Nanoemulsion Using the Industrial Waste of Orange (Citrus reticulate) Peel to Improve in Vitro Bioaccessibility of Carotenoids and Use as Natural Food Colorant. J. Food Process Preserv. 2020, 44, e14429. [Google Scholar] [CrossRef]
  120. Sedeek, M.S.; Al-Mahallawi, A.M.; Hussien, R.A.A.; Ali, A.M.A.; Naguib, I.A.; Mansour, M.K. Hexosomal Dispersion: A Nano-Based Approach to Boost the Antifungal Potential of Citrus Essential Oils against Plant Fungal Pathogens. Molecules 2021, 26, 6284. [Google Scholar] [CrossRef]
  121. Abdel-Bar, H.M.; Khater, S.E.; Ghorab, D.M.; Al-mahallawi, A.M. Hexosomes as Efficient Platforms for Possible Fluoxetine Hydrochloride Repurposing with Improved Cytotoxicity against HepG2 Cells. ACS Omega 2020, 5, 26697–26709. [Google Scholar] [CrossRef]
  122. Feng, K.; Zhu, X.; Liu, G.; Kan, Q.; Chen, T.; Chen, Y.; Cao, Y. Dietary Citrus Peel Essential Oil Ameliorates Hypercholesterolemia and Hepatic Steatosis by Modulating Lipid and Cholesterol Homeostasis. Food Funct. 2020, 11, 7217–7230. [Google Scholar] [CrossRef]
  123. Asikin, Y.; Shimizu, K.; Iwasaki, H.; Oku, H.; Wada, K. Stress Amelioration and Anti-Inflammatory Potential of Shiikuwasha (Citrus depressa Hayata) Essential Oil, Limonene, and γ-Terpinene. J. Food Drug Anal. 2022, 30, 454–465. [Google Scholar] [CrossRef]
  124. Torimiro, N.; Adegun, B.R.; Abioye, O.E.; Omole, R.K. Antibacterial Activity of Essential Oil from Citrus aurantifolia (Christm.) Swingle Peels against Multidrug-Resistant Bacterial Isolates. AiM 2020, 10, 214–223. [Google Scholar] [CrossRef]
  125. Sreepian, A.; Popruk, S.; Nutalai, D.; Phutthanu, C.; Sreepian, P.M. Antibacterial Activities and Synergistic Interaction of Citrus Essential Oils and Limonene with Gentamicin against Clinically Isolated Methicillin-Resistant Staphylococcus aureus. Sci. World J. 2022, 2022, 8418287. [Google Scholar] [CrossRef]
  126. Guo, J.; Gao, Z.; Li, G.; Fu, F.; Liang, Z.; Zhu, H.; Shan, Y. Antimicrobial and Antibiofilm Efficacy and Mechanism of Essential Oil from Citrus Changshan-Huyou Y. B. Chang against Listeria monocytogenes. Food Control 2019, 105, 256–264. [Google Scholar] [CrossRef]
  127. Restuccia, C.; Oliveri Conti, G.; Zuccarello, P.; Parafati, L.; Cristaldi, A.; Ferrante, M. Efficacy of Different Citrus Essential Oils to Inhibit the Growth and B1 Aflatoxin Biosynthesis of Aspergillus flavus. Environ. Sci. Pollut. Res. 2019, 26, 31263–31272. [Google Scholar] [CrossRef] [PubMed]
  128. Bava, R.; Castagna, F.; Piras, C.; Palma, E.; Cringoli, G.; Musolino, V.; Lupia, C.; Perri, M.R.; Statti, G.; Britti, D.; et al. In Vitro Evaluation of Acute Toxicity of Five Citrus Spp. Essential Oils towards the Parasitic Mite Varroa destructor. Pathogens 2021, 10, 1182. [Google Scholar] [CrossRef] [PubMed]
  129. Oyedeji, A.O.; Okunowo, W.O.; Osuntoki, A.A.; Olabode, T.B.; Ayo-folorunso, F. Insecticidal and Biochemical Activity of Essential Oil from Citrus sinensis Peel and Constituents on Callosobrunchus maculatus and Sitophilus zeamais. Pestic. Biochem. Physiol. 2020, 168, 104643. [Google Scholar] [CrossRef]
  130. Visakh, N.U.; Pathrose, B.; Narayanankutty, A.; Alfarhan, A.; Ramesh, V. Utilization of Pomelo (Citrus maxima) Peel Waste into Bioactive Essential Oils: Chemical Composition and Insecticidal Properties. Insects 2022, 13, 480. [Google Scholar] [CrossRef] [PubMed]
  131. Fadilah, N.Q.; Jittmittraphap, A.; Leaungwutiwong, P.; Pripdeevech, P.; Dhanushka, D.; Mahidol, C.; Ruchirawat, S.; Kittakoop, P. Virucidal Activity of Essential Oils From Citrus x aurantium L. Against Influenza A Virus H1N1: Limonene as a Potential Household Disinfectant Against Virus. Nat. Product. Commun. 2022, 17, 1934578X2110727. [Google Scholar] [CrossRef]
  132. Asaad, G.; Abdelhameed, M.; Elraey, M.; Roshdy, W.; Elgamal, A.; Moamen, Y. Citrus clementine Peels Essential Oil Exhibited Anti-SARS-CoV-2 and Its Modulatory Effect against Cytokine Storm: Evidence from in Vitro and in Silico Studies. Egypt. J. Chem. 2022, 10, 419–427. [Google Scholar] [CrossRef]
  133. Uçar, Y. Antioxidant Effect of Nanoemulsions Based on Citrus Peel Essential Oils: Prevention of Lipid Oxidation in Trout. Eur. J. Lipid Sci. Technol. 2020, 122, 1900405. [Google Scholar] [CrossRef]
  134. Kosker, A.R. The Effects of Nanoemulsions Based on Citrus Essential Oils on the Formation of Biogenic Amines in Trout Fillets Stored at 4 ± 2 °C. J. Food Saf. 2020, 40, e12762. [Google Scholar] [CrossRef]
  135. Lin, L.-Y.; Chuang, C.-H.; Chen, H.-C.; Yang, K.-M. Lime (Citrus aurantifolia (Christm.) Swingle) Essential Oils: Volatile Compounds, Antioxidant Capacity, and Hypolipidemic Effect. Foods 2019, 8, 398. [Google Scholar] [CrossRef] [Green Version]
  136. Magalhães, M.L.; Ionta, M.; Ferreira, G.Á.; Campidelli, M.L.L.; Nelson, D.L.; Ferreira, V.R.F.; Rezende, D.A.D.C.S.; Cardoso, M.D.G. Biological Activities of the Essential Oil from the Moro Orange Peel (Citrus sinensis (L.) Osbeck). Flavour. Fragr. J. 2020, 35, 294–301. [Google Scholar] [CrossRef]
  137. Viana, M.D.M.; Silva Neto, G.J.D.; Lima, A.A.D.; Leite, A.B.; Souza, I.T.; Santana, A.E.G.; Campesatto, E.A.; Moreira, M.S.A. Citrus limon (L.) Burm f. Essential Oil Has Anxiolytic and Sedative Properties by Modulating GABAA-Receptors. Braz. Arch. Biol. Technol. 2020, 63, e20200206. [Google Scholar] [CrossRef]
  138. Kwangjai, J.; Cheaha, D.; Manor, R.; Sa-ih, N.; Samerphob, N.; Issuriya, A.; Wattanapiromsakul, C.; Kumarnsit, E. Modification of Brain Waves and Sleep Parameters by Citrus reticulata Blanco. Cv. Sai-Nam-Phueng Essential Oil. Biomed. J. 2021, 44, 727–738. [Google Scholar] [CrossRef] [PubMed]
  139. Bi, W.; Zhou, J.; Zhao, L.; Wang, C.; Wu, W.; Zhang, L.; Ji, B.; Zhang, N.; Zhou, F. Preventive Effect of Different Citrus Essential Oils on Primary Dysmenorrhea: In Vivo and in Vitro Study. Food Biosci. 2021, 42, 101135. [Google Scholar] [CrossRef]
  140. Ayuningtyas, N.F.; Hendarti, H.T.; Soebadi, B.; Condro Surboyo, M.D.; Hadi, P.; Ganesha, R.; Ernawati, D.S.; Marsetyo, R.I. Expression of VEGF and CD-31 in Traumatic Ulcer of Diabetic Wistar Rats after Application of Citrus limon Peel Essential Oil. J. Oral. Biol. Craniofacial Res. 2023, 13, 380–385. [Google Scholar] [CrossRef] [PubMed]
  141. Disson, O.; Lecuit, M. Targeting of the Central Nervous System by Listeria monocytogenes. Virulence 2012, 3, 213–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Freitas, C.M.P.; Coimbra, J.S.R.; Souza, V.G.L.; Sousa, R.C.S. Structure and Applications of Pectin in Food, Biomedical, and Pharmaceutical Industry: A Review. Coatings 2021, 11, 922. [Google Scholar] [CrossRef]
  143. Minzanova, S.; Mironov, V.; Arkhipova, D.; Khabibullina, A.; Mironova, L.; Zakirova, Y.; Milyukov, V. Biological Activity and Pharmacological Application of Pectic Polysaccharides: A Review. Polymers 2018, 10, 1407. [Google Scholar] [CrossRef] [Green Version]
  144. Míguez, B.; Gómez, B.; Gullón, P.; Gullón, B.; Alonso, J.L. Pectic Oligosaccharides and Other Emerging Prebiotics. In Probiotics and Prebiotics in Human Nutrition and Health; Rao, V., Rao, L.G., Eds.; InTech: Houston, TX, USA, 2016; ISBN 978-953-51-2475-7. [Google Scholar]
  145. Zhang, S.; Hu, H.; Wang, L.; Liu, F.; Pan, S. Preparation and Prebiotic Potential of Pectin Oligosaccharides Obtained from Citrus Peel Pectin. Food Chem. 2018, 244, 232–237. [Google Scholar] [CrossRef]
  146. Lee, T.; Chang, Y.H. Structural, Physicochemical, and in-Vitro Release Properties of Hydrogel Beads Produced by Oligochitosan and de-Esterified Pectin from Yuzu (Citrus junos) Peel as a Quercetin Delivery System for Colon Target. Food Hydrocoll. 2020, 108, 106086. [Google Scholar] [CrossRef]
  147. Jacob, E.M.; Borah, A.; Jindal, A.; Pillai, S.C.; Yamamoto, Y.; Maekawa, T.; Kumar, D.N.S. Synthesis and Characterization of Citrus-Derived Pectin Nanoparticles Based on Their Degree of Esterification. J. Mater. Res. 2020, 35, 1514–1522. [Google Scholar] [CrossRef]
  148. Han, H.; Song, K.B. Antioxidant Activities of Mandarin (Citrus unshiu) Peel Pectin Films Containing Sage (Salvia officinalis) Leaf Extract. Int. J. Food Sci. Technol. 2020, 55, 3173–3181. [Google Scholar] [CrossRef]
  149. Al-Enazi, N.M.; Alsamhary, K.; Ameen, F. Evaluation of Citrus Pectin Capped Copper Sulfide Nanoparticles against Candidiasis Causing Candida Biofilms. Environ. Res. 2023, 225, 115599. [Google Scholar] [CrossRef] [PubMed]
  150. Mao, G.; Li, S.; Orfila, C.; Shen, X.; Zhou, S.; Linhardt, R.J.; Ye, X.; Chen, S. Depolymerized RG-I-Enriched Pectin from Citrus Segment Membranes Modulates Gut Microbiota, Increases SCFA Production, and Promotes the Growth of Bifidobacterium spp., Lactobacillus spp. and Faecalibaculum spp. Food Funct. 2019, 10, 7828–7843. [Google Scholar] [CrossRef] [PubMed]
  151. Rajulapati, V.; Dhillon, A.; Goyal, A. Enzymatically Produced Pectic-Oligosaccharides from Fruit Waste of Citrus reticulata (Mandarin) Peels Display Cytotoxicity against Colon Cancer Cells. Bioresour. Technol. Rep. 2021, 15, 100740. [Google Scholar] [CrossRef]
  152. Ishisono, K.; Mano, T.; Yabe, T.; Kitaguchi, K. Dietary Fiber Pectin Ameliorates Experimental Colitis in a Neutral Sugar Side Chain-Dependent Manner. Front. Immunol. 2019, 10, 2979. [Google Scholar] [CrossRef] [Green Version]
  153. Meneguzzo, F.; Ciriminna, R.; Zabini, F.; Pagliaro, M. Review of Evidence Available on Hesperidin-Rich Products as Potential Tools against COVID-19 and Hydrodynamic Cavitation-Based Extraction as a Method of Increasing Their Production. Processes 2020, 8, 549. [Google Scholar] [CrossRef]
  154. Hu, H.; Zhang, S.; Liu, F.; Zhang, P.; Muhammad, Z.; Pan, S. Role of the Gut Microbiota and Their Metabolites in Modulating the Cholesterol-Lowering Effects of Citrus Pectin Oligosaccharides in C57BL/6 Mice. J. Agric. Food Chem. 2019, 67, 11922–11930. [Google Scholar] [CrossRef]
  155. Flori, L.; Albanese, L.; Calderone, V.; Meneguzzo, F.; Pagliaro, M.; Ciriminna, R.; Zabini, F.; Testai, L. Cardioprotective Effects of Grapefruit IntegroPectin Extracted via Hydrodynamic Cavitation from By-Products of Citrus Fruits Industry: Role of Mitochondrial Potassium Channels. Foods 2022, 11, 2799. [Google Scholar] [CrossRef]
  156. Rojas-Lema, S.; Torres-Giner, S.; Quiles-Carrillo, L.; Gomez-Caturla, J.; Garcia-Garcia, D.; Balart, R. On the Use of Phenolic Compounds Present in Citrus Fruits and Grapes as Natural Antioxidants for Thermo-Compressed Bio-Based High-Density Polyethylene Films. Antioxidants 2020, 10, 14. [Google Scholar] [CrossRef]
  157. Kaurinovic, B.; Vastag, D. Flavonoids and Phenolic Acids as Potential Natural Antioxidants. In Antioxidants; Shalaby, E., Ed.; IntechOpen: London, UK, 2019; ISBN 978-1-78923-919-5. [Google Scholar]
  158. Tripoli, E.; Guardia, M.L.; Giammanco, S.; Majo, D.D.; Giammanco, M. Citrus Flavonoids: Molecular Structure, Biological Activity and Nutritional Properties: A Review. Food Chem. 2007, 104, 466–479. [Google Scholar] [CrossRef]
  159. Shimamura, Y.; Sei, S.; Nomura, S.; Masuda, S. Protective Effects of Dried Mature Citrus unshiu Peel (Chenpi) and Hesperidin on Aspirin-Induced Oxidative Damage. J. Clin. Biochem. Nutr. 2021, 68, 149–155. [Google Scholar] [CrossRef] [PubMed]
  160. Cryer, B.; Mahaffey, K. Gastrointestinal Ulcers, Role of Aspirin, and Clinical Outcomes: Pathobiology, Diagnosis, and Treatment. JMDH 2014, 7, 137–146. [Google Scholar] [CrossRef] [Green Version]
  161. Sharaf, M.; Arif, M.; Hamouda, H.I.; Khan, S.; Abdalla, M.; Shabana, S.; Rozan, H.E.; Khan, T.U.; Chi, Z.; Liu, C. Preparation, Urease Inhibition Mechanisms, and Anti-Helicobacter pylori Activities of Hesperetin-7-Rhamnoglucoside. Curr. Res. Microb. Sci. 2022, 3, 100103. [Google Scholar] [CrossRef] [PubMed]
  162. Chandra Babu, T.M.; Rajesh, S.S.; Bhaskar, B.V.; Devi, S.; Rammohan, A.; Sivaraman, T.; Rajendra, W. Molecular Docking, Molecular Dynamics Simulation, Biological Evaluation and 2D QSAR Analysis of Flavonoids from Syzygium alternifolium as Potent Anti-Helicobacter pylori Agents. RSC Adv. 2017, 7, 18277–18292. [Google Scholar] [CrossRef] [Green Version]
  163. Wang, Y.; Chen, Y.; Zhang, H.; Chen, J.; Cao, J.; Chen, Q.; Li, X.; Sun, C. Polymethoxyflavones from Citrus Inhibited Gastric Cancer Cell Proliferation through Inducing Apoptosis by Upregulating RARβ, Both in Vitro and in Vivo. Food Chem. Toxicol. 2020, 146, 111811. [Google Scholar] [CrossRef]
  164. Tajaldini, M.; Samadi, F.; Khosravi, A.; Ghasemnejad, A.; Asadi, J. Protective and Anticancer Effects of Orange Peel Extract and Naringin in Doxorubicin Treated Esophageal Cancer Stem Cell Xenograft Tumor Mouse Model. Biomed. Pharmacother. 2020, 121, 109594. [Google Scholar] [CrossRef]
  165. Deenonpoe, R.; Prayong, P.; Thippamom, N.; Meephansan, J.; Na-Bangchang, K. Anti-Inflammatory Effect of Naringin and Sericin Combination on Human Peripheral Blood Mononuclear Cells (HPBMCs) from Patient with Psoriasis. BMC Complement. Altern. Med. 2019, 19, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Wang, Y.; Ji, S.; Zang, W.; Wang, N.; Cao, J.; Li, X.; Sun, C. Identification of Phenolic Compounds from a Unique Citrus Species, Finger Lime (Citrus australasica) and Their Inhibition of LPS-Induced NO-Releasing in BV-2 Cell Line. Food Chem. Toxicol. 2019, 129, 54–63. [Google Scholar] [CrossRef]
  167. Smeriglio, A.; Cornara, L.; Denaro, M.; Barreca, D.; Burlando, B.; Xiao, J.; Trombetta, D. Antioxidant and Cytoprotective Activities of an Ancient Mediterranean Citrus (Citrus lumia Risso) Albedo Extract: Microscopic Observations and Polyphenol Characterization. Food Chem. 2019, 279, 347–355. [Google Scholar] [CrossRef] [PubMed]
  168. Kusumawati, I.G.A.W.; Putra, I.M.W.A.; Yogeswara, I.B.A. In Vitro ACE Inhibitory Activity and Bioactive Compounds of Aqueous Extract of Citrus amblycarpa. Trad. Med. J. 2021, 26, 117. [Google Scholar] [CrossRef]
  169. Wang, Y.; Zang, W.; Ji, S.; Cao, J.; Sun, C. Three Polymethoxyflavones Purified from Ougan (Citrus reticulata Cv. Suavissima) Inhibited LPS-Induced NO Elevation in the Neuroglia BV-2 Cell Line via the JAK2/STAT3 Pathway. Nutrients 2019, 11, 791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Al-Saman, M.A.; Abdella, A.; Mazrou, K.E.; Tayel, A.A.; Irmak, S. Antimicrobial and Antioxidant Activities of Different Extracts of the Peel of Kumquat (Citrus japonica Thunb). Food Meas. 2019, 13, 3221–3229. [Google Scholar] [CrossRef]
  171. Ahmed, O.M.; Fahim, H.I.; Ahmed, H.Y.; Al-Muzafar, H.M.; Ahmed, R.R.; Amin, K.A.; El-Nahass, E.-S.; Abdelazeem, W.H. The Preventive Effects and the Mechanisms of Action of Navel Orange Peel Hydroethanolic Extract, Naringin, and Naringenin in N-Acetyl-p-Aminophenol-Induced Liver Injury in Wistar Rats. Oxidative Med. Cell. Longev. 2019, 2019, 2745352. [Google Scholar] [CrossRef] [Green Version]
  172. Sung, J.; Suh, J.H.; Wang, Y. Effects of Heat Treatment of Mandarin Peel on Flavonoid Profiles and Lipid Accumulation in 3T3-L1 Adipocytes. J. Food Drug Anal. 2019, 27, 729–735. [Google Scholar] [CrossRef]
  173. Karthikeyan, A.; Kim, H.H.; Preethi, V.; Moniruzzaman, M.; Lee, K.H.; Kalaiselvi, S.; Kim, G.S.; Min, T. Assessment of Anti-Inflammatory and Antioxidant Effects of Citrus unshiu Peel (CUP) Flavonoids on LPS-Stimulated RAW 264.7 Cells. Plants 2021, 10, 2209. [Google Scholar] [CrossRef] [PubMed]
  174. Fathy, H.M.; Abd El-Maksoud, A.A.; Cheng, W.; Elshaghabee, F.M.F. Value-Added Utilization of Citrus Peels in Improving Functional Properties and Probiotic Viability of Acidophilus-bifidus-thermophilus (ABT)-Type Synbiotic Yoghurt during Cold Storage. Foods 2022, 11, 2677. [Google Scholar] [CrossRef]
  175. Babu, V.; Binwal, M.; Ranjana; Kumari, R.; Sen, S.; Kumar, A.; Mugale, M.N.; Shanker, K.; Kumar, N.; Bawankule, D.U. Hesperidin-rich Ethanol Extract from Waste Peels of Citrus limetta Mitigates Rheumatoid Arthritis and Related Complications. Phytother. Res. 2021, 35, 3325–3336. [Google Scholar] [CrossRef]
  176. Ben Abdallah, M.; Chadni, M.; M’hiri, N.; Brunissen, F.; Rokbeni, N.; Allaf, K.; Besombes, C.; Ioannou, I.; Boudhrioua, N. Intensifying Effect of Instant Controlled Pressure Drop (DIC) Pre-Treatment on Hesperidin Recovery from Orange Byproducts: In Vitro Antioxidant and Antidiabetic Activities of the Extracts. Molecules 2023, 28, 1858. [Google Scholar] [CrossRef] [PubMed]
  177. Nakashima, M.; Goda, N.; Tenno, T.; Kotake, A.; Inotsume, Y.; Amaya, M.; Hiroaki, H. Pharmacologic Comparison of High-Dose Hesperetin and Quercetin on MDCK II Cell Viability, Tight Junction Integrity, and Cell Shape. Antioxidants 2023, 12, 952. [Google Scholar] [CrossRef]
  178. Lu, Q.; Kishi, H.; Zhang, Y.; Morita, T.; Kobayashi, S. Hesperetin Inhibits Sphingosylphosphorylcholine-Induced Vascular Smooth Muscle Contraction by Regulating the Fyn/Rho-Kinase Pathway. J. Cardiovasc. Pharmacol. 2022, 79, 456–466. [Google Scholar] [CrossRef]
  179. Yao, Y.; Lin, M.; Liu, Z.; Liu, M.; Zhang, S.; Zhang, Y. Hesperidin Inhibits Lung Cancer In Vitro and In Vivo Through PinX1. Front. Pharmacol. 2022, 13, 918665. [Google Scholar] [CrossRef]
  180. Sánchez-Recillas, A.; González-Rivero, N.A.; Barrera-Canto, V.; Ibarra-Barajas, M.; Estrada-Soto, S.; Ortiz-Andrade, R. Vasorelaxant and Antihypertensive Activities of Citroflavonoids (Hesperidin/Naringenin Mixture): Potential Prophylactic of Cardiovascular Endothelial Dysfunction. Pharmacogn. Mag. 2019, 15, 84. [Google Scholar]
  181. Jeong, S.A.; Yang, C.; Song, J.; Song, G.; Jeong, W.; Lim, W. Hesperidin Suppresses the Proliferation of Prostate Cancer Cells by Inducing Oxidative Stress and Disrupting Ca2+ Homeostasis. Antioxidants 2022, 11, 1633. [Google Scholar] [CrossRef] [PubMed]
  182. Lee, J.; Kim, D.-H.; Kim, J.H. Combined Administration of Naringenin and Hesperetin with Optimal Ratio Maximizes the Anti-Cancer Effect in Human Pancreatic Cancer via down Regulation of FAK and P38 Signaling Pathway. Phytomedicine 2019, 58, 152762. [Google Scholar] [CrossRef] [PubMed]
  183. Ammar, N.M.; Hassan, H.A.; Abdallah, H.M.I.; Afifi, S.M.; Elgamal, A.M.; Farrag, A.R.H.; El-Gendy, A.E.-N.G.; Farag, M.A.; Elshamy, A.I. Protective Effects of Naringenin from Citrus sinensis (Var. Valencia) Peels against CCl4-Induced Hepatic and Renal Injuries in Rats Assessed by Metabolomics, Histological and Biochemical Analyses. Nutrients 2022, 14, 841. [Google Scholar] [CrossRef]
  184. Shi, X.; Luo, X.; Chen, T.; Guo, W.; Liang, C.; Tang, S.; Mo, J. Naringenin Inhibits Migration, Invasion, Induces Apoptosis in Human Lung Cancer Cells and Arrests Tumour Progression in Vitro. J. Cell. Mol. Med. 2021, 25, 2563–2571. [Google Scholar] [CrossRef]
  185. Chen, J.; Li, H.; Yang, C.; He, Y.; Arai, T.; Huang, Q.; Liu, X.; Miao, L. Citrus Naringenin Increases Neuron Survival in Optic Nerve Crush Injury Model by Inhibiting JNK-JUN Pathway. IJMS 2021, 23, 385. [Google Scholar] [CrossRef] [PubMed]
  186. Lee, D.-H.; Woo, J.-K.; Heo, W.; Huang, W.-Y.; Kim, Y.; Chung, S.; Lee, G.-H.; Park, J.-W.; Han, B.-K.; Shin, E.-C.; et al. Citrus junos Tanaka Peel Extract and Its Bioactive Naringin Reduce Fine Dust-Induced Respiratory Injury Markers in BALB/c Male Mice. Nutrients 2022, 14, 1101. [Google Scholar] [CrossRef]
  187. Singh, S.; Maurya, A.K.; Meena, A.; Mishra, N.; Luqman, S. Narirutin. A Flavonoid Found in Citrus Fruits Modulates Cell Cycle Phases and Inhibits the Proliferation of Hormone-Refractory Prostate Cancer Cells by Targeting Hyaluronidase. Food Chem. Toxicol. 2023, 174, 113638. [Google Scholar] [CrossRef]
  188. Patel, P.; Barve, K.; Bhatt, L.K. Narirutin-Rich Fraction from Grape Fruit Peel Protects against Transient Cerebral Ischemia Reperfusion Injury in Rats. Nutr. Neurosci. 2022, 25, 920–930. [Google Scholar] [CrossRef]
  189. Yousof Ali, M.; Zaib, S.; Mizanur Rahman, M.; Jannat, S.; Iqbal, J.; Kyu Park, S.; Seog Chang, M. Poncirin, an Orally Active Flavonoid Exerts Antidiabetic Complications and Improves Glucose Uptake Activating PI3K/Akt Signaling Pathway in Insulin Resistant C2C12 Cells with Anti-Glycation Capacities. Bioorganic Chem. 2020, 102, 104061. [Google Scholar] [CrossRef]
  190. Cao, X.; Guo, X.; Fang, X.; Ru, S.; Li, E. Effects of Poncirin, a Citrus Flavonoid and Its Aglycone, Isosakuranetin, on the Gut Microbial Diversity and Metabolomics in Mice. Molecules 2022, 27, 3641. [Google Scholar] [CrossRef] [PubMed]
  191. Deepika, M.S.; Thangam, R.; Vijayakumar, T.S.; Sasirekha, R.; Vimala, R.T.V.; Sivasubramanian, S.; Arun, S.; Babu, M.D.; Thirumurugan, R. Antibacterial Synergy between Rutin and Florfenicol Enhances Therapeutic Spectrum against Drug Resistant Aeromonas Hydrophila. Microb. Pathog. 2019, 135, 103612. [Google Scholar] [CrossRef]
  192. Ahmad, T.; Javed, A.; Khan, T.; Althobaiti, Y.S.; Ullah, A.; Almutairi, F.M.; Shah, A.J. Investigation into the Antihypertensive Effects of Diosmetin and Its Underlying Vascular Mechanisms Using Rat Model. Pharmaceuticals 2022, 15, 951. [Google Scholar] [CrossRef]
  193. Park, S.; Bong, S.-K.; Lee, J.W.; Park, N.-J.; Choi, Y.; Kim, S.M.; Yang, M.H.; Kim, Y.K.; Kim, S.-N. Diosmetin and Its Glycoside, Diosmin, Improve Atopic Dermatitis- Like Lesions in 2,4-Dinitrochlorobenzene-Induced Murine Models. Biomol. Ther. 2020, 28, 542–548. [Google Scholar] [CrossRef] [PubMed]
  194. Roy, P.; Mohanty, A.K.; Dick, P.; Misra, M. A Review on the Challenges and Choices for Food Waste Valorization: Environmental and Economic Impacts. ACS Environ. Au 2023, 3, 58–75. [Google Scholar] [CrossRef]
  195. Nirmal, N.P.; Khanashyam, A.C.; Mundanat, A.S.; Shah, K.; Babu, K.S.; Thorakkattu, P.; Al-Asmari, F.; Pandiselvam, R. Valorization of Fruit Waste for Bioactive Compounds and Their Applications in the Food Industry. Foods 2023, 12, 556. [Google Scholar] [CrossRef]
  196. Niluxsshun, M.C.D.; Masilamani, K.; Mathiventhan, U. Green Synthesis of Silver Nanoparticles from the Extracts of Fruit Peel of Citrus tangerina, Citrus sinensis, and Citrus limon for Antibacterial Activities. Bioinorg. Chem. Appl. 2021, 2021, 6695734. [Google Scholar] [CrossRef] [PubMed]
  197. Osonga, F.J.; Akgul, A.; Yazgan, I.; Akgul, A.; Eshun, G.B.; Sakhaee, L.; Sadik, O.A. Size and Shape-Dependent Antimicrobial Activities of Silver and Gold Nanoparticles: A Model Study as Potential Fungicides. Molecules 2020, 25, 2682. [Google Scholar] [CrossRef]
  198. Alkhulaifi, M.M.; Alshehri, J.H.; Alwehaibi, M.A.; Awad, M.A.; Al-Enazi, N.M.; Aldosari, N.S.; Hatamleh, A.A.; Abdel- Raouf, N. Green Synthesis of Silver Nanoparticles Using Citrus limon Peels and Evaluation of Their Antibacterial and Cytotoxic Properties. Saudi J. Biol. Sci. 2020, 27, 3434–3441. [Google Scholar] [CrossRef]
  199. Prabhu, S.; Poulose, E.K. Silver Nanoparticles: Mechanism of Antimicrobial Action, Synthesis, Medical Applications, and Toxicity Effects. Int. Nano Lett. 2012, 2, 32. [Google Scholar] [CrossRef] [Green Version]
  200. Rizzello, L.; Pompa, P.P. Nanosilver-Based Antibacterial Drugs and Devices: Mechanisms, Methodological Drawbacks, and Guidelines. Chem. Soc. Rev. 2014, 43, 1501–1518. [Google Scholar] [CrossRef] [PubMed]
  201. Zhao, Q.; Song, W.; Zhao, B.; Yang, B. Spectroscopic Studies of the Optical Properties of Carbon Dots: Recent Advances and Future Prospects. Mater. Chem. Front. 2020, 4, 472–488. [Google Scholar] [CrossRef]
  202. Šafranko, S.; Goman, D.; Stanković, A.; Medvidović-Kosanović, M.; Moslavac, T.; Jerković, I.; Jokić, S. An Overview of the Recent Developments in Carbon Quantum Dots—Promising Nanomaterials for Metal Ion Detection and (Bio)Molecule Sensing. Chemosensors 2021, 9, 138. [Google Scholar] [CrossRef]
  203. Gudimella, K.K.; Appidi, T.; Wu, H.-F.; Battula, V.; Jogdand, A.; Rengan, A.K.; Gedda, G. Sand Bath Assisted Green Synthesis of Carbon Dots from Citrus Fruit Peels for Free Radical Scavenging and Cell Imaging. Colloids Surf. B Biointerfaces 2021, 197, 111362. [Google Scholar] [CrossRef]
  204. Šafranko, S.; Stanković, A.; Hajra, S.; Kim, H.-J.; Strelec, I.; Dutour-Sikirić, M.; Weber, I.; Herak Bosnar, M.; Grbčić, P.; Kraljević Pavelić, S.; et al. Preparation of Multifunctional N-Doped Carbon Quantum Dots from Citrus clementina Peel: Investigating Targeted Pharmacological Activities and the Potential Application for Fe3+ Sensing. Pharmaceuticals 2021, 14, 857. [Google Scholar] [CrossRef]
  205. Liu, Y.; Yang, F.; Zhao, X.; Wang, S.; Yang, Q.; Zhang, X. Crystal Structure, Solubility, and Pharmacokinetic Study on a Hesperetin Cocrystal with Piperine as Coformer. Pharmaceutics 2022, 14, 94. [Google Scholar] [CrossRef] [PubMed]
  206. Stanisic, D.; Liu, L.H.B.; Dos Santos, R.V.; Costa, A.F.; Durán, N.; Tasic, L. New Sustainable Process for Hesperidin Isolation and Anti-Ageing Effects of Hesperidin Nanocrystals. Molecules 2020, 25, 4534. [Google Scholar] [CrossRef]
  207. Elghani, E.M.A.; Omar, F.A.; Emam, M.M.A.-A.; Al-Mahallawi, A.M.; Tadros, S.H.; Soliman, F.M.; ElSayed, A.M. Hesperidin Hexosomal Loaded Nanodispersion: Insights of Its Antimycobacterial, Cytotoxic and Anti-HCoV Effects. Nat. Product. Res. 2023, 37, 1719–1724. [Google Scholar] [CrossRef]
  208. Gao, L.; Mei, S.; Ma, H.; Chen, X. Ultrasound-Assisted Green Synthesis of Gold Nanoparticles Using Citrus Peel Extract and Their Enhanced Anti-Inflammatory Activity. Ultrason. Sonochemistry 2022, 83, 105940. [Google Scholar] [CrossRef] [PubMed]
  209. Pradhan, S.P.; Sahoo, S.; Behera, A.; Sahoo, R.; Sahu, P.K. Memory Amelioration by Hesperidin Conjugated Gold Nanoparticles in Diabetes Induced Cognitive Impaired Rats. J. Drug Deliv. Sci. Technol. 2022, 69, 103145. [Google Scholar] [CrossRef]
  210. Tshireletso, P.; Ateba, C.N.; Fayemi, O.E. Spectroscopic and Antibacterial Properties of CuONPs from Orange, Lemon and Tangerine Peel Extracts: Potential for Combating Bacterial Resistance. Molecules 2021, 26, 586. [Google Scholar] [CrossRef] [PubMed]
  211. Julianti Wijayadi, L.; Rusliati Rusli, T. Encapsulated Lime Peel Essential Oil (Citrus hystrix) Into Chitosan Nanoparticle: New Entity to Enhanced Effectivity Against Propionilbacterium acne in Vitro. IOP Conf. Ser. Mater. Sci. Eng. 2020, 852, 012016. [Google Scholar] [CrossRef]
  212. Stanly, C.; Moubarak, M.; Fiume, I.; Turiák, L.; Pocsfalvi, G. Membrane Transporters in Citrus clementina Fruit Juice-Derived Nanovesicles. IJMS 2019, 20, 6205. [Google Scholar] [CrossRef] [Green Version]
  213. Stanly, C.; Alfieri, M.; Ambrosone, A.; Leone, A.; Fiume, I.; Pocsfalvi, G. Grapefruit-Derived Micro and Nanovesicles Show Distinct Metabolome Profiles and Anticancer Activities in the A375 Human Melanoma Cell Line. Cells 2020, 9, 2722. [Google Scholar] [CrossRef]
Figure 1. Anatomical and structural characteristics of citrus fruits.
Figure 1. Anatomical and structural characteristics of citrus fruits.
Pharmaceuticals 16 01081 g001
Figure 2. The abundant phytochemicals in citrus fruits and their by-products. Reprinted from ref. [11].
Figure 2. The abundant phytochemicals in citrus fruits and their by-products. Reprinted from ref. [11].
Pharmaceuticals 16 01081 g002
Figure 3. Schematic presentation of the role of cuticular wax in fruits. Reprinted from ref. [115].
Figure 3. Schematic presentation of the role of cuticular wax in fruits. Reprinted from ref. [115].
Pharmaceuticals 16 01081 g003
Figure 4. L. monocytogenes biofilm investigation by using SEM and CLSM imaging after 8, 16, and 24 h of Huyou essential oil treatment; the control sample represents untreated biofilm cells: (A) SEM imaging, (B) CLSM imaging, and (C) COMSTAT analysis; bars represent the mean values (mean ± SD; n = 3), “∗” pointed to significantly enrichment. p < 0.001 were labeled as “∗∗∗”, p < 0.01 were labeled as “∗∗”, and p < 0.05 were labeled as “∗”. Reprinted with permission from ref. [126]. Copyright 2019 Elsevier.
Figure 4. L. monocytogenes biofilm investigation by using SEM and CLSM imaging after 8, 16, and 24 h of Huyou essential oil treatment; the control sample represents untreated biofilm cells: (A) SEM imaging, (B) CLSM imaging, and (C) COMSTAT analysis; bars represent the mean values (mean ± SD; n = 3), “∗” pointed to significantly enrichment. p < 0.001 were labeled as “∗∗∗”, p < 0.01 were labeled as “∗∗”, and p < 0.05 were labeled as “∗”. Reprinted with permission from ref. [126]. Copyright 2019 Elsevier.
Pharmaceuticals 16 01081 g004
Figure 5. Schematic illustration of the hydrogel beads used as a quercetin delivery system for the colon target. Reprinted with permission from ref. [146]. Copyright 2020 Elsevier.
Figure 5. Schematic illustration of the hydrogel beads used as a quercetin delivery system for the colon target. Reprinted with permission from ref. [146]. Copyright 2020 Elsevier.
Pharmaceuticals 16 01081 g005
Figure 6. (A) Effects of citrus extract and hesperidin on the gastric mucosa in aspirin-induced lesions in rats. (B) Photomicrographs showing the macroscopic appearance of the stomach. (C) Gastric bleeding score in rats by applying different formulations. The data represent the mean ± SD (n = 5, per group). Statistical analysis was carried out with the Steel–Dwass test; * p < 0.05. Reprinted from ref. [159].
Figure 6. (A) Effects of citrus extract and hesperidin on the gastric mucosa in aspirin-induced lesions in rats. (B) Photomicrographs showing the macroscopic appearance of the stomach. (C) Gastric bleeding score in rats by applying different formulations. The data represent the mean ± SD (n = 5, per group). Statistical analysis was carried out with the Steel–Dwass test; * p < 0.05. Reprinted from ref. [159].
Pharmaceuticals 16 01081 g006
Figure 7. Bioimaging of pure CQDs (designated as CD in the figure) and CQDs conjugated with folic acid (designated as FA-CD) in MCF-7 cell lines. Reprinted with permission from ref. [203]. Copyright 2020 Elsevier.
Figure 7. Bioimaging of pure CQDs (designated as CD in the figure) and CQDs conjugated with folic acid (designated as FA-CD) in MCF-7 cell lines. Reprinted with permission from ref. [203]. Copyright 2020 Elsevier.
Pharmaceuticals 16 01081 g007
Table 1. Summary of the literature reporting bioactivities of citrus-based essential oils of the past six years (2018–2023).
Table 1. Summary of the literature reporting bioactivities of citrus-based essential oils of the past six years (2018–2023).
SourceFormulation/Chemical AnalyteBioactivityTesting SubjectsReferences
C. aurantifolia peelThe essential oil isolated by hydrodistillation using a Clevenger apparatusAntimicrobial activityIn vitro on multi-drug resistant bacterial isolates[124]
C. reticulata Blanco, C. aurantifolia (Christm.) Swingle
peel
Essential oils prepared by hydrodistillationAntimicrobial activityIn vitro against S. aureus, including MSSA 1, MRSA 2, and MDR 3 phenotypes, and clinically isolated MRSA and MSSA[125]
C. aurantium “Changshan-huyou” peelEssential oils isolated by steam distillationAntimicrobial activityIn vitro against L. monocytogenes[126]
C. lemon, C. aurantifolia, C. maxima, and C. sinensis peelsNano-hexosomal dispersions of citrus essential oilsAntifungal activityIn vitro against phytopathogenic fungi (R. solani, S. rolfsii, F. solani, F. oxysporum, F. semtectium, B. cinerea, and A. alternata)[120]
C. bergamia
Risso, C. aurantium L., C. sinensis (L.) Osbeck., C. deliciosa Ten., and C. limon (L.) Burm. f. peels
Cold-pressed essential oilsAntifungal activityIn vitro against aflatoxin B1 (AFB1)[127]
C. bergamia, C. sinensis, C. limon, C. reticulata, and C. paradisi peelEssential oils obtained by distillationAntiparasitic activityIn vitro against Varroa destructor[128]
C. sinensis peelThe essential oil isolated by
hydrodistillation using Clevenger apparatus
Insecticidal activityIn vitro against Callosobrunchus maculatus and Sitophilus zeamais; studies on the inhibitory effects on acetylcholinesterase (AChE), Na+/K+-ATPase and glutathione-S- transferase (GST) activity[129]
C. maxima peelEssential oils prepared by hydrodistillationInsecticidal (larvicidal) activityIn vitro against Culex tritaeniorhynchus and Aedes aegypti species of mosquitoes[130]
C. aurantium peelEssential oils prepared by solvent extractionAntiviral activityIn vitro against influenza A virus H1N1[131]
C. clementine peelEssential oil prepared by solvent extractionAntiviral activityIn vitro on Vero-E6 cell lines; SARS-CoV-2 propagated in tested cell line[132]
Orange, lemon, mandarin, and
grapefruit peels 4
Commercially purchased essential oils; prepared nanoemulsionsAntioxidant activityLipid and fatty acid methyl ester analysis; trout[133]
C. reticulata peelEssential oil prepared by continuous phase transition extractionPrevention of hypercholesterolemia and hepatic steatosisIn vivo on male Sprague-Dawley rats on a high-fat diet[122]
Orange, lemon, mandarin, and
grapefruit peels 4
Commercially purchased essential oils; prepared nanoemulsionsSuppressive effect on the biogenic amine formationTrout fillets[134]
C. aurantifolia (Christm.) Swingle peelEssential oils prepared by steam distillationAntioxidant capacity and hypolipidemic effectDPPH, ABTS•+ assay; lipid-induced hyperlipidemia in a rat model[135]
C. sinensis (L.) OsbeckThe essential oil isolated by hydrodistillation using a Clevenger apparatusAntifungal and antitumor activityAntifungal: Aspergillus
carbonarius and Aspergillus flavus/antitumor: Tumor cells (A549, lung adenocarcinoma; MCF-7, breast adenocarcinoma;
and HT-144, melanoma) and normal cells (fibroblasts derived from normal human skin, CCD-1059Sk)
[136]
C. depressa Hayata pulpThe essential oil isolated by hydrodistillation using a Clevenger apparatusStress reduction activity and anti-inflammatory potentialIn vivo on nine healthy female panelists (ECG and EEG monitoring); nitric oxide (NO) and interleukin-1β markers[123]
C. limon (L.) Burm f. peelCommercially purchased essential oilAnxiolytic and sedative propertiesIn vivo on Swiss mice model[137]
C. reticulata Blanco peelsThe essential oil obtained by supercritical CO2 extractionMood disorder/relaxing agentIn vivo on adult male Wistar rats; inhalation[138]
C. sinensis, C. bergamia, C. paradisi, C. grandis, C. reticulata Blanco, C. japonica, C. limon, C. aurantifolia, and immature C. aurantium L. peelsEssential oils prepared by hydrodistillationTreatment of dysmenorrheaIn vivo on female Sprague Dawley rats/in vitro on the RL95-2 (human endometrial carcinoma) cells[139]
C. limon peelEssential oil prepared by steam distillationThe healing effect of traumatic ulcers induced by diabetesIn vivo on diabetic Wistar rats[140]
1 Methicillin-susceptible S. aureus; 2 methicillin-resistant S. aureus; 3 multidrug-resistant; 4 species not specified.
Table 4. Literature reporting the preparation of different (nano)formulations by using citrus by-products as precursors (2018–2023).
Table 4. Literature reporting the preparation of different (nano)formulations by using citrus by-products as precursors (2018–2023).
SourceFormulationApplication/BioactivityTesting SubjectsReferences
Citrus peel 1Carbon quantum dots conjugated with folic acidBioimaging in MCF-7 cell lines, antiradical activityIn vitro on MCF-7 (human breast carcinoma), L929 (mice fibroblasts)[203]
C. clementina peelAmino acid-functionalized carbon quantum dotsAntiradical activity, bioimaging in MCF-7 cell lines, antitumor activity in pancreatic cancer cell linesAntiradical activity: DPPH/in vitro on HepG2 (hepatocellular carcinoma), MCF-7 (breast adenocarcinoma, metastatic), HCT-116 (colorectal carcinoma), CFPAC-1 (cystic fibrosis pancreatic adenocarcinoma, metastatic), and HFF-1 (human foreskin fibroblasts)[204]
Commercial productHesperetin cocrystals with piperineDrug deliveryIn vivo bioavailability on Sprague–Dawley rats[205]
C. sinensis peelHesperidin nanocrystalsCosmeticsIn vitro on artificial skin[206]
C. sinensis L. Osbeck var. Valencia peelHesperidin hexosomal loaded nanodispersionAntimycobacterial, cytotoxic, and anti-HCov activityAntimycobacterial: Mycobacterium (M.) tuberculosis (MTB)/cytotoxic: against A-549 (human pulmonary adenocarcinoma) cell lines/antiviral: human coronavirus 229E[207]
C. reticulata peelHesperidin encapsulated in magnetic casein-CaFe2O4 nanohybrid carrierDrug delivery, antitumor activityIn vitro drug release/in vitro on SKOV-3 (human ovarian cancer cell line) and MDA-MB-231 TNBC (human breast cancer cell line)[56]
C. sinensis var. Valencia peelGold nanoparticles (AuNPs)Anti-inflammatory activityNitric oxide inhibitory activity, qRT-PCR 2, Western blot[208]
Orange peel 1Hesperidin gold nanoparticles (Hes-AuNPs)Neuroprotective and antioxidant effectsIn vivo on Wistar rats/antioxidant: DPPH and in vivo studies[209]
C. tangerina, C. sinensis, and C. limon peelSilver nanoparticles (AgNPs)Antimicrobial activityAntimicrobial: E. coli and S. aureus[196]
C. limon peelSilver nanoparticles (AgNPs)Antimicrobial activityAntimicrobial: A. baumannii, S. typhimurium, E. coli, P. aeruginosa, S. aureus, and P. vulgaris[198]
Lemon, tangerine, and orange peel 1Copper oxide nanoparticles (CuONPs)Antimicrobial activityAntimicrobial: five
strains of Gram-positive (Enterococcus (E.) faecalis, S. aureus, L. monocytogenes, S. pneumonia and Clostridium (C.) perfringens) and five strains
of Gram-negative (E. coli, Moraxella (M.) catarrhalis, Salmonella (S.) enterica subsp. diarizonae, Campylobacter (C.) coli, and P. aeruginosa) bacteria
[210]
C. hystrix peelEncapsulated essential oil into chitosan nanoparticleAntimicrobial activityAntimicrobial: Propionilbacterium (P.) Acnes[211]
C. clementine vesiclesExosome-like nano-sized vesiclesMolecular deliveryProteomic and bioinformatic studies[212]
C. sinensis, C. limon, C. paradise, C. aurantium isolated vesiclesMicro- and nano-sized vesiclesAntitumor activityIn vitro on breast adenocarcinoma (MCF7), human melanoma (A375), lung adenocarcinoma (A549), and human normal skin keratinocyte (HaCat) cells[213]
1 Species not specified; 2 real-time quantitative reverse transcription PCR.
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

Šafranko, S.; Šubarić, D.; Jerković, I.; Jokić, S. Citrus By-Products as a Valuable Source of Biologically Active Compounds with Promising Pharmaceutical, Biological and Biomedical Potential. Pharmaceuticals 2023, 16, 1081. https://doi.org/10.3390/ph16081081

AMA Style

Šafranko S, Šubarić D, Jerković I, Jokić S. Citrus By-Products as a Valuable Source of Biologically Active Compounds with Promising Pharmaceutical, Biological and Biomedical Potential. Pharmaceuticals. 2023; 16(8):1081. https://doi.org/10.3390/ph16081081

Chicago/Turabian Style

Šafranko, Silvija, Drago Šubarić, Igor Jerković, and Stela Jokić. 2023. "Citrus By-Products as a Valuable Source of Biologically Active Compounds with Promising Pharmaceutical, Biological and Biomedical Potential" Pharmaceuticals 16, no. 8: 1081. https://doi.org/10.3390/ph16081081

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