Abstract
Carotenoids constitute compounds of significant biological interest due to their multiple biological activities, such as antimicrobial, anticancer, antiadipogenic, antidiabetic, and antioxidant properties. Metabolic syndrome (MetS) comprehends a series of metabolic abnormalities (e.g., hypertension, obesity, and atherogenic dyslipidemia) that can affect children, adolescents, and the elderly. The treatment of MetS involves numerous medications, which, despite their efficacy, pose challenges due to prolonged use, high costs, and various side effects. Carotenoids and their derivatives have been proposed as alternative treatments to MetS because they reduce serum triglyceride concentrations, promote insulin response, inhibit adipogenesis, and downregulate angiotensin-converting enzyme activity. However, carotenoids are notably sensitive to pH, light exposure, and temperature. This review addresses the activity of carotenoids such as lycopene, lutein, fucoxanthin, astaxanthin, crocin, and β-carotene towards MetS. It includes a discussion of sources, extraction methods, and characterization techniques for analyzing carotenoids. Encapsulation approaches are critically reviewed as alternatives to prevent degradation and improve the biological performance of carotenoids. A brief overview of the physiopathology and epidemiology of the diseases, including MetS, is also provided.
Keywords: carotenoids, metabolic disorders, encapsulation, biological activity, stability, bioavailability
1. Introduction
Metabolic syndrome (MetS), a complex of physiological and metabolic disorders, is a significant health concern. It includes atherogenic dyslipidemia, dysglycemia, abdominal obesity, cardiovascular diseases, hyperglycemia, and fatty liver [1]. The development of MetS is influenced by both physiological and behavioral factors. Smoking, poor diet, and limited physical activity fall under the former, while high blood cholesterol levels and high body mass index are associated with the latter [2].
The prevalence of MetS can vary depending on factors such as age, socioeconomic status, ethnic groups, gender, and diagnosed disease [3]. Based on the latter, it has been estimated that MetS affects approximately 20–25% of global adults and 0–19.2% of children [4,5]. Current treatment approaches for MetS are primarily focused on changes in dietary habits and the intensity of physical exercises [6]. Regarding drug therapy, commonly prescribed medications for the treatment of MetS include angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), statins, and metformin [7].
Carotenoids represent a unique class of organic molecules extensively present in higher plants, algae, bacteria, and fungi. Depending on the organisms that synthesize them, carotenoids can regulate internal signaling mechanisms, mediate stress responses, and modulate complex biochemical processes such as photosynthesis [8]. In nature, carotenoids can be found in peaches, papaya, cereal grains, dairy goods, and insects [9]. Despite their wide distribution, carotenoids’ biosynthesis can be influenced by high or low altitude, climate conditions, humidity, and exposure to light [10,11].
From biological sources, carotenoids can be extracted through standard laboratory techniques such as maceration and Soxhlet extraction using non-polar (such as hexane and petroleum ether) and polar solvents (such as acetone and ethanol), separated using chromatography techniques, including column chromatography and high-performance liquid chromatography [12]. Identification of carotenoids can be achieved using spectroscopy methods such as nuclear magnetic resonance, infrared spectroscopy, and UV-Vis spectroscopy [13].
Chemically, carotenoids are a broad classification of isoprenoids characterized by extended hydrocarbon chains containing multiple single or double bonds and featuring cyclic or linear structures at their termini. Additionally, they can contain several functional groups in their chemical architecture, such as acetate, hydroxyl, epoxide, carboxylic, sulfate, and lactone groups [14]. Based on these features, carotenoids are classified into acyclic carotenes (e.g., lycopene and ζ-carotene), cyclic carotenes (e.g., α-carotene and β-carotene), carotenols (e.g., lutein and zeaxanthin), epoxycarotenoids (e.g., auroxanthin and luteoxanthin), crocetin glycosides (e.g., crocin), monoketo β-carotenes (e.g., echinenone), and 4,4′-diketo derivatives of β-carotene (e.g., canthaxanthin) [15].
Economically, the global market impact of carotenoids amounts to USD 1.44 billion. Among carotenoids, those with the highest industrial production comprehend astaxanthin, annatto, β-carotene, lutein, and lycopene, significant products used in dietary supplements, cosmetics, foods, and beverages [16]. Biologically, carotenoids are desirable compounds due to their ability to inhibit the growth of pathogenic Gram-positive (e.g., Staphylococcus aureus and Clostridioides difficile) and Gram-negative (e.g., Pseudomonas aeruginosa and Klebsiella oxytoca) strains [17]. Additionally, they regulate the levels of reactive oxygen species (ROS), scavenge free radicals, induce apoptosis of cancer cells, diminish the formation of new blood vessels, reduce the secretion of pro-inflammatory cytokines, and protect neurons against degradation [18].
In the clinical pipeline, carotenoids have been utilized to formulate multiple products to treat various disorders. Some of these drugs or supplements currently available in international markets include Pregvit®, Soriatane®, and Epuris®, which are indicated for reducing photosensitivity in patients, treating psoriasis, and managing severe recalcitrant nodular acne, respectively [19]. Due to their clinical safety and tolerability, carotenoids have been suggested as important adjuvant drugs in the treatment of COVID-19 [20], melanoma [21], breast cancer [22], diabetic retinopathy [23], and MetS [24].
In combating MetS, carotenoids have been shown to reduce the risk of diabetes and insulin resistance [25], downregulate blood pressure in hypertensive patients [26], reduce abdominal and epididymal adipose tissue weights [27], and slow the rise of blood glucose levels [28]. However, their broader applications are hindered by significant challenges, including low water solubility, high susceptibility to multifactorial degradation, high rates of oxidative degradation, and susceptibility to trans–cis isomerization [29].
In recent decades, micro- and nanotechnologies have emerged as active research fields to overcome significant challenges in the biomedical applications of bioactive natural products like carotenoids. Materials employed in both areas to improve these products’ performance, stability, and bioavailability include biopolymeric matrices such as chitosan, alginates, casein, or gelatin, as well as lipidic structures like liposomes or emulsions [30]. In the specific case of carotenoids, commonly utilized micro- and nanomaterials are gum Arabic [31], maltodextrin [32], fructo-oligosaccharides and inulin [33], and poly(vinylpyrrolidone) [34].
Given the need for a comprehensive review that addresses the potential of carotenoids as an alternative treatment in preventing and managing MetS, this review provides recent scientific evidence covering the extraction and characterization of carotenoids to their biological evaluation in vitro and in vivo using models that resemble conditions related to MetS disorders. In addition, the fundamentals of micro- and nanoencapsulation to protect the chemical integrity and preserve the therapeutic functionality of carotenoids are included. The search systems included Web of Science, PubMed, Springer, Wiley Online Library, and Google Scholar from 2020 to 2024. Keywords used to retrieve evidence were carotenoids, metabolic syndrome, microtechnology, nanotechnology, and structure–activity relationship. The number of publications related to the use of carotenoids against Mets and their number of citations are illustrated in Figure 1.
Figure 1.
Time distribution of published papers related to the investigation of carotenoids against MetS.
2. MetS: An Overview about Its Epidemiology, Oxidative Stress, and Treatment
2.1. Epidemiology
MetS encompasses various diseases, including hypertension, obesity, and diabetes mellitus. Figure 2 illustrates other diseases that are part of or associated with MetS, such as polycystic ovary syndrome, cardiovascular diseases (CVDs), dementia, non-alcoholic fatty liver disease (NAFLD), hypertension, and cancer. Discussing the epidemiology of MetS is challenging due to its association with various diseases, which can vary depending on age, gender, and ethnicity. However, it is documented that hypertension has been associated with over ~8.5 million deaths as of 2015, primarily in low- and middle-income countries [12]. In comparison, obesity is reported to affect ~650 million adults, particularly those in high- and middle-income countries [35], while diseases such as diabetes mellitus have affected ∼451 million people in recent decades [36].
Figure 2.
Cluster of diseases constituting metabolic syndrome. Adapted from BioRender.com.
2.2. Risk Factors
The development of MetS can be influenced by various factors, including oxidative stress, chronic inflammation, excessive alcohol and cigarette consumption, family history, and insufficient physical activity [37]. Although each factor plays a role in the progression of MetS-related diseases, the precise mechanisms remain elusive, with oxidative stress being the most frequently implicated. In patients diagnosed with MetS, oxidative stress may arise from mitochondrial and metabolic dysfunction caused by elevated levels of fatty acid or glucose and reduced insulin secretion or decreased levels of antioxidant enzymes in β-cells [38].
Free radicals such as ROS (H2O2, OH·, and 1O2), ONOO−, HNO2, and NO∙ can disrupt cell signaling pathways such as the polyol or protein kinase C pathways. This disruption leads to dysregulated mitochondrial energy levels and over-secretion of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukins-2, -6, and -1β [39,40].
In the case of hypertension, recent studies have indicated that insulin resistance promotes increased lipid uptake by cells, such as myocardial cells, to meet energy demands, as glucose uptake is insufficient. This leads to more significant amounts of fatty acids for oxidation, potentially resulting in lipotoxicity. Consequently, ROS subsequently increases, leading to oxidative stress and damage to myocardial tissue, contributing to systolic dysfunction. Similarly, obese patients have been shown to have elevated levels of free fatty acids, increased secretion of pro-inflammatory cytokines (e.g., TNF-α and interleukin-6), and reduced activity of antioxidant enzymes (e.g., catalase and superoxide dismutase) [41]. Conversely, conditions like insulin resistance and type 2 diabetes (DM2) involve systemic inflammation and elevated ROS levels, resulting in various systemic damages such as extracellular matrix migration and deposition, endothelial dysfunction, cardiac hypertrophy and dysfunction, and fibrosis [42].
2.3. Treatment and Limitations
The treatment of MetS consists of administering antihypertensive drugs, lipid-lowering medications, antidiabetic medicines, and a combination of a healthy diet and physical activity. Current therapeutic regimens for hypertension include the administration of ARBs such as losartan [43], ACE inhibitors like lisinopril [44], or calcium channel blockers such as amlodipine [45].
Against diabetes, frequently administrated drugs include metformin, insulin, sulfonylureas such as glyburide [46], glucagon-like peptide-1 (GLP-1) receptor agonists like liraglutide [47], and sodium–glucose cotransporter 2 (SGLT2) inhibitors such as dapagliflozin [48]. In the case of anti-obesity drugs, common agents include lipase inhibitors such as orlistat [49], sympathomimetic appetite suppressants like phentermine [50], GLP-1 receptor agonists such as semaglutide [51], and serotonin receptor agonists like lorcaserin [52].
In clinical practice, administering ARBs, ACE inhibitors, antidiabetic agents, GLP-1 receptor agonists, or SGLT2 inhibitors can lead to limitations and adverse effects (AEs) that may affect patients’ well-being and prognosis. Co-administration of ACE inhibitors and ARBs has been associated with major cardiovascular events, all-cause mortality, gastrointestinal disorders, hypotension, angioedema, and hyperkalemia [53]. Similarly, the administration of GPL-1 agonists like semaglutide has been linked to serious AEs such as vomiting, pancreatitis, and diarrhea, while liraglutide use has resulted in upper abdominal pain [54]. Given these concerns, it is crucial to continue evaluating alternative treatments for MetS-related diseases.
3. Carotenoids: Sources, Extraction, Characterization, and Activities against MetS
3.1. Sources, Extraction, and Characterization
The advantages of carotenoids stem from their broad spectrum of biological activities and widespread presence in various sources.
Conventional extraction techniques, while effective, are associated with high energy consumption, long extraction periods, and low yield of bioactive products [55]. In contrast, green extraction methods, in line with the principles of green chemistry, offer an attractive alternative. These methods are versatile, allowing for decreased energy consumption and the use of new-generation solvents, while ensuring the obtention of high-quality products. Importantly, they can be performed with low volumes of green solvents such as ionic liquids, deep eutectic solvents, and natural deep eutectic solvents, which are non-volatile and biodegradable substances with the capacity to be recycled [56].
In recent years, green extraction methods have emerged as innovative techniques, offering a promising future for carotenoid extraction. These methods, such as ultrasound-assisted extraction (UAE), microwave-assisted extraction (MAE), and supercritical fluid extraction (SFE), can be optimized to yield high quantities of bioactive products while maintaining their eco-friendly nature and safety [57]. For instance, carotenoids such as 13-β-carotene, 9-β-carotene, and γ-carotene have been successfully extracted from peach palm (Bactris gasipaes Kunth), an Amazonian fruit rich in carbohydrates and oil by UAE [58]. Following the same technique, carotenoids have been obtained from orange peel utilizing olive oil as a solvent by controlling extraction time, temperature, and liquid-to-solid ratio [59].
Through UAE, carotenoids can be extracted from carrot pomace using hexane, acetone, or ethyl acetate while controlling experimental parameters such as liquid-to-solid ratio, ultrasonic power, and time [60]. In comparison with UAE, the efficacy of MAE to extract carotenoids from Rhodotorula glutinis using solvents of different polarity (dichloromethane, diethyl ether, dimethyl carbonate, and ethyl acetate) has been compared [57]. On the other hand, SFE with carbon dioxide (CO2) has been documented as a toxic waste-free process that does not cause the degradation of thermolabile compounds. The experimental setting of a CO2-SFE is depicted in Figure 3A, whereas Figure 3B illustrates the use of solvent extraction-based techniques for the recovery of carotenoids from palm-pressed fiber (PPF). CO2-SFE has been utilized to extract carotenoids from the cells of yeast Rhodotorula spp., strain ELP2022 under various operating parameters (pressure and temperature), and further separated by chromatography methods [61].
Figure 3.
(A) Representation of a CO2-SFE system for the extraction of lycopene from ripe grapefruit endocarp; reprinted and adapted with permission from [62]. (B) Solvent extraction system of carotenoids from palm-pressed fiber (PFF) utilizing a peristaltic pump, thermostatic bath, and thermometer; reprinted and adapted with permission from [63].
From microorganisms, carotenoids such as β-carotene can be extracted from yeasts like Yarrowia lipolytica using solid–liquid extraction with a solvent mixture (acetone, ethanol, and water), followed by solid–liquid–liquid extraction [64]. Alternatively, carotenoids such as astaxanthin have been extracted from genetically modified cell-derived cultures of microalgae such as Haematococcus lacustris [65], Dunaliella salina, Chromochloris zofingiensis, Chloromonas krienitzii, and Sanguina nivaloides [66]. Hydroxylated derivatives of astaxanthin, such as 2,2’-dihydroxy-astaxanthin and 2-hydroxy-astaxanthin, can be extracted with acetone from Brevundimonas aurantiaca M3d10, a bacterial strain found in wild olive flies [67,68]. Other genetically modified organisms to obtain astaxanthin encompass HEK293T cells, which are human embryonic kidney cells that have been proposed as an optimized efficient in vitro model for the biosynthesis of carotenoids [69]. Following extraction from these sources, carotenoids are chromatographically separated, quantified, and spectroscopically identified.
The separation of carotenoids is typically conducted using optimized column chromatography techniques such as high-performance liquid chromatography (HPLC), ultra-high-performance liquid chromatography (UHPLC), or ultra-performance liquid chromatography (UPLC). In contrast, to other chromatography methods, HPLC, UHPLC, and UPLC systems can be coupled to diode array detectors (DAD), photodiode array (PDA) detectors, and mass spectrometers (MS) to efficiently characterize the presence of bioactive compounds [70]. During the analysis of complex mixtures, it is relevant to consider the main features of each technique. For example, UHPLC and UPLC systems can operate at higher pressures, utilize columns with particle sizes less than ~2 μm, possess increased separation efficiency, and execute faster time analyses than HPLC systems. Despite their advantages, the instrumentation of UHPLC and UPLC systems is complex, and analysis of samples tends to be expensive [71].
For the evaluation of natural products, the use of DAD and PDA detectors is necessary since they enable the detection and identification of compounds based on their UV-Vis absorption spectra, which is a process with higher sensitivity compared to other detectors such as UV-Vis or refractive index detectors [72,73]. Even though DAD and PDA detectors are exploited in the investigation of bioactive compounds, it is crucial to mention that PDA detectors are preferred since they exhibit higher sensitivity, better signal-to-noise ratio, high data acquisition rates, higher spectral resolution, and advanced data processing [74]. In contrast, DAD detectors enable basic data processing features at slower scanning speeds with lower spectral resolution.
Adequate separation and further identification of carotenoids can be achieved by coupling HPLC, UHPLC, or UPLC systems with MS detectors. MS detectors are superior in identifying carotenoids, as they yield detailed information about the fragmentation patterns and molecular weight of molecules [75]. In contrast to DAD or PDA detectors, MS can be used with ionization techniques to quantify the presence of carotenoids among samples and confirm their identity [76]. Natural products can be identified through MS detectors by considering two or more stages of mass analysis within a mass spectrometer. The techniques derived from this process are known as tandem mass detection methods or MS/MS; a representative example includes a quadrupole–time-of-flight MS [77].
In the same context, column selection is another essential feature to consider when using chromatography methods. Column choice determines the successful separation of compounds [78]. In HPLC-based systems, columns exhibit different affinities for analytes in terms of polarity and charge, and they must possess compatible features with the composition of the mobile phase and parameters of the designed method, such as temperature and pressure [79]. Standard columns employed during HPLC analyses include C18 and C30 columns, which vary by their alkyl chain length, hydrophobicity, operating conditions, and selectivity. For the separation of carotenoids, representative chromatography columns comprehend platinum C18 100A and betasil C18 columns [80].
To illustrate the extraction, characterization, and separation of carotenoids, Figure 4A represents the obtention of carotenoids from haloarchaeal strains and their characterization by UV-Vis spectroscopy, where it can be noticed that carotenoids exhibit major peaks at bands located at 385–522 nm [81]. Figure 4B depicts the HPLC analysis of lutein, zeaxanthin, and β-cryptoxanthin synthesized by a plant xanthophyll acyltransferase (XAT) by adding acyl donors [82]. Moreover, Figure 4C illustrates the extraction of carotenoids from Rhodosporidium sp., their identification by thin-layer chromatography (TLC) and HPLC, and evaluation against HEK293T cells and in silico modeling interactions with the vascular endothelial growth factor receptor 1 (VEGFR2) [83].
Figure 4.
(A) Carotenoids isolated from twelve strains of haloarchaeal strains categorized into two main categories (yellow and orange) and their absorbance profiles obtained by UV-Vis spectroscopy analyses; figure was reprinted and adapted with permission from [81]. (B) HPLC analysis of lutein, zeaxanthin, and β-cryptoxanthin synthesized by a plant xanthophyll acyltransferase (XAT) by adding acyl donors [82]. (C) Cultivation of red yeast strains, extraction, separation with hexane, and chromatography analysis of carotenoids by thin-layer chromatography (TLC) and HPLC, and their effect in HEK293T cells and in silico simulation; figure was reprinted and adapted with permission from [83].
For the separation of carotenoids, an ultra-rapid resolution HPLC-DAD method was employed to detect free carotenoids like lutein, violaxanthin, and lycopene through liquid–liquid extraction from juices of mango, guava, pineapple, watermelon, and grape cultivars from Brazil [84]. This same technique has been utilized to analyze the presence of ketocarotenoids, such as astaxanthin, and 3-hydroxyechinenone in an acetone extract from Balaustium muroum, a free-living mite with a red body [85]. In contrast, fucoxanthin extracted from genetically modified cultures of Nanofrustulum shiloi was purified using preparative HPLC-DAD and subsequently analyzed by liquid chromatography–tandem mass spectrometry (LC-MS/MS) [86]. Table 1 provides an overview of carotenoids extracted from different sources and the techniques used for their separation and characterization.
Table 1.
Sources, extraction techniques, separation methods, and identification approaches of carotenoids.
| Carotenoid | Source | Extraction Technique | Separation Method | Identification Approach | References |
|---|---|---|---|---|---|
| Haematococcus pluvialis | Liquid–liquid extraction | HPLC | UV-Vis spectroscopy FTIR spectroscopy |
[87] | |
| Astaxanthin | Corynebacterium glutamicum | Maceration | HPLC | N.I. | [88] |
| Phaffia rhodozyma | Solid–liquid extraction | N.I. | UV-Vis spectroscopy | [89] | |
| Tisochrysis lutea | Solid–liquid US-assisted extraction | HPLC and CPC | N.I. | [90] | |
| Fucoxanthin | Undaria pinnatifida | Heat extraction | HPLC | UV-Vis spectroscopy | [91] |
| Tomato (Solanum lycopersicum) | Enzyme-assisted extraction | UHPLC | N.I. | [92] | |
| Carrot (Daucus carota) | Organic solvent extraction | N.I. | UV-Vis spectroscopy | [93] | |
| Lycopene | Elaeagnus umbellata | UAE | UHPLC | UV-Vis spectroscopy FTIR spectroscopy MS |
[94] |
| Red papaya | SC-CO2 | N.I. | N.I. | [95] | |
| Pistachio waste | Soxhlet extraction | LC-MS/MS | MS | [96] | |
| Lutein | Fruit juices | Liquid–liquid extraction | HPLC | N.I. | [84] |
| Marigold flowers | Surfactant-based ATPS extraction | N.I. | UV-Vis spectroscopy | [97] | |
| Lycium barbarum | Liquid–liquid extraction | HPLC HSCCC |
N.I. | [98] | |
| Zeaxanthin | Dried corn silk | Solid–liquid extraction | HPLC CC |
UV-Vis spectroscopy FTIR spectroscopy NMR spectroscopy |
[99] |
| Chlorella | PLE | HPLC | UV-Vis spectroscopy | [100] |
Abbreviations: SC-CO2, supercritical carbon dioxide; ATPS, two-phase system; PLE, pressurized liquid extraction; HPLC, high-performance liquid chromatography; CPC, centrifugal partition chromatography; UHPLC, ultra-high-performance liquid chromatography (HPLC), liquid chromatography–tandem mass spectrometry; HSCCCU, high-speed countercurrent chromatography; UV-Vis, ultraviolet–visible; FTIR, Fourier transform infrared spectroscopy; MS, mass spectrometry; N.I., not indicated.
Spectroscopy and microscopy techniques have been widely used to characterize carotenoids in laboratory settings and clinical studies. For example, FTIR spectroscopy was employed to analyze the functional groups present in flour derived from the peels of passion fruit (Passiflora edulis f. flavicarpa), known for its natural abundance of carotenoids [101]. In another study, nuclear magnetic resonance (NMR) was utilized to investigate changes in the chemical structure of carotenoids like cryptoxanthin and zeaxanthin from chili oil subjected to different frying temperatures: 30, 150, 170, and 190 °C [102]. In cross-sectional studies, spectroscopy approaches such as Raman spectroscopy have been used to monitor Korean adults’ carotenoid intake [103]. Furthermore, in parallel controlled trials, pressure-mediated reflection spectroscopy has been applied to detect changes in skin carotenoid levels, such as lycopene, lutein, phytoene, and phytofluene, in adults diagnosed with obesity [104]. Regarding the use of microscopy methods, fluorescence lifetime imaging microscopy has been implemented to analyze and correlate carotenogenesis events among cellular structures from Bracteacoccus aggregatus cultures [105].
3.2. Activities against MetS
Against hypertension, dietary carotenoids have been shown to reduce blood pressure and atherosclerosis in patients diagnosed with CVDs. Lycopene has been implicated in exerting preventive effects during the treatment of CVDs. It can downregulate the generation of ROS, modulate the activation of NF-κB, a major pro-inflammatory pathway, and thereby regulate the expression of cell adhesion factors and vascular permeability [106]. Similarly, high serum levels of α-carotene, β-carotene, β-cryptoxanthin, lycopene, lutein, and zeaxanthin have been associated with a decreased risk of mortality caused by CVDs among adults in the USA [26]. Moreover, it has been revealed that low serum lycopene concentrations are linked to an increased risk of death from CVDs, while moderate serum levels are associated with protective effects against cardiovascular death in recent cross-sectional survey studies [107].
The consideration of carotenoids as useful bioactive nature products against obesity is attributed to their capacity to affect adipogenesis, modulate metabolic capacity, and decrease the release of inflammatory products [108]. In C57BL/6J obese mice models, treatment with lutein was found to decrease epididymal and abdominal adipose tissue weights, reduce serum cholesterol, low-density lipoprotein cholesterol (LDC-C) concentration, hepatic triglycerides, and cholesterol, as well as blood glucose levels [27]. These effects were also analyzed in combination with orlistat. Similarly, C57BL/6N mice treated with carotenoids from red paprika (e.g., α-carotene, β-carotene, capsorubin, lutein, and zeaxanthin) showed enhanced endurance exercise capacity, increased muscle weights of limbs, and diminished intramyocellular fat accumulation, prevented the degradation of muscle protein through the activation of the mTOR pathway, and enhanced mitochondrial dysfunction [109].
The effect of carotenoids on diabetic patients or clinical models has been an active research field over the last decades. For instance, it was demonstrated that treatment with astaxanthin decreased the levels of low-density lipoproteins and cholesterol while improving whole-body glucose disposal and insulin sensitivity without causing apparent AEs among prediabetic patients [110]. Similarly, fucoxanthin, an algae-specific xanthophyll, was documented through a randomized, double-blind placebo-controlled clinical trial to increase the total and first phase of insulin secretion while decreasing body weight, body mass index, waist circumference, systolic blood pressure, diastolic blood pressure, and triglyceride levels among patients with MetS [111].
Against other diseases that constitute MetS, such as cancer, high circulating levels of carotenoids such as β-carotene and vitamin A have been demonstrated to reduce breast cancer risk while promoting the production of metabolites involved in redox balance, immune response regulation, and synthesis of macromolecules. This was reported through metabolomics analyses [112]. In the case of disorders that arise from cognitive decline, such as dementia, α- and β-carotene, lutein, and zeaxanthin have been associated with improved cognitive functionality [113]. On the other hand, treatment with lycopene has been demonstrated to prevent the development of NAFLD in C57BL/6J mice by suppressing the hepatic NFκ-B/NLRP3 inflammasome pathway [114]. In contrast, treatment with zeaxanthin has been shown to decrease the generation of ROS, inhibit ferroptosis, enhance mitochondrial dysfunction, and diminish lipid peroxidation in free fatty acid-induced HepG2 cells. These effects are promising for further validating the role of carotenoids in NAFLD intervention [115].
4. Encapsulation of Carotenoids
Encapsulation is the process whereby bioactive substances and their physicochemical features are protected from environmental factors (e.g., temperature, light, and moisture) or physiological changes (e.g., transport mechanisms, enzyme degradation, and binding proteins) by trapping them within matrices synthesized from different materials [116]. As illustrated in Figure 5A, the release of therapeutic cargo from micro- and nanostructures can be triggered by external and internal stimuli such as heat, ultrasound, light, magnetic field alterations, pH, redox conditions, and enzyme activity. In contrast, via linker chains, their uptake by tissues or cells can be facilitated by attaching natural or synthetic targeting moieties such as enzymes, antibodies, folic acid, or polyethylene glycol [117]. This event is also depicted in Figure 5A.
Figure 5.
(A) Schematic representation of functionalization of micro- or nanomaterials with natural and synthetic molecules to improve their targeting capacity and release of therapeutic cargo through external (heat, ultrasound, light, and magnetic field) and internal stimuli (pH, redox conditions, and enzyme activity). (B) Classification of materials as gels, polymeric particles, polymeric micelles, liposomes, nanorods, and spheres. (C) Administration routes of micro- and nanomaterials via ocular (drops, ointments, implants, and inserts), oral (tablets, capsules, thin films, syrups, and solutions), inhalational (dry powders, liquid sprays, and aerosols), and topical/transdermal (ointments, gel, cremes, patches, and microneedles). Adapted from BioRender.com.
Microstructures range from 1 to 1000 μm, and they can be manufactured from synthetic, semi-synthetic, and natural components through physicochemical routes, such as solvent evaporation, spray-drying, and anionic polymerization methods, respectively [118,119,120]. As represented in Figure 5C, microparticles are convenient as they can be manipulated to formulate solid, semi-solid, and liquid products that can be administered orally, intravenously, intramuscularly, or subcutaneously to successfully deliver and enhance the efficacy and bioavailability of one or more drugs [121].
In contrast, nanomaterials are conventionally found from 10 to 500 nm and are frequently exploited for the development of drug delivery systems [122], bioactive structures [123], and platforms for disease diagnosis [124]. Like microparticles, nanomaterials are synthesized from organic and inorganic substances through top-down (e.g., laser ablation, lithography, and vapor deposition) and bottom-up (e.g., flame spraying, sol–gel, and micro-emulsion) approaches [125]. They can be administered through various routes, such as intranasal, intraventricular, and intraparenchymal [126]. The administration routes of nanomaterials are depicted in Figure 5C. However, the difference between both categories lies in the fact that nanomaterials are preferred because they can mediate molecular and cellular phenomena, cross endothelial cells of major anatomical structures, and interact with receptors or recognize ligands [126,127].
Given their versatility in the encapsulation of bioactive products, nanoparticles are frequently synthesized to improve the therapeutic activities of carotenoids. As noted in Figure 6A, carbon nanoparticles containing carotenoids can be used to develop multifunctional systems with antiproliferative effects towards melanoma and breast cancer cell lines and 3D imaging capacity in skin tissues [128]. As mentioned, these events can arise if nanoparticles are tailored with molecules, which, in the case of Figure 6A, were attributed to the incorporation of phospholipids onto the surface of carbon nanoparticles. The functionalization of carbon nanoparticles resulted from a complex process that included nucleation, passivation, membrane-freeze, and sonication stages. This can be observed in Figure 6A.
Figure 6.
(A) Development of carbon nanoparticles (C3-NP) containing carotenoids and coated with phospholipids to enhance their antiproliferative effect against melanoma and cancer cell lines, imaging capacity, and cellular entry; figure was reprinted and adapted with permission from [128]. (B) Dynamic light scattering (DLS) analysis of cationic liposomes prepared with β-carotene; figure was reprinted and adapted with permission from [129]. (C) Transmission electron microscopy (TEM) analysis of cationic liposomes loaded with lutein or β-carotene; figure was reprinted and adapted with permission from [129].
The physical features such as size and morphology dictate their fate in organisms. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) are frequently used to determine these parameters. As observed in Figure 6B, DLS analyses of cationic liposomes loaded with carotenoids (lutein and β-carotene) can reveal their size distribution patterns and demonstrate the presence of structures with diverse sizes. In the case of TEM, it is used to determine the morphological arrangement of micro- or nanostructures. As observed in Figure 6C, cationic liposomes loaded with lutein and β-carotene have been demonstrated to adopt oval shapes and small aggregates.
The encapsulation of carotenoids is necessary as they tend to be rapidly degraded upon exposure to heat, oxygen, and light. Other factors that alter their stability include transition metals and radical species [130]. Various micromaterials have been used to prevent the degradation of carotenoids or their isomers; for example, supramolecular oligosaccharides such as cyclodextrins [131], soluble fibers such as pectin [132], and protein residues from cereal grains such as barley [133].
There is scientific evidence regarding using micro- and nanomaterials to treat, in vitro or in vivo. However, Table 2 compiles recent studies where micro- and nanomaterials have been used to encapsulate carotenoids and exhibit potential functional activities to decrease or mitigate molecular disorders related to MetS.
Table 2.
Examples of microencapsulation systems of carotenoids and the observed activities.
| Carotenoid | Encapsulation Technique | Raw Materials | Observed Activities | References |
|---|---|---|---|---|
| Astaxanthin | Spray drying | Gum Arabic | Increased solubility, stability, and enhanced bioavailability in simulated GIT. | [134] |
| Multilayer O/W emulsion and spray drying | ι-carrageenan, chitosan, lupin protein isolate, and sunflower oil | Augmented astaxanthin retention, storage stability, and water solubility. | [135] | |
| Ionic gelation | Low-methoxyl pectin, chitosan, and alginate | Improved particle sphericity and limited oil oxidation during formulation. Increased thermal stability and bioavailability in simulated GIT. |
[132] | |
| Fucoxanthin | Spray drying and freeze drying | Maltodextrin, soy lecithin, and gum Arabic | Microcapsules exhibited high encapsulation efficiency, increased bioavailability, and ABTS•+ scavenging activity. | [136] |
| Sequential coating modification | Maltodextrin and gum Arabic | Enhanced encapsulation efficiency and fucoxanthin stability. | [137] | |
| Complex carriers | Gum Arabic, gelatin, and alginate hydrogel | In vitro increased bioavailability and fucoxanthin protection against SGF. In vivo oral administration demonstrated lowering the blood lipid and oxidative stress levels. |
[138] | |
| Lycopene | Spray drying | Gum Arabic and inulin | Microcapsules demonstrated high lycopene release in simulated gastric fluid. | [139,140] |
| Ionotropic gelation | Alginate and κ-carrageenan | Microspheres exhibited in vitro α-amylase and α-glucosidase inhibitory activity. | [141] | |
| Complex coacervation and freeze drying | Whey proteins and acacia gum | Formulation increased antioxidant activity and showed in vitro α-amylase and α-glucosidase inhibitory activity. | [142] | |
| Lutein | Spray drying | Inulin and modified starch | Microencapsulated powders significantly increased lutein stability and thermal degradation resistance. | [143] |
| Citric acid-esterified potato starch and whey protein | Increased encapsulation efficiency, embedding effect, lutein aqueous solubility, and thermal resistance. | [144] | ||
| Electrostatic complexation | Sodium caseinate and sodium alginate | Reduced lutein decomposition during storage and increased FFA release and lutein bio accessibility in a simulated GIT. | [145] | |
| Crocin | Spray drying | Gelatin | Increased encapsulation efficiency and preserved crocin physicochemical properties. | [146] |
| Ionotropic gelation | Alginate | High encapsulation efficiency and fast kinetics release. | [147] | |
| Chitosan, gelatin, and oxidized alginate | Hydrogel exhibited high encapsulation efficiency, sustained crocin release, and superior mucoadhesive strength. | [148] | ||
| β-carotene | Spray drying | Gum Arabic, maltodextrin, modified starch, and whey protein | Increased β-carotene half-life and decreased the β-carotene encapsulated fraction. | [149] |
| Complex coacervation | Amaranth carboxymethyl starch and lactoferrin | Enhanced encapsulation efficiency and thermal and photolytic stability. High intestinal release in oily matrices. | [150] | |
| Ionotropic gelation | Sodium caseinate and κ-carrageenan | High encapsulation efficiency and β-carotene retention. Low viscosity. | [151] | |
| W/O/W emulsion | Aggregated insoluble soybean protein hydrolysate and xanthan gum | Increased encapsulation efficiency and good pH, ionic, and thermal stability. Enhanced bioavailability in simulated GIT. | [152] |
Abbreviations: O/W, oil/water; GIT, gastrointestinal tract; ABTS•+, 2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt radical cation; SGF, simulated gastric fluid; FFA, free fatty acids; W/O/W; water/oil/water.
In comparison, common nanomaterials used to entrap carotenoids include polymer-coated liposomes [153], gelatin-based nanoparticles (NPs) [154], and hydrogels synthesized from starch blends [155]. Scientific evidence regarding common carotenoids entrapped into micro- or nanomaterials to treat diseases related to MetS is limited, but it is compiled in the following sections. Table 3 provides examples of nanomaterials used to entrap carotenoids and their biological properties.
Table 3.
Examples of nanoencapsulation systems of carotenoids and the observed activities.
| Carotenoid | Encapsulation System | Raw Materials | Observed Activities | References |
|---|---|---|---|---|
| Astaxanthin | Nanocapsules | Formaldehyde and lysine | Nanocapsules reduced the production of H2O2 and maintain mitochondrial membrane potential. Nanocapsules exhibited stability against high temperatures, pH, and UV radiation. |
[156] |
| NPs | Chitosan and sodium triphosphate | NPs exhibited sustained in vitro release in simulated gastric and intestinal conditions. NPs loaded with astaxanthin executed prolonged residence, time levels, and antioxidant activities in Sprague-Dawley rats. |
[157] | |
| Nanoemulsion | Soybean protein isolate and sodium alginate | Nanoemulsion presented scavenging activity against H2O2 and DPPH radicals. Nanoemulsion exhibited stability upon thermal, light, storage, and gastrointestinal digestion exposure. |
[158] | |
| Fucoxanthin | NPs | Alginate, casein, and chitosan | NPs improved the release of fucoxanthin under simulated gastrointestinal digestion conditions. The membrane permeability of fucoxanthin was enhanced. NPs loaded with fucoxanthin exhibited enhanced plasma levels after oral administration. |
[159] |
| Nanocomplexes | Whey protein | Nanocomplexes protected fucoxanthin against UV-B radiation, heat, and pH. Enhanced ROS accumulation, caused mitochondrial damage, and regulated apoptosis. |
[156] | |
| Lycopene | Nanofibers | Gelatin | Improved water solubility. Enhanced antioxidant activity during 14-day storage. |
[160] |
| NPs | Alginate phosphatidylcholine | Improved lycopene bioavailability upon cellular uptake by Caco-2 cells. Increased efficiency, stability, and dispersion of lycopene. |
[161] | |
| Lipid-core nanocapsules | Tween 80, span 60, poly-ɛ-caprolactone, and coconut oil | Decreased the viability of MCF-7 cells after 24 and 72 h of exposure. Inhibited the activation of NF-κB and reduced ROS production in microglial (HMC3) cells. |
[162] | |
| Lutein | NPs | Stevioside | Entrapment into NPs improved the bioavailability of lutein. Lutein-loaded NPs enter cells by clathrin-mediated endocytosis. |
[163] |
| Nanoemulsion | Linoleic acid, oleic acid, sodium taurocholate, and mono-oleoyl glycerol | Enhanced the aqueous solubility of lutein. Improved the tissue distribution pattern of lutein in liver and eyes of mice models. |
[164] | |
| Crocin | Nanocapsules | Lecithin and chitosan | The sustained release of crocin was promoted. Encapsulation of crocin protective its integrity under in vitro digestion conditions. |
[165] |
| Nanoemulsion | Chitosan and alginate | Nanoemulsion exhibited high stability under stimulated gastric conditions (pH 2) and executed sustained release of crocin. | [166] |
Abbreviations: H2O2, hydrogen peroxide; DPPH, 2,2-diphenyl-1-picrylhydrazyl; NPs, nanoparticles; UV-B, ultraviolet B; ROS, reactive oxygen species; NF-κB; nuclear factor kappa B.
4.1. Astaxanthin
Astaxanthin is a carotenoid composed of forty carbon atoms, fifty-two hydrogen atoms, and four oxygen atoms (C40H52O4). Modern advances have involved encapsulating astaxanthin in micro- and nanomaterials. For instance, NPs prepared with astaxanthin and anthocyanins have demonstrated enhanced activity of superoxidase dismutase (SOD), glutathione peroxidase (GSH-PX), and catalase while reducing fat content, lipofuscin accumulation, and ROS levels on high-fat Caenorhabditis elegans cultures. These properties are appealing for the treatment of DM2 [167]. In obesity, emulsion-based delivery systems containing astaxanthin have been shown to reduce body fat accumulation, hepatic fatty acid, and hepatic lipid levels in obese mice models. These effects are attributed to the high oral absorbability of the prepared system [168].
In other studies, nanocarriers containing astaxanthin, formulated with galactose, whey protein isolate, and triphenyl-phosphonium, have been shown to target mitochondria in steatotic HepG2 cells exhibiting potent anti-adipogenic and antioxidative activities. Moreover, these carriers altered blood lipid levels in NAFLD mouse models and reduced liver lipid accumulation [169]. In rat models treated with CCl4, astaxanthin NPs prepared with lecithin ameliorated hepatic damage and decreased plasma biomarker levels such as aminotransferase and aspartate aminotransferase [170]. Against potential neurological disorders, NPs or nanostructured lipid carriers containing astaxanthin demonstrated protective effects against MPP+ (an active metabolite that mimics Parkinson’s disease), ferric iron, and tert-butyl hydroperoxide in SH-SY5Y cells [171].
On the other hand, microencapsulation has also been explored with this carotenoid. Spray drying utilizing gum Arabic has shown promising effects, including increased solubility, stability, and improved bioavailability in the simulated gastrointestinal tract (GIT). It was observed that variations in inlet and outlet temperatures could impact the encapsulation efficiency, with higher microencapsulation efficiency noted at higher inlet temperatures ranging from 180 to 190 °C. This is attributed to the accelerated formation rate of microcapsules under these conditions [134]. In another study, multilayer O/W emulsions were formed using ι-carrageenan, chitosan, lupine protein isolate, and sunflower oil, followed by spray drying. The effects of this technique include an increase in astaxanthin retention and storage stability, achieved by providing protection against oxidative damage of astaxanthin. It was observed that O/W emulsions exhibit greater physical stability in two- and three-membrane systems compared to single-membrane systems. Additionally, a greater solubility in water was noted [135]. On the other hand, ionic gelation can be employed using low-methoxyl pectin, chitosan, and alginate to form microcapsules. This microencapsulation method improves particle sphericity and limits oil oxidation during formulation. Increased thermal stability and bioavailability in simulated GIT were also observed. The authors concluded that the appropriate ratio of alginate to pectin could result in ideal encapsulation efficiency and bioaccessibility, with these values depending on the concentrations of the polymers [132].
4.2. β-Carotene
The evidence regarding micro- or nanomaterials prepared with isolated or commercially available β-carotene (C40H56) is limited. Previous studies have shown that NPs loaded with carotenoid-rich extracts from cantaloupe melon enhanced the levels of hepatic retinol in Wistar rats with obesity without affecting tissue integrity [172]. On the other hand, β-carotene-based nanoemulsions combined with quercetin have been reported to protect hepatic and pancreatic β-cells against damage, reduce blood sugar levels, influence oral glucose tolerance, control body weight, decrease tissue damage markers such as aspartate aminotransferase and alanine aminotransferase, and enhance SOD levels in diabetic albino Wistar rats [173]. In another study, calcium–alginate beads were fabricated using the extrusion process to enhance the stability and preserve the DPPH antioxidant activity of olive oil, which was enriched with carotenoids such as β-carotene [59].
Similarly, protein-based nanoemulsions have demonstrated the capacity to increase the bioavailability of β-carotene and retinol using in vitro (simulated gastrointestinal digestion) and in vivo (female Sprague-Dawley rats) models [174]. Other formulations to enhance the bioavailability of β-carotene have been observed with emulsion formed with insoluble soybean, protein hydrolysate, and xanthan gum, which exhibited enhanced thermal, ionic, and pH stability while promoting the bioavailability of this compound under simulated gastric conditions [152].
The microencapsulation of this carotenoid with gum Arabic, maltodextrin, modified starch, and whey protein was tested using the spray drying technique. The results showed an increased half-life of β-carotene and a decreased fraction of encapsulated β-carotene. It was also observed that the agglomeration of the microparticles positively affected their solubility in water [149]. Another technique tested with this carotenoid was complex coacervation with Amaranth carboxymethyl starch and lactoferrin. Enhanced encapsulation efficiency, thermal protection at 50 °C, and photolytic stability against UV radiation were observed. Likewise, a high intestinal release in oily matrices was observed, along with carotenoid protection against gastrointestinal conditions [150].
Likewise, ionotropic gelation was tested using sodium caseinate and κ-carrageenan as materials. High encapsulation efficiency, β-carotene retention, and low viscosity were observed. Some losses of the carotenoid were noted due to probable escapes of oil droplets during the formation of the microparticles in the gelation process. Additionally, it was observed that the polysaccharide concentration influenced the morphology of the particles, while the pH did not significantly affect characteristics such as morphology and encapsulation efficiency [151]. On the other hand, aggregated insoluble soybean protein hydrolysate and xanthan gum were used to create a W/O/W emulsion. An increased encapsulation efficiency (70.56 ± 0.06%) and suitable pH values (5 to 11), ionic stability, and thermal stability (25 to 85 °C) were observed. Similarly, the technique enhanced bioavailability in simulated GIT. An advantage of the technique is its ability to protect and deliver both hydrophilic and hydrophobic components, as is the case of this carotenoid [152].
4.3. Crocin
The effect of microencapsulation on crocin (C44H64O24) has been tested using spray drying with gelatin as an ingredient. A direct proportionality was noted between the gelatin concentration and particle size, making obtaining smaller microcapsules with lower gelatin concentrations possible. As a result, increased encapsulation efficiency and preserved crocin physicochemical properties were observed [146]. On the other hand, ionotropic gelation was used with alginate as the primary material. This resulted in high encapsulation efficiency and fast kinetics release for the microparticles obtained. Crocin, unlike the carotenoids discussed above, is soluble in water. However, with microencapsulation, it was observed that the release of the molecule in an aqueous environment could be controlled, presenting an advantage of using this technique [147]. In another study, the ionotropic gelation technique was used with chitosan, gelatin, and oxidized alginate as materials. The observed effects of the hydrogel were high encapsulation efficiency, sustained crocin release, and superior mucoadhesive strength. It was also proven that with a lower concentration of chitosan, there was a more efficient gelation process since the mixture’s viscosity was reduced, and the solubility of crocin was favored [148].
4.4. Fucoxanthin
Fucoxanthin is a carotenoid with forty-two carbon, fifty-eight hydrogen, and six oxygen atoms (C42H58O6). Over the last decades, fucoxanthin has been entrapped into structures of different materials. Recently, it was documented that fucoxanthin and zein hydrolysate encapsulated into NPs diminished liver pathology, decreased blood glucose levels, influenced the expression of genes (e.g., GLP-1, GLUT2, and PI3K) related to glucose metabolism, and improved the restoration of intestinal microorganisms correlated to the gut microbiota in C57BL/6J mice [175]. In other studies, fucoxanthin into hydrolyzed zein nanocomplexes has been reported to decrease and restore fasting blood glucose levels in DM2 mice (C57BL/6) while affecting oxidative stress, enhancing SOD and GSH-PX levels, upregulating GLUT2 expression, promoting repair of hepatocyte and pancreatic β-cell damage, and regulating glycogen synthesis in the same model [176].
Using gums and proteins of natural origin, gum Arabic/gelatin microcapsules containing fucoxanthin and incorporated into alginate hydrogel beads decreased body weight. It lowered blood lipid content (high-density lipoprotein cholesterol, total cholesterol, and low-density lipoprotein cholesterol) and oxidative stress levels in specific-pathogen-free (SPF) grade Kunming (KM) mice models after oral administration. In the same study, treatment with microcapsules was biocompatible with L929 cells [138]. On the other hand, microencapsulation with maltodextrin, soy lecithin, and gum Arabic through spray drying and freeze drying has resulted in microcapsules with high encapsulation efficiency and increased bioavailability, as well as ABTS•+ scavenging activity.
Furthermore, it was observed that the microcapsules obtained by freeze drying had a higher encapsulation efficiency than those obtained by spray drying, resulting in a higher concentration of the carotenoid and increased observed activity [136]. In another study, microencapsulation through sequential coating modification using maltodextrin and gum Arabic enhanced encapsulation efficiency and fucoxanthin stability. The authors describe that this technique increased stability against damage by light and heat, making it viable for use in several industries where fucoxanthin is exposed to these conditions [137]. Additionally, the efficacy of complex carriers was evaluated using gum Arabic, gelatin, and alginate hydrogel as materials, and their in vitro and in vivo activity was tested. Increased bioavailability and fucoxanthin protection against simulated gastric fluid (SGF) were observed in in vitro tests. On the other hand, in in vivo tests, a decrease in blood lipids and oxidative stress levels was demonstrated after oral administration. Furthermore, fucoxanthin accumulation in the intestinal tract also decreased weight in rats fed a high-lipid diet [138].
On the other hand, nanocomplexes loaded with fucoxanthin composed of gelatin and chitosan oligosaccharides have been shown to reduce the generation of ROS, decrease mitochondrial damage upon exposure to H2O2, inhibit apoptosis, and regulate lipid metabolism in L02 cells [177]. In another study, fucoxanthin isolated from Sargassum wightii Greville exhibited antidiabetic and antihypertensive properties by reducing oxidative stress, preserving pancreatic tissue morphology, and reducing hyperglycemic condition [178].
4.5. Lycopene
Lycopene (C40H56), a carotenoid commonly present in tomato skin, is susceptible to environmental factors such as light and oxygen degradation. In recent studies, lycopene was incorporated into liposomes loaded with tobramycin to develop a multifunctional antibiotic hydrogel. This hydrogel demonstrated the ability to inhibit bacterial proliferation, promote cell migration and angiogenesis, stimulate collagen deposition, and enhance wound healing in an infected diabetic wound model in rats [179].
In another study, lycopene was initially encapsulated within liposomes and then integrated into a porous chitosan microgel along with nicotinamide mononucleotide. This formulation was found to exert protective effects against acute liver injury in C547BL/6 mice and regulate gut microbiota [180]. The protective effect on the liver was attributed to the microgel’s ability to inhibit the TLR4/NF-κB signaling pathway and interact with the TLR4/MD2 complex, reducing inflammatory and oxidative stress responses. Similarly, sea buckthorn juices containing lycopene were entrapped within microspheres derived from sodium alginate and κ-carrageenan. These microspheres were reported to regulate the release and control of blood glucose levels by inhibiting the activity of α-amylase and α-glucosidase, respectively [141].
Regarding nanomaterials, lycopene loaded into H-ferritin nanocages, along with triphenylphosphonium, has been reported as a novel targeted therapy for intracerebral and intra-neuronal treatment to address memory loss and neuronal dysfunction. This formulation can regulate mitochondrial function in nerve cells, preserve mitochondrial morphology, prevent memory decline, and promote synaptic plasticity in aging mice [181]. Similarly, treatment with sequence-targeted lycopene nanodots encapsulated within H-ferritin nanocages protected mitochondrial functionality by enhancing ROS scavenging activity and promoting neural enrichment and mitochondrial regulation in C57/BL mice [182]. Additionally, these nanodots exhibited pro-survival mitophagy activities and induced the degradation of pathogenic α-synuclein.
Since microencapsulation has been another method of protecting molecules with specific bioactivity, experiments have been reported on preparing lycopene microcapsules. One study evaluated microencapsulation with gum Arabic and inulin using spray drying. The microcapsules demonstrated high lycopene release in simulated gastric fluid. However, a decrease in antioxidant activity was observed, probably due to exposure of the carotenoid-containing solutions to environmental factors such as oxygen and light during the microcapsule preparation process. Furthermore, the encapsulated extract showed more excellent stability against light and oxygen than the pure extract when both were stored for 27 days [139,140]. On the other hand, lycopene microencapsulation was achieved through ionotropic gelation using alginate and κ-carrageenan in another experiment. In a separate experiment, whey proteins and acacia gum were utilized to produce microcapsules through complex coacervation and freeze drying. The observed effect was increased antioxidant activity, and in vitro α-amylase and α-glucosidase inhibitory activity was demonstrated, with a greater effect on α-amylase. The observed enzymatic activity can be categorized as a preventive effect against DM2 and MetS [142].
4.6. Lutein
Lutein is a linear xanthophyll with forty carbon atoms, fifty-six hydrogen atoms, and two oxygens (C40H56O2). In an innovative study, lutein was encapsulated into chitosan-coated liposomes and further reported to decrease the levels of ROS and enhance the performance of SOD and GHS-PX in vitro. In the same study, treatment with lutein diminished total cholesterol and triglycerides levels, body weight, and fat accumulation in epididymal adipose tissue and liver of high-fat diet C57BL/6J mice [183]. Similarly, lutein-coated poly (lactic-co-glycolic acid) NPs coated with macrophage membranes inhibited angiotensin II-induced primary cardiac fibroblast proliferation, improved cardiac function and structure, and regulated cardiac fibrosis without causing significant toxicity in C57BL/6 male mice [184].
Regarding microencapsulation, inulin and modified starch were used to produce powdered microcapsules via spray drying, enhancing lutein stability and resistance to thermal degradation. In recent studies, the spray drying technique was employed with citric acid-esterified potato starch and whey protein as the main components. Microcapsules produced via this method exhibited increased encapsulation efficiency, embedding effect, lutein aqueous solubility, and thermal resistance. It was observed that smaller microcapsules had better solubility in water. Therefore, it can be inferred that the choice of materials can influence the effectiveness of the encapsulation technique, even when the same spray drying method is utilized [144]. Similarly, sodium caseinate and alginate were utilized to microencapsulate lutein through electrostatic complexation. The observed effects included reduced lutein decomposition during storage compared to carotenoid emulsions. It was evident that degradation due to oxidative damage was mitigated by the protective barrier, preventing free radicals from affecting the lutein molecule. Furthermore, increased FFA release and lutein bioaccessibility were observed in simulated GIT [145].
5. Conclusions
Carotenoids, ubiquitous bioactive compounds, have been extensively utilized in formulating products significantly impacting global health care and economic sectors. Evidence regarding the prevalence of MetS primarily focuses on patients diagnosed with diabetes, obesity, or cardiovascular diseases. Present therapeutic approaches are tailored to specific MetS-related conditions, often presenting due to the potential for AEs.
In recent decades, the utilization of carotenoids has emerged as an appealing option for mitigating or preventing the onset of MetS. This is attributed to their advantageous role in regulating lipid metabolism, reducing blood sugar levels, augmenting the activity of antioxidant enzymes, and averting tissue and cellular damage. Nonetheless, their application is impeded by their vulnerability to degradation caused by environmental and physiological factors.
From the available literature, it is evident that carotenoids are primarily sourced from biological materials, with the UAE method being the preferred extraction technique. However, their characterization using spectroscopy techniques is still limited, which poses a challenge in gaining a comprehensive understanding of their chemical features and biological performance. This limitation opens avenues for innovative research. Moreover, the use of micro- and nanomaterials to address these limitations and their evaluation in models that simulate MetS conditions is a relatively new approach, with only a few studies meeting these criteria. Furthermore, in cases where studies focus on the use of carotenoids and materials with micro- or nanostructured arrangements, they often struggle to establish clear relationships between morphology or surface charge and the activity of the entrapped substance against MetS models. This highlights the need for further investigation in this area to elucidate the mechanisms underlying their therapeutic potential.
Acknowledgments
J.L.M.-M. is grateful to Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT) for his doctoral fellowship. All figures were created using BioRender.com.
Author Contributions
Conceptualization, A.E.O.-R., C.A.d.P. and J.D.L.-R.; validation, J.D.L.-R.; formal analysis, J.A.M.-T., K.S.-C. and J.L.M.-M.; investigation, A.E.O.-R., J.A.M.-T., K.S.-C. and J.L.M.-M.; resources, A.E.O.-R., C.A.d.P. and J.D.L.-R.; data curation, A.E.O.-R., J.A.M.-T., K.S.-C. and J.L.M.-M.; writing—original draft preparation, A.E.O.-R., J.A.M.-T., K.S.-C. and J.L.M.-M.; writing—review and editing, A.E.O.-R., C.A.d.P., E.R.L.-M. and J.D.L.-R.; visualization, K.S.-C. and J.L.M.-M.; supervision, A.E.O.-R., C.A.d.P. and J.D.L.-R.; project administration, A.E.O.-R. and J.D.L.-R.; funding acquisition, C.A.d.P., E.R.L.-M. and J.D.L.-R. All authors have read and agreed to the published version of the manuscript.
Conflicts of Interest
The authors declare no conflicts of interest.
Funding Statement
This research received no external funding.
Footnotes
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.
References
- 1.Mohamed S.M., Shalaby M.A., El-Shiekh R.A., El-Banna H.A., Emam S.R., Bakr A.F. Metabolic Syndrome: Risk Factors, Diagnosis, Pathogenesis, and Management with Natural Approaches. Food Chem. Adv. 2023;3:100335. doi: 10.1016/j.focha.2023.100335. [DOI] [Google Scholar]
- 2.Al Shehri H.A., Al Asmari A.K., Khan H.A., Al Omani S., Kadasah S.G., Horaib G.B., Al Buraidi A., Al Sharif A.A., Mohammed F.S., Abbasmanthiri R., et al. Association between Preventable Risk Factors and Metabolic Syndrome. Open Med. 2022;17:341–352. doi: 10.1515/med-2021-0397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Liang X., Or B., Tsoi M.F., Cheung C.L., Cheung B.M.Y. Prevalence of Metabolic Syndrome in the United States National Health and Nutrition Examination Survey 2011–18. Postgrad. Med. J. 2023;99:985–992. doi: 10.1093/postmj/qgad008. [DOI] [PubMed] [Google Scholar]
- 4.Manaf M.R.A., Nawi A.M., Tauhid N.M., Othman H., Rahman M.R.A., Yusoff H.M., Safian N., Ng P.Y., Manaf Z.A., Kadir N.B.A., et al. Prevalence of Metabolic Syndrome and Its Associated Risk Factors among Staffs in a Malaysian Public University. Sci. Rep. 2021;11:8132. doi: 10.1038/s41598-021-87248-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Belete R., Ataro Z., Abdu A., Sheleme M. Global Prevalence of Metabolic Syndrome among Patients with Type I Diabetes Mellitus: A Systematic Review and Meta-Analysis. Diabetol. Metab. Syndr. 2021;13:25. doi: 10.1186/s13098-021-00641-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zujko M.E., Rożniata M., Zujko K. Individual Diet Modification Reduces the Metabolic Syndrome in Patients before Pharmacological Treatment. Nutrients. 2021;13:2102. doi: 10.3390/nu13062102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Aguilar-Salinas C.A., Viveros-Ruiz T. Recent Advances in Managing/Understanding the Metabolic Syndrome. F1000Research. 2019;8:370. doi: 10.12688/f1000research.17122.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.von Lintig J., Moon J., Lee J., Ramkumar S. Carotenoid Metabolism at the Intestinal Barrier. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids. 2020;1865:158580. doi: 10.1016/j.bbalip.2019.158580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Crupi P., Faienza M.F., Naeem M.Y., Corbo F., Clodoveo M.L., Muraglia M. Overview of the Potential Beneficial Effects of Carotenoids on Consumer Health and Well-Being. Antioxidants. 2023;12:1069. doi: 10.3390/antiox12051069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zheng W., Yu S., Zhang W., Zhang S., Fu J., Ying H., Pingcuo G., Liu S., Zhao F., Wu Q., et al. The Content and Diversity of Carotenoids Associated with High-Altitude Adaptation in Tibetan Peach Fruit. Food Chem. 2023;398:133909. doi: 10.1016/j.foodchem.2022.133909. [DOI] [PubMed] [Google Scholar]
- 11.Dzomeku B.M., Wald J.P., Wünsche J.N., Nohr D., Biesalski H.K. Climate Change Enhanced Carotenoid Pro-Vitamin A Levels of Selected Plantain Cultivars. Plants. 2020;9:541. doi: 10.3390/plants9040541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhou B., Perel P., Mensah G.A., Ezzati M. Global Epidemiology, Health Burden and Effective Interventions for Elevated Blood Pressure and Hypertension. Nat. Rev. Cardiol. 2021;18:785–802. doi: 10.1038/s41569-021-00559-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Generalić Mekinić I., Šimat V., Rathod N.B., Hamed I., Čagalj M. Algal Carotenoids: Chemistry, Sources, and Application. Foods. 2023;12:2768. doi: 10.3390/foods12142768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Meléndez-Martínez A.J., Mapelli-Brahm P., Hornero-Méndez D., Vicario I.M. Structures, Nomenclature and General Chemistry of Carotenoids and Their Esters. In: Mercadante A.Z., editor. Foods: Physical, Chemical and Biological Properties. Royal Society of Chemistry; London, UK: 2019. [DOI] [Google Scholar]
- 15.Riaz M., Zia-Ul-Haq M., Dou D. Chemistry of Carotenoids. In: Zia-Ul-Haq M., Dewanjee S., Riaz M., editors. Carotenoids: Structure and Function in the Human Body. Springer International Publishing; Cham, Switzerland: 2021. pp. 43–76. [Google Scholar]
- 16.Saini R.K., Prasad P., Lokesh V., Shang X., Shin J., Keum Y.-S., Lee J.-H. Carotenoids: Dietary Sources, Extraction, Encapsulation, Bioavailability, and Health Benefits—A Review of Recent Advancements. Antioxidants. 2022;11:795. doi: 10.3390/antiox11040795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sereti F., Alexandri M., Papadaki A., Papapostolou H., Kopsahelis N. Carotenoids Production by Rhodosporidium paludigenum Yeasts: Characterization of Chemical Composition, Antioxidant and Antimicrobial Properties. J. Biotechnol. 2024;386:52–63. doi: 10.1016/j.jbiotec.2024.03.011. [DOI] [PubMed] [Google Scholar]
- 18.Polyakov N.E., Focsan A.L., Gao Y., Kispert L.D. The Endless World of Carotenoids—Structural, Chemical and Biological Aspects of Some Rare Carotenoids. Int. J. Mol. Sci. 2023;24:9885. doi: 10.3390/ijms24129885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Metibemu D.S., Ogungbe I.V. Carotenoids in Drug Discovery and Medicine: Pathways and Molecular Targets Implicated in Human Diseases. Molecules. 2022;27:6005. doi: 10.3390/molecules27186005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Popov A.M., Kozlovskaya E.P., Klimovich A.A., Rutckova T.A., Vakhrushev A.I., Hushpulian D.M., Gazaryan I.G., Makhankov V.V., Son O.M., Tekutyeva L.A. Carotenoids from Starfish Patiria Pectinifera: Therapeutic Activity in Models of Inflammatory Diseases. Mar. Drugs. 2023;21:470. doi: 10.3390/md21090470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ferraz C.A.A., Grougnet R., Nicolau E., Picot L., de Oliveira R.G., Jr. Carotenoids from Marine Microalgae as Antimelanoma Agents. Mar. Drugs. 2022;20:618. doi: 10.3390/md20100618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Malhão F., Macedo A.C., Costa C., Rocha E., Ramos A.A. Fucoxanthin Holds Potential to Become a Drug Adjuvant in Breast Cancer Treatment: Evidence from 2D and 3D Cell Cultures. Molecules. 2021;26:4288. doi: 10.3390/molecules26144288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Robles-Rivera R.R., Castellanos-González J.A., Olvera-Montaño C., Flores-Martin R.A., López-Contreras A.K., Arevalo-Simental D.E., Cardona-Muñoz E.G., Roman-Pintos L.M., Rodríguez-Carrizalez A.D. Adjuvant Therapies in Diabetic Retinopathy as an Early Approach to Delay Its Progression: The Importance of Oxidative Stress and Inflammation. Oxidative Med. Cell. Longev. 2020;2020:e3096470. doi: 10.1155/2020/3096470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Islam F., Khan J., Zehravi M., Das R., Haque M.A., Banu A., Parwaiz S., Nainu F., Nafady M.H., Shahriar S.M.S., et al. Synergistic Effects of Carotenoids: Therapeutic Benefits on Human Health. Process Biochem. 2024;136:254–272. doi: 10.1016/j.procbio.2023.11.033. [DOI] [Google Scholar]
- 25.Lim M., Kim J. Association between Fruit and Vegetable Consumption and Risk of Metabolic Syndrome Determined Using the Korean Genome and Epidemiology Study (KoGES) Eur. J. Nutr. 2020;59:1667–1678. doi: 10.1007/s00394-019-02021-5. [DOI] [PubMed] [Google Scholar]
- 26.Zhu X., Cheang I., Tang Y., Shi M., Zhu Q., Gao R., Liao S., Yao W., Zhou Y., Zhang H., et al. Associations of Serum Carotenoids with Risk of All-Cause and Cardiovascular Mortality in Hypertensive Adults. J. Am. Heart Assoc. 2023;12:e027568. doi: 10.1161/JAHA.122.027568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gopal S.S., Sukhdeo S.V., Vallikannan B., Ponesakki G. Lutein Ameliorates High-Fat Diet-Induced Obesity, Fatty Liver, and Glucose Intolerance in C57BL/6J Mice. Phytother. Res. 2023;37:329–341. doi: 10.1002/ptr.7615. [DOI] [PubMed] [Google Scholar]
- 28.Yamamoto Y., Narumi K., Yamagishi N., Nishi T., Ito T., Iseki K., Kobayashi M., Kanai Y. Oral Administration of Linoleic Acid Immediately before Glucose Load Ameliorates Postprandial Hyperglycemia. Front. Pharmacol. 2023;14:1197743. doi: 10.3389/fphar.2023.1197743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Boonlao N., Ruktanonchai U.R., Anal A.K. Enhancing Bioaccessibility and Bioavailability of Carotenoids Using Emulsion-Based Delivery Systems. Colloids Surf. B Biointerfaces. 2022;209:112211. doi: 10.1016/j.colsurfb.2021.112211. [DOI] [PubMed] [Google Scholar]
- 30.Carotenoid-Loaded Nanocarriers: A Comprehensive Review—ScienceDirect. [(accessed on 15 April 2024)]. Available online: https://www.sciencedirect.com/science/article/pii/S0001868619302738?via%3Dihub.
- 31.González-Peña M.A., Lozada-Ramírez J.D., Ortega-Regules A.E. Antioxidant Activities of Spray-Dried Carotenoids Using Maltodextrin-Arabic Gum as Wall Materials. Bull. Natl. Res. Cent. 2021;45:58. doi: 10.1186/s42269-021-00515-z. [DOI] [Google Scholar]
- 32.Tamtürk F., Gürbüz B., Toker Ö.S., Dalabasmaz S., Malakjani N., Durmaz Y., Konar N. Optimization of Chlorella vulgaris Spray Drying Using Various Innovative Wall Materials. Algal Res. 2023;72:103115. doi: 10.1016/j.algal.2023.103115. [DOI] [Google Scholar]
- 33.Cabezas-Terán K., Grootaert C., Ortiz J., Donoso S., Ruales J., Van Bockstaele F., Van Camp J., Van de Wiele T. In Vitro Bioaccessibility and Uptake of β-Carotene from Encapsulated Carotenoids from Mango by-Products in a Coupled Gastrointestinal Digestion/Caco-2 Cell Model. Food Res. Int. 2023;164:112301. doi: 10.1016/j.foodres.2022.112301. [DOI] [PubMed] [Google Scholar]
- 34.Honda M., Zhang Y., Kageyama H., Hibino T., Goto M., Nishida Y. Formation and Characterization of Z-Isomer-Enriched Carotenoid-Loaded Microparticles with Poly(Vinylpyrrolidone) Using a Spray Drying Technique. Ind. Eng. Chem. Res. 2024;63:383–393. doi: 10.1021/acs.iecr.3c03450. [DOI] [Google Scholar]
- 35.Sørensen T.I.A., Martinez A.R., Jørgensen T.S.H. Epidemiology of Obesity. In: Eckel J., Clément K., editors. From Obesity to Diabetes. Springer International Publishing; Cham, Switzerland: 2022. pp. 3–27. [Google Scholar]
- 36.Shi M., Zhang X., Wang H. The Prevalence of Diabetes, Prediabetes and Associated Risk Factors in Hangzhou, Zhejiang Province: A Community-Based Cross-Sectional Study. Diabetes Metab. Syndr. Obes. 2022;15:713–721. doi: 10.2147/DMSO.S351218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Åberg F., Byrne C.D., Pirola C.J., Männistö V., Sookoian S. Alcohol Consumption and Metabolic Syndrome: Clinical and Epidemiological Impact on Liver Disease. J. Hepatol. 2023;78:191–206. doi: 10.1016/j.jhep.2022.08.030. [DOI] [PubMed] [Google Scholar]
- 38.Martemucci G., Fracchiolla G., Muraglia M., Tardugno R., Dibenedetto R.S., D’Alessandro A.G. Metabolic Syndrome: A Narrative Review from the Oxidative Stress to the Management of Related Diseases. Antioxidants. 2023;12:2091. doi: 10.3390/antiox12122091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Raut S.K., Khullar M. Oxidative Stress in Metabolic Diseases: Current Scenario and Therapeutic Relevance. Mol. Cell. Biochem. 2023;478:185–196. doi: 10.1007/s11010-022-04496-z. [DOI] [PubMed] [Google Scholar]
- 40.Kumar A., Prajapati P., Singh G., Kumar D., Mishra V., Kim S.-C., Raorane C.J., Raj V., Kushwaha S. Salbutamol Attenuates Diabetic Skeletal Muscle Atrophy by Reducing Oxidative Stress, Myostatin/GDF-8, and Pro-Inflammatory Cytokines in Rats. Pharmaceutics. 2023;15:2101. doi: 10.3390/pharmaceutics15082101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cojocaru K.-A., Luchian I., Goriuc A., Antoci L.-M., Ciobanu C.-G., Popescu R., Vlad C.-E., Blaj M., Foia L.G. Mitochondrial Dysfunction, Oxidative Stress, and Therapeutic Strategies in Diabetes, Obesity, and Cardiovascular Disease. Antioxidants. 2023;12:658. doi: 10.3390/antiox12030658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Huang Y., Kyriakides T.R. The Role of Extracellular Matrix in the Pathophysiology of Diabetic Wounds. Matrix Biol. Plus. 2020;6–7:100037. doi: 10.1016/j.mbplus.2020.100037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Santos I.S.R., Martin-Pastor M., Tavares Júnior A.G., Queiroz K.A., da Silva Sólon L.G., Sousa F.F.O.d. Metabolomic Profile and Its Correlation with the Plasmatic Levels of Losartan, EXP3174 and Blood Pressure Control in Hypertensive and Chronic Kidney Disease Patients. Int. J. Mol. Sci. 2023;24:9832. doi: 10.3390/ijms24129832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fishel Bartal M., Blackwell S.C., Pedroza C., Lawal D., Amro F., Samuel J., Chauhan S.P., Sibai B.M. Oral Combined Hydrochlorothiazide and Lisinopril vs Nifedipine for Postpartum Hypertension: A Comparative-Effectiveness Pilot Randomized Controlled Trial. Am. J. Obstet. Gynecol. 2023;228:571.e1–571.e10. doi: 10.1016/j.ajog.2023.01.015. [DOI] [PubMed] [Google Scholar]
- 45.Li L., Tong X., Ma Z., Lv L., Liu H., Chen G.L. Folic Acid Enhances the Cardiovascular Protective Effect of Amlodipine in Renal Hypertensive Rats with Elevated Homocysteine. Clin. Exp. Hypertens. 2023;45:2205058. doi: 10.1080/10641963.2023.2205058. [DOI] [PubMed] [Google Scholar]
- 46.Tandon S., Sarkar S. Glipizide Ameliorates Human Poly(Q) Mediated Neurotoxicity by Upregulating Insulin Signalling in Drosophila Disease Models. Biochem. Biophys. Res. Commun. 2023;645:88–96. doi: 10.1016/j.bbrc.2023.01.022. [DOI] [PubMed] [Google Scholar]
- 47.Liu Z. Efficacy of Metformin Combined with Liraglutide on the Glucose and Lipid Metabolism, Vascular Endothelial Function, and Oxidative Stress of Patients with T2DM and Metabolic Syndrome. Pak. J. Med. Sci. 2024;40:26–30. doi: 10.12669/pjms.40.1.7936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jin Z.-J., Wang G.-Z. Clinical Efficacy of Dapagliflozin in the Treatment of Patients with Diabetic Nephropathy and Its Effect on Proteinuria Level. Diabetes Metab. Syndr. Obes. 2023;16:2167–2175. doi: 10.2147/DMSO.S421579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Liu S., Lin X., Tao M., Chen Q., Sun H., Han Y., Yang S., Gao Y., Qu S., Chen H. Efficacy and Safety of Orlistat in Male Patients with Overweight/Obesity and Hyperuricemia: Results of a Randomized, Double-Blind, Placebo-Controlled Trial. Lipids Health Dis. 2024;23:77. doi: 10.1186/s12944-024-02047-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bays H.E., Hsia D.S., Nguyen L.T., Peterson C.A., Varghese S.T. Effects of Phentermine/Topiramate Extended-Release, Phentermine, and Placebo on Ambulatory Blood Pressure Monitoring in Adults with Overweight or Obesity: A Randomized, Multicenter, Double-Blind Study. Obes. Pillars. 2024;9:100099. doi: 10.1016/j.obpill.2024.100099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Murvelashvili N., Xie L., Schellinger J.N., Mathew M.S., Marroquin E.M., Lingvay I., Messiah S.E., Almandoz J.P. Effectiveness of Semaglutide versus Liraglutide for Treating Post-Metabolic and Bariatric Surgery Weight Recurrence. Obesity. 2023;31:1280–1289. doi: 10.1002/oby.23736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wagner S., Brierley D.I., Leeson-Payne A., Jiang W., Chianese R., Lam B.Y.H., Dowsett G.K.C., Cristiano C., Lyons D., Reimann F., et al. Obesity Medication Lorcaserin Activates Brainstem GLP-1 Neurons to Reduce Food Intake and Augments GLP-1 Receptor Agonist Induced Appetite Suppression. Mol. Metab. 2023;68:101665. doi: 10.1016/j.molmet.2022.101665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Insani W.N., Whittlesea C., Ju C., Man K.K., Adesuyan M., Chapman S., Wei L. Impact of ACEIs and ARBs-Related Adverse Drug Reaction on Patients’ Clinical Outcomes: A Cohort Study in UK Primary Care. Br. J. Gen. Pr. 2023;73:e832–e842. doi: 10.3399/BJGP.2023.0153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Liu L., Chen J., Wang L., Chen C., Chen L. Association between Different GLP-1 Receptor Agonists and Gastrointestinal Adverse Reactions: A Real-World Disproportionality Study Based on FDA Adverse Event Reporting System Database. Front. Endocrinol. 2022;13:1043789. doi: 10.3389/fendo.2022.1043789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Koina I.M., Sarigiannis Y., Hapeshi E. Green Extraction Techniques for the Determination of Active Ingredients in Tea: Current State, Challenges, and Future Perspectives. Separations. 2023;10:121. doi: 10.3390/separations10020121. [DOI] [Google Scholar]
- 56.Martins R., Barbosa A., Advinha B., Sales H., Pontes R., Nunes J. Green Extraction Techniques of Bioactive Compounds: A State-of-the-Art Review. Processes. 2023;11:2255. doi: 10.3390/pr11082255. [DOI] [Google Scholar]
- 57.Majid I., Khan S., Aladel A., Dar A.H., Adnan M., Khan M.I., Mahgoub Awadelkareem A., Ashraf S.A. Recent Insights into Green Extraction Techniques as Efficient Methods for the Extraction of Bioactive Components and Essential Oils from Foods. CyTA J. Food. 2023;21:101–114. doi: 10.1080/19476337.2022.2157492. [DOI] [Google Scholar]
- 58.Menezes Silva J.V., Silva Santos A., Araujo Pereira G., Campos Chisté R. Ultrasound-Assisted Extraction Using Ethanol Efficiently Extracted Carotenoids from Peels of Peach Palm Fruits (Bactris Gasipaes Kunth) without Altering Qualitative Carotenoid Profile. Heliyon. 2023;9:e14933. doi: 10.1016/j.heliyon.2023.e14933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Savic Gajic I.M., Savic I.M., Gajic D.G., Dosic A. Ultrasound-Assisted Extraction of Carotenoids from Orange Peel Using Olive Oil and Its Encapsulation in Ca-Alginate Beads. Biomolecules. 2021;11:225. doi: 10.3390/biom11020225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vo T.P., Tran H.K.L., Ta T.M.N., Nguyen H.T.V., Phan T.H., Nguyen T.H.P., Nguyen V.K., Dang T.C.T., Nguyen L.G.K., Chung T.Q., et al. Extraction and Emulsification of Carotenoids from Carrot Pomaces Using Oleic Acid. ACS Omega. 2023;8:39523–39534. doi: 10.1021/acsomega.3c05301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Larocca V., Martino M., Trupo M., Magarelli R.A., Spagnoletta A., Ambrico A. Evaluation of Carbon Dioxide Supercritical Fluid Extraction (CO2-SFE) on Carotenoids Recovery from Red Yeast Cells. Biomass Conv. Bioref. 2023 doi: 10.1007/s13399-023-04434-z. [DOI] [Google Scholar]
- 62.Priyadarsani S., Patel A.S., Kar A., Dash S. Process Optimization for the Supercritical Carbondioxide Extraction of Lycopene from Ripe Grapefruit (Citrus paradisi) Endocarp. Sci. Rep. 2021;11:10273. doi: 10.1038/s41598-021-89772-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Alvarenga G.L., Cuevas M.S., Capellini M.C., Crevellin E.J., de Moraes L.A.B., Rodrigues C.E.d.C. Extraction of Carotenoid-Rich Palm Pressed Fiber Oil Using Mixtures of Hydrocarbons and Short Chain Alcohols. Food Res. Int. 2020;128:108810. doi: 10.1016/j.foodres.2019.108810. [DOI] [PubMed] [Google Scholar]
- 64.Naveira-Pazos C., Veiga M.C., Mussagy C.U., Farias F.O., Kennes C., Pereira J.F.B. Carotenoids Production and Extraction from Yarrowia lipolytica Cells: A Biocompatible Approach Using Biosolvents. Sep. Purif. Technol. 2024;343:127136. doi: 10.1016/j.seppur.2024.127136. [DOI] [Google Scholar]
- 65.Radice R.P., Padula M.C., Liguori A., D’Arienzo G., Martelli G. Genetic Improvement to Obtain Specialized Haematococcus Pluvialis Genotypes for the Production of Carotenoids, with Particular Reference to Astaxanthin. Int. J. Plant Biol. 2023;14:276–285. doi: 10.3390/ijpb14010023. [DOI] [Google Scholar]
- 66.Chekanov K. Diversity and Distribution of Carotenogenic Algae in Europe: A Review. Mar. Drugs. 2023;21:108. doi: 10.3390/md21020108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Centini M., Martinez-Sañudo I., Biagi M., Dreassi E., Mazzon L., Marri L. Brevundimonas Aurantiaca M3d10, Isolated from the Olive Fly, Produces Hydroxylated Astaxanthin. Cosmetics. 2023;10:103. doi: 10.3390/cosmetics10040103. [DOI] [Google Scholar]
- 68.Asker D. Isolation and Characterization of a Novel, Highly Selective Astaxanthin-Producing Marine Bacterium. J. Agric. Food Chem. 2017;65:9101–9109. doi: 10.1021/acs.jafc.7b03556. [DOI] [PubMed] [Google Scholar]
- 69.Mohammed Y., Ye D., He M., Wang H., Zhu Z., Sun Y. Production of Astaxanthin by Animal Cells via Introduction of an Entire Astaxanthin Biosynthetic Pathway. Bioengineering. 2023;10:1073. doi: 10.3390/bioengineering10091073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.LaCourse W.R., LaCourse M.E. General Instrumentation in HPLC. In: Fanali S., Chankvetadze B., Haddad P.R., Poole C.F., Riekkola M.-L., editors. Liquid Chromatography. 3rd ed. Volume 1. Elsevier; Amsterdam, The Netherlands: 2023. pp. 61–73. Handbooks in Separation Science. [Google Scholar]
- 71.Nahar L., Onder A., Sarker S.D. A Review on the Recent Advances in HPLC, UHPLC and UPLC Analyses of Naturally Occurring Cannabinoids (2010–2019) Phytochem. Anal. 2020;31:413–457. doi: 10.1002/pca.2906. [DOI] [PubMed] [Google Scholar]
- 72.Queral-Beltran A., Marín-García M., Lacorte S., Tauler R. UV-Vis Absorption Spectrophotometry and LC-DAD-MS-ESI(+)-ESI(−) Coupled to Chemometrics Analysis of the Monitoring of Sulfamethoxazole Degradation by Chlorination, Photodegradation, and Chlorination/Photodegradation. Anal. Chim. Acta. 2023;1276:341563. doi: 10.1016/j.aca.2023.341563. [DOI] [PubMed] [Google Scholar]
- 73.Lv J., Wu Q., Li S., Yi H., Xie F. Development and Validation of a UPLC-PDA Method for Quantifying Ceftazidime in Dried Blood Spots. J. Pharm. Biomed. Anal. 2024;239:115928. doi: 10.1016/j.jpba.2023.115928. [DOI] [PubMed] [Google Scholar]
- 74.Xie F., Liu L., Wang Y., Peng Y., Li S. An UPLC-PDA Assay for Simultaneous Determination of Seven Antibiotics in Human Plasma. J. Pharm. Biomed. Anal. 2022;210:114558. doi: 10.1016/j.jpba.2021.114558. [DOI] [PubMed] [Google Scholar]
- 75.Muchiri R.N., van Breemen R.B. Drug Discovery from Natural Products Using Affinity Selection-Mass Spectrometry. Drug Discov. Today Technol. 2021;40:59–63. doi: 10.1016/j.ddtec.2021.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zeng Q., Xia M.-C., Yin X., Cheng S., Xue Z., Tan S., Gong X., Ye Z. Recent Developments in Ionization Techniques for Single-Cell Mass Spectrometry. Front. Chem. 2023;11:1293533. doi: 10.3389/fchem.2023.1293533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Abdullahi A.D., Unban K., Saenjum C., Kodchasee P., Kangwan N., Thananchai H., Shetty K., Khanongnuch C. Antibacterial Activities of Miang Extracts against Selected Pathogens and the Potential of the Tannin-Free Extracts in the Growth Inhibition of Streptococcus Mutans. PLoS ONE. 2024;19:e0302717. doi: 10.1371/journal.pone.0302717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Misiurek J., Plech T., Kaproń B., Makuch-Kocka A., Szultka-Młyńska M., Buszewski B., Petruczynik A. Determination of Some Isoquinoline Alkaloids in Extracts Obtained from Selected Plants of the Ranunculaceae, Papaveraceae and Fumarioideae Families by Liquid Chromatography and In Vitro and In Vivo Investigations of Their Cytotoxic Activity. Molecules. 2023;28:3503. doi: 10.3390/molecules28083503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Baranyika J.B., Bakire S., Shoucheng P., Meihao S., Hirwa H. Application of the Selected Macroporous Resin for the Separation and Identification of Flavonoids from Chinese Radix Pueraria lobata by HPLC-Q-TOF-MS. Microchem. J. 2024;196:109662. doi: 10.1016/j.microc.2023.109662. [DOI] [Google Scholar]
- 80.Ligor M., Kováčová J., Gadzała-Kopciuch R.M., Studzińska S., Bocian S., Lehotay J., Buszewski B. Study of RP HPLC Retention Behaviours in Analysis of Carotenoids. Chromatographia. 2014;77:1047–1057. doi: 10.1007/s10337-014-2657-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Verma D.K., Chaudhary C., Singh L., Sidhu C., Siddhardha B., Prasad S.E., Thakur K.G. Isolation and Taxonomic Characterization of Novel Haloarchaeal Isolates from Indian Solar Saltern: A Brief Review on Distribution of Bacteriorhodopsins and V-Type ATPases in Haloarchaea. Front. Microbiol. 2020;11:554927. doi: 10.3389/fmicb.2020.554927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Watkins J.L., Li M., McQuinn R.P., Chan K.X., McFarlane H.E., Ermakova M., Furbank R.T., Mares D., Dong C., Chalmers K.J., et al. A GDSL Esterase/Lipase Catalyzes the Esterification of Lutein in Bread Wheat. Plant Cell. 2019;31:3092–3112. doi: 10.1105/tpc.19.00272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sinha S., Das S., Saha B., Paul D., Basu B. Anti-Microbial, Anti-Oxidant, and Anti-Breast Cancer Properties Unraveled in Yeast Carotenoids Produced via Cost-Effective Fermentation Technique Utilizing Waste Hydrolysate. Front. Microbiol. 2022;13:1088477. doi: 10.3389/fmicb.2022.1088477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.de Barros-Santos R.G., Pimentel T.C., Amorim T.A., da Silva Nogueira E.T., de Oliveira Vilar S.B., de Souza M.E.A.O., de Brito Araújo Carvalho A.J., Magnani M., dos Santos Lima M. Ultra-Fast Determination of Free Carotenoids in Fruit Juices by Rapid Resolution Liquid Chromatography (RRLC): Method Validation and Characterization of Brazilian Whole Fruit Juices. Food Anal. Methods. 2023;16:808–818. doi: 10.1007/s12161-023-02458-9. [DOI] [Google Scholar]
- 85.Osakabe M., Shimano S. The Flashy Red Color of the Red Velvet Mite Balaustium Murorum (Prostigmata: Erythraeidae) Is Caused by High Abundance of the Keto-Carotenoids, Astaxanthin and 3-Hydroxyechinenone. Exp. Appl. Acarol. 2023;89:1–14. doi: 10.1007/s10493-022-00766-z. [DOI] [PubMed] [Google Scholar]
- 86.Erdoğan A., Karataş A.B., Demir D., Demirel Z., Aktürk M., Çopur Ö., Conk-Dalay M. Manipulation in Culture Conditions of Nanofrustulum Shiloi for Enhanced Fucoxanthin Production and Isolation by Preparative Chromatography. Molecules. 2023;28:1988. doi: 10.3390/molecules28041988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Nemani N., Dehnavi S.M., Pazuki G. Extraction and Separation of Astaxanthin with the Help of Pre-Treatment of Haematococcus Pluvialis Microalgae Biomass Using Aqueous Two-Phase Systems Based on Deep Eutectic Solvents. Sci. Rep. 2024;14:5420. doi: 10.1038/s41598-024-55630-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Seeger J., Wendisch V.F., Henke N.A. Extraction and Purification of Highly Active Astaxanthin from Corynebacterium Glutamicum Fermentation Broth. Mar. Drugs. 2023;21:530. doi: 10.3390/md21100530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Silva P.G.P., Mussagy C.U., Lima C.d.A., Santos-Ebinuma V.C., Burkert J.F.d.M., Santos L.O. Sustainable Approach to Recover β-Carotene and Astaxanthin from Phaffia rhodozyma Grown in a Stirred-Tank Bioreactor under the Influence of Magnetic Fields. Bioresour. Technol. 2023;390:129906. doi: 10.1016/j.biortech.2023.129906. [DOI] [PubMed] [Google Scholar]
- 90.Pajot A., Chollet S., Nicolau E., Marchal L. Improving the Extraction and the Purification of Fucoxanthin from Tisochrysis lutea Using Centrifugal Partition Chromatography. Algal Res. 2023;74:103174. doi: 10.1016/j.algal.2023.103174. [DOI] [Google Scholar]
- 91.Lourenço-Lopes C., Silva A., Garcia-Oliveira P., Soria-Lopez A., Echave J., Grosso C., Cassani L., Barroso M.F., Simal-Gandara J., Fraga-Corral M., et al. Kinetic Extraction of Fucoxanthin from Undaria Pinnatifida Using Ethanol as a Solvent. Mar. Drugs. 2023;21:414. doi: 10.3390/md21070414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Tran Q.T.N., Nguyen H.V.H. Optimization of Enzyme-Assisted Lycopene Extraction from Tomato (Lycopersicon esculentum) Peel Using Rice Bran Oil. Food Meas. 2023;17:5154–5162. doi: 10.1007/s11694-023-02029-w. [DOI] [Google Scholar]
- 93.Ge B., Wang W., Gao Y., Chen X. Optimization of Extraction of Lycopene from Carrot and Determination of Its Antioxidant Activity. Food Meas. 2023;17:5497–5505. doi: 10.1007/s11694-023-02046-9. [DOI] [Google Scholar]
- 94.Surmanidze N., Vanidze M., Djafaridze I., Davitadze R., Qarcivadze I., Khakhutaishvili M., Kalandia A. Optimization of the Method of Ultrasonic Extraction of Lycopene with a Green Extract from the Fruit of Elaeagnus Umbellata, Common in Western Georgia. Food Sci. Nutr. 2024;12:3593–3601. doi: 10.1002/fsn3.4030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Dhakane-Lad J., Kar A., Patel A.S. SC-CO2 Extraction of Lycopene from Red Papaya Using Rice Bran Oil as a Co-Solvent Lessens Its Degradation during Storage. Sep. Sci. Technol. 2023;58:2357–2368. doi: 10.1080/01496395.2023.2255738. [DOI] [Google Scholar]
- 96.Ahmadi R., Honarvar M., Ghavami M., Daali Y. Optimization of Lutein Extraction from Pistachio Waste Using Experimental Design and Ultrasonic Method. Waste Biomass Valor. 2023;15:3593–3601. doi: 10.1007/s12649-023-02363-6. [DOI] [Google Scholar]
- 97.Maheshwari N., Arya R.K., Verros G.D., Dhamole P.B., Kannan A. Surfactant-Enhanced Extraction of Lutein from Marigold Petals Using an Aqueous Two-Phase System. Separations. 2023;10:133. doi: 10.3390/separations10020133. [DOI] [Google Scholar]
- 98.Sun X., Ma L., Muhire J., Zhang F.-X., Huang X.-Y., Liu J.-F., Pei D., Di D.-L. An Integrated Strategy for Combining Three-Phase Liquid-Liquid Extraction with Continuous High-Speed Countercurrent Chromatography: Highly Efficient in Isolating and Purifying Zeaxanthin from the Industrial Crop Lycium barbarum L. Ind. Crops Prod. 2023;206:117641. doi: 10.1016/j.indcrop.2023.117641. [DOI] [Google Scholar]
- 99.Yang L., Zi C., Li Y., Huang J., Gu Z., Wang C., Hu J.-M., Jiang Z., Zhang W. An In-Depth Investigation of Molecular Interaction in Zeaxanthin/Corn Silk Glycan Complexes and Its Positive Role in Hypoglycemic Activity. Food Chem. 2024;438:137986. doi: 10.1016/j.foodchem.2023.137986. [DOI] [PubMed] [Google Scholar]
- 100.Wang M., Morón-Ortiz Á., Zhou J., Benítez-González A., Mapelli-Brahm P., Meléndez-Martínez A.J., Barba F.J. Effects of Pressurized Liquid Extraction with Dimethyl Sulfoxide on the Recovery of Carotenoids and Other Dietary Valuable Compounds from the Microalgae Spirulina, Chlorella and Phaeodactylum tricornutum. Food Chem. 2023;405:134885. doi: 10.1016/j.foodchem.2022.134885. [DOI] [PubMed] [Google Scholar]
- 101.Macedo M.C.C., Correia V.T.d.V., Silva V.D.M., Pereira D.T.V., Augusti R., Melo J.O.F., Pires C.V., de Paula A.C.C.F.F., Fante C.A. Development and Characterization of Yellow Passion Fruit Peel Flour (Passiflora edulis f. Flavicarpa) Metabolites. 2023;13:684. doi: 10.3390/metabo13060684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bai X., Wan P., Liu J., Yao J., Chen D.-W. Investigation on the Changes of Carotenoids and Capsaicinoids in Chili Oil at Different Frying Temperature by Using 1H NMR. Curr. Res. Food Sci. 2023;6:100411. doi: 10.1016/j.crfs.2022.100411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ahn S., Ahn S., Jang H., Eom K., Kim Y.J., Hwang J.-E., Chung J.I., Park J.-Y., Nam S., Choi Y.-H., et al. Validation of Resonance Raman Spectroscopy-Measured Skin Carotenoid Status as a Biomarker for Fruit and Vegetable Intake in Korean Adults. Br. J. Nutr. 2023;130:1993–2001. doi: 10.1017/S0007114523001058. [DOI] [PubMed] [Google Scholar]
- 104.Casperson S.L., Roemmich J.N., Larson K.J., Hess J.M., Palmer D.G., Jahns L. Sensitivity of Pressure-Mediated Reflection Spectroscopy to Detect Changes in Skin Carotenoids in Adults without Obesity in Response to Increased Carotenoid Intake: A Randomized Controlled Trial. J. Nutr. 2023;153:588–597. doi: 10.1016/j.tjnut.2023.01.002. [DOI] [PubMed] [Google Scholar]
- 105.Solovchenko A., Lobakova E., Semenov A., Gorelova O., Fedorenko T., Chivkunova O., Parshina E., Maksimov G., Sluchanko N.N., Maksimov E. Multimodal Non-Invasive Probing of Stress-Induced Carotenogenesis in the Cells of Microalga Bracteacoccus Aggregatus. Protoplasma. 2024 doi: 10.1007/s00709-024-01956-9. [DOI] [PubMed] [Google Scholar]
- 106.Ba W., Xu W., Deng Z., Zhang B., Zheng L., Li H. The Antioxidant and Anti-Inflammatory Effects of the Main Carotenoids from Tomatoes via Nrf2 and NF-κB Signaling Pathways. Nutrients. 2023;15:4652. doi: 10.3390/nu15214652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Han X., Liu J., Gu Y., Li Y., Zhang W., Lv N., Dang A. Comparing Lycopene’s Impact on Mortality in Adults with or without Obesity. Food Funct. 2024;15:3340–3352. doi: 10.1039/D3FO04533A. [DOI] [PubMed] [Google Scholar]
- 108.Moreno L.G., César N.R., Melo D.S., Figueiró M.T.O., dos Santos E.C., Evangelista-Silva P.H., de Sousa Santos C., Costa K.B., Rocha-Vieira E., Dias-Peixoto M.F., et al. A MUFA/Carotenoid-Rich Oil Ameliorated Insulin Resistance by Improving Inflammation and Oxidative Stress in Obese Rats. Mol. Cell. Endocrinol. 2024;581:112110. doi: 10.1016/j.mce.2023.112110. [DOI] [PubMed] [Google Scholar]
- 109.Kim Y.I., Kim J.-S., Lee H., Jung C.H., Ahn J. Whole Red Paprika (Capsicum annuum L.) and Its Orange-Red Pigment Capsanthin Ameliorate Obesity-Induced Skeletal Muscle Atrophy in Mice. J. Funct. Foods. 2023;107:105624. doi: 10.1016/j.jff.2023.105624. [DOI] [Google Scholar]
- 110.Ciaraldi T.P., Boeder S.C., Mudaliar S.R., Giovannetti E.R., Henry R.R., Pettus J.H. Astaxanthin, a Natural Antioxidant, Lowers Cholesterol and Markers of Cardiovascular Risk in Individuals with Prediabetes and Dyslipidaemia. Diabetes Obes. Metab. 2023;25:1985–1994. doi: 10.1111/dom.15070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.López-Ramos A., González-Ortiz M., Martínez-Abundis E., Pérez-Rubio K.G. Effect of Fucoxanthin on Metabolic Syndrome, Insulin Sensitivity, and Insulin Secretion. J. Med. Food. 2023;26:521–527. doi: 10.1089/jmf.2022.0103. [DOI] [PubMed] [Google Scholar]
- 112.Peng C., Zeleznik O.A., Shutta K.H., Rosner B.A., Kraft P., Clish C.B., Stampfer M.J., Willett W.C., Tamimi R.M., Eliassen A.H. A Metabolomics Analysis of Circulating Carotenoids and Breast Cancer Risk. Cancer Epidemiol. Biomark. Prev. 2022;31:85–96. doi: 10.1158/1055-9965.EPI-21-0837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhong Q., Sun W., Qin Y., Xu H. Association of Dietary α-Carotene and β-Carotene Intake with Low Cognitive Performance in Older Adults: A Cross-Sectional Study from the National Health and Nutrition Examination Survey. Nutrients. 2023;15:239. doi: 10.3390/nu15010239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Gao X., Zhao X., Liu M., Zhao H., Sun Y. Lycopene Prevents Non-Alcoholic Fatty Liver Disease through Regulating Hepatic NF-κB/NLRP3 Inflammasome Pathway and Intestinal Microbiota in Mice Fed with High-Fat and High-Fructose Diet. Front. Nutr. 2023;10:1120254. doi: 10.3389/fnut.2023.1120254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Liu H., Yan J., Guan F., Jin Z., Xie J., Wang C., Liu M., Liu J. Zeaxanthin Prevents Ferroptosis by Promoting Mitochondrial Function and Inhibiting the P53 Pathway in Free Fatty Acid-Induced HepG2 Cells. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids. 2023;1868:159287. doi: 10.1016/j.bbalip.2023.159287. [DOI] [PubMed] [Google Scholar]
- 116.Li Z., Liu W., Sun C., Wei X., Liu S., Jiang Y. Gastrointestinal pH-Sensitive Pickering Emulsions Stabilized by Zein Nanoparticles Coated with Bioactive Glycyrrhizic Acid for Improving Oral Bioaccessibility of Curcumin. ACS Appl. Mater. Interfaces. 2023;15:14678–14689. doi: 10.1021/acsami.2c21549. [DOI] [PubMed] [Google Scholar]
- 117.Tian H., Zhang T., Qin S., Huang Z., Zhou L., Shi J., Nice E.C., Xie N., Huang C., Shen Z. Enhancing the Therapeutic Efficacy of Nanoparticles for Cancer Treatment Using Versatile Targeted Strategies. J. Hematol. Oncol. 2022;15:132. doi: 10.1186/s13045-022-01320-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Vera-Vázquez F., Ramírez-Bribiesca J.E., Cruz-Monterrosa R.G., Crosby-Galvan M.M., Barcena-Gama J.R., Ramírez D.T., Mejía-Méndez J.L., Vallejo-Hernández L.H., López-Mena E.R. Enhancing Pectin Particles with Polymer Additives: Mitigating Rumen Degradation and Minimizing Yellowish Milk Color in Grazed Cows. Polymers. 2024;16:106. doi: 10.3390/polym16010106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Murueva A.V., Shershneva A.M., Shishatskaya E.I., Volova T.G. Characteristics of Microparticles Based on Resorbable Polyhydroxyalkanoates Loaded with Antibacterial and Cytostatic Drugs. Int. J. Mol. Sci. 2023;24:14983. doi: 10.3390/ijms241914983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Joshi S., Klier J., Beltramo P.J. Encapsulation of Inorganic Nanoparticles by Anionic Emulsion Polymerization of Diethyl Methylene Malonate for Developing Hybrid Microparticles with Tailorable Composition. Colloids Interfaces. 2024;8:10. doi: 10.3390/colloids8010010. [DOI] [Google Scholar]
- 121.da Silva R.Y.P., Menezes D.L.B.d., Oliveira V.d.S., Converti A., Lima Á.A.N.d. Microparticles in the Development and Improvement of Pharmaceutical Formulations: An Analysis of In Vitro and In Vivo Studies. Int. J. Mol. Sci. 2023;24:5441. doi: 10.3390/ijms24065441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sathiyabama M., Boomija R.V., Muthukumar S., Gandhi M., Salma S., Prinsha T.K., Rengasamy B. Green Synthesis of Chitosan Nanoparticles Using Tea Extract and Its Antimicrobial Activity against Economically Important Phytopathogens of Rice. Sci. Rep. 2024;14:7381. doi: 10.1038/s41598-024-58066-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sánchez-López A.L., Perfecto-Avalos Y., Sanchez-Martinez A., Ceballos-Sanchez O., Sepulveda-Villegas M., Rincón-Enríquez G., Rodríguez-González V., Garcia-Varela R., Lozano L.M., Eloyr Navarro-López D., et al. Influence of Erbium Doping on Zinc Oxide Nanoparticles: Structural, Optical and Antimicrobial Activity. Appl. Surf. Sci. 2022;575:151764. doi: 10.1016/j.apsusc.2021.151764. [DOI] [Google Scholar]
- 124.Zhang X., Centurion F., Misra A., Patel S., Gu Z. Molecularly Targeted Nanomedicine Enabled by Inorganic Nanoparticles for Atherosclerosis Diagnosis and Treatment. Adv. Drug Deliv. Rev. 2023;194:114709. doi: 10.1016/j.addr.2023.114709. [DOI] [PubMed] [Google Scholar]
- 125.Chandrakala V., Aruna V., Angajala G. Review on Metal Nanoparticles as Nanocarriers: Current Challenges and Perspectives in Drug Delivery Systems. Emergent Mater. 2022;5:1593–1615. doi: 10.1007/s42247-021-00335-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Afzal O., Altamimi A.S.A., Nadeem M.S., Alzarea S.I., Almalki W.H., Tariq A., Mubeen B., Murtaza B.N., Iftikhar S., Riaz N., et al. Nanoparticles in Drug Delivery: From History to Therapeutic Applications. Nanomaterials. 2022;12:4494. doi: 10.3390/nano12244494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ghobashy M.M., Alkhursani S.A., Alqahtani H.A., El-damhougy T.K., Madani M. Gold Nanoparticles in Microelectronics Advancements and Biomedical Applications. Mater. Sci. Eng. B. 2024;301:117191. doi: 10.1016/j.mseb.2024.117191. [DOI] [Google Scholar]
- 128.Misra S.K., Mukherjee P., Chang H.-H., Tiwari S., Gryka M., Bhargava R., Pan D. Multi-Functionality Redefined with Colloidal Carotene Carbon Nanoparticles for Synchronized Chemical Imaging, Enriched Cellular Uptake and Therapy. Sci. Rep. 2016;6:29299. doi: 10.1038/srep29299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Elkholy N.S., Shafaa M.W., Mohammed H.S. Biophysical Characterization of Lutein or Beta Carotene-Loaded Cationic Liposomes. RSC Adv. 2020;10:32409–32422. doi: 10.1039/D0RA05683A. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Boon C.S., McClements D.J., Weiss J., Decker E.A. Factors Influencing the Chemical Stability of Carotenoids in Foods. Crit. Rev. Food Sci. Nutr. 2010;50:515–532. doi: 10.1080/10408390802565889. [DOI] [PubMed] [Google Scholar]
- 131.Wulandari S., Choi J., Kurniawan R.G., Sugiarto J.R., Myint A.A., Kwak S.K., Kim J. Synthesis of Highly Stable Encapsulated Astaxanthin/β-Cyclodextrin Microparticles Using Supercritical CO2 as an Antisolvent. J. CO2 Util. 2023;75:102575. doi: 10.1016/j.jcou.2023.102575. [DOI] [Google Scholar]
- 132.Vakarelova M., Zanoni F., Donà G., Fierri I., Chignola R., Gorrieri S., Zoccatelli G. Microencapsulation of Astaxanthin by Ionic Gelation: Effect of Different Gelling Polymers on the Carotenoid Load, Stability and Bioaccessibility. Int. J. Food Sci. Technol. 2023;58:2489–2497. doi: 10.1111/ijfs.16389. [DOI] [Google Scholar]
- 133.Meira A.C.F.D.O., Morais L.C.D., Figueiredo J.D.A., Veríssimo L.A.A., Botrel D.A., Resende J.V.D. Microencapsulation of β-Carotene Using Barley Residue Proteins from Beer Waste as Coating Material. J. Microencapsul. 2023;40:171–185. doi: 10.1080/02652048.2023.2183277. [DOI] [PubMed] [Google Scholar]
- 134.Sánchez C.A.O., Zavaleta E.B., García G.R.U., Solano G.L., Díaz M.P.R. Krill Oil Microencapsulation: Antioxidant Activity, Astaxanthin Retention, Encapsulation Efficiency, Fatty Acids Profile, in Vitro Bioaccessibility and Storage Stability. LWT. 2021;147:111476. doi: 10.1016/j.lwt.2021.111476. [DOI] [Google Scholar]
- 135.Morales E., Burgos-Díaz C., Zúñiga R.N., Jorkowski J., Quilaqueo M., Rubilar M. Influence of O/W Emulsion Interfacial Ionic Membranes on the Encapsulation Efficiency and Storage Stability of Powder Microencapsulated Astaxanthin. Food Bioprod. Process. 2021;126:143–154. doi: 10.1016/j.fbp.2020.12.014. [DOI] [Google Scholar]
- 136.Foo S.C., Khong N.M.H., Yusoff F.M. Physicochemical, Microstructure and Antioxidant Properties of Microalgae-Derived Fucoxanthin Rich Microcapsules. Algal Res. 2020;51:102061. doi: 10.1016/j.algal.2020.102061. [DOI] [Google Scholar]
- 137.Oliyaei N., Moosavi-Nasab M., Tamaddon A.M., Fazaeli M. Double Encapsulation of Fucoxanthin Using Porous Starch through Sequential Coating Modification with Maltodextrin and Gum Arabic. Food Sci. Nutr. 2020;8:1226–1236. doi: 10.1002/fsn3.1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Li Y., Dou X., Pang J., Liang M., Feng C., Kong M., Liu Y., Cheng X., Wang Y., Chen X. Improvement of Fucoxanthin Oral Efficacy via Vehicles Based on Gum Arabic, Gelatin and Alginate Hydrogel: Delivery System for Oral Efficacy Enhancement of Functional Food Ingredients. J. Funct. Foods. 2019;63:103573. doi: 10.1016/j.jff.2019.103573. [DOI] [Google Scholar]
- 139.Corrêa-Filho L.C., Lourenço S.C., Duarte D.F., Moldão-Martins M., Alves V.D. Microencapsulation of Tomato (Solanum lycopersicum L.) Pomace Ethanolic Extract by Spray Drying: Optimization of Process Conditions. Appl. Sci. 2019;9:612. doi: 10.3390/app9030612. [DOI] [Google Scholar]
- 140.Corrêa-Filho L.C., Santos D.I., Brito L., Moldão-Martins M., Alves V.D. Storage Stability and In Vitro Bioaccessibility of Microencapsulated Tomato (Solanum lycopersicum L.) Pomace Extract. Bioengineering. 2022;9:311. doi: 10.3390/bioengineering9070311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Tkacz K., Turkiewicz I.P., Nowicka P., Wojdyło A. Microspheres as Carriers of Sea Buckthorn Carotenoids and Tocols with Antidiabetic Potential: Effect of Biopolymers, Cross-Linking and Storage. Food Biosci. 2024;59:103995. doi: 10.1016/j.fbio.2024.103995. [DOI] [Google Scholar]
- 142.Gheonea (Dima) I., Aprodu I., Cîrciumaru A., Râpeanu G., Bahrim G.E., Stănciuc N. Microencapsulation of Lycopene from Tomatoes Peels by Complex Coacervation and Freeze-Drying: Evidences on Phytochemical Profile, Stability and Food Applications. J. Food Eng. 2021;288:110166. doi: 10.1016/j.jfoodeng.2020.110166. [DOI] [Google Scholar]
- 143.Ding Z., Tao T., Yin X., Prakash S., Wang X., Zhao Y., Han J., Wang Z. Improved Encapsulation Efficiency and Storage Stability of Spray Dried Microencapsulated Lutein with Carbohydrates Combinations as Encapsulating Material. LWT. 2020;124:109139. doi: 10.1016/j.lwt.2020.109139. [DOI] [Google Scholar]
- 144.Zhao W., Zhang B., Liang W., Liu X., Zheng J., Ge X., Shen H., Lu Y., Zhang X., Sun Z., et al. Lutein Encapsulated in Whey Protein and Citric Acid Potato Starch Ester: Construction and Characterization of Microcapsules. Int. J. Biol. Macromol. 2022;220:1–12. doi: 10.1016/j.ijbiomac.2022.08.068. [DOI] [PubMed] [Google Scholar]
- 145.Hao J., Xu J., Zhang W., Li X., Liang D., Xu D., Cao Y., Sun B. The Improvement of the Physicochemical Properties and Bioaccessibility of Lutein Microparticles by Electrostatic Complexation. Food Hydrocoll. 2022;125:107381. doi: 10.1016/j.foodhyd.2021.107381. [DOI] [Google Scholar]
- 146.Ganjali E., Elhamirad A.H., Hosseini F., Mahfoozi M. The Improvement of Crocin Stability in Rock Candy (Nabat) By Microencapsulation. J. Nutr. Fasting Health. 2020;8:280–287. doi: 10.22038/jnfh.2020.50824.1286. [DOI] [Google Scholar]
- 147.Essifi K., Brahmi M., Berraaouan D., Ed-Daoui A., El Bachiri A., Fauconnier M.-L., Tahani A. Influence of Sodium Alginate Concentration on Microcapsules Properties Foreseeing the Protection and Controlled Release of Bioactive Substances. J. Chem. 2021;2021:e5531479. doi: 10.1155/2021/5531479. [DOI] [Google Scholar]
- 148.Merati F., Mehryab F., Mortazavi S.A., Haeri A. An Experimental Design Approach for Development of Crocin-Loaded Microparticles Embedded in Gelatin/Oxidized Alginate-Based Hydrogel. J. Pharm. Innov. 2023;18:1812–1826. doi: 10.1007/s12247-023-09755-0. [DOI] [Google Scholar]
- 149.Dłużewska E., Florowska A., Domian E., Wojciechowska M., Maszewska M. The Influence of the Agglomeration Process on Stability of Microencapsulated β-Carotene. Int. J. Food Eng. 2020;16:20180310. doi: 10.1515/ijfe-2018-0310. [DOI] [Google Scholar]
- 150.Constantino A.B.T., Garcia-Rojas E.E. Microencapsulation of Beta-Carotene by Complex Coacervation Using Amaranth Carboxymethyl Starch and Lactoferrin for Application in Gummy Candies. Food Hydrocoll. 2023;139:108488. doi: 10.1016/j.foodhyd.2023.108488. [DOI] [Google Scholar]
- 151.Perrechil F.A., Maximo G.J., Sato A.C.K., Cunha R.L. Microbeads of Sodium Caseinate and κ-Carrageenan as a β-Carotene Carrier in Aqueous Systems. Food Bioprocess. Technol. 2020;13:661–669. doi: 10.1007/s11947-020-02426-9. [DOI] [Google Scholar]
- 152.Xu Z., Zhang X., Wu X., Ma D., Huang Y., Zhao Q., Zhang S., Li Y. Co-Delivery of Vitamin C and β-Carotene in W/O/W Emulsions Stabilized by Modified Aggregated Insoluble Soybean Protein Hydrolysate-Xanthan Gum Complexes. Int. J. Biol. Macromol. 2024;261:129855. doi: 10.1016/j.ijbiomac.2024.129855. [DOI] [PubMed] [Google Scholar]
- 153.Wang Y., Zhou Q., Zheng J., Xiong H., Zhao L., Xu Y., Bai C. Fabricating Pectin and Chitosan Double Layer Coated Liposomes to Improve Physicochemical Stability of Beta-Carotene and Alter Its Gastrointestinal Fate. Int. J. Biol. Macromol. 2023;247:125780. doi: 10.1016/j.ijbiomac.2023.125780. [DOI] [PubMed] [Google Scholar]
- 154.dos Santos Pais T., Luchiari A.C., de Souza A.M., Medeiros I., Silva M.G.F.R., dos Santos Y.L., Silva-Maia J.K., Passos T.S., Morais A.H.d.A. Assessment of Acute Toxicity of Crude Extract Rich in Carotenoids from Cantaloupe Melon (Cucumis melo L.) and the Gelatin-Based Nanoparticles Using the Zebrafish (Danio rerio) Model. Food Chem. Toxicol. 2023;181:114091. doi: 10.1016/j.fct.2023.114091. [DOI] [PubMed] [Google Scholar]
- 155.Kim D., Jung Y., Rho S.-J., Kim Y.-R. Improved Stability and in Vitro Bioavailability of β-Carotene in Filled Hydrogel Prepared from Starch Blends with Different Granule Sizes. Food Hydrocoll. 2023;139:108546. doi: 10.1016/j.foodhyd.2023.108546. [DOI] [Google Scholar]
- 156.Guo M., Cui W., Li Y., Fei S., Sun C., Tan M., Su W. Microfluidic Fabrication of Size-Controlled Nanocarriers with Improved Stability and Biocompatibility for Astaxanthin Delivery. Food Res. Int. 2023;170:112958. doi: 10.1016/j.foodres.2023.112958. [DOI] [PubMed] [Google Scholar]
- 157.Kim E.S., Baek Y., Yoo H.-J., Lee J.-S., Lee H.G. Chitosan-Tripolyphosphate Nanoparticles Prepared by Ionic Gelation Improve the Antioxidant Activities of Astaxanthin in the In Vitro and In Vivo Model. Antioxidants. 2022;11:479. doi: 10.3390/antiox11030479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ge Y., Zhou Y., Li S., Yan J., Chen H., Qin W., Zhang Q. Astaxanthin Encapsulation in Soybean Protein Isolate–Sodium Alginate Complexes-Stabilized Nanoemulsions: Antioxidant Activities, Environmental Stability, and in Vitro Digestibility. J. Sci. Food Agric. 2024;104:1539–1552. doi: 10.1002/jsfa.13036. [DOI] [PubMed] [Google Scholar]
- 159.Koo S.Y., Hwang K.T., Hwang S., Choi K.Y., Park Y.J., Choi J.-H., Truong T.Q., Kim S.M. Nanoencapsulation Enhances the Bioavailability of Fucoxanthin in Microalga Phaeodactylum tricornutum Extract. Food Chem. 2023;403:134348. doi: 10.1016/j.foodchem.2022.134348. [DOI] [PubMed] [Google Scholar]
- 160.İnanç Horuz T., Belibağlı K.B. Nanoencapsulation by Electrospinning to Improve Stability and Water Solubility of Carotenoids Extracted from Tomato Peels. Food Chem. 2018;268:86–93. doi: 10.1016/j.foodchem.2018.06.017. [DOI] [PubMed] [Google Scholar]
- 161.Shwetha H.J., Arathi B.P., Beto Mukherjee M., Ambedkar R., Shivaprasad S., Raichur A.M., Lakshminarayana R. Zein-Alginate-Phosphatidylcholine Nanocomplex Efficiently Delivers Lycopene and Lutein over Dietary-Derived Carotenoid Mixed Micelles in Caco-2 Cells. J. Agric. Food Chem. 2022;70:15474–15486. doi: 10.1021/acs.jafc.2c05008. [DOI] [PubMed] [Google Scholar]
- 162.Vasconcelos A.G., Valim M.O., Amorim A.G.N., do Amaral C.P., de Almeida M.P., Borges T.K.S., Socodato R., Portugal C.C., Brand G.D., Mattos J.S.C., et al. Cytotoxic Activity of Poly-ɛ-Caprolactone Lipid-Core Nanocapsules Loaded with Lycopene-Rich Extract from Red Guava (Psidium guajava L.) on Breast Cancer Cells. Food Res. Int. 2020;136:109548. doi: 10.1016/j.foodres.2020.109548. [DOI] [PubMed] [Google Scholar]
- 163.Dai Z., Song J., Chen Y., Feng L., Xu Y., Li D., Wu C., Zhang Z., Liu J. Study on the Bioavailability of Stevioside-Encapsulized Lutein and Its Mechanism. Food Chem. 2021;354:129528. doi: 10.1016/j.foodchem.2021.129528. [DOI] [PubMed] [Google Scholar]
- 164.Toragall V., Srirangam P., Jayapala N., Baskaran V. Lutein Encapsulated Oleic—Linoleic Acid Nanoemulsion Boosts Oral Bioavailability of the Eye Protective Carotenoid Lutein in Rat Model. Mater. Today Commun. 2021;28:102522. doi: 10.1016/j.mtcomm.2021.102522. [DOI] [Google Scholar]
- 165.Saroglu O., Atalı B., Yıldırım R.M., Karadag A. Characterization of Nanoliposomes Loaded with Saffron Extract: In Vitro Digestion and Release of Crocin. Food Meas. 2022;16:4402–4415. doi: 10.1007/s11694-022-01526-8. [DOI] [Google Scholar]
- 166.Nasrpour S., Yousefi G., Niakosari M., Aminlari M. Nanoencapsulation of Saffron Crocin into Chitosan/Alginate Interpolyelectrolyte Complexes for Oral Delivery: A Taguchi Approach to Design Optimization. J. Food Sci. 2022;87:1148–1160. doi: 10.1111/1750-3841.16052. [DOI] [PubMed] [Google Scholar]
- 167.Yu D., Guo M., Tan M., Su W. Lipid-Lowering and Antioxidant Effects of Self-Assembled Astaxanthin–Anthocyanin Nanoparticles on High-Fat Caenorhabditis Elegans. Foods. 2024;13:514. doi: 10.3390/foods13040514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Gao Y., Yuan S., Chen Y., Liu F., Wei Z., Cao W., Li R.W., Xu J., Xue C., Tang Q. The Improvement Effect of Astaxanthin-Loaded Emulsions on Obesity Is Better than That of Astaxanthin in the Oil Phase. Food Funct. 2022;13:3720–3731. doi: 10.1039/D1FO03185F. [DOI] [PubMed] [Google Scholar]
- 169.Che T., Song Y., Su W., Xing S., Wang H., Tan M. Hepatic Parenchymal Cell and Mitochondrial-Targeted Astaxanthin Nanocarriers for Relief of High Fat Diet-Induced Nonalcoholic Fatty Liver Disease. Food Funct. 2023;14:2908–2920. doi: 10.1039/D2FO04036K. [DOI] [PubMed] [Google Scholar]
- 170.Ghosh A., Banik S., Yamada K., Misaka S., Prud’homme R.K., Sato H., Onoue S. Stabilized Astaxanthin Nanoparticles Developed Using Flash Nanoprecipitation to Improve Oral Bioavailability and Hepatoprotective Effects. Pharmaceutics. 2023;15:2562. doi: 10.3390/pharmaceutics15112562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Torres J., Pereira J.M., Marques-Oliveira R., Costa I., Gil-Martins E., Silva R., Remião F., Peixoto A.F., Sousa Lobo J.M., Silva A.C. An In Vitro Evaluation of the Potential Neuroprotective Effects of Intranasal Lipid Nanoparticles Containing Astaxanthin Obtained from Different Sources: Comparative Studies. Pharmaceutics. 2023;15:1035. doi: 10.3390/pharmaceutics15041035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.de Carvalho Gomes C., Lima M.S.R., de Oliveira G.L.R., Medeiros I., Xavier H.S.T., dos Santos Pais T., de Sousa Costa I., de Carvalho F.M.C., Serquiz A.C., de Souza Lima M.C.J., et al. Nanoparticles Loaded with a Carotenoid-Rich Extract from Cantaloupe Melon Improved Hepatic Retinol Levels in a Diet-Induced Obesity Preclinical Model. ACS Omega. 2023;8:28475–28486. doi: 10.1021/acsomega.3c02750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Mahadev M., Dubey A., Shetty A. Ultrasonically Fabricated Beta-Carotene Nanoemulsion: Optimization, Characterization and Evaluation of Combinatorial Effect with Quercetin on Streptozotocin-Induced Diabetic Rat Model. Pharmaceutics. 2023;15:574. doi: 10.3390/pharmaceutics15020574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Teixé-Roig J., Oms-Oliu G., Odriozola-Serrano I., Martín-Belloso O. Enhancing in Vivo Retinol Bioavailability by Incorporating β-Carotene from Alga Dunaliella salina into Nanoemulsions Containing Natural-Based Emulsifiers. Food Res. Int. 2023;164:112359. doi: 10.1016/j.foodres.2022.112359. [DOI] [PubMed] [Google Scholar]
- 175.Zhang X., Dong J., Yin H., Chen L., He Y., Qin Q., Zhou Y., Fan M., Wang D. Exploiting Fucoxanthin Mono-Carrier Nanoparticles to Modulate Digestion and Metabolic Regulation in an Obesity Model. Food Biosci. 2024;57:103466. doi: 10.1016/j.fbio.2023.103466. [DOI] [Google Scholar]
- 176.Zhang X., Fan M., Luo K., Xu W., Dong J., Wang D., Chen L., Yu J. In Vivo Assessment of the Effects of Mono-Carrier Encapsulated Fucoxanthin Nanoparticles on Type 2 Diabetic C57 Mice and Their Oxidative Stress. Antioxidants. 2022;11:1976. doi: 10.3390/antiox11101976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Wang C., Bai Y., Yin W., Qiu B., Jiang P., Dong X., Qi H. Nanoencapsulation Motivates the High Inhibitive Ability of Fucoxanthin on H2O2-Induced Human Hepatocyte Cell Line (L02) Apoptosis via Regulating Lipid Metabolism Homeostasis. J. Agric. Food Chem. 2023;71:6087–6098. doi: 10.1021/acs.jafc.3c01160. [DOI] [PubMed] [Google Scholar]
- 178.Raji V., Loganathan C., Ramesh T., Thayumanavan P. Dual Antidiabetic and Antihypertensive Activity of Fucoxanthin Isolated from Sargassum wightii Greville in in Vivo Rat Model. Food Sci. Hum. Wellness. 2023;12:1693–1700. doi: 10.1016/j.fshw.2023.02.037. [DOI] [Google Scholar]
- 179.Fang Y., Nie T., Li G., Wang L., Du J., Wu J. Multifunctional Antibiotic Hydrogel Doped with Antioxidative Lycopene-Based Liposome for Accelerative Diabetic Wound Healing. Chem. Eng. J. 2024;480:147930. doi: 10.1016/j.cej.2023.147930. [DOI] [Google Scholar]
- 180.Ge J., Ye L., Cheng M., Xu W., Chen Z., Guan F. Preparation of Microgels Loaded with Lycopene/NMN and Their Protective Mechanism against Acute Liver Injury. Food Funct. 2024;15:809–822. doi: 10.1039/D3FO03293K. [DOI] [PubMed] [Google Scholar]
- 181.Xia X., Li H., Xu X., Wang Z., Zhao G., Du M. Improving Mitochondrial Function for Alleviating Memory Decline of Aging Mice via Dual-Delivering Lycopene Nanoparticles. Appl. Mater. Today. 2024;37:102132. doi: 10.1016/j.apmt.2024.102132. [DOI] [Google Scholar]
- 182.Xia X., Li H., Xu X., Zhao G., Du M. Facilitating Pro-Survival Mitophagy for Alleviating Parkinson’s Disease via Sequence-Targeted Lycopene Nanodots. ACS Nano. 2023;17:17979–17995. doi: 10.1021/acsnano.3c04308. [DOI] [PubMed] [Google Scholar]
- 183.Zhang G., Zhang M., Pei Y., Qian K., Xie J., Huang Q., Liu S., Xue N., Zu Y., Wang H. Enhancing Stability of Liposomes Using High Molecular Weight Chitosan to Promote Antioxidative Stress Effects and Lipid-Lowering Activity of Encapsulated Lutein in Vivo and in Vitro. Int. J. Biol. Macromol. 2023;253:126564. doi: 10.1016/j.ijbiomac.2023.126564. [DOI] [PubMed] [Google Scholar]
- 184.Guo T., Chen L., Li F., Cao Y., Li D., Xiong Q., Ling Z. Biomimetic Nanoparticles Loaded Lutein Functionalized by Macrophage Membrane for Targeted Amelioration Pressure Overload-Induced Cardiac Fibrosis. Biomed. Pharmacother. 2023;167:115579. doi: 10.1016/j.biopha.2023.115579. [DOI] [PubMed] [Google Scholar]






