Previous Article in Journal
Alpha-Mangostin Enhances Radiosensitivity in HeLa Cervical Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements in Encapsulation Technologies: The Potential of Polyphenols as an Antidiabetic Therapy

by
Rigoberto Cabanillas-Ponce de León
1,
Feliznando Isidro Cardenas-Torres
2,
Noe Ontiveros
3,
Laura Aracely Contreras-Angulo
1,
Cristina Alicia Elisande-Romero
4,
Nayely Leyva-López
5,
Manuel de Jesús Bernal-Millán
1,
Jose Basilio Heredia
1 and
Erick Paul Gutiérrez-Grijalva
6,*
1
Laboratory of Functional Foods and Nutraceuticals, Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a El Dorado Km 5.5, Col. Campo El Diez, Culiacán 80110, Mexico
2
Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán 80019, Mexico
3
Laboratorio de Análisis Clínicos e Investigación (LACIUS, U.N), Departamento de Ciencias Químico-Biológicas y Agropecuarias, Facultad de Ciencia Biológicas y de Salud, Universidad de Sonora, Navojoa 85880, Mexico
4
Unidad Regional Culiacán, Universidad Autónoma de Occidente, Blvd. Lola Beltrán y Blvd. Rolando Arjona, 4 de Marzo, Culiacán 80020, Mexico
5
Posdoc SECIHTI-Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a El Dorado Km 5.5, Col. Campo El Diez, Culiacán 80110, Mexico
6
Programa de Investigadoras e Investigadores por México SECIHTI-Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a El Dorado Km 5.5, Col. Campo El Diez, Culiacán 80110, Mexico
*
Author to whom correspondence should be addressed.
Sci. Pharm. 2025, 93(4), 55; https://doi.org/10.3390/scipharm93040055
Submission received: 16 September 2025 / Revised: 19 October 2025 / Accepted: 24 October 2025 / Published: 28 October 2025

Abstract

Diabetes mellitus (DM) is a disease that affects over 537 million people worldwide and results in 6.7 million deaths annually. Conventional treatment of this disease focuses on lifestyle changes and drug administration. However, very few people can adhere to a healthier lifestyle, and drugs are difficult to access, especially in low- and middle-income countries. An alternative as an adjuvant to the treatment of DM is the phenolic compounds from plants with reported anti-diabetic effects. However, the bioavailability of these compounds is very low since they are affected by the gastrointestinal tract and xenobiotic metabolism. To improve the availability of these compounds, an emerging technology such as encapsulation is being used since it has been reported that the encapsulation of phenolic compounds improves both their bioaccessibility and bioavailability, as well as their bioactivity. In this review, we will focus on compiling the most up-to-date information on the different encapsulation processes of phenolic compounds and the antidiabetic effect of encapsulated phenolic compounds using the databases PubMed, Scopus, Web of Science, and Google Scholar. We will discuss the mechanisms, pathways, and receptors involved in the modulation of DM, especially those related to inflammation, oxidative stress, and insulin resistance.

1. Introduction

Diabetes mellitus (DM) is considered a group of metabolic disorders; this includes not only the inappropriate utilization of glucose as the main energy source but also impaired glucogenesis, gluconeogenesis, and insulin secretion and signalization. According to the American Diabetes Association (ADA), there are four types of diabetes mellitus categorized in Figure 1 [1,2]. When DM is not well-treated, several complications can develop, such as fatty liver, renal disease, metabolic syndrome, infertility, and neurological diseases, among others [3,4].
Worldwide, it is estimated that more than 422 million people live with DM, and incidence has been increasing steadily, especially in low and middle-income regions. This disease is considered a global health emergency, with projections expecting that by 2030, cases will reach 643 million and 783 million by 2045. In countries such as the United States, the incidence of DM in the young population continues to rise, with estimates of 18,200 cases of type 1 DM and 5300 cases of type 2 DM. Nonetheless, worldwide, it is considered that 98% of diagnoses are for type 2 DM, and the rest are other types [1,5,6].
Considering type 2 DM (T2DM) is the most common diagnosis, most investigations have been aimed at it to understand psychopathology, complications, and treatments. But firstly, a correct diagnosis is necessary, considering for non-pregnant individuals the following tests: plasma A1C, fasting plasma glucose (FPG) value, 2 h glucose (2 h PG) value during a 75 g oral glucose tolerance test (OGTT), or random glucose value. It is also crucial to consider symptoms of hyperglycemia, such as polyuria, polydipsia, unexplained weight loss, and increased appetite [1]. Contrary to Type 1 DM, T2DM can be prevented and well as managed by modifying risk habits and conditions. T2DM is closely linked to obesity and, therefore, to inflammation and oxidative stress. These three factors are commonly found in all non-communicable diseases [7,8].
Treatment and prevention of DM include exercise, a correct diet, drugs, and insulin in specific cases. Managing obesity and being overweight in diabetic patients also plays an important role. Access to treatment is key for individuals with DM to increase life quality and expectancy [9,10,11]. Nowadays, including other therapies and adjuvants to control or reduce oxidative stress and inflammation related to obesity and diabetes is a promising strategy. In this regard, several natural compounds, supplements, extracts, and other nutraceuticals have been studied to determine effective and safe doses for individuals with diabetes and prediabetes [12,13,14,15].
Several compounds, especially polyphenols, have been studied in vivo and in vitro as agents for prevention, management, and co-adjuvants in DM, regarding their proven and potential health benefits. Some activities attributed to polyphenols in this manner are antioxidants, anti-inflammatory, hypoglycemic, antiadipogenic, anti-gluconeogenesis, modulation of glucagon, insulin, and others [3,16,17,18,19].
Polyphenolic compounds are a heterogeneous class of phytochemicals characterized by multiple phenolic structural units, which are widely distributed in various plant-derived foods. Polyphenols are classified into two primary categories: flavonoids and non-flavonoids. Flavonoids include subcategories, such as anthocyanins, flavanols, flavanones, flavonols, and isoflavones, each characterized by distinct structural variations. In contrast, non-flavonoids comprise phenolic acids, xanthones, stilbenes, lignans, and tannins [20].
Polyphenols are abundantly present in fruits, vegetables, cereals, and other natural plant matrices, being integral to the sensory profiles and serving as biochemical markers for several foods and processing methods. For instance, in wines, polyphenols contribute to the complexity of flavor, color, and astringency while also providing insights into the authenticity and geographical origin of the product [21]. Furthermore, these compounds are pivotal to human health, with numerous studies elucidating their potential therapeutic benefits and prophylactic effects by several mechanisms against various pathologies [22]. Their bioactive properties have been associated with the modulation of oxidative stress [23], mitigating oxidative stress at the cellular level via scavenging and neutralizing reactive oxygen species [24,25,26]. For instance, it has been reported that dill (Anethum graveolens L.) extract exhibits antidiarrheal, anti-inflammatory, and antioxidant properties associated with its polyphenols [23]. Pumpkin pulp has been reported as a natural reservoir of polyphenolic compounds, showing antioxidant and antimicrobial properties, making it an attractive candidate for developing products to promote health [27]. In another study, it was reported that there are anti-inflammatory and antioxidant properties in several types of beans [28]. Similarly, a combination of vitamin C, resveratrol, and astaxanthin showed anti-inflammatory and antioxidant properties [29]. In other studies, resveratrol, in combination with urapidil, has neuroprotective effects by downregulating neurodegeneration [30].
On the other hand, a recent study suggests that tannic acid is a versatile candidate for preventing doxorubicin (DOX)-induced hepatotoxicity, potentially through the preservation of cellular physiology, viability, and, notably, redox homeostasis [31]. Another beneficial effect of polyphenols is the modulation of intestinal microbiota. A study showed how epigallocatechin-3-gallate has been shown to elevate the production of short-chain fatty acids, enhance amino acid metabolism, and downregulate pathways associated with intestinal inflammation. Additionally, this compound modulates the gut microbiota and mitigates Clostridioides difficile infection, offering novel insights into potential therapeutic interventions [32]. The study by Zhao et al. [33] suggests that polyphenols can modulate the gut microbiota, significantly impacting the production of microbial metabolites like isovaleric acid and isobutyric acid.
In the case of metabolic diseases, polyphenols, such as catechins, proanthocyanidins, hydroxybenzoic acids, and lignans, have been associated with a minor risk of developing type 2 diabetes [34]. A study conducted by Liao et al. [35] showed that Chinese jujube polyphenols exhibit significant hypoglycemic and antioxidative effects in rats with T2DM, thereby ameliorating glucose metabolism disorders and oxidative damage. Aqueous extracts of cinnamon and clove demonstrated greater potency than acarbose in inhibiting alpha-glucosidase activity and exhibited the highest antioxidant activity. The polyphenol content strongly correlated with antioxidant capacity, suggesting that these spices hold potential for the prevention and treatment of DM [36]. A study on the low-temperature aqueous extract of sea mustard (Undaria pinnatifida) at 50 °C indicates potential antihyperglycemic effects. This effect is mediated by the modulation of glucose uptake via specific glucose transporters, suggesting the extract’s capacity to mitigate postprandial hyperglycemia [37]. Also, Liu et al. [38] indicate that the continuous administration of theaflavins (100 mg/kg) significantly suppressed blood glucose levels, reduced insulin resistance, and decreased the expression of oxidative stress markers and inflammatory cytokines in Goto-Kakizaki rats. Furthermore, consuming theaflavins facilitated the restoration of intestinal microbial community structure by reducing the abundance of pathogenic bacteria and increasing the prevalence of beneficial microorganisms.
In a study designed to evaluate the effects of a six-year nutritional and lifestyle intervention on oxidative and inflammatory markers in individuals aged 55 and older who are at high risk of cardiovascular diseases [39], the results showed that increased polyphenol intake was associated with a greater reduction in body mass index among participants. This suggests that polyphenols may play a role in promoting weight loss or maintaining healthier body weight, which is crucial for reducing the risk of obesity-related diseases such as T2DM, and at the same time, the study suggests that adherence to a low-calorie Mediterranean diet with increased polyphenol intake could contribute to positive health outcomes in a synergistic way.
These studies prove that polyphenols confer numerous health benefits and contribute to disease prevention and management. However, one of their primary limitations is the low bioavailability of these compounds [40]. In this sense, bioavailability is enhanced by (1) the physiological dose, (2) reduced particle size and thermal treatment, which facilitate compound release from the matrix, and (3) the presence of lipids, with minimal proteins and indigestible carbohydrates in the matrix [40]. For these reasons, it is particularly pertinent to explore strategies aimed at enhancing the bioavailability of these compounds to maximize the potential health benefits associated with their consumption.
Micro- and nano-encapsulated PC delivery systems have been developed for this purpose. The wall material and micro- or nanoparticle preparation method should be selected considering the PCs’ biological properties, physicochemical characteristics, and purity to be encapsulated [41,42]. The present review is focused on the technologies used to encapsulate PCs that could serve as nutraceuticals for the prevention of T2DM or adjuncts in the therapeutic management of this chronic disease. The mechanism involved in the PCs-induced antidiabetic, anti-inflammatory, and antioxidant effects is also discussed.

2. Polyphenols

Most of the bioactive properties of polyphenols depend on their bioavailability, as non-bioavailable polyphenols can still exert preventive properties in the onset of colorectal cancer and modulate the intestinal microbiota. In vivo, many polyphenols that exhibit promising in vitro bioactivity suffer from low intestinal absorption and rapid elimination, resulting in limited systemic exposure.
A critical factor contributing to the variability in polyphenol bioavailability is the interindividual differences in absorption, distribution, metabolism, and excretion (ADME). These differences may stem from genetic polymorphisms affecting intestinal enzymes or xenobiotic transporters [43,44]. Additionally, variations in dietary habits, physiological conditions, and the permeability of biological barriers between healthy individuals and those with compromised health may further influence polyphenol bioavailability [45,46].
Enhancing polyphenol bioavailability, particularly in the context of dietary intake, is critically influenced by the interaction between nutritional lipids and polyphenols. Dietary fats, notably, have been recognized for their ability to enhance the solubility of polyphenols, potentially increasing their bioavailability [47]. Hydrophobic polyphenols, such as curcumin, demonstrate improved bioavailability when co-administered with dietary lipids [40]. This evidence suggests that integrating dietary fats with polyphenol consumption could optimize their absorption and subsequent physiological effects.
Furthermore, macronutrients such as carbohydrates and fats have been identified as intestinal absorption enhancers, which can modulate the time required to reach peak plasma concentrations of polyphenols [47]. The gut microbiota also plays a pivotal role in the metabolism and bioavailability of polyphenols. It has been demonstrated that gut bacteria can generate bioactive metabolites from polyphenols, thereby modulating various physiological processes and enhancing overall bioavailability [48,49,50,51,52].
The interaction between polyphenols and gut microbiota is bidirectional. Gut microbiota can convert polyphenols into simpler, more absorbable forms [53,54]. Conversely, polyphenols can influence gut microbiota composition, promote the growth of beneficial bacteria while suppressing pathogenic strains, thereby supporting a balanced gut microbiome [55]. Moreover, the complexation of polyphenols with proteins has improved their bioaccessibility and bioavailability. This mechanism protects polyphenols during gastrointestinal transit, enabling them to reach the colon, where they undergo further metabolism by gut microbiota, thus amplifying their health benefits [56].

3. Microencapsulation

Microencapsulation is one of the most promising technologies for directed therapeutic treatments used in recent years [57]. It is a process in which bioactive compounds like phenolics are trapped in an encapsulating material to create particles with a semipermeable membrane [58]. These bioactive agents can be encapsulated in their solid, liquid, and gaseous forms to obtain microcapsules with a size between 1 and 1000 μm [59]. Furthermore, the morphologies depend on the microencapsulation method, obtaining microspheres, microcapsules, and microparticles [60].
Microencapsulation of phenolic compounds protects them from environmental factors (light, humidity, temperature, and oxygen), provides controlled release over time, improves bioaccessibility, and increases shelf life and ease of storage [61]. There are several conventional and emerging microencapsulation techniques. Generally, the most used are physical methods, such as spray drying, freeze-drying, and extrusion, and physicochemical methods, such as liposomes, coacervation, ionic gelation, and co-crystallization [60,62]. Some microencapsulation examples of phenolic compounds are summarized in Table 1, and their basis/foundation is described later.

3.1. Physical Methods

Spray drying is one of the best technologies for microencapsulating phenolic compounds. This technique consists of the atomization of a liquid mixture integrated by the core material (phenolic compounds) and the wall (encapsulating agent) in a stream of hot air, generating water evaporation and obtaining dry microparticles with a size between 1 and 100 µm [63,64]. This method offers several advantages, including simplicity, flexibility, low cost, easy scaling, high stability of the final product, and high encapsulation efficiency. Additionally, it is suitable for heat-sensitive compounds due to its short exposure times at high temperatures [65,66]. However, the selection of coat materials is important since it can affect the properties of the microparticles. Among the main encapsulating agents, carbohydrates, gums, pectin, proteins, and mixtures stand out [65,67].
Freeze-drying: is the most efficient technique for the encapsulation of bioactive compounds; it is the most popular drying process used for compounds that are heat sensitive. The method is based on the freezing and later sublimation of water from the solid/frozen state directly to the gaseous state, applying a vacuum. Exposure to low temperatures causes lyophilized products to retain their initial nutraceutical properties; however, the microencapsulation efficiency of this technology depends on the used wall materials, among them polymers, sugars such as maltodextrin, mannose, and trehalose, milk, polyols, and others [58,68,69].
Extrusion: is a physical method in which phenolic compounds are encapsulated in hydrocolloid materials. Extrusion using natural polymers is a technique that improves bioactive compounds’ stability, limits the use of high temperatures and organic solvents, and is also low-cost [70]. Generally, extrusion microencapsulation includes three processes: (1) melt injection, (2) melt-extrusion, and (3) centrifugal extrusion (coextrusion) [71]. This process is used to produce microcapsules by forcing a stream of shell material to surround the core material, a process based on the forced pass of a solution containing phenolic compounds through nozzles using droplet-generating equipment [58,59].

3.2. Physico-Chemical Methods

Liposomes, also called lipid vesicles, are spherical microscopic structures that consist of one or more phospholipid bilayers trapping an aqueous compartment in which lipophilic and hydrophilic agents can be dissolved in the lipid membrane and in the nucleus, respectively. The size of these lipid vesicles can vary from a few nanometers to several micrometers [72,73]. Due to their size, amphiphilic character, and biocompatibility, liposomes have been used as delivery vehicles for different phenolic compounds. Its application as a carrier system for phenolic compounds depends strictly on the physicochemical properties of its membranes, its size, the nature of its components, surface charge, and lipid organization [73,74].
Coacervation is a technique that consists of the phase separation of a colloidal system in the liquid–liquid phase of a polymer or a mixture of these with opposite charges in an aqueous solution caused by electrostatic interactions, hydrogen bonds, hydrophobic interactions, and enzymatic cross-linking agents (altering ionic strength, pH, or temperature). The process includes three basic steps: the formation of three immiscible phases, the deposition of the coating, and finally, the solidification of the coating [58,75,76]. Coacervation can be simple or complex, depending on the number of polymers used. Simple coacervation employs a simple polymer that absorbs at the interface between the colloidal solution and the solvent. Complex coacervation uses two or more polymer solutions for the formation of walls around an active core [58,76]. Proteins and polysaccharides are generally used as covering materials [77,78].
Co-crystallization: is a method that uses sucrose as a matrix to incorporate bioactive compounds, it includes the preparation of a supersaturated sucrose solution, the addition of central materials, uniform mixing, and heating the mixture up to crystallization temperature [79]. This is a drying process where core materials in liquid form are directly converted into dry powder without the need for an additional drying step. Co-crystallization improves solubility, humectability, uniformity, dispersibility, hydration, anti-agglomeration, stability, and fluidity of the encapsulated bioactive compound [80].
Ionic gelation is a physicochemical method for the encapsulation of phenolic compounds. This method can be performed through atomization, electrostatic deposition, or drop procedures. It fundamentally consists of trapping an active substance and releasing it through gel phase changes (pH, mechanical wear, enzymes, and osmosis). Encapsulation starts with an aqueous polymeric solution, with low molecular mass ions that interact with polyelectrolytes of opposite charges, reacting and forming an insoluble gel [81,82]. Ion gelation is a simple procedure that does not require specialized equipment, uses relatively low temperatures and slow agitation, does not use organic solvents, and is low-cost, allowing the encapsulation of compounds that would degrade under other conditions. However, a disadvantage of this method is the low retention of hydrophilic compounds; hence, it is important to apply strategies like emulsion systems and cover material to enhance the encapsulation efficiency [83].
Table 1. Microencapsulation methodologies used in extracts rich in phenolic compounds.
Table 1. Microencapsulation methodologies used in extracts rich in phenolic compounds.
SourceEncapsulation MethodWall MaterialConditionsResultsReference
Tucuma Coproduct
(Astrocaryum vulgare Mart.) Almonds
Spray DryingMaltodextrin (5%)Temperature: 100°
C; flow rate: 7.5 mL/min, and pressure: 6 bar.
The microparticles showed spherical and heterogeneous structures and good encapsulation efficiency.[67]
Blackberry Pomace (Rubus fruticosus)Spray DryingMaltodextrin DE 10, in a 1:1 (w/w) ratioInlet drying air temperature: 170 °C; atomization pressure: 4 bar; drying air flow: 3.5 m3/h, and flow rate: 0.5 L/h.Microparticles have a rounded outer structure and are agglomerated into different sizes.[64]
Chipilin (Crotalaria longirostrata) methanolic extractsSpray DryingMaltodextrin, Arabic gum, Cajanus gum, cocoa shell pectin, Cajanus protein, and soy protein.Inlet air temperature: 120 °C; feed flow: 3 mL min−1; drop pressure: 1.35 barMicrocapsules with mostly irregular amorphous structures, smooth surfaces, and depressions. Size between 3 and 8 μm[65]
Sambucus nigra L. (elderberry)Spray DryingModified chitosan, sodium alginate, and Arabic gum.Flow rate: 4 mL/min (15%); inlet temperature: 115 °C; air pressure: 5–6 bar, and aspiration rate: 100% (36 m3/h)Very small particles (between 5 and 19 μm).[66]
Extract from Lippia citriodora leavesSpray DryingMaltodextrin and inulinInlet air temperature 135–195 °C; airflow: 0.30 m3/min; feeding flow: 2 mL/min, atomization air flow: 13 L/minInulin increased powder and polar compounds recovery, whereas maltodextrin achieved a higher encapsulation efficiency.[63]
Ciriguela (Spondias purpurea L.)Freeze-dryingMaltodextrin 10 DE and arabic gum48 h in a freeze dryer at −80 °C and 0.28 mbar chamber pressure.Microcapsules with irregular shape, extensive wrinkles, and a serrated surface.[84]
Blackberry (Rubus fruticosus)Freeze-dryingChitosan, xanthan, β-cyclodextrin, and hydrogelMixture: 0.003 mol of polymer and the same proportion of extract, diluted in 50 mL of water. The solution was frozen at −80 °C for 24 h, with subsequent lyophilization.Only chitosan and xanthan showed the characteristic shape.[68]
Blueberry (Vaccinium myrtillus) JuiceFreeze-dryingHP-β-cyclodextrin and β-cyclodextrinβ-CD in 15% (w/w) ratio to hot (75 °C) blueberry juice. The precipitated product was freeze-dried at −50 °CFormation of amorphous material and a 78.1% product yield.[69]
Pomegranate (Punica granatum L.)Freeze-dryingMaltodextrin (20 DE)The extract and maltodextrin mixture (1:2 (w/w)) was lyophilized at −30 °C and vacuum pressure: 0.04 mbar.Homogeneous coating on particle surface.[85]
Black chokeberry
(Aronia melanocarpa)
Indirect extrusionSodium alginate, low-molecular-weight chitosan, carrageenan, Low-methoxyl pectinAlginate was mixed in equal proportions (1:1 g/g) with other encapsulants. Encapsulator; vibrating nozzle: 150 m; pressure: 200 mbar; frequency: 400 Hz; electrode: 1000 V; solidification temperature: 30 °C and complexation time: 10 min.Hydrogel beads differ in shape and structure. The most regular capsules were obtained with the mixture of alginate + carrageenan.[70]
Papaya fruit (Carica papaya L.)ExtrusionPectin-alginateThe papaya extract was encapsulated through the in situ and two-step methodologies. Alginate:pectin ratio was 55:45.Bioactive compounds are dispersed in the encapsulation matrix, improving their thermal stability.[86]
Proanthocyanidin cinnamon extractComplex coacervationGelatin and five different polysaccharides (gum Arabic, pectin, cashew tree gum, carboxymethylcellulose, and κ-carrageenanThe proanthocyanidin-rich cinnamon extract was dispersed in distilled water. The gelatin dispersion was added, and then the polysaccharide solution. The decanted material was frozen at −20 °C and dried in freeze-dryer.Particles presented resistance when submitted to different stress conditions, except pH lower than 2 and temperatures higher than 50 °C.[77]
Polyphenols from oat branComplex coacervationWhey protein concentrates 10%
Maltodextrin 10%
The wall materials were mixed in ratios 10:0, 8:2, 6:4, 4:6, and 2:8 by gentle magnetic stirring for 1 h. BAS extract was then added to the wall material at 10% (1:10 ratio) and the microcapsules solution was formed using a Magnetic Stirrer for 15 min.The encapsulation efficiency was 95.28%. The release percentage of polyphenols coated in a capsule ranged between 70 and 83% after 2 h of digestion.[87]
(−)-Epigallocatechin gallate (≥94%)LiposomesPhospholiponPhospholipon and Epigallocatechin gallate were dissolved in ethanol. Citric acid (0.1%) was added while stirring, and the mixture was heated to 60 °C. The microparticles were prepared using an encapsulator.Encapsulation efficiency (>97%) and sustained release; in 14 days, no more than 15% of EGCG was released. The sizes of the liposomes were estimated at 1–2 μm.[88]
Grape-seed extractLiposomesSoy lecithinGrape-seed extract was incorporated into liposomes (1.1% w/w soy lecithin) using high-pressure homogenization (22,500 psi).Entrapment efficiency for uncoated liposomes was 88.2 ± 4.7%. The release rate after 24 h from uncoated liposomes was 0.55*h.[72]
Green tea extract (C. sinensis)Ionic gelationAmidated low methoxyl pectin, calcium chloride,
hydrogenated palm oil
Association of a double emulsion (water/oil/water) with ionic gelation. The final emulsion was sprayed through a double-fluid atomizer on a CaCl2 crosslinking solution acidified with citric acid (pH 3).72.6 ± 0.4% encapsulation efficiency for ionic gelation microparticles.[81]
Anthocyanins from Hibiscus sabdariffa L. calycesIonic gelationRapeseed oil, pectin, calcium chlorideIonic gelation using two techniques: drip-extrusion and atomization, both using a double emulsion (Hibiscus extract/rapeseed oil/pectin) and a cross-linked solution (CaCl2).The median diameter (D50) of the particles ranged from 78 to 1100 μm, and encapsulation efficiency ranged from 67.9 to 93.9%.[83]
Securigera securidaca (L) seed extractCo-crystallizationSaccharoseSucrose and S. securidaca extract were mixed on a heater at 132 °C. The co-crystallized product was dried in an oven at 40 °C for 15 h, then ground and sieved.The production efficiency and moisture content of the extract-containing co-crystallized powder were 84% and 0.14%, respectively.[89]
Pomegranate Peel
Extract
Co-CrystallizationFood-grade crystal sucroseSucrose solution and extract were mixed at 700 rpm. The mixture is placed in a water bath and stirred until it reaches 45 °C. The powder is kept in a desiccator for 24 h.The co-crystallized powder had low moisture content (0.59%), low hygroscopicity (0.011%), high apparent density (0.803 g/cm3) and solubility (61 s).[90]

4. Nanoencapsulation

One emergent technology used to entrap polyphenolic compounds is nanotechnology; this science involves the design of nanoscale systems (particle size 1–100 nm). The size allows it to pass through the tissue and reach the sites of interest, since it increases the surface–volume ratio; therefore, it is used in different disciplines such as biology, chemistry, and medicine [91,92,93]. The main objective of nanoencapsulation is protect an active ingredient (gas, solid, or liquid) with a matrix or shell, to form different types of nanoparticles, such as nanosphere, nanocapsules, nanoemulsion, nanoliposome, and nanoniosome, using different nanoencapsulation techniques such as deprotonation, ionic crosslinking, pH-regulated self-aggregation, polyelectrolyte complexation, ionic gelation, and hydrophobic modification, coacervation, nanoprecipitation, emulsification, layer-by-layer, sonication, desolvation, reverse-phase evaporation, supercritical fluid, electrospray, nano spray drying [94,95,96,97]. Depending on the characteristics mentioned above and the encapsulation technique, the polyphenolic compound can be found dissolved within the nanoparticle, dispersed, trapped, or adsorbed [98]. On the other hand, there are many wall materials, some of the most commonly used being chitosan, gold (chloroauric acid), silver, mesoporous silica, hyaluronic acid, sodium alginate, polylactides (PLA), albumin, gelatin, poly(lactide)-poly(ethylene glycol) (PLA-PEG), poly(lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG), polyglycolides (PGA), lecithin, polyglutamic acid, wheat protein, β-Lactoglobulin, among others [93,94,95]. All these materials must meet certain safety requirements, such as being non-toxic, easily degradable, and having physicochemical properties compatible with the polyphenol for better release [95].
In this sense, the wall material, polyphenol, and the nanoencapsulation technique are considered in the design of nanoformulations (Table 2, Figure 2) to improve the release of polyphenolic compounds in the specific targets of action. It has been shown that polyphenols influence different non-communicable diseases such as diabetes. For this reason various researchers have dedicated efforts to developing nanoformulations loaded with these compounds, due to the great advantages offered by nanoencapsulation, such as increasing its effectiveness by having a smaller size, improving solubility, in addition to protecting the compound from the degradation process caused by environmental factors such as light, changes in pH, temperature and radiation, and lastly and most importantly, a better bioavailability of the compounds is achieved. This could reduce the negative effects and help achieve greater specificity of the active compound or polyphenol to enhance its therapeutic action [91,99,100].

5. Current Evidence Regarding the Efficacy of Encapsulated Polyphenols

5.1. In Vitro

As mentioned above, diabetes mellitus is one of the most prevalent diseases worldwide, which is why many studies have focused on investigating this disease and how to improve its symptoms. In vitro analysis is one of the most widely used techniques to determine the beneficial effect of encapsulated polyphenols against diabetes. One of the more commonly used in vitro studies is simulated digestion, as phenolic compounds have a major bioavailability problem. To mention some examples, Verônica Cardoso de Souza et al. [123] studied Bauhinia forficata, a plant rich in polyphenols that is mainly used for its hypoglycemic activity, which is related to its antioxidant and anti-inflammatory potential, performed nanoencapsulation of infusion and decoction of B. forficata leaves using spray drive using maltodextrin and colloidal silicon dioxide as wall material, reporting that the nanoencapsulated flavonoid compounds were bioaccessible after the gastric phase (49.38% and 64.17% of polyphenols and 64.08% and 36.61% of flavonoids) and duodenal (52.68% and 79.06% of polyphenols and 13.24% and 139.03% of flavonoids), with a variation of 52.27% to 70.55% of the antioxidant activity maintained, by the ORAC method, after gastric digestion and still 25% after duodenal, concluding that nanoencapsulation is a very viable technique for the conservation of bioactive compounds.
On the other hand, another of the most widely used in vitro techniques is the inhibition of enzymes related to carbohydrate metabolism. A clear example is the research carried out by Kerbab et al. [124], which studied the effect of the shrub Halimium halimifolium as an antidiabetic agent, finding that the phenolic compounds of this shrub have great antioxidant capacity and antidiabetic potential by inhibiting the enzymes α-amylase and α-glucosidase (IC50 = 0.82 mg/mL and 25.01 μg/mL, respectively); likewise, the authors performed microencapsulation of the compounds to optimize stability, handling, and delivery of bioactive compounds, using microencapsulation through spray drying and cellulose acetate phthalate as wall material. Likewise, Zorzenon et al. [125] evaluated maltodextrin microcapsules containing ethanolic extract of Stevia, by means of driver spray encapsulation, analyzed physicochemical parameters, antidiabetic activity (through inhibition of α-amylase), cytotoxicity, bioaccessibility of the compounds by in vitro digestion, as well as the structure of the microcapsules by scanning electron microscopy, the microcapsules showed greater solubility (~35%), lower moisture content (~29%) and maltodextrin DE10 had higher efficiency as an encapsulating agent (87%) compared to DE19 (76%) and showed well-defined spherical structures. Microencapsulation preserved the phenolic compound content and antioxidant activity present in the extract (7.2% and 87.5%, respectively). De Silva et al. [126] evaluated nanoencapsulated compounds of Bael fruit, using nanoencapsulation by ionic gelation using alginate as wall material; the authors report that with this nanoencapsulation, the compounds were more stable and that it enhanced the antidiabetic, antioxidant, and anti-inflammatory effects by having a slower and more controlled release profile with respect to non-encapsulated compounds.
Another of the routes studied for the determination of the antidiabetic effect of polyphenols is the glucose transporters GLUT-4. Chauhan et al. [127] studied chitosan-encapsulated nanocurcumin and the impact it has on the translocation of this glucose transporter, reporting that chitosan-nanocurcumin capsules caused an increase in the translocation of GLUT-4 to the cell surface in L6 skeletal muscle cells. The effect was associated with an increase in the phosphorylation of AKT (Ser-473) and its subsequent target GSK-3β (Ser-9).

5.2. In Vivo

In recent years, there has been an interest in studying the in vivo effect of encapsulated polyphenols, namely phenolic acids or flavonoids, and the polyphenolic-rich extracts from natural sources, as antidiabetic agents. According to Pandey and Dvorakova [128], the most common in vivo model for testing antidiabetic drugs is the induction of diabetes in rodents using streptozotocin (STZ). This drug has been used in several doses (10–150 mg/kg) and is usually administered orally or intraperitoneally. The mode of action of STZ to induce diabetes is to selectively damage the pancreatic β-cells present in the islets of Langerhans through several mechanisms, so the pancreas stops producing insulin, consequently inducing type I diabetes in a single dose [129]. Type I diabetes is, therefore, the most studied in rodent models. Inducing type II diabetes in vivo in rats or mice is more laborious. Several strategies are used together with the STZ administration, for example, using high-fat diets, nicotinamide, or rodents genetically susceptible to developing diabetes [128]. Even though these models show some disadvantages, such as high cost and variability, they are still relevant to studying diabetes and potential antidiabetic drugs.
Regarding the use of individual polyphenols as antidiabetic agents Panwar, Raghuwanshi, Srivastava, Sharma, and Pruthi [130] evaluated the antihyperglycemic effect of chitosan-encapsulated ferulic acid in diabetic Wistar rats. These authors reported that encapsulated ferulic acid significantly (compared with the diabetic group) reduced the levels of blood glucose and increased the secretion of insulin, as well as restoration of the pancreatic islets of Langerhans. Additionally, they observed a reduction in total cholesterol and triglycerides, which are biochemical markers of hyperlipidemia caused by diabetes complications. Nanoparticles of the flavonoid hesperidin were evaluated in a nicotinamide + STZ-induced diabetic model in male albino rats. After administering encapsulated hesperidin, the rats showed significantly lower plasma glucose concentration and increased insulin levels than the diabetic control group. Furthermore, the pancreatic islets of Langerhans were restored in rats treated with the nanoparticles of hesperidin. In contrast, rats treated with metformin still showed degeneration in the pancreatic cell clusters caused by the STZ. Other research evaluating the antidiabetic effect of diverse individual polyphenols can be reviewed in Table 3. It is important to mention that most in vivo studies have reported that encapsulating phenolic compounds improves their antidiabetic effects compared to free phenolics.
Encapsulated phenolic extracts from several herbs and plants are also investigated in models of rodents for their antidiabetic properties. The advantage of studying plant extracts over individual polyphenols could provide information regarding the synergistic effect that several compounds found in an extract might exert, therefore potentiating their biological effect [131]. In this sense, a poly-herbal (Justicia glabr, Adhatoda zeylanica, Andrographis paniculata, Gymnema sylvestre, Andrographis alata, and Syzygium cumini) ethanolic extract encapsulated with chitosan (particle size 62.6 ± 2.15 nm) was administered to diabetic rats for 30 days. After the experimental period, the rats exhibited significantly lower glucose concentrations and HbA1c levels, along with increased insulin and liver glycogen levels compared to the diabetic control group [132].
Table 3. Recently reported in vivo effects of encapsulated polyphenols as antidiabetic agents.
Table 3. Recently reported in vivo effects of encapsulated polyphenols as antidiabetic agents.
CompoundPolymer/Particle SizeDosageIn Vivo ModelEffect *Ref.
ChrysinPLGA/176.0 ± 2.1 nmOne administration of 20 mg/kgSTZ-induced diabetes in male albino rats (180–200 g)↓ Blood glucose
↓ TG, LDL
↑ HDL
[133]
CurcuminChitosan/n.s.150 mg/kg once a day, for 28 daysSTZ-induced type 1 diabetes in C57Bl/6 mice↓ Blood glucose
↑ Insulin secretion
↓ Fibrosis in the kidney
[134]
Ferulic acidChitosan/211.3 ± 5.1 nm10 mg/kg once a day, for 14 daysSTZ-induced diabetes in Wistar albino rats (110–150 g)↓ Blood glucose
↑ Plasma insulin levels
↓ TC, TG
-Recovered islets of Langerhans in the pancreas
[130]
HesperidinMgAl-double layered hydroxide/330–380 nm50 mg/kg once a day, for 30 daysNicotinamide + STZ-induced diabetes in male albino rats (200–300 g)↓ Plasma glucose, HbA1c
↑ Insulin, HOMA-B
-Restored the pancreatic Islets of Langerhans
[135]
LiquiritinPhospholipid complex/91.8 ± 1.9 nm200 mg/kg once a day, for 28 daysSTZ-diabetes induced in male ICR mice (18–22 g)↓ Blood glucose
-Improved the glomerular
and renal cortical structure of the kidney
[136]
MangiferinLabrafil M 2130 CS/138.4 ± 3.4 nmOne administration of 40 mg/kgHigh-fat diet + STZ-diabetes induced in male Wistar rats (250 g)↓ Blood glucose
↓ TC, TG
↑ HDL
↓ AST, ALT
[137]
MangiferinNSC-alginate/124 nm10 mg/kg once a day, for 28 daysSTZ-induced diabetes in Wistar rats (100–150 g)↓ Blood glucose
↓ TC, TG, LDL
↑ HDL
[138]
MyricetinChitosan/184.4 ± 4.1 nm50 mg/kg once a day, for 28 daysSTZ-induced diabetes in male Wistar rats (~250 g)↓ Blood glucose
↓ TG, TC
↑ BW
[139]
NaringeninPhospholipid LECIVA-S70/564.4 nmSingle dose of 25 mg/kg or
50 mg/kg, for 28 days
STZ-induced diabetes in male Sprague Dawley rats (180–220 g)↓ Plasma glucose level
↓ TC, TG, BUN
↓ ALT, AST
[140]
NaringeninPLGA/129 nmOne dose of 10 mg/kg, and a second dose after 10 days, period of 7–49 daysSTZ-induced diabetes in male Wistar rats↓ Blood glucose
↑ Insulin level
↓ HbA1c
-Restored pancreas and kidney cells
[141]
QuercetinEudragit L-100/144.7 ± 1.7 nm200 mg/kg once a day, during 21 daysSTZ-induced diabetes in albino female Wistar rats (150–200 g)↓ Blood glucose
↓ TG, TC, LDL
↓ ALP, ALT, AST
↓ cellular damage in the pancreas
[142]
QuercetinPLGA/179.9 ± 11.2 nm150 mg/kg every 5th day, during
15 days
STZ-stimulated male Sprague-Dawley rats (~250 g)↓ Blood glucose[143]
QuercetinPoloxamer-180-stearic acid/157.1 to 528.2 nm5 or 10 mg/kg, for 21 daysSTZ-induced diabetic retinopathy in male adult zebra fish (<8 months)↓ Plasma glucose[144]
ResveratrolChitosan/38.0 nm100 mg/kg, for 28 daysSTZ-indued gestational diabetes mellitus in Wistar albino rats (180–200 g)↓ Blood glucose
↑ Insulin level
↓ TC, TG, LDL
↑ HDL
[145]
* Compared with the diabetic control group. ALP: alkaline phosphate; ALT: alanine aminotransferase; AST: aspartate aminotransferase; BUN: blood urea nitrogen; BW: body weight; HbA1c: glycosylated hemoglobin; HDL: high density lipoproteins; HOMA-B: homeostasis model assessment of β-cell function; LDL: low density lipoproteins; n.s.: not specified; NSC: N-succinylated chitosan; PLGA: DL-polylactide/glycolide copolymer 75/25; STZ: streptozotocin; TC: total cholesterol; TG: triglycerides. ↑ means increased. ↓ means decreased.
Furthermore, encapsulated (maltodextrin + whey protein) extracts from coffee parchment containing chlorogenic acid significantly reduced the glucose and HOMA-IR levels in obese male Wistar rats. Rats treated with the encapsulated extracts exhibited better biochemical parameters, since the TC, TG, and AST and ALT levels were significantly lowered; furthermore, TC and TG in the liver were also diminished [146]. An encapsulated anthocyanin-rich extract from the fruit of Vaccinium meridionale was administered to obese C57BL/6 mice, which significantly reduced the glucose and TC levels, when compared with the obese control group [147]. Other research evaluating the antidiabetic effect of polyphenolic-containing extracts is mentioned in Table 4.
It has been stated that polyphenols reduce ROS levels, inflammation, and oxidative stress in pancreatic-damaged cells, which helps to restore and maintain the correct functionality of β-cells and the regulation of insulin secretion. Other proposed antidiabetic mechanisms of polyphenols are related to the inhibition of: (1) digestive enzymes, (2) dipeptidyl-peptidase IV, (3) glycation of proteins, and (4) diabetic-related complications, among others [148]. The evidence suggests that encapsulated polyphenols, both individually and in extract, show promising attributes to be considered in the management of diabetes.
Table 4. Recently reported in vivo effects of encapsulated polyphenolic extracts from plants as antidiabetic agents.
Table 4. Recently reported in vivo effects of encapsulated polyphenolic extracts from plants as antidiabetic agents.
Plant SpecieComponents of the ExtractEncapsulating Material/Particle SizeIn Vivo Model, DosageEffect *Ref.
Cinnamomoum osmophloeum KanehiraCinnamaldehyde, benzoic acid, caffeic acid, caffeoylquinic acid, cinnamic acid, coumaric acid, rutin, kaempferol, eugenol, quercetin, and derivativesNanoemulsion (soybean oil, lecithin and Tween 80)/
36.6 nm
Nicotinamide + STZ-induced diabetes in male Wistar rats (7 weeks old), 60 mg/kg (cinammaldehyde equivalents)↓ Blood glucose, HOMA-IR
↓ TC, TG, AST, ALT, BUN
[149]
Coccinia grandisPhenolics and flavonoidsGelatin/
468 ± 14 nm
High-fat diet + STZ-induced diabetes in male Wistar rats (135–165 g), single dose of 330 mg/kg↓ Plasma glucose[150]
Coffea arabicaCaffeine, chlorogenic acidMaltodextrin + whey protein/
1–2 µm
Fructose-induced obesity in male Wistar rats (85–120 g), 100 mg/kg per day (during 28 days)↓ Glucose, HOMA-IR
↓ TC, TG, AST, ALT
↓ Liver-TG, liver-TC
[146]
Murraya koenigiiPhenolics and flavonoidsGelatin/
520 ± 33 nm
High-fat diet + STZ-induced diabetes in male Wistar rats (135–165 g), single dose of 65 mg/kg↓ Plasma glucose[150]
Posidonia oceanicaHydroxybenzoic acid, protocatechuic acid, ferulic acid, gallic acid, coumaric acid, sinapic acid, vanillic acid, catechin, epicatechin, luteolin, naringenin, apigenin, among others.Bovine gelatine/
274.7 ± 30.5
STZ-induced diabetes in male Wistar albino rats (150–170 g), 100 mg/kg (for 28 days)↓ Glucose, HOMA-IR
↑ GLUT4
[151]
Senna auriculataPhenolics and flavonoidsGelatin/
563 ± 4 nm
High-fat diet + STZ-induced diabetes in male Wistar rats (135–165 g), Single dose of 45 mg/kg↓ Plasma glucose[150]
Vaccinium meridionaleDelphinidin 3-hexoside, cyanidin-3-galactoside, cyanidin-3-glucoside, cyanidin 3-arabinosidePro-nanosome Nio-N/219.7 ± 3.1 nmHigh-fat diet-induced obesity in C57BL/6 mice, 160 µg/mL (during 28 days)↓ Glucose
↓ TC, leptin
[147]
* Compared with the diabetic control group. ALT: aAlanine aminotransferase; AST: aAs-partate aminotransferase; BUN: bBlood urea nitrogen; GLUT4: iInsulin-regulated glucose transporter; HOMA-IR: hHomeostasis model assessment-insulin resistance; STZ: sStrep-tozotocin; TC: tTotal cholesterol; TG: tTriglycerides. ↑ means increased. ↓ means decreased.

6. ADMET Analysis of Polyphenols with Antidiabetic Properties

Some of the chemical characteristics of a potential drug agent can be used to evaluate the drug-likeness of a molecule; this is called Lipinski’s rule of 5 [152]; these are (1) molecular weight below 500, (2) the molecule has no more than 5 hydrogen bond donors, (3) the molecule has no more than 10 hydrogen bond acceptors, and (4) the partition coefficient (Log p) is under 5.
These characteristics can help us predict the passive absorption of a molecule. In Table 5, we summarize the polyphenols with antidiabetic properties and their potential bioavailability using the rule of 5, and in Figure 3 we show the chemical structures of the polyphenols reported with antidiabetic properties.
One important point to consider about bioactive compounds is their potential toxicity, which is why in Table 6, we provide a prediction of the toxicity of phenolic compounds with possible antidiabetic activity. ProTox 3.0 was used for this prediction. This program incorporates molecular similarity, fragment propensities, most frequent features, and (fragment similarity-based CLUSTER cross-validation) machine-learning, based on a total of 61 models for the prediction of toxicity endpoints such as acute toxicity, organ toxicity, toxicological endpoints, molecular initiating events, metabolism, adverse outcomes (Tox21) pathways, and toxicity targets [153].
Among the interactions of greatest interest for understanding the metabolic profile and pharmacological safety of bioactive compounds are those with cytochrome P450 (CYP) isoenzymes, mainly CYP1A2, CYP2C19, CYP2C9, CYP2D6, CYP3A4, and CYP2B6. These enzymes play a key role in the metabolism of xenobiotics and drugs, directly influencing their half-life, bioavailability, and the efficiency of the compounds. For polyphenols with antidiabetic activity, analyzing their ability to inhibit these isoforms helps us predict possible drug interactions and unwanted effects, as well as optimize their stability [154,155,156,157]. Table 7 shows the possible interactions that these polyphenols may have with CYP.
In turn, to understand the biological behavior of polyphenolic compounds, it is a priority to evaluate pharmacokinetic parameters such as stability in human liver microsomes (HLM stability), permeability through the blood–brain barrier (BBB), inhibition of P-glycoprotein (P-gp), and gastrointestinal absorption (GI absorption). For example, HLM stability provides an estimate of the potential hepatic metabolic susceptibility, a key factor in determining the bioavailability and half-life of these compounds [158]. BBB is important because of the possible involvement of the central nervous system in metabolic regulation and modulation of oxidative stress in diabetes [159,160]. On the other hand, the inhibition of P-gp is involved in both tissue disposition and therapeutic efficacy, since it can modify the active transport and elimination of these compounds [161,162]. Likewise, adequate gastrointestinal absorption is key to ensuring effective plasma levels, since many compounds have low solubility. That is why Table 8 presents the predictions for polyphenolic compounds with potential antidiabetic activity in relation to these pharmacokinetic parameters.

7. Future Directions and Perspectives for Clinical Translation

The growing evidence reviewed in this article shows that encapsulation technologies can enhance the bioavailability and antidiabetic efficacy of polyphenols. However, most studies are limited to in vitro experiments, and in vivo experiments only yield short-term results, which hinder clinical translation.
Therefore, translating the summarized results into clinical applications of polyphenol antidiabetic agents requires coordinated advances, such as selecting optimal wall materials, polyphenol interaction with these materials, and standardizing individual polyphenol concentrations to find effective doses. For instance, researchers must optimize encapsulation techniques for specific polyphenols, depending on their chemical structures and properties, and align these techniques with formulation parameters while considering pharmacokinetic goals and pharmacological applications. Novel delivery systems, such as nanoparticles and multi-component systems, hold significant potential as targeted systems to be released with physiological triggers or to co-deliver polyphenols with enzyme inhibitors or hormones. The natural carriers, such as plant-derived exosomes, demonstrate multi-target activity in animal models as reported in the literature, but currently face challenges in standardization, targeting, and long-term safety. Particularly, long-term safety is not reported as in vivo studies are mostly designed for short-term responses.
Furthermore, many studies mentioned in this review use crude plant extracts or polyphenol-rich mixtures, which causes variability in the preparations and complicates the interpretation and translation to clinical applications. In silico molecular docking and molecular docking dynamic simulations can guide the selection of optimal wall materials and crosslinkers by predicting interactions with polyphenols. Future studies should correlate physicochemical parameters with pharmacokinetic outcomes to facilitate translation.

8. Conclusions

Diabetes mellitus is a global health emergency affecting millions of individuals; it impacts society at an economic level, but also affects life expectancy and life quality. Diagnosing and treatment of prediabetes and DM are crucial. Still, limited access to medications and health care in middle and low-income populations influences treatment adherence and increases the risk of developing other health complications. In this regard, research aiming to create safe and effective alternatives obtained from natural sources represents a promising strategy. Research and technology have made it possible to protect compounds such as polyphenols by encapsulating them in different materials and with other methods. Choosing the appropriate and safe polyphenol dosages to achieve the antidiabetic effect is important. As discussed in this paper, selecting the proper encapsulation material, specific delivery, and polyphenols (isolated, mixed, or in addition to other bioactive compounds) must also be a priority to ensure bioavailability and nutraceutical properties.

Author Contributions

R.C.-P.d.L. and E.P.G.-G. designed the work; R.C.-P.d.L., F.I.C.-T., N.O., C.A.E.-R., L.A.C.-A., N.L.-L., M.d.J.B.-M., J.B.H., and E.P.G.-G. prepared the review article; E.P.G.-G. supervised the manuscript work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

All the data collected can be found in this document.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAAmerican Diabetes Association
ADMEAbsorption, distribution, metabolism, and excretion
ALPAlkaline phosphate
ALTAlanine aminotransferase
ASTAspartate aminotransferase
BBBBlood–brain barrier
BUNBlood urea nitrogen
BWBody weight
CATCatalase
DMDiabetes mellitus
FPGFasting plasma glucose
GLUT4Insulin-regulated glucose transporter
GPXGlutathione peroxidase
Hb1A1cGlycosylated hemoglobin
HDLHigh density lipoproteins
HLMThe human liver microsomal
HOMA-BHomeostasis model assessment of β-cell function
HOMA-IRHomeostasis model assessment-insulin resistance
IC50Inhibitory Concentration 50
LUVUnilamellar vesicles
LDLLow density lipoproteins
LMPHLongzhua mushroom polysaccharide hydrogel
MLVMultilamellar vesicles
NLCsNanostructured Lipid Carriers
NSCN-succinylated chitosan
OGTTOral glucose tolerance test
PGApolyglycolides
P-gpP-glycoprotein
PLAPolylactides
PLA-PEGpoly(lactide)-poly(ethylene glycol)
PLGADL-polylactide/glycolide copolymer
PLGA-PEGpoly(lactide-co-glycolide)-poly(ethylene glycol)
SODSuperoxide dismutase
SLNsSolid Lipid Nanoparticles
STZstreptozotocin
TBRASThiobarbituric acid reactive substances
TCTotal cholesterol
TGTriglycerides
T2DMType 2 Diabetes mellitus
2-hPG2 hPlasma glucose

References

  1. American Diabetes Association Professional Practice Committee. 2. Diagnosis and Classification of Diabetes: Standards of Care in Diabetes—2024. Diabetes Care 2023, 47, S20–S42. [Google Scholar] [CrossRef] [PubMed]
  2. Popoviciu, M.S.; Paduraru, L.; Nutas, R.M.; Ujoc, A.M.; Yahya, G.; Metwally, K.; Cavalu, S. Diabetes Mellitus Secondary to Endocrine Diseases: An Update of Diagnostic and Treatment Particularities. Int. J. Mol. Sci. 2023, 24, 12676. [Google Scholar] [CrossRef]
  3. Sharma, P.; Hajam, Y.A.; Kumar, R.; Rai, S. Complementary and alternative medicine for the treatment of diabetes and associated complications: A review on therapeutic role of polyphenols. Phytomed Plus 2022, 2, 100188. [Google Scholar] [CrossRef]
  4. Vlacho, B.; Rossell-Rusiñol, J.; Granado-Casas, M.; Mauricio, D.; Julve, J. Overview on chronic complications of diabetes mellitus. In Chronic Complications of Diabetes Mellitus; Elsevier: Amsterdam, The Netherlands, 2024; pp. 1–10. [Google Scholar]
  5. International Diabetes Federation. Diabetes Atlas. Available online: https://diabetesatlas.org/atlas-reports (accessed on 7 June 2025).
  6. World Health Organization. Diabetes. Available online: https://www.who.int/health-topics/diabetes#tab=tab_1 (accessed on 1 August 2024).
  7. Chandrasekaran, P.; Weiskirchen, R. The Role of Obesity in Type 2 Diabetes Mellitus—An Overview. Int. J. Mol. Sci. 2024, 25, 1882. [Google Scholar] [CrossRef]
  8. Rohm, T.V.; Meier, D.T.; Olefsky, J.M.; Donath, M.Y. Inflammation in obesity, diabetes, and related disorders. Immunity 2022, 55, 31–55. [Google Scholar] [CrossRef]
  9. Li, Y.; Schoufour, J.; Wang, D.D.; Dhana, K.; Pan, A.; Liu, X.; Song, M.; Liu, G.; Shin, H.J.; Sun, Q. Healthy lifestyle and life expectancy free of cancer, cardiovascular disease, and type 2 diabetes: Prospective cohort study. BMJ 2020, 368, l6669. [Google Scholar] [CrossRef]
  10. Lingvay, I.; Sumithran, P.; Cohen, R.V.; le Roux, C.W. Obesity management as a primary treatment goal for type 2 diabetes: Time to reframe the conversation. Lancet 2022, 399, 394–405. [Google Scholar] [CrossRef]
  11. Magkos, F.; Hjorth, M.F.; Astrup, A. Diet and exercise in the prevention and treatment of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2020, 16, 545–555. [Google Scholar] [CrossRef]
  12. Akbari, B.; Baghaei-Yazdi, N.; Bahmaie, M.; Mahdavi Abhari, F. The role of plant-derived natural antioxidants in reduction of oxidative stress. BioFactors 2022, 48, 611–633. [Google Scholar] [CrossRef] [PubMed]
  13. Pasupuleti, V.R.; Arigela, C.S.; Gan, S.H.; Salam, S.K.N.; Krishnan, K.T.; Rahman, N.A.; Jeffree, M.S. A review on oxidative stress, diabetic complications, and the roles of honey polyphenols. Oxidative Med. Cell. Longev. 2020, 2020, 8878172. [Google Scholar] [CrossRef] [PubMed]
  14. Pérez-Torres, I.; Castrejón-Téllez, V.; Soto, M.E.; Rubio-Ruiz, M.E.; Manzano-Pech, L.; Guarner-Lans, V. Oxidative stress, plant natural antioxidants, and obesity. Int. J. Mol. Sci. 2021, 22, 1786. [Google Scholar] [CrossRef]
  15. Unuofin, J.O.; Lebelo, S.L. Antioxidant effects and mechanisms of medicinal plants and their bioactive compounds for the prevention and treatment of type 2 diabetes: An updated review. Oxidative Med. Cell. Longev. 2020, 2020, 1356893. [Google Scholar] [CrossRef]
  16. Arkoub-Djermoune, L.; Boulekbache-Makhlouf, L.; Zeghichi-Hamri, S.; Bellili, S.; Boukhalfa, F.; Madani, K. Influence of the Thermal Processing on the Physico-Chemical Properties and the Antioxidant Activity of A Solanaceae Vegetable: Eggplant. J. Food Qual. 2016, 39, 181–191. [Google Scholar] [CrossRef]
  17. Aryal, D.; Joshi, S.; Thapa, N.K.; Chaudhary, P.; Basaula, S.; Joshi, U.; Bhandari, D.; Rogers, H.M.; Bhattarai, S.; Sharma, K.R.; et al. Dietary phenolic compounds as promising therapeutic agents for diabetes and its complications: A comprehensive review. Food Sci. Nutr. 2024, 12, 3025–3045. [Google Scholar] [CrossRef]
  18. Naz, R.; Saqib, F.; Awadallah, S.; Wahid, M.; Latif, M.F.; Iqbal, I.; Mubarak, M.S. Food Polyphenols and Type II Diabetes Mellitus: Pharmacology and Mechanisms. Molecules 2023, 28, 3996. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, Y.; Alkhalidy, H.; Liu, D. The Emerging Role of Polyphenols in the Management of Type 2 Diabetes. Molecules 2021, 26, 703. [Google Scholar] [CrossRef]
  20. Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A concise overview on the chemistry, occurrence, and human health. Phytother. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef] [PubMed]
  21. Sánchez Arribas, A.; Moreno, M.; Moreno, G.A.; Bermejo, E.; Zapardiel, A.; Chicharro, M. Characterization of White Wines by Electrochemical Indexes Obtained Using Carbon Nanotube-modified Electrodes. Electroanalysis 2018, 30, 1461–1471. [Google Scholar] [CrossRef]
  22. Barbosa, S.; Pardo-Mates, N.; Hidalgo-Serrano, M.; Saurina, J.; Puignou, L.; Núñez, O. Detection and Quantitation of Frauds in the Authentication of Cranberry-Based Extracts by UHPLC-HRMS (Orbitrap) Polyphenolic Profiling and Multivariate Calibration Methods. J. Agric. Food Chem. 2018, 66, 9353–9365. [Google Scholar] [CrossRef]
  23. Brinsi, C.; Jedidi, S.; Sammari, H.; Selmi, H.; Sebai, H. Antidiarrheal, anti-inflammatory and antioxidant effects of Anethum graveolens L. fruit extract on castor oil-induced diarrhea in rats. Neurogastroenterol. Motil. 2024, 36, e14892. [Google Scholar] [CrossRef] [PubMed]
  24. Arslaner, A.; Türkoğlu, Z. A potential antiviral and food-derived healty ingredient: Resveratrol. Food Health 2021, 7, 54–63. [Google Scholar] [CrossRef]
  25. Santos Sánchez, N.; Salas-Coronado, R.; Villanueva, C.; Hernandez-Carlos, B. Antioxidant Compounds and Their Antioxidant Mechanism; InTech Open: Rijeka, Croatia, 2019. [Google Scholar]
  26. Xuan Hoan, N.; Anh, L.; Quan, D.; Cuong, D.; Thai Ha, H.; Minh, N.; Hieu, D.; Thuat, N.; Pham, T.; Tuyen, D. Functional-Antioxidant Food; InTech Open: Rijeka, Croatia, 2021. [Google Scholar]
  27. Pinna, N.; Ben Abbou, S.; Ianni, F.; Angeles Flores, G.; Pietercelie, A.; Perretti, G.; Blasi, F.; Angelini, P.; Cossignani, L. Phenolic compounds from pumpkin pulp: Extraction optimization and biological properties. Food Chem. X 2024, 23, 101628. [Google Scholar] [CrossRef] [PubMed]
  28. Upadhyaya, B.; Moreau, R.; Majumder, K. Antioxidant and Anti-Inflammatory Capacities of Three Dry Bean Varieties after Cooking and In Vitro Gastrointestinal Digestion. J. Agric. Food Chem. 2024, 72, 18445–18454. [Google Scholar] [CrossRef]
  29. Marzagalli, M.; Battaglia, S.; Raimondi, M.; Fontana, F.; Cozzi, M.; Ranieri, F.R.; Sacchi, R.; Curti, V.; Limonta, P. Anti-Inflammatory and Antioxidant Properties of a New Mixture of Vitamin C, Collagen Peptides, Resveratrol, and Astaxanthin in Tenocytes: Molecular Basis for Future Applications in Tendinopathies. Mediat. Inflamm. 2024, 2024, 5273198. [Google Scholar] [CrossRef]
  30. Çetin, R.; Bahadir, S.; Basar, İ.; Aslanoglu, B.; Atlas, B.; Kaya, S.; Güzel, B.C.; Turan, Y. Neuroprotective effects of the combined treatment of resveratrol and urapidil in experimental cerebral ischemia-reperfusion injury in rats. Acta Cir. Bras. 2024, 39, e395329. [Google Scholar] [CrossRef]
  31. Özturk, N.; Ceylan, H.; Demir, Y. The hepatoprotective potential of tannic acid against doxorubicin-induced hepatotoxicity: Insights into its antioxidative, anti-inflammatory, and antiapoptotic mechanisms. J. Biochem. Mol. Toxicol. 2024, 38, e23798. [Google Scholar] [CrossRef]
  32. Wu, Z.; Shen, J.; Xu, Q.; Xiang, Q.; Chen, Y.; Lv, L.; Zheng, B.; Wang, Q.; Wang, S.; Li, L. Epigallocatechin-3-Gallate Improves Intestinal Gut Microbiota Homeostasis and Ameliorates Clostridioides difficile Infection. Nutrients 2022, 14, 3756. [Google Scholar] [CrossRef] [PubMed]
  33. Zhao, X.; Guo, J.; Wang, Y.; Yi, X. High-tannin food enhances spatial memory and scatter-hoarding in rodents via the microbiota-gut-brain axis. Microbiome 2024, 12, 140. [Google Scholar] [CrossRef] [PubMed]
  34. Tresserra-Rimbau, A.; Castro-Barquero, S.; Vitelli-Storelli, F.; Becerra-Tomas, N.; Vázquez-Ruiz, Z.; Díaz-López, A.; Corella, D.; Castañer, O.; Romaguera, D.; Vioque, J.; et al. Associations between Dietary Polyphenols and Type 2 Diabetes in a Cross-Sectional Analysis of the PREDIMED-Plus Trial: Role of Body Mass Index and Sex. Antioxidants 2019, 8, 537. [Google Scholar] [CrossRef] [PubMed]
  35. Liao, M.; Wang, X. Ameliorating effect of Chinese jujube polyphenol on blood glucose oxidative stress in type 2 diabetic rats. J. Diabetes Complicat. 2024, 38, 108804. [Google Scholar] [CrossRef] [PubMed]
  36. Choockong, C.; Itharat, A.; Pipatrattanaseree, W.; Ninlaor, T.; Piwngam, K.; Intharit, N.; Sukkhum, S.; Davies, N.M. The most commonly used spices in Thai traditional medicine: In vitro evaluation of anti-hyperglycemic, antioxidant, polyphenol content, and nitric oxide production inhibitory activities. Res. Pharm. Sci. 2024, 19, 13–28. [Google Scholar] [CrossRef]
  37. Lee, S.M.; Park, S.Y.; Kim, J.Y. Comparative evaluation of the antihyperglycemic effects of three extracts of sea mustard (Undaria pinnatifida): In vitro and in vivo studies. Food Res. Int. 2024, 190, 114623. [Google Scholar] [CrossRef]
  38. Liu, C.; Zeng, H.; Cui, W.; Ouyang, J.; Zhou, F.; Wen, S.; Fang, W.; Zhang, S.; Huang, J.; Liu, Z. Theaflavins mitigate diabetic symptoms in GK rats by modulating the INSR/PI3K-Akt/GSK-3 pathway and intestinal microbiota. Int. J. Biol. Macromol. 2024, 277, 134331. [Google Scholar] [CrossRef] [PubMed]
  39. Quetglas-Llabrés, M.M.; Monserrat-Mesquida, M.; Bouzas, C.; García, S.; Mateos, D.; Ugarriza, L.; Gómez, C.; Sureda, A.; Tur, J.A. Long-Term Impact of Nutritional Intervention with Increased Polyphenol Intake and Physical Activity Promotion on Oxidative and Inflammatory Profiles in Patients with Metabolic Syndrome. Nutrients 2024, 16, 2121. [Google Scholar] [CrossRef] [PubMed]
  40. Bohn, T. Dietary factors affecting polyphenol bioavailability. Nutr. Rev. 2014, 72, 429–452. [Google Scholar] [CrossRef] [PubMed]
  41. Castro-López, C.; Espinoza-González, C.; Ramos-González, R.; Boone-Villa, V.D.; Aguilar-González, M.A.; Martínez-Ávila, G.C.G.; Aguilar, C.N.; Ventura-Sobrevilla, J.M. Spray-drying encapsulation of microwave-assisted extracted polyphenols from Moringa oleifera: Influence of tragacanth, locust bean, and carboxymethyl-cellulose formulations. Food Res. Int. 2021, 144, 110291. [Google Scholar] [CrossRef]
  42. Costa, M.; Sezgin-Bayindir, Z.; Losada-Barreiro, S.; Paiva-Martins, F.; Saso, L.; Bravo-Díaz, C. Polyphenols as Antioxidants for Extending Food Shelf-Life and in the Prevention of Health Diseases: Encapsulation and Interfacial Phenomena. Biomedicines 2021, 9, 1909. [Google Scholar] [CrossRef]
  43. Gerloff, T. Impact of genetic polymorphisms in transmembrane carrier-systems on drug and xenobiotic distribution. Naunyn Schmiedebergs Arch. Pharmacol. 2004, 369, 69–77. [Google Scholar] [CrossRef]
  44. Lampe, J.W. Interindividual differences in response to plant-based diets: Implications for cancer risk. Am. J. Clin. Nutr. 2009, 89, 1553S–1557S. [Google Scholar] [CrossRef]
  45. Hidalgo-Liberona, N.; González-Domínguez, R.; Vegas, E.; Riso, P.; Del Bo’, C.; Bernardi, S.; Peron, G.; Guglielmetti, S.; Gargari, G.; Kroon, P.A. Increased intestinal permeability in older subjects impacts the beneficial effects of dietary polyphenols by modulating their bioavailability. J. Agric. Food Chem. 2020, 68, 12476–12484. [Google Scholar] [CrossRef]
  46. Margalef, M.; Pons, Z.; Iglesias-Carres, L.; Bravo, F.I.; Muguerza, B.; Arola-Arnal, A. Flavanol plasma bioavailability is affected by metabolic syndrome in rats. Food Chem. 2017, 231, 287–294. [Google Scholar] [CrossRef]
  47. Zhang, H.; Yu, D.; Sun, J.; Liu, X.; Jiang, L.; Guo, H.; Ren, F. Interaction of plant phenols with food macronutrients: Characterisation and nutritional–physiological consequences. Nutr. Res. Rev. 2014, 27, 1–15. [Google Scholar] [CrossRef]
  48. Iqbal, Y.; Cottrell, J.J.; Suleria, H.A.; Dunshea, F.R. Gut microbiota-polyphenol interactions in chicken: A review. Animals 2020, 10, 1391. [Google Scholar] [CrossRef]
  49. Li, D.; Yang, Y.; Yang, X.; Wang, Z.; Yao, X.; Guo, Y. Enhanced bioavailability and anti-hyperglycemic activity of young apple polyphenols by complexation with whey protein isolates. J. Food Sci. 2022, 87, 1257–1267. [Google Scholar] [CrossRef]
  50. Pasinetti, G.M.; Singh, R.; Westfall, S.; Herman, F.; Faith, J.; Ho, L. The role of the gut microbiota in the metabolism of polyphenols as characterized by gnotobiotic mice. J. Alzheimer’s Dis. 2018, 63, 409–421. [Google Scholar] [CrossRef]
  51. Wang, Q.; Xu, K.; Cai, X.; Wang, C.; Cao, Y.; Xiao, J. Rosmarinic acid restores colonic mucus secretion in colitis mice by regulating gut microbiota-derived metabolites and the activation of inflammasomes. J. Agric. Food Chem. 2023, 71, 4571–4585. [Google Scholar] [CrossRef] [PubMed]
  52. Zhou, N.; Gu, X.; Zhuang, T.; Xu, Y.; Yang, L.; Zhou, M. Gut microbiota: A pivotal hub for polyphenols as antidepressants. J. Agric. Food Chem. 2020, 68, 6007–6020. [Google Scholar] [CrossRef] [PubMed]
  53. Corrêa, T.A.F.; Rogero, M.M.; Hassimotto, N.M.A.; Lajolo, F.M. The Two-Way Polyphenols-Microbiota Interactions and Their Effects on Obesity and Related Metabolic Diseases. Front. Nutr. 2019, 6, 188. [Google Scholar] [CrossRef] [PubMed]
  54. Shivashankara, K.; Acharya, S. Bioavailability of dietary polyphenols and the cardiovascular diseases. Open Nutraceuticals J. 2010, 3, 227–241. [Google Scholar] [CrossRef]
  55. Li, M.; Zheng, Y.; Zhao, J.; Liu, M.; Shu, X.; Li, Q.; Wang, Y.; Zhou, Y. Polyphenol mechanisms against gastric cancer and their interactions with gut microbiota: A review. Curr. Oncol. 2022, 29, 5247–5261. [Google Scholar] [CrossRef]
  56. Lila, M.A.; Hoskin, R.T.; Grace, M.H.; Xiong, J.; Strauch, R.; Ferruzzi, M.; Iorizzo, M.; Kay, C. Boosting the bioaccessibility of dietary bioactives by delivery as protein–polyphenol aggregate particles. J. Agric. Food Chem. 2022, 70, 13017–13026. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, L.; Gnanaraj, C.; Arulselvan, P.; El-Seedi, H.; Teng, H. A review on advanced microencapsulation technology to enhance bioavailability of phenolic compounds: Based on its activity in the treatment of Type 2 Diabetes. Trends Food Sci. Technol. 2019, 85, 149–162. [Google Scholar] [CrossRef]
  58. Frakolaki, G.; Giannou, V.; Kekos, D.; Tzia, C. A review of the microencapsulation techniques for the incorporation of probiotic bacteria in functional foods. Crit. Rev. Food Sci. Nutr. 2021, 61, 1515–1536. [Google Scholar] [CrossRef]
  59. Arenas-Jal, M.; Suñé-Negre, J.M.; García-Montoya, E. An overview of microencapsulation in the food industry: Opportunities, challenges, and innovations. Eur. Food Res. Technol. 2020, 246, 1371–1382. [Google Scholar] [CrossRef]
  60. Mohammadalinejhad, S.; Kurek, M.A. Microencapsulation of anthocyanins-critical review of techniques and wall materials. Appl. Sci. 2021, 11, 3936. [Google Scholar] [CrossRef]
  61. Peanparkdee, M.; Iwamoto, S.; Yamauchi, R. Microencapsulation: A review of applications in the food and pharmaceutical industries. Rev. Agric. Sci. 2016, 4, 56–65. [Google Scholar] [CrossRef]
  62. Mehta, N.; Kumar, P.; Verma, A.K.; Umaraw, P.; Kumar, Y.; Malav, O.P.; Sazili, A.Q.; Domínguez, R.; Lorenzo, J.M. Microencapsulation as a Noble Technique for the Application of Bioactive Compounds in the Food Industry: A Comprehensive Review. Appl. Sci. 2022, 12, 1424. [Google Scholar] [CrossRef]
  63. Leyva-Jiménez, F.J.; Lozano-Sánchez, J.; de la Luz Cádiz-Gurrea, M.; Fernández-Ochoa, Á.; Arráez-Román, D.; Segura-Carretero, A. Spray-drying microencapsulation of bioactive compounds from lemon verbena green extract. Foods 2020, 9, 1547. [Google Scholar] [CrossRef] [PubMed]
  64. Santos, S.S.d.; Rodrigues, L.M.; da Costa, S.C.; Bergamasco, R.d.C.; Madrona, G.S. Microencapsulation of bioactive compounds from blackberry pomace (Rubus fruticosus) by spray drying technique. Int. J. Food Eng. 2017, 13, 20170047. [Google Scholar] [CrossRef]
  65. Navarro-Flores, M.J.; Ventura-Canseco, L.M.C.; Meza-Gordillo, R.; Ayora-Talavera, T.R.; Abud-Archila, M. Spray drying encapsulation of a native plant extract rich in phenolic compounds with combinations of maltodextrin and non-conventional wall materials. J. Food Sci. Technol. 2020, 57, 4111–4122. [Google Scholar] [CrossRef]
  66. Ribeiro, A.M.; Estevinho, B.N.; Rocha, F. Spray Drying Encapsulation of Elderberry Extract and Evaluating the Release and Stability of Phenolic Compounds in Encapsulated Powders. Food Bioprocess Technol. 2019, 12, 1381–1394. [Google Scholar] [CrossRef]
  67. Ferreira, L.M.D.M.C.; Pereira, R.R.; Carvalho-Guimarães, F.B.D.; Remígio, M.S.D.N.; Barbosa, W.L.R.; Ribeiro-Costa, R.M.; Silva-Júnior, J.O.C. Microencapsulation by Spray Drying and Antioxidant Activity of Phenolic Compounds from Tucuma Coproduct (Astrocaryum vulgare Mart.) Almonds. Polymers 2022, 14, 2905. [Google Scholar] [CrossRef] [PubMed]
  68. Da Rosa, C.G.; Borges, C.D.; Zambiazi, R.C.; Rutz, J.K.; da Luz, S.R.; Krumreich, F.D.; Benvenutti, E.V.; Nunes, M.R. Encapsulation of the phenolic compounds of the blackberry (Rubus fruticosus). LWT 2014, 58, 527–533. [Google Scholar] [CrossRef]
  69. Wilkowska, A.; Ambroziak, W.; Czyzowska, A.; Adamiec, J. Effect of Microencapsulation by Spray-Drying and Freeze-Drying Technique on the Antioxidant Properties of Blueberry (Vaccinium myrtillus) Juice Polyphenolic Compounds. Pol. J. Food Nutr. Sci. 2016, 66, 11–16. [Google Scholar] [CrossRef]
  70. Stach, M.; Kolniak-Ostek, J. The Influence of the Use of Different Polysaccharide Coatings on the Stability of Phenolic Compounds and Antioxidant Capacity of Chokeberry Hydrogel Microcapsules Obtained by Indirect Extrusion. Foods 2023, 12, 515. [Google Scholar] [CrossRef]
  71. Bakry, A.M.; Abbas, S.; Ali, B.; Majeed, H.; Abouelwafa, M.Y.; Mousa, A.; Liang, L. Microencapsulation of Oils: A Comprehensive Review of Benefits, Techniques, and Applications. Compr. Rev. Food Sci. Food Saf. 2016, 15, 143–182. [Google Scholar] [CrossRef]
  72. Gibis, M.; Ruedt, C.; Weiss, J. In vitro release of grape-seed polyphenols encapsulated from uncoated and chitosan-coated liposomes. Int. Food Res. 2016, 88, 105–113. [Google Scholar] [CrossRef]
  73. Has, C.; Sunthar, P. A comprehensive review on recent preparation techniques of liposomes. J. Liposome Res. 2020, 30, 336–365. [Google Scholar] [CrossRef] [PubMed]
  74. Bozzuto, G.; Molinari, A. Liposomes as nanomedical devices. Int. J. Nanomed. 2015, 10, 975–999. [Google Scholar] [CrossRef] [PubMed]
  75. Choudhury, N.; Meghwal, M.; Das, K. Microencapsulation: An overview on concepts, methods, properties and applications in foods. Food Front. 2021, 2, 426–442. [Google Scholar] [CrossRef]
  76. Ozkan, G.; Franco, P.; De Marco, I.; Xiao, J.; Capanoglu, E. A review of microencapsulation methods for food antioxidants: Principles, advantages, drawbacks and applications. Food Chem. 2019, 272, 494–506. [Google Scholar] [CrossRef]
  77. de Souza, V.B.; Thomazini, M.; Echalar Barrientos, M.A.; Nalin, C.M.; Ferro-Furtado, R.; Genovese, M.I.; Favaro-Trindade, C.S. Functional properties and encapsulation of a proanthocyanidin-rich cinnamon extract (Cinnamomum zeylanicum) by complex coacervation using gelatin and different polysaccharides. Food Hydrocoll. 2018, 77, 297–306. [Google Scholar] [CrossRef]
  78. Soliman, T.N.; Mohammed, D.M.; El-Messery, T.M.; Elaaser, M.; Zaky, A.A.; Eun, J.B.; Shim, J.H.; El-Said, M.M. Microencapsulation of Plant Phenolic Extracts Using Complex Coacervation Incorporated in Ultrafiltered Cheese Against AlCl3-Induced Neuroinflammation in Rats. Front. Nutr. 2022, 9, 929977. [Google Scholar] [CrossRef]
  79. Sarabandi, K.; Mahoonak, A.S.; Akbari, M. Physicochemical properties and antioxidant stability of microencapsulated marjoram extract prepared by co-crystallization method. J. Food Process Eng. 2019, 42, e12949. [Google Scholar] [CrossRef]
  80. Ali, N.A.; Dash, K.K.; Pandey, V.K.; Tripathi, A.; Mukarram, S.A.; Harsányi, E.; Kovács, B. Extraction and Encapsulation of Phytocompounds of Poniol Fruit via Co-Crystallization: Physicochemical Properties and Characterization. Molecules 2023, 28, 4764. [Google Scholar] [CrossRef]
  81. Cutrim, C.S.; Alvim, I.D.; Cortez, M.A.S. Microencapsulation of green tea polyphenols by ionic gelation and spray chilling methods. J. Food Technol. 2019, 56, 3561–3570. [Google Scholar] [CrossRef] [PubMed]
  82. Kurozawa, L.E.; Hubinger, M.D. Hydrophilic food compounds encapsulation by ionic gelation. Curr. Opin. Food Sci. 2017, 15, 50–55. [Google Scholar] [CrossRef]
  83. de Moura, S.C.S.R.; Berling, C.L.; Germer, S.P.M.; Alvim, I.D.; Hubinger, M.D. Encapsulating anthocyanins from Hibiscus sabdariffa L. calyces by ionic gelation: Pigment stability during storage of microparticles. Food Chem. 2018, 241, 317–327. [Google Scholar] [CrossRef]
  84. da Silva Júnior, M.E.; Araújo, M.V.R.L.; Martins, A.C.S.; dos Santos Lima, M.; da Silva, F.L.H.; Converti, A.; Maciel, M.I.S. Microencapsulation by spray-drying and freeze-drying of extract of phenolic compounds obtained from ciriguela peel. Sci. Rep. 2023, 13, 15222. [Google Scholar] [CrossRef] [PubMed]
  85. Thakur, N.S.; Thakur, A. Microencapsulation of wild pomegranate flavedo phenolics by lyophilization: Effect of maltodextrin concentration, structural morphology, functional properties, elemental composition and ingredient for development of functional beverage. LWT 2020, 133, 110077. [Google Scholar] [CrossRef]
  86. Vallejo-Castillo, V.; Rodríguez-Stouvenel, A.; Martínez, R.; Bernal, C. Development of alginate-pectin microcapsules by the extrusion for encapsulation and controlled release of polyphenols from papaya (Carica papaya L.). J. Food Biochem. 2020, 44, e13331. [Google Scholar] [CrossRef]
  87. Bannikova, A.; Zyainitdinov, D.; Evteev, A.; Drevko, Y.; Evdokimov, I. Microencapsulation of polyphenols and xylooligosaccharides from oat bran in whey protein-maltodextrin complex coacervates: In-vitro evaluation and controlled release. Bioact. Carbohydr. Diet. Fibre 2020, 23, 100236. [Google Scholar] [CrossRef]
  88. Istenič, K.; Cerc Korošec, R.; Poklar Ulrih, N. Encapsulation of (-)-epigallocatechin gallate into liposomes and into alginate or chitosan microparticles reinforced with liposomes. J. Sci. Food Agric. 2016, 96, 4623–4632. [Google Scholar] [CrossRef]
  89. Nik, A.B.; Vazifedoost, M.; Didar, Z.; Hajirostamloo, B. The antioxidant and physicochemical properties of microencapsulated bioactive compounds in Securigera securidaca (L.) seed extract by co-crystallization. Food Qual. Saf. 2019, 3, 243–250. [Google Scholar] [CrossRef]
  90. Chezanoglou, E.; Goula, A.M. Properties and Stability of Encapsulated Pomegranate Peel Extract Prepared by Co-Crystallization. Appl. Sci. 2023, 13, 8680. [Google Scholar] [CrossRef]
  91. Anandharamakrishnan, C. Techniques for Nanoencapsulation of Food Ingredients; Springer: Berlin/Heidelberg, Germany, 2014; Volume 8. [Google Scholar]
  92. Contreras-Angulo, L.A.; Gutiérrez-Grijalva, E.P.; Cabanillas-Bojórquez, L.A.; Jiménez-Ortega, L.A.; Heredia, J.B. Chapter 7—Nanoformulations applied to the delivery of alkaloids. In Phytochemical Nanodelivery Systems as Potential Biopharmaceuticals; Heredia, J.B., Gutiérrez-Grijalva, E.P., Licea-Claverie, A., Gutierrez-Uribe, J.A., Patra, J.K., Eds.; Elsevier: Amsterdam, The Netherlands, 2023; pp. 257–281. [Google Scholar]
  93. Garcia-Carrasco, M.; Parra-Aguilar, I.F.; Gutiérrez-Grijalva, E.P.; Licea-Claverie, A.; Basilio Heredia, J. Chapter 18—Nano-formulations in drug delivery. In Food, Medical, and Environmental Applications of Nanomaterials; Pal, K., Sarkar, A., Sarkar, P., Bandara, N., Jegatheesan, V., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 473–491. [Google Scholar]
  94. Pedrozo, R.C.; Antônio, E.; Khalil, N.M.; Mainardes, R.M. Bovine serum albumin-based nanoparticles containing the flavonoid rutin produced by nano spray drying. Braz. J. Pharm. 2020, 56, e17692. [Google Scholar] [CrossRef]
  95. Rambaran, T.F. Nanopolyphenols: A review of their encapsulation and anti-diabetic effects. SN Appl. Sci. 2020, 2, 1335. [Google Scholar] [CrossRef]
  96. Rehman, A.; Ahmad, T.; Aadil, R.M.; Spotti, M.J.; Bakry, A.M.; Khan, I.M.; Zhao, L.; Riaz, T.; Tong, Q. Pectin polymers as wall materials for the nano-encapsulation of bioactive compounds. Trends Food Sci. Technol. 2019, 90, 35–46. [Google Scholar] [CrossRef]
  97. Ribeiro, E.F.; de Barros-Alexandrino, T.T.; Assis, O.B.G.; Junior, A.C.; Quiles, A.; Hernando, I.; Nicoletti, V.R. Chitosan and crosslinked chitosan nanoparticles: Synthesis, characterization and their role as Pickering emulsifiers. Carbohydr. Polym. 2020, 250, 116878. [Google Scholar] [CrossRef]
  98. Jiang, T.; Jin, K.; Liu, X.; Pang, Z. 8—Nanoparticles for tumor targeting. In Biopolymer-Based Composites; Jana, S., Maiti, S., Jana, S., Eds.; Woodhead Publishing: Cambridge, UK, 2017; pp. 221–267. [Google Scholar]
  99. Costa-Almeida, R.; Soares, R.; Costa, R. Polyphenol-Based Nanoparticles as Multifaceted Diabetes Modulators. In Functional Bionanomaterials: From Biomolecules to Nanoparticles; Thangadurai, D., Sangeetha, J., Prasad, R., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 251–270. [Google Scholar]
  100. Rai, V.K.; Gupta, G.D.; Pottoo, F.H.; Barkat, M.A. Potential of Nano-Structured Drug Delivery System for Phytomedicine Delivery. In Nanophytomedicine: Concept to Clinic; Beg, S., Barkat, M.A., Ahmad, F.J., Eds.; Springer: Singapore, 2020; pp. 89–111. [Google Scholar]
  101. Wu, M.; Jin, J.; Jin, P.; Xu, Y.; Yin, J.; Qin, D.; Wang, K.; Du, Q. Epigallocatechin gallate-β-lactoglobulin nanoparticles improve the antitumor activity of EGCG for inducing cancer cell apoptosis. J. Funct. Foods 2017, 39, 257–263. [Google Scholar] [CrossRef]
  102. Palacio, J.; Monsalve, Y.; Ramírez-Rodríguez, F.; López, B. Study of encapsulation of polyphenols on succinyl-chitosan nanoparticles. J. Drug Deliv. Sci. Technol. 2020, 57, 101610. [Google Scholar] [CrossRef]
  103. Gadkari, P.V.; Balaraman, M. Extraction of catechins from decaffeinated green tea for development of nanoemulsion using palm oil and sunflower oil based lipid carrier systems. J. Food Eng. 2015, 147, 14–23. [Google Scholar] [CrossRef]
  104. Harwansh, R.K.; Mukherjee, P.K.; Kar, A.; Bahadur, S.; Al-Dhabi, N.A.; Duraipandiyan, V. Enhancement of photoprotection potential of catechin loaded nanoemulsion gel against UVA induced oxidative stress. J. Photochem. Photobiol. B Biol. 2016, 160, 318–329. [Google Scholar] [CrossRef]
  105. Antônio, E.; Khalil, N.M.; Mainardes, R.M. Bovine Serum Albumin Nanoparticles Containing Quercetin: Characterization and Antioxidant Activity. J. Nanosci. Nanotechnol. 2016, 16, 1346–1353. [Google Scholar] [CrossRef]
  106. Javani, R.; Hashemi, F.S.; Ghanbarzadeh, B.; Hamishehkar, H. Quercetin-loaded niosomal nanoparticles prepared by the thin-layer hydration method: Formulation development, colloidal stability, and structural properties. LWT 2021, 141, 110865. [Google Scholar] [CrossRef]
  107. Romeo, A.; Musumeci, T.; Carbone, C.; Bonaccorso, A.; Corvo, S.; Lupo, G.; Anfuso, C.D.; Puglisi, G.; Pignatello, R. Ferulic Acid-Loaded Polymeric Nanoparticles for Potential Ocular Delivery. Pharmaceutics 2021, 13, 687. [Google Scholar] [CrossRef] [PubMed]
  108. Nallamuthu, I.; Devi, A.; Khanum, F. Chlorogenic acid loaded chitosan nanoparticles with sustained release property, retained antioxidant activity and enhanced bioavailability. Asian J. Pharm. Anal. 2015, 10, 203–211. [Google Scholar] [CrossRef]
  109. Neelakandan, M.; Manoharan, S.; Muralinaidu, R.; Thara, J.M. Tumor preventive and antioxidant efficacy of chlorogenic acid–loaded chitosan nanoparticles in experimental skin carcinogenesis. Naunyn Schmiedebergs Arch. Pharmacol. 2023, 396, 533–546. [Google Scholar] [CrossRef] [PubMed]
  110. Mariadoss, A.V.A.; Vinayagam, R.; Senthilkumar, V.; Paulpandi, M.; Murugan, K.; Xu, B.; Gothandam, K.M.; Kotakadi, V.S.; David, E. Phloretin loaded chitosan nanoparticles augments the pH-dependent mitochondrial-mediated intrinsic apoptosis in human oral cancer cells. Int. J. Biol. Macromol. 2019, 130, 997–1008. [Google Scholar] [CrossRef]
  111. Kuai, L.; Liu, F.; Ma, Y.; Goff, H.D.; Zhong, F. Regulation of nano-encapsulated tea polyphenol release from gelatin films with different Bloom values. Food Hydrocoll. 2020, 108, 106045. [Google Scholar] [CrossRef]
  112. Gaber Ahmed, G.H.; Fernández-González, A.; Díaz García, M.E. Nano-encapsulation of grape and apple pomace phenolic extract in chitosan and soy protein via nanoemulsification. Food Hydrocoll. 2020, 108, 105806. [Google Scholar] [CrossRef]
  113. Soleimanifar, M.; Jafari, S.M.; Assadpour, E. Encapsulation of olive leaf phenolics within electrosprayed whey protein nanoparticles; production and characterization. Food Hydrocoll. 2020, 101, 105572. [Google Scholar] [CrossRef]
  114. Oskoueian, E.; Karimi, E.; Noura, R.; Ebrahimi, M.; Shafaei, N.; Karimi, E. Nanoliposomes encapsulation of enriched phenolic fraction from pistachio hulls and its antioxidant, anti-inflammatory, and anti-melanogenic activities. J. Microencapsul. 2020, 37, 1–13. [Google Scholar] [CrossRef]
  115. Gharehbeglou, P.; Jafari, S.M.; Homayouni, A.; Hamishekar, H.; Mirzaei, H. Fabrication of double W1/O/W2 nano-emulsions loaded with oleuropein in the internal phase (W1) and evaluation of their release rate. Food Hydrocoll. 2019, 89, 44–55. [Google Scholar] [CrossRef]
  116. Grama, C.N.; Suryanarayana, P.; Patil, M.A.; Raghu, G.; Balakrishna, N.; Kumar, M.N.V.R.; Reddy, G.B. Efficacy of Biodegradable Curcumin Nanoparticles in Delaying Cataract in Diabetic Rat Model. PLoS ONE 2013, 8, e78217. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, J.; Li, S.; An, F.-F.; Liu, J.; Jin, S.; Zhang, J.-C.; Wang, P.C.; Zhang, X.; Lee, C.-S.; Liang, X.-J. Self-carried curcumin nanoparticles for in vitro and in vivo cancer therapy with real-time monitoring of drug release. Nanoscale 2015, 7, 13503–13510. [Google Scholar] [CrossRef]
  118. Chockalingam, S.; Thada, R.; Dhandapani, R.K.; Panchamoorthy, R. Biogenesis, characterization, and the effect of vicenin-gold nanoparticles on glucose utilization in 3T3-L1 adipocytes: A bioinformatic approach to illuminate its interaction with PTP 1B and AMPK. Biotechnol. Prog. 2015, 31, 1096–1106. [Google Scholar] [CrossRef] [PubMed]
  119. Das, S.; Roy, P.; Pal, R.; Auddy, R.G.; Chakraborti, A.S.; Mukherjee, A. Engineered silybin nanoparticles educe efficient control in experimental diabetes. PLoS ONE 2014, 9, e101818. [Google Scholar] [CrossRef]
  120. Salah, M.; Mansour, M.; Zogona, D.; Xu, X. Nanoencapsulation of anthocyanins-loaded β-lactoglobulin nanoparticles: Characterization, stability, and bioavailability in vitro. Food Res. Int. 2020, 137, 109635. [Google Scholar] [CrossRef] [PubMed]
  121. Sun, J.; Chen, J.; Mei, Z.; Luo, Z.; Ding, L.; Jiang, X.; Bai, W. Synthesis, structural characterization, and evaluation of cyanidin-3-O-glucoside-loaded chitosan nanoparticles. Food Chem. 2020, 330, 127239. [Google Scholar] [CrossRef]
  122. Liang, T.; Guan, R.; Shen, H.; Xia, Q.; Liu, M. Optimization of Conditions for Cyanidin-3-OGlucoside (C3G) Nanoliposome Production by Response Surface Methodology and Cellular Uptake Studies in Caco-2 Cells. Molecules 2017, 22, 457. [Google Scholar] [CrossRef]
  123. Verônica Cardoso de Souza, B.; de Morais Sousa, M.; Augusto Gasparotto Sattler, J.; Cristina Sousa Gramoza Vilarinho Santana, A.; Bruno Fonseca de Carvalho, R.; de Sousa Lima Neto, J.; de Matos Borges, F.; Angelica Neri Numa, I.; Braga Ribeiro, A.; César Cunha Nunes, L. Nanoencapsulation and bioaccessibility of polyphenols of aqueous extracts from Bauhinia forficata link. Food Chem. 2022, 5, 100144. [Google Scholar] [CrossRef]
  124. Kerbab, K.; Sansone, F.; Zaiter, L.; Esposito, T.; Celano, R.; Franceschelli, S.; Pecoraro, M.; Benayache, F.; Rastrelli, L.; Picerno, P.; et al. Halimium halimifolium: From the Chemical and Functional Characterization to a Nutraceutical Ingredient Design. Planta Med. 2019, 85, 1024–1033. [Google Scholar] [CrossRef]
  125. Zorzenon, M.R.T.; Formigoni, M.; da Silva, S.B.; Hodas, F.; Piovan, S.; Ciotta, S.R.; Jansen, C.A.; Dacome, A.S.; Pilau, E.J.; Mareze-Costa, C.E.; et al. Spray drying encapsulation of stevia extract with maltodextrin and evaluation of the physicochemical and functional properties of produced powders. J. Food Sci. 2020, 85, 3590–3600. [Google Scholar] [CrossRef] [PubMed]
  126. De Silva, N.D.; Attanayake, A.P.; Karunaratne, D.N.; Arawwawala, L.; Pamunuwa, G.K. Bael (Aegle marmelos L. Correa) fruit extracts encapsulated alginate nanoparticles as a potential dietary supplement with improved bioactivities. J. Food Sci. 2023, 88, 4942–4961. [Google Scholar] [CrossRef] [PubMed]
  127. Chauhan, P.; Tamrakar, A.K.; Mahajan, S.; Prasad, G. Chitosan encapsulated nanocurcumin induces GLUT-4 translocation and exhibits enhanced anti-hyperglycemic function. Life Sci. 2018, 213, 226–235. [Google Scholar] [CrossRef]
  128. Pandey, S.; Dvorakova, C.M. Future Perspective of Diabetic Animal Models. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 25–38. [Google Scholar] [CrossRef] [PubMed]
  129. Goyal, S.N.; Reddy, N.M.; Patil, K.R.; Nakhate, K.T.; Ojha, S.; Patil, C.R.; Agrawal, Y.O. Challenges and issues with streptozotocin-induced diabetes—A clinically relevant animal model to understand the diabetes pathogenesis and evaluate therapeutics. Chem. Biol. Interact. 2016, 244, 49–63. [Google Scholar] [CrossRef]
  130. Panwar, R.; Raghuwanshi, N.; Srivastava, A.K.; Sharma, A.K.; Pruthi, V. In-vivo sustained release of nanoencapsulated ferulic acid and its impact in induced diabetes. Mater. Sci. Eng. C 2018, 92, 381–392. [Google Scholar] [CrossRef]
  131. Yao, Y.; Luong, T.N.; Lepik, M.; Aftab, N.; Fong, V.H.; Vieira, A. Synergism of Antioxidant Phytochemicals: Comparisons Among Purified Polyphenols and Dietary-Plant Extracts; International Society for Horticultural Science: Orlando, FL, USA, 2012; pp. 121–127. [Google Scholar]
  132. Revathi, G.; Elavarasi, S.; Saravanan, K.; Ashokkumar, M.; Egbuna, C. Greater efficiency of polyherbal drug encapsulated biosynthesized chitosan nano-biopolymer on diabetes and its complications. Int. J. Biol. Macromol. 2023, 240, 124445. [Google Scholar] [CrossRef]
  133. El-Hussien, D.; El-Zaafarany, G.M.; Nasr, M.; Sammour, O. Chrysin nanocapsules with dual anti-glycemic and anti-hyperlipidemic effects: Chemometric optimization, physicochemical characterization and pharmacodynamic assessment. Int. J. Pharm. 2021, 592, 120044. [Google Scholar] [CrossRef]
  134. Sudirman, S.; Lai, C.-S.; Yan, Y.-L.; Yeh, H.-I.; Kong, Z.-L. Histological evidence of chitosan-encapsulated curcumin suppresses heart and kidney damages on streptozotocin-induced type-1 diabetes in mice model. Sci. Rep. 2019, 9, 15233. [Google Scholar] [CrossRef]
  135. El-Shahawy, A.A.G.; Abdel-Moneim, A.; Ebeid, A.S.M.; Eldin, Z.E.; Zanaty, M.I. A novel layered double hydroxide-hesperidin nanoparticles exert antidiabetic, antioxidant and anti-inflammatory effects in rats with diabetes. Mol. Biol. Rep. 2021, 48, 5217–5232. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, Q.; Wei, C.; Weng, W.; Bao, R.; Adu-Frimpong, M.; Toreniyazov, E.; Ji, H.; Xu, X.-M.; Yu, J. Enhancement of oral bioavailability and hypoglycemic activity of liquiritin-loaded precursor liposome. Int. J. Pharm. 2021, 592, 120036. [Google Scholar] [CrossRef]
  137. Foudah, A.I.; Ayman Salkini, M.; Alqarni, M.H.; Alam, A. Preparation and evaluation of antidiabetic activity of mangiferin-loaded solid lipid nanoparticles. Saudi J. Biol. Sci. 2024, 31, 103946. [Google Scholar] [CrossRef]
  138. Wang, Y.; Karmakar, T.; Ghosh, N.; Basak, S.; Gopal Sahoo, N. Targeting mangiferin loaded N-succinyl chitosan-alginate grafted nanoparticles against atherosclerosis—A case study against diabetes mediated hyperlipidemia in rat. Food Chem. 2022, 370, 131376. [Google Scholar] [CrossRef]
  139. Upadhyay, M.; Hosur, R.V.; Jha, A.; Bharti, K.; Mali, P.S.; Jha, A.K.; Mishra, B.; Kumar, A. Myricetin encapsulated chitosan nanoformulation for management of type 2 diabetes: Preparation, optimization, characterization and in vivo activity. Biomater. Adv. 2023, 153, 213542. [Google Scholar] [CrossRef]
  140. Joshi, R.; Laddha, A.P.; Kulkarni, Y.A.; Wairkar, S. Improved performance of naringenin herbosomes over naringenin in streptozotocin-induced diabetic rats: In vitro and in vivo evaluation. Asian Pac. J. Trop. Biomed. 2021, 11, 385–393. [Google Scholar] [CrossRef]
  141. Maity, S.; Chakraborti, A.S. Formulation, physico-chemical characterization and antidiabetic potential of naringenin-loaded poly D, L lactide-co-glycolide (N-PLGA) nanoparticles. Eur. Polym. J. 2020, 134, 109818. [Google Scholar] [CrossRef]
  142. Ahmad, M.Z.; Pathak, K.; Das, R.J.; Saikia, R.; Sarma, H.; Gogoi, N.; Gogoi, U.; Das, A.; Alasiri, A.S.; Abdel-Wahab, B.A.; et al. Design and optimization of quercetin-loaded polymeric Eudragit L-100 nanoparticles for anti-diabetic activity with improved oral delivery: In-vitro and in-vivo evaluation. J. Inorg. Organomet. Polym. Mater. 2023, 33, 2411–2428. [Google Scholar] [CrossRef]
  143. Chitkara, D.; Nikalaje, S.K.; Mittal, A.; Chand, M.; Kumar, N. Development of quercetin nanoformulation and in vivo evaluation using streptozotocin induced diabetic rat model. Drug Deliv. Transl. Res. 2012, 2, 112–123. [Google Scholar] [CrossRef]
  144. Wang, S.; Du, S.; Wang, W.; Zhang, F. Therapeutic investigation of quercetin nanomedicine in a zebrafish model of diabetic retinopathy. Biomed. Pharmacother. 2020, 130, 110573. [Google Scholar] [CrossRef]
  145. Du, S.; Lv, Y.; Li, N.; Huang, X.; Liu, X.; Li, H.; Wang, C.; Jia, Y.-F. Biological investigations on therapeutic effect of chitosan encapsulated nano resveratrol against gestational diabetes mellitus rats induced by streptozotocin. J. Drug Deliv. 2020, 27, 953–963. [Google Scholar] [CrossRef] [PubMed]
  146. Benyelles, M.; Merzouk, H.; Merzouk, A.Z.; Imessaoudene, A.; Medjdoub, A.; Mebarki, A. Valorization of Encapsulated Coffee Parchment Extracts as Metabolic Control for High Fructose Diet-Induced Obesity, Using Wistar Rat as Animal Model. Waste Biomass Valori 2024, 15, 265–281. [Google Scholar] [CrossRef]
  147. Colorado, D.; Fernandez, M.; Orozco, J.; Lopera, Y.; Muñoz, D.L.; Acín, S.; Balcazar, N. Metabolic activity of anthocyanin extracts loaded into non-ionic niosomes in diet-induced obese mice. Pharm. Res. 2020, 37, 152. [Google Scholar] [CrossRef]
  148. de Paulo Farias, D.; de Araújo, F.F.; Neri-Numa, I.A.; Pastore, G.M. Antidiabetic potential of dietary polyphenols: A mechanistic review. Int. Food Res. 2021, 145, 110383. [Google Scholar] [CrossRef]
  149. Huang, Y.-C.; Chen, B.-H. A comparative study on improving streptozotocin-induced type 2 diabetes in rats by hydrosol, extract and nanoemulsion prepared from cinnamon leaves. Antioxidants 2023, 12, 29. [Google Scholar] [CrossRef]
  150. Wickramasinghe, A.S.D.; Attanayake, A.P.; Kalansuriya, P. Gelatine nanoparticles encapsulating three edible plant extracts as potential nanonutraceutical agents against type 2 diabetes mellitus. J. Microencapsul. 2024, 41, 94–111. [Google Scholar] [CrossRef]
  151. Ammar, N.M.; Hassan, H.A.; Mohammed, M.A.; Serag, A.; Abd El-Alim, S.H.; Elmotasem, H.; El Raey, M.; El Gendy, A.N.; Sobeh, M.; Abdel-Hamid, A.-H.Z. Metabolomic profiling to reveal the therapeutic potency of Posidonia oceanica nanoparticles in diabetic rats. RSC Adv. 2021, 11, 8398–8410. [Google Scholar] [CrossRef] [PubMed]
  152. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  153. Protox3.0. Available online: https://tox-new.charite.de/protox3/index.php?site=home (accessed on 15 October 2025).
  154. Korobkova, E.A. Effect of Natural Polyphenols on CYP Metabolism: Implications for Diseases. Chem. Res. Toxicol. 2015, 28, 1359–1390. [Google Scholar] [CrossRef] [PubMed]
  155. Basheer, L.; Kerem, Z. Interactions between CYP3A4 and Dietary Polyphenols. Oxidative Med. Cell. Longev. 2015, 2015, 854015. [Google Scholar] [CrossRef]
  156. Sprouse, A.A.; van Breemen, R.B. Pharmacokinetic Interactions between Drugs and Botanical Dietary Supplements. Drug Metab. Dispos. 2016, 44, 162–171. [Google Scholar] [CrossRef] [PubMed]
  157. Sang, R.; Jiang, W.; Zhang, C.; Yin, R.; Ouyang, Z.; Wei, Y. Effect of food components on cytochrome P450 expression and activity. Hum. Nutr. Metab. 2025, 40, 200304. [Google Scholar] [CrossRef]
  158. Stromsnes, K.; Lagzdina, R.; Olaso-Gonzalez, G.; Gimeno-Mallench, L.; Gambini, J. Pharmacological Properties of Polyphenols: Bioavailability, Mechanisms of Action, and Biological Effects in In Vitro Studies, Animal Models, and Humans. Biomedicines 2021, 9, 1074. [Google Scholar] [CrossRef] [PubMed]
  159. Kim, Y.; Cho, A.Y.; Kim, H.C.; Ryu, D.; Jo, S.A.; Jung, Y.-S. Effects of Natural Polyphenols on Oxidative Stress-Mediated Blood-Brain Barrier Dysfunction. Antioxidants 2022, 11, 197. [Google Scholar] [CrossRef]
  160. Shimazu, R.; Anada, M.; Miyaguchi, A.; Nomi, Y.; Matsumoto, H. Evaluation of Blood–Brain Barrier Permeability of Polyphenols, Anthocyanins, and Their Metabolites. J. Agric. Food Chem. 2021, 69, 11676–11686. [Google Scholar] [CrossRef]
  161. Singh, K.; Tarapcsák, S.; Gyöngy, Z.; Ritter, Z.; Batta, G.; Bosire, R.; Remenyik, J.; Goda, K. Effects of Polyphenols on P-Glycoprotein (ABCB1) Activity. Pharmaceutics 2021, 13, 2062. [Google Scholar] [CrossRef]
  162. Nabekura, T.; Kawasaki, T.; Furuta, M.; Kaneko, T.; Uwai, Y. Effects of Natural Polyphenols on the Expression of Drug Efflux Transporter P-Glycoprotein in Human Intestinal Cells. ACS Omega 2018, 3, 1621–1626. [Google Scholar] [CrossRef]
Figure 1. Classification of diabetes mellitus according to the ADA.
Figure 1. Classification of diabetes mellitus according to the ADA.
Scipharm 93 00055 g001
Figure 2. Encapsulation techniques for polyphenols with antidiabetic potential to enhance their bioavailability and bioactivity.
Figure 2. Encapsulation techniques for polyphenols with antidiabetic potential to enhance their bioavailability and bioactivity.
Scipharm 93 00055 g002
Figure 3. 3D chemical structures of polyphenols with antidiabetic properties.
Figure 3. 3D chemical structures of polyphenols with antidiabetic properties.
Scipharm 93 00055 g003
Table 2. Design of nanoformulations loaded with polyphenol compounds.
Table 2. Design of nanoformulations loaded with polyphenol compounds.
Polyphenols LoadedNanosystemEncapsulating MaterialTechnique NanoencapsulationEffect *Ref.
Epigallocatechin-3-gallateNanoparticleBovine β-lactoglobulinCo-assembled with preheated↑ Inhibition of proliferation of human malignant melanoma cells and esophageal carcinoma cells[101]
Epigallocatechin-3-gallateNanoparticlesSuccinyl-chitosan (modified chitosan), pentasodium tripolyphosphateIonic crosslinking65% nanoencapsulation efficiency[102]
Propyl gallateNanoparticlesSuccinyl-chitosan (modified chitosan), pentasodium tripolyphosphateIonic crosslinking27% nanoencapsulation efficiency[102]
Gallic acidNanoparticlesSuccinyl-chitosan (modified chitosan), pentasodium tripolyphosphateIonic crosslinking88% nanoencapsulation efficiency[102]
CatechinNanoemulsionPalm oil and sunflower oilNanoemulsificationPhysically and chemically stable emulsions, with no significant variation in droplet diameter, conductivity, refractive index and pH[103]
CatechinNanoemulsionEthyl oleate, the surfactant spans 80, and the cosurfactant trancutol CGNanoemulsification↑ skin permeability
↑ %relative bioavailability
↑ SOD, GPX and CAT
↓ TBRAS
[104]
RutinNanoparticleBovine serum albuminsNanospray drying32% nanoencapsulation efficiency
↑ antioxidant effect for ABTS radical
[94]
QuercetinNanoparticleBovine serum albumins and glutaraldehyde as a crosslinking agentDesolvation85% nanoencapsulation efficiency
↑ antioxidant effect for ABTS radical
[105]
QuercetinNanoniosomeSurfactants (span 60 and 80, tween 60 and 80), polymers (polyethylene glycol, propylene glycol, glycerol, and cholesterol).Thin-layer hydration combined with sonicationTween 60/Span 60 showed better nanoencapsulation efficiency[106]
Trans-Ferulic acidNanoparticleNanoparticle A: poly (lactic acid)
Nanoparticle B: poly (lactic acid)/poly (lactic-co-glycolic acid)
Nanoprecipitation75% nanoencapsulation efficiency
Controlled liberation in in vitro analysis
No toxic effects at concentrations of 2.5 mg/ml
[107]
Chlorogenic acidNanoparticleChitosan, pentasodium tripolyphosphateIonic gelation59% nanoencapsulation efficiency
69% release after 100 h
[108,109]
PhloretinNanoparticleChitosan, sodium tripolyphosphateIonotropic gelation↑ mitochondrial-mediated apoptotic
↓ oxidative stress
[110]
Tea PolyphenolNanoparticleChitosan, sulfobutylether-β-cyclodextrinInclusion complexes↑ antioxidant activity
Chemically stable nanocapsule
[111]
Phenolics of grape pomaceNanocapsulesChitosan, soy proteinNanoemulsification95 and 75% nanoencapsulation efficiency
↑ antioxidant capacity
[112]
Phenolics of apple pomaceNanocapsulesChitosan, soy proteinNanoemulsification95 and 75% nanoencapsulation efficiency
↑ antioxidant capacity
[112]
Olive leaf phenolicsNanoparticleWhey protein concentrate and tween 20 as surfactantElectrospray232.3–659.8 nm nanoparticle size
0.074–0.65 Polydispersity index
[113]
Phenolics of pistachio hullsNanoliposomeLecithinSonication↑ antioxidant capacity
↑ anti-inflammatory activity
Anti-melanogenic activity
[114]
OleuropinNanoemulsionSoybean oil, span 80 (surfactant), whey protein concentrate, and pectinDouble emulsification91% nanoencapsulation efficiency
40.4% liberation
[115]
CurcuminNanoparticlePolyvinyl alcohol,
Poly(lactide-co-glycolic) acid
Modified emulsion- diffusion-evaporation method↑ bioavailability
↑ delay diabetic cataract in rats
[116]
CurcuminNanoparticlePoly (maleic anhydride-alt-1-octadecene), poly (ethylene glycol)-amine and
1-Ethyl-3-(3-dimethylaminopropyl)-carbodiimide
Sonication↑ dispersibility
↑ stability
↑ therapeutic efficiency
[117]
Vicenin-2NanoparticleChloroauric acidUltrasonicationCell viability 78.21%
↑ glucose uptake
Strong interaction with PTP1B and AMPK
[118]
SylbinNanoparticleChitosan, poly(lactide-co-glycolic) acid, pluronic F-127Solvent diffusion and polyelectrolyte deposition92.11% nanoencapsulation efficiency
↓ blood glucose
↓ glycosylated hemoglobin
[119]
Anthocyanin from raspberryNanoparticleβ-Lactoglobulin, N-(3-Dimethylaminopropyl)-N-ethyl carbodiimide hydrochloride (cross-linking)Desolvation77% nanoencapsulation efficiency
↑ antioxidant activity
↑ stability, bioavailability
[120]
Cyanidin 3-O-GlucosideNanoparticleNanoparticle1: Chitosan, and PGA
Nanoparticle 2: Chitosan oligosaccharide and polyglutamic acid
Nanoparticle 3: Carboxymethyl chitosan, CaCl2
Ionic crosslinking53.88% nanoencapsulation efficiency
75% release at pH 5.3
[121]
Cyanidin 3-O-GlucosideNanoliposomePhosphatidylcholine and cholesterolReverse-phase evaporationOptimal conditions: concentration 0.17 mg/mL, temperature 41.41 °C, and relation 2.87[122]
* Compared with the non-encapsulation control group. SOD: superoxide dismutase; GPX: glutathione peroxidase; CAT: catalase; TBRAS: thiobarbituric acid reactive substances. ↑ means increased. ↓ means decreased.
Table 5. Lipinski’s rule of 5 evaluation of polyphenols with antidiabetic properties.
Table 5. Lipinski’s rule of 5 evaluation of polyphenols with antidiabetic properties.
MoleculeClass of CompoundPubChem CIDChemical FormulaMolecular WeightH Bond DonorsH Bond AcceptorLog pLipinski Rule of 5
Cyanidin 3-glucosideAnthocyanin197081C21H21ClO11484.8811−1.5No
CurcuminCurcuminoids969516C21H20O6368.4263.2Yes
(+)-CatechinFlavanol9064C15H14O6290.27561.4Yes
(−)-EpicatechinFlavanol72276C15H14O6290.27561.8Yes
LiquiritinFlavanone503737C21H22O9418.4590.4Yes
NaringeninFlavanone439246C15H12O5272.25352.2Yes
ChrysinFlavone5281607C15H10O4254.24242.5Yes
HesperidinFlavone10621C28H34O15610.6815−1.1No
LuteolinFlavone5280445C15H10O6286.24462.0Yes
MyricetinFlavonol5281672C15H10O8318.23681.6No
QuercetinFlavonol5280343C15H10O7302.23571.5Yes
MangiferinGlucosylxanthone5281647C19H18O11422.3811−0.4No
Benzoic acidHydroxybenzoic acid243C7H6O2122.12121.87Yes
Hydroxybenzoic acidHydroxybenzoic acid135C7H6O3138.12231.58yes
Gallic acidHydroxybenzoic acid370C7H6O5170.12450.7Yes
Ferulic acidHydroxycinnamic acid445858C10H10O4194.18241.5Yes
Cinnamic acidHydroxycinnamic acid444539C9H8O2148.16122.1Yes
Caffeic acidHydroxycinnamic acid689043C9H8O4180.16341.2Yes
Coumaric acidHydroxycinnamic acid637542C9H8O3164.16231.5Yes
Rosmarinic acidHydroxycinnamic acid5281792C18H16O8360.3582.4Yes
ResveratrolStilbene445154C14H12O3228.24333.1Yes
Table 6. Predicted LDL50 and toxicity of polyphenols with antidiabetic potential.
Table 6. Predicted LDL50 and toxicity of polyphenols with antidiabetic potential.
MoleculePredicted LD50 (mg/kg)Predicted Toxicity ClassHepatotoxicityNeuro ToxicityNephrotoxicityRespiratory ToxicityCardiotoxicityCarcinogenicityInmuno ToxicityMutagenicityCytotoxicityClinical ToxicityNutricional Toxicity
Cyanidin 3-glucoside50005InactiveInactiveActiveActiveInactiveInactiveActiveInactiveInactiveInactiveActive
Curcumin20004InactiveInactiveActiveInactiveActiveInactiveActiveInactiveInactiveActiveInactive
(+)-Catechin10,0006InactiveInactiveActiveActiveInactiveInactiveInactiveInactiveInactiveActiveActive
(−)-Epicatechin10,0006InactiveInactiveActiveActiveInactiveInactiveInactiveInactiveInactiveActiveActive
Liquiritin23005InactiveInactiveActiveActiveInactiveInactiveActiveInactiveInactiveActiveActive
Naringenin20004InactiveInactiveActiveActiveInactiveInactiveInactiveInactiveActiveActiveActive
Chrysin39195InactiveInactiveActiveActiveInactiveInactiveInactiveInactiveInactiveInactiveInactive
Hesperidin12,0006InactiveInactiveActiveActiveInactiveInactiveActiveInactiveInactiveActiveActive
Luteolin39195InactiveInactiveActiveActiveInactiveActiveInactiveActiveInactiveInactiveActive
Myricetin1593InactiveInactiveActiveActiveInactiveActiveInactiveActiveInactiveInactiveActive
Quercetin1593InactiveInactiveActiveActiveInactiveActiveInactiveActiveInactiveInactiveActive
Mangiferin21InactiveInactiveActiveActiveInactiveInactiveActiveActiveInactiveActiveActive
Benzoic acid2903ActiveInactiveActiveInactiveInactiveInactiveInactiveInactiveInactiveInactiveInactive
Hydroxybenzoic acid22005InactiveInactiveActiveInactiveInactiveInactiveInactiveInactiveInactiveInactiveInactive
Gallic acid20004InactiveInactiveActiveActiveInactiveActiveInactiveInactiveInactiveActiveInactive
Ferulic acid17724InactiveInactiveActiveInactiveInactiveInactiveActiveInactiveInactiveActiveInactive
Cinnamic acid25005ActiveInactiveActiveInactiveInactiveInactiveInactiveInactiveInactiveInactiveInactive
Caffeic acid29805InactiveInactiveActiveInactiveInactiveActiveInactiveInactiveInactiveActiveInactive
Coumaric acid28505InactiveInactiveActiveInactiveInactiveActiveInactiveInactiveInactiveInactiveInactive
Rosmarinic acid50005InactiveInactiveActiveInactiveInactiveInactiveActiveInactiveInactiveActiveInactive
Resveratrol15604InactiveInactiveActiveInactiveActiveInactiveInactiveInactiveInactiveInactiveInactive
Predicted toxicity classScipharm 93 00055 i001
Table 7. Prediction of CYP inhibition of phenolic compounds with antidiabetic potential.
Table 7. Prediction of CYP inhibition of phenolic compounds with antidiabetic potential.
MoleculeInhibitor
CYP1A2CYP2C19CYP2C9CYP2D6CYP3A4CYP2B6CYP2C8
Cyanidin 3-glucosideNoNoNoNoNoNoYes
CurcuminNoNoYesNoNoYesYes
(+)-CatechinNoNoNoNoNoNoYes
(−)-EpicatechinNoNoNoNoNoYesYes
LiquiritinNoNoNoNoNoNoNo
NaringeninYesYesYesNoYesYesYes
ChrysinYesNoNoYesYesYesYes
HesperidinNoNoNoNoNoNoNo
LuteolinYesNoNoYesYesYesYes
MyricetinYesNoYesNoYesNoYes
QuercetinYesNoNoNoYesNoYes
MangiferinNoNoNoNoNoNoYes
Benzoic acidNoNoNoNoNoNoNo
Hydroxybenzoic acidNoNoNoNoNoNoNo
Gallic acidNoNoNoNoNoNoNo
Ferulic acidNoNoNoNoNoNoYes
Cinnamic acidNoNoNoNoNoNoYes
Caffeic acidNoNoNoNoNoNoYes
Coumaric acidNoNoNoNoNoNoYes
Rosmarinic acidNoNoNoNoNoNoNo
ResveratrolYesNoNoNoYesNoYes
Table 8. Pharmacokinetic properties of polyphenols with antidiabetic properties.
Table 8. Pharmacokinetic properties of polyphenols with antidiabetic properties.
MoleculeHLM Stability *BBB *P-gp Inhibitor *GI Absorption *
Cyanidin 3-glucosideNoNoNoLow
CurcuminYesNoNoHigh
(+)-CatechinNoNoNoHigh
(−)-EpicatechinNoNoNoHigh
LiquiritinNoNoNoLow
NaringeninYesNoYesHigh
ChrysinYesNoNoHigh
HesperidinNoNoNoLow
LuteolinYesNoNoHigh
MyricetinYesNoNoLow
QuercetinYesNoNoHigh
MangiferinNoNoNoLow
Benzoic acidNoNoNoHigh
Hydroxybenzoic acidNoNoNoHigh
Gallic acidNoNoNoHigh
Ferulic acidNoNoNoHigh
Cinnamic acidNoNoNoHigh
Caffeic acidNoNoNoHigh
Coumaric acidNoNoNoHigh
Rosmarinic acidNoNoNoLow
ResveratrolNoNoNoHigh
* HLM: The human liver microsomal; BBB: Blood–brain barrier; P-gp: P-glycoprotein; GI: Gastrointestinal.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cabanillas-Ponce de León, R.; Cardenas-Torres, F.I.; Ontiveros, N.; Contreras-Angulo, L.A.; Elisande-Romero, C.A.; Leyva-López, N.; Bernal-Millán, M.d.J.; Heredia, J.B.; Gutiérrez-Grijalva, E.P. Advancements in Encapsulation Technologies: The Potential of Polyphenols as an Antidiabetic Therapy. Sci. Pharm. 2025, 93, 55. https://doi.org/10.3390/scipharm93040055

AMA Style

Cabanillas-Ponce de León R, Cardenas-Torres FI, Ontiveros N, Contreras-Angulo LA, Elisande-Romero CA, Leyva-López N, Bernal-Millán MdJ, Heredia JB, Gutiérrez-Grijalva EP. Advancements in Encapsulation Technologies: The Potential of Polyphenols as an Antidiabetic Therapy. Scientia Pharmaceutica. 2025; 93(4):55. https://doi.org/10.3390/scipharm93040055

Chicago/Turabian Style

Cabanillas-Ponce de León, Rigoberto, Feliznando Isidro Cardenas-Torres, Noe Ontiveros, Laura Aracely Contreras-Angulo, Cristina Alicia Elisande-Romero, Nayely Leyva-López, Manuel de Jesús Bernal-Millán, Jose Basilio Heredia, and Erick Paul Gutiérrez-Grijalva. 2025. "Advancements in Encapsulation Technologies: The Potential of Polyphenols as an Antidiabetic Therapy" Scientia Pharmaceutica 93, no. 4: 55. https://doi.org/10.3390/scipharm93040055

APA Style

Cabanillas-Ponce de León, R., Cardenas-Torres, F. I., Ontiveros, N., Contreras-Angulo, L. A., Elisande-Romero, C. A., Leyva-López, N., Bernal-Millán, M. d. J., Heredia, J. B., & Gutiérrez-Grijalva, E. P. (2025). Advancements in Encapsulation Technologies: The Potential of Polyphenols as an Antidiabetic Therapy. Scientia Pharmaceutica, 93(4), 55. https://doi.org/10.3390/scipharm93040055

Article Metrics

Back to TopTop