Next Article in Journal
Synbiotic Microencapsulation of Lactobacillus Strains from Mexican Fermented Beverages for Enhanced Probiotic Functionality
Previous Article in Journal
A Study on the Electrochemical Performance of SLMed Al6061/TiB2 Composite Anodes Caused by Laser Power
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability

1
Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China
2
Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(5), 1184; https://doi.org/10.3390/molecules30051184
Submission received: 24 January 2025 / Revised: 4 March 2025 / Accepted: 4 March 2025 / Published: 6 March 2025

Abstract

:
Flavonoids represent a class of natural plant secondary metabolites with multiple activities including antioxidant, antitumor, anti-inflammatory, and antimicrobial properties. However, due to their structural characteristics, they often exhibit low bioavailability in vivo. In this review, we focus on the in vivo study of flavonoids, particularly the effects of gut microbiome on flavonoids, including common modifications such as methylation, acetylation, and dehydroxylation, etc. These modifications aim to change the structural characteristics of the original substances to enhance absorption and bioavailability. In order to improve the bioavailability of flavonoids, we discuss two feasible methods, namely dosage form modification and chemical modification, and hope that these approaches will offer new insights into the application of flavonoids for human health. In this article, we also introduce the types, plant sources, and efficacy of flavonoids. In conclusion, this is a comprehensive review on how to improve the bioavailability of flavonoids.

Graphical Abstract

1. Introduction

Flavonoids are a class of natural polyphenolic compounds widely found in plants and are part of the secondary metabolites of plants [1]. They are mainly present in various parts of fruits, vegetables, tea leaves, nuts, flowers and other plants. Citrus fruits (such as oranges and lemons), grapes, apples, and berries are rich in flavonoids, including hesperidin, anthocyanins, and quercetin. Green leafy vegetables such as spinach, kale, onions, and broccoli are also important sources of flavonoids [2]. In addition, theaflavins and catechins in tea, quercetin in nuts, and isoflavones in soybeans (such as soy isoflavones) are typical sources of flavonoids [3,4].
Flavonoids in nature connect to sugar molecules (glycosyl) through glycosidic linkage to form flavonoid glycosides. Aglycones are non-sugar parts of glycosides, for example, quercitrin (quercetin-3-O-glucoside) are glycosides, while quercetin are aglycones. The activities of flavonoid glycosides and their aglycones are quite different. In general, flavonoid glycosides exhibit greater hydrophilicity compared to their corresponding aglycones, while they tend to have lower lipophilicity than the aglycones. Flavonoids (aglycones) exhibit high bioactivity, but their stability is very poor. When the sugar substituent binds to the aglycones through glycosidic linkage, the bioactivity of the flavonoids decreases but their stability is improved [5]. In terms of drug activity, flavonoid glycosides may be better than their aglycones [6]. In addition, glycosylation can increase the hydrophilicity of flavonoid aglycones, reduce toxic side effects, and improve the specific targeting ability of the drug [6]. In terms of bioavailability, flavonoid glycosides are generally lower than those of aglycones [7]. But this phenomenon is not absolute, because the plasma concentration of flavonoid-C glycosides seems to be significantly higher in the body than their aglycones [8]. Furthermore, glycosylation is also believed to protect plants from self-oxidative damage, although the biological activity of flavonoids may decrease [8].
As natural products, flavonoids have various positive effects on plants. These compounds are the natural coloring agents of plants, especially anthocyanins, which are important players in plants’ ability to display their vibrant colors [9]. At the same time, the dazzling colors that are made possible by the presence of flavonoids can attract animals that spread seeds on behalf of the plant itself [10]. Flavonoids are also associated with plant growth and development and play a role in the growth of plant leaves through their antioxidant capacity and regulatory properties [11]. Of course, as essential players in nature, not all plants are at the top of the pyramid. Most face numerous biotic or abiotic stresses, such as being “tasty meals” for herbivores, invasive pathogens, UV radiation, drought, and more. It seems that flavonoids have become the main means by which plants combat these stresses [12]. Flavonoids play a significant role in protecting plants from herbivores by altering their taste, reducing their digestibility, and increasing toxicity at high concentrations [13]. Flavonoids have also been found to combat fungal or bacterial infections in plants by inducing apoptosis and mitochondrial dysfunction, as well as by blocking cell wall synthesis [14,15]. Another example is the finding that flavonoids from maize species enhance the plant’s drought tolerance by regulating drought-induced oxidative damage and stomatal movement [16].
Meanwhile, they also have positive effects on human health through dietary intake, including antioxidant, antitumor, anti-inflammatory, antimicrobial and other bioactivities [2]. Although increasing studies have shown the great potential of flavonoids in promoting human health, the limiting factors of their bioavailability are often overlooked when explaining their “human-friendly” bioactivities. In other words, certain flavonoids can indeed promote human health, but the questions of how, how much, and how effectively the body can use flavonoids are often understated. In fact, the bioavailability of flavonoids is relatively low, mainly due to the following factors. First, the interaction between flavonoids and other nutrients, for example, fat intake will increase its bioavailability, while protein intake will reduce its bioavailability [17]; the second is the metabolic behavior of the liver (Phase I and Phase II), such as the methylation, sulfation or glucuronidation of flavonoids [18]; the third is the interaction with the gut microbiome. It is worth noting that after being “processed” by microorganisms, the bioactivity of some flavonoids may even improve [18]. Additionally, factors such as diet, genetics, and metabolic diseases can significantly affect flavonoid bioavailability, influencing how effectively these compounds are absorbed and utilized by the body. In short, when considering the bioactivity of flavonoids, their bioavailability must be taken into account first, because their bioactivity depends largely on their bioavailability.
Research on the structural characteristics, bioactivities and in vivo processes of flavonoids is the key point for trying to improve their bioavailability. Modern science and technology have enabled the modification of drugs or compounds through formulation technology or chemical modification methods, allowing certain physical and chemical properties of the original compound to be altered towards an ideal state. In this review, we primarily discuss how to improve the bioavailability of flavonoids and propose that it can be achieved by changing the dosage form of the drug and through chemical modification, providing a theoretical basis for the long-term development of flavonoids in the field of medical health.
In order to complete this manuscript, we performed an exhaustive and methodical review of the literature by querying several prominent scientific databases, including PubMed, ScienceDirect, Web of Science, Scopus, Google Scholar, SpringerLink, Wiley Online Library. The search was conducted using a combination of key terms, such as flavonoids, biological activity, plant sources, gut microbiome, nano preparation, and structural modifications. This comprehensive search strategy facilitated the acquisition of the most relevant and current research, ensuring a thorough and up-to-date understanding of the biological properties, health-promoting potential, and applications of flavonoids. Furthermore, particular attention was given to the exploration of plant-based sources, the interaction between gut microbes and flavonoid metabolism, and the latest advancements in pharmaceutical technologies, which collectively provided a well-rounded and nuanced perspective on the subject matter.

2. Classification of Flavonoids

Flavonoids are a class of compounds with diphenyl chromone as the basic nucleus. They are C6–C3–C6 structural substances consisting of two benzene rings (A and B rings) and one pyran ring (C ring, a three-carbon structure) [19] (Figure 1).
It is worth noting that there are two ways for plants to synthesize flavonoids, one is the shikimic acid pathway, which produces a phenylpropanoid (C6–C3) skeleton, and the other is the acetate pathway, which provides building blocks for polymerizing two-carbon units [1]. The acetate pathway generates ring A, the shikimic acid pathway generates ring B, ring A and ring B condense to form chalcone (a precursor of flavonoids), which then undergoes catalysis by chalcone isomerase to form the heterocyclic (C-ring) flavanone. And flavanone is the starting compound for the synthesis of other flavonoids [1,20] (Figure 2).
To date, more than 10,000 natural flavonoids have been identified [21]. According to the connection point between the benzene ring (B ring) and the pyran ring, and whether the central three-carbon chain is cyclized and the degree of oxidation, flavonoids can be roughly divided into the following subcategories: flavones, isoflavones, flavonols, flavanols, flavanones chalcones, and anthocyanidins [22] (Table 1). Catechins are the most abundant flavanols in tea. While flavanones are mainly derived from citrus fruits, among which hesperidin, naringenin and paclitaxel are the most representative. Flavones such as luteolin and apigenin can be found in celery, tea, red pepper, and oranges [22]. The main sources of isoflavones are beans and their derivatives, with genistein and glycitein being the representative compounds. Quercetin is the most widely studied flavonol compound, mainly derived from onions, kale, leeks, and other dietary sources. Chalcone is a natural open-chain flavonoid and a precursor to all other flavonoids [23]. Xanthohumol and Corylifolinin are the most representative chalcone compounds owing to their rich pharmacological activities. Anthocyanidins are natural dyes found in plants, giving flowers and fruits their attractive colors [24]. The most abundant anthocyanidins include delphinidin, cyanidin, petunidin, peonydin, malvidin, and pelargonidin [25].

3. Efficacy of Flavonoids

3.1. Antioxidant

In 1985, a monograph titled “Oxidative Stress” first introduced the concept of oxidative stress, in which oxidative stress is described as oxidative damage to cells and organs [47]. Oxidative stress generally refers to the phenomenon in which the balance between oxidation and anti-oxidation is disrupted or reactive oxygen species (ROS)/reactive nitrogen species (RNS) are excessively produced in organisms to induce inflammatory infiltration of neutrophils and trigger a series of cellular damage and pathological reactions. ROS include superoxide anion, hydroxyl radical, hydrogen peroxide and singlet oxygen; RNS include nitric oxide, nitrogen dioxide and peroxynitrite [48]. Correspondingly, according to the nature of antioxidants, the system that antagonizes oxidative stress in the body can be roughly divided into enzymatic and non-enzymatic systems. The enzymatic antioxidant system includes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px). The non-enzymatic antioxidant system includes vitamin C, vitamin E, carotenoids and some trace elements such as selenium and zinc. Some views in modern molecular biology suggest that oxidative stress is the result of the disruption of the thiol redox pathway (controlled by thioredoxin Trx, glutathione GSH and cystedin Cys). The influence of the thiol redox pathway on some pathways, such as HRas, PTP-18, Nrf2, NF-κB, etc., can significantly interfere with normal physiological activities of the human body [49,50].
Flavonoid compounds can directly remove excessive ROS in the body due to their special structural morphology. The specific activity intensity is arranged in the following order: Flavanol (Flavan-3-ols) (4.21 mM) > Flavonol (1.83 mM) > Chalcone (1.51 mM) > Flavonoid (0.91 mM) > Flavanone (0.16 mM) > Isoflavone (0.08 mM) [51]. In short, the activity of flavonoids in scavenging ROS depends on their own structure, the number and arrangement of hydroxyl groups play an important role in the antioxidant effect of flavonoids [52,53]. This effect may be attributed to the hydrogen atoms provided by the hydroxyl structure or the electron delocalization effect triggered by conjugation with the oxygen group, which leads to the scavenging of free radicals [51], and the hydroxyl structure of the B ring is the most important factor in determining the clearance of ROS and RNS compared to the A and C rings [54].
Flavonoids can also play an antioxidant role by activating antioxidant enzymes. Studies have shown that flavonoids activate electrophile-responsive element (EpRE) to regulate multiple phase II detoxification enzymes including NAD(P)H quinone oxidoreductase (NQO), glutathione S-transferase (GST), and UDP glucuronyl transferase (UGT), thereby resisting oxidative stress [54]. Correspondingly, these substances can also exert their antioxidant stress resistance by inhibiting oxidases. Currently, flavonoid compounds have been shown to inhibit cyclooxygenase, lipoxygenase, microsomal monooxygenase, nicotinamide adenine dinucleotide (NADH) oxidase, xanthine oxidase, and others [22]. Studies have also shown that quercetin can inhibit the burst of oxidative stress by downregulating the protein kinase C (PKC) pathway to block its phosphorylation process of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) subunit p47phox [55].
In addition, flavonoids are also considered to form chelates with metal ions to combat oxidative stress. One study pointed out that quercetin and morin exert antioxidant effects by antagonizing the decrease in the activities of SOD and GSH induced by cadmium (Cd). Compared with the Cd-only group, the SOD and GSH activities in mice receiving quercetin + Cd or mulberry + Cd groups showed significantly higher activity (p ⩽ 0.001) [56].
Oxidation of human low-density lipoprotein (LDL) is associated with atherosclerosis, and α-tocopherol is an antioxidant for LDL. Studies have shown that flavonoids such as quercetin, epigallocatechin, epigallocatechin gallate, epicatechin, epicatechin gallate and naringin can act as hydrogen donors to reduce α-tocopherol free radicals or promote the production of α-tocopherol, thereby delaying the oxidation of LDL [57,58].
Of course, there are some relatively new views suggesting that flavonoid compounds play an antioxidant role by increasing the level of uric acid in plasma, but this view still lacks specific data support [59]. Some studies have also shown that when β-carotene is combined with flavonoid compounds such as hesperidin, rutin, and quercetin, it exhibits an anti-DNA-damage effect under UV-A irradiation [60].
In summary, the mechanisms of flavonoids against oxidative stress can be summarized as follows:
  • The hydroxyl structure of flavonoids acts as a hydrogen atom donor to neutralize free radicals to directly remove ROS.
  • The oxo group of flavonoids participates in the conjugated system to enhance electron delocalization.
  • Activation of antioxidant enzymes activity.
  • Inhibition of oxidase activity.
  • Formation of metal chelates.
Finally, it is worth noting that under pathological conditions, high levels of free radicals can damage molecules such as nucleic acids, proteins, and lipids, ultimately leading to cell aging and death, as well as promoting the development of tumors [61,62,63]. Therefore, the antioxidant biological properties of flavonoids may be the prerequisite for their other pharmacological activities, especially antitumor activity [64].

3.2. Anticancer

Uncontrolled abnormal proliferation of normal cells in the body is called cancer [65]. Various cellular activities such as growth, proliferation, apoptosis, and metastasis of tumor cells are regulated by multiple important signal cascades such as Nuclear factor erythroid 2-related factor 2 (Nrf2), Cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING), WNT/β-catenin, Rat Sarcoma/Rapidly Accelerated Fibrosarcoma/Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase (Ras/Raf/MEK/ERK), Epidermal growth factor receptor (EGFR), Phosphoinositide-specific Phospholipase C/Protein kinase C (PLC/PKC), Phosphoinositide 3-Kinase/Protein kinase B (PI3K/AKT), Notch Receptor (NOTCH), Platelet-derived growth factor (PDGF), Vascular Endothelial Growth Factors (VEGF), Cellular-mesenchymal epithelial transition factor (c-MET), Yes-associated protein/Transcriptional Enhanced Associate Domains (YAP/TEAD), Phospho P38 mitogen-activated protein kinase/Mitogen-activated protein kinase/Extracellular regulated protein kinases (P38/MAPK/Erk), Toll-like receptors (TLR) etc.) [66,67,68,69,70,71,72,73,74,75]. It is worth noting that these pathways are not involved in regulation in isolation. Pathways are closely related to each other to form signaling cascades. Usually, inhibition of such signaling pathways/cascades can significantly inhibit the development of cancer and even overcome the problem of drug resistance [76,77,78].
Currently, a large number of studies have shown that flavonoid compounds can exert a powerful tumor-inhibitory effect, including the regulation of multi-level signaling pathways, key proteins, and factors [79]. Take Taxifolin (TAX) as an example: TAX is a flavonoid derivative found in natural plants. Studies have shown that TAX exerts tumor suppression by regulating the MAPK signaling pathway. Specifically, the regulation of the MAPK pathway includes the inhibition of pERK, pJNK, p-p38, Rac1, and c-Myc. This result is supported by Western blotting experiments [80]. The study also found that TAX inhibited the nuclear translocation of the tumor cell division gene β-catenin by inhibiting Rac1/JNK signaling. The expression of the β-catenin downstream gene, microphthalmia transcription factor (MITF) was inhibited in B16F10 and A375 cell lines treated with TAX [80]. Research by Manigandan et al. [81] revealed a potential mechanism for TAX in fighting rectal cancer. TAX stimulates the expression of the Nrf2 protein by inhibiting Wnt/β-catenin signaling, while Nrf2 protects cells from oxidative stress and DNA damage by enhancing the expression of antioxidant enzymes, which is believed to reduce the risk of cancer development. However, excessive activation of Nrf2 may promote the malignant proliferation of cancer cells, enhance the invasiveness and metastasis of tumor cells, and induce drug resistance. Therefore, it remains to be determined whether TAX-induced Nrf2 expression also plays a positive role in other types of cancer besides rectal cancer [82]. A previous study found that both the flavonoid biochanin A and the chemotherapy drug temozolomide inhibited the cell viability of human glioblastoma U87 MG cells in a dose-dependent manner. Compared to the control group, the inhibitory effect of combined administration of the two was more significant (p ≤ 0.05), subsequent Western blotting experiments confirmed the specific inhibitory mechanism. The experiment found that the combined drug treatment inhibited the levels of p-ERK, p-AKT, EGFR and c-myc, and upregulated the expression of phosphorylated p53 (p-p53) [83]. In addition, the inhibition of flavonoids such as hesperidin, naringin, quercetin, luteolin, and apigenin on NF-κB, ERK1/2, AP1, TNF-α, PKC, and Nrf2 pathways has been proved to have significant pharmacological activities on a variety of cancers, including hepatocellular carcinoma, colon cancer, breast cancer, lung cancer, cervical cancer, and bladder cancer. Of course, these activities may be attributed to the antioxidant activity of flavonoids [64]. The issue of tumor resistance has long been a hot topic of research. Recently, some scholars have pointed out that flavonoids and their conjugates interact with efflux proteins (mainly the ABC transporter family), especially P-glycoprotein and breast cancer resistance protein (BCRP). The interaction between flavonoids and these ABC transporters (ATP binding cassettes) may be an important way to enhance the bioavailability of chemotherapeutic drugs and overcome tumor resistance [78].

3.3. Anti-Inflammatory

Inflammation is a complex pathological and physiological process associated with a variety of diseases, involving the activation of both immune and non-immune cells [84]. Through the inflammatory response, invading pathogens or dysfunctional cells are eliminated to promote tissue repair and restoration of homeostasis [85]. Flavonoids have been shown to have strong anti-inflammatory effects and can inhibit the development of inflammation by inhibiting the production of inflammatory mediators such as prostaglandins and leukotrienes. Quercetin is a flavonoid with multiple physiological activities, including antitumor and anti-inflammatory effects. A recent study found that quercetin inhibited the expression of cyclooxygenase-2 (COX-2) protein and COX-2 promoter activity in MDA-MB-231 cells in a dose-dependent manner [86]. Nobiletin was found to block the mRNA expression of inducible nitric oxide synthase (iNOS) and COX-2 and significantly inhibited LPS-induced nitric oxide (NO) and prostaglandin E2 (PGE2) production [87].
In addition, flavonoids inhibit the progression of inflammation by regulating the nuclear factor κB (NF-κB) signaling pathway [88,89,90]. NF-κB is an important transcription factor, involved in the regulation of various inflammatory reactions [91]. During an inflammatory response, NF-κB promotes the release of pro-inflammatory factors such as TNF-α, IL-1β and IL-6 by activating the transcription of downstream genes [92]. Flavonoids can modulate the NF-κB pathway through multiple mechanisms, thereby reducing the production of pro-inflammatory factors. Green tea polyphenols have been found to alleviate silicosis by inhibiting the expression of proinflammatory factors TNF-α, IL-1β, and IL-6 by inhibiting the IL-17/NF-κBp65 signaling cascade [93]. Other studies have shown that flavonoids can bind to IκB kinase (IKK) and inhibit the nuclear translocation of NF-κB, thereby significantly reducing inflammatory responses [94,95].
It is worth noting that flavonoids can also play an anti-inflammatory role as antioxidants. Flavonoids inhibit inflammatory factors or promote the expression of antioxidant genes by regulating classic inflammatory response pathways (such as TLR4-NF-κB, PI3K-AKT and Nrf2/HO-1 signaling pathways), thus playing an anti-inflammatory role in the cardiovascular system [96].

3.4. Antimicrobial

In recent years, as the problem of antibiotic resistance has become increasingly serious [97], researchers have paid more and more attention to the potential of flavonoids as natural antimicrobial agents. Flavonoids can combat bacterial infection through many mechanisms, including inhibiting bacterial cell wall synthesis, damaging bacterial cell membranes, inducing oxidative stress, inhibiting protein synthesis, and interfering with bacterial DNA replication and transcription [98]. A recent study explained the antibacterial mechanism of the flavonoids sophoraflavanone G and kurarinone. The results showed that these two substances inhibited the bioactivity of Staphylococcus aureus in a dose-dependent manner, and even their MIC90 (the minimum concentration of the tested drug that inhibits 90% bacterial growth) remained stable after 20 consecutive subcultures. This indicates that Staphylococcus aureus has a higher sensitivity to these two substances. Further studies have found that these two substances mainly exert their corresponding antibacterial effects by targeting the cell membrane of Staphylococcus aureus, causing structural damage to the membrane, and inhibiting the biosynthesis of the cell membrane and cell wall [99]. Pyocyanin and elastase are important virulence factors of Pseudomonas aeruginosa, which are related to its biofilm formation and immune evasion, respectively [100,101]. Naringenin was found to exert its antibacterial properties by inhibiting both pyocyanin and elastase [102]. In addition, some studies have shown that flavonoids such as epigallocatechin gallate (EGCG) and theaflavin-3,3′-digallate (TF3) exert anti-Clostridium perfringens effects in a concentration-dependent manner by inhibiting proteins involved in septum formation, DNA separation, and cell division [103]. It is worth noting that Lee et al. revealed an unusual antibacterial mechanism of flavonoids for the first time. In this study, it was found that the flavonoid propolin D from Macaranga tanarius (L.) inhibited the formation of Candida albicans biofilm by reducing the formation of hyphae and inhibited the formation of Enterohemorrhagic Escherichia coli (EHEC) biofilm by reducing the formation of curly fimbriae. The results were demonstrated by qRT-PCR. These results showed that propolin D could significantly downregulate the gene expression of ECE1, HWP1, and curli subunits (csgA and csgB) to inhibit the formation of biofilm [104].

4. Pharmacokinetics of Flavonoids

Most flavonoids exist in glycoside form (except flavanol) [105]. Flavonoids are absorbed and metabolized in the form of glycosides or aglycones, but the specific mechanisms underlying the difference in absorption ability between glycosides and aglycones are still unknown [18]. Usually, these compounds are absorbed in the small intestine (except flavan-3-ols and proanthocyanidins) [106]. The absorption mechanism was divided into two types based on the cleavage enzymes present in the brush border epithelial cells of the small intestine. One mechanism is thought to be the conversion of flavonoids to their aglycone forms by the enzyme lactase phlorizin hydrolase (LPH), which, due to their increased lipid solubility, allows them to enter the intestinal epithelial cells via passive diffusion. The second mechanism involves the transfer of hydrophilic glycosides by certain membrane transporters, such as sodium-dependent glucose transporter 1 (SGLT-1) and glucose transporter 2 (GLUT-2) [107]. After transport by SGLT-1 or GLUT-2, cytosolic β-glucosidase (CBG) or LPH hydrolyzes flavonoid glycosides into their aglycones for absorption [108,109]. Both aglycones and their corresponding glycosides can be absorbed through these two pathways, but in comparison, the absorption efficiency of flavonoid aglycones is higher than that of their corresponding glycosides. In a Caco-2BBe1 cell model, it was found that the metabolic kinetics of quercetin and anthocyanins differ significantly from their respective monoglycosides or bisglycosides. High levels of aglycone substances rather than glycoside substances were detected in cells and on the basolateral side. This suggested that aglycone substances have higher cellular uptake capacity and transport efficiency. However, when glycone groups are introduced into the aglycone structure, lower cellular uptake and transport behavior can be observed, and the greater the number of glycone groups, the lower the uptake and transport efficiency [107].
After the administration of quercetin or quercitrin, it is difficult to detect the components of these two substances in plasma, but their glucuronides, sulfates or methyl conjugates can be detected, which reminds us that some unexpected changes may occur in flavonoids after oral administration [110]. Studies have shown that such substances undergo hepatic metabolism by phase I hydroxylation of the aromatic ring and phase II metabolism by binding to UPD-glucuronosyltransferase (Glucuronidation), glutathione S-transferase (sulfation), and catechol-O-methyltransferase (methylation) after ingestion [111]. Glucuronidation and sulfation aim to increase the water solubility of flavonoids for renal excretion. The ortho-hydroxyl groups on the structure of flavonoids are methylated by catechol-O-methyltransferase, and the methylated flavonoids are primarily excreted through the kidneys [111]. In summary, glucuronidation, sulfation, and methylation serve to enhance the water solubility of these substances, promoting their excretion through bile and urine [105]. Furthermore, this process is also a key mechanism that limits the toxic potential of flavonoids [105]. Of course, some very interesting phenomena were revealed during the research process. Flavonoids seem to be more inclined to undergo sulfation rather than glucuronidation, but they can also transform to glucuronidation after sulfation. Of course, such a transformation needs to be established under the condition of large-scale intake [112]. The balance between sulfation and glucuronidation also seems to be affected by a number of factors, such as gender, species, and whether or not the individual is fasting [113].
Notably, flavonoids that have been absorbed by the small intestine enter the liver through the portal vein and are metabolized by hepatic enzymes and released into circulation, increasing the amount of polar conjugates excreted in the urine or returned to the duodenum through the gallbladder [22,114]. Meanwhile, most unabsorbed flavonoids enter the large intestine, where abundant gut microbiomes cleave their pyranone rings, undergo dehydroxylation, decarboxylation, and other reactions, further metabolizing the flavonoids, which are subsequently absorbed or excreted in the feces [115,116] (Figure 3). Of course, gut microbiomes may also change the structure of flavonoids to enhance their bioactivity and improve their bioavailability. But interestingly, some compounds that have been metabolized in the blood may even re-enter the liver circulation for further metabolism and eventually be excreted from the body in urine [108].

5. Flavonoids and Gut Microbiome

Some studies have suggested that the interaction between the gut microbiome and flavonoids may be a key factor in promoting the beneficial effects of flavonoids. That is, the gut microbiome may affect the utilization or biotransformation of flavonoids, and flavonoids may also affect the growth of microorganisms present in the intestine [117]. We may hypothesize that if the population and number of microorganisms in the intestinal tract that are “friendly” to flavonoids change, then the flavonoids ingested orally may lose most of their physiological activities that are beneficial to the human body, and this is a concern that requires vigilance. Previous studies have found that there are a large number of microorganisms in the intestine (about 105 in the small intestine, about 103 to 107 in the ileum, and about 1012 in the colon) [118]. These microorganisms will produce a large number of enzymes during growth and metabolism. Under the action of these enzymes, the flavonoids entering the intestinal tract will undergo reactions such as hydrolysis, reduction, deketolization, decarboxylation, etc. generating new microbial metabolites (Figure 4). These “processed” flavonoids undergo structural changes, which contribute to alterations in their bioactivity and bioavailability [119,120]. For example, flavonoid aglycones with 5-hydroxyl groups in their structure can be degraded faster in the intestine than structures without 5-hydroxyl groups [121], and structures without methoxy groups can be degraded faster than structures with methoxy groups [122].

5.1. Hydrolysis Reaction

In nature, flavonoids mainly exist in the form of glycosides combined with glycone groups. The glycosidic bond between the flavonoid and the glycone groups dissociates, resulting in the release of the flavonoid aglycone [123]. A group of active ingredients metabolically produced by gut microbiome and referred to as fecal microbial enzymes, such as α-rhamnosidase, β-glucosidase and β-glucuronidase, have been shown to be major players in the hydrolytic reactions of flavonoid glycosides in the intestinal tract. Higher levels of β-glucosidase and β-glucuronidase were observed in a diarrhea piglet model, both enzymes are derived from Escherichia spp. The increase in the number of Escherichia spp. under diarrheal conditions promoted the activity of these two enzymes. In subsequent experiments, it was also found that β-glucosidase and β-glucuronidase promoted the metabolism of daidzin, baicalin and wogonoside etc., because their corresponding daidzein, baicalein and wogonin etc. were detected in the body, and flavonoids in the form of aglycones seemed to have better therapeutic effects in fighting diarrhea [124]. Previously, a strain called PUE was found to convert [6″, 6″-D2] puerarin containing C-glucose into daidzein and [6″, 6″-D2] glucose (labeled with deuterium), the process believed to be caused by C-1 hydroxylation hydrolysis triggered by PUE” [125]. A study conducted some time ago found that the Bacteroides distasonis strain was present in the feces provided by volunteers, and this strain participated in the hydrolysis of flavonoids by providing α-rhamnosidase and β-galactosidase. It is reported that under the action of these two hydrolases, quercitrin and rutin were hydrolyzed to quercetin, and robinin was further hydrolyzed to kaempferol [126]. In general, based on previous learning experience, there are several rules regarding the difficulty of hydrolysis of flavonoid glycosides:
  • Depending on the type of glycoside bond, flavonoid glycosides undergo hydrolysis in roughly the following order of difficulty C-glycosides, S-glycosides, O-glycosides, and N-glycosides.
  • Among pyranosides, the hydrolysis rates are ranked from fast to slow as follows: pentose, methylpentose, hexose, heptose, glucuronide.
  • Amino-glycosides are more difficult to hydrolyze than hydroxy-glycosides, which are more difficult to hydrolyze than deoxyglycosides: 2-aminoglycosides < 2-hydroxyglycosides < 3-deoxyglycosides < 2-deoxyglycosides < 2, 3-deoxyglycosides.
  • Ketoglycosides are more easily hydrolyzed than aldosides.
  • Aromatic glycosides are more easily hydrolyzed than aliphatic glycosides.
  • Furanosides are more easily hydrolyzed than pyranosides.

5.2. Reduction Reaction

There are a large number of anaerobic bacteria in the gastrointestinal tract [127], and the double bond between C2 and C3 of flavonoids is easily hydrogenated and reduced [128]. A recent study attracted attention, reporting a previously undiscovered a distinct class of ene-reductases called flavonoid reductase (FLR). The researchers evaluated the metabolites of apigenin by co-culturing it with Escherichia coli expressing the A4U99_05915 gene (encoding the KGF53654.1 protein). As a result, the consumption of apigenin and the production of naringenin were observed in the supernatant. Further studies have shown that the FLR encoded by the A4U99_05915 gene has extremely high catalytic properties for flavonoids and flavonols, and it is catalytically active only against these two types of substrates and can catalyze the hydrogenation of the C2 = C3 double bond on the C ring of flavonoids and flavonols, generating dihydroflavones and dihydroflavonols [129]. In addition, flavonoids can also undergo a reduction reaction between the C2-O bonds under the action of the gut microbiome, resulting in the cleavage of the C ring. After ring opening, the O-position and C3-position of some flavonoids, such as flavonols, will recombine to form a five-membered ring [129,130]. There are also some more complex reactions, such as the opening of the C2 and O positions of apigenin under the action of E.ramulus leading to the formation of phloretin (chalcone). And the chalcone is further reduced to form dihydrochalcone, which ultimately produces phloroglucinol and p-hydroxyphenylpropionic acid [131].

5.3. (De)Methylation, Acetylation, Dehydroxylation Reactions

Flavone glycosides are hydrolyzed to produce their aglycone part (flavonoid parent core). In addition to being partially absorbed by the small intestine, the unused flavonoid parent core structure is prone to methylation, demethylation, acetylation, and dehydroxylation in the small intestine. It has been reported that after methylation, the bioactivity of flavonoids will be greatly improved [132,133]. The flavonoid O-methyltransferase (SaOMT-2) has been cloned from Streptomyces avermitilis and has been found to have a wide range of substrate properties, capable of methylation of various flavonoids, including isoflavones, flavonoids, flavonols, and flavanones [134]. Of course, flavonoids will undergo demethylation reactions under the influence of certain intestinal flora. For example, the intestinal bacterium Blautia sp. MRG-PMF1 produces a methyltransferase that catalyzes the complete demethylation of 5,7-dimethoxyflavone (5,7-DMF), 3,5,7-trimethoxyflavone (3,5,7-TMF), 3,5,7,3′, 4′-pentamethoxyflavone and other substances to produce their corresponding demethylation products [135]. In addition to methylation, acetylation is another common modification by intestinal bacteria. The main metabolites of hyperoside in the intestine were identified using ultra-high-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. The results showed that in the presence of intestinal bacteria hyperoside was metabolized into its corresponding acetylated products and dehydroxylated products [136]. Some bacteria can also catalyze the dehydroxylation of flavonoids. The dehydroxylation reaction is unique in the biotransformation between catechins and gut microbiome because it requires specific sites for dehydroxylation. For example, the premise for Eggerthella sp. CAT-1 to cleave the C ring of catechins is to ensure that the B ring has a para-hydroxyl group, and the prerequisite for removing the para-hydroxyl group of the B ring is the cleavage of the C ring [137].

6. Bioavailability of Flavonoids

The chemical structure of flavonoids consists of an organic skeleton with a C6–C3–C6 aromatic ring, and the bioavailability of flavonoids is usually very low [120,138]. Structure, solute–solvent interactions, crystallinity, and thermodynamic properties are important determinants of the aqueous solubility of flavonoids [139]. The degree of methoxylation and glycosylation may also affect the solubility of these compounds; for example, the presence of glycone groups and unsubstituted hydroxyl groups increases polarity and thus aqueous solubility, whereas methoxylation generally decreases polarity. Furthermore, these compounds behave as weak acids (e.g., rutin has an overall pKa of 6.37 and contains four phenolic acid groups with pKa values ranging from 7.1 to 11.65). Therefore, the intestinal pH is insufficient to allow them to be completely dissolved. Because these are weak acids, their protonated forms are less soluble. Passive diffusion is also unlikely due to the low lipophilicity of the bound compound at intestinal pH [139]. In general, the bioavailability of hydrophobic flavonoids increases with the increase in glycosides, but their pharmacological activities may decrease [139]. Generally speaking, the bioavailability, metabolism, and bioactivity of flavonoids depend on their structural composition, number of hydroxyl groups, and the presence of structurally substitutable functional groups [140,141]. Previous studies have pointed out that the bioavailability of some flavonoids differs across various categories and grades, as follows: phenolic acids > isoflavones > flavonols > catechins > flavanones, proanthocyanidins > anthocyanins [142,143,144,145]. The bioavailability of different flavonoids has also been demonstrated in a study in which the minimum absorption of several flavonoids was assessed using urinary excretion (equivalent to a percentage of intake) as the primary indicator. The results showed that the minimum absorption of daidzein was 42.3 ± 3.0%, while the corresponding aglycone (daidzein) was only 27.5%. The minimum absorption of genistin was 15.6 ± 1.8%, while the corresponding aglycone (genistein) was 8.6%. In addition, the study also pointed out the urinary excretion of other flavonoids such as gallic acid (37.7 ± 1.0%), anthocyanidins (0.4 ± 0.3%), hesperidin (8.6 ± 4.0%), naringin (8.8 ± 3.17%), etc. [142]. However, it is worth noting that the detection of urinary excretion has a strong correlation with the intake dose of flavonoids, so these data may only have some reference value at present, and the specific differences need to be further explored. Glycosylation is a key modification of most flavonoids, and the modification of glycosidic substances by glycosylation enhances the bioactivity and bioavailability of natural products [146,147]. But interestingly, the absorption of flavonoid aglycones seems to be higher than that of their corresponding flavonoid glycosides to some extent, because aglycones can be absorbed from the small intestine, while most flavonoid glycosides need to be broken down by intestinal enzymes or intestinal flora before they can be directly absorbed [105]. These results, however, demonstrate that the bioavailability of some flavonoid glycosides appears to be higher than that of their corresponding aglycone counterparts [142]. Again, the absorption efficiency of quercetin glucoside is higher than that of its aglycone form [148], and the bioavailability of daidzin is higher than that of its corresponding aglycone [149]. These conclusions suggest that our research on the bioavailability of flavonoids may be more complicated than we initially imagined. In addition, factors such as diet, genetics, and metabolic diseases may also play a significant role in flavonoid bioavailability. The composition and activity of intestinal microorganisms may differ significantly between individuals, resulting in differences in the absorption, distribution, metabolism, and excretion of flavonoids, especially considering that dietary intake of flavonoids can promote more favorable composition of human intestinal microorganisms [150]. In addition, the food matrix will have a great impact on the bioavailability of flavonoids, and high-fat diets have been shown to change the structure and diversity of intestinal microorganisms [151]. For example, quercetin is lipophilic, and a high-fat diet can enhance its absorption [152]. Genetic polymorphisms of enzymes associated with flavonoid metabolism may also lead to inter-individual differences in bioavailability [153]. In addition, factors such as age, gender, race, and (pathological) physiological status may also affect the bioavailability of flavonoids [153]. Therefore, when facing special groups, the dosage and method of flavonoids need to be selected with great care [154]. Metabolic diseases are another factor affecting the bioavailability of flavonoids. In one study, it was found that intake of quercetin affects gut microorganisms and reduces the progression of atherosclerosis, but these effects are influenced by the presence of dietary plant polysaccharides and metabolic diseases [155]. In another study, 400 mg/kg of mangiferin was administered to diabetic and normal rats. The plasma area under the curve (AUC) of diabetic rats was significantly higher than that of the control group. In addition, the metabolic products of diabetic rats were also more than those of regular rats [156].

6.1. Formulation Modification

Modern formulation techniques have made it possible to significantly improve the therapeutic efficacy, release control, and bioavailability of drugs by preparing drugs or carriers at the nanoscale. Such technologies are often referred to as nanomedicine preparations or nanocarrier drug delivery systems. The combination of modern formulation techniques with flavonoids represents a promising area of research for improving their bioavailability [157,158]. Therefore, in this chapter, we mainly review nano-flavonoid drugs that improve pharmacokinetics, with the aim of illustrating the enormous potential of nanotechnology in enhancing the bioavailability of flavonoids. By preparing flavonoid compounds such as luteolin, naringenin, quercetin, etc. into corresponding nano-suspensions, solid lipid nanoparticles, liposomes, gel systems, micelles, and other nano-preparations, it becomes evident that the bioavailability can be significantly improved compared to the original drug [157,159,160,161,162,163] (Table 2).
In addition, there are several relatively novel multifunctional flavonoid nano-preparations, which are primarily used to meet personalized treatment needs and even visual treatment needs. For example, HA-RES-OPC-MMP NPs, a rhythmic flavonoid nano-preparation prepared in response to myocardial ischemia/reperfusion injury (rhythmic disease), has been shown to mitigate the damage during myocardial ischemia/reperfusion by targeting activated rhythm genes, as demonstrated in a pharmacodynamic study in mice [164]. In the case of L-EGCG-Mn, a flavonoid preparation that is both pH-responsive and MRI-imaging, the chelation of Mn2+ by EGCG is greatly reduced in acidic environments, which prompts its release for MRI capability [165]. These preparations, while improving the bioavailability of the original drug, can also be designed into highly effective nano-preparations with targeting, imaging, and even photothermal therapeutic effects, which are effective against a wide range of diseases including antioxidant, antitumor and, anti-inflammatory [166,167,168,169,170,171,172]. The nanoparticle-mediated flavonoid molecular targeting therapy strategy is another research direction with great potential. This technology is considered to overcome the limitations of traditional drugs, such as poor efficacy, insufficient specificity, and high toxicity [173]. The polymeric nanomicelles loaded with quercetin (P-gp inhibitor) and the estrogen receptor antagonist tamoxifen increased the oral bioavailability of quercetin by 2.9 times. Additionally, this formulation demonstrated superior in vivo antitumor efficacy [174]. Currently, a large number of nanoparticle formulations of various flavonoids have entered clinical stages, such as NCT02029352: Sinecatechins for the treatment of skin cancer, NCT03278925: Catechins for the treatment of liver cancer, and NCT01732393: Quercetin for the treatment of tumor inflammation. Of particular interest is the potential of EGCG in cancer therapy, with numerous clinical records already available, including NCT00917735, NCT02580279, and NCT02891538 [175].
Table 2. Nano preparation for improving bioavailability of flavonoids.
Table 2. Nano preparation for improving bioavailability of flavonoids.
TypeSubstanceNameAdministrationDoseAnimal/Cell LineDiseasePharmacokinetics (Compared with Pure Substances)Ref.
NanosuspensionsBaicalinBG-NS and BG-MSOral administration85, 170, 340 and 680 mg/kgWistar rats---AUC(0–t) increased by about 2.22 times and 1.37 times respectively; Vz/F reduced by about 2.27 times and 1.28 times respectively…[176]
LuteolinSLNCOral gavage20 mg/kgMale Sprague Dawley rats---Bioavailability increased by about 3.48 times[177]
DaidzeinF-A and F-BCo-incubation50–400 μMRG2Brain glioma---[178]
NaringeninTPNSOral administration30 mg/kgMale Sprague Dawley rats---Cmax increased by about 2.1 times, AUMC0–∞ increased by about 3.76 times…[179]
SilybinSPCs-NPsOral gavage50 mg/kgMale Sprague Dawley ratsLiver protectionAUC0–∞ increased by about 124.70 times; CL reduced by about 124.58 times; Tmax increased by about 8.82 times…[180]
KaempferolTPGS-KAE-NSpsOral gavage and vein injection15 mg/kg, ig;
5 mg/kg, iv
Female Balb/c miceBreast cancerCmax increased by about 2.41 times; AUC0–t increased by about 4.83 times, T1/2 increased by about 2.64 times.[181]
Hydroxy genkwaninHGK-NSpsvein injection10, 20, 40 mg/kgFemale NU/NU nude miceBreast cancer---[182]
NaringeninNRG-NSOral administration20 mg/kgFemale Wistar rats---Cmax increased by about 2 times; AUC0–24 h increased by about 1.8 times;[183]
BaicalinBCA-NS/NCCSOral administration10 mg/mLMale Wister rats---AUC(0–24) increased by 1.85 times; Cmax increased by about 1.90 times…[184]
LiposomesQuercetinQuercetin liposomesCo-incubation
---RBL-2H3AllergicAnti-allergic activity is higher than that of raw drug[185]
Fisetin---Intraperitoneal injection21 mg/kgC57BL/6J miceLung cancerRelative bioavailability increased by about 47 times[186]
RutinMP-LROral administration16.15 mgC57 BL/6N miceObesityThe RQ-AUC value (the lower the ratio indicates better fatty acid metabolism) is significantly lower than the raw material drug (p < 0.05)[187]
RutinRGD-RUT-LIPO and ABX-RUT-LIPOTail vein injection5 mg/kg (Rutin equivalent)Male Sprague Dawley ratsThrombusShortened clotting time; Relative bioavailability increased by about 3 times.[188]
Licochalcone ALCA-LiposomesOral administration30, 60 mg/kgMale Sprague Dawley ratsRenal injuryAUC0–24 increased by about 2.86 times; Cmax increased by about 2.49 times; MRT0–t increased by about 1.26 times;[189]
Solid lipid nanoparticlesMorin hydrateMSNOral gavage50 mg/kgMale Sprague Dawley ratsCervical cancerCmax increased by about 2.95 times; AUC increased by about 3.10 times;MRT increased by about 2.04 times;[190]
Hydroxysafflor yellow AHSYA SLNOral administration20 mg/kgMale Sprague Dawley ratsNerve injuryCmax increased by about 7.76 times; AUC increased by about 3.99 times; Oral absorption in rats increased by about 3.97 times.[191]
BaicalinOX26-PEG-CSLNvein injection4.42 mg/kgMale Sprague Dawley ratsCerebral ischemia reperfusion injuryAUC increased by about 5.69 times; Cmax increased by about 6.84 times;[192]
NaringeninNrg-SLNsOral administration6 mg/mLWistar ratsinflammation; AntioxidantAUC0→∞ increased by about 17.44 times; MRT0→∞ increased by about 8.81 times; The overall bioavailability is increased by about 12 times;[193]
NaringeninNRG-SLNsintratracheal instillation20 mg/kgMale Sprague Dawley rats---Cmax increased by about 1.62 times; AUC0→∞ increased by about 3.66 times; MRT increased by about 3.33 times; Relative bioavailability increased by about 2.53 times;[194]
PuerarinPue-SLNOral gavage20 mg/kgSprague Dawley rats---AUC0→∞ increased by about 3.00 times; MRT increased by about 1.79 times; CL reduced by about 3.21 times…[195]
NanoemulsionsBreviscapine---Oral administration---Male Wister rats---Cmax increased by about 2.87 times; AUC(0–t) ncreased by about 2.57 times; Relative bioavailability reached 249.70%;[196]
BaicaleinBCL-NEsOral gavage25 mg/kgSprague Dawley rats---AUC0–t increased by about 5.25 times; Cmax increased by about 7.66 times; Relative bioavailability reached 524.7%[197]
Acetylpuerarin---Oral administration30 mg/kgWistar ratsCerebral ischemic reperfusion injuryCmax increased by about 2.89 times; AUC0–t increased by about 2.57 times;[198]
LuteolinNECh-LUTintranasal administration32 μg/kgMale Wistar ratsNeuroblastomaAUC0-∞increased by about 4.40 times; T1/2 increased by about 10 times;[199]
Fisetin---intraperitoneal injection112.5 mg/kgC57BL/6J miceLung cancerRelative bioavailability increased by about 24 times[200]
IsoliquiritigeninISL-NEOcular instillation50 μL 0.2% (w/v)Male New Zealand white rabbitsCorneal neovascularizaionCmax of tear, cornea, Conjunctiva Aqueous humor increased by about 8.70, 3.95, 298.75 and 1.88 times respectively; AUC0–8 h increased by about 5.76, 7.80, 356.57 and 2.13 times respectively…[201]
HydrogelCatechinCA-NG4Transdermal administration5 mg (CA equivalent)male Wistar ratsAntioxidantAUC0–∞increased by about 10.33 times; Relative bioavailability increased to 894.73%[202]
Metal-organic frameworksBaicalinPEG-FA@ZIF-8@BANvein injection---female BALB/c miceBreast cancer Stronger tumor suppressor effect. [203]
NanoparticlesEpigallocatechin gallateCE-HK NPIntratumoral injection20 mg/kg and 40 mg/kgMale BALB/c NudeLiver cancerTumor suppression effect increased by about 2.77 times.[204]
Metal nanoparticlesHesperetinAu-mPEG(5000)-S-HP NPsIntraperitoneal injection(IP)1.5 mg/0.5 mLMale, Wistar strain albino ratsLiver cancer---[205]
Magnetic nanoparticlesQuercetinFe3O4@PCA-PEG-FACo-incubation
50, 100, 200 μg/mLMDA-MB-231 and HeLa------[206]
NABreviscapineBVP-NS, BVP-LP, BVP-PLCOral gavage20 mg/kgMale Sprague-Dawley rats---The relative bioavailability increased to 245.97%, 237.51%, and 471.32, respectively;[207]
NanogelBreviscapineBRE-NGintranasal administration3, 10, 50, 100 mg/mLMale Sprague-Dawley ratsCerebral ischemia reperfusion injuryAbsolute bioavailability increased by about 142.80 times[208]

6.2. Structural Modification

Structural modification of flavonoids is an important approach to promote their development in the field of medicine. Modern research shows that altering the skeleton structure of flavonoids has a significant impact on their absorption, metabolism, and distribution. Common modification methods include chemical modification, microbial methods, and enzymatic methods [209]. Generally speaking, chemical modification is the most direct and simple modification method, while the other two methods are more commonly used in large-scale industrial production. Chemical modification aims to replace selected chemical parts with other functional groups to achieve more desirable properties. As a key strategy to improve the bioavailability of natural products or drugs, improve the pharmacological effects and pharmacokinetic properties, it has broad application prospects. By performing various chemical modifications on flavonoids, their solubility, stability, and bioactivity can be optimized, thereby enhancing the efficacy of these compounds and expanding their range of applications.

6.2.1. Acetylation

The bioavailability of flavonoids is closely related to their lipophilicity, and the methylene groups in acetamide derivatives result in increased lipophilicity of the compounds, thus improving their bioavailability. Isika et al. [210] converted all hydroxyl groups in the structures of quercetin, apigenin, and luteolin into acetamide groups and evaluated the bioavailability of both original substances and their acetylated derivatives (quercetin tetra acetamide, apigenin di acetamide, and luteolin tri acetamide) in vitro. The results showed that acetylated flavonoids had higher bioavailability compared with unmodified compounds. Of course, this could all be attributed to the presence of methylene and amide groups [211]. In addition, the researchers synthesized acetylated arbutin [212], epigallocatechin-3-gallate [213], and acylated puerarin (puerarin esters) [214], in order to improve the lipid solubility of the drugs. In particular, the pharmacokinetics of acylated puerarin were investigated in SD rats. The results clearly showed that esters can improve the bioavailability of the original drug puerarin [215]. This is because enhanced lipid solubility allows these compounds to more easily penetrate the lipid bilayer of the cell membrane [216]. However, it is still important to note that acetylated flavonoids may exhibit certain changes in bioactivity. For example, acetylated flavonoid glycosides reduce the original activity against diet-induced obesity (DIO) and hepatic steatosis [217].

6.2.2. Glycosylation

Most natural flavonoids exist in the form of glycosides. Aglycones (flavonoids) are combined with glycone groups through glycosidic bonds to form flavonoid glycosides. Most flavonoid glycosides are metabolized in the body into corresponding aglycones with smaller analytical structures before they are absorbed. Therefore, some opinions think that the bioavailability of flavonoid aglycones is always higher than that of their corresponding glycosides. However, a previous study found that the absorption efficiency of quercetin glucoside was higher than that of its aglycone form [148]. Other studies also have reported that glucosylation may increase the bioavailability of flavonoids such as anthocyanins [218]. Of course, the impact of glycosylation on the bioavailability of flavonoids cannot be determined solely based on individual studies. The type, amount and nature of glycosidic bond may all have a greater impact on the bioavailability of flavonoids [219].

6.2.3. Methyl Etherification

Flavonoids such as EGCG have multiple phenolic hydroxyl groups. A large number of active phenolic hydroxyl groups contribute to their drug activity but are also the culprits for their reduced bioavailability [220]. For the structural modification of EGCG, the methyl etherification pathway was used to achieve higher bioavailability by converting some or all of the eight active phenolic hydroxyl groups in the A, B, and D rings into methyl ether derivatives [221,222,223]. It is worth noting that methyl etherification may not only have beneficial effects on the human body but also play a certain role in protecting plants against pests and diseases. For example, methyl etherified naringenin has been found to be used to fight fungal pathogens that can infect corn [224].

6.2.4. Esterification

Esterification of the carboxyl groups of flavonoids to increase their lipid solubility and enhance their affinity for cell membranes is an effective method to improve the bioavailability of flavonoids. The researchers prepared a series of baicalin ester derivatives by adding fatty alcohols to baicalin in a non-aqueous medium for the esterification reaction. Interestingly, the esterified baicalin not only exhibits stronger antibacterial activity, but also increase the lipophilicity of the original drug. Of course, the researchers also pointed out that fatty alcohol chains that are too long may cause excessive lipophilic activity and form extracellular aggregates, thereby reducing their activity [225]. In addition, the researchers also synthesized baicalin ester derivatives with different fatty acid chain lengths through a whole-cell catalytic esterification reaction and evaluated the absorption of baicalin esters compared to baicalin using a Caco-2 cell model. The results showed that the absorption efficiency of baicalin ester was much greater than that of baicalin, with a maximum difference of up to 10 times [226].

6.2.5. Acylation

Due to the poor stability and low solubility of some flavonoids in lipids, acylation modification has become the main means to improve their bioactivity. Compared with unacylated flavonoids, acylated flavonoids have greatly improved their activity. Phloridzin docosahexaenoate (PZ-DHA) is an omega-3 fatty acid ester of a flavonoid precursor. Recent research has shown that PZ-DHA, in contrast to pure Phloridzin, demonstrates significant antitumor metastatic activity. This is achieved through the inhibition of angiogenesis both in vitro and in vivo, as well as by reducing the proliferation and migration of human umbilical vein endothelial cells (HUVECs) (p < 0.01) and human microvascular venous endothelial cells (HMVECs) (p < 0.05) [227]. Moreover, PZ-DHA currently demonstrates significant advantages in the treatment of skin cancer, triple-negative breast cancer, overcoming tumor resistance in triple-negative breast cancer, and combating leukemia [227,228,229,230]. In conclusion, acylation increases the lipophilicity of flavonoids, making them more easily absorbed by cells, and acylated flavonoids have demonstrated significantly greater pharmacological activity in antiviral, antioxidant, anti-inflammatory, antimicrobial, anticancer and other. However, some studies have pointed out that high doses of acylated quercitrin, compared to its non-acylated form, may even exacerbate cellular oxidative stress. This suggests that precise dosage control is essential, especially when considering flavonoid compounds modified by acylation [231].

7. Prospect

Despite showing significant bioactivity in vitro, the bioavailability of flavonoids in vivo remains a major bottleneck that limits their widespread application [232]. To address this issue, future research may explore various approaches, including optimizing drug delivery technologies, chemically modifying flavonoid molecules, and examining the influence of gut microbiome, among others.
With advances in nanotechnology, delivery systems for flavonoids are undergoing rapid innovation. Techniques such as nanoparticles, liposomes, and solid dispersions have significantly enhanced their bioavailability [233]. Research indicates that carriers like nanoparticles and liposomes can effectively protect flavonoids from degradation in the digestive system and enhance their therapeutic efficacy through targeted delivery mechanisms [234]. Furthermore, the chemical modification of flavonoid compounds is a significant strategy for enhancing their bioavailability. Structural alterations, such as introducing new functional groups to the flavonoid molecule, can improve solubility, stability, and lipophilicity, thereby facilitating absorption and distribution [235]. Future integration of efficient molecular design and synthesis methods may significantly enhance the bioavailability of flavonoid compounds. Moreover, recent studies have shown that the gut microbiome plays a crucial role in the metabolism of flavonoid compounds. The gut microbiome not only generates bioactive metabolites from flavonoid metabolism but also influences their absorption and biological effects within the host organism [127]. This finding offers new insights into personalized nutrition and the application of flavonoid compounds. Future research could explore the functions of different gut microbiome communities to further investigate their interactions with flavonoid compounds and develop personalized health management strategies based on microbiome-drug interactions [236]. For example, the probiotic strain Bifidobacterium pseudocatenulatum B7003 not only provides benefits for gut transit but also enhances the antioxidant activity and bioavailability of flavonoids by converting flavonoid glycosides in dairy-like products into their aglycone form [237]. It is worth noting that flavonoids are also considered a nutritional source for probiotics by providing energy and nutrients to promote the growth and reproduction of probiotics [238,239]. The addition of banana peel polyphenol extract to yogurt enhanced the vitality of B. lactis and L. acidophilus strains [240]. Undoubtedly, this strategy holds significant growth potential, as the appeal of a win–win situation is difficult to overlook. The strategy also has been implemented by Shehata et al., who developed functional beverages from taro leaf extract (TLE) and probiotics. This study highlighted that the polyphenols (including flavonoids) in these extracts play a significant role in enhancing the viability and stability of probiotics. Research has demonstrated that functional beverages are capable of maintaining probiotic concentrations exceeding 7.00 log cfu/mL after 30 days of storage, thereby supporting essential health-promoting functions. Additionally, the presence of probiotics has been shown to enhance the antioxidant capacity of polyphenolic compounds [241]. In the future, advancements in gut health, immune function, cardiovascular health, and so on, may all be positively influenced by ongoing research into the combination of flavonoids and probiotics [242,243,244,245].

8. Conclusions

This review comprehensively explores the multifaceted nature of botanical flavonoids, emphasizing their diverse bioactivities and the challenges associated with their low bioavailability. We highlight the critical role of gut microbiome in the metabolism and bioactivation of flavonoids, revealing how microbial modifications can enhance their therapeutic potential. Furthermore, it delves into innovative strategies such as nano-formulations and chemical modifications (e.g., acylation, glycosylation) to improve the bioavailability and bioactivity of flavonoids. These approaches offer new avenues for optimizing the pharmacokinetics and therapeutic efficacy of these natural compounds. We underscore the need for future research to focus on advanced delivery systems and structural modifications, aiming to translate the promising in vitro findings of flavonoids into clinical and health applications. By addressing the bioavailability bottleneck and leveraging the interactions between flavonoids and the gut microbiome, this work paves the way for personalized nutrition and therapeutic strategies, potentially impacting gut health, immune function, and cardiovascular health.

Author Contributions

L.H.: Writing-original draft, Investigation. Y.L.: Investigation, Supervision. J.Y.: Funding acquisition. C.C.: Investigation, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from National Natural Science Foundation of China (No.82260746). The authors gratefully acknowledge financial supports from Jiangxi Province Double Thousand Talent-Leader of Natural Science Project (jxsq2023101038), Jiangxi Province Urgently Overseas Talent Project (2022BCJ25027 & 2023BBG70014), Natural Science Foundation of Jiujiang (S2024KXJJ0001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data was used for the research described in the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AKT: Protein kinase B; AP1: Activator protein 1; BCRP: Breast cancer resistance protein; CAT: Catalase; CBG: Cytosolic β-glucosidase; Cd: Cadmium; c-MET: Cellular-mesenchymal epithelial transition factor; C-Myc: Cellular Myelocytomatosis oncogene Gene; cGAS-STING: Cyclic GMP-AMP synthase-stimulator of interferon genes; COX-2: Cyclooxygenase-2; csgA/B: Curli subunits A/B; DIO: Diet-induced obesity; EGCG: Epigallocatechin gallate; EGFR: Epidermal growth factor receptor; EHEC: Escherichia coli; EpRE: Electrophile-responsive element; ERK1/2: extracellular regulated protein kinases1/2; FLR: flavonoid reductase; GLUT-2: Glucose transporter 2; GSH-Px: Glutathione peroxidase; HO-1: Heme oxygenase 1 LDL: Low-density lipoprotein; IL-6: Interleukin-6; IL-1β: Interleukin-1β; IL-17: Interleukin-17; IKK: IκB kinase; iNOS: Inducible nitric oxide synthase; LPH: Lactase phlorizin hydrolase; MAPK/Erk: Mitogen-activated protein kinase/Extracellular regulated protein kinases; MITF: Microphthalmia transcription factor; NADH: Nicotinamide adenine dinucleotide; NADPH: Nicotinamide adenine dinucleotide phosphate; NF-κB: Nuclear factor κB; NOTCH: Notch Receptor; NOX: Nicotinamide adenine dinucleotide phosphate oxidase; Nrf2: Nuclear factor erythroid 2-related factor 2; PDGF: Platelet-derived growth factor; Perk: Phospho-ERK; PGE2: Prostaglandin E2; PLC/PKC: Phosphoinositide-specific Phospholipase C/Protein kinase C; PI3K: Phosphoinositide 3-Kinase; pJNK: Phospho c-Jun N-terminal kinase; P-P38: Phospho P38 mitogen-activated protein kinase; Rac1: Ras-related C3 botulinum toxin substrate 1; Raf: Rapidly Accelerated Fibrosarcoma; Ras: Rat Sarcoma; RNS: Reactive nitrogen species; ROS: Reactive oxygen species; SOD: Superoxide dismutase; SGLT-1: Sodium-dependent glucose transporter 1; TAX: Taxifolin; TF3: Theaflavin-3,3′-digallate; TLR: Toll-like receptors; TNF-α: Tumour necrosis factor-α; VEGF: Vascular Endothelial Growth Factors; YAP/TEAD: Yes-associated protein/Transcriptional Enhanced Associate Domains.

References

  1. Nabavi, S.M.; Šamec, D.; Tomczyk, M.; Milella, L.; Russo, D.; Habtemariam, S.; Suntar, I.; Rastrelli, L.; Daglia, M.; Xiao, J.; et al. Flavonoid biosynthetic pathways in plants: Versatile targets for metabolic engineering. Biotechnol. Adv. 2020, 38, 107316. [Google Scholar] [CrossRef] [PubMed]
  2. Hasnat, H.; Shompa, S.A.; Islam, M.M.; Alam, S.; Richi, F.T.; Emon, N.U.; Ashrafi, S.; Ahmed, N.U.; Chowdhury, M.N.R.; Fatema, N.; et al. Flavonoids: A treasure house of prospective pharmacological potentials. Heliyon 2024, 10, e27533. [Google Scholar] [CrossRef]
  3. Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. 2017, 8, 423–435. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, M.; Zhang, Z.; Zhu, M.; Liu, K.; Farag, M.A.; Song, L.; Gao, F.; Tao, H. Biofortification of flavonoids in nuts along the agro-food chain for improved nutritional and health benefits, a comprehensive review and future prespectives. Food Chem. 2025, 464, 141754. [Google Scholar] [CrossRef] [PubMed]
  5. Xie, L.; Deng, Z.; Zhang, J.; Dong, H.; Wang, W.; Xing, B.; Liu, X. Comparison of Flavonoid O-Glycoside, C-Glycoside and Their Aglycones on Antioxidant Capacity and Metabolism during In Vitro Digestion and In Vivo. Foods 2022, 11, 882. [Google Scholar] [CrossRef]
  6. Maaiden, E.E.; Ullah, N.; Ezzariai, A.; Mazar, A.; Boukcim, H.; Hirich, A.; Nasser, B.; Qarah, N.; Kouisni, L.; Kharrassi, Y.E. Comparing antioxidant and cytoprotective effects: Quercetin glycoside vs. aglycone from Ephedra alata. Phytomedicine Plus 2024, 4, 100603. [Google Scholar] [CrossRef]
  7. Bartnik, M.; Facey, P.C. Chapter 8—Glycosides. In Pharmacognosy; Badal, S., Delgoda, R., Eds.; Academic Press: Boston, MA, USA, 2017; pp. 101–161. [Google Scholar]
  8. Xiao, J. Dietary flavonoid aglycones and their glycosides: Which show better biological significance? Crit. Rev. Food Sci. Nutr. 2017, 57, 1874–1905. [Google Scholar] [CrossRef]
  9. Sunil, L.; Shetty, N.P. Biosynthesis and regulation of anthocyanin pathway genes. Appl. Microbiol. Biotechnol. 2022, 106, 1783–1798. [Google Scholar] [CrossRef]
  10. Valenta, K.; Nevo, O.; Martel, C.; Chapman, C.A. Plant attractants: Integrating insights from pollination and seed dispersal ecology. Evol. Ecol. 2017, 31, 249–267. [Google Scholar] [CrossRef]
  11. Golovatskaya, I.F.; Medvedeva, Y.V.; Kadyrbaev, M.K.; Boyko, E.V. Specificity of Growth and Accumulation of Flavonoids in Plants and Cell Cultures of Lychnis chalcedonica Obtained from Explants of Different Organs. Russ. J. Plant Physiol. 2024, 71, 24. [Google Scholar] [CrossRef]
  12. Patil, J.R.; Mhatre, K.J.; Yadav, K.; Yadav, L.S.; Srivastava, S.; Nikalje, G.C. Flavonoids in plant-environment interactions and stress responses. Discov. Plants 2024, 1, 68. [Google Scholar] [CrossRef]
  13. Mierziak, J.; Kostyn, K.; Kulma, A. Flavonoids as important molecules of plant interactions with the environment. Molecules 2014, 19, 16240–16265. [Google Scholar] [CrossRef]
  14. Das, A.; Choudhury, S.; Gopinath, V.; Majeed, W.; Chakraborty, S.; Bhairavi, K.S.; Chowdhury, S.; Dubey, V.K.; Akhtar, M.S. Functions of Flavonoids in Plant, Pathogen, and Opportunistic Fungal Interactions. In Opportunistic Fungi, Nematode and Plant Interactions: Interplay and Mechanisms; Akhtar, M.S., Ed.; Springer Nature: Singapore, 2024; pp. 91–123. [Google Scholar]
  15. Zheng, X.; Zhang, X.; Zeng, F. Biological Functions and Health Benefits of Flavonoids in Fruits and Vegetables: A Contemporary Review. Foods 2025, 14, 155. [Google Scholar] [CrossRef]
  16. Li, B.; Fan, R.; Sun, G.; Sun, T.; Fan, Y.; Bai, S.; Guo, S.; Huang, S.; Liu, J.; Zhang, H.; et al. Flavonoids improve drought tolerance of maize seedlings by regulating the homeostasis of reactive oxygen species. Plant Soil 2021, 461, 389–405. [Google Scholar] [CrossRef]
  17. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef]
  18. Cassidy, A.; Minihane, A.-M. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am. J. Clin. Nutr. 2017, 105, 10–22. [Google Scholar] [CrossRef]
  19. Wang, T.Y.; Li, Q.; Bi, K.S. Bioactive flavonoids in medicinal plants: Structure, activity and biological fate. Asian J. Pharm. Sci. 2018, 13, 12–23. [Google Scholar] [CrossRef]
  20. Wang, J.; Chen, C.; Guo, Q.; Gu, Y.; Shi, T.Q. Advances in Flavonoid and Derivative Biosynthesis: Systematic Strategies for the Construction of Yeast Cell Factories. ACS Synth. Biol. 2024, 13, 2667–2683. [Google Scholar] [CrossRef]
  21. Agati, G.; Azzarello, E.; Pollastri, S.; Tattini, M. Flavonoids as antioxidants in plants: Location and functional significance. Plant Sci. Int. J. Exp. Plant Biol. 2012, 196, 67–76. [Google Scholar] [CrossRef]
  22. Shen, N.; Wang, T.; Gan, Q.; Liu, S.; Wang, L.; Jin, B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022, 383, 132531. [Google Scholar] [CrossRef]
  23. Rudrapal, M.; Khan, J.; Dukhyil, A.A.B.; Alarousy, R.; Attah, E.I.; Sharma, T.; Khairnar, S.J.; Bendale, A.R. Chalcone Scaffolds, Bioprecursors of Flavonoids: Chemistry, Bioactivities, and Pharmacokinetics. Molecules 2021, 26, 7177. [Google Scholar] [CrossRef]
  24. Cruz, L.; Basílio, N.; Mateus, N.; de Freitas, V.; Pina, F. Natural and Synthetic Flavylium-Based Dyes: The Chemistry Behind the Color. Chem. Rev. 2022, 122, 1416–1481. [Google Scholar] [CrossRef]
  25. Tena, N.; Martín, J.; Asuero, A.G. State of the Art of Anthocyanins: Antioxidant Activity, Sources, Bioavailability, and Therapeutic Effect in Human Health. Antioxidants 2020, 9, 451. [Google Scholar] [CrossRef]
  26. Wang, M.; Firrman, J.; Liu, L.; Yam, K. A Review on Flavonoid Apigenin: Dietary Intake, ADME, Antimicrobial Effects, and Interactions with Human Gut Microbiota. BioMed Res. Int. 2019, 2019, 7010467. [Google Scholar] [CrossRef]
  27. Osigwe, C.; Akah, P.; Nworu, C.; Okoye, F. Apigenin: A methanol fraction component of Newbouldia laevis leaf, as a potential antidiabetic agent. J. Phytopharm. 2017, 6, 38–44. [Google Scholar] [CrossRef]
  28. Ginwala, R.; Bhavsar, R.; Chigbu, D.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef]
  29. Li, S.-G.; Yang, R.; Lu, M.-M.; Wang, S.-M.; Meng, J. A new isoflavone from processed root barks of Paeonia suffruticosa and their procoagulant activity. Nat. Prod. Res. 2024, 11, 1–7. [Google Scholar] [CrossRef]
  30. Li, P.; Ding, W.; Chen, F.; Zhou, F.; Ruan, Z.; Li, J.; Wu, Y. Isoflavones relieve intestinal motility disorders in colitis rats by regulating 5-hydroxytryptamine and interstitial cells of Cajal. Food Biosci. 2024, 63, 105587. [Google Scholar] [CrossRef]
  31. Selepe, M.A. Isoflavone Derivatives as Potential Anticancer Agents: Synthesis and Bioactivity Studies. ChemMedChem 2024, 19, e202400420. [Google Scholar] [CrossRef]
  32. Kozłowska, A.; Szostak-Węgierek, D. Targeting Cardiovascular Diseases by Flavonols: An Update. Nutrients 2022, 14, 1439. [Google Scholar] [CrossRef]
  33. Felice, M.R.; Maugeri, A.; De Sarro, G.; Navarra, M.; Barreca, D. Molecular Pathways Involved in the Anti-Cancer Activity of Flavonols: A Focus on Myricetin and Kaempferol. Int. J. Mol. Sci. 2022, 23, 4411. [Google Scholar] [CrossRef]
  34. Dajas, F.; Andrés, A.C.; Florencia, A.; Carolina, E.; Felicia, R.M. Neuroprotective actions of flavones and flavonols: Mechanisms and relationship to flavonoid structural features. Cent. Nerv. Syst. Agents Med. Chem. 2013, 13, 30–35. [Google Scholar] [CrossRef]
  35. Duan, Y.; Eduardo Melo Santiago, F.; Rodrigues Dos Reis, A.; de Figueiredo, M.A.; Zhou, S.; Thannhauser, T.W.; Li, L. Genotypic variation of flavonols and antioxidant capacity in broccoli. Food Chem. 2021, 338, 127997. [Google Scholar] [CrossRef]
  36. Yang, C.P.; Shie, P.H.; Huang, G.J.; Chien, S.C.; Kuo, Y.H. New Anti-inflammatory Flavonol Glycosides from Lindera akoensis Hayata. Molecules 2019, 24, 563. [Google Scholar] [CrossRef]
  37. Lea, M.A. Flavonol regulation in tumor cells. J. Cell. Biochem. 2015, 116, 1190–1194. [Google Scholar] [CrossRef]
  38. Wisetsai, A.; Choodej, S.; Ngamrojanavanich, N.; Pudhom, K. Fatty acid acylated flavonol glycosides from the seeds of Nephelium lappaceum and their nitric oxide suppression activity. Phytochemistry 2022, 201, 113262. [Google Scholar] [CrossRef]
  39. Motallebi, M.; Bhia, M.; Rajani, H.F.; Bhia, I.; Tabarraei, H.; Mohammadkhani, N.; Pereira-Silva, M.; Kasaii, M.S.; Nouri-Majd, S.; Mueller, A.L.; et al. Naringenin: A potential flavonoid phytochemical for cancer therapy. Life Sci. 2022, 305, 120752. [Google Scholar] [CrossRef]
  40. Cai, J.; Wen, H.; Zhou, H.; Zhang, D.; Lan, D.; Liu, S.; Li, C.; Dai, X.; Song, T.; Wang, X.; et al. Naringenin: A flavanone with anti-inflammatory and anti-infective properties. Biomed. Pharmacother. = Biomed. Pharmacother. 2023, 164, 114990. [Google Scholar] [CrossRef]
  41. Tutunchi, H.; Naeini, F.; Ostadrahimi, A.; Hosseinzadeh-Attar, M.J. Naringenin, a flavanone with antiviral and anti-inflammatory effects: A promising treatment strategy against COVID-19. Phytother. Res. PTR 2020, 34, 3137–3147. [Google Scholar] [CrossRef]
  42. Zhou, K.; Yang, S.; Li, S.M. Naturally occurring prenylated chalcones from plants: Structural diversity, distribution, activities and biosynthesis. Nat. Prod. Rep. 2021, 38, 2236–2260. [Google Scholar] [CrossRef]
  43. Tuli, H.S.; Aggarwal, V.; Parashar, G.; Aggarwal, D.; Parashar, N.C.; Tuorkey, M.J.; Varol, M.; Sak, K.; Kumar, M.; Buttar, H.S. Xanthohumol: A Metabolite with Promising Anti-Neoplastic Potential. Anti-Cancer Agents Med. Chem. 2022, 22, 418–432. [Google Scholar] [CrossRef]
  44. Sun, W.; Yue, J.; Xu, T.; Cui, Y.; Huang, D.; Shi, H.; Xiong, J.; Sun, W.; Yi, Q. Xanthohumol alleviates palmitate-induced inflammation and prevents osteoarthritis progression by attenuating mitochondria dysfunction/NLRP3 inflammasome axis. Heliyon 2023, 9, e21282. [Google Scholar] [CrossRef]
  45. Lee, Y.M.; Yoon, Y.; Yoon, H.; Park, H.M.; Song, S.; Yeum, K.J. Dietary Anthocyanins against Obesity and Inflammation. Nutrients 2017, 9, 1089. [Google Scholar] [CrossRef]
  46. Mohammadi, N.; Farrell, M.; O'Sullivan, L.; Langan, A.; Franchin, M.; Azevedo, L.; Granato, D. Effectiveness of anthocyanin-containing foods and nutraceuticals in mitigating oxidative stress, inflammation, and cardiovascular health-related biomarkers: A systematic review of animal and human interventions. Food Funct. 2024, 15, 3274–3299. [Google Scholar] [CrossRef]
  47. Sies, H. Oxidative Stress; Academic Press Inc.: Cambridge, MA, USA, 1985. [Google Scholar]
  48. Del Río, L.A. ROS and RNS in plant physiology: An overview. J. Exp. Bot. 2015, 66, 2827–2837. [Google Scholar] [CrossRef]
  49. Jones, D.P. Radical-free biology of oxidative stress. Am. J. Physiol. Cell Physiol. 2008, 295, C849–C868. [Google Scholar] [CrossRef]
  50. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  51. Cai, Y.Z.; Mei, S.; Jie, X.; Luo, Q.; Corke, H. Structure-radical scavenging activity relationships of phenolic compounds from traditional Chinese medicinal plants. Life Sci. 2006, 78, 2872–2888. [Google Scholar] [CrossRef]
  52. Sarian, M.N.; Ahmed, Q.U.; Mat So’ad, S.Z.; Alhassan, A.M.; Murugesu, S.; Perumal, V.; Syed Mohamad, S.N.A.; Khatib, A.; Latip, J. Antioxidant and Antidiabetic Effects of Flavonoids: A Structure-Activity Relationship Based Study. BioMed Res. Int. 2017, 2017, 8386065. [Google Scholar] [CrossRef]
  53. Zuo, A.R.; Dong, H.H.; Yu, Y.Y.; Shu, Q.L.; Zheng, L.X.; Yu, X.Y.; Cao, S.W. The antityrosinase and antioxidant activities of flavonoids dominated by the number and location of phenolic hydroxyl groups. Chin. Med. 2018, 13, 51. [Google Scholar] [CrossRef]
  54. Procházková, D.; Boušová, I.; Wilhelmová, N. Antioxidant and prooxidant properties of flavonoids. Fitoterapia 2011, 82, 513–523. [Google Scholar] [CrossRef] [PubMed]
  55. de Araújo, G.R.; Rabelo, A.C.; Meira, J.S.; Rossoni-Júnior, J.V.; Castro-Borges, W.; Guerra-Sá, R.; Batista, M.A.; Silveira-Lemos, D.D.; Souza, G.H.; Brandão, G.C.; et al. Baccharis trimera inhibits reactive oxygen species production through PKC and down-regulation p47 (phox) phosphorylation of NADPH oxidase in SK Hep-1 cells. Exp. Biol. Med. 2017, 242, 333–343. [Google Scholar] [CrossRef]
  56. Chlebda, E.; Magdalan, J.; Merwid-Ląd, A.; Trocha, M.; Kopacz, M.; Kuźniar, A.; Nowak, D.; Szeląg, A. Influence of water-soluble flavonoids, quercetin-5′-sulfonic acid sodium salt and morin-5′-sulfonic acid sodium salt, on antioxidant parameters in the subacute cadmium intoxication mouse model. Exp. Toxicol. Pathol. 2010, 62, 105–108. [Google Scholar] [CrossRef]
  57. Zhang, S.; Li, L.; Chen, W.; Xu, S.; Feng, X.; Zhang, L. Natural products: The role and mechanism in low-density lipoprotein oxidation and atherosclerosis. Phytother. Res. 2021, 35, 2945–2967. [Google Scholar] [CrossRef] [PubMed]
  58. Hanasaki, Y.; Ogawa, S.; Fukui, S. The correlation between active oxygens scavenging and antioxidative effects of flavonoids. Free. Radic. Biol. Med. 1994, 16, 845–850. [Google Scholar] [CrossRef]
  59. Wang, S.; Fang, Y.; Yu, X.; Guo, L.; Zhang, X.; Xia, D. The flavonoid-rich fraction from rhizomes of Smilax glabra Roxb. ameliorates renal oxidative stress and inflammation in uric acid nephropathy rats through promoting uric acid excretion. Biomed. Pharmacother. 2019, 111, 162–168. [Google Scholar] [CrossRef]
  60. Yeh, S.-L.; Wang, W.-Y.; Huang, C.-H.; Hu, M.-L. Pro-oxidative effect of β-carotene and the interaction with flavonoids on UVA-induced DNA strand breaks in mouse fibroblast C3H10T1/2 cells. J. Nutr. Biochem. 2005, 16, 729–735. [Google Scholar] [CrossRef]
  61. Forni, C.; Rossi, M.; Borromeo, I.; Feriotto, G.; Platamone, G.; Tabolacci, C.; Mischiati, C.; Beninati, S. Flavonoids: A Myth or a Reality for Cancer Therapy? Molecules 2021, 26, 3583. [Google Scholar] [CrossRef] [PubMed]
  62. Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef]
  63. Mitra, S.; Nguyen, L.N.; Akter, M.; Park, G.; Choi, E.H.; Kaushik, N.K. Impact of ROS Generated by Chemical, Physical, and Plasma Techniques on Cancer Attenuation. Cancers 2019, 11, 1030. [Google Scholar] [CrossRef]
  64. Slika, H.; Mansour, H.; Wehbe, N.; Nasser, S.A.; Iratni, R.; Nasrallah, G.; Shaito, A.; Ghaddar, T.; Kobeissy, F.; Eid, A.H. Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed. Pharmacother. 2022, 146, 112442. [Google Scholar] [CrossRef] [PubMed]
  65. Roy, P.S.; Saikia, B.J. Cancer and cure: A critical analysis. Indian J. Cancer 2016, 53, 441–442. [Google Scholar] [CrossRef] [PubMed]
  66. Hossain, M.A. Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur. J. Pharmacol. 2024, 979, 176727. [Google Scholar] [PubMed]
  67. Gao, Y.; Hou, J.; Fei, X.; Ren, L.; Lu, R.; Liu, P.; Liu, S.; Zhu, C.; Wang, X.; Pan, Y. TAGLN2 targeted control of ARPC5-mediated activation of the MEK/ERK signaling pathway influences the proliferation, invasion, and metastasis of pancreatic cancer cells. Cell. Signal. 2024, 120, 111227. [Google Scholar] [CrossRef]
  68. Wu, L.; Hu, Z.; Song, X.-F.; Liao, Y.-J.; Xiahou, J.-H.; Li, Y.; Zhang, Z.-H. Targeting Nrf2 signaling pathways in the role of bladder cancer: From signal network to targeted therapy. Biomed. Pharmacother. 2024, 176, 116829. [Google Scholar] [CrossRef]
  69. Liang, J.-L.; Jin, X.-K.; Deng, X.-C.; Huang, Q.-X.; Zhang, S.-M.; Chen, W.-H.; Zhang, X.-Z. Targeting activation of cGAS-STING signaling pathway by engineered biomaterials for enhancing cancer immunotherapy. Mater. Today 2024, 78, 251–296. [Google Scholar] [CrossRef]
  70. Zhou, W.; Zeng, T.; Chen, J.; Tang, X.; Yuan, Y.; Hu, D.; Zhang, Y.; Li, Y.; Zou, J. Aberrant angiogenic signaling pathways: Accomplices in ovarian cancer progression and treatment. Cell. Signal. 2024, 120, 111240. [Google Scholar] [CrossRef]
  71. Khan, I.R.; Sadida, H.Q.; Hashem, S.; Singh, M.; Macha, M.A.; Al-Shabeeb Akil, A.S.; Khurshid, I.; Bhat, A.A. Therapeutic implications of signaling pathways and tumor microenvironment interactions in esophageal cancer. Biomed. Pharmacother. 2024, 176, 116873. [Google Scholar] [CrossRef]
  72. Chu, J.; Yuan, C.; Zhou, L.; Zhao, Y.; Wu, X.; Yan, Y.; Liu, Y.; Liu, X.; Jing, L.; Dong, T.; et al. JianPiTongLuo (JPTL) Recipe regulates anti-apoptosis and cell proliferation in colorectal cancer through the PI3K/AKT signaling pathway. Heliyon 2024, 10, e35490. [Google Scholar] [CrossRef]
  73. Jin, X.; Wang, S.; Luo, L.; Yan, F.; He, Q. Targeting the Wnt/β-catenin signal pathway for the treatment of gastrointestinal cancer: Potential for advancement. Biochem. Pharmacol. 2024, 227, 116463. [Google Scholar] [CrossRef]
  74. Napolitano, S.; Martini, G.; Ciardiello, D.; Del Tufo, S.; Martinelli, E.; Troiani, T.; Ciardiello, F. Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol. Hepatol. 2024, 9, 664–676. [Google Scholar] [CrossRef]
  75. Yang, M.H.; Basappa, B.; Deveshegowda, S.N.; Ravish, A.; Mohan, A.; Nagaraja, O.; Madegowda, M.; Rangappa, K.S.; Deivasigamani, A.; Pandey, V.; et al. A novel drug prejudice scaffold-imidazopyridine-conjugate can promote cell death in a colorectal cancer model by binding to β-catenin and suppressing the Wnt signaling pathway. J. Adv. Res. 2024; in press. [Google Scholar] [CrossRef]
  76. Bharathiraja, P.; Yadav, P.; Sajid, A.; Ambudkar, S.V.; Prasad, N.R. Natural medicinal compounds target signal transduction pathways to overcome ABC drug efflux transporter-mediated multidrug resistance in cancer. Drug Resist. Updates 2023, 71, 101004. [Google Scholar] [CrossRef] [PubMed]
  77. Hemmati-Dinarvand, M.; Ahmadvand, H.; Seghatoleslam, A. Nitazoxanide and Cancer Drug Resistance: Targeting Wnt/β-catenin Signaling Pathway. Arch. Med. Res. 2022, 53, 263–270. [Google Scholar] [CrossRef]
  78. Morris, M.E.; Zhang, S. Flavonoid–drug interactions: Effects of flavonoids on ABC transporters. Life Sci. 2006, 78, 2116–2130. [Google Scholar] [CrossRef]
  79. Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.-H.; Jaremko, M. Important Flavonoids and Their Role as a Therapeutic Agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef] [PubMed]
  80. Xu, L.; Zhang, L.; Zhang, S.; Yang, J.; Zhu, A.; Sun, J.; Kalvakolanu, D.V.; Cong, X.; Zhang, J.; Tang, J.; et al. Taxifolin inhibits melanoma proliferation/migration impeding USP18/Rac1/JNK/β-catenin oncogenic signaling. Phytomedicine 2024, 123, 155199. [Google Scholar] [CrossRef]
  81. Manigandan, K.; Manimaran, D.; Jayaraj, R.L.; Elangovan, N.; Dhivya, V.; Kaphle, A. Taxifolin curbs NF-κB-mediated Wnt/β-catenin signaling via up-regulating Nrf2 pathway in experimental colon carcinogenesis. Biochimie 2015, 119, 103–112. [Google Scholar] [CrossRef]
  82. Wu, S.; Lu, H.; Bai, Y. Nrf2 in cancers: A double-edged sword. Cancer Med. 2019, 8, 2252–2267. [Google Scholar] [CrossRef]
  83. Desai, V.; Jain, A.; Shaghaghi, H.; Summer, R.; Lai, J.C.K.; Bhushan, A. Combination of Biochanin A and Temozolomide Impairs Tumor Growth by Modulating Cell Metabolism in Glioblastoma Multiforme. Anticancer. Res. 2019, 39, 57–66. [Google Scholar] [CrossRef]
  84. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From inflammation to cancer. Ir. J. Med. Sci. 2017, 186, 57–62. [Google Scholar] [CrossRef]
  85. Fuster, J.J.; Zuriaga, M.A.; Fuster, V. Inflammation as a Driver of Disease. In Encyclopedia of Cell Biology, 2nd ed.; Bradshaw, R.A., Hart, G.W., Stahl, P.D., Eds.; Academic Press: Oxford, UK, 2023; pp. 495–501. [Google Scholar]
  86. Xiao, X.; Shi, D.; Liu, L.; Wang, J.; Xie, X.; Kang, T.; Deng, W. Quercetin suppresses cyclooxygenase-2 expression and angiogenesis through inactivation of P300 signaling. PLoS ONE 2011, 6, e22934. [Google Scholar] [CrossRef]
  87. Murata, T.; Ishiwa, S.; Lin, X.; Nakazawa, Y.; Tago, K.; Funakoshi-Tago, M. The citrus flavonoid, nobiletin inhibits neuronal inflammation by preventing the activation of NF-κB. Neurochem. Int. 2023, 171, 105613. [Google Scholar] [CrossRef] [PubMed]
  88. Zulkefli, N.; Che Zahari, C.N.M.; Sayuti, N.H.; Kamarudin, A.A.; Saad, N.; Hamezah, H.S.; Bunawan, H.; Baharum, S.N.; Mediani, A.; Ahmed, Q.U.; et al. Flavonoids as Potential Wound-Healing Molecules: Emphasis on Pathways Perspective. Int. J. Mol. Sci. 2023, 24, 4607. [Google Scholar] [CrossRef]
  89. Atreya, I.; Atreya, R.; Neurath, M.F. NF-kappaB in inflammatory bowel disease. J. Intern. Med. 2008, 263, 591–596. [Google Scholar] [CrossRef]
  90. Kubatka, P.; Mazurakova, A.; Samec, M.; Koklesova, L.; Zhai, K.; Al-Ishaq, R.; Kajo, K.; Biringer, K.; Vybohova, D.; Brockmueller, A.; et al. Flavonoids against non-physiologic inflammation attributed to cancer initiation, development, and progression-3PM pathways. EPMA J. 2021, 12, 559–587. [Google Scholar] [CrossRef] [PubMed]
  91. Van der Heiden, K.; Cuhlmann, S.; Luong Le, A.; Zakkar, M.; Evans, P.C. Role of nuclear factor kappaB in cardiovascular health and disease. Clin. Sci. 2010, 118, 593–605. [Google Scholar] [CrossRef]
  92. Karin, M.; Greten, F.R. NF-κB: Linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol. 2005, 5, 749–759. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, Y.; Ding, Q.; Xie, Y.; Zhang, Q.; Zhou, Y.; Sun, H.; Qian, R.; Wang, L.; Chen, X.; Gao, Y.; et al. Green tea polyphenol alleviates silica particle-induced lung injury by suppressing IL-17/NF-κB p65 signaling-driven inflammation. Phytomedicine 2024, 135, 156238. [Google Scholar] [CrossRef]
  94. Wu, D.-G.; Yu, P.; Li, J.-W.; Jiang, P.; Sun, J.; Wang, H.-Z.; Zhang, L.-D.; Wen, M.-B.; Bie, P. Apigenin potentiates the growth inhibitory effects by IKK-β-mediated NF-κB activation in pancreatic cancer cells. Toxicol. Lett. 2014, 224, 157–164. [Google Scholar] [CrossRef]
  95. Chen, L.; Teng, H.; Jia, Z.; Battino, M.; Miron, A.; Yu, Z.; Cao, H.; Xiao, J. Intracellular signaling pathways of inflammation modulated by dietary flavonoids: The most recent evidence. Crit. Rev. Food Sci. Nutr. 2018, 58, 2908–2924. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, Y.; Luo, J.; Peng, L.; Zhang, Q.; Rong, X.; Luo, Y.; Li, J. Flavonoids: Potential therapeutic agents for cardiovascular disease. Heliyon 2024, 10, e32563. [Google Scholar] [CrossRef] [PubMed]
  97. Roope, L.S.J.; Smith, R.D.; Pouwels, K.B.; Buchanan, J.; Abel, L.; Eibich, P.; Butler, C.C.; Tan, P.S.; Walker, A.S.; Robotham, J.V.; et al. The challenge of antimicrobial resistance: What economics can contribute. Science 2019, 364, eaau4679. [Google Scholar] [CrossRef] [PubMed]
  98. Biharee, A.; Sharma, A.; Kumar, A.; Jaitak, V. Antimicrobial flavonoids as a potential substitute for overcoming antimicrobial resistance. Fitoterapia 2020, 146, 104720. [Google Scholar] [CrossRef] [PubMed]
  99. Weng, Z.; Zeng, F.; Wang, M.; Guo, S.; Tang, Z.; Itagaki, K.; Lin, Y.; Shen, X.; Cao, Y.; Duan, J.-A.; et al. Antimicrobial activities of lavandulylated flavonoids in Sophora flavences against methicillin-resistant Staphylococcus aureus via membrane disruption. J. Adv. Res. 2024, 57, 197–212. [Google Scholar] [CrossRef]
  100. Das, T.; Kutty, S.K.; Kumar, N.; Manefield, M. Pyocyanin facilitates extracellular DNA binding to Pseudomonas aeruginosa influencing cell surface properties and aggregation. PLoS ONE 2013, 8, e58299. [Google Scholar] [CrossRef]
  101. Casilag, F.; Lorenz, A.; Krueger, J.; Klawonn, F.; Weiss, S.; Häussler, S. The LasB Elastase of Pseudomonas aeruginosa Acts in Concert with Alkaline Protease AprA To Prevent Flagellin-Mediated Immune Recognition. Infect. Immun. 2016, 84, 162–171. [Google Scholar] [CrossRef]
  102. Tao, J.; Yan, S.; Wang, H.; Zhao, L.; Zhu, H.; Wen, Z. Antimicrobial and antibiofilm effects of total flavonoids from Potentilla kleiniana Wight et Arn on Pseudomonas aeruginosa and its potential application to stainless steel surfaces. LWT 2022, 154, 112631. [Google Scholar] [CrossRef]
  103. Noor Mohammadi, T.; Maung, A.T.; Sato, J.; Sonoda, T.; Masuda, Y.; Honjoh, K.; Miyamoto, T. Mechanism for antibacterial action of epigallocatechin gallate and theaflavin-3,3′-digallate on Clostridium perfringens. J. Appl. Microbiol. 2019, 126, 633–640. [Google Scholar] [CrossRef]
  104. Lee, J.-H.; Kim, Y.-G.; Khadke, S.K.; Yamano, A.; Woo, J.-T.; Lee, J. Antimicrobial and antibiofilm activities of prenylated flavanones from Macaranga tanarius. Phytomedicine 2019, 63, 153033. [Google Scholar] [CrossRef]
  105. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
  106. Donovan, J.L.; Manach, C.; Faulks, R.M.; Kroon, P.A. Absorption and Metabolism of Dietary Plant Secondary Metabolites. In Plant Secondary Metabolites; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2006; pp. 303–351. [Google Scholar]
  107. Zhang, H.; Hassan, Y.I.; Liu, R.; Mats, L.; Yang, C.; Liu, C.; Tsao, R. Molecular Mechanisms Underlying the Absorption of Aglycone and Glycosidic Flavonoids in a Caco-2 BBe1 Cell Model. ACS Omega 2020, 5, 10782–10793. [Google Scholar] [CrossRef]
  108. Crozier, A.; Del Rio, D.; Clifford, M.N. Bioavailability of dietary flavonoids and phenolic compounds. Mol. Asp. Med. 2010, 31, 446–467. [Google Scholar] [CrossRef]
  109. Walgren, R.A.; Lin, J.-T.; Kinne, R.K.H.; Walle, T. Cellular Uptake of Dietary Flavonoid Quercetin 4′-β-Glucoside by Sodium-Dependent Glucose Transporter SGLT111This study was supported by National Institutes of Health Grant GM55561. J. Pharmacol. Exp. Ther. 2000, 294, 837–843. [Google Scholar] [CrossRef] [PubMed]
  110. Day, A.J.; Mellon, F.; Barron, D.; Sarrazin, G.; Morgan, M.R.A.; Williamson, G. Human metabolism of dietary flavonoids: Identification of plasma metabolites of quercetin. Free. Radic. Res. 2001, 35, 941–952. [Google Scholar] [CrossRef] [PubMed]
  111. Swanson, H. Flavonoids, Inflammation and Cancer; World Scientific: Singapore, 2015. [Google Scholar]
  112. Koster, H.; Halsema, I.; Scholtens, E.; Knippers, M.; Mulder, G.J. Dose-dependent shifts in the sulfation and glucuronidation of phenolic compounds in the rat in vivo and in isolated hepatocytes. The role of saturation of phenolsulfotransferase. Biochem. Pharmacol. 1981, 30, 2569–2575. [Google Scholar] [CrossRef]
  113. Piskula, M.K. Soy Isoflavone Conjugation Differs in Fed and Food-Deprived Rats. J. Nutr. 2000, 130, 1766–1771. [Google Scholar] [CrossRef] [PubMed]
  114. Williamson, G.; Kay, C.D.; Crozier, A. The Bioavailability, Transport, and Bioactivity of Dietary Flavonoids: A Review from a Historical Perspective. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1054–1112. [Google Scholar] [CrossRef]
  115. Skibola, C.F.; Smith, M.T. Potential health impacts of excessive flavonoid intake. Free. Radic. Biol. Med. 2000, 29, 375–383. [Google Scholar] [CrossRef]
  116. Velderrain-Rodríguez, G.R.; Palafox-Carlos, H.; Wall-Medrano, A.; Ayala-Zavala, J.F.; Chen, C.Y.; Robles-Sánchez, M.; Astiazaran-García, H.; Alvarez-Parrilla, E.; González-Aguilar, G.A. Phenolic compounds: Their journey after intake. Food Funct. 2014, 5, 189–197. [Google Scholar] [CrossRef]
  117. Wan, M.L.Y.; Co, V.A.; El-Nezami, H. Dietary polyphenol impact on gut health and microbiota. Crit. Rev. Food Sci. Nutr. 2021, 61, 690–711. [Google Scholar] [CrossRef]
  118. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef] [PubMed]
  119. Aura, A.-M. Microbial metabolism of dietary phenolic compounds in the colon. Phytochem. Rev. 2008, 7, 407–429. [Google Scholar] [CrossRef]
  120. Billowria, K.; Ali, R.; Rangra, N.K.; Kumar, R.; Chawla, P.A. Bioactive Flavonoids: A Comprehensive Review on Pharmacokinetics and Analytical Aspects. Crit. Rev. Anal. Chem. 2024, 54, 1002–1016. [Google Scholar] [CrossRef]
  121. Lin, Y.-T.; Hsiu, S.-L.; Hou, Y.-C.; Chen, H.-Y.; Chao, P.-D.L. Degradation of Flavonoid Aglycones by Rabbit, Rat and Human Fecal Flora. Biol. Pharm. Bull. 2003, 26, 747–751. [Google Scholar] [CrossRef]
  122. Simons, A.L.; Renouf, M.; Hendrich, S.; Murphy, P.A. Human Gut Microbial Degradation of Flavonoids:  Structure−Function Relationships. J. Agric. Food Chem. 2005, 53, 4258–4263. [Google Scholar] [CrossRef]
  123. Rechner, A.R.; Smith, M.A.; Kuhnle, G.; Gibson, G.R.; Debnam, E.S.; Srai, S.K.; Moore, K.P.; Rice-Evans, C.A. Colonic metabolism of dietary polyphenols: Influence of structure on microbial fermentation products. Free. Radic. Biol. Med. 2004, 36, 212–225. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, C.S.; Liang, X.; Wei, X.H.; Chen, F.L.; Tang, Q.F.; Tan, X.M. Comparative metabolism of the eight main bioactive ingredients of gegen qinlian decoction by the intestinal flora of diarrhoeal and healthy piglets. Biomed. Chromatogr. BMC 2019, 33, e4421. [Google Scholar] [CrossRef] [PubMed]
  125. Nakamura, K.; Nishihata, T.; Jin, J.S.; Ma, C.M.; Komatsu, K.; Iwashima, M.; Hattori, M. The C-glucosyl bond of puerarin was cleaved hydrolytically by a human intestinal bacterium strain PUE to yield its aglycone daidzein and an intact glucose. Chem. Pharm. Bull. 2011, 59, 23–27. [Google Scholar] [CrossRef]
  126. Bokkenheuser, V.D.; Shackleton, C.H.; Winter, J. Hydrolysis of dietary flavonoid glycosides by strains of intestinal Bacteroides from humans. Biochem. J. 1987, 248, 953–956. [Google Scholar] [CrossRef]
  127. Jalili-Firoozinezhad, S.; Gazzaniga, F.S.; Calamari, E.L.; Camacho, D.M.; Fadel, C.W.; Bein, A.; Swenor, B.; Nestor, B.; Cronce, M.J.; Tovaglieri, A.; et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nat. Biomed. Eng. 2019, 3, 520–531. [Google Scholar] [CrossRef]
  128. Xiong, H.H.; Lin, S.Y.; Chen, L.L.; Ouyang, K.H.; Wang, W.J. The Interaction between Flavonoids and Intestinal Microbes: A Review. Foods 2023, 12, 320. [Google Scholar] [CrossRef] [PubMed]
  129. Yang, G.; Hong, S.; Yang, P.; Sun, Y.; Wang, Y.; Zhang, P.; Jiang, W.; Gu, Y. Discovery of an ene-reductase for initiating flavone and flavonol catabolism in gut bacteria. Nat. Commun. 2021, 12, 790. [Google Scholar] [CrossRef] [PubMed]
  130. Braune, A.; Engst, W.; Elsinghorst, P.W.; Furtmann, N.; Bajorath, J.; Gütschow, M.; Blaut, M. Chalcone Isomerase from Eubacterium ramulus Catalyzes the Ring Contraction of Flavanonols. J. Bacteriol. 2016, 198, 2965–2974. [Google Scholar] [CrossRef] [PubMed]
  131. Kaluzhskiy, L.; Ershov, P.; Yablokov, E.; Shkel, T.; Grabovec, I.; Mezentsev, Y.; Gnedenko, O.; Usanov, S.; Shabunya, P.; Fatykhava, S.; et al. Human Lanosterol 14-Alpha Demethylase (CYP51A1) Is a Putative Target for Natural Flavonoid Luteolin 7,3′-Disulfate. Molecules 2021, 26, 2237. [Google Scholar] [CrossRef]
  132. Cui, M.Y.; Lu, A.R.; Li, J.X.; Liu, J.; Fang, Y.M.; Pei, T.L.; Zhong, X.; Wei, Y.K.; Kong, Y.; Qiu, W.Q.; et al. Two types of O-methyltransferase are involved in biosynthesis of anticancer methoxylated 4′-deoxyflavones in Scutellaria baicalensis Georgi. Plant Biotechnol. J. 2022, 20, 129–142. [Google Scholar] [CrossRef]
  133. Wen, L.; Jiang, Y.; Yang, J.; Zhao, Y.; Tian, M.; Yang, B. Structure, bioactivity, and synthesis of methylated flavonoids. Ann. New York Acad. Sci. 2017, 1398, 120–129. [Google Scholar] [CrossRef]
  134. Kim, B.G.; Jung, B.R.; Lee, Y.; Hur, H.G.; Lim, Y.; Ahn, J.H. Regiospecific flavonoid 7-O-methylation with Streptomyces avermitilis O-methyltransferase expressed in Escherichia coli. J. Agric. Food Chem. 2006, 54, 823–828. [Google Scholar] [CrossRef]
  135. Burapan, S.; Kim, M.; Han, J. Demethylation of Polymethoxyflavones by Human Gut Bacterium, Blautia sp. MRG-PMF1. J. Agric. Food Chem. 2017, 65, 1620–1629. [Google Scholar] [CrossRef]
  136. Yang, J.; Qian, D.; Guo, J.; Jiang, S.; Shang, E.-X.; Duan, J.-A.; Xu, J. Identification of the major metabolites of hyperoside produced by the human intestinal bacteria using the ultra performance liquid chromatography/quadrupole-time-of-flight mass spectrometry. J. Ethnopharmacol. 2013, 147, 174–179. [Google Scholar] [CrossRef]
  137. Wang, Y.; Yu, F.; Liu, M.Y.; Zhao, Y.K.; Wang, D.M.; Hao, Q.H.; Wang, X.L. Isolation and Characterization of a Human Intestinal Bacterium Eggerthella sp. AUH-JLD49s for the Conversion of (-)-3′-Desmethylarctigenin. J. Agric. Food Chem. 2017, 65, 4051–4056. [Google Scholar] [CrossRef]
  138. Thilakarathna, S.H.; Rupasinghe, H.P. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients 2013, 5, 3367–3387. [Google Scholar] [CrossRef] [PubMed]
  139. Premathilaka, R.; Rashidinejad, A.; Golding, M.; Singh, J. Oral delivery of hydrophobic flavonoids and their incorporation into functional foods: Opportunities and challenges. Food Hydrocoll. 2022, 128, 107567. [Google Scholar] [CrossRef]
  140. Jucá, M.M.; Cysne Filho, F.M.S.; de Almeida, J.C.; Mesquita, D.D.S.; Barriga, J.R.M.; Dias, K.C.F.; Barbosa, T.M.; Vasconcelos, L.C.; Leal, L.; Ribeiro, J.E.; et al. Flavonoids: Biological activities and therapeutic potential. Nat. Prod. Res. 2020, 34, 692–705. [Google Scholar] [CrossRef]
  141. Rahaman, S.T.; Mondal, S. Flavonoids: A vital resource in healthcare and medicine. Pharm. Pharmacol. Int. J. 2020, 8, 91–104. [Google Scholar] [CrossRef]
  142. Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Rémésy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81 (Suppl. S1), 230s–242s. [Google Scholar] [CrossRef]
  143. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and Human Health: The Role of Bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef]
  144. Rodríguez-García, C.; Sánchez-Quesada, C.; Gaforio, J.J. Dietary Flavonoids as Cancer Chemopreventive Agents: An Updated Review of Human Studies. Antioxidants 2019, 8, 137. [Google Scholar] [CrossRef] [PubMed]
  145. Spencer, J.P.E.; Chaudry, F.; Pannala, A.S.; Srai, S.K.; Debnam, E.; Rice-Evans, C. Decomposition of Cocoa Procyanidins in the Gastric Milieu. Biochem. Biophys. Res. Commun. 2000, 272, 236–241. [Google Scholar] [CrossRef]
  146. Zong, G.; Fei, S.; Liu, X.; Li, J.; Gao, Y.; Yang, X.; Wang, X.; Shen, Y. Crystal structures of rhamnosyltransferase UGT89C1 from Arabidopsis thaliana reveal the molecular basis of sugar donor specificity for UDP-β-l-rhamnose and rhamnosylation mechanism. Plant J. Cell Mol. Biol. 2019, 99, 257–269. [Google Scholar] [CrossRef]
  147. Wu, Y.; Wang, H.; Liu, Y.; Zhao, L.; Pei, J. An efficient preparation and biocatalytic synthesis of novel C-glycosylflavonols kaempferol 8-C-glucoside and quercetin 8-C-glucoside through using resting cells and macroporous resins. Biotechnol. Biofuels Bioprod. 2022, 15, 129. [Google Scholar] [CrossRef]
  148. Morand, C.; Manach, C.; Crespy, V.; Remesy, C. Quercetin 3-O-beta-glucoside is better absorbed than other quercetin forms and is not present in rat plasma. Free Radic. Res. 2000, 33, 667–676. [Google Scholar] [CrossRef] [PubMed]
  149. Setchell, K.D.; Brown, N.M.; Desai, P.; Zimmer-Nechemias, L.; Wolfe, B.E.; Brashear, W.T.; Kirschner, A.S.; Cassidy, A.; Heubi, J.E. Bioavailability of pure isoflavones in healthy humans and analysis of commercial soy isoflavone supplements. J. Nutr. 2001, 131 (Suppl. S4), 1362s–1375s. [Google Scholar] [CrossRef]
  150. Mao, S.; Ren, Y.; Ye, X.; Tian, J. The metabolites of flavonoids with typical structure enhanced bioactivity through gut microbiota. Food Biosci. 2024, 59, 104165. [Google Scholar] [CrossRef]
  151. Zhou, M.; Ma, J.; Kang, M.; Tang, W.; Xia, S.; Yin, J.; Yin, Y. Flavonoids, gut microbiota, and host lipid metabolism. Eng. Life Sci. 2024, 24, 2300065. [Google Scholar] [CrossRef] [PubMed]
  152. Makino, R.; Takano, K.; Kita, K.; Nishimukai, M. Influence of long-term feeding of high-fat diet on quercetin and fat absorption from the small intestine in lymph duct-cannulated rats. Biosci. Biotechnol. Biochem. 2018, 82, 2007–2011. [Google Scholar] [CrossRef] [PubMed]
  153. Favari, C.; Rinaldi de Alvarenga, J.F.; Sánchez-Martínez, L.; Tosi, N.; Mignogna, C.; Cremonini, E.; Manach, C.; Bresciani, L.; Del Rio, D.; Mena, P. Factors driving the inter-individual variability in the metabolism and bioavailability of (poly)phenolic metabolites: A systematic review of human studies. Redox Biol. 2024, 71, 103095. [Google Scholar] [CrossRef]
  154. Sime, F.B.; Roberts, M.S.; Roberts, J.A. Optimization of dosing regimens and dosing in special populations. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2015, 21, 886–893. [Google Scholar] [CrossRef]
  155. Kasahara, K.; Kerby, R.L.; Aquino-Martinez, R.; Evered, A.H.; Cross, T.-W.L.; Everhart, J.; Ulland, T.K.; Kay, C.D.; Bolling, B.W.; Bäckhed, F.; et al. Gut microbes modulate the effects of the flavonoid quercetin on atherosclerosis. NPJ Biofilms Microbiomes 2025, 11, 12. [Google Scholar] [CrossRef]
  156. Liu, H.; Wu, B.; Pan, G.; He, L.; Li, Z.; Fan, M.; Jian, L.; Chen, M.; Wang, K.; Huang, C. Metabolism and pharmacokinetics of mangiferin in conventional rats, pseudo-germ-free rats, and streptozotocin-induced diabetic rats. Drug Metab. Dispos. Biol. Fate Chem. 2012, 40, 2109–2118. [Google Scholar] [CrossRef]
  157. Dobrzynska, M.; Napierala, M.; Florek, E. Flavonoid Nanoparticles: A Promising Approach for Cancer Therapy. Biomolecules 2020, 10, 1268. [Google Scholar] [CrossRef]
  158. Wang, J.; Feng, X.; Li, Z.; Liu, Y.; Yang, W.; Zhang, T.; Guo, P.; Liu, Z.; Qi, D.; Pi, J. The Flavonoid Components of Scutellaria baicalensis: Biopharmaceutical Properties and their Improvement using Nanoformulation Techniques. Curr. Top. Med. Chem. 2023, 23, 17–29. [Google Scholar]
  159. Bhia, M.; Motallebi, M.; Abadi, B.; Zarepour, A.; Pereira-Silva, M.; Saremnejad, F.; Santos, A.C.; Zarrabi, A.; Melero, A.; Jafari, S.M.; et al. Naringenin Nano-Delivery Systems and Their Therapeutic Applications. Pharmaceutics 2021, 13, 291. [Google Scholar] [CrossRef]
  160. Joma, N.; Bielawski, P.B.; Saini, A.; Kakkar, A.; Maysinger, D. Nanocarriers for natural polyphenol senotherapeutics. Aging Cell 2024, 23, e14178. [Google Scholar] [CrossRef] [PubMed]
  161. Cai, Z.Y.; Li, X.M.; Liang, J.P.; Xiang, L.P.; Wang, K.R.; Shi, Y.L.; Yang, R.; Shi, M.; Ye, J.H.; Lu, J.L.; et al. Bioavailability of Tea Catechins and Its Improvement. Molecules 2018, 23, 2346. [Google Scholar] [CrossRef]
  162. Sysak, S.; Czarczynska-Goslinska, B.; Szyk, P.; Koczorowski, T.; Mlynarczyk, D.T.; Szczolko, W.; Lesyk, R.; Goslinski, T. Metal Nanoparticle-Flavonoid Connections: Synthesis, Physicochemical and Biological Properties, as Well as Potential Applications in Medicine. Nanomaterials 2023, 13, 1531. [Google Scholar] [CrossRef] [PubMed]
  163. Chen, S.; Wang, X.; Cheng, Y.; Gao, H.; Chen, X. A Review of Classification, Biosynthesis, Biological Activities and Potential Applications of Flavonoids. Molecules 2023, 28, 4982. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, B.; Wang, C.; Guo, M.; Zhu, F.; Yu, Z.; Zhang, W.; Li, W.; Zhang, Y.; Tian, W. Circadian Rhythm-Dependent Therapy by Composite Targeted Polyphenol Nanoparticles for Myocardial Ischemia-Reperfusion Injury. ACS Nano 2024, 18, 28154–28169. [Google Scholar] [CrossRef]
  165. Li, J.; Jiang, X.; Shang, L.; Li, Z.; Yang, C.; Luo, Y.; Hu, D.; Shen, Y.; Zhang, Z. L-EGCG-Mn nanoparticles as a pH-sensitive MRI contrast agent. Drug Deliv. 2021, 28, 134–143. [Google Scholar] [CrossRef]
  166. Xu, C.; Wang, Y.; Yu, H.; Tian, H.; Chen, X. Multifunctional Theranostic Nanoparticles Derived from Fruit-Extracted Anthocyanins with Dynamic Disassembly and Elimination Abilities. ACS Nano 2018, 12, 8255–8265. [Google Scholar] [CrossRef]
  167. Fu, S.; Cai, Z.; Gu, H.; Lui, S.; Ai, H.; Song, B.; Wu, M. Rutin-coated ultrasmall manganese oxide nanoparticles for targeted magnetic resonance imaging and photothermal therapy of malignant tumors. J. Colloid Interface Sci. 2024, 670, 499–508. [Google Scholar] [CrossRef]
  168. Guo, C.; Sun, J.; Dong, J.; Cai, W.; Zhao, X.; Song, B.; Zhang, R. A natural anthocyanin-based multifunctional theranostic agent for dual-modal imaging and photothermal anti-tumor therapy. J. Mater. Chem. B 2021, 9, 7447–7460. [Google Scholar] [CrossRef]
  169. Zhang, J.; Xie, H.; Wang, T.; Zhang, H.; Yang, Z.; Yang, P.; Li, Y.; Ma, X.; Gu, Z. Epicatechin-assembled nanoparticles against renal ischemia/reperfusion injury. J. Mater. Chem. B 2022, 10, 6965–6973. [Google Scholar] [CrossRef] [PubMed]
  170. Pradhan, S.P.; Swain, S.; Sa, N.; Pilla, S.N.; Behera, A.; Sahu, P.K.; Chandra Si, S. Photocatalysis of environmental organic pollutants and antioxidant activity of flavonoid conjugated gold nanoparticles. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2022, 282, 121699. [Google Scholar] [CrossRef]
  171. Hassani, S.; Maghsoudi, H.; Fattahi, F.; Malekinejad, F.; Hajmalek, N.; Sheikhnia, F.; Kheradmand, F.; Fahimirad, S.; Ghorbanpour, M. Flavonoids nanostructures promising therapeutic efficiencies in colorectal cancer. Int. J. Biol. Macromol. 2023, 241, 124508. [Google Scholar] [CrossRef] [PubMed]
  172. Wen, E.; Cao, Y.; He, S.; Zhang, Y.; You, L.; Wang, T.; Wang, Z.; He, J.; Feng, Y. The mitochondria-targeted Kaempferol nanoparticle ameliorates severe acute pancreatitis. J. Nanobiotechnology 2024, 22, 148. [Google Scholar] [CrossRef] [PubMed]
  173. Aiello, P.; Consalvi, S.; Poce, G.; Raguzzini, A.; Toti, E.; Palmery, M.; Biava, M.; Bernardi, M.; Kamal, M.A.; Perry, G.; et al. Dietary flavonoids: Nano delivery and nanoparticles for cancer therapy. Semin. Cancer Biol. 2021, 69, 150–165. [Google Scholar] [CrossRef]
  174. Jain, A.K.; Thanki, K.; Jain, S. Co-encapsulation of Tamoxifen and Quercetin in Polymeric Nanoparticles: Implications on Oral Bioavailability, Antitumor Efficacy, and Drug-Induced Toxicity. Mol. Pharm. 2013, 10, 3459–3474. [Google Scholar] [CrossRef]
  175. Sharma, T.; Singh, D.; Mahapatra, A.; Mohapatra, P.; Sahoo, S.; Sahoo, S.K. Advancements in clinical translation of flavonoid nanoparticles for cancer treatment. OpenNano 2022, 8, 100074. [Google Scholar] [CrossRef]
  176. Shi-Ying, J.; Jin, H.; Shi-Xiao, J.; Qing-Yuan, L.; Jin-Xia, B.; Chen, H.G.; Rui-Sheng, L.; Wei, W.; Hai-Long, Y. Characterization and evaluation in vivo of baicalin-nanocrystals prepared by an ultrasonic-homogenization-fluid bed drying method. Chin. J. Nat. Med. 2014, 12, 71–80. [Google Scholar] [CrossRef]
  177. Liu, J.; Sun, Y.; Cheng, M.; Liu, Q.; Liu, W.; Gao, C.; Feng, J.; Jin, Y.; Tu, L. Improving Oral Bioavailability of Luteolin Nanocrystals by Surface Modification of Sodium Dodecyl Sulfate. AAPS PharmSciTech 2021, 22, 133. [Google Scholar] [CrossRef]
  178. Uğur Kaplan, A.B.; Öztürk, N.; Çetin, M.; Vural, İ.; Öznülüer Özer, T. The Nanosuspension Formulations of Daidzein: Preparation and In Vitro Characterization. Turk. J. Pharm. Sci. 2022, 19, 84–92. [Google Scholar] [CrossRef] [PubMed]
  179. Singh, M.K.; Pooja, D.; Ravuri, H.G.; Gunukula, A.; Kulhari, H.; Sistla, R. Fabrication of surfactant-stabilized nanosuspension of naringenin to surpass its poor physiochemical properties and low oral bioavailability. Phytomedicine 2018, 40, 48–54. [Google Scholar] [CrossRef] [PubMed]
  180. Chi, C.; Zhang, C.; Liu, Y.; Nie, H.; Zhou, J.; Ding, Y. Phytosome-nanosuspensions for silybin-phospholipid complex with increased bioavailability and hepatoprotection efficacy. Eur. J. Pharm. Sci. 2020, 144, 105212. [Google Scholar] [CrossRef]
  181. He, W.; Zhang, J.; Ju, J.; Wu, Y.; Zhang, Y.; Zhan, L.; Li, C.; Wang, Y. Preparation, characterization, and evaluation of the antitumor effect of kaempferol nanosuspensions. Drug Deliv. Transl. Res. 2023, 13, 2885–2902. [Google Scholar] [CrossRef]
  182. Ao, H.; Li, Y.; Li, H.; Wang, Y.; Han, M.; Guo, Y.; Shi, R.; Yue, F.; Wang, X. Preparation of hydroxy genkwanin nanosuspensions and their enhanced antitumor efficacy against breast cancer. Drug Deliv. 2020, 27, 816–824. [Google Scholar] [CrossRef]
  183. Gera, S.; Talluri, S.; Rangaraj, N.; Sampathi, S. Formulation and Evaluation of Naringenin Nanosuspensions for Bioavailability Enhancement. AAPS PharmSciTech 2017, 18, 3151–3162. [Google Scholar] [CrossRef]
  184. Xie, J.; Luo, Y.; Liu, Y.; Ma, Y.; Yue, P.; Yang, M. Novel redispersible nanosuspensions stabilized by co-processed nanocrystalline cellulose-sodium carboxymethyl starch for enhancing dissolution and oral bioavailability of baicalin. Int. J. Nanomed. 2019, 14, 353–369. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, Y.; Guan, R.; Huang, H. Anti-Allergic Effects of Quercetin and Quercetin Liposomes in RBL-2H3 Cells. Endocr. Metab. Immune Disord. Drug Targets 2023, 23, 692–701. [Google Scholar] [CrossRef]
  186. Seguin, J.; Brullé, L.; Boyer, R.; Lu, Y.M.; Ramos Romano, M.; Touil, Y.S.; Scherman, D.; Bessodes, M.; Mignet, N.; Chabot, G.G. Liposomal encapsulation of the natural flavonoid fisetin improves bioavailability and antitumor efficacy. Int. J. Pharm. 2013, 444, 146–154. [Google Scholar] [CrossRef]
  187. Li, Z.; Liang, S.; Sun, H.; Bao, C.; Li, Y. Antilipogenesis Effect of Rutin-Loaded Liposomes Using a Microneedle Delivery System. ACS Appl. Mater. Interfaces 2023, 15, 54294–54303. [Google Scholar] [CrossRef]
  188. Priya, V.; Singh, S.K.; Revand, R.; Kumar, S.; Mehata, A.K.; Sushmitha, P.; Mahto, S.K.; Muthu, M.S. GPIIb/IIIa Receptor Targeted Rutin Loaded Liposomes for Site-Specific Antithrombotic Effect. Mol. Pharm. 2023, 20, 663–679. [Google Scholar] [CrossRef] [PubMed]
  189. Liu, J.; Zhu, Z.; Yang, Y.; Adu-Frimpong, M.; Chen, L.; Ji, H.; Toreniyazov, E.; Wang, Q.; Yu, J.; Xu, X. Preparation, characterization, pharmacokinetics, and antirenal injury activity studies of Licochalcone A-loaded liposomes. J. Food Biochem. 2022, 46, e14007. [Google Scholar] [CrossRef]
  190. Karamchedu, S.; Tunki, L.; Kulhari, H.; Pooja, D. Morin hydrate loaded solid lipid nanoparticles: Characterization, stability, anticancer activity, and bioavailability. Chem. Phys. Lipids 2020, 233, 104988. [Google Scholar] [CrossRef]
  191. Zhao, B.; Gu, S.; Du, Y.; Shen, M.; Liu, X.; Shen, Y. Solid lipid nanoparticles as carriers for oral delivery of hydroxysafflor yellow A. Int. J. Pharm. 2018, 535, 164–171. [Google Scholar] [CrossRef]
  192. Liu, Z.; Zhang, L.; He, Q.; Liu, X.; Okeke, C.I.; Tong, L.; Guo, L.; Yang, H.; Zhang, Q.; Zhao, H.; et al. Effect of Baicalin-loaded PEGylated cationic solid lipid nanoparticles modified by OX26 antibody on regulating the levels of baicalin and amino acids during cerebral ischemia-reperfusion in rats. Int. J. Pharm. 2015, 489, 131–138. [Google Scholar] [CrossRef]
  193. Zaheer, Y.; Ali, M.A.; Rehman, M.; Iftikhar, M.; Anwar, S.; Ali, A.; Mobeen, A.; Iqbal, M.; Iqbal, S.; Younis, M.R.; et al. Naringenin loaded solid lipid nanoparticles alleviate oxidative stress and enhance oral bioavailability of naringenin. Colloids Surf. B Biointerfaces 2024, 247, 114423. [Google Scholar] [CrossRef]
  194. Ji, P.; Yu, T.; Liu, Y.; Jiang, J.; Xu, J.; Zhao, Y.; Hao, Y.; Qiu, Y.; Zhao, W.; Wu, C. Naringenin-loaded solid lipid nanoparticles: Preparation, controlled delivery, cellular uptake, and pulmonary pharmacokinetics. Drug Des. Dev. Ther. 2016, 10, 911–925. [Google Scholar]
  195. Luo, C.-F.; Yuan, M.; Chen, M.-S.; Liu, S.-M.; Zhu, L.; Huang, B.-Y.; Liu, X.-W.; Xiong, W. Pharmacokinetics, tissue distribution and relative bioavailability of puerarin solid lipid nanoparticles following oral administration. Int. J. Pharm. 2011, 410, 138–144. [Google Scholar] [CrossRef] [PubMed]
  196. Ma, Y.; Li, H.; Guan, S. Enhancement of the oral bioavailability of breviscapine by nanoemulsions drug delivery system. Drug Dev. Ind. Pharm. 2015, 41, 177–182. [Google Scholar] [CrossRef]
  197. Yin, J.; Xiang, C.; Wang, P.; Yin, Y.; Hou, Y. Biocompatible nanoemulsions based on hemp oil and less surfactants for oral delivery of baicalein with enhanced bioavailability. Int. J. Nanomed. 2017, 12, 2923–2931. [Google Scholar] [CrossRef]
  198. Sun, D.; Wei, X.; Xue, X.; Fang, Z.; Ren, M.; Lou, H.; Zhang, X. Enhanced oral absorption and therapeutic effect of acetylpuerarin based on D-α-tocopheryl polyethylene glycol 1000 succinate nanoemulsions. Int. J. Nanomed. 2014, 9, 3413–3423. [Google Scholar]
  199. Diedrich, C.; Camargo Zittlau, I.; Schineider Machado, C.; Taise Fin, M.; Maissar Khalil, N.; Badea, I.; Mara Mainardes, R. Mucoadhesive nanoemulsion enhances brain bioavailability of luteolin after intranasal administration and induces apoptosis to SH-SY5Y neuroblastoma cells. Int. J. Pharm. 2022, 626, 122142. [Google Scholar] [CrossRef] [PubMed]
  200. Ragelle, H.; Crauste-Manciet, S.; Seguin, J.; Brossard, D.; Scherman, D.; Arnaud, P.; Chabot, G.G. Nanoemulsion formulation of fisetin improves bioavailability and antitumour activity in mice. Int. J. Pharm. 2012, 427, 452–459. [Google Scholar] [CrossRef]
  201. Zhang, R.; Yang, J.; Luo, Q.; Shi, J.; Xu, H.; Zhang, J. Preparation and in vitro and in vivo evaluation of an isoliquiritigenin-loaded ophthalmic nanoemulsion for the treatment of corneal neovascularization. Drug Deliv. 2022, 29, 2217–2233. [Google Scholar] [CrossRef] [PubMed]
  202. 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]
  203. Mi, X.; Hu, M.; Dong, M.; Yang, Z.; Zhan, X.; Chang, X.; Lu, J.; Chen, X. Folic Acid Decorated Zeolitic Imidazolate Framework (ZIF-8) Loaded with Baicalin as a Nano-Drug Delivery System for Breast Cancer Therapy. Int. J. Nanomed. 2021, 16, 8337–8352. [Google Scholar] [CrossRef] [PubMed]
  204. Tang, P.; Sun, Q.; Yang, H.; Tang, B.; Pu, H.; Li, H. Honokiol nanoparticles based on epigallocatechin gallate functionalized chitin to enhance therapeutic effects against liver cancer. Int. J. Pharm. 2018, 545, 74–83. [Google Scholar] [CrossRef]
  205. Krishnan, G.; Subramaniyan, J.; Chengalvarayan Subramani, P.; Muralidharan, B.; Thiruvengadam, D. Hesperetin conjugated PEGylated gold nanoparticles exploring the potential role in anti-inflammation and anti-proliferation during diethylnitrosamine-induced hepatocarcinogenesis in rats. Asian J. Pharm. Sci. 2017, 12, 442–455. [Google Scholar] [CrossRef]
  206. Mashhadi Malekzadeh, A.; Ramazani, A.; Tabatabaei Rezaei, S.J.; Niknejad, H. Design and construction of multifunctional hyperbranched polymers coated magnetite nanoparticles for both targeting magnetic resonance imaging and cancer therapy. J. Colloid Interface Sci. 2017, 490, 64–73. [Google Scholar] [CrossRef]
  207. Song, Z.; Yin, J.; Xiao, P.; Chen, J.; Gou, J.; Wang, Y.; Zhang, Y.; Yin, T.; Tang, X.; He, H. Improving Breviscapine Oral Bioavailability by Preparing Nanosuspensions, Liposomes and Phospholipid Complexes. Pharmaceutics 2021, 13, 132. [Google Scholar] [CrossRef]
  208. Chen, Y.; Liu, Y.; Xie, J.; Zheng, Q.; Yue, P.; Chen, L.; Hu, P.; Yang, M. Nose-to-Brain Delivery by Nanosuspensions-Based in situ Gel for Breviscapine. Int. J. Nanomed. 2020, 15, 10435–10451. [Google Scholar] [CrossRef]
  209. Sajid, M.; Channakesavula, C.N.; Stone, S.R.; Kaur, P. Synthetic Biology towards Improved Flavonoid Pharmacokinetics. Biomolecules 2021, 11, 754. [Google Scholar] [CrossRef] [PubMed]
  210. Isika, D.K.; Sadik, O.A. Selective Structural Derivatization of Flavonoid Acetamides Significantly Impacts Their Bioavailability and Antioxidant Properties. Molecules 2022, 27, 8133. [Google Scholar] [CrossRef] [PubMed]
  211. Isika, D.K.; Özkömeç, F.N.; Çeşme, M.; Sadik, O.A. Synthesis, biological and computational studies of flavonoid acetamide derivatives. RSC Adv. 2022, 12, 10037–10050. [Google Scholar] [CrossRef] [PubMed]
  212. Jiang, L.; Wang, D.; Zhang, Y.; Li, J.; Wu, Z.; Wang, Z.; Wang, D. Investigation of the pro-apoptotic effects of arbutin and its acetylated derivative on murine melanoma cells. Int. J. Mol. Med. 2018, 41, 1048–1054. [Google Scholar] [CrossRef]
  213. Lambert, J.D.; Sang, S.; Hong, J.; Kwon, S.J.; Lee, M.J.; Ho, C.T.; Yang, C.S. Peracetylation as a means of enhancing in vitro bioactivity and bioavailability of epigallocatechin-3-gallate. Drug Metab. Dispos. Biol. Fate Chem. 2006, 34, 2111–2116. [Google Scholar] [CrossRef]
  214. Li, X.-F.; Yuan, T.; Xu, H.; Xin, X.; Zhao, G.; Wu, H.; Xiao, X. Whole-Cell Catalytic Synthesis of Puerarin Monoesters and Analysis of Their Antioxidant Activities. J. Agric. Food Chem. 2019, 67, 299–307. [Google Scholar] [CrossRef]
  215. Xiao, C.; Li, J.; Dong, X.; He, X.; Niu, X.; Liu, C.; Zhong, G.; Bauer, R.; Yang, D.; Lu, A. Anti-oxidative and TNF-α suppressive activities of puerarin derivative (4AC) in RAW264.7 cells and collagen-induced arthritic rats. Eur. J. Pharmacol. 2011, 666, 242–250. [Google Scholar] [CrossRef]
  216. Yuan, D.; Guo, Y.; Pu, F.; Yang, C.; Xiao, X.; Du, H.; He, J.; Lu, S. Opportunities and challenges in enhancing the bioavailability and bioactivity of dietary flavonoids: A novel delivery system perspective. Food Chem. 2024, 430, 137115. [Google Scholar] [CrossRef]
  217. Dai, J.; Liang, K.; Zhao, S.; Jia, W.; Liu, Y.; Wu, H.; Lv, J.; Cao, C.; Chen, T.; Zhuang, S.; et al. Chemoproteomics reveals baicalin activates hepatic CPT1 to ameliorate diet-induced obesity and hepatic steatosis. Proc. Natl. Acad. Sci. USA 2018, 115, E5896–E5905. [Google Scholar] [CrossRef]
  218. Milbury, P.E.; Vita, J.A.; Blumberg, J.B. Anthocyanins are bioavailable in humans following an acute dose of cranberry juice. J. Nutr. 2010, 140, 1099–1104. [Google Scholar] [CrossRef]
  219. Zhao, C.L.; Chen, Z.J.; Bai, X.S.; Ding, C.; Long, T.J.; Wei, F.G.; Miao, K.R. Structure–activity relationships of anthocyanidin glycosylation. Mol. Divers. 2014, 18, 687–700. [Google Scholar] [CrossRef] [PubMed]
  220. Sang, S.; Lambert, J.D.; Yang, C.S. Bioavailability and stability issues in understanding the cancer preventive effects of tea polyphenols. J. Sci. Food Agric. 2006, 86, 2256–2265. [Google Scholar] [CrossRef]
  221. Lotito, S.B.; Zhang, W.J.; Yang, C.S.; Crozier, A.; Frei, B. Metabolic conversion of dietary flavonoids alters their anti-inflammatory and antioxidant properties. Free. Radic. Biol. Med. 2011, 51, 454–463. [Google Scholar] [CrossRef]
  222. Ha, T.; Kim, M.K.; Park, K.S.; Jung, W.; Choo, H.; Chong, Y. Structural Modification of (-)-Epigallocatechin Gallate (EGCG) Shows Significant Enhancement in Mitochondrial Biogenesis. J. Agric. Food Chem. 2018, 66, 3850–3859. [Google Scholar] [CrossRef] [PubMed]
  223. Forester, S.C.; Lambert, J.D. The catechol-O-methyltransferase inhibitor, tolcapone, increases the bioavailability of unmethylated (-)-epigallocatechin-3-gallate in mice. J. Funct. Foods 2015, 17, 183–188. [Google Scholar] [CrossRef]
  224. Calla, B. Diverse defenses: O-methylated flavonoids contribute to the maize arsenal against fungal pathogens. Plant Physiol. 2022, 188, 24–25. [Google Scholar] [CrossRef] [PubMed]
  225. Xin, X.; Zhang, M.; Li, X.-F.; Zhao, G. Biocatalytic Synthesis of Lipophilic Baicalin Derivatives as Antimicrobial Agents. J. Agric. Food Chem. 2019, 67, 11684–11693. [Google Scholar] [CrossRef]
  226. Zhang, M.; Xin, X.; Zhao, G.; Zou, Y.; Li, X.-F. In vitro absorption and lipid-lowering activity of baicalin esters synthesized by whole-cell catalyzed esterification. Bioorganic Chem. 2022, 120, 105628. [Google Scholar] [CrossRef]
  227. Fernando, W.; Clark, R.F.; Rupasinghe, H.P.V.; Hoskin, D.W.; Coombs, M.R.P. Phloridzin Docosahexaenoate Inhibits Spheroid Formation by Breast Cancer Stem Cells and Exhibits Cytotoxic Effects against Paclitaxel-Resistant Triple Negative Breast Cancer Cells. Int. J. Mol. Sci. 2023, 24, 14577. [Google Scholar] [CrossRef]
  228. Arumuggam, N.; Melong, N.; Too, C.K.; Berman, J.N.; Rupasinghe, H.V. Phloridzin docosahexaenoate, a novel flavonoid derivative, suppresses growth and induces apoptosis in T-cell acute lymphoblastic leukemia cells. Am. J. Cancer Res. 2017, 7, 2452–2464. [Google Scholar]
  229. Fernando, W.; Coyle, K.; Marcato, P.; Vasantha Rupasinghe, H.P.; Hoskin, D.W. Phloridzin docosahexaenoate, a novel fatty acid ester of a plant polyphenol, inhibits mammary carcinoma cell metastasis. Cancer Lett. 2019, 465, 68–81. [Google Scholar] [CrossRef]
  230. Mantso, T.; Trafalis, D.T.; Botaitis, S.; Franco, R.; Pappa, A.; Rupasinghe, H.P.V.; Panayiotidis, M.I. Novel Docosahexaenoic Acid Ester of Phloridzin Inhibits Proliferation and Triggers Apoptosis in an In Vitro Model of Skin Cancer. Antioxidants 2018, 7, 188. [Google Scholar] [CrossRef] [PubMed]
  231. Warnakulasuriya, S.N.; Ziaullah; Rupasinghe, H.P.V. Long Chain Fatty Acid Esters of Quercetin-3-O-glucoside Attenuate H2O2-induced Acute Cytotoxicity in Human Lung Fibroblasts and Primary Hepatocytes. Molecules 2016, 21, 452. [Google Scholar] [CrossRef]
  232. Chen, L.; Cao, H.; Huang, Q.; Xiao, J.; Teng, H. Absorption, metabolism and bioavailability of flavonoids: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 7730–7742. [Google Scholar] [CrossRef] [PubMed]
  233. Chowdhury, A.; Gorain, B.; Mitra Mazumder, P. Recent advancements in drug delivery system of flavonoids with a special emphasis on the flavanone naringenin: Exploring their application in wound healing and associated processes. Inflammopharmacology 2024, 33, 69–90. [Google Scholar] [CrossRef] [PubMed]
  234. Khan, H.; Ullah, H.; Martorell, M.; Valdes, S.E.; Belwal, T.; Tejada, S.; Sureda, A.; Kamal, M.A. Flavonoids nanoparticles in cancer: Treatment, prevention and clinical prospects. Semin. Cancer Biol. 2021, 69, 200–211. [Google Scholar] [CrossRef]
  235. Li, C.; Dai, T.; Chen, J.; Chen, M.; Liang, R.; Liu, C.; Du, L.; McClements, D.J. Modification of flavonoids: Methods and influences on biological activities. Crit. Rev. Food Sci. Nutr. 2023, 63, 10637–10658. [Google Scholar] [CrossRef]
  236. Baky, M.H.; Elshahed, M.; Wessjohann, L.; Farag, M.A. Interactions between dietary flavonoids and the gut microbiome: A comprehensive review. Br. J. Nutr. 2022, 128, 577–591. [Google Scholar] [CrossRef]
  237. Di Gioia, D.; Strahsburger, E.; Lopez de Lacey, A.M.; Bregola, V.; Marotti, I.; Aloisio, I.; Biavati, B.; Dinelli, G. Flavonoid bioconversion in Bifidobacterium pseudocatenulatum B7003: A potential probiotic strain for functional food development. J. Funct. Foods 2014, 7, 671–679. [Google Scholar] [CrossRef]
  238. Pei, R.; Liu, X.; Bolling, B. Flavonoids and gut health. Curr. Opin. Biotechnol. 2020, 61, 153–159. [Google Scholar] [CrossRef] [PubMed]
  239. Filannino, P.; Bai, Y.; Di Cagno, R.; Gobbetti, M.; Gänzle, M.G. Metabolism of phenolic compounds by Lactobacillus spp. during fermentation of cherry juice and broccoli puree. Food Microbiol. 2015, 46, 272–279. [Google Scholar] [CrossRef] [PubMed]
  240. Mahomud, M.S.; Islam, M.N.; Hossen, D.; Wazed, M.A.; Yasmin, S.; Sarker, M.S.H. Innovative probiotic yogurt: Leveraging green banana peel for enhanced quality, functionality, and sensory attributes. Heliyon 2024, 10, e38781. [Google Scholar] [CrossRef] [PubMed]
  241. Shehata, M.G.; Abd El-Aziz, N.M.; Mehany, T.; Simal-Gandara, J. Taro leaves extract and probiotic lactic acid bacteria: A synergistic approach to improve antioxidant capacity and bioaccessibility in fermented milk beverages. LWT 2023, 187, 115280. [Google Scholar] [CrossRef]
  242. Zhang, X.; Miao, Q.; Pan, C.; Yin, J.; Wang, L.; Qu, L.; Yin, Y.; Wei, Y. Research advances in probiotic fermentation of Chinese herbal medicines. iMeta 2023, 2, e93. [Google Scholar] [CrossRef]
  243. Wang, Y.; Wang, C.; Shi, J.; Zhang, Y. Effects of derivatization and probiotic transformation on the antioxidative activity of fruit polyphenols. Food Chem. X 2024, 23, 101776. [Google Scholar] [CrossRef]
  244. Pereira, E.P.R.; Ferreira, B.M.; Freire, L.; Neri-Numa, I.A.; Guimarães, J.T.; Rocha, R.S.; Pastore, G.M.; Cruz, A.G.; Sant’ana, A.S. Enhancing the functionality of yogurt: Impact of exotic fruit pulps addition on probiotic viability and metabolites during processing and storage. Food Res. Int. 2024, 196, 115057. [Google Scholar] [CrossRef]
  245. Mafe, A.N.; Edo, G.I.; Majeed, O.S.; Gaaz, T.S.; Akpoghelie, P.O.; Isoje, E.F.; Igbuku, U.A.; Owheruo, J.O.; Opiti, R.A.; Garba, Y.; et al. A review on probiotics and dietary bioactives: Insights on metabolic well-being, gut microbiota, and inflammatory responses. Food Chem. Adv. 2025, 6, 100919. [Google Scholar] [CrossRef]
Figure 1. Basic skeleton and active sites of flavonoids.
Figure 1. Basic skeleton and active sites of flavonoids.
Molecules 30 01184 g001
Figure 2. The synthetic pathway of flavonoids.
Figure 2. The synthetic pathway of flavonoids.
Molecules 30 01184 g002
Figure 3. Metabolic pathways of flavonoids.
Figure 3. Metabolic pathways of flavonoids.
Molecules 30 01184 g003
Figure 4. Processing of intestinal microbiome.
Figure 4. Processing of intestinal microbiome.
Molecules 30 01184 g004
Table 1. Classification, chemical characterization, common biological sources, representatives, and applications of flavonoids.
Table 1. Classification, chemical characterization, common biological sources, representatives, and applications of flavonoids.
SubtypeStructure BackboneChemical CharacterizationCommon Biological SourcesRepresentativesApplicationsRef.
FlavoneMolecules 30 01184 i001There is a double bond between the C2 and C3 positions; a ketone group is at the C4 position; and the C2 position is connected to the B ring.Celery, tea, red peppers, and orangesApigenin; LuteolinCancer, cardiovascular disease, neuroinflammation inflammation, anti-diabetic, antibacterial, antioxidant, and antiviral, etc.[26,27,28]
IsoflavoneMolecules 30 01184 i002There is a double bond between the C2 and C3 positions; a ketone group is at the C4 position; and the C3 position is connected to the B ring.Soybeans, and soy-derived productsGenistein; DaidzeinAntioxidant, diarrhea relief, procoagulant activity, and anticancer, etc.[29,30,31]
FlavonolMolecules 30 01184 i003There is a double bond between the C2 and C3 positions; a ketone group is at the C4 position; a hydroxyl group is connected to the C3 position; and the C2 position is connected to the B ring.Apples, cherries, plums, apricots, berries, onions, kale, and leeksQuercetin; Myricetin; KaempferolCardiovascular diseases, anticancer, antioxidation, and neuroprotection, etc.[32,33,34,35]
FlavanolMolecules 30 01184 i004There is no double bond between the C2 and C3 positions; there is no ketone group at the C4 position; a hydroxyl group is connected to the C3 position; the C2 position is connected to the B ring.Broccoli, onions, asparagus, apples, and teaEpicatechin; EpigallocatechinAntioxidant, anticancer, and anti-inflammatory, etc.[36,37,38]
FlavanoneMolecules 30 01184 i005There is no double bond between the C2 and C3 positions; there is a ketone group at C4 position; and the C2 position is connected to the B ring.Citrus FruitsHesperidin; Naringin;
Paclitaxel
Anticancer, anti-inflammatory, antibacterial, antioxidant, antiviral, and lipid-lowering, etc.[2,39,40,41]
ChalconeMolecules 30 01184 i006No C ring (open-chain flavonoids).Leguminosae, Moraceae, Zingiberaceae, and CannabaceaeXanthohumol; CorylifolininAntioxidant, antibacterial, anti-inflammatory, antiviral, and anticancer, etc.[42,43,44]
AnthocyanidinMolecules 30 01184 i0072-phenylbenzopyranyl cation structure; there is a double bond between the C1 and C2; there is a double bond between the C3 and C4. The C2 position is connected to the B ring.Blueberries, red cabbage, tomatoes, purple sweet potatoes, and eggplantDelphinidin; Cyanidin; Petunidin; Peonidin; Malvidin; PelargonidinEye health, cardiovascular disease, antiobesity, antidiabetes, antibacterial, anticancer activity and neurodegenerative diseases, etc.[25,45,46]
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

Hu, L.; Luo, Y.; Yang, J.; Cheng, C. Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability. Molecules 2025, 30, 1184. https://doi.org/10.3390/molecules30051184

AMA Style

Hu L, Luo Y, Yang J, Cheng C. Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability. Molecules. 2025; 30(5):1184. https://doi.org/10.3390/molecules30051184

Chicago/Turabian Style

Hu, Lei, Yiqing Luo, Jiaxin Yang, and Chunsong Cheng. 2025. "Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability" Molecules 30, no. 5: 1184. https://doi.org/10.3390/molecules30051184

APA Style

Hu, L., Luo, Y., Yang, J., & Cheng, C. (2025). Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability. Molecules, 30(5), 1184. https://doi.org/10.3390/molecules30051184

Article Metrics

Back to TopTop