Next Article in Journal
FTIR Detection of Ce3+ Sites on Shape-Controlled Ceria Nanoparticles Using Adsorbed 15N2 as a Probe Molecule
Previous Article in Journal
Volatile Compounds Profiling of Fresh R. alba L. Blossom by Headspace—Solid Phase Microextraction and Gas Chromatography
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress and Prospects of Flavonoids in the Treatment of Hyperlipidemia: A Narrative Review

School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(15), 3103; https://doi.org/10.3390/molecules30153103
Submission received: 1 July 2025 / Revised: 19 July 2025 / Accepted: 22 July 2025 / Published: 24 July 2025
(This article belongs to the Section Natural Products Chemistry)

Abstract

Hyperlipidemia (HLP) is a disorder of human lipid metabolism or transport, primarily characterized by abnormally elevated levels of total cholesterol (TC), triglycerides (TGs), and low-density lipoprotein cholesterol (LDL-C) in the blood. It is a key factor contributing to the development of non-alcoholic fatty liver disease, obesity, diabetes, atherosclerosis, and cardiovascular and cerebrovascular diseases. Statistics show that the prevalence of dyslipidemia among Chinese adults is as high as 35.6%, and it has shown a trend of younger onset in recent years, posing a serious threat to public health. Therefore, the prevention and treatment of dyslipidemia carry significant social significance. The pathogenesis of hyperlipidemia is complex and diverse, and currently used medications are often accompanied by side effects during treatment, making the research and development of new therapeutic approaches a current focus. Numerous studies have shown that flavonoids, which are abundant in most medicinal plants, fruits, and vegetables, exert effects on regulating lipid homeostasis and treating hyperlipidemia through a multi-target mechanism. These compounds have demonstrated significant effects in inhibiting lipid synthesis, blocking lipid absorption, promoting cholesterol uptake, enhancing reverse cholesterol transport, and suppressing oxidative stress, inflammation, and intestinal microbiota disorders. This article reviews the latest progress in the mechanisms of flavonoids in the treatment of hyperlipidemia, providing a theoretical basis for future research on drugs for hyperlipidemia.

1. Introduction

Hyperlipidemia is a disorder of lipid metabolism, clinically characterized by dyslipidemia, including elevated levels of total cholesterol (TC), triglycerides (TGs), and low-density lipoprotein cholesterol (LDL-C) or decreased high-density lipoprotein cholesterol (HDL-C). It serves as a risk factor for obesity, non-alcoholic fatty liver disease, and cardiovascular and cerebrovascular diseases such as atherosclerosis [1]. Current clinical medications for hyperlipidemia mainly include statins, fibrates, ezetimibe, PCSK9 inhibitors, and bile acid sequestrants [2]. Although these drugs effectively reduce lipid levels, they are often associated with side effects. For example, statins may cause myotoxicity and hepatotoxicity [3], while fibrates can lead to liver function deterioration and increased creatinine levels [2]. Due to the numerous adverse reactions associated with these drugs, research on natural products has received significant attention. A growing body of studies has demonstrated that traditional Chinese medicines and their extracts, as well as dietary flavonoids and their derivatives, hold great value and potential in the treatment of hyperlipidemia [4]. This paper is a narrative review that aims to summarize the research progress on the mechanism of flavonoids in the treatment of hyperlipidemia by reviewing the relevant literature.
Current research has gaps in mechanisms: for instance, the specific mechanisms of flavonoids in hyperlipidemia subtypes remain unclear, and the exploration of new targets such as circRNA is insufficient. Future studies should focus on these directions and promote clinical trials to clarify their clinical positioning in hyperlipidemia.

2. Flavonoids

In recent years, increasing attention has been paid to the research and application of traditional Chinese medicines, their extracts, and active ingredients in diets. Studies have shown that natural products in many traditional Chinese medicines, fruits, and vegetables can regulate lipids through multiple pathways, and there is ample evidence confirming that certain natural compounds have therapeutic potential for diseases related to hyperlipidemia [5]. These chemical substances offer obvious advantages, such as defined components, easy preparation, and relatively clear toxicity and side effects, making them important targets for studying lipid metabolism regulation. Numerous studies have found that traditional Chinese medicines, fruits, and vegetables contain a rich variety of flavonoids [6,7,8,9,10], which have obvious regulatory effects on lipid metabolism [11]. Flavonoids are a class of natural secondary metabolites widely present in TCMs, with multiple biological activities. They have a 2-phenylchromone structure and can be divided into seven main types based on the degree of oxidation of ring C (central three-carbon chain), ring connection patterns, and different substituents in their nuclear structure. These types include flavones, flavonols, dihydroflavones, isoflavones, flavanols, chalcones, and anthocyanidins [12]. Most flavonoids exhibit lipid-lowering effects, and the representative flavonoids with lipid-lowering activities in their main types will be described in detail below.

2.1. Flavones

Flavones, including apigenin, luteolin, chrysin, acacetin, baicalein, robinin, and chrysoeriol, have been extensively studied. Among these, apigenin and luteolin stand out for their pronounced lipid-lowering properties. Apigenin is a natural flavonoid compound widely present in Leonurus japonicus, olive leaves, chamomile, parsley, celery, Basella alba L., artichoke, etc. [13,14,15,16]. Structurally, its A-ring bears hydroxyl (-OH) substituents at the 5- and 7-positions, while the B-ring harbors a hydroxyl group at the 4’-position. These hydroxyl moieties endow apigenin with high polarity, facilitating interactions with water molecules and other polar entities. More importantly, they serve as critical functional groups underlying its multifaceted biological activities. Emerging evidence has highlighted apigenin’s diverse properties, including anti-inflammatory [17], antioxidant [18], anticancer [19], and anti-apoptotic properties [20] and its ability to reduce lipid levels and insulin resistance [21].
Luteolin: Luteolin is present in Taraxacum mongolicum, Chrysanthemum morifolium, lotus stems, Lonicera japonica, cabbage, celery, apples, oranges, pomegranates, lemons, etc. [22,23,24,25,26,27]. Its structure features multiple hydroxyl groups, which serve as critical functional moieties for antioxidant and other biological activities. Emerging research has revealed its diverse bioactivities, including anticancer effects [28], lipid-lowering and anti-inflammatory properties [29], modulation of intestinal microbiota [30], and immunoregulatory functions [31]. These attributes endow luteolin with promising potential for drug development and functional food applications.

2.2. Flavonols

Flavonol compounds comprise quercetin, kaempferol, morin, galangin, myricetin, rutin, isorhamnetin, etc., with quercetin and kaempferol being most representative for their lipid-lowering properties. Quercetin: Quercetin is commonly found in traditional Chinese medicines such as Astragalus membranaceus, Trichosanthes kirilowii, Albizia julibrissin flowers, and Morus alba leaves, as well as in fruits and vegetables including grapes, onions, carrots, and potatoes [32,33,34,35,36,37,38,39,40]. Its basic nucleus is phenyl benzoyl ketone, forming a C6-C3-C6 structure where two benzene rings (A and B) are linked by an oxygen-containing pyran ring (C ring). The planar configuration of the three-ring system confers relative molecular polarity, enabling diverse biochemical interactions. As a pivotal flavonol, Quercetin exhibits multifaceted bioactivities: ameliorating metabolic syndrome [41], antibacterial effects [42], neuroprotection [43], anti-ferroptotic activity [44], hypoglycemic and anti-obesity properties [45], and antirheumatic effects [46]. It also plays a critical role in chemotherapy by mitigating the adverse effects of cisplatin, daunorubicin, and tert-butyl hydroperoxide [47]. Furthermore, quercetin demonstrates synergistic effects with various compounds and drugs, such as menadione and amoxicillin [48]. Owing to its diverse biological activities and therapeutic potential, quercetin is widely recognized as one of the most significant flavonols in dietary sources and pharmaceutical research.
Kaempferol: It has been reported that kaempferol is present in Ginkgo biloba leaves, Carthamus tinctorius, Cnidium monnieri, blueberry leaves, figs, legumes, cauliflower, cabbage, strawberries, tea, and tomatoes [49,50,51,52,53,54,55]. Its core structure features the typical tricyclic system of flavonols, with two benzene rings (A and B rings) linked by a central three-carbon chain (C ring) to form a C6-C3-C6 skeleton. Kaempferol exhibits diverse pharmacological properties, including anticancer [56], anti-inflammatory [57], antioxidant [58], and anti-thrombotic activities and pleiotropic cardiovascular protective effects [59]. It also demonstrates therapeutic efficacy in Alzheimer’s disease [60]. Notably, kaempferol is being applied in cancer chemotherapy [61], holding significant research value in cancer treatment.

2.3. Dihydroflavones

Dihydroflavone derivatives include hesperidin, sakuranetin, isoliquiritigenin, naringenin, etc., with hesperidin being a paradigmatic example for its lipid-lowering activities. Hesperidin: Hesperidin is a flavonoid naturally abundant in citrus fruits such as Citrus sinensis and Citrus reticulata [62]. Structurally, it features the canonical tricyclic framework of flavonoids, where two benzene rings (A and B rings) are linked by a central three-carbon chain (C ring) to form a C6-C3-C6 skeleton. The C ring exists as a pyran ring with a single bond between C-2 and C-3 positions, adopting a cis configuration. Accumulating evidence indicates that hesperidin confers multifaceted benefits, including lipid-lowering [63], antioxidant [64], and anti-inflammatory [65] effects and neuroprotective effects against neurodegenerative disorders [31]. Therefore, daily intake of citrus fruits rich in hesperidin is associated with many health benefits.

2.4. Isoflavones

Isoflavone compounds include formononetin, isoliquiritin, maackiain, soy isoflavones, daidzein, calycosin, etc., among which formononetin is a representative example for its lipid-lowering effects. Formononetin: Also known as formononetin, it widely exists in leguminous plants (such as Astragalus membranaceus, Pueraria lobata, Glycyrrhiza uralensis, etc.), coffee beans, strawberries, and grapes [66,67,68,69,70]. Structurally, its C-ring serves as the bridge connecting the A and B rings, forming an unsaturated six-membered chromone ring. A critical feature is the double bond between C-2 and C-3 positions of the C-ring, endowing it with unsaturation—a defining structural characteristic of isoflavones. Studies from 1998 to 2019 have highlighted formononetin’s therapeutic potential in preventing and treating various diseases, including cancer, obesity, lipid-lowering, osteoarthritis, and neurodegenerative diseases [71,72].

2.5. Flavanols

Flavanol compounds include catechin, epicatechin, afzelechin, epigallocatechin, proanthocyanidins, trimeric flavanols, etc., with catechin being a representative example for its lipid-lowering effects. Catechin: It is a class of flavonoid compounds widely present in Ampelopsis japonica, Lycium barbarum, green tea, kiwifruit, etc. [73,74,75,76]. Its core structure is 2-phenylbenzopyran (flavanol), where two benzene rings (A and B rings) are linked by a three-carbon chain to form a pyran ring (C ring). The molecule contains multiple hydroxyl groups, endowing it with strong polarity and antioxidant properties. Studies have shown that catechin acts as a reactive oxygen species (ROS) scavenger and metal ion chelator, exerting antioxidant activity indirectly [77]. It also demonstrates the ability to delay muscle atrophy and enhance exercise capacity, thereby preventing, alleviating, delaying, or even treating muscle-related disorders caused by aging and diseases [78]. Additionally, catechin exhibits biological activities such as anticancer and lipid-lowering effects [79,80], holding promising potential for further development in pharmaceutical and food industries.

2.6. Chalcones

Chalcone compounds include licochalcone A, aurones, sulfuretin, carthamin, isoliquiritigenin A, morusin, etc., among which licochalcone A is a representative example for its lipid-lowering effects. Licochalcone A: Licochalcone A is predominantly found in Glycyrrhiza uralensis [81]. Its structure features two benzene rings linked by a three-carbon chain containing a central carbon–carbon double bond, forming an α,β-unsaturated ketone moiety. This unique conjugated system represents the key structural core for its biological activities. Owing to its beneficial effects, such as hypoglycemic regulation, lipid-lowering [82], and anti-allergic properties [83], licochalcone A has gained popularity in dietary applications.

2.7. Anthocyanidins

Anthocyanidin compounds include cyanidin-3-glucoside, anthocyanins, delphinidin, peonidin, malvidin, delphinidin-3-rutinoside, etc., among which cyanidin-3-glucoside is a representative example for its lipid-lowering effects. Cyanidin-3-glucoside: It is an important anthocyanin, mainly present in Artemisia argyi, mulberries, black wolfberries, purple cabbage, purple sweet potatoes, grapes, etc. [84,85,86]. Its molecular structure features a pyranidinium ring conjugated with two benzene rings, forming an extended conjugated system that dictates the compound’s color and chemical properties. Reported studies have shown that cyanidin-3-glucoside exhibits multiple biological activities, including anticancer [87] and lipid-lowering [88] activities and modulation of intestinal microbiota [89].
In conclusion, the seven classes of flavonoid compounds discussed above exhibit diverse biological activities, including lipid-lowering, anticancer, anti-inflammatory, and antioxidant effects, as summarized in Table 1. They play critical roles in the treatment of cancer, lipid metabolism disorders, and neurodegenerative diseases.

3. Mechanisms of Flavonoids in Treating Hyperlipidemia

Hyperlipidemia refers to elevated levels of total cholesterol (TC), triglycerides (TGs), or low-density lipoprotein cholesterol (LDL-C) in the blood, with pathological mechanisms involving pathophysiological processes across multiple systems. Flavonoids exert therapeutic effects against hyperlipidemia through multiple mechanisms, primarily by inhibiting lipid synthesis and absorption. The following summarizes the action mechanisms of the representative lipid-lowering flavonoids discussed above in treating hyperlipidemia in recent years.

3.1. Inhibition of Lipid Synthesis

3.1.1. Inhibition of Triglyceride Synthesis

SREBP has three subtypes: SREBP-1a, SREBP-1c, and SREBP-2. Among them, SREBP-1a is responsible for the synthesis of lipids and cholesterol; SREBP-1c mainly promotes the transcription of genes related to fatty acid and TG synthesis; SREBP-2 is a regulatory factor that regulates the transcription of HMGCR (3-Hydroxy-3-Methylglutaryl-Coenzyme A Reductase) and the expression of LDL receptors [107]. SREBP needs to go through a series of biological processes, and it can only exert its transcriptional regulatory effect after being cleaved into active fragments and entering the nucleus. First, SREBP structurally binds to SCAP (SREBP cleavage-activating protein), and the formed SCAP-SREBP complex is retained on the endoplasmic reticulum membrane by INSIG1 (Insulin-Induced Gene 1). Under certain conditions, after SCAP is recognized by the COPΙΙ complex, the SCAP-SREBP complex dissociates from INSIG1, allowing SREBP to be transported from the endoplasmic reticulum to the Golgi apparatus under the escort of SCAP. In the Golgi apparatus, SREBP is cleaved by Site-1 protease (S1P) and Site-2 protease (S2P) to form active fragments, and nSREBP is formed to enter the nucleus [108]. Studies have found that apigenin can promote the expression level of INSIG1 by inhibiting miR-363-3p in cells and inhibit the expression of mature SREBP1 and its transcriptional effect on downstream lipid synthesis genes ACC1, FASN, and SCD, thereby regulating lipid synthesis [109]. Licochalcone A can also affect lipid synthesis and treat hyperlipidemia by inhibiting SERBP1 [110].
The PI3K-Akt signaling pathway is closely associated with lipid synthesis. Inhibition of PI3K-Akt pathway activation blocks insulin metabolism and suppresses cellular lipid synthesis [111,112]. PPARγ (peroxisome proliferator-activated receptor gamma) serves as a key adipogenic transcription factor, regulating the expression of triglyceride synthesis-related key enzymes such as fatty acid synthase (FAS) [113]. Studies have shown that flavonoids including apigenin, quercetin, and luteolin can effectively inhibit the PI3K-Akt signaling pathway and PPARγ protein expression, thereby regulating intracellular triglyceride levels [114].
The AKT/mTOR/SREBP1 signaling pathway represents an essential axis in lipid metabolism. In this pathway, AKT activates mTOR to enhance SREBP1 expression, thereby accelerating fatty acid production [115]. Through a combination of network pharmacology, metabolomics, molecular docking, and in vitro experiments, Tao Chen et al. validated that quercetin effectively treats hyperlipidemia by inhibiting the AKT/mTOR/SREBP1-mediated fatty acid synthesis pathway [116].
Adiponectin (Adipo), an adipokine, specifically binds to adiponectin receptor 1 (AdipoR1) and adiponectin receptor 2 (AdipoR2). This binding activates the AMPK and PPAR-α signaling pathways, thereby inhibiting fatty acid synthase enzymes (SREBP-1c and FASN) [117]. Additionally, flavonoids such as kaempferol and hesperidin have been reported to regulate triglycerides by targeting AKT [118,119]. Licochalcone A inhibits the PPARγ/SREBP1 axis to block adipocyte differentiation and lipogenesis [120], while cyanidin-3-O-β-glucoside mediates transcription of triglyceride lipases [121]. Quercetin activates the Adipo/AdipoR2 signaling pathway to reduce adipogenesis [122].
Lipoprotein lipase (LPL), which hydrolyzes triglycerides (TGs) into free fatty acids (FFAs), serves as a critical determinant of TG metabolism. Studies have shown that phosphorylation of AMP-activated protein kinase (pAMPK) inhibits LPL, making the pAMPK-LPL signaling pathway one of the key regulators of TG metabolism. Cyanidin-3-O-β-glucoside modulates TG metabolism by acting on the pAMPK-LPL signaling pathway [123].

3.1.2. Inhibition of Cholesterol Synthesis

Endogenous cholesterol synthesis is one of the primary sources of cholesterol in the human body. The cholesterol biosynthesis pathway consists of approximately 33 steps, with HMGCR (3-Hydroxy-3-Methylglutaryl-Coenzyme A Reductase) serving as the rate-limiting enzyme, strictly regulating cholesterol homeostasis. INSIG-1 participates not only in the transcriptional and translational regulation of HMGCR but also in its ubiquitination-mediated degradation, dynamically modulating HMGCR activity based on intracellular cholesterol levels to maintain cholesterol homeostasis [124,125]. HMGCR activity can be inhibited to suppress cholesterol synthesis. For instance, AMPK phosphorylates HMGCR at the Ser872 site, thereby inhibiting its enzymatic activity [126]. Through transcriptomic analysis, Prasanth Puthanveetil et al. discovered that apigenin reduces cholesterol levels by inhibiting multiple transcripts involved in the cholesterol biosynthesis pathway, suggesting it may act by suppressing SREBP transcriptional targets [127]. Quercetin has also been reported to inhibit HMGCR activity to lower cholesterol levels [128]. 7-Dehydrocholesterol, a key intermediate in the cholesterol biosynthesis pathway, is converted to cholesterol under the catalysis of 7-dehydrocholesterol reductase (DHCR7) [129]. Cole Cochran leveraged untargeted metabolomics to prognosticate the therapeutic potential of apigenin, with in vitro experiments corroborating that apigenin downregulates 7-dehydrocholesterol—an intermediate in cholesterol biosynthesis—thus modulating lipid homeostasis [130].

3.2. Inhibition of Lipid Absorption

Niemann–Pick C1-like 1 (NPC1L1), predominantly expressed in intestinal epithelial cells, mediates endocytic uptake of exogenous cholesterol from the intestinal lumen [131]. Additionally, NPC1L1 contributes to the reabsorption of intestinal bile acids [132]. Reports have shown that luteolin exerts ezetimibe-mimetic effects by directly binding to NPC1L1, thereby inhibiting cholesterol absorption and treating hypercholesterolemia within a short timeframe [133]. Mari Nekohashi et al. demonstrated that both luteolin and quercetin suppress NPC1L1-mediated cholesterol uptake in Caco-2 and HEK293T cells [134]. Cholecystokinin (CCK), an appetite-regulating hormone, is commonly used to suppress appetite in obesity therapy. Hesperetin and hesperidin stimulate CCK release from enteroendocrine STC-1 cells, thereby reducing lipid intake [135].

3.3. Promotion of Cholesterol Uptake

Cholesterol uptake refers to the process by which cells acquire cholesterol from the bloodstream or the surrounding environment. Low-density lipoprotein (LDL) binds to LDL receptors (LDLRs) on the cell membrane, triggering endocytosis and the formation of endosomes. Under the acidic conditions within endosomes, LDL is released, while LDLRs recycle back to the cell surface for continuous LDL binding. Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the LDLR/LDL complex, inhibiting LDLR release and promoting its lysosomal degradation, thereby disrupting LDLR recycling [136,137]. Majambu Mbikay et al. supplemented the high-fat diet of mice with quercetin-3-glucoside and observed that it suppressed PCSK9 expression and secretion, enhanced LDLR-mediated cholesterol uptake, and reversed diet-induced steatohepatitis, hyperlipidemia, and hyperinsulinemia. These beneficial effects were also replicated in pancreatic β-cells of the same mice [138]. Additionally, studies have reported that catechins can treat hyperlipidemia by modulating LDL uptake [139].

3.4. Promotion of Reverse Cholesterol Transport (RCT)

Reverse cholesterol transport (RCT) is the process by which cholesterol is transported from peripheral tissues (e.g., macrophages, vascular wall cells) to the liver via plasma lipoproteins, followed by excretion as bile acids or free cholesterol. The major steps include cholesterol efflux, HDL (high-density lipoprotein) maturation and transport, hepatic uptake and metabolism, and cholesterol excretion [140]. ATP-Binding Cassette A1 (ABCA1) facilitates cholesterol efflux, relieving lipid accumulation by accelerating cellular cholesterol efflux to promote macrophage RCT and HDL formation. Kun Ren et al. established an atherosclerosis model using ApoE−/−mice fed a high-fat diet and injected with LPS and then treated these mice with apigenin. The results showed that apigenin reduced serum TG, total cholesterol (CHO), and LDL levels while increasing HDL levels, indicating its role in regulating plasma lipoprotein cholesterol. In vitro experiments further revealed that apigenin significantly enhanced ABCA1-mediated cholesterol efflux and decreased cholesterol (CHO), free cholesterol (FC), and cholesteryl ester (CE) levels in macrophage-derived foam cells [141]. ABCG1 (ATP-Binding Cassette G1) shares similar functions with ABCA1, transporting cholesterol to mature HDL to form CE-rich HDL particles. Scavenger receptor class B type 1 (SRB1) primarily mediates hepatic uptake of HDL-derived CE, converting it to bile acids for excretion—the final step of RCT [142]. Liver X receptor α (LXRα), abundantly expressed in the liver, adipose tissue, and macrophages, directly promotes RCT by upregulating ABCA1, ABCG1, and CYP7A1 [143]. Luteolin modulates cholesterol levels by regulating LXRα, ABCG1, and SRB1 expression in HepG2 hepatocytes and livers of diet-induced obese mice [144]. Bile acid metabolism represents the final step of RCT, with bile acid biosynthesis accounting for ~40% of cholesterol elimination [145]. Cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme in the classical bile acid synthesis pathway, is a promising target for hyperlipidemia therapy [146]. Dongliang Wang et al. demonstrated in vivo that cyanidin-3-O-glucoside upregulates hepatic cholesterol 7α-hydroxylase expression in high-fat diet-fed ApoE−/−male mice, reducing hypercholesterolemia [147].

3.5. Inhibition of Oxidative Stress

In a hyperlipidemic state, enhanced lipid peroxidation and reduced anti-lipid peroxidase activity occur. When oxygen radicals attack lipids, they disrupt hepatic lipid metabolism, triggering peroxidation reactions that generate excessive malondialdehyde (MDA). This further suppresses antioxidant enzyme activity, ultimately exacerbating oxidative stress and worsening hyperlipidemia [148]. Nrf2 (nuclear factor erythroid 2-related factor 2) resides primarily in the cytoplasm, chelated with Kelch-like Ech-associated protein (Keap1) and subjected to ubiquitination-mediated degradation. Upon activation by electrophilic agents like ROS (reactive oxygen species), Keap1 undergoes a conformational change, losing its ability to promote Nrf2 ubiquitination. This dissociates Keap1 from Nrf2, allowing Nrf2 accumulation and translocation to the nucleus, where it induces expression of antioxidant enzymes such as superoxide dismutase (SOD) and heme oxygenase-1 (HO-1) [149]. Jin-Ting Yang et al. found that luteolin not only reduced the myocardial lipid peroxidation product MDA but also enhanced nuclear translocation of Nrf2, upregulating downstream antioxidant genes NQO1 (quinone oxidoreductase 1) and HO-1 [150]. Hesperidin has been shown to protect against redox imbalance induced by hyperlipidemia in rats [151,152].

3.6. Promotion of Fatty Acid β-Oxidation

Fatty acid oxidation represents a critical metabolic process for energy production via fatty acid catabolism, playing a pivotal role in lipid homeostasis. Excessive fatty acid activation reduces β-oxidation, ultimately leading to hepatic lipid deposition. Fatty acid β-oxidation (FAβO), mediated by key catabolic enzymes, promotes β-oxidation of fatty acids to regulate lipid metabolism [153,154]. Jihan Sun et al. demonstrated that luteolin effectively lowers blood lipid levels in hyperlipidemic rats, enhances antioxidant enzyme activity, and reduces lipid peroxidation products. The mechanism may involve modulation of key enzymes regulating triglyceride levels, such as FAβO and fatty acid synthase (FAS) [155].

3.7. Regulation of Autophagy

Autophagic degradation of intracellular lipid droplets, termed lipophagy, decomposes triglycerides into free fatty acids (FFAs) within autolysosomes, thereby maintaining lipid homeostasis and preventing intracellular lipid accumulation [156]. Transcription factor EB (TFEB) serves as a key regulator of the autophagy–lysosome pathway, whose activity is inhibited by mTOR-mediated phosphorylation, leading to attenuated lipophagy [157]. AMPK activation phosphorylates and suppresses mTOR, counteracting this inhibitory effect [158]. Formononetin promotes the AMPK/TFEB pathway to ameliorate lipid accumulation in free fatty acid-stimulated HepG2 cells and primary mouse hepatocytes [159]. Zixuan Hu et al. treated atherosclerotic ApoE−/−mice with apigenin and observed reduced lipid levels. Western blot analysis of autophagy-related proteins demonstrated that apigenin may regulate lipid metabolism by activating autophagy in ApoE−/−mice [160].

3.8. Inhibition of Apoptosis

LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) serves as a specific receptor for oxidized low-density lipoprotein (ox-LDL), mediating ox-LDL uptake and intracellular signal transduction. This process promotes macrophage foam cell formation, induces lipid metabolism disorders, and contributes to diseases such as hyperlipidemia and atherosclerosis [161]. The Bcl-2 protein family plays a pivotal role in regulating apoptosis, with cellular apoptosis governed by the gene regulation of Bcl-2 and Bax. Consequently, most studies use the Bax/Bcl-2 gene ratio as an indicator of apoptotic status. Excessive hepatic lipid accumulation triggers hepatocyte apoptosis and injury, exacerbating hyperlipidemia and ultimately leading to liver dysfunction [162]. Qian Xu et al. demonstrated in vivo that apigenin effectively reduces serum lipid levels in hyperlipidemic rats, inhibits LOX-1 gene expression, and increases the Bcl-2/Bax ratio. These effects underlie its therapeutic actions against hyperlipidemia and prevention of atherosclerosis [163].

3.9. Inhibition of Inflammation

Activation of the NLRP3 (NOD-like receptor protein 3) inflammasome triggers the production of proinflammatory cytokines, such as interleukin (IL)-1β and IL-18. NLRP3 inflammasome involvement has been observed in cardiovascular diseases (CVDs), including hyperlipidemia, obesity, and atherosclerosis [164,165]. Zheng Lu et al. investigated the mechanism by which apigenin alleviates atherosclerosis and non-alcoholic fatty liver disease by inhibiting the NLRP3 inflammasome in mice. They found that apigenin reduces lipid accumulation by suppressing the NLRP3/NF-κB signaling pathway in HepG2 cells [166]. Other flavonoids, such as quercetin and hesperidin, exhibit similar effects [167,168].

3.10. Regulation of Gut Microbiota

Metabolomics and gut microbiomics have been widely applied to explore the mechanisms of drug therapy. The gut microbiota, composed of diverse microbial communities, produces metabolites that influence host metabolism. Emerging evidence has highlighted the critical role of the gut microbiota in the pathogenesis of hyperlipidemia [169,170]. Tongtong Wang et al. investigated the mechanism of quercetin in treating hyperlipidemia through plasma metabolomics and gut microbiome analysis. The study revealed that quercetin reduces lipid levels and improves liver function, potentially by regulating metabolism and the gut microbiota [171]. Tieqiao Wang explored the anti-obesity effects of kaempferol in high-fat diet (HFD)-fed mice and its impact on the gut microbiota, finding that kaempferol lowers blood lipid levels and modulates the gut microbiota to prevent obesity [172].
In conclusion, extensive studies have identified ten major mechanisms by which flavonoids treat hyperlipidemia, as illustrated in Figure 1 and Table 2. These mechanisms include the following: inhibition of lipid synthesis, suppression of lipid absorption, promotion of cholesterol uptake, enhancement of reverse cholesterol transport, and inhibition of oxidative stress. Additionally, flavonoids modulate fatty acid β-oxidation, autophagy, apoptosis, inflammation, and the gut microbiota.

4. Bioavailability of Flavonoids

Apigenin is a type of flavonoid with poor oral bioavailability, low solubility, and high intestinal membrane permeability. It has low solubility in non-polar solvents (0.001–1.63 mg/mL in non-polar solvents) or high hydrophilicity (1.35 μg/mL in pure water). At pH 7.5, the maximum solubility in phosphate buffer is 2.16 μg/mL [173,174,175,176]. Apigenin is mainly transported through passive and active carrier-mediated saturation mechanisms in the duodenum and jejunum, as well as passive transport mechanisms in the ileum and colon. The aglycone apigenin can be rapidly absorbed in the perfused rat intestinal model. Studies have shown that after a single oral administration of radiolabeled apigenin, 51.0% of the radioactivity was recovered in urine, 12.0% in feces, 1.2% in blood, 0.4% in the kidneys, and 9.4% in the intestines within 10 days. Various results such as glucuronic acid and sulfated conjugate metabolites collectively indicate that apigenin has slow metabolism, slow absorption, and a slow elimination phase [173,177]. Due to the limitations of apigenin in absorption and bioavailability, there is a need to develop new carriers to improve its oral bioavailability, for example, by incorporating it into phospholipid preparations, nanosuspensions, polymeric nanoparticles, and nanocrystals [16].
Studies have reported that after oral administration of luteolin, the plasma luteolin level (3.04 ± 0.60 μg/mL) reaches a peak at 0.42 h, indicating that it can be rapidly absorbed. In addition, luteolin (60 mg/kg) is rapidly absorbed after intraperitoneal injection in rats. In contrast, its concentration peaks at 0.083 h (71.99 ± 11.04 μg/mL) with a longer half-life (3.2 ± 0.7 h), which shows that the absorption of luteolin varies with the administration method [178,179,180]. Luteolin has a strong metabolic effect, so its bioavailability is low after oral administration. The poor solubility and stability of luteolin are important obstacles to its clinical development. Therefore, a large number of studies have attempted to modify the structure of luteolin to improve its bioavailability, such as luteolin tetraphosphate and metal complexes of luteolin [181,182]. In terms of distribution, the distribution of luteolin may be affected by its binding to plasma proteins. In plasma, luteolin has a high affinity for human serum albumin, thus limiting its distribution in tissues with low protein-binding capacity [183]. Luteolin and its metabolites are mainly excreted and eliminated through the kidneys and biliary tract. Studies have reported that after oral administration, the half-life of luteolin is between 1 and 6 h, and most of the drug is excreted within 24 h. The main metabolite detected in urine is glucuronide, accounting for about 60% of the total excretion [184,185,186].
Quercetin is sensitive to pH and highly hydrophobic, and it can precipitate in gastric and intestinal juices, resulting in low bioavailability or bioaccessibility. Studies have shown that most of the biological activities of quercetin can only be achieved after quercetin is absorbed by the human body. However, the absorption of intact quercetin by healthy adults is relatively low [187,188]. In order to improve its bioavailability, most researchers have integrated nanoemulsion technology into preparations to solve the shortcomings of poor water solubility and poor oral absorption [189]. Quercetin biotransformation occurs through xenobiotic metabolism, which consists of three phases that act independently and/or additively to limit the absorption and accumulation of xenobiotics: phase I, modification; phase II, conjugation; and phase III, elimination. Its phase II metabolites include quercetin monoglucuronide, quercetin diglucuronide, quercetin sulfate, etc. The phase II metabolites of quercetin secreted from the small intestine reach the liver through the portal vein for further metabolism. Ingested quercetin can be rapidly excreted through feces and urine [190,191,192,193]. Finally, most quercetin-derived metabolites are identified as 3-hydroxyphenylacetic acid, benzoic acid, and hippuric acid [194].
Kaempferol is a highly polar glycoside, so its absorption is poor, and it is directly absorbed into the hepatic portal vein [57,195,196]. Studies have shown that after oral intake of kaempferol, kaempferol exists in plasma at nanomolar concentrations. Kaempferol found in chicory was also administered to eight healthy volunteers (246 mg kaempferol per kg of chicory). At 5.8 h after oral administration of chicory containing 8.65 mg kaempferol, high plasma concentrations of 100 nM kaempferol-3-glucoside (79%), 3-glucoside (14%), and 3-(6-malonyl)-glucoside (7%) were observed. Kaempferol metabolites are excreted through urine and bile. Proportions of 1.9% and 2.5% of the total dose are excreted through urine and bile, respectively [197].
Hesperidin has poor solubility and faces obstacles to its absorption in the gastrointestinal tract [198,199]. Unlike aglycones, hesperidin has poor membrane permeability and is mainly absorbed through the paracellular pathway [200,201]. Studies have shown that in the case of neurodegenerative diseases, hesperidin can cross the blood–brain barrier to reach the disease site and act directly where pathological processes occur [202].
Formononetin is rapidly absorbed orally, with the maximum plasma concentration occurring between 30 and 60 min and a half-life of about 2 h. It is metabolized by the liver through cytochrome P450 enzymes and phase II conjugation [203]. Studies have shown that after oral administration of formononetin to rats, the oral bioavailability of formononetin is 21.8%, and its absorption in the small intestine is better than that in the large intestine [204]. The plasma protein binding of formononetin is 93.61 ± 0.44% and 96.14 ± 0.15%. The bioavailability of free/relatively unchanged formononetin is only about 3%. In addition, it was found that the clearance rate of formononetin is as high as 5.13 L/h/kg [205,206]. At present, most studies use combined nano-formulations to enhance the bioavailability of formononetin for better application [207,208].
Catechin has low bioavailability due to its low gastrointestinal absorption rate [209]. Studies have shown that the concentration of catechin metabolites decreases sharply within 12 to 24 h after an experimental meal. Piskula et al. reported that the plasma epicatechin metabolite concentration decreased rapidly 8 h after oral administration of catechin in rats, indicating that catechin has a fast metabolic rate [210].
Although licochalcone A has many physiological functions, its wide application is limited due to its low solubility and bioavailability. There are few reports on the bioavailability of licochalcone A. Studies have reported that licochalcone A has an impact on P-glycoprotein (P-gp) as well as CYP3A4 and 2C9 activities. Licochalcone A inhibits CYP3A4 and CYP2C9 enzyme activities, with 50% inhibitory concentrations (IC50) of 2.0 and 0.1 μM, respectively [211]. In recent years, studies have also reported the development of self-microemulsifying drug delivery systems loaded with licochalcone A and licochalcone A-integrated casein–pectin nano-delivery systems to enhance its oral bioavailability and improve intestinal digestibility [212,213].
The bioavailability of cyanidin-3-glucoside is low. The small intestine is the site with the highest absorption rate of cyanidin-3-glucoside. It has been reported that after rats eat food rich in cyanidin-3-glucoside for 15 days, the content of C3G in the jejunum is 605 nmol/g, and the metabolites are combined with glucuronic acid [214]. Although the bioavailability of cyanidin-3-glucoside in the human body is too low, a large number of experiments have shown that cyanidin-3-glucoside can be detected in tissues and organs. For example, after rats were fed with cyanidin-3-glucoside-rich blackberries for 15 days, cyanidin-3-glucoside was found in many organs such as the liver, brain, jejunum, stomach, kidney, and even bones [215,216].
In conclusion, although various flavonoids have a variety of biological activities, their bioavailability is not ideal, which greatly limits their clinical application. Current studies have shown that methods such as combined nano-formulations can be used to improve their bioavailability for clinical practice.

5. Safety of Flavonoids

Currently, there are no reports on the toxic and side effects of apigenin on the human body. For example, Shoubaky et al. evaluated the acute toxicity of apigenin and found that there were no deaths or signs of toxicity in mice or rats with oral doses as high as 5000 mg/kg [217,218]. Luteolin has low toxicity, and due to its anticancer biological activity, it has a certain degree of toxic and side effects on cancer cells [219,220]. Ding S et al. found that luteolin can promote the apoptosis of hepatocellular carcinoma cells but has no effect on normal cells [221]. In addition, it has been reported that long-term and excessive use of luteolin can induce the consumption of glutathione, activate the metabolism of CYP450 (such as CYP3A), and mediate the formation of o-benzoquinone metabolites, thereby causing cytotoxicity in primary rat hepatocytes [222]. Adverse reactions related to quercetin only exist in a few cases. On the whole, quercetin has few toxic and side effects. Studies have shown that no adverse events occurred in subjects after continuous intake of 500 mg of quercetin per day for 4 to 8 weeks, 730 mg for 4 consecutive weeks, 1000 mg for 5 consecutive days, or at least 2 weeks or 12 consecutive weeks of such intake [223,224,225]. Through research, Ruscinc N et al. found that kaempferol can cause adverse reactions of vascularization in the chorioallantoic membrane, which indicates that kaempferol is a non-irritating compound [226]. In addition, because kaempferol can act as a pro-oxidant, some studies have found that kaempferol has mutagenic and genotoxic effects [57]. Some potential benefits of hesperidin use include its safety, non-accumulation, and limited side effects. In safety studies, it was administered to mice at a dose as high as 5%, and even with a relatively long administration time, no mutagenic, toxic, or carcinogenic effects were reported [227]. Formononetin showed mortality at an acute dose of 300 mg/kg and an LD50 of 103.6 mg/kg BW, with a NOAEL of 50 mg/kg BW. All other acute and subacute doses are safe when administered intraperitoneally [228]. In addition, studies have shown that acute and subacute toxicity tests of formononetin were conducted by intraperitoneal injection in mice. The results showed that the acute dose of FMN was 300 mg/kg, the LD50 was 103.6 mg/kg, and the NOAEL was 50 mg/kg. In the subacute toxicity test, there were no changes in the animals’ body weight, food intake, water intake, and behavior and no toxic effects or pathological damage to the organs. Therefore, it is proven that formononetin is safe and non-toxic and can be used for pharmacological and therapeutic purposes [228]. Through clinical trials, Matsuyama T et al. found that children who consumed beverages rich in catechins every day did not show any growth impairment due to catechin intake, but instead showed an effect of improving obesity [229]. Studies have shown that licochalcone A (197.1 μM) can scavenge 77.92% of free radicals. Licochalcone A at a concentration of 147.75 μM or higher can cause cytotoxicity in Chinese hamster ovary fibroblasts; however, there was no significant toxicity in biochemical markers and body weight [230]. Cyanidin-3-glucoside is generally similar to the above substances in terms of safety. Studies have reported that no serious adverse events occurred during the 2-week human study of black bean seed coat extract Cyanidin-3-glucoside, and there was only one minor adverse event, which may be unrelated to the consumption of black beans [231].
In conclusion, various flavonoid compounds generally show good safety, but there are some differences. Apigenin and hesperidin have outstanding safety with no reported obvious toxic or side effects, even at high doses or with long-term use; only a few cases or mild reactions have been reported for luteolin, cyanidin-3-glucoside, etc. Overall, the safety of flavonoids is relatively high, but the safety of each compound still needs to be explored systematically and scientifically.

6. Conclusions

Hyperlipidemia, a disorder characterized by abnormal lipid metabolism, stands as a key risk factor for cardiovascular diseases (CVDs), encompassing non-alcoholic fatty liver disease (NAFLD), obesity, and atherosclerosis. Despite the fact that flavonoids exert multifaceted therapeutic effects on hyperlipidemia, with the seven classes of flavonoid compounds discussed in this paper possessing diverse biological activities such as lipid-lowering, anticancer, anti-inflammatory, and antioxidant properties and playing significant roles in the treatment of cancer, lipid metabolism disorders, and neurodegenerative diseases, their underlying mechanisms remain incompletely elucidated. Currently, ten major mechanisms by which flavonoids treat hyperlipidemia have been identified, including inhibition of lipid synthesis, suppression of lipid absorption, promotion of cholesterol uptake, enhancement of reverse cholesterol transport, inhibition of oxidative stress, regulation of fatty acid β-oxidation, autophagy, apoptosis, inflammation, and modulation of the gut microbiota. Nevertheless, issues such as the specific mechanism by which apigenin downregulates 7-dehydrocholesterol reductase (DHCR7) and the precise pathway through which hesperidin protects hyperlipidemic rats against redox imbalance remain to be addressed. Furthermore, although flavonoids have been shown to treat hyperlipidemia through multiple mechanisms, their overall bioavailability is less than ideal. To tackle this problem of low bioavailability, most current studies have incorporated them into technologies such as nanosuspensions, polymeric nanoparticles, and nanocrystals to enhance their clinical applicability. In terms of safety, flavonoids exhibit low toxic and side effects and high safety. Although only a few cases of adverse reactions have been reported, the safety evaluation of flavonoids still requires systematic and scientific investigation. Statistical data reveal that CVDs cause 17.9 million deaths annually, accounting for 31% of global mortality, thus ranking as the leading cause of death worldwide [232]. Therefore, continuously deepening the exploration of the mechanisms underlying the improvement of hyperlipidemia by flavonoids holds great significance, providing a theoretical basis for the development of novel therapeutic approaches for hyperlipidemia.

Author Contributions

Conceptualization, X.C. and J.Y.; methodology, X.C.; validation, X.C., J.Y. and Y.Z. (Yunyue Zhou); formal analysis, Q.W.; investigation, S.X.; resources, X.C.; data curation, J.Y.; writing—original draft preparation, X.C.; writing—review and editing, Wenying Niu; visualization, Wenying Niu; supervision, Y.Z. (Yukun Zhang); project administration, W.N. and Y.Z. (Yukun Zhang); funding acquisition, All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Natural Science Foundation of China (grant number 82274405) and the “Basic Research Support Program for Outstanding Young Teachers” of Heilongjiang Provincial Undergraduate Universities (grant number YQJH2024227).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare that there are no conflicts of interest in the article.

References

  1. Wang, K.X.; Xu, W.J.; Zhang, F.L.; Ding, M.Z.; Tan, X.M.; Xia, T. Investigation of Simiao Pill in Improving Lipid Deposition in Free Fatty Acid-Induced Hyperlipidemic HepG2 Cell Model Based on PIK3CA/AKT1/PPARG/CYP7A1 Signaling Pathway. Drug Eval. Res. 2025, 48, 1425–1437. [Google Scholar]
  2. Tian, Q.S.; Li, Y.L.; Pei, H.Y.; Tian, Y.Y.; Zuo, Z.P.; Zhao, X.Y.; Liu, C.; Wang, Z.B. Pathogenesis of Hyperlipidemia and Drug Therapy. Chem. Life 2022, 42, 2237–2247. [Google Scholar] [CrossRef]
  3. Zhou, Z.; Curtis, A.J.; Breslin, M.; Nelson, M. Regarding Article, Statin Toxicity: Mechanistic Insights and Clinical Implications. Circ. Res. 2019, 124, e120. [Google Scholar] [CrossRef] [PubMed]
  4. Shuai, P.; Cao, N.N.; Li, X.; Zang, W.J.; Yang, W.J.; Xiao, X.F. Research Progress on Signal Pathways Involved in Hyperlipidemia Intervened by Traditional Chinese Medicine. Drug Eval. Res. 2022, 45, 177–185. [Google Scholar]
  5. Nei, Y.; Zhang, M.; Huang, Y.W. Research Progress on Natural Compounds Inhibiting HMG-CoA Reductase for Improving Cardiovascular and Cerebrovascular Diseases. J. Food Saf. Qual. 2021, 12, 7333–7341. [Google Scholar] [CrossRef]
  6. Rupasinghe, H.P.V. Special Issue “Flavonoids and Their Disease Prevention and Treatment Potential”: Recent Advances and Future Perspectives. Molecules 2020, 25, 4746. [Google Scholar] [CrossRef] [PubMed]
  7. Manach, C.; Donovan, J.L. Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Radic. Res. 2004, 38, 771. [Google Scholar] [CrossRef] [PubMed]
  8. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 29, e47. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  9. Laveriano-Santos, E.P.; Arancibia-Riveros, C.; Tresserra-Rimbau, A.; Castro-Barquero, S.; Ruiz-León, A.M.; Estruch, R.; Casas, R.; Bodega, P.; de Miguel, M.; de Cos-Gandoy, A.; et al. Flavonoid Intake From Cocoa-Based Products and Adiposity Parameters in Adolescents in Spain. Front. Nutr. 2022, 6, 931171. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Perk, J.; De Backer, G.; Gohlke, H.; Graham, I.; Graham, I.; Reiner, Z.; Verschuren, M.; Albus, C.; Benlian, P.; Boysen, G.; et al. European Guidelines on cardiovascular disease prevention in clinical practice (version 2012): The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts). Atherosclerosis 2012, 223, 1635–1701. [Google Scholar]
  11. Chen, Z.; Zhang, M.; He, R.R.; Xu, J.; Nie, H. Research progress on the prevention and treatment of hyperlipidemia by plant flavones of medicinal and food homology. J. Liaoning Univ. Tradit. Chin. Med. 2024, 26, 215–220. [Google Scholar] [CrossRef]
  12. 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] [PubMed Central]
  13. Mo, X.X.; Chang, G.X.; Yuan, Y.; Yu, J.J.; Zheng, Y.G.; Wang, Y. The Research Progress on Flavonoid Chemical Constituents, Content Determination and Pharmacological Effects of Plants in Leonurus Genus. J. Yunnan Natl. Univ. Nat. Sci. Ed. 2025, 1–18. Available online: http://kns.cnki.net/kcms/detail/53.1192.n.20250319.1524.002.html (accessed on 19 March 2025).
  14. Wu, M.J.; Ge, J.X.; Yang, Y.F.; Wu, H.Z. The Research Progress on Chemical Constituents and Pharmacological Effects of Olea europaea Leaves and Predictive Analysis of Its Quality Markers. Cent. South Pharm. 2025, 23, 1726–1734. Available online: http://kns.cnki.net/kcms/detail/43.1408.R.20250427.1642.007.html (accessed on 19 March 2025).
  15. Li, J.J.; Chen, S.Y.; Lan, W. The Research Progress on Chemical Constituents and Pharmacological Effects of Chamomile and Prediction Analysis of Its Quality Markers. Chin. Pharm. J. 2024, 59, 664–675. [Google Scholar]
  16. Majma Sanaye, P.; Mojaveri, M.R.; Ahmadian, R.; Sabet Jahromi, M.; Bahramsoltani, R. Apigenin and its dermatological applications: A comprehensive review. Phytochemistry 2022, 203, 113390. [Google Scholar] [CrossRef] [PubMed]
  17. Yoon, J.H.; Kim, M.-Y.; Cho, J.Y. Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer. Int. J. Mol. Sci. 2023, 24, 1498. [Google Scholar] [CrossRef] [PubMed]
  18. Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef]
  19. Imran, M.; Aslam Gondal, T.; Atif, M.; Shahbaz, M.; Batool Qaisarani, T.; Hanif Mughal, M.; Salehi, B.; Martorell, M.; Sharifi-Rad, J. Apigenin as an anticancer agent. Phytother. Res. 2020, 34, 1812–1828. [Google Scholar] [CrossRef] [PubMed]
  20. Zafar, A.; Alruwaili, N.K.; Imam, S.S.; Alsaidan, O.A.; Ahmed, M.M.; Yasir, M.; Warsi, M.H.; Alquraini, A.; Ghoneim, M.M.; Alshehri, S. Development and Optimization of Hybrid Polymeric Nanoparticles of Apigenin: Physicochemical Characterization, Antioxidant Activity and Cytotoxicity Evaluation. Sensors 2022, 22, 1364. [Google Scholar] [CrossRef] [PubMed]
  21. Jiang, J.Y.; Tang, R.Y.; Zhang, J.N.; Chen, W.T.; Jia, D.Z.; Xiao, G.L. The Exploration of Apigenin on Hepatic Protection and Mechanisms in Hyperlipidemic Mice Based on Metabolomics Approach. Chin. J. New Drugs Clin. Pharmacol. 2024, 35, 1344–1351. [Google Scholar] [CrossRef]
  22. Ren, Y.R. The Study on Chemical Characterization of Taraxacum mongolicum in Vitro and in Vivo and Its Potential Mechanisms for Improving Hyperlipidemia. Beijing Univ. Chin. Med. 2023. [Google Scholar] [CrossRef]
  23. Yang, S.B.; Lu, J.Q.; Dong, L.; Xiao, X.; Mao, M.L.; Pang, Y.; Chen, J.Y.; Zhang, Y.L.; Chen, W.; Chen, L.D.; et al. The Research Progress on Chemical Components of Chrysanthemum and Its Prevention and Treatment of Cardiovascular Diseases. J. Liaoning Univ. Tradit. Chin. Med. 2025, 27, 95–99. [Google Scholar]
  24. Zhou, C.S.; Rao, G.X. The Study on Chemical Constituents of Nelumbo nucifera Stem. Chin. J. Ethnomed. Ethnopharm. 2024, 33, 28–31. [Google Scholar]
  25. Feng, W.S.; Chen, X.; Zheng, X.K.; Zhang, C.L.; Li, D.D. Study on Chemical Constituents of Honeysuckle. J. Chin. Pharm. Sci. 2011, 46, 338–340. [Google Scholar]
  26. Shuang, F.; Yaling, J.; Jinhai, L.; Cui, G.Y. Research progress on luteolin derivatives. Chem. Bull. 2024, 87, 300–309. [Google Scholar] [CrossRef]
  27. Xin, Z.; Fan, X.; Jun, Z.; He, Y.F.; Pei, X.J. Research progress on physiological functions and preparations of luteolin. China Surfactant Deterg. Cosmet. 2023, 53, 437–444. [Google Scholar]
  28. Rath, P.; Chauhan, A.; Ranjan, A.; Aggarwal, D.; Rani, I.; Choudhary, R.; Shahwan, M.; Ramniwas, S.; Joshi, H.; Haque, S.; et al. Luteolin: A promising modulator of apoptosis and survival signaling in liver cancer. Pathol. Res. Pract. 2024, 260, 155430. [Google Scholar] [CrossRef] [PubMed]
  29. Nabavi, S.F.; Braidy, N.; Gortzi, O.; Sobarzo-Sanchez, E.; Daglia, M.; Skalicka-Woźniak, K.; Nabavi, S.M. Luteolin as an anti-inflammatory and neuroprotective agent: A brief review. Brain Res. Bull. 2015, 119, 1–11. [Google Scholar] [CrossRef] [PubMed]
  30. Li, B.; Du, P.; Du, Y.; Zhao, D.; Cai, Y.; Yang, Q.; Guo, Z. Luteolin alleviates inflammation and modulates gut microbiota in ulcerative colitis rats. Life Sci. 2021, 269, 119008. [Google Scholar] [CrossRef] [PubMed]
  31. Huang, L.; Kim, M.Y.; Cho, J.Y. Immunopharmacological Activities of Luteolin in Chronic Diseases. Int. J. Mol. Sci. 2023, 24, 2136. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Liu, Y.L. Screening and Mechanistic Study of Active Components from Astragalus membranaceus Against Gastric Cancer. Ph.D. Thesis, Gansu Agricultural University, Lanzhou, China, 2024. [Google Scholar] [CrossRef]
  33. Li, X.; Cheng, B.B.; Tan, J.L.; Wan, J.Q.; Wang, H.Y.; Dai, A.G. Study on Quercetin, the Main Component of Astragalus membranaceus, in Regulating Ferroptosis of PASMCs via MAPK Pathway for Anti-Hypoxic Pulmonary Hypertension. J. Chin. Pharm. Sci. 2024, 33, 714–729. [Google Scholar]
  34. Sun, Y.N.; Yang, K.; Zhang, Z.Q. Study on Content Changes of Five Chemical Constituents in Trichosanthes kirilowii Before and After Compatibility with Aconitum kusnezoffii. Chin. Arch. Tradit. Chin. Med. 2022, 40, 240–246. [Google Scholar] [CrossRef]
  35. Li, R.; Tian, J.F.; Luo, X.F.; Li, M.R. Research Progress on Chemical Constituents and Pharmacological Effects of Albizia julibrissin Flowers. Tianjin Pharm. 2022, 34, 66–71. [Google Scholar]
  36. Lin, Q.F.; Zhang, N.; Chen, Z.W.; Zhang, L. Study on Chemical Constituents of Red Mulberry Leaves. J. Chin. Med. Mater. 2020, 43, 1364–1367. [Google Scholar] [CrossRef]
  37. Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  38. Changjiang, G.; Jing, X.; Jingyu, W.; Yang, J.J.; Wu, J.Q.; Gao, W.N. Contents of flavonoids in common vegetables in China. Acta Nutr. Sinica 2009, 31, 185–190. [Google Scholar]
  39. Junjie, Z. Identification and Metabolic Diversity of Flavonoids in Citrus Fruits. Master’s Thesis, Huazhong Agricultural University, Wuhan, China, 2018. [Google Scholar]
  40. Jingfang, W.; Ke, L.; Jinhai, L.; Cui, G.Y. Natural antioxidant-quercetin. Chem. Adhes. 2024, 46, 611–615+630. [Google Scholar]
  41. Hosseini, A.; Razavi, B.M.; Banach, M.; Hosseinzadeh, H. Quercetin and metabolic syndrome: A review. Phytother. Res. 2021, 35, 5352–5364. [Google Scholar] [CrossRef] [PubMed]
  42. Nguyen, T.L.A.; Bhattacharya, D. Antimicrobial Activity of Quercetin: An Approach to Its Mechanistic Principle. Molecules 2022, 27, 2494. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Chiang, M.C.; Tsai, T.Y.; Wang, C.J. The Potential Benefits of Quercetin for Brain Health: A Review of Anti-Inflammatory and Neuroprotective Mechanisms. Int. J. Mol. Sci. 2023, 24, 6328. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Wang, Y.; Wan, R.; Peng, W.; Zhao, X.; Bai, W.; Hu, C. Quercetin alleviates ferroptosis accompanied by reducing M1 macrophage polarization during neutrophilic airway inflammation. Eur. J. Pharmacol. 2023, 938, 175407. [Google Scholar] [CrossRef] [PubMed]
  45. Yi, R.; Liu, Y.; Zhang, X.; Sun, X.; Wang, N.; Zhang, C.; Deng, H.; Yao, X.; Wang, S.; Yang, G. Unraveling Quercetin’s Potential: A Comprehensive Review of Its Properties and Mechanisms of Action, in Diabetes and Obesity Complications. Phytother. Res. 2024, 38, 5641–5656. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, F.; Zhang, Y.; Zhou, J.; Cai, Y.; Li, Z.; Sun, J.; Xie, Z.; Hao, G. Metabolic effects of quercetin on inflammatory and autoimmune responses in rheumatoid arthritis are mediated through the inhibition of JAK1/STAT3/HIF-1α signaling. Mol. Med. 2024, 30, 170. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Alía, M.; Ramos, S.; Mateos, R.; Granado-Serrano, A.B.; Bravo, L.; Goya, L. Quercetin protects human hepatoma HepG2 against oxidative stress induced by tert-butyl hydroperoxide. Toxicol. Appl. Pharmacol. 2006, 212, 110. [Google Scholar] [CrossRef] [PubMed]
  48. Li, Q.C.; Liang, Y.; Hu, G.R.; Tian, Y. Enhanced therapeutic efficacy and amelioration of cisplatin-induced nephrotoxicity by quercetin in 1,2-dimethyl hydrazine-induced colon cancer in rats. Indian J. Pharmacol. 2016, 48, 168. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Dong, H.L. Study on Flavonoid Glycosides in the Bulk Drug of Ginkgo Leaf Injection. Master’s Thesis, Ningxia Medical University, Yinchuan, China, 2020. [Google Scholar] [CrossRef]
  50. Tursun, E.; Zhou, Z.B.; Wan, C.X. Isolation and Identification of Chemical Constituents from Xinjiang Safflower and Its Seed Oil. J. Tarim. Univ. 2017, 29, 1–6. [Google Scholar]
  51. Zheng, D.D. Study on Chemical Constituents of Cnidium monnieri. Master’s Thesis, Tianjin University of Traditional Chinese Medicine, Tianjin, China, 2020. [Google Scholar] [CrossRef]
  52. Jiao, H.Y.; Liu, H.; Xue, X.; Chen, X.F. Research Progress on Chemical Constituents and Biological Activities of Polyphenols from Blueberry Leaves. Guangzhou Chem. Ind. 2022, 50, 10–12+20. [Google Scholar]
  53. Dong, L.M.; Kong, L.X.; Chen, D.X.; Lan, L.P.; Wu, X.Y.; Zhao, T. Study on Flavonoid Chemical Constituents and Antioxidant Activity of Ficus carica. China Food Saf. Mag. 2023, 34, 93–95+99. [Google Scholar] [CrossRef]
  54. Calderón-Montaño, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A review on the dietary flavonoid kaempferol. Mini. Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef] [PubMed]
  55. Yang, Y.; Chen, Z.; Zhao, X.; Xie, H.; Du, L.; Gao, H.; Xie, C. Mechanisms of Kaempferol in the treatment of diabetes: A comprehensive and latest review. Front. Endocrinol. 2022, 13, 990299. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Nejabati, H.R.; Roshangar, L. Kaempferol: A potential agent in the prevention of colorectal cancer. Physiol. Rep. 2022, 10, e15488. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules 2020, 25, 4073. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  58. Hussain, M.S.; Altamimi, A.S.A.; Afzal, M.; Almalki, W.H.; Kazmi, I.; Alzarea, S.I.; Gupta, G.; Shahwan, M.; Kukreti, N.; Wong, L.S.; et al. Kaempferol: Paving the path for advanced treatments in aging-related diseases. Exp. Gerontol. 2024, 188, 112389. [Google Scholar] [CrossRef] [PubMed]
  59. Adebamowo, C.A.; Cho, E.; Sampson, L.; Katan, M.B.; Spiegelman, D.; Willett, W.C.; Holmes, M.D. Dietary flavonols and flavonol-rich foods intake and the risk of breast cancer. Int. J. Cancer 2005, 114, 628–633. [Google Scholar] [CrossRef] [PubMed]
  60. Dong, X.; Zhou, S.; Nao, J. Kaempferol as a therapeutic agent in Alzheimer’s disease: Evidence from preclinical studies. Ageing Res. Rev. 2023, 87, 101910. [Google Scholar] [CrossRef] [PubMed]
  61. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Aslam Gondal, T.; Saeed, F.; Imran, A.; Shahbaz, M.; Tsouh Fokou, P.V.; Umair Arshad, M.; Khan, H.; et al. Kaempferol: A Key Emphasis to Its Anticancer Potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, J.J.; Shu, J.B.; Chen, J.P.; Su, W.Y.; Wu, Q.; Zhu, W.D. Quality Evaluation of Tangerine Peel from Different Botanical Origins. Chin. Arch. Tradit. Chin. Med. 2025, 1–21. Available online: http://kns.cnki.net/kcms/detail/21.1546.R.20250609.1519.013.html (accessed on 10 June 2025).
  63. Wang, Q.R.; Liu, R.X.; He, X.; Zhang, T.; Zhao, X.H. Research Progress on Pharmacological Effects and Solubility-Enhancing Technologies of Hesperidin. Prog. Vet. Med. 2025, 46, 91–96. [Google Scholar] [CrossRef]
  64. Pyrzynska, K. Hesperidin: A Review on Extraction Methods, Stability and Biological Activities. Nutrients 2022, 14, 2387. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Buzdağlı, Y.; Eyipınar, C.D.; Kacı, F.N.; Tekin, A. Effects of hesperidin on anti-inflammatory and antioxidant response in healthy people: A meta-analysis and meta-regression. Int. J. Environ. Health Res. 2023, 33, 1390–1405. [Google Scholar] [CrossRef] [PubMed]
  66. Li, X.; Huang, W.; Tan, R.; Xu, C.; Chen, X.; Li, S.; Liu, Y.; Qiu, H.; Cao, H.; Cheng, Q. The benefits of hesperidin in central nervous system disorders, based on the neuroprotective effect. Biomed. Pharmacother. 2023, 159, 114222. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, L.; Deng, S.J.; Zhang, Y.G.; Cui, X.H.; Yu, W.B. Study on Extraction, Isolation and Action Mechanism of Anti-Inflammatory Active Components from Pueraria lobata. J. Tonghua Norm. Univ. 2025, 46, 68–80. [Google Scholar] [CrossRef]
  68. Lu, H.Y. Study on Formononetin from Astragalus membranaceus Combined with Intermittent Exercise in Alleviating Myocardial Injury in Myocardial Infarction Rats. Mol. Plant Breed. 2024, 22, 7556–7563. [Google Scholar] [CrossRef]
  69. Tian, J.M.; Liu, H.; Bai, X.M.; Jia, T.J. Determination of Formononetin Content in Flavonoids of Glycyrrhiza uralensis by High Performance Liquid Chromatography. Chin. Tradit. Pat. Med. 2007, 8, 1232–1233. [Google Scholar]
  70. Aliya, S.; Alhammadi, M.; Park, U.; Tiwari, J.N.; Lee, J.H.; Han, Y.K.; Huh, Y.S. The potential role of formononetin in cancer treatment: An updated review. Biomed. Pharmacother. 2023, 168, 115811. [Google Scholar] [CrossRef] [PubMed]
  71. Machado Dutra, J.; Espitia, P.J.P.; Andrade Batista, R. Formononetin: Biological effects and uses—A review. Food Chem. 2021, 359, 129975. [Google Scholar] [CrossRef] [PubMed]
  72. Jia, C.; Hu, F.; Lu, D.; Jin, H.; Lu, H.; Xue, E.; Wu, D. Formononetin inhibits IL-1β-induced inflammation in human chondrocytes and slows the progression of osteoarthritis in rat model via the regulation of PTEN/AKT/NF-κB pathway. Int. Immunopharmacol. 2022, 113 Pt A, 109309. [Google Scholar] [CrossRef] [PubMed]
  73. Zheng, H.X.; Niu, Q.Y.; Zhang, X.; Wen, X.L.; Huang, S.H.; Chen, L.P.; Huang, L.P.; Liu, N.H. Research Progress on Chemical Constituents, Pharmacological Effects, Medicinal Property and Prediction of Quality Markers (Q-Markers) of Ampelopsis japonica. Pharmacol. Clin. Chin. Mater. Medica 2025, 1–16. [Google Scholar] [CrossRef]
  74. Mi, J.; Wei, J.Y.; Lu, L.; Zhang, L.T.; Jin, B.; Li, X.Y.; Yan, Y.M. Comparison of Total Flavonoid Content and Antioxidant Activity in Lycium barbarum Determined by Different Colorimetric Methods. Food Res. Dev. 2025, 46, 155–162. [Google Scholar]
  75. Kang, Y.S.; Ryu, C.; Suguri, M.; Park, S.B.; Kishino, S.; Onoyama, H. Estimating the catechin concentrations of new shoots in green tea fields using ground-based hyperspectral imagery. Food Chem. 2022, 370, 130987. [Google Scholar] [CrossRef] [PubMed]
  76. Arts, I.C.; van de Putte, B.; Hollman, P.C. Catechin contents of foods commonly consumed in The Netherlands. 1. Fruits, vegetables, staple foods, and processed foods. J. Agric. Food Chem. 2000, 48, 1746. [Google Scholar] [CrossRef] [PubMed]
  77. Bernatoniene, J.; Kopustinskiene, D.M. The Role of Catechins in Cellular Responses to Oxidative Stress. Molecules 2018, 23, 965. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  78. Li, P.; Liu, A.; Xiong, W.; Lin, H.; Xiao, W.; Huang, J.; Zhang, S.; Liu, Z. Catechins enhance skeletal muscle performance. Crit. Rev. Food Sci. Nutr. 2020, 60, 515–528. [Google Scholar] [CrossRef] [PubMed]
  79. Liu, Y.; Long, Y.; Fang, J.; Liu, G. Advances in the Anti-Atherosclerotic Mechanisms of Epigallocatechin Gallate. Nutrients 2024, 16, 2074. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  80. Yang, C.S.; Wang, H. Cancer Preventive Activities of Tea Catechins. Molecules 2016, 21, 1679. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  81. Zhang, Z.; Yang, J.Y.; Wang, X.X.; Wang, Y.; Liu, R.X.; Chen, W.; Li, H. Determination of Total Flavonoids and Liquiritigenin A in Licorice Residue and Optimization of Extraction-Purification Process. Chem. Bio-Eng. 2024, 41, 55–61. [Google Scholar]
  82. Li, M.T.; Xie, L.; Jiang, H.M.; Huang, Q.; Tong, R.S.; Li, X.; Xie, X.; Liu, H.M. Role of Licochalcone A in Potential Pharmacological Therapy: A Review. Front. Pharmacol. 2022, 13, 878776. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Shu, J.; Cui, X.; Liu, X.; Yu, W.; Zhang, W.; Huo, X.; Lu, C. Licochalcone A inhibits IgE-mediated allergic reaction through PLC/ERK/STAT3 pathway. Int. J. Immunopathol. Pharmacol. 2022, 36, 3946320221135462. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Chen, C.J.; Du, H.Z.; Miao, Y.H.; Fang, Y.; Zhao, T.T.; Liu, D.H. Predictive Analysis of Quality Markers and Resource Evaluation of Artemisia argyi Based on Chemical Components and Network Pharmacology. China J. Chin. Mater. Medica 2023, 48, 5474–5486. [Google Scholar] [CrossRef]
  85. Liu, Y.; Tan, L.L.; Lan, Y.Y. Research Progress on Chemical Constituents, Pharmacological Effects and Quality Markers of Fructus Mori. Chongqing Med. 2021, 50, 1063–1067. [Google Scholar]
  86. Gan, X.N.; Wang, H.J.; Li, Y.Z.; Li, B. UPLC-Triple TOF/MS Analysis of Chemical Components and Determination of Total Anthocyanins in Lycium ruthenicum. Food Sci. 2021, 42, 185–190. [Google Scholar]
  87. Safdar, M.A.; Aslam, R.M.N.; Shakeel, A.; Shiza, W.M.; Jmail, A.; Mehmood, M.H.; Gul, H. Cyanidin as potential anticancer agent targeting various proliferative pathways. Chem. Biol. Drug. Des. 2023, 101, 438–452. [Google Scholar] [CrossRef] [PubMed]
  88. Li, X.; Shi, Z.; Zhu, Y.; Shen, T.; Wang, H.; Shui, G.; Loor, J.J.; Fang, Z.; Chen, M.; Wang, X.; et al. Cyanidin-3-O-glucoside improves non-alcoholic fatty liver disease by promoting PINK1-mediated mitophagy in mice. Br. J. Pharmacol. 2020, 177, 3591–3607. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Cheng, Z.; Si, X.; Tan, H.; Zang, Z.; Tian, J.; Shu, C.; Sun, X.; Li, Z.; Jiang, Q.; Meng, X.; et al. Cyanidin-3-O-glucoside and its phenolic metabolites ameliorate intestinal diseases via modulating intestinal mucosal immune system: Potential mechanisms and therapeutic strategies. Crit. Rev. Food Sci. Nutr. 2023, 63, 1629–1647. [Google Scholar] [CrossRef] [PubMed]
  90. Zhang, F.; Li, F.; Chen, G. Neuroprotective effect of apigenin in rats after contusive spinal cord injury. Neurol. Sci. 2014, 35, 583–588. [Google Scholar] [CrossRef] [PubMed]
  91. Han, Y.; Zhang, T.; Su, J.; Zhao, Y.; Wang, C.C.; Li, X. Apigenin attenuates oxidative stress and neuronal apoptosis in early brain injury following subarachnoid hemorrhage. J. Clin. Neurosci. 2017, 40, 157–162. [Google Scholar] [CrossRef] [PubMed]
  92. Wu, L.; Guo, T.; Deng, R.; Liu, L.; Yu, Y. Apigenin Ameliorates Insulin Resistance and Lipid Accumulation by Endoplasmic Reticulum Stress and SREBP-1c/SREBP-2 Pathway in Palmitate-Induced HepG2 Cells and High-Fat Diet-Fed Mice. J. Pharmacol. Exp. Ther. 2021, 377, 146–156. [Google Scholar] [CrossRef] [PubMed]
  93. Jiang, Z.B.; Wang, W.J.; Xu, C.; Xie, Y.J.; Wang, X.R.; Zhang, Y.Z.; Huang, J.M.; Huang, M.; Xie, C.; Liu, P.; et al. Luteolin and its derivative apigenin suppress the inducible PD-L1 expression to improve anti-tumor immunity in KRAS-mutant lung cancer. Cancer Lett. 2021, 515, 36–48. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, J.F.; Ma, Y.; Wang, Y.; Du, Z.Y.; Shen, J.K.; Peng, H.L. Reduction of lipid accumulation in HepG2 cells by luteolin is associated with activation of AMPK and mitigation of oxidative stress. Phytother. Res. 2011, 25, 588–596. [Google Scholar] [CrossRef] [PubMed]
  95. Hung, T.W.; Chen, P.N.; Wu, H.C.; Wu, S.W.; Tsai, P.Y.; Hsieh, Y.S.; Chang, H.R. Kaempferol Inhibits the Invasion and Migration of Renal Cancer Cells through the Downregulation of AKT and FAK Pathways. Int. J. Med. Sci. 2017, 14, 984–993. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  96. Zhu, X.; Wang, X.; Ying, T.; Li, X.; Tang, Y.; Wang, Y.; Yu, T.; Sun, M.; Zhao, J.; Du, Y.; et al. Kaempferol alleviates the inflammatory response and stabilizes the pulmonary vascular endothelial barrier in LPS-induced sepsis through regulating the SphK1/S1P signaling pathway. Chem. Biol. Interact. 2022, 368, 110221. [Google Scholar] [CrossRef] [PubMed]
  97. Kouhestani, S.; Jafari, A.; Babaei, P. Kaempferol attenuates cognitive deficit via regulating oxidative stress and neuroinflammation in an ovariectomized rat model of sporadic dementia. Neural. Regen. Res. 2018, 13, 1827–1832. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  98. Chen, H.; Nie, T.; Zhang, P.; Ma, J.; Shan, A. Hesperidin attenuates hepatic lipid accumulation in mice fed high-fat diet and oleic acid induced HepG2 via AMPK activation. Life Sci. 2022, 296, 120428. [Google Scholar] [CrossRef] [PubMed]
  99. Megahed, A.; Gadalla, H.; Filimban, W.A.; Albukhari, T.A.; Sembawa, H.; Bagadood, R.M.; Sindi, G.; Abdelhamid, F.M.; El-Boshy, M.E.; Risha, E.F. Hesperidin ameliorates thioacetamide-induced liver fibrosis via antioxidative and anti-inflammatory mechanisms targeting TGF-β/α-SMA pathways in rats. Int. J. Immunopathol. Pharmacol. 2024, 38, 3946320241309004. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  100. Yurtal, Z.; Altug, M.E.; Unsaldi, E.; Secinti, I.E.; Kucukgul, A. Investigation of Neuroprotective and Therapeutic Effects of Hesperidin in Experimental Spinal Cord Injury. Turk. Neurosurg. 2020, 30, 899–906. [Google Scholar] [CrossRef] [PubMed]
  101. Ma, C.; Xia, R.; Yang, S.; Liu, L.; Zhang, J.; Feng, K.; Shang, Y.; Qu, J.; Li, L.; Chen, N.; et al. Formononetin attenuates atherosclerosis via regulating interaction between KLF4 and SRA in apoE mice. Theranostics 2020, 10, 1090–1106. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  102. Ng, T.K.; Chu, K.O.; Wang, C.C.; Pang, C.P. Green Tea Catechins as Therapeutic Antioxidants for Glaucoma Treatment. Antioxidants 2023, 12, 1320. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  103. Luo, Z.; Guo, Z.; Xiao, T.; Liu, H.; Su, G.; Zhao, Y. Enrichment of total flavones and licochalcone A from licorice residues and its hypoglycemic activity. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1114–1115, 134–145. [Google Scholar] [CrossRef] [PubMed]
  104. Quan, H.Y.; Kim, S.J.; Kim, D.Y.; Jo, H.K.; Kim, G.W.; Chung, S.H. Licochalcone A regulates hepatic lipid metabolism through activation of AMP-activated protein kinase. Fitoterapia 2013, 86, 208. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, C.F.; Wu, C.Y.; Lin, C.F.; Liu, Y.W.; Lin, T.C.; Liao, H.J.; Chang, G.R. The anticancer effects of cyanidin 3-O-glucoside combined with 5-fluorouracil on lung large-cell carcinoma in nude mice. Biomed. Pharmacother. 2022, 151, 113128. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, T.; Jiang, G.; Lv, S.; Xiao, Y.; Fan, C.; Zou, M.; Wang, Y.; Guo, Q.; Kabir, M.A.; Peng, X. Avian safety guardian: Luteolin restores Mycoplasma gallisepticum-induced immunocompromise to improve production performance via inhibiting the IL-17/NF-kB pathway. Int. Immunopharmacol. 2023, 124 Pt B, 110946. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, L.L.; Zhang, S.J.; Ma, P.; Dan, J.H. Research Progress on the Regulation of SREBP2 Expression and Activity. Chin. J. Cell. Biol. 2021, 43, 2441–2448. [Google Scholar]
  108. Zhang, S.J.; Wang, S.; Zhang, T.T.; Jia, S.T.; Dan, J.H. Research Progress on the Regulation of HMGCR Expression and Activity. Chin. J. Cell. Biol. 2023, 45, 990–996. [Google Scholar]
  109. Wang, L.C. Mechanistic Study on Apigenin Inhibiting Non-Alcoholic Fatty Liver Disease. Master’s Thesis, Tianjin University of Science and Technology, Tianjin, China, 2023. [Google Scholar] [CrossRef]
  110. Oh, G.S.; Lee, G.G.; Yoon, J.; Oh, W.K.; Kim, S.W. Selective inhibition of liver X receptor α-mediated lipogenesis in primary hepatocytes by licochalcone A. Chin. Med. 2015, 10, 8. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  111. Zhang, X.; He, B.; Chen, E.; Lu, J.; Wang, J.; Cao, H.; Li, L. The aryl hydrocarbon receptor ligand ITE inhibits cell proliferation and migration and enhances sensitivity to drug-resistance in hepatocellular carcinoma. J. Cell. Physiol. 2021, 236, 178–192. [Google Scholar] [CrossRef] [PubMed]
  112. Kim, G.S.; Park, H.J.; Woo, J.H.; Kim, M.K.; Koh, P.O.; Min, W.; Ko, Y.G.; Kim, C.H.; Won, C.K.; Cho, J.H. Citrus aurantium flavonoids inhibit adipogenesis through the Akt signaling pathway in 3T3-L1 cells. BMC Complement Altern. Med. 2012, 12, 31. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  113. Farmer, S.R. Transcriptional control of adipocyte formation. Cell. Metab. 2006, 4, 263. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  114. Li, T.; Zhang, L.; Jin, C.; Xiong, Y.; Cheng, Y.Y.; Chen, K. Pomegranate flower extract bidirectionally regulates the proliferation, differentiation and apoptosis of 3T3-L1 cells through regulation of PPARγ expression mediated by PI3K-AKT signaling pathway. Biomed. Pharmacother. 2020, 131, 110769. [Google Scholar] [CrossRef] [PubMed]
  115. Yin, F.; Sharen, G.; Yuan, F.; Peng, Y.; Chen, R.; Zhou, X.; Wei, H.; Li, B.; Jing, W.; Zhao, J. TIP30 regulates lipid metabolism in hepatocellular carcinoma by regulating SREBP1 through the Akt/mTOR signaling pathway. Oncogenesis 2017, 6, e347. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Chen, T.; Wang, T.; Shi, Y.; Deng, J.; Yan, X.; Zhang, C.; Yin, X.; Liu, W. Integrated network pharmacology, metabolomics and molecular docking analysis to reveal the mechanisms of quercetin in the treatment of hyperlipidemia. J. Pharm. Biomed. Anal. 2025, 252, 116507. [Google Scholar] [CrossRef] [PubMed]
  117. Iwabu, M.; Okada-Iwabu, M.; Yamauchi, T.; Kadowaki, T. Adiponectin/AdipoR Research and Its Implications for Lifestyle-Related Diseases. Front. Cardiovasc. Med. 2019, 6, 116. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  118. Hoang, M.H.; Jia, Y.; Lee, J.H.; Kim, Y.; Lee, S.J. Kaempferol reduces hepatic triglyceride accumulation by inhibiting Akt. J. Food Biochem. 2019, 43, e13034. [Google Scholar] [CrossRef] [PubMed]
  119. Morikawa, K.; Nonaka, M.; Mochizuki, H.; Handa, K.; Hanada, H.; Hirota, K. Naringenin and hesperetin induce growth arrest, apoptosis, and cytoplasmic fat deposit in human preadipocytes. J. Agric. Food Chem. 2008, 56, 11030. [Google Scholar] [CrossRef] [PubMed]
  120. Quan, H.Y.; Baek, N.I.; Chung, S.H. Licochalcone A prevents adipocyte differentiation and lipogenesis via suppression of peroxisome proliferator-activated receptor γ and sterol regulatory element-binding protein pathways. J. Agric. Food Chem. 2012, 60, 5112. [Google Scholar] [CrossRef] [PubMed]
  121. Guo, H.; Guo, J.; Jiang, X.; Li, Z.; Ling, W. Cyanidin-3-O-β-glucoside, a typical anthocyanin, exhibits antilipolytic effects in 3T3-L1 adipocytes during hyperglycemia: Involvement of FoxO1-mediated transcription of adipose triglyceride lipase. Food Chem. Toxicol. 2012, 50, 3040. [Google Scholar] [CrossRef] [PubMed]
  122. Sannappa Gowda, N.G.; Shiragannavar, V.D.; Karunakara, S.H.; Veeranna, R.P.; Suvarna, D.; Kumar, D.P.; Santhekadur, P.K. Novel role of Quercetin in ameliorating metabolic syndrome via VDR mediated activation of adiponectin/AdipoR2 signaling. Biochem. Biophys. Rep. 2024, 39, 101754. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  123. Wei, X.; Wang, D.; Yang, Y.; Xia, M.; Li, D.; Li, G.; Zhu, Y.; Xiao, Y.; Ling, W. Cyanidin-3-O-β-glucoside improves obesity and triglyceride metabolism in KK-Ay mice by regulating lipoprotein lipase activity. J. Sci. Food Agric. 2011, 91, 1006. [Google Scholar] [CrossRef] [PubMed]
  124. Bai, X.; Wang, S.; Shu, L.; Cao, Q.Y.; Hu, H.M.; Zhu, Y.C. Hawthorn leaf flavonoids alleviate the deterioration of atherosclerosis by inhibiting SCAP-SREBP2-LDLR pathway through sPLA2-ⅡA signaling in macrophages in mice. J. Ethnopharmacol. 2024, 327, 118006. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, X.T.; Yang, J.B.; Zhou, Y.Y.; Yang, S.H.; Yang, R.H.; Fang, S.J.; Ma, Y.X.; Niu, W.Y. Research Progress on the Regulatory Mechanisms of 3-Hydroxy-3-Methylglutaryl-Coenzyme A Reductase. Chin. J. Pathophysiol. 2025, 41, 791–797. [Google Scholar]
  126. Clarke, P.R.; Hardie, D.G. Regulation of HMG-CoA reductase: Identification of the site phosphorylated by the AMP—Activated protein kinase in vitro and in intact rat liver. EMBO J. 1990, 9, 2439–2446. [Google Scholar] [CrossRef] [PubMed]
  127. Puthanveetil, P.; Kong, X.; Bräse, S.; Voros, G.; Peer, W.A. Transcriptome analysis of two structurally related flavonoids; Apigenin and Chrysin revealed hypocholesterolemic and ketogenic effects in mouse embryonic fibroblasts. Eur. J. Pharmacol. 2021, 893, 173804. [Google Scholar] [CrossRef] [PubMed]
  128. Yi, H.; Peng, H.; Wu, X.; Xu, X.; Kuang, T.; Zhang, J.; Du, L.; Fan, G. The Therapeutic Effects and Mechanisms of Quercetin on Metabolic Diseases: Pharmacological Data and Clinical Evidence. Oxid. Med. Cell. Longev. 2021, 2021, 6678662. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  129. Cui, S.; Ye, J. 7-Dehydrocholesterol: A sterol shield against an iron sword. Mol. Cell 2024, 84, 1183–1185. [Google Scholar] [CrossRef] [PubMed]
  130. Cochran, C.; Martin, K.; Rafferty, D.; Choi, J.; Leontyev, A.; Shetty, A.; Kurup, S.; Puthanveetil, P. Untargeted Metabolomics Based Prediction of Therapeutic Potential for Apigenin and Chrysin. Int. J. Mol. Sci. 2023, 24, 4066. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  131. Zhang, R.; Liu, W.; Zeng, J.; Meng, J.; Shi, L.; Yang, S.; Chang, J.; Wang, C.; Xing, K.; Wen, J.; et al. Recent advances in the screening methods of NPC1L1 inhibitors. Biomed. Pharmacother. 2022, 155, 113732. [Google Scholar] [CrossRef] [PubMed]
  132. Xiao, J.; Dong, L.W.; Liu, S.; Meng, F.H.; Xie, C.; Lu, X.Y.; Zhang, W.J.; Luo, J.; Song, B.L. Bile acids-mediated intracellular cholesterol transport promotes intestinal cholesterol absorption and NPC1L1 recycling. Nat. Commun. 2023, 14, 6469. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  133. Kobayashi, S. The Effect of Polyphenols on Hypercholesterolemia through Inhibiting the Transport and Expression of Niemann-Pick C1-Like 1. Int. J. Mol. Sci. 2019, 20, 4939. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  134. Nekohashi, M.; Ogawa, M.; Ogihara, T.; Nakazawa, K.; Kato, H.; Misaka, T.; Abe, K.; Kobayashi, S. Luteolin and quercetin affect the cholesterol absorption mediated by epithelial cholesterol transporter niemann-pick c1-like 1 in caco-2 cells and rats. PLoS ONE 2014, 9, e97901. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  135. Dudhia, Z.; Louw, J.; Muller, C.; Joubert, E.; de Beer, D.; Kinnear, C.; Pheiffer, C. Cyclopia maculata and Cyclopia subternata (honeybush tea) inhibits adipogenesis in 3T3-L1 pre-adipocytes. Phytomedicine 2013, 20, 401. [Google Scholar] [CrossRef] [PubMed]
  136. Han, L.; Wu, L.; Yin, Q.; Li, L.; Zheng, X.; Du, S.; Huang, X.; Bai, L.; Wang, Y.; Bian, Y. A promising therapy for fatty liver disease: PCSK9 inhibitors. Phytomedicine 2024, 128, 155505. [Google Scholar] [CrossRef] [PubMed]
  137. Qi, C.; Fan, D.; Wang, L.; Guo, L.; Jiang, H.; Wang, L. Mechanisms of action and therapeutic potential of PCSK9-regulating drugs. Pharm. Biol. 2025, 63, 428–446. [Google Scholar] [CrossRef] [PubMed]
  138. Mbikay, M.; Mayne, J.; Sirois, F.; Fedoryak, O.; Raymond, A.; Noad, J.; Chrétien, M. Mice Fed a High-Cholesterol Diet Supplemented with Quercetin-3-Glucoside Show Attenuated Hyperlipidemia and Hyperinsulinemia Associated with Differential Regulation of PCSK9 and LDLR in their Liver and Pancreas. Mol. Nutr. Food Res. 2018, 62, e1700729. [Google Scholar] [CrossRef] [PubMed]
  139. Hui, C.K.; Majid, N.I.; Yusof, H.M.; Zainol, K.M.; Mohamad, H.; Mohd Zin, Z. Catechin profile and hypolipidemic activity of Morinda citrifolia leaf water extract. Heliyon 2020, 6, e04337. [Google Scholar] [CrossRef] [PubMed]
  140. Franceschini, G.; Maderna, P.; Sirtori, C.R. Reverse cholesterol transport: Physiology and pharmacology. Atherosclerosis 1991, 88, 99–107. [Google Scholar] [CrossRef] [PubMed]
  141. Ren, K.; Jiang, T.; Zhou, H.F.; Liang, Y.; Zhao, G.J. Apigenin Retards Atherogenesis by Promoting ABCA1-Mediated Cholesterol Efflux and Suppressing Inflammation. Cell Physiol. Biochem. 2018, 47, 2170–2184. [Google Scholar] [CrossRef] [PubMed]
  142. Dergunov, A.D.; Savushkin, E.V.; Dergunova, L.V.; Litvinov, D.Y. Significance of Cholesterol-Binding Motifs in ABCA1, ABCG1, and SR-B1 Structure. J. Membr. Biol. 2019, 252, 41–60. [Google Scholar] [CrossRef] [PubMed]
  143. Zhao, C.; Dahlman-Wright, K. Liver X receptor in cholesterol metabolism. J. Endocrinol. 2010, 204, 233. [Google Scholar] [CrossRef] [PubMed]
  144. Park, H.S.; Lee, K.; Kim, S.H.; Hong, M.J.; Jeong, N.J.; Kim, M.S. Luteolin improves hypercholesterolemia and glucose intolerance through LXRα-dependent pathway in diet-induced obese mice. J. Food Biochem. 2020, 44, e13358. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, S.; Sheng, F.; Zou, L.; Xiao, J.; Li, P. Hyperoside attenuates non-alcoholic fatty liver disease in rats via cholesterol metabolism and bile acid metabolism. J. Adv. Res. 2021, 34, 109–122. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  146. Lim, M.Y.C.; Kiat Ho, H. Pharmacological modulation of cholesterol 7α-hydroxylase (CYP7A1) as a therapeutic strategy for hypercholesterolemia. Biochem. Pharmacol. 2024, 220, 115985. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, D.; Xia, M.; Gao, S.; Li, D.; Zhang, Y.; Jin, T.; Ling, W. Cyanidin-3-O-β-glucoside upregulates hepatic cholesterol 7α-hydroxylase expression and reduces hypercholesterolemia in mice. Mol. Nutr. Food Res. 2012, 56, 610. [Google Scholar] [CrossRef] [PubMed]
  148. Yue, P.P. Effects of Acupuncture Therapy on Hepatic Oxidative Stress and Nrf2/Keap1/HO-1 Signaling Pathway in Hyperlipidemic Rats. Master’s Thesis, Changchun University of Chinese Medicine, Changchun, China, 2024. [Google Scholar] [CrossRef]
  149. Yang, J.B.; Chen, X.T.; Zhou, Y.Y.; Yang, S.H.; Yang, R.H.; Wang, Q.; Xue, S.; Niu, W.Y. Research Progress on the Preventive and Therapeutic Effects of Traditional Chinese Medicine on Metabolic-Associated Fatty Liver Disease Based on Nrf2 Signaling Pathway. Drug Eval. Res. 2024, 47, 2681–2687. [Google Scholar]
  150. Yang, J.T.; Wang, J.; Zhou, X.R.; Xiao, C.; Lou, Y.Y.; Tang, L.H.; Zhang, F.J.; Qian, L.B. Luteolin alleviates cardiac ischemia/reperfusion injury in the hypercholesterolemic rat via activating Akt/Nrf2 signaling. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2018, 391, 719–728. [Google Scholar] [CrossRef] [PubMed]
  151. Kumar, R.; Akhtar, F.; Rizvi, S.I. Hesperidin attenuates altered redox homeostasis in an experimental hyperlipidaemic model of rat. Clin. Exp. Pharmacol. Physiol. 2020, 47, 571–582. [Google Scholar] [CrossRef] [PubMed]
  152. Kumar, R.; Khan, M.I.; Ashfaq, F.; Alsayegh, A.A.; Khatoon, F.; Altamimi, T.N.; Rizvi, S.I. Hesperidin Supplementation Improves Altered PON -1, LDL Oxidation, Inflammatory Response and Hepatic Function in an Experimental Rat Model of Hyperlipidemia. Indian J. Clin. Biochem. 2024, 39, 257–263. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  153. Cherkaoui-Malki, M.; Surapureddi, S.; El-Hajj, H.I.; Vamecq, J.; Andreoletti, P. Hepatic steatosis and peroxisomal fatty acid beta-oxidation. Curr. Drug. Metab. 2012, 13, 1412. [Google Scholar] [CrossRef] [PubMed]
  154. Li, P.; Li, X.B.; Fu, S.X.; Wu, C.C.; Wang, X.X.; Yu, G.J.; Long, M.; Wang, Z.; Liu, G.W. Alterations of fatty acid β-oxidation capability in the liver of ketotic cows. J. Dairy Sci. 2012, 95, 1759. [Google Scholar] [CrossRef] [PubMed]
  155. Sun, J.; Wang, Z.; Chen, L.; Sun, G. Hypolipidemic Effects and Preliminary Mechanism of Chrysanthemum Flavonoids, Its Main Components Luteolin and Luteoloside in Hyperlipidemia Rats. Antioxidants 2021, 10, 1309. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  156. Sathyanarayan, A.; Mashek, M.T.; Mashek, D.G. ATGL Promotes Autophagy/Lipophagy via SIRT1 to Control Hepatic Lipid Droplet Catabolism. Cell Rep. 2017, 19, 1–9. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  157. Pastore, N.; Vainshtein, A.; Klisch, T.J.; Armani, A.; Huynh, T.; Herz, N.J.; Polishchuk, E.V.; Sandri, M.; Ballabio, A. TFE3 regulates whole-body energy metabolism in cooperation with TFEB. EMBO Mol. Med. 2017, 9, 605–621. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  158. Liu, W.; Zhao, Y.; Wang, G.; Feng, S.; Ge, X.; Ye, W.; Wang, Z.; Zhu, Y.; Cai, W.; Bai, J.; et al. TRIM22 inhibits osteosarcoma progression through destabilizing NRF2 and thus activation of ROS/AMPK/mTOR/autophagy signaling. Redox. Biol. 2022, 53, 102344. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  159. Wang, Y.; Zhao, H.; Li, X.; Wang, Q.; Yan, M.; Zhang, H.; Zhao, T.; Zhang, N.; Zhang, P.; Peng, L.; et al. Formononetin alleviates hepatic steatosis by facilitating TFEB-mediated lysosome biogenesis and lipophagy. J. Nutr. Biochem. 2019, 73, 108214. [Google Scholar] [CrossRef] [PubMed]
  160. Hu, Z.; Li, Y.; Yao, N.; Gan, H.; Zeng, Q.; Huang, X.; Huang, D.; Cai, D.; Chen, Y. Apigenin attenuates the atherosclerotic lesions through enhancing selective autophagy/lipophagy and promoting RCT process. Pharm. Biol. 2025, 63, 387–401. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  161. Yan, M.; Mehta, J.L.; Hu, C. LOX-1 and obesity. Cardiovasc. Drugs Ther. 2011, 25, 469. [Google Scholar] [CrossRef] [PubMed]
  162. Xia, Z.H.; Chen, W.B.; Shi, L.; Jiang, X.; Li, K.; Wang, Y.X.; Liu, Y.Q. The Underlying Mechanisms of Curcumin Inhibition of Hyperglycemia and Hyperlipidemia in Rats Fed a High-Fat Diet Combined With STZ Treatment. Molecules 2020, 25, 271. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  163. Xu, Q.; Li, Y.C.; Du, C.; Wang, L.N.; Xiao, Y.H. Effects of Apigenin on the Expression of LOX-1, Bcl-2, and Bax in Hyperlipidemia Rats. Chem. Biodivers. 2021, 18, e2100049. [Google Scholar] [CrossRef] [PubMed]
  164. Ding, P.; Song, Y.; Yang, Y.; Zeng, C. NLRP3 inflammasome and pyroptosis in cardiovascular diseases and exercise intervention. Front. Pharmacol. 2024, 15, 1368835. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  165. Ramachandran, R.; Manan, A.; Kim, J.; Choi, S. NLRP3 inflammasome: A key player in the pathogenesis of life-style disorders. Exp. Mol. Med. 2024, 56, 1488–1500. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  166. Lu, Z.; Liu, L.; Zhao, S.; Zhao, J.; Li, S.; Li, M. Apigenin attenuates atherosclerosis and non-alcoholic fatty liver disease through inhibition of NLRP3 inflammasome in mice. Sci. Rep. 2023, 13, 7996. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  167. Xie, L.; Chi, X.; Wang, H.; Dai, A.; Dong, J.; Liu, S.; Zhang, D. Mechanism of action of buckwheat quercetin in regulating lipid metabolism and intestinal flora via Toll-like receptor 4 or nuclear factor κB pathway in rats on a high-fat diet. Nutrition 2023, 115, 112148. [Google Scholar] [CrossRef] [PubMed]
  168. Xiong, H.; Wang, J.; Ran, Q.; Lou, G.; Peng, C.; Gan, Q.; Hu, J.; Sun, J.; Yao, R.; Huang, Q. Hesperidin: A Therapeutic Agent For Obesity. Drug Des. Devel. Ther. 2019, 13, 3855–3866. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  169. Zhang, J.X.; Ma, J.J.; Li, Y.J.; Dong, S.Q.; Yang, B.; Fan, H.R. Research Progress on the Mechanisms of Traditional Chinese Medicine in Treating Hyperlipidemia Based on Intestinal Microbiota. Chin. Herb. Med. 2025, 56, 2171–2183. [Google Scholar]
  170. Liu, Z.X.; Kui, L.L. Exploring the Prevention and Treatment of Hyperlipidemia by Integrative Chinese and Western Medicine Based on Intestinal Microbiota. Asia-Pac. Tradit. Med. 2024, 20, 191–197. [Google Scholar]
  171. Wang, T.; Liu, L.; Deng, J.; Jiang, Y.; Yan, X.; Liu, W. Analysis of the mechanism of action of quercetin in the treatment of hyperlipidemia based on metabolomics and intestinal flora. Food Funct. 2023, 14, 2112–2127. [Google Scholar] [CrossRef] [PubMed]
  172. Wang, T.; Wu, Q.; Zhao, T. Preventive Effects of Kaempferol on High-Fat Diet-Induced Obesity Complications in C57BL/6 Mice. Biomed. Res. Int. 2020, 2020, 4532482. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  173. Zhang, J.J.; Liu, D.P.; Huang, Y.T.; Yuan, G.; Shuai, Q. Biopharmaceutics classification and intestinal absorption study of apigenin. Int. J. Pharm. 2012, 436, 311–317. [Google Scholar] [CrossRef] [PubMed]
  174. Li, B.; Robinson, D.H.; Birt, D.F. Evaluation of properties of apigenin and [G-3H]apigenin and analytic method development. J. Pharm. Sci. 1997, 86, 721–725. [Google Scholar] [CrossRef] [PubMed]
  175. Tang, D.; Chen, K.; Huang, L.; Li, J. Pharmacokinetic properties and drug interactions of apigenin, a natural flavone. Expert Opin. Drug Metab. Toxicol. 2017, 13, 323–330. [Google Scholar] [CrossRef] [PubMed]
  176. Liu, Y.; Hu, M. Absorption and Metabolism of Flavonoids in the Caco-2 Cell Culture Model and a Perused Rat Intestinal Model, Drug Metabolism and Disposition. Drug. Metab. Dispos. 2002, 30, 370. [Google Scholar] [CrossRef] [PubMed]
  177. Gradolatto, A.; Basly, J.P.; Berges, R.; Teyssier, C.; Chagnon, M.C.; Siess, M.H.; Canivenc-Lavier, M.C. Pharmacokinetics and metabolism of apigenin in female and male rats after a single oral administration. Drug Metab. Dispos. 2005, 33, 49–54. [Google Scholar] [CrossRef] [PubMed]
  178. Ge, X.; He, X.; Lin, Z.; Zhu, Y.; Jiang, X.; Zhao, L.; Zeng, F.; Chen, L.; Xu, W.; Liu, T.; et al. 6,8-(1,3-Diaminoguanidine) luteolin and its Cr complex show hypoglycemic activities and alter intestinal microbiota composition in type 2 diabetes mice. Food Funct. 2022, 13, 3572–3589. [Google Scholar] [CrossRef] [PubMed]
  179. Zhu, M.; Sun, Y.; Su, Y.; Guan, W.; Wang, Y.; Han, J.; Wang, S.; Yang, B.; Wang, Q.; Kuang, H. Luteolin: A promising multifunctional natural flavonoid for human diseases. Phytother. Res. 2024, 38, 3417–3443. [Google Scholar] [CrossRef] [PubMed]
  180. Shi, F.; Pan, H.; Lu, Y.; Ding, L. An HPLC-MS/MS method for the simultaneous determination of luteolin and its major metabolites in rat plasma and its application to a pharmacokinetic study. J. Sep. Sci. 2018, 41, 3830–3839. [Google Scholar] [CrossRef] [PubMed]
  181. Lin, L.C.; Pai, Y.F.; Tsai, T.H. Isolation of Luteolin and Luteolin-7-O-glucoside from Dendranthema morifolium Ramat Tzvel and Their Pharmacokinetics in Rats. J. Agric. Food Chem. 2015, 63, 7700. [Google Scholar] [CrossRef] [PubMed]
  182. Miyashita, A.; Ito, J.; Parida, I.S.; Syoji, N.; Fujii, T.; Takahashi, H.; Nakagawa, K. Improving water dispersibility and bioavailability of luteolin using microemulsion system. Sci. Rep. 2022, 12, 11949. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  183. Kaci, H.; Bodnárová, S.; Fliszár-Nyúl, E.; Lemli, B.; Pelantová, H.; Valentová, K.; Bakos, É.; Özvegy-Laczka, C.; Poór, M. Interaction of luteolin, naringenin, and their sulfate and glucuronide conjugates with human serum albumin, cytochrome P450 (CYP2C9, CYP2C19, and CYP3A4) enzymes and organic anion transporting polypeptide (OATP1B1 and OATP2B1) transporters. Biomed. Pharmacother. 2023, 157, 114078. [Google Scholar] [CrossRef] [PubMed]
  184. Chen, X.; Liu, L.; Sun, Z.; Liu, Y.; Xu, J.; Liu, S.; Huang, B.; Ma, L.; Yu, Z.; Bi, K. Pharmacokinetics of luteolin and tetra-acetyl-luteolin assayed by HPLC in rats after oral administration. Biomed. Chromatogr. 2010, 24, 826. [Google Scholar] [CrossRef] [PubMed]
  185. Zhou, P.; Li, L.P.; Luo, S.Q.; Jiang, H.D.; Zeng, S. Intestinal absorption of luteolin from peanut hull extract is more efficient than that from individual pure luteolin. J. Agric. Food Chem. 2008, 56, 296–300. [Google Scholar] [CrossRef] [PubMed]
  186. Yao, C.; Dai, S.; Wang, C.; Fu, K.; Wu, R.; Zhao, X.; Yao, Y.; Li, Y. Luteolin as a potential hepatoprotective drug: Molecular mechanisms and treatment strategies. Biomed. Pharmacother. 2023, 167, 115464. [Google Scholar] [CrossRef] [PubMed]
  187. Rodrigues, D.B.; Marques, M.C.; Hacke, A.; Loubet Filho, P.S.; Cazarin, C.B.B.; Mariutti, L.R.B. Trust your gut: Bioavailability and bioaccessibility of dietary compounds. Curr. Res. Food Sci. 2022, 5, 228–233. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  188. Liu, L.; Barber, E.; Kellow, N.J.; Williamson, G. Improving quercetin bioavailability: A systematic review and meta-analysis of human intervention studies. Food Chem. 2025, 477, 143630. [Google Scholar] [CrossRef] [PubMed]
  189. Che, Z.H. Preparation and in Vitro and in Vivo Evaluation of Quercetin Nanoemulsion. Master’s Thesis, Anhui Medical University, Hefei, China, 2022. [Google Scholar] [CrossRef]
  190. Arts, I.C.; Sesink, A.L.; Faassen-Peters, M.; Hollman, P.C. The type of sugar moiety is a major determinant of the small intestinal uptake and subsequent biliary excretion of dietary quercetin glycosides. Br. J. Nutr. 2004, 91, 841. [Google Scholar] [CrossRef] [PubMed]
  191. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  192. Omiecinski, C.J.; Vanden Heuvel, J.P.; Perdew, G.H.; Peters, J.M. Xenobiotic metabolism, disposition, and regulation by receptors: From biochemical phenomenon to predictors of major toxicities. Toxicol. Sci. 2011, 120 (Suppl. S1), S49–S75. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  193. de Boer, V.C.; Dihal, A.A.; van der Woude, H.; Arts, I.C.; Wolffram, S.; Alink, G.M.; Rietjens, I.M.; Keijer, J.; Hollman, P.C. Tissue distribution of quercetin in rats and pigs. J. Nutr. 2005, 135, 1718. [Google Scholar] [CrossRef] [PubMed]
  194. Mullen, W.; Rouanet, J.M.; Auger, C.; Teissèdre, P.L.; Caldwell, S.T.; Hartley, R.C.; Lean, M.E.; Edwards, C.A.; Crozier, A. Bioavailability of [2-(14)C]quercetin-4’-glucoside in rats. J. Agric. Food Chem. 2008, 56, 12127. [Google Scholar] [CrossRef] [PubMed]
  195. 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 Safety 2018, 17, 1054–1112. [Google Scholar] [CrossRef] [PubMed]
  196. Alkandahri, M.Y.; Pamungkas, B.T.; Oktoba, Z.; Shafirany, M.Z.; Sulastri, L.; Arfania, M.; Anggraeny, E.N.; Pratiwi, A.; Astuti, F.D.; Indriyani; et al. Hepatoprotective Effect of Kaempferol: A Review of the Dietary Sources, Bioavailability, Mechanisms of Action, and Safety. Adv. Pharmacol. Pharm. Sci. 2023, 2023, 1387665. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  197. DuPont, M.S.; Day, A.J.; Bennett, R.N.; Mellon, F.A.; Kroon, P.A. Absorption of kaempferol from endive, a source of kaempferol-3-glucuronide, in humans. Eur. J. Clin. Nutr. 2004, 58, 947–954. [Google Scholar] [CrossRef] [PubMed]
  198. Wdowiak, K.; Walkowiak, J.; Pietrzak, R.; Bazan-Woźniak, A.; Cielecka-Piontek, J. Bioavailability of Hesperidin and Its Aglycone Hesperetin-Compounds Found in Citrus Fruits as a Parameter Conditioning the Pro-Health Potential (Neuroprotective and Antidiabetic Activity)-Mini-Review. Nutrients 2022, 14, 2647. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  199. Cao, R.; Zhao, Y.; Zhou, Z.; Zhao, X. Enhancement of the water solubility and antioxidant activity of hesperidin by chitooligosaccharide. J. Sci. Food Agric. 2018, 98, 2422–2427. [Google Scholar] [CrossRef] [PubMed]
  200. Serra, H.; Mendes, T.; Bronze, M.R.; Simplício, A.L. Prediction of intestinal absorption and metabolism of pharmacologically active flavones and flavanones. Bioorg. Med. Chem. 2008, 16, 4009–4018. [Google Scholar] [CrossRef] [PubMed]
  201. 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, S230–S242. [Google Scholar] [CrossRef] [PubMed]
  202. Makarova, N.M. Bioavailability and metabolism of flavonoids. Eksp. Klin. Farmakol. 2011, 74, 33–40. [Google Scholar] [PubMed]
  203. Sharma, N.; Kabra, A. Formononetin: Pharmacological properties and therapeutic potential. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2025, 1–21. [Google Scholar] [CrossRef] [PubMed]
  204. Luo, L.Y.; Fan, M.X.; Zhao, H.Y.; Li, M.X.; Wu, X.; Gao, W.Y. Pharmacokinetics and Bioavailability of the Isoflavones Formononetin and Ononin and Their in vitro Absorption in Using Chamber and Caco-2 Cell Models. J. Agric. Food Chem. 2018, 66, 2917–2924. [Google Scholar] [CrossRef] [PubMed]
  205. Singh, S.P.; Wahajuddin Tewari, D.; Pradhan, T.; Jain, G.K. PAMPA permeability, plasma protein binding, blood partition, pharmacokinetics and metabolism of formononetin, a methoxylated isoflavone. Food Chem. Toxicol. 2011, 49, 1056–1062. [Google Scholar] [CrossRef] [PubMed]
  206. Almatroodi, S.A.; Almatroudi, A.; Khan, A.A.; Rahmani, A.H. Potential Therapeutic Targets of Formononetin, a Type of Methoxylated Isoflavone, and Its Role in Cancer Therapy through the Modulation of Signal Transduction Pathways. Int. J. Mol. Sci. 2023, 24, 9719. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  207. Dong, Z.; Wang, Y.; Guo, J.; Tian, C.; Pan, W.; Wang, H.; Yan, J. Prostate Cancer Therapy Using Docetaxel and Formononetin Combination: Hyaluronic Acid and Epidermal Growth Factor Receptor Targeted Peptide Dual Ligands Modified Binary Nanoparticles to Facilitate the in vivo Anti-Tumor Activity. Drug. Des. Dev. Ther. 2022, 16, 2683–2693. [Google Scholar] [CrossRef] [PubMed]
  208. Xiong, W.; Lan, Q.; Liang, X.; Zhao, J.; Huang, H.; Zhan, Y.; Qin, Z.; Jiang, X.; Zheng, L. Cartilage-targeting poly(ethylene glycol) (PEG)-formononetin (FMN) nanodrug for the treatment of osteoarthritis. J. Nanobiotechnol. 2021, 19, 197. [Google Scholar] [CrossRef] [PubMed]
  209. Tang, G.Y.; Meng, X.; Gan, R.Y.; Zhao, C.N.; Liu, Q.; Feng, Y.B.; Li, S.; Wei, X.L.; Atanasov, A.G.; Corke, H.; et al. Health Functions and Related Molecular Mechanisms of Tea Components: An Update Review. Int. J. Mol. Sci. 2019, 20, 6196. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  210. Manach, C.; Texier, O.; Morand, C.; Crespy, V.; Régérat, F.; Demigné, C.; Rémésy, C. Comparison of the bioavailability of quercetin and catechin in rats. Free Radic. Biol. Med. 1999, 27, 1259. [Google Scholar] [CrossRef] [PubMed]
  211. Choi, J.S.; Choi, J.S.; Choi, D.H. Effects of licochalcon A on the pharmacokinetics of losartan and its active metabolite, EXP-3174, in rats. Pharmazie 2013, 68, 882. [Google Scholar] [PubMed]
  212. Li, M.; Yang, S.; Yi, H.; Wang, Z.; Xu, B.; Li, G.; Ma, C.; Yuan, C.; Wang, Z. Preparation and evaluation of licochalcone A-integrated casein-pectin nanodelivery system: Insights into its gastrointestinal digestibility and bioavailability. Food Chem. 2025, 486, 144633. [Google Scholar] [CrossRef] [PubMed]
  213. Zhu, Z.; Liu, J.; Yang, Y.; Adu-Frimpong, M.; Ji, H.; Toreniyazov, E.; Wang, Q.; Yu, J.; Xu, X. SMEDDS for improved oral bioavailability and anti-hyperuricemic activity of licochalcone A. J. Microencapsul. 2021, 38, 459–471. [Google Scholar] [CrossRef] [PubMed]
  214. Cai, H.; Thomasset, S.C.; P-Berry, D.; Garcea, G.; Brown, K.; Steward, W.P.; Gescher, A.J. Determination of anthocyanins in the urine of patients with colorectal liver metastases after administration of bilberry extract. Biomed. Chromatogr. 2011, 25, 660. [Google Scholar] [CrossRef] [PubMed]
  215. Kalt, W.; Blumberg, J.B.; McDonald, J.E.; Vinqvist-Tymchuk, M.R.; Fillmore, S.A.; Graf, B.A.; O’Leary, J.M.; Milbury, P.E. Identification of anthocyanins in the liver, eye, and brain of blueberry-fed pigs. J. Agric. Food Chem. 2008, 56, 705. [Google Scholar] [CrossRef] [PubMed]
  216. Yang, M.; Abdullah, A.N.; Hussain, M.; Lu, X.; Xu, J.; Zhong, H.; Guan, R. A review of recent advances on cyanidin-3-glucoside: The biotransformation, absorption, bioactivity and applications of nano-encapsulation. Food Funct. 2023, 14, 6320–6345. [Google Scholar] [CrossRef] [PubMed]
  217. Shoubaky, G.A.E.; Abdel-Daim, M.M.; Mansour, M.H.; Salem, E.A. Isolation and identification of a flavone apigenin from marine red alga Acanthophora spicifera with antinociceptive and anti-Inflammatory activities. J. Exp. Neurosci. 2016, 10, 21–29. [Google Scholar] [CrossRef] [PubMed]
  218. Allemailem, K.S.; Almatroudi, A.; Alharbi, H.O.A.; AlSuhaymi, N.; Alsugoor, M.H.; Aldakheel, F.M.; Khan, A.A.; Rahmani, A.H. Apigenin: A Bioflavonoid with a Promising Role in Disease Prevention and Treatment. Biomedicines 2024, 12, 1353. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  219. Wang, C.; Li, Q.; Xiao, B.; Fang, H.; Huang, B.; Huang, F.; Wang, Y. Luteolin enhances the antitumor efficacy of oncolytic vaccinia virus that harbors IL-24 gene in liver cancer cells. J. Clin. Lab. Anal. 2021, 35, e23677. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  220. Im, E.; Yeo, C.; Lee, E.O. Luteolin induces caspase-dependent apoptosis via inhibiting the AKT/osteopontin pathway in human hepatocellular carcinoma SK-Hep-1 cells. Life Sci. 2018, 209, 259–266. [Google Scholar] [CrossRef] [PubMed]
  221. Ding, S.; Hu, A.; Hu, Y.; Ma, J.; Weng, P.; Dai, J. Anti-hepatoma cells function of luteolin through inducing apoptosis and cell cycle arrest. Tumour. Biol. 2014, 35, 3053. [Google Scholar] [CrossRef] [PubMed]
  222. Shi, F.; Zhao, P.; Li, X.; Pan, H.; Ma, S.; Ding, L. Cytotoxicity of luteolin in primary rat hepatocytes: The role of CYP3A-mediated ortho-benzoquinone metabolite formation and glutathione depletion. J. Appl. Toxicol. 2015, 35, 1372. [Google Scholar] [CrossRef] [PubMed]
  223. Javadi, F.; Ahmadzadeh, A.; Eghtesadi, S.; Aryaeian, N.; Zabihiyeganeh, M.; Rahimi Foroushani, A.; Jazayeri, S. The effect of quercetin on inflammatory factors and clinical symptoms in women with rheumatoid arthritis: A double-blind, randomized controlled trial. J. Am. Coll. Nutr. 2016, 36, 9–15. [Google Scholar] [CrossRef] [PubMed]
  224. Shi, Y.; Williamson, G. Quercetin lowers plasma uric acid in pre-hyperuricaemic males: A randomised, double-blinded, placebo-controlled, cross-over trial. Br. J. Nutr. 2016, 115, 800–806. [Google Scholar] [CrossRef] [PubMed]
  225. Andres, S.; Pevny, S.; Ziegenhagen, R.; Bakhiya, N.; Schäfer, B.; Hirsch-Ernst, K.I.; Lampen, A. Safety Aspects of the Use of Quercetin as a Dietary Supplement. Mol. Nutr. Food Res. 2018, 62, 1700447. [Google Scholar] [CrossRef] [PubMed]
  226. Ruscinc, N.; Massarico Serafim, R.A.; Almeida, C.; Rosado, C.; Baby, A.R. Challenging the safety and efficacy of topically applied chlorogenic acid, apigenin, kaempferol, and naringenin by HET-CAM, HPLC-TBARS-EVSC, and laser Doppler flowmetry. Front. Chem. 2024, 12, 1400881. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  227. Garg, A.; Garg, S.; Zaneveld, L.J.D.; Singla, A.K. Chemistry and pharmacology of the citrus bioflavonoid hesperidin. Phyther. Res. 2001, 15, 655–669. [Google Scholar] [CrossRef] [PubMed]
  228. Pingale, T.D.; Gupta, G.L. Acute and sub-acute toxicity study reveals no dentrimental effect of formononetin in mice upon repeated i.p. dosing. Toxicol. Mech. Methods 2023, 33, 688–697. [Google Scholar] [CrossRef] [PubMed]
  229. Matsuyama, T.; Tanaka, Y.; Kamimaki, I.; Nagao, T.; Tokimitsu, I. Catechin safely improved higher levels of fatness, blood pressure, and cholesterol in children. Obesity 2008, 16, 1338. [Google Scholar] [CrossRef] [PubMed]
  230. de Freitas, K.S.; Squarisi, I.S.; Acésio, N.O.; Nicolella, H.D.; Ozelin, S.D.; Reis Santos de Melo, M.; Guissone, A.P.P.; Fernandes, G.; Silva, L.M.; da Silva Filho, A.A.; et al. Licochalcone A a licorice flavonoid: Antioxidant, cytotoxic, genotoxic, and chemopreventive potential. J. Toxicol. Environ. Health A 2020, 83, 673–686. [Google Scholar] [CrossRef] [PubMed]
  231. Jeon, S.; Han, S.; Lee, J.; Hong, T.; Yim, D.S. The safety and pharmacokinetics of cyanidin-3-glucoside after 2-week administration of black bean seed coat extract in healthy subjects. Korean J. Physiol. Pharmacol. 2012, 16, 249. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  232. Li, Q.S.; Li, X.C.; Wang, X.M.; Guo, J.F.; Wu, L.L.; Qin, L.L.; Liu, T.H. Research Progress on Traditional Chinese Medicine Intervening in PPARγ Signaling Pathway for Improving Hyperlipidemia. China J. Tradit. Chin. Med. Pharm. 2025, 40, 1851–1855. [Google Scholar]
Figure 1. Mechanisms of flavonoids in treating hyperlipidemia.
Figure 1. Mechanisms of flavonoids in treating hyperlipidemia.
Molecules 30 03103 g001
Table 1. Biological activities of representative flavonoids with lipid-lowering effects.
Table 1. Biological activities of representative flavonoids with lipid-lowering effects.
CategoryHCAsStructureSourceBiological Activity/ApplicationExperimental ModelDose RangesRef.
FlavonesApigenin
(C15H10O6)
Molecules 30 03103 i001Leonurus japonicus, olive leaves, chamomile, parsley, celery, Basella alba L., artichoke, etc.anti-inflammatoryin vitro10, 20, 30 μM[17]
antioxidationin vivo20 mg/kg[90]
anticancerin vivo20–200 mg/kg[19]
anti-apoptosisin vivo10 mg/kg[91]
improve lipid metabolismin vitro40 μmol/L[21]
insulin resistancein vivo10 mg/kg[92]
Luteolin
(C15H10O7)
Molecules 30 03103 i002Taraxacum mongolicum, Chrysanthemum morifolium, lotus stems, Lonicera japonica, cabbage, celery, apples, oranges, pomegranates, lemons, etc.including anticancerin vivo30 mg/kg[93]
lipid-loweringin vitro10–20 mm[94]
anti-inflammatoryin vitro10 μm, 100 μm[29]
modulation of intestinal microbiotain vivo17.3 mg/mL[30]
immunoregulatory functionsin vivo100 mg/kg[95]
FlavonolsQuercetin
(C15H10O7)
Molecules 30 03103 i003Astragalus membranaceus, Trichosanthes kirilowii, Albizia julibrissin flowers, and Morus alba leaves, as well as in fruits and vegetables including grapes, onions, carrots, and potatoesameliorating metabolic syndromein vivo5, 10 mg/kg[41]
antibacterial effectsin vitro1–8 mg/mL[42]
neuroprotectionin vivo50, 100 mg/kg[43]
anti-ferroptotic activityin vivo25 mg/kg[44]
hypoglycemicclinical400 mg/kg[41]
anti-obesity propertiesin vivo240 mg/kg[41]
antirheumatic effectsin vitro20, 40, 80 μmol/L[46]
Kaempferol
(C15H10O6)
Molecules 30 03103 i004Ginkgo biloba leaves, Carthamus tinctorius, Cnidium monnieri, blueberry leaves, figs, legumes, cauliflower, cabbage, strawberries, tea, and tomatoesincluding anticancerin vitro0, 25, 50, 75, 100 μM[96]
anti-inflammatoryin vivo3, 5, 9 mg/kg[97]
antioxidantin vivo10 mg/kg[98]
Alzheimer’s diseasein vivo10 mg/kg[60]
DihydroflavonesHesperidin
(C28H34O15)
Molecules 30 03103 i005citrus fruits such as Citrus sinensis and Citrus reticulatalipid-loweringin vivo150, 300 mg/kg[99]
antioxidantin vivo50 mg/kg[64]
anti-inflammatoryin vivo200 mg/kg[100]
neuroprotective effectsin vivo200 mg/kg[101]
IsoflavonesFormononetin
(C16H12O4)
Molecules 30 03103 i006leguminous plants (such as Astragalus membranaceus, Pueraria lobata, Glycyrrhiza uralensis, etc.), coffee beans, strawberries, and grapesincluding anticancerin vivo10 mg[71]
anti-obesityclinical30[71]
lipid-loweringin vivo10 mg/kg[102]
anti-osteoarthritisin vitro200 μM[72]
neuroprotective effectsin vitro10 µM[71]
FlavanolsCatechin
(C15H14O6)
Molecules 30 03103 i007Ampelopsis japonica, Lycium barbarum, green tea, kiwifruit, etc.antioxidationin vitro50 μM[103]
including anticancerin vitro25 μMol/L[79]
lipid-loweringclinical400 mg/d[80]
ChalconesLicochalcone A(C21H22O4)Molecules 30 03103 i008Glycyrrhiza uralensislower blood sugarin vivo100, 200, 300 mg/kg[104]
lipid-loweringin vivo5, 10 mg/kg[105]
anti-allergic propertiesin vivo20, 40, 80 mg/kg[83]
AnthocyanidinsCyanidin-3-glucoside
(C21H21ClO11)
Molecules 30 03103 i009Artemisia argyi, mulberries, black wolfberries, purple cabbage, purple sweet potatoes, grapes, etc.including anticancerin vivo5 mg/kg[106]
lipid-loweringin vitro100 μM[88]
modulation of intestinal microbiotain vivo250 mg/kg[89]
Table 2. Molecular-level regulation of lipid metabolism by various flavonoids.
Table 2. Molecular-level regulation of lipid metabolism by various flavonoids.
CategoryHCAsMechanisms of ActionClassification of MechanismsExperimental ModelDose RangesRef.
FlavonesApigenin
(C15H10O6)
miR-363-3p↓, INSIG1↑, SREBP1, ACC1, FASN, SCD↓Inhibition of Triglyceride SynthesisHepG2 cells
C57BL/6J mice
20, 40 μmol/L
150, 250 mg/kg
[109]
PI3K, p-Akt, PPARγ↓3T3-L1 preadipocytes20, 40 μM[114]
SREBP2, HMGCR↓Inhibition of Cholesterol Synthesismouse embryonic fibroblasts50 μM[127]
7-Dehydrocholesterol, Xanthine↓mouse embryonic fibroblasts25 μM[130]
miR-33↓, ABCA1↑Promotion of Cholesterol UptakeRAW264.7 cells
ApoE–/–mice
10, 20, 40 μM
10 mg/kg
[141]
ULK1, UVRAG, beclin-1↑Promote AutophagyC57BL/6J mice6.25, 12.5, 25 mg/kg[160]
LOX-1↑, Bcl-2, Bax↓Inhibition of ApoptosisSprague Dawley rats20, 40, 80 mg/kg[163]
NLRP3, NF-κB↓Inhibition of InflammationHepG2 cells
Ldlr−/−mice
25, 50 μM
5 mg/kg
[166]
Luteolin
(C15H10O7)
PI3K, p-Akt, PPARγ↓Inhibition of Triglyceride Synthesis3T3-L1 preadipocytes10 μM[114]
NPC1L1↓Inhibition of Lipid AbsorptionCaco-2 cells25–100 μM[133,134]
LXRα, ABCG1, SRB1↑Promotion of Cholesterol UptakeHepG2 cells
C57BL/6J mice
10–50 μM[144]
Akt, Nrf2, NQO1, HO-1↑, mPTP↓Inhibition of Oxidative StressSprague Dawley rats100 mg/kg[150]
FAβO↑, FAS↓Promotion of Fatty Acid β-OxidationSprague Dawley rats50 mg/kg[155]
FlavonolsQuercetin
(C15H10O7)
PI3K, p-Akt, PPARγ↓Inhibition of Triglyceride Synthesis3T3-L1 preadipocytes10 μM[114]
AKT, mTOR, SREBP1↓Sprague Dawley rats150 mg/kg[116]
Adipo, AdipoR2↑3T3-L1 preadipocytes
Swiss albino mice
5, 10, 20 μM
100 mg/kg
[122]
HMGCR↓Inhibition of Cholesterol SynthesisC57BL/6J mice5 mg/kg[128]
TLR4, NF-κB↓Inhibition of InflammationSprague Dawley rats200 mg/kg[138]
beneficial bacteria↑, pathogenic bacteria↓, Firmicutes/Bacteroidetes↓Regulation of Gut MicrobiotaSprague Dawley rats10, 100, 200 mg/kg[171]
Kaempferol
(C15H10O6)
AKT, SREBP1↓Inhibition of Triglyceride SynthesisHepG2, THP-1, cco2 cells10, 20 μM[118]
relative abundance of Firmicutes↓, relative abundance of Bacteroidetes↑Regulation of Gut MicrobiotaC57BL/6J mice200 mg/kg[172]
DihydroflavonesHesperidin
(C28H34O15)
p-Akt, NF-κB, Bcl-2↓Inhibition of Triglyceride SynthesisAML-1 human preadipocyte cell line100, 500 µM[119]
STC-1, CCK↑Inhibition of Lipid Absorption3T3-L1 preadipocytes0–1600 μg/mL[135]
LDL oxidation, PON-1↓Inhibition of Oxidative StressWistar rat100 mg/kg[151]
IL-6↓; TNF-α↓Inhibition of InflammationSprague Dawley rats50 mg/kg[168]
IsoflavonesFormononetin
(C16H12O4)
AMPK↑, TFEB↓Promote AutophagyHepG2 cells
C57BL/6J mice
40 μM
100 mg/kg
[159]
FlavanolsCatechin
(C15H14O6)
LDL-c uptake↑Promotion of Cholesterol UptakeHepG2 cells4 μg/mL[139]
ChalconesLicochalcone A(C21H22O4)LXRα, SREBP1↓Inhibition of Triglyceride SynthesisHepG2 cells10 μg/mL[110]
PPARγ, SREBP1↓3T3-L1 preadipocytes
ICR mouse
5, 10 μM
5, 10 mg/kg
[120]
AnthocyanidinsCyanidin-3-glucoside
(C21H21ClO11)
O-glycosylation of FoxO1↑, FFAs, glycerol, ATGL↓Inhibition of Triglyceride Synthesis3T3-L1 preadipocytes50 μM[121]
pAMPK↑, LPL↓skeletal muscle cell, adipocyte
KK-Ay mouse
10, 50, 100 µmol/L[123]
LXRα, CYP7A1↑Promotion of Cholesterol Uptakehuman aortic endothelial cell
ApoE–/–mice
0.5, 5, 50 μM
food (0.06% w/w)
[147]
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

Chen, X.; Yang, J.; Zhou, Y.; Wang, Q.; Xue, S.; Zhang, Y.; Niu, W. Research Progress and Prospects of Flavonoids in the Treatment of Hyperlipidemia: A Narrative Review. Molecules 2025, 30, 3103. https://doi.org/10.3390/molecules30153103

AMA Style

Chen X, Yang J, Zhou Y, Wang Q, Xue S, Zhang Y, Niu W. Research Progress and Prospects of Flavonoids in the Treatment of Hyperlipidemia: A Narrative Review. Molecules. 2025; 30(15):3103. https://doi.org/10.3390/molecules30153103

Chicago/Turabian Style

Chen, Xingtong, Jinbiao Yang, Yunyue Zhou, Qiao Wang, Shuang Xue, Yukun Zhang, and Wenying Niu. 2025. "Research Progress and Prospects of Flavonoids in the Treatment of Hyperlipidemia: A Narrative Review" Molecules 30, no. 15: 3103. https://doi.org/10.3390/molecules30153103

APA Style

Chen, X., Yang, J., Zhou, Y., Wang, Q., Xue, S., Zhang, Y., & Niu, W. (2025). Research Progress and Prospects of Flavonoids in the Treatment of Hyperlipidemia: A Narrative Review. Molecules, 30(15), 3103. https://doi.org/10.3390/molecules30153103

Article Metrics

Back to TopTop