Open Access

The protective effects and mechanism of myricetin in liver diseases (Review)

  • Authors:
    • Mi Chen
    • Shengnan Zhang
    • Xingqiong Huang
    • Dandan Zhang
    • Dan Zhu
    • Changhan Ouyang
    • Yankun Li
  • View Affiliations

  • Published online on: February 3, 2025     https://doi.org/10.3892/mmr.2025.13452
  • Article Number: 87
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liver diseases have become one of the significant threats to global health. However, there is a lack of effective targeted therapeutic drugs in this field and the existing drugs used for liver disease treatment usually have side‑effects. Traditional Chinese medicine (TCM) has the distinctive advantages of multi‑target and low side‑effects. As a flavonoid with various pharmacological activities such as anti‑tumour, anti‑oxidant, anti‑inflammatory and anti‑bacterial, the TCM myricetin has been widely used in liver disease research. The present work focuses on the role and molecular mechanism of myricetin in liver diseases such as acute liver injury, fatty liver, liver fibrosis and hepatocellular carcinoma. It is a promising reference for further research and application of myricetin in the treatment of liver diseases.

Introduction

As the metabolic center of the body, the liver plays a vital role in glucose metabolism, lipid metabolism, bile secretion and vitamin storage hormone synthesis (1,2). Consequently, this leads to its constant exposure to potentially pathogenic molecules or endotoxins produced by digestive products and gut microbiota. Moreover, the liver is the target of most hepatotoxic drugs and hepatitis viruses (3), which makes it highly susceptible to damage that can induce different types of liver diseases. Epidemiological reports indicate that >2 million individuals die annually worldwide from liver diseases (4). Although medicines are already available for the treatment of various common liver diseases such as acute liver injury, fatty liver, liver fibrosis and hepatocellular carcinoma (HCC), they usually induce serious adverse effects during treatment. For instance, obeticholic acid, a drug for treating liver fibrosis, can cause itching, while sorafenib, a drug for treating liver cancer, can cause diarrhea (5,6). Therefore, it is urgent to explore safer medicines with fewer side-effects for the treatment of liver diseases.

Traditional Chinese medicine (TCM) has received increasing attention from researchers in the treatment of liver diseases due to its high efficiency, multi-targeting and low side effects. As a monomer of TCM, myricetin (3,3′,4′,5,5′,7-hexahydroxyflavone) is a natural polyhydroxy flavonoid extracted from various plants (7). It has a good safety profile for consumption, as well as being an important ingredient in food and juice drinks (8,9). The US Food and Drug Administration has approved myricetin as being a healthy product (10). Recent studies have revealed that myricetin plays a vital role in the treatment of liver diseases (11,12). Given its edible safety and low side-effects, myricetin is probably a promising candidate for the treatment of liver diseases. However, a review of the efficacy and mechanism of myricetin in the treatment of various liver diseases has not been made, to the best of the authors' knowledge. In view of this, the present study summarized the progress of research on myricetin in the treatment of liver diseases to provide a direction and basis for further exploration of the potential of myricetin in the clinical treatment of liver diseases.

Sources and characteristics of myricetin

Myricetin, initially extracted from the bark of Myrica nagi by Perkin and Hummel, is widely present in a variety of plants, such as Rosa canina L. (Rose Hip), Urtica dioica L. (Nettle) and Portulaca oleracea L. (Purslane) (13,14). The structural formula of myricetin is shown in Fig. 1. Its relative molecular mass is 318.24 and its melting point is within the range of 324.0–325.5°C. It is soluble in methanol, ethanol and ethyl acetate, slightly soluble in water and insoluble in chloroform and petroleum ether (15). It has been found that berries, vegetables and tea also contain myricetin and there are two main methods to obtain myricetin, namely plant extraction and chemical synthesis (16,17). Moreover, myricetin possesses multiple biological characteristics such as anti-bacterial, anti-inflammatory, anti-oxidant, anti-tumour and immunomodulatory effects (1820). In view of this, the present study focused on the efficacy and mechanism of myricetin in liver diseases.

Role of myricetin in acute liver injury

Acute liver injury is a liver dysfunction disease with rapid onset, complex causative factors and significant clinical symptoms. As the major component of the outer membrane of Gram-negative bacteria, lipopolysaccharide (LPS) is an important virulence factor inducing acute liver injury (21). It can be recognized and bound by the Toll-like receptor (TLR) 4, which activates the NF-κB signalling pathway to induce an inflammatory response (22). Inactive NF-κB will bind to IκB and remain in the cytoplasm, but when NF-κB in the conjugate is activated, the two become phosphorylated and separate (23). Subsequently, phosphorylated (p-)NF-κB is translocated to the nucleus, thereby inducing the expression of immune-associated factors such as cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), IL-6, TNF-α and IL-1β (24). Notably, Berköz et al (25) found that myricetin inhibits the expression of p-NF-κB and p-IκB, as well as the inflammation-associated factors COX-2, iNOS, IL-6, TNF-α and IL-1β in a rat model of LPS-induced acute liver injury (Fig. 2). As members of the MAPK cascade system family, ERK, JNK and P38 play essential roles in acute liver injury (26). D-galactosamine (D-GalN), a chemical hepatotoxic agent, is often combined with LPS for the study of acute liver injury. Unexpectedly, Lv et al (27) found that myricetin inhibits the expression of TLR4, p-IκB and MAPK members (p-ERK, p-JNK and p-P38) in LPS/D-GalN-induced acute liver injury, thereby attenuating inflammation-induced liver injury (Fig. 2).

In addition, LPS/D-GalN disrupts the mitochondrial structure of hepatocytes and generates large amounts of reactive oxygen species (ROS), thereby inducing oxidative stress (28). Nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) is an important regulatory pathway for liver oxidative stress, in which HO-1 degrades heme and releases biliverdin, CO and ferrous ions to alleviate liver oxidative stress (29). Excessive ROS in hepatocytes induces Nrf2 into the nucleus, promoting the expression of the downstream anti-oxidant gene HO-1 (30). In acute liver injury, myricetin with strong anti-oxidant and free radical scavenging ability promotes the expression of Nrf2 and Ho-1 to alleviate liver oxidative stress injury (Fig. 2) (20,27). Excessive ROS induces the expression and phosphorylation of P53 in hepatocytes (31). P53 promotes the expression of Bax (a pro-apoptotic gene) and inhibits the expression of Bcl-2 (an anti-apoptotic gene), which induces an increase in mitochondrial membrane permeability, resulting in the release of pro-apoptotic factors, such as cytochrome c, from the mitochondria into the cytoplasm (32). In the cytoplasm, cytochrome c forms an apoptotic complex with apoptotic protease-activating factor-1, which activates caspase-9 and caspase-3, thereby triggering the mitochondrial apoptotic pathway and leading to aberrant apoptosis in hepatocytes (33). Lv et al (27) found that myricetin reduces the protein expression of activated caspase-3 (cleaved caspase-3) and caspase-9 (cleaved caspase-9) to inhibit aberrant apoptosis in acute liver injury (Fig. 2). In addition, caspase-3 activates endonucleases in apoptosis, cleaving nuclear DNA and inducing DNA damage (34). STAT3 is involved in DNA damage repair and its phosphorylation (p-STAT3) promotes the expression of FOXM1, which initiates the expression of proteins involved in the regulation of the DNA damage repair system (35). Significantly, Matić et al (36) found that myricetin promotes the expression of p-STAT3 and p-Akt to inhibit DNA damage in pyrogallol-induced acute liver injury (Fig. 2).

Role of myricetin in fatty liver disease

Fatty liver disease is a metabolic disease characterized by hepatocellular steatosis, which mainly consists of non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD). With the change of the lifestyle and dietary structure of an individual, the incidence of fatty liver disease has shown a significant growing trend and has become a major threat to health in recent years. It has been found that myricetin not only relieves acute liver injury but also plays a vital role in treating AFLD and NAFLD (37,38).

AFLD

AFLD is a chronic liver disease caused by prolonged and heavy alcohol consumption. Studies have shown that disorders of lipid metabolism, oxidative stress and inflammatory responses are all associated with the development of AFLD (39,40).

In the liver, alcohol is first converted to acetaldehyde by alcohol dehydrogenase (ADH), then to acetic acid by acetaldehyde dehydrogenase (ALDH) and then enters the tricarboxylic acid cycle as acetyl-CoA, which is eventually converted to CO2 and H2O (41). In addition, cytochrome P450 2E1 (CYP2E1) in the liver can be activated by ethanol to oxidize ethanol into acetaldehyde and produce large amounts of ROS, which can induce mitochondrial dysfunction, inhibit fatty acid β-oxidation and exacerbate the formation of FLD (42). Myricetin attenuates ROS-induced ALFD by decreasing ADH and CYP2E1 activities and alcohol-induced oxidative damage in the liver by decreasing ROS and MDA levels (Fig. 3) (38,43).

Dysregulation of lipid metabolism due to aberrant expression of metabolic enzymes is another key factor in the pathogenesis of AFLD. Acetyl coenzyme A carboxylase, the rate-limiting enzyme for de novo synthesis of fatty acids and carnitine palmitoyltransferase 1 (CPT1), the rate-limiting enzyme for fatty acid oxidation, work together to maintain lipid homeostasis (44). AMP-activated protein kinase (AMPK) can promote lipid metabolism in the liver by inactivating acetyl coenzyme A carboxylase through phosphorylation and promoting the expression of CPT1; moreover, p-AMPK can reduce fatty acid synthesis by decreasing the expression of Srebp-1 (45). Guo et al (38) found that myricetin promotes the expression of phosphorylated AMPKa and reduces ethanol-induced lipid synthesis and accumulation in the liver (Fig. 3).

NAFLD

First proposed by Ludwig et al (46) in 1980, NAFLD is a liver disease caused by steatosis in the absence of alcohol intake. It is currently the most common liver disease worldwide and a significant risk factor for the development of HCC. Similar to AFLD, disorders of lipid metabolism, oxidative stress, inflammatory response and apoptosis are also the pathogenic mechanisms in NAFLD (47).

The accumulation of triglycerides in hepatocytes is known as steatosis, which is the initial stage in the development of NAFLD. As a member of the ligand-activated nuclear receptor superfamily, peroxisome proliferator-activated receptor gamma (PPAR-γ) plays a vital role in the development of NAFLD. It has been found that PPARγ upregulates the expression of CD36, which promotes fatty acid transmembrane transport and exacerbates liver lipid accumulation (48). It also upregulates the expression of Srebp-1, which promotes liver lipid synthesis and exacerbates the development of NAFLD (49). Myricetin was found to inhibit the expression of PPARγ and CD36 and alleviate high-fat diet-induced liver steatosis, as well as inhibit the expression of Srebp-1 and reduce liver fat content in ob/ob mice (Fig. 3) (50,51). Nrf2 protects the liver by regulating the expression of anti-oxidant enzymes nicotinamide adenine dinucleotide phosphate: quinone oxidoreductase 1 (NQO1) and HO-1 to scavenge ROS (52). In NAFLD, myricetin promotes the expression of NRF2 and its downstream genes NQO1 and HO-1, thereby alleviating liver oxidative stress (Fig. 3) (53).

The imbalance of intestinal flora structure is another crucial factor in the occurrence and development of NAFLD. It will affect the levels of metabolites such as short-chain fatty acids, bile acids and inflammatory factors (54). Acetic acid, propionic acid and butyric acid are the majority of short-chain fatty acids produced in the intestinal environment and their content ratio is ~3:1:1 (55). Butyric acid can limit ATP synthesis by regulating uncoupling and participate in liver fatty acid oxidation to reduce lipid accumulation (56,57). Sun et al (58) found that myricetin can increase the abundance of Bacteroides to increase the butyric acid content and inhibit liver fat accumulation induced by a high-fat diet (Fig. 3).

Tight junctions [claudins, occludin and (ZO)], adherens junctions and intercellular junctions consisting of desmosomes are important components of the intestinal barrier, effectively preventing the invasion of harmful substances and bacteria. Decreased expression of tight junction proteins induces significant enlargement of cell pores and increased intestinal mucosal permeability leads to impairment of the intestinal barrier (59). Intestinal bacterial metabolites (such as LPS) can be released into the blood and liver via the portal venous system, stimulating the release of inflammatory factors from the liver, thereby inducing NAFLD (60). Peña-Rodríguez et al (61) found that butyric acid upregulates the expression of occludin and ZO-1 and maintains the integrity of the intestinal mucosal barrier, thereby inhibiting liver inflammation. Similarly, Sun et al (58) found that myricetin can increase butyric acid content and promote the expression of ZO-1, thereby inhibiting high-fat diet-induced liver inflammation (Fig. 3). LPS can bind to TLR2/TLR4 on the surface of macrophages, inducing macrophages to polarize to the M1 type and releasing large amounts of pro-inflammatory factors, such as TNF-α, IL-1β and IL-6 (62). Myricetin can alleviate macrophage polarization to the M1 type and promote its polarization to the M2 type with an anti-inflammatory effect by reducing the expression of TLR2/TLR4 in macrophages and thus attenuating the inflammatory response in NAFLD (Fig. 3) (63).

Role of myricetin in liver fibrosis

Liver fibrosis is usually recognized as the pathological process of massive synthesis and sustained accumulation of extracellular matrix (ECM) following chronic liver injury. The activation process of hepatic stellate cells (HSCs) expressing α-smooth muscle actin (α-SMA) and producing large amounts of ECM is considered as the key link in the development of liver fibrosis (64). TGF-β1 is the most potent HSC activator. During HSC activation, first, TGF-β1 binds to the type II TGF-β receptors (TβRII), recruiting type I TGF-β receptors (TβRI) to form the TGF-β1/TβRII/TβRI complex (65). Then, the complex generated in the previous step induces a conformational change in TGF-β1R, which promotes the binding of Smad2 to Smad3 and phosphorylation to form the p-Smad2/p-Smad3 complex (66). Finally, the complex generated in the previous step binds to either α-SMA of the nucleus or DNA of connective tissue growth factor, regulating their expression to promote the activation of HSCs (67). Notably, myricetin inhibits the expression of TGF-β1, p-Smad2, p-Smad3 and α-SMA in the livers of mice infected with S. japonicum, thereby alleviating the development of liver fibrosis (Fig. 4) (68).

In addition, platelet-derived growth factor (PDGF) secreted by platelets promotes the activation of HSCs and the deposition of ECM, contributing to the development of liver fibrosis. PDGF-BB promotes the activation and proliferation of HSCs by activating PI3K/Akt and Ras-ERK signaling pathways via promoting the expression of p-AKT and p-ERK in CFSC-8 cells (rat hepatic stellate cells) cells (69). Geng et al (70) found that myricetin reverses the PDGF-BB-induced increase in the expression of p-AKT and p-ERK and decreases the expression of α-SMA and collagen type I (Collagen I) in CFSC-8 cells (Fig. 4). Similarly, myricetin inhibits the expression of liver TGF-β1 and α-SMA in the Ccl4-induced mouse liver fibrosis model, thereby alleviating liver fibrosis (Fig. 4) (71).

Role of myricetin in HCC

HCC is one of the most common cancers worldwide. The signalling pathways that regulate its occurrence and formation have become the focus of researchers' attention in recent years. The hippo signalling pathway controls liver tumorigenesis by regulating the balance between cell proliferation and apoptosis. In HCC, inactivation of the hippo signalling pathway promotes the entry of yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif into the nucleus, leading to the abnormal proliferation of HCC cells (72). Myricetin alleviates the development of HCC by inhibiting cell proliferation and promoting apoptosis (Fig. 5) (73). Mechanistically, myricetin promotes the phosphorylation of large tumor suppressor 1/2 to degrade YAP protein and reduce its nuclear translocation (73). This leads to a decrease in the expression of proliferation-promoting genes and an increase in the expression of anti-apoptotic genes in HCC cells (73). As a serine/threonine protein kinase, p21-activated kinase 1 (PAK1) activates the β-catenin signalling pathway to promote the proliferation of cancer cells and inhibit their apoptosis (74). Myricetin inhibits the expression of PKA1 and β-catenin in the diethylinitrosamine-induced rat HCC model, contributing to decreased proliferation and increased apoptosis of HCC cells (75).

Mitochondria are double-membrane organelles that play an important role in cellular energy production, metabolism and signal transduction. The liver, as a major metabolic organ, is rich in mitochondria. Mitochondrial dysfunction in hepatocytes leads to an abnormal increase in ROS, which promotes the activation of oncogenes, inducing the development of HCC (28,76). Moreover, ROS can promote oxidative stress in the liver, causing telomere shortening and chromosomal abnormalities and inducing the transformation of normal hepatocytes into HCC cells (77). However, Chandel and Tuveson (78) found that high levels of ROS can also promote apoptosis and necrosis in HCC cells, thus inhibiting the development of HCC. Analogously, Seydi et al (79) found that myricetin can target mitochondria and increase ROS levels and caspase-3 activity in HCC cells, thereby promoting apoptosis of HCC cells (Fig. 5). Briefly, myricetin inhibits the development of liver cancer by regulating the proliferation and apoptosis of HCC cells.

Limitations and future prospects

As the main constituent of medicinal and edible bayberry, myricetin possesses biological characteristics such as anti-tumour, anti-oxidation, anti-inflammatory and anti-bacterial properties (1820). In liver diseases, the mechanisms of myricetin's action on different types of liver diseases show diversity. However, these mechanisms still cannot fully explain the role of myricetin in various liver diseases and the specific molecular mechanisms need to be further explored. Although a large number of studies have confirmed that myricetin notably improves various liver diseases, this is only at the stage of animal or cellular experiments. To date, clinical trials with myricetin for the treatment of liver diseases have not yet been conducted. Therefore, the future work on myricetin in liver diseases needs to be more invested in clinical research. In addition, the stability of myricetin is limited, which leads to its lower bioavailability. Therefore, future research on the pharmaceutical dosage form of myricetin should be intensified to improve its bioavailability

As a monomer of TCM, myricetin is generally considered to be a safe drug. Yang et al (80) found no side effects in mice treated with 1,000 mg/kg of myricetin. In addition, human umbilical vein endothelial cells did not show significant cytotoxicity following treatment with myricetin (81). However, Canada et al (82) found that the cellular activity of isolated guinea pig intestinal cells was reduced and significant cellular damage occurred following treatment with myricetin. There are also those who consider that myricetin is capable of autoxidation, potentially leading to the development of oxidative stress (83). Therefore, there is an urgent need to further investigate the potential toxicity mechanisms of myricetin.

Conclusions

As a common disease, liver disease has become a serious threat to the health and lives of individuals all over the world. Although there are a wide variety of medications available for the treatment of liver diseases, they all possess significant side-effects and their efficacy remains unsatisfactory. Given the obvious advantages of TCM in research and development cost and therapeutic efficacy, the treatment of liver diseases with TCM has gradually been increasingly accepted and emphasized by researchers in recent years. The present study discussed the effectiveness and mechanism of myricetin in acute liver injury, fatty liver, liver fibrosis and HCC, which can facilitate further research on therapeutic drugs for liver diseases (Table I). It is hoped that the present review will draw the attention of liver disease research scholars to TCM.

Table I.

Role of myricetin in different liver diseases.

Table I.

Role of myricetin in different liver diseases.

First author, yearType of liver diseaseAnimal/cell modelFunctionsRole(Refs.)
Xiao, 2021Acute liver injury Lipopolysaccharide-induced Balb/c mice modelInhibits oxidative stress, inflammationAlleviates liver injury(24)
Kim, 2023Acute liver injury D-galactosamine-induced C57BL/6J mice model, HepG2 cellInhibits oxidative stress, inflammation, apoptosisAlleviates liver injury(26)
Shih, 2022Acute liver injuryPyrogallol-induced Wistar rat modelInhibit DNA damageAlleviates liver injury(35)
Chang, 2012AFLDEthanol-induced AML12 cellsInhibits inflammation, fatty acid synthesisAlleviates liver injury(37)
Leung and Nieto, 2013AFLDEthanol-induced Wistar rat modelPromote ethanol metabolism; Inhibits oxidative stress, inflammationAlleviates liver injury(42)
Chen, 2023NAFLDHigh-fat-fed C57BL/6J mice modelPromote lipid metabolismAlleviates liver injury(49)
Xia, 2019NAFLDob/ob mice modelInhibits oxidative stress, lipid accumulationAlleviates liver injury(50)
Lei, 2024NAFLDHigh-fat-fed C57BL/6J mice modelInhibits oxidative stress, lipid accumulationAlleviates liver injury(52)
Zhang, 2023NAFLDHigh-fat-fed rat modelInhibits inflammation, lipid synthesisAlleviates liver injury(57)
Peña-Rodríguez, 2022NAFLDHigh-fat-fed C57BL/6J mice model, RAW264.7 cellsInhibit lipid accumulation, liver fibrosis, cell death, inflammationAlleviates liver injury(61)
Dewidar, 2019Liver fibrosisSchistosoma japonicum-Infected BALB/c mice modelInhibits inflammation and fibrosisAlleviates liver injury(67)
Li, 2020Liver fibrosisCcl4-induced BALB/c mice model and CFSC-8B cellsInhibits HSC proliferation, migration, ECM accumulationAttenuates liver fibrosis(69)
Geng, 2017Liver fibrosisCcl4-induced BALB/cN mice modelInhibits oxidative stress, inflammation, fibrosis; Promote proliferationAlleviates liver injury(70)
Lee, 2021HCCHepG2 and Huh-7Inhibits HCC cell proliferation, Promote apoptosisAlleviates liver fibrosis(72)
Li, 2019HCCDEN-induced rat model, HepG2Promotes apoptosisInhibits HCC(73)
Chandel and Tuveson, 2014HCCDEN/2-AAF-induced rat modelPromotes oxidative stress and apoptosisInhibits HCC(78)

[i] AFLD, alcoholic fatty liver disease; NAFLD, non-alcoholic fatty liver disease; HCC, hepatocellular carcinoma.

Acknowledgements

Not applicable.

Funding

The present review was supported by the Natural Science Foundation of Hubei Province (grant no. 2024AFB494 to Mi Chen), the Foundation of Hubei University of Science and Technology Science (grant no. BK202336 to Mi Chen).

Availability of data and materials

Not applicable.

Author's contributions

CO and YL conceptualized the manuscript and revised and edited the manuscript. MC and SZ drafted the manuscript and prepared the figures. XH, DZha and DZhu supervised and revised the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Frazier K, Manzoor S, Carroll K, DeLeon O, Miyoshi S, Miyoshi J, St George M, Tan A, Chrisler EA, Izumo M, et al: Gut microbes and the liver circadian clock partition glucose and lipid metabolism. J Clin Invest. 133:e1625152023. View Article : Google Scholar : PubMed/NCBI

2 

Sinha RA, Singh BK and Yen PM: Direct effects of thyroid hormones on hepatic lipid metabolism. Nat Rev Endocrinol. 14:259–269. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Sun J, Yu X, Weng Z, Jin L and Yang J, Zhang H, Gu J, Wang N and Yang J: The impact of hepatotoxic drugs on the outcome of patients with acute deterioration of hepatitis B virus-related chronic disease. Eur J Gastroenterol Hepatol. 34:782–790. 2022.PubMed/NCBI

4 

Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E and Kamath PS: Global burden of liver disease: 2023 update. J Hepatol. 79:516–537. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Pate J, Gutierrez JA, Frenette CT, Goel A, Kumar S, Manch RA, Mena EA, Pockros PJ, Satapathy SK, Yimam KK and Gish RG: Practical strategies for pruritus management in the obeticholic acid-treated patient with PBC: Proceedings from the 2018 expert panel. BMJ Open Gastroenterol. 6:e0002562019. View Article : Google Scholar : PubMed/NCBI

6 

Zhu YJ, Zheng B, Wang HY and Chen L: New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacol Sin. 38:614–622. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Semwal DK, Semwal RB, Combrinck S and Viljoen A: Myricetin: A dietary molecule with diverse biological activities. Nutrients. 8:902016. View Article : Google Scholar : PubMed/NCBI

8 

Nardini M and Garaguso I: Characterization of bioactive compounds and antioxidant activity of fruit beers. Food Chem. 305:1254372020. View Article : Google Scholar : PubMed/NCBI

9 

Jiang C, Xie L, Wang Y, Liang J, Li H, Luo L, Li T, Liang Z, Tang L, Ning D, et al: Highly sensitive electrochemical detection of myricetin in food samples based on the enhancement effect of Al-MOFs. Anal Methods. 14:3521–3528. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Klimek-Szczykutowicz M, Gaweł-Bęben K, Rutka A, Blicharska E, Tatarczak-Michalewska M, Kulik-Siarek K, Kukula-Koch W, Malinowska MA and Szopa A: Moringa oleifera (drumstick tree)-nutraceutical, cosmetological and medicinal importance: A review. Front Pharmacol. 15:12883822024. View Article : Google Scholar : PubMed/NCBI

11 

Wang M, Ren S, Bi Z, Zhang L, Cui M, Sun R, Bao J, Gao D, Yang B, Li X, et al: Myricetin reverses epithelial-endothelial transition and inhibits vasculogenic mimicry and angiogenesis of hepatocellular carcinoma by directly targeting PAR1. Phytother Res. 36:1807–1821. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Rostami A, Baluchnejadmojarad T and Roghani M: Hepatoprotective effect of myricetin following Lipopolysaccharide/DGalactosamine: Involvement of autophagy and sirtuin 1. Curr Mol Pharmacol. 16:419–433. 2023. View Article : Google Scholar : PubMed/NCBI

13 

Perkin AG and Hummel JJ: LXXVI-The colouring principle contained in the bark of Myrica nagi part I. J Chem Soc Trans. 69:1287–1294. 1896. View Article : Google Scholar

14 

Ozcan C and Yaman M: Determination of Myricetin in medicinal plants by high-performance liquid chromatography. In strum Sci Technol. 43:44–52. 2015.

15 

Perkin AG: XXI.-Myricetin. Part II. J Chem Soc Trans. 81:203–210. 1902. View Article : Google Scholar

16 

He J, Wang Y, Chang AK, Xu L, Wang N, Chong X, Li H, Zhang B, Jones GW and Song Y: Myricetin prevents fibrillogenesis of hen egg white lysozyme. J Agric Food Chem. 62:9442–9449. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Jiang M, Zhu M, Wang L and Yu S: Anti-tumor effects and associated molecular mechanisms of myricetin. Biomed Pharmacother. 120:1095062019. View Article : Google Scholar : PubMed/NCBI

18 

Zhou JL, Chen HH, Xu J, Huang MY, Wang JF, Shen HJ, Shen SX, Gao CX and Qian CD: Myricetin acts as an inhibitor of type II NADH Dehydrogenase from Staphylococcus aureus. Molecules. 29:23542024. View Article : Google Scholar : PubMed/NCBI

19 

Hou DD, Gu YJ, Wang DC, Niu Y, Xu ZR, Jin ZQ, Wang XX and Li SJ: Therapeutic effects of myricetin on atopic dermatitis in vivo and in vitro. Phytomedicine. 102:1542002022. View Article : Google Scholar : PubMed/NCBI

20 

Li Q, Tan Q, Ma Y, Gu Z and Chen S: Myricetin suppresses ovarian cancer in vitro by activating the p38/Sapla signaling pathway and suppressing intracellular oxidative stress. Front Oncol. 12:9033942022. View Article : Google Scholar : PubMed/NCBI

21 

Shrivastava R and Chng SS: Lipid trafficking across the Gram-negative cell envelope. J Biol Chem. 294:14175–14184. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Wang P, Han X, Mo B, Huang G and Wang C: LPS enhances TLR4 expression and IFN-γ production via the TLR4/IRAK/NF-κB signaling pathway in rat pulmonary arterial smooth muscle cells. Mol Med Rep. 16:3111–3116. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Fuchs O: Transcription factor NF-κB inhibitors as single therapeutic agents or in combination with classical chemotherapeutic agents for the treatment of hematologic malignancies. Curr Mol Pharmacol. 3:98–122. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Xiao P, Li M, Zhou M, Zhao X, Wang C, Qiu J, Fang Q, Jiang H, Dong H and Zhou R: TTP protects against acute liver failure by regulating CCL2 and CCL5 through m6A RNA methylation. JCI Insight. 6:e1492762021. View Article : Google Scholar : PubMed/NCBI

25 

Berköz M, Ünal S, Karayakar F, Yunusoğlu O, Özkan-Yılmaz F, Özlüer-Hunt A and Aslan A: Prophylactic effect of myricetin and apigenin against lipopolysaccharide-induced acute liver injury. Mol Biol Rep. 48:6363–6373. 2021. View Article : Google Scholar : PubMed/NCBI

26 

Kim JM, Cho SS, Kang S, Moon C, Yang JH and Ki SH: Castanopsis sieboldii extract alleviates acute liver injury by antagonizing inflammasome-mediated pyroptosis. Int J Mol Sci. 24:119822023. View Article : Google Scholar : PubMed/NCBI

27 

Lv H, An B, Yu Q, Cao Y, Liu Y and Li S: Prophylactic effect of myricetin and apigenin against lipopolysaccharide-induced acute liver injury lipopolysaccharide and D-galactosamine-induced fulminant hepatitis. Int J Biol Macromol. 155:1092–1104. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Lee HJ, Oh YK, Rhee M, Lim JY, Hwang JY, Park YS, Kwon Y, Choi KH, Jo I, Park SI, et al: The role of STAT1/IRF-1 on synergistic ROS production and loss of mitochondrial transmembrane potential during hepatic cell death induced by LPS/d-GalN. J Mol Biol. 369:967–984. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Sheftel AD, Kim SF and Ponka P: Non-heme induction of heme oxygenase-1 does not alter cellular iron metabolism. J Biol Chem. 282:10480–10486. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Lin L, Wu Q, Lu F, Lei J, Zhou Y, Liu Y, Zhu N, Yu Y, Ning Z, She T and Hu M: Nrf2 signaling pathway: Current status and potential therapeutic targetable role in human cancers. Front Oncol. 13:11840792023. View Article : Google Scholar : PubMed/NCBI

31 

Huang J, Wu L, Tashiro S, Onodera S and Ikejima T: Reactive oxygen species mediate oridonin-induced HepG2 apoptosis through p53, MAPK, and mitochondrial signaling pathways. J Pharmacol Sci. 107:370–379. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Miyashita T, Krajewski S, Krajewska M, Wang HG, Lin HK, Liebermann DA, Hoffman B and Reed JC: Tumor suppressor p53 is a regulator of bcl-2 and bax gene expression in vitro and in vivo. Oncogene. 9:1799–1805. 1994.PubMed/NCBI

33 

Garrido C, Galluzzi L, Brunet M, Puig PE, Didelot C and Kroemer G: Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 13:1423–1433. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Nagata S, Nagase H, Kawane K, Mukae N and Fukuyama H: Degradation of chromosomal DNA during apoptosis. Cell Death Differ. 10:108–116. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Shih PC: The role of the STAT3 signaling transduction pathways in radioresistance. Pharmacol Ther. 234:1081182022. View Article : Google Scholar : PubMed/NCBI

36 

Matić S, Stanić S, Bogojević D, Vidaković M, Grdović N, Dinić S, Solujić S, Mladenović M, Stanković N and Mihailović M: Methanol extract from the stem of Cotinus coggygria Scop., and its major bioactive phytochemical constituent myricetin modulate pyrogallol-induced DNA damage and liver injury. Mutat Res. 755:81–89. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Chang CJ, Tzeng TF, Liou SS, Chang YS and Liu IM: myricetin increases hepatic peroxisome proliferator-activated receptor α protein expression and decreases plasma lipids and adiposity in rats. Evid Based Complement Alternat Med. 2012:7871522012. View Article : Google Scholar : PubMed/NCBI

38 

Guo C, Xue G, Pan B, Zhao M, Chen S, Gao J, Chen T and Qiu L: Myricetin ameliorates ethanol-induced lipid accumulation in liver cells by reducing fatty acid biosynthesis. Mol Nutr Food Res. 63:e18013932019. View Article : Google Scholar : PubMed/NCBI

39 

Wang W, Zhai T, Luo P, Miao X, Wang J and Chen Y: Beneficial effects of silibinin on serum lipids, bile acids, and gut microbiota in methionine-choline-deficient diet-induced mice. Front Nutr. 10:12571582023. View Article : Google Scholar : PubMed/NCBI

40 

Wang C, Gong B, Peng D, Liu Y, Wu Y and Wei J: Agarwood extract mitigates alcoholic fatty liver in C57 mice via anti oxidation and anti inflammation. Mol Med Rep. 28:2102023. View Article : Google Scholar : PubMed/NCBI

41 

Osna NA, Rasineni K, Ganesan M, Donohue TM Jr and Kharbanda KK: Pathogenesis of alcohol-associated liver disease. J Clin Exp Hepatol. 12:1492–1513. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Leung TM and Nieto N: CYP2E1 and oxidant stress in alcoholic and non-alcoholic fatty liver disease. J Hepatol. 58:395–398. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Ahmad SB, Rashid SM, Wali AF, Ali S, Rehman MU, Maqbool MT, Nadeem A, Ahmad SF and Siddiqui N: Myricetin (3,3′,4′,5,5′,7-hexahydroxyflavone) prevents ethanol-induced biochemical and inflammatory damage in the liver of Wistar rats. Hum Exp Toxicol. 41:96032712110668432022. View Article : Google Scholar : PubMed/NCBI

44 

Schlaepfer IR and Joshi M: CPT1A-mediated fat oxidation, mechanisms, and therapeutic potential. Endocrinology. 161:bqz0462020. View Article : Google Scholar : PubMed/NCBI

45 

Fang C, Pan J, Qu N, Lei Y, Han J, Zhang J and Han D: The AMPK pathway in fatty liver disease. Front Physiol. 13:9702922022. View Article : Google Scholar : PubMed/NCBI

46 

Ludwig J, Viggiano TR, McGill DB and Oh BJ: Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 55:434–438. 1980. View Article : Google Scholar : PubMed/NCBI

47 

Lian CY, Zhai ZZ, Li ZF and Wang L: High fat diet-triggered non-alcoholic fatty liver disease: A review of proposed mechanisms. Chem Biol Interact. 330:1091992020. View Article : Google Scholar : PubMed/NCBI

48 

Li L, Sun H, Chen J, Ding C, Yang X, Han H and Sun Q: Mitigation of non-alcoholic steatohepatitis via recombinant Orosomucoid 2, an acute phase protein modulating the Erk1/2-PPARγ-Cd36 pathway. Cell Rep. 42:1126972023. View Article : Google Scholar : PubMed/NCBI

49 

Chen H, Tan H, Wan J, Zeng Y, Wang J, Wang H and Lu X: PPAR-γ signaling in nonalcoholic fatty liver disease: Pathogenesis and therapeutic targets. Pharmacol Ther. 245:1083912023. View Article : Google Scholar : PubMed/NCBI

50 

Xia SF, Qiu YY, Chen LM, Jiang YY, Huang W, Xie ZX, Tang and Sun J: Myricetin alleviated hepatic steatosis by acting on microRNA-146b/thyroid hormone receptor b pathway in high-fat diet fed C57BL/6J mice. Food Funct. 10:1465–1477. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Choi HN, Shin JY and Kim JI: Ameliorative effect of myricetin on nonalcoholic fatty liver disease in ob/ob Mice. J Med Food. 24:1092–1099. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Lei ZY, Li ZH, Lin DN, Cao J, Chen JF, Meng SB, Wang JL, Liu J, Zhang J and Lin BL: Med1 inhibits ferroptosis and alleviates liver injury in acute liver failure via Nrf2 activation. Cell Biosci. 14:542024. View Article : Google Scholar : PubMed/NCBI

53 

Xia SF, Le GW, Wang P, Qiu YY, Jiang YY and Tang X: Regressive effect of myricetin on hepatic steatosis in mice fed a high-fat diet. Nutrients. 8:7992016. View Article : Google Scholar : PubMed/NCBI

54 

Tang WHW, Li DY and Hazen SL: Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol. 16:137–154. 2019. View Article : Google Scholar : PubMed/NCBI

55 

van der Hee B and Wells JM: Microbial regulation of host physiology by short-chain fatty acids. Trends Microbiol. 29:700–712. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Beauvieux MC, Tissier P, Gin H, Canioni P and Gallis JL: Butyrate impairs energy metabolism in isolated perfused liver of fed rats. J Nutr. 131:1986–1992. 2001. View Article : Google Scholar : PubMed/NCBI

57 

Zhang L, Chen N, Zhan L, Bi T, Zhou W, Zhang L and Zhu L: Erchen Decoction alleviates obesity-related hepatic steatosis via modulating gut microbiota-drived butyric acid contents and promoting fatty acid β-oxidation. J Ethnopharmacol. 317:1168112023. View Article : Google Scholar : PubMed/NCBI

58 

Sun WL, Li XY, Dou HY, Wang XD, Li JD, Shen L and Ji HF: Myricetin supplementation decreases hepatic lipid synthesis and inflammation by modulating gut microbiota. Cell Rep. 36:1096412021. View Article : Google Scholar : PubMed/NCBI

59 

Chelakkot C, Ghim J and Ryu SH: Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med. 50:1–9. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Xu M, Luo K, Li J, Li Y, Zhang Y, Yuan Z, Xu Q and Wu X: Role of intestinal microbes in chronic liver diseases. Int J Mol Sci. 23:126612022. View Article : Google Scholar : PubMed/NCBI

61 

Peña-Rodríguez M, Vega-Magaña N, García-Benavides L, Zepeda-Nuño JS, Gutierrez-Silerio GY, González-Hernández LA, Andrade-Villanueva JF, Del Toro-Arreola S, Pereira-Suárez AL and Bueno-Topete MR: Butyrate administration strengthens the intestinal epithelium and improves intestinal dysbiosis in a cholestasis fibrosis model. J Appl Microbiol. 132:571–583. 2022. View Article : Google Scholar : PubMed/NCBI

62 

Hirschfeld M, Weis JJ, Toshchakov V, Salkowski CA, Cody MJ, Ward DC, Qureshi N, Michalek SM and Vogel SN: Signaling by toll-like receptor 2 and 4 agonists results in differential gene expression in murine macrophages. Infect Immun. 69:1477–1482. 2001. View Article : Google Scholar : PubMed/NCBI

63 

Yao Q, Li S, Li X, Wang F and Tu C: myricetin modulates macrophage polarization and mitigates liver inflammation and fibrosis in a murine model of nonalcoholic steatohepatitis. Front Med (Lausanne). 7:712020. View Article : Google Scholar : PubMed/NCBI

64 

Yuan S, Wei C, Liu G, Zhang L, Li J, Li L, Cai S and Fang L: Sorafenib attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis via HIF-1α/SLC7A11 pathway. Cell Prolif. 55:e131582022. View Article : Google Scholar : PubMed/NCBI

65 

Yakymovych I, Yakymovych M, Hamidi A, Landström M and Heldin CH: The type II TGF-β receptor phosphorylates Tyr182 in the type I receptor to activate downstream Src signaling. Sci Signal. 15:eabp95212022. View Article : Google Scholar : PubMed/NCBI

66 

Hu Y, Li Z, Gong L and Song Z: β-Asarone suppresses TGF-β/Smad signaling to reduce the invasive properties in esophageal squamous cancer cells. Med Oncol. 39:2432022. View Article : Google Scholar : PubMed/NCBI

67 

Dewidar B, Meyer C, Dooley S and Meindl-Beinker AN: TGF-β in hepatic stellate cell activation and liver fibrogenesis-updated 2019. Cells. 8:14192019. View Article : Google Scholar : PubMed/NCBI

68 

Huang P, Zhou M, Cheng S, Hu Y, Gao M, Ma Y, Limpanont Y, Zhou H, Dekumyoy P, Cheng Y and Lv Z: myricetin possesses anthelmintic activity and attenuates hepatic fibrosis via modulating TGFβ1 and Akt signaling and shifting Th1/Th2 balance in schistosoma japonicum-infected mice. Front Immunol. 11:5932020. View Article : Google Scholar : PubMed/NCBI

69 

Li HG, You PT, Xia Y, Cai Y, Tu YJ, Wang MH, Song WC, Quan TM, Ren HY, Liu YW, et al: Yu Gan Long ameliorates hepatic fibrosis by inhibiting PI3K/AKT, Ras/ERK and JAK1/STAT3 signaling pathways in CCl4-induced liver fibrosis rats. Curr Med Sci. 40:539–547. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Geng Y, Sun Q, Li W, Lu ZM, Xu HY, Shi JS and Xu ZH: The common dietary flavonoid myricetin attenuates liver fibrosis in carbon tetrachloride treated mice. Mol Nutr Food Res. 61:2016003922017. View Article : Google Scholar

71 

Domitrović R, Rashed K, Cvijanović O, Vladimir-Knežević S, Škoda M and Višnić A: Myricitrin exhibits antioxidant, anti-inflammatory and antifibrotic activity in carbon tetrachloride-intoxicated mice. Chem Biol Interact. 230:21–29. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Lee NH, Kim SJ and Hyun J: MicroRNAs regulating Hippo-YAP signaling in liver cancer. Biomedicines. 9:3472021. View Article : Google Scholar : PubMed/NCBI

73 

Li M, Chen J, Yu X, Xu S, Li D, Zheng Q and Yin Y: Myricetin suppresses the propagation of hepatocellular carcinoma via down-regulating expression of YAP. Cells. 8:3582019. View Article : Google Scholar : PubMed/NCBI

74 

He H, Huynh N, Liu KH, Malcontenti-Wilson C, Zhu J, Christophi C, Shulkes A and Baldwin GS: P-21 activated kinase 1 knockdown inhibits β-catenin signalling and blocks colorectal cancer growth. Cancer Lett. 317:65–71. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Iyer SC, Gopal A and Halagowder D: Myricetin induces apoptosis by inhibiting P21 activated kinase 1 (PAK1) signaling cascade in hepatocellular carcinoma. Mol Cell Biochem. 407:223–237. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Lee HY, Nga HT, Tian J and Yi HS: Mitochondrial metabolic signatures in hepatocellular carcinoma. Cells. 10:19012021. View Article : Google Scholar : PubMed/NCBI

77 

Cardin R, Piciocchi M, Bortolami M, Kotsafti A, Barzon L, Lavezzo E, Sinigaglia A, Rodriguez-Castro KI, Rugge M and Farinati F: Oxidative damage in the progression of chronic liver disease to hepatocellular carcinoma: An intricate pathway. World J Gastroenterol. 20:3078–3086. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Chandel NS and Tuveson DA: The promise and perils of antioxidants for cancer patients. N Engl J Med. 371:177–178. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Seydi E, Rasekh HR, Salimi A, Mohsenifar Z and Pourahmad J: Myricetin selectively induces apoptosis on cancerous hepatocytes by directly targeting their mitochondria. Basic Clin Pharmacol Toxicol. 119:249–258. 2016. View Article : Google Scholar : PubMed/NCBI

80 

Yang Y, Choi JK, Jung CH, Koh HJ, Heo P, Shin JY, Kim S, Park WS, Shin HJ and Kweon DH: SNARE-wedging polyphenols as small molecular botox. Planta Med. 8:233–236. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Kim JD, Liu L, Guo W and Meydani M: Chemical structure of flavonols in relation to modulation of angiogenesis and immune-endothelial cell adhesion. J Nutr Biochem. 7:165–176. 2006. View Article : Google Scholar

82 

Canada AT, Watkins WD and Nguyen TD: The toxicity of flavonoids to guinea pig enterocytes. Toxicol Appl Pharmacol. 99:357–361. 1989. View Article : Google Scholar : PubMed/NCBI

83 

Canada AT, Giannella E, Nguyen TD and Mason RP: The production of reactive oxygen species by dietary flavonols. Free Radic Biol Med. 9:441–449. 1990. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2025
Volume 31 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen M, Zhang S, Huang X, Zhang D, Zhu D, Ouyang C and Li Y: The protective effects and mechanism of myricetin in liver diseases (Review). Mol Med Rep 31: 87, 2025.
APA
Chen, M., Zhang, S., Huang, X., Zhang, D., Zhu, D., Ouyang, C., & Li, Y. (2025). The protective effects and mechanism of myricetin in liver diseases (Review). Molecular Medicine Reports, 31, 87. https://doi.org/10.3892/mmr.2025.13452
MLA
Chen, M., Zhang, S., Huang, X., Zhang, D., Zhu, D., Ouyang, C., Li, Y."The protective effects and mechanism of myricetin in liver diseases (Review)". Molecular Medicine Reports 31.4 (2025): 87.
Chicago
Chen, M., Zhang, S., Huang, X., Zhang, D., Zhu, D., Ouyang, C., Li, Y."The protective effects and mechanism of myricetin in liver diseases (Review)". Molecular Medicine Reports 31, no. 4 (2025): 87. https://doi.org/10.3892/mmr.2025.13452