Next Article in Journal
An Integrated Framework for Drought Stress in Plants
Previous Article in Journal
A Combined Proteomic and Transcriptomic Signature Is Predictive of Response to Anti-PD-1 Treatment: A Retrospective Study in Metastatic Melanoma Patients
Previous Article in Special Issue
High Resolution HLA-A, HLA-B, and HLA-C Allele Frequencies in Romanian Hematopoietic Stem Cell Donors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems

1
“Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
2
Department of Molecular and Nanopharmaceutics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
3
Faculty of Pharmacy, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
4
Faculty of Medicine, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(17), 9346; https://doi.org/10.3390/ijms25179346
Submission received: 5 August 2024 / Revised: 21 August 2024 / Accepted: 27 August 2024 / Published: 28 August 2024

Abstract

:
Chronic liver injuries often lead to hepatic fibrosis, a condition characterized by excessive extracellular matrix accumulation and abnormal connective tissue hyperplasia. Without effective treatment, hepatic fibrosis can progress to cirrhosis or hepatocellular carcinoma. Current treatments, including liver transplantation, are limited by donor shortages and high costs. As such, there is an urgent need for effective therapeutic strategies. This review focuses on the potential of plant-based therapeutics, particularly polyphenols, phenolic acids, and flavonoids, in treating hepatic fibrosis. These compounds have demonstrated anti-fibrotic activities through various signaling pathways, including TGF-β/Smad, AMPK/mTOR, Wnt/β-catenin, NF-κB, PI3K/AKT/mTOR, and hedgehog pathways. Additionally, this review highlights the advancements in nanoparticulate drug delivery systems that enhance the pharmacokinetics, bioavailability, and therapeutic efficacy of these bioactive compounds. Methodologically, this review synthesizes findings from recent studies, providing a comprehensive analysis of the mechanisms and benefits of these plant-based treatments. The integration of novel drug delivery systems with plant-based therapeutics holds significant promise for developing effective treatments for hepatic fibrosis.

1. Introduction

Chronic liver injuries are a primary manifestation of hepatic fibrosis [1], which represents an abnormal wound healing response characterized by excessive extracellular matrix (ECM) accumulation and abnormal connective tissue hyperplasia [2]. Without effective treatment, hepatic fibrosis can advance to cirrhosis or hepatocellular carcinoma [3]. Currently, liver transplantation is the most effective treatment for cirrhosis, but its clinical application is limited due to the shortage of donors and high costs [4]. There is no specific medication for treating hepatic fibrosis, and many hepatic anti-fibrotic drugs are still in the research and development phase [1]. Considering the severe consequences of hepatic fibrosis, understanding the underlying mechanisms leading to its development and progression is crucial. This understanding is essential for developing effective therapeutic strategies [2].
Polyphenols are increasingly gaining attention for the development of potential drugs for liver disease treatment. Numerous polyphenols have demonstrated hepatic anti-fibrotic activity by inhibiting the activity of hepatic stellate cells (HSCs) [2]. These bioactive compounds operate through various pathways, including the TGF-β/Smad signaling pathways, AMPK/mTOR, Wnt/β-catenin, NF-κB, PI3K/AKT/mTOR, hedgehog pathways, and other factors associated with hepatic fibrosis.
Hepatic fibrosis can be mitigated using medicinal plants, plant extracts, and bioactive compounds derived from plants that inhibit the activation of hepatic stellate cells and reduce ECM deposition [5,6]. Plant extracts are a mixture of bioactive compounds and pharmacokinetic synergists [7]. These biologically active compounds can work synergistically to enhance the therapeutic efficacy of plant-based medicines [8]. Medicinal plants and their phytocompounds can protect the liver through various mechanisms, including the inhibition of fibrogenesis, oxidative stress, and tumor growth [9].
As most liver injuries are chronic conditions that require long-term treatment, it is important to minimize the side effects of hepatoprotective drugs. All bioactive compounds, including plant-based drugs, can have adverse effects. Therefore, further research on plant-based drugs with hepatic anti-fibrotic effects is necessary [10]. Despite significant progress in understanding the pathogenesis of hepatic fibrosis, no effective agent has been developed yet to prevent or directly reverse the fibrotic process [11]. The administered dose of biologically active compounds significantly influences the clinical response. Higher doses of such compounds have shown superior clinical efficacy but are associated with increased toxicity in various organs [12]. Many plant-based drugs and plant extracts have poor absorption and low bioavailability due to their poor lipid solubility or improper molecular sizes [13].
The aim of this review is to comprehensively analyze and synthesize current research on the anti-fibrotic effects of polyphenols, specifically phenolic acids and flavonoids, and to evaluate the advancements in nanoparticulate drug delivery systems that enhance the pharmacokinetics and therapeutic efficacy of these bioactive compounds. By exploring the molecular mechanisms through which polyphenols modulate key signaling pathways implicated in hepatic fibrosis, and by assessing the potential of novel delivery systems to improve their bioavailability and reduce side effects, this review seeks to provide a detailed understanding of the potential therapeutic applications in the treatment of hepatic fibrosis and to identify future research directions in this field.

2. Polyphenols and Hepatic Fibrosis

The intricate mechanisms driving hepatic fibrosis highlight the need for combined therapeutic approaches that target multiple signaling pathways. In addition to chemical compounds, various natural products have shown effectiveness in treating hepatic fibrosis [14]. Polyphenols, which are secondary metabolites naturally found in many plant-derived foods and beverages commonly consumed in the human diet, are particularly noteworthy. Based on their chemical structure, polyphenols are classified into several categories: phenolic acids (including hydroxycinnamic and hydroxybenzoic acids), flavonoids, stilbenes, tannins, and lignans [15,16,17,18,19].

2.1. Phenolic Acids

Phenolic acids are the simplest phenolic compounds, characterized by a single phenolic ring with multiple hydroxyl or methoxyl groups attached [20]. They are divided into two main categories: derivatives of hydroxycinnamic acid and derivatives of hydroxybenzoic acid [21].
Hydroxycinnamic acids are aromatic carboxylic acids with an unsaturated side chain [22]. In these acids, the carboxylic acid functional group is separated from the phenol ring by a double bond (C=C) [21]. Cinnamic acids function as phytohormones and are precursors to chalcones, flavonoids, anthocyanins, and stilbenes [22]. Hydroxycinnamic acids associated with hepatic fibrosis include chlorogenic acid, ferulic acid, isochlorogenic acid, p-coumaric acid, rosmarinic acid, salvianolic acids A and B, and sinapic acid (Table 1).
Hydroxybenzoic acids are phenols substituted with a carboxylic acid functional group directly bonded to the phenol ring [21]. These acids are less abundant in plants and are components of complex structures such as tannins and lignins [23]. This category includes p-hydroxybenzoic, protocatechuic, vanillic, syringic, and gallic acids [24]. Hydroxybenzoic acids with hepatic anti-fibrotic activity include gallic acid, protocatechuic acid, and vanillic acid (Table 1).
Table 1. Pharmacological effects of phenolic acids in liver fibrosis.
Table 1. Pharmacological effects of phenolic acids in liver fibrosis.
Class of
Phenolic Acids
Bioactive CompoundsCell Lines/
Animal Model
Pharmacological EffectsReference
Hydroxycinnamic acidsChlorogenic acidLX-2 cells
Sprague-Dawley rats
-
inhibited the mRNA expression of miR-21, CTGF, α-SMA, TIMP-1, and TGF-β1 and the protein expression of p-Smad2, p-Smad3, p-Smad2/3, CTGF, α-SMA, TIMP-1, and TGF-β1 both in vitro and in vivo
-
↑ the mRNA and protein expression of Smad7 and MMP-9
-
↓ the degree of liver fibrosis
-
↓ α-SMA and Col-1 expression in liver tissue
-
↓ TGF-β1 in serum
[25]
Sprague-Dawley rats
-
attenuated CCl4-induced liver damage
-
↓ ALT, AST
-
↑ ALB
-
alleviated the degree of liver fibrogenesis and formation of pseudo-lobulus
-
↓ α-SMA and Col-1
-
↓ the expression levels of TLR4, MyD88, iNOS, and COX-2
-
↑ BAMBI
-
suppressed CCl4-induced NF-κB activation
-
↓ hepatic mRNA expression and serum levels of TNF-α, IL-6, and IL-1β
[26]
Ferulic acidMPHs, RAW 264.7 cells, and LX-2 cells
C57BL/6J mice
-
ameliorated CCl4-induced inflammation and fibrotic liver damage in mice
-
↓ ALT, AST
-
↓ MDA
-
↑ SOD
-
↓ collagen deposition
-
↓ fibronectin, Col-1, TGF-β, Acta2
-
↑ the phosphorylation of AMPK and ERK1/2
-
inhibited hepatic oxidative stress, macrophage activation, and HSC activation via AMPK phosphorylation in different liver cells
-
↓ MDA, NOX2
-
↑ SOD
-
↓ ROS production in MPHs
-
↓ IL-1β, F4/80, and Cd11b
-
↓ pro-inflammatory gene transcript levels of Ccl2 and TNF-α, IL-6, and iNOS
-
promoted the translocation of NF-κB P50 and P65 from nucleus to cytoplasm
-
inhibited the activity of PTP1B
[27]
Isochlorogenic acid BC57BL/6 mice
-
improved the pathological lesions of liver fibrosis
-
↓ serum ALT, AST, HYP, cholesterol, triglycerides
-
inhibited HSC activation
-
↓ the expressions of hepatic genes involved in liver fibrosis: LOX, TGF-β1, MCP-1, Col1α1, TIMP-1
-
attenuated liver oxidative stress through Nrf2 signaling pathway
[28]
p-Coumaric acidLX-2 cells
C57BL/6 mice
-
improved systemic insulin sensitivity without altering adiposity
-
↓ ALT, AST
-
attenuated hepatic signaling pathways associated with NLRP3 inflammasome activation: TLR4/NF-κB, and endoplasmic reticulum/oxidative stress
-
↓ circulating IL-1β levels
-
ameliorated hepatic fibrosis
-
↓ the excessive deposition of collagen fibers
-
↓ α-SMA
-
normalized the expression of TGF-β, Col1α2, Col3α1, Col4α1, and TIMP-1
-
↓ NLRP3 activation and caspase-1 cleavage
[29]
Rosmarinic acidSprague-Dawley rats
HSC-T6
-
inhibited HSC proliferation
-
inhibited TGF-β1, CTGF, and α-SMA expression in cultured HSCs
-
↓ collagen deposition
-
ameliorated hepatocyte degeneration, necrosis, and infiltration of inflammatory cells
-
↓ serum levels of HA, LN, and PCIII
-
↑ ALB/GLB
-
↓ ALT, AST, HYP
-
inhibited TGF-β1, CTGF expression in vivo
[30]
Salvianolic acid ASprague-Dawley rats
-
↓ liver fibrosis by inhibiting liver function, liver fibrosis index, and collagen deposition in vivo
-
↓ ALT, AST, HA, CIV, LN, PIIIP
-
↓ HYP
-
↓ α-SMA, TGF-β1, PDGF-βR, CTGF, desmin, and vimentin
-
inhibited the PI3K/AKT/mTOR signaling cascade
-
↓ p-AKT, p-mTOR, p-p70S6K1
-
prevented the stimulation of hepatic stellate cells and the synthesis of ECM
-
↓ the hepatocyte apoptosis
-
↑ Bcl-2
-
↓ Bax
-
↓ caspase-3 and cleaved caspase-3
[31]
Salvianolic acid BC57BL/6 mice
LO2 cells
-
attenuated liver fibrosis in CCl4-induced mice
-
↓ ALT, AST
-
↓ the hepatic inflammatory cell infiltration
-
↓ collagen levels
-
alleviated liver fibrosis in mice by targeting up-regulation of Ecm1 and inhibiting hepatocyte ferroptosis
[32]
C57BL/6 mice
LX2 and WRL68 cells
-
alleviated hepatic fibrogenesis by inhibiting the activation of HSCs and collagen deposition
-
↓ ALT, AST
-
displayed anti-inflammatory effects in CCl4-induced liver fibrosis
-
↓ IL-1β, IL-6
-
↓ the infiltration of CD68 and CD11b cells in liver
-
↓ γH2AX
[33]
HSC-LX-2 cells
BALB/c mice
-
had a good binding ability to PDGFRβ
-
inhibited the activation of HSCs in vitro
-
↓ the mRNA expression levels of α-SMA and Col-1
-
attenuated HSC activation by targeting PDGFRβ pathways
-
↓ p-AKT/AKT, p-ERK/ERK, and p-p38/p38 signaling pathways
-
inhibited the migration and proliferation and promoted apoptosis of HSCs
-
suppressed PDGF-BB-induced HSC activation and the PDGFBB/PDGFRβ pathway in vitro
-
↓ α-SMA and Col-1
-
↓ the expression of p-PDGFRβ/PDGFRβ, p-AKT/AKT, p-ERK/ERK, and p-p38/p38 proteins
-
inhibited PDGFRβ signaling pathway, HSC activation improved CCl4-induced liver fibrosis and inflammation in vivo
-
↓ ALT, AST
-
↓ α-SMA and Col-1
-
↓ the expression of p-AKT, p-ERK, and p-p38 proteins
-
↓ the mRNA expression levels of inflammatory factors IL-1β, IL-6, TNF-α, TGF-β, and COX-2
[34]
JS1 and LX2 cells
-
inhibited autophagy of HSCs induced by TGF-β1
-
inhibited the protein expression of LC3B II
-
induced the expression of C-Caspase 3
-
inhibited activation of JS1 through repressing autophagy of JS1 induced by TGF-β1
-
↓ LC3B II, Atg5, α-SMA, and Col-I protein expressions
-
inhibited activation and autophagy of HSCs by down-regulating the ERK, p38, and JNK pathways
-
↓ p-ERK, p-JNK, and p-p38 MAPK protein expressions
[35]
Sprague-Dawley rats
-
inhibited the CCl4-induced histopathological deterioration of the liver
-
↓ α-SMA
-
↓ liver damage caused by CCl4
-
↓ serum ALT, AST, TBIL
-
↑ ALB
-
↓ TGF-β1
-
inhibited the CCl4-induced activation of the Hh signaling pathway
-
↓ Shh, Ptch1, Smo, Gli1
[36]
LX-2 and T6 cells
BALB/c mice
-
attenuated HSC activation
-
↓ the mRNA levels of α-SMA and Col-1 in LX-2 and T6 cells
-
↓ the expression of LncRNA-ROR in vitro
-
inhibited HSC proliferation via LncRNA-ROR
-
inhibited HSC activation via LncRNA-ROR-mediated NF-κB signaling
-
attenuated primary HSC activation and down-regulated LncRNA-ROR mRNA expression
-
↓ α-SMA
-
↓ the expression of LncRNA-ROR via miR-6499-3p
-
ameliorated liver function, attenuated fibrosis severity, inhibited HSC activation, and regulated LncRNA-ROR and NF-κB signaling in CCl4-induced experimental mice
-
↓ serum ALT, AST, TBIL
-
↓ α-SMA and Col-1
-
↓ the phosphorylation of NF-κB p65
-
↓ the phosphorylation levels of NF-κB p65, IκBα, and IKKα
-
↓ IL-1β, IL-6, TNF-α, TGF-β1, COX-2
-
inhibited the mRNA level of α-SMA and LncRNA-ROR in liver tissues
[37]
LX-2 cells
-
promotes FGF19 secretion by LX-2 cells
-
inhibited LPS-induced HSC proliferation and activation
-
↓ α-SMA and Col1A1
-
↓ polymerization of actin F filaments in LX-2 cells
-
restored LPS-induced decrease in FGF19 and FGFR4 expression levels
[38]
HSC-T6 and LX-2 cells
-
ameliorated histopathological characteristics and hepatic fibrosis markers in mice
-
↓ α-SMA, Col-1, TGF-β1
-
inhibited activation of MAPK and P-Smad2/3L and P-Smad2C
-
↑ phosphorylation of P-Smad3C
-
modulated MAPK pathway activation and Smad2/3 phosphorylation in TGF-β1-stimulated HSCs
-
↓ P-ERK1/2, P-JNK1/2, P-p38, P-Smad2C, P-Smad2L, P-Smad3C, and P-Smad3L
-
↑ P-Smad3C
-
inhibited the expression of PAI in TGF-β1-stimulated HSCs
[39]
Sinapic acidSprague-Dawley rats
-
prevented DMN-induced loss of body weight
-
↓ AST, ALT
-
↓ hepatic HYP content
-
↓ MDA
-
↓ TGF-β1, Col-1, α-SMA
-
↓ NF-κB p65
[40]
Hydroxybenzoic acidsGallic acid and dodecyl gallateWistar albino rats
-
prevented the increase in relative liver weight and levels of triglycerides
-
restored serum hepatic enzyme activities
-
↓ TBIL, ALT, AST, γ-GT
-
↓ TBARS levels
-
↑ GSH
-
↓ lipid peroxidation levels
-
↑ catalase, GPx, GR, GST
-
improved histopathologic alterations
-
↑ the expression of p53 gene
[41]
Protocatechuic acidHSC-T6 cells
C57BL/6 mice
-
regulated cell viability in TNF-α-induced HSC-T6 cells via regulation on TGF-β signaling pathway
-
↓ TGF-β, p-Smad2, p-ERK, and c-Jun
-
attenuated the alteration of phenotype associated with TAA-induced liver damage and fibrosis in mice
-
↓ collagen
-
attenuated TAA-induced liver damage and fibrosis in mice
-
played a protective role in liver fibrosis through regulation of the TGF-β signaling pathway
-
↓ the protein expression of p-Smad2, p-ERK, and c-Jun
-
inhibited the mRNA level of IL-6 and TNF-α in TAA-induced mice
[42]
Vanillic acidSprague-Dawley rats
HSC-T6 cells
-
attenuated CCl4-induced liver fibrosis
-
ameliorated adipose degeneration of hepatocytes
-
↓ infiltration of inflammatory cells
-
inhibited the MIF/CD74 signaling pathway in vivo
-
↓ the mRNA and protein levels of MIF and CD74
-
suppressed autophagy and activity of HSCs in vivo
-
↓ α-SMA and LC3B
-
inhibited the MIF/CD74 signaling pathway and autophagy of HSCs in vitro
-
↓ MIF, CD74, α-SMA, LC3B, and Col-1
-
suppressed the proliferation and the migration of HSCs
[43]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; Akt, protein kinase B; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AMPK, adenosine monophosphate-activated protein kinase; AST, aspartate aminotransferase; Atg5, autophagy-related gene 5; BAMBI, “bone morphogenetic protein” activin membrane-bound inhibitor; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma-2; CAT, catalase; Ccl2, chemokine (C-C motif) ligand 2; CCl4, carbon tetrachloride; CIV, type IV collagen; Col-1, collagen 1; Col1α1, collagen type 1 alpha 1; Col1α2, collagen type 1 alpha 2; Col3α1, collagen type 3 alpha 1; Col4α1, collagen type 4 alpha 1; COX-2, cyclooxygenase-2; CTGF, connective tissue growth factor; DMN, dimethylnitrosamine; Ecm1, extracellular matrix protein 1; ERK, extracellular signal-regulated protein kinase; ERK1/2, extracellular signal-regulated kinases 1/2; FGF19, fibroblast growth factor; FGFR4, fibroblast growth factor receptor 4; GLB, globulin; Gli1, transcription factor glioma-associated oncogene homolog 1; GPx, glutathione peroxidase; GR, glutathione reductase; GSH, glutathione; GST, glutathione-S-transferase; HA, hyaluronic acid; HSCs, hepatic stellate cells; HYP, hydroxyproline; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; JNK, c-Jun N-terminal kinase; LC3B, microtubule-associated protein 2 light chain 3 type B; LN, laminin; LncRNA, long non-coding RNA; LOX, lysyloxidase; LPS, lipopolysaccharide; LX-2, human hepatic stellate cell line; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; MIF, macrophage migration inhibitory factor; MMP-9, matrix metalloproteinase 9; MPHs, mouse primary hepatocytes; mTOR, mammalian target of rapamycin; MYD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa B; NLRP3, NLR family pyrin domain containing 3; NOX2, nicotinamide adenine dinucleotide phosphate oxidase-2; Nrf2, nuclear factor erythroid 2-related factor 2; PAI, plasminogen activator inhibitor; p-Akt, phosphorylated protein kinase B; PCIII, procollagen type III; PDGFRβ, platelet-derived growth factor receptor beta; PDGF-βR, platelet-derived growth factor receptor beta; p-ERK, phosphorylated extracellular signal-regulated protein kinase; PI3K, phosphatidylinositol 3-kinase; PIIIP, procollagen III peptide; p-JNK, phosphorylated c-Jun N-terminal kinase; p-mTOR, phosphorylated mammalian target of rapamycin; p-Smad2, phosphorylated Smad2; P-Smad2/3L, phosphorylation of Smad2/3 at linker regions; P-Smad2C, phosphorylation of Smad2 at C-terminal linker regions; p-Smad3, phosphorylated Smad3; P-Smad3C, phosphorylation of Smad3 at C-terminal linker regions; Ptch1, membrane protein receptor protein patched homolog 1; PTP1B, protein tyrosine phosphatase 1B; ROR, regulator of reprogramming; ROS, reactive oxygen species; Shh, Sonic hedgehog protein; Smo, membrane protein receptor Smoothened; SOD, superoxide dismutase; TAA, thioacetamide; TBIL, total bilirubin; TGF-β1, transforming growth factor beta 1; TIMP-1, tissue inhibitor of metalloproteinases 1; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor alpha; α-SMA, alpha smooth muscle actin; γ-GT, γ-glutamyl transpeptidase.

2.2. Flavonoids

Flavonoids are a class of plant pigments increasingly utilized in drug development and nutraceutical applications [44]. These natural phenolic compounds feature a phenyl benzo (γ) pyrone-derived structure, comprising two benzene rings (A and B) connected by a pyrane ring (C) [45].
The effectiveness of orally administered flavonoids is limited due to their low dissolution rate, partial degradation in the acidic gastric environment, reduced permeability, and extensive first-pass metabolism before reaching systemic circulation [46].
Flavonoids are categorized into different subclasses based on the specific carbon atom of ring C to which ring B is attached and the degree of unsaturation and oxidation of ring C. These subclasses include flavan-3-ols (also known as flavanols or catechins), flavonols, flavones, flavanones, isoflavones, anthocyanidins, and chalcones [47,48].

2.2.1. Flavanols

Flavanols (IUPAC name: 3-hydroxy-2-phenylchromen-4-one) are a subclass of flavonoids and serve as secondary metabolites in plants [49]. Their chemical structure includes a hydroxyl group (-OH) on the third carbon atom (C3) and a carbonyl group (C=O) on the fourth carbon atom of the central heterocyclic ring [50].
Table 2 summarizes the pharmacological effects of several flavanol compounds on liver fibrosis, highlighting their ability to reduce fibrosis, oxidative stress, inflammation, and hepatic stellate cell activation. Each compound—epigallocatechin-3-gallate (EGCG), dihydromyricetin, hesperetin and its derivatives, hesperidin, liquiritigenin, naringenin, and naringin—demonstrates protective effects against liver damage through various mechanisms, including the modulation of signaling pathways like TGF-β1/Smad, PI3K/Akt, and cGAS-STING, ultimately contributing to the attenuation of liver fibrosis and improvement in liver function.

2.2.2. Flavonols

Flavonols are bioavailable compounds with multiple therapeutic benefits, such as hepatoprotective activity, free radical scavenging, cardioprotective, antiviral, antibacterial, and antineoplastic properties [73]. Flavonols contain a central structure of 3-hydroxyflavones, also known as 3-hydroxy-2-phenylchromen-4-one [74,75]. Flavonols are distinguished from other groups of flavonoids by the hydroxylation of one of the benzene rings. Each flavonol presents a distinct pattern of hydroxylation of the benzene ring [76,77]. The free forms of flavonols are called aglycones. The latter have a common structure of a 3-hydroxyflavone backbone and are distinguished by the position of the hydroxyl groups. The number of hydroxyl groups significantly contributes to the bioactivity of these compounds [78,79].
Table 3 summarizes the pharmacological effects of various flavonols in reducing liver fibrosis. These compounds—fisetin, galangin, isorhamnetin, kaempferol, dihydrokaempferol, morin, myricetin, myricitrin, and quercetin—work primarily by inhibiting key fibrotic pathways, including PI3K/Akt, Wnt/β-catenin, TGF-β1-Smad, and ERK1/2 signaling pathways, by reducing inflammation and oxidative stress, suppressing hepatic stellate cell activation, and enhancing antioxidant defenses.

2.2.3. Flavones

Flavones constitute another subclass of flavonoids. Their core structure includes a double bond between the C2 and C3 positions and a ketone group at the C4 position on ring C. The molecular formula for flavones is in [20,105,106]. Typically, flavones have a hydroxyl group at the fifth position of ring A, with additional hydroxylation potentially occurring at other positions, such as the seventh position of ring A or the 3′ and 4′ positions of ring B [107].
Table 4 provides a detailed summary of the pharmacological effects of flavones in combating liver fibrosis. These flavones act by reducing oxidative stress, inflammation, and collagen deposition, primarily through the inhibition of key fibrogenic signaling pathways like TGF-β/Smad and PI3K/Akt (baicalein, chrysin, isovitexin). They also suppress the activation and proliferation of hepatic stellate cells (HSCs), promote apoptosis and autophagy, and restore the balance of extracellular matrix (ECM) components (diosmin, isoorientin, ligustroflavone, isovitexin). Additionally, some flavones enhance antioxidant defenses and modulate pathways such as Nrf2 (alpinetin), cGAS-STING (oroxylin A), and Hippo/YAP and autophagy pathways (nobiletin), repressing the miR-17-5p/Wnt/β-catenin signaling (diosmin), suppressing TGF-β1-induced Smad and AKT signaling (luteolin), inhibiting the TLR2/TLR4 pathway (luteoloside), blocking the p38 MAPK and PDGF-Rβ signaling pathways (tricin), and contributing to the attenuation of liver fibrosis and the restoration of normal liver structure and function in various cell and animal models.

2.2.4. Flavanones

Flavanones are a subclass of flavonoids characterized by their distinct chemical structure. They have a flavan nucleus with a saturated three-carbon chain and a hydroxyl group attached to the second carbon (C2). This structure differentiates them from other flavonoids, such as flavones and flavonols, which have a double bond between C2 and C3. Flavanones are commonly found in citrus fruits such as oranges, lemons, and grapefruits, and they contribute to the characteristic bitterness of these fruits. Examples of flavanones include naringenin, hesperetin, and eriodictyol.
Flavanones have been studied for their potential health benefits, including antioxidant, anti-inflammatory, and anticancer properties. Their bioavailability and metabolism are subjects of ongoing research to better understand their therapeutic potential.
Table 5 highlights how flavanones mitigate liver fibrosis by inhibiting key fibrogenic pathways, via the SIRT1/TGF-β1/Smad3 signaling pathway, enhancing autophagy, and inhibiting the PI3K/AKT/mTOR pathway (ampelopsin), by reversing activated HSCs to their quiescent state, modulating autophagy, and inhibiting the mTOR signaling pathway (naringin), or by reducing oxidative stress and inflammation, enhancing antioxidant defenses via the Nrf2/HO-1 pathway, and inhibiting HSC activation through the TGF-β/Smad and PI3K/Akt signaling pathways (pinocembrin), leading to reduced fibrosis and improved liver function.

2.2.5. Isoflavones

Isoflavones are a subclass of flavonoids, which are naturally occurring compounds found in plants. Unlike other flavonoids, isoflavones have a unique structural feature where the B ring is attached to the C3 position of the central C ring, rather than the C2 position. This structural variation imparts distinct biological activities to isoflavones.
Isoflavones are predominantly found in legumes, with soybeans being the most significant source. Some of the well-known isoflavones include genistein, daidzein, and glycitein.
Table 6 highlights how isoflavones mitigate liver fibrosis by reducing oxidative stress, inflammation, and collagen deposition, while inhibiting key fibrogenic pathways such as JAK2/STAT3, TGF-β/Smad, and ERK1/2 (calycosin, genistein, puerarin, glabridin, soy isoflavone, tectorigenin). These compounds also promote the balance of matrix metalloproteinases (MMPs) and tissue inhibitors (TIMPs), enhance antioxidant defenses, and modulate HSC activation, contributing to the attenuation of fibrosis and improvement in liver function.

2.2.6. Anthocyanidins

Anthocyanidins are pigments responsible for the colors in plants, flowers, and fruits [48]. Anthocyanins from blueberries have been shown to regulate the epigenetic modifications of hepatic stellate cells (HSCs), thereby intervening in the treatment of hepatic fibrosis [154,155,156].
Table 7 highlights how anthocyanidins reduce liver fibrosis by inhibiting key fibrotic processes such as HSC activation, inflammation, oxidative stress, and collagen deposition, while enhancing antioxidant defenses, promoting apoptosis, and modulating signaling pathways such as Nrf2 and TGF-β/Smad, contributing to improved liver function and reduced fibrosis.

2.2.7. Chalcones

Chalcones are a subclass of flavonoids characterized by their unique open-chain structure. Unlike other flavonoids that have a closed ring system, chalcones consist of two aromatic rings (A and B) joined by a three-carbon α,β-unsaturated carbonyl system.
Chalcones are found in various plant species and are known for their bright yellow pigments. They serve as precursors in the biosynthesis of other flavonoids and isoflavonoids through the chalcone isomerase-catalyzed cyclization.
Table 8 demonstrates that chalcones effectively combat liver fibrosis by targeting several critical mechanisms. These compounds inhibit the activation of hepatic stellate cells (HSCs), reduce oxidative stress, and suppress inflammatory responses. Additionally, chalcones promote apoptosis in fibrotic cells, decrease collagen deposition, and modulate key signaling pathways such as Nrf2/HO-1, NF-κB, and metabolic processes like glycolysis. Together, these actions lead to significant reductions in fibrosis and improvements in liver function across various experimental models.

2.3. Stilbenes

Stilbenes are organic compounds characterized by a 1,2-diphenylethylene structure, commonly found in plants such as grapes, berries, and peanuts. Known for their potent antioxidant properties, these phytochemicals have been extensively researched for their potential health benefits. Resveratrol, a notable stilbene, offers liver protection against damage from chemicals, cholestasis, and alcohol; improves glucose metabolism and lipid profiles; and reduces liver fibrosis and steatosis [173]. It regulates fibrogenesis by reducing portal pressure, inhibiting hepatic stellate cell activation, and enhancing endothelial function, as well as modulating key signaling pathways like NF-κB and PI3K/Akt [174,175], and inducing autophagy via the microRNA-20a/PTEN/PI3K/AKT axis [176].
In liver fibrosis models, such as those induced by dimethylnitrosamine (DMN), resveratrol reduces inflammatory cell infiltration and fibrosis, lowering MDA levels, increasing GPx and SOD levels, and inhibiting inflammatory mediators like NO, TNF-α, and IL-1β [177,178,179]. Additionally, resveratrol alleviates mercury-induced liver fibrosis by activating the Sirt1/PGC-1α pathway and regulating the microbiota–gut–liver axis, enhancing the abundance of Bifidobacterium [180]. In obstructive jaundice, it protects liver function by modulating lipid metabolism, reducing oxidative stress, and down-regulating targets like mTOR and CYP enzymes [181]. Piceatannol, a resveratrol analog, effectively protects against CCl4-induced liver fibrosis in mice by improving liver function, reducing collagen deposition, and suppressing fibrosis markers via the TGF-β/Smad pathway. It also alleviates oxidative damage, highlighting its potential as a preventive agent for liver fibrosis [182].
Pterostilbene, found in grapes and berries, inhibits DMN-induced liver fibrosis in rats by improving liver function, reducing fibrotic changes, and decreasing hepatic stellate cell activation. It lowers serum ALT and AST levels, improves histopathology, and reduces fibrosis markers like α-SMA, TGF-β1, and MMP2, likely through the inhibition of the TGF-β1/Smad signaling pathway [183]. In another study, pterostilbene with a superior pharmacokinetic profile showed stronger anti-fibrotic effects than hydroxylated stilbenes in a CCl4-induced rat liver fibrosis model. It significantly reduced fibrosis markers and down-regulated key signaling pathways, demonstrating more potent protective activity.
Another stilbene, mulberroside A, reduces CCl4-induced liver fibrosis in mice by inhibiting the pro-inflammatory response and cytokine expression, providing significant liver protection without directly affecting hepatic stellate cell proliferation [184].

3. Polyphenol-Based Drug Delivery Systems and Hepatic Fibrosis

Nanoparticulate drug delivery systems significantly enhance the pharmacokinetics of drugs, including their absorption, metabolism, and excretion [185,186,187]. Compared to traditional formulations, nanoencapsulation offers several advantages, such as improved solubility and bioavailability, targeted drug delivery, consistent drug release, reduced dosage, and fewer side effects [188].
Nanoparticles (NPs) play a crucial role in improving the efficiency of co-delivery methods due to their ability to easily cross cell membranes because of their small size, enhance drug kinetics, and escape lysosomal degradation following endocytosis [189,190,191]. The main challenge of co-delivery systems is using carriers to simultaneously transport drugs with different properties [192]. Various modified carriers, such as liposomes, micelles, and polymeric NPs, have been employed to enhance the efficiency of co-delivery systems [193].
Polymeric nanoparticles, solid lipid nanoparticles, polymeric micelles, dendrimers, liposomes, nanocapsules, nanogels, nano-emulsions, and carbon nanotubes are examples of novel drug delivery systems (NDDSs). These systems, composed of biocompatible and biodegradable materials, offer numerous advantages over conventional dosage forms, such as controlled drug release, improved stability, and reduced adverse effects [194,195].
Nanocarriers ensure site-specific delivery of therapeutics, thereby improving bioavailability, stability, solubility, controlled release of active ingredients, and prolonged drug action [196,197]. Additionally, nanocarriers protect drugs from metabolic degradation [198]. Examples of nanocarriers include nanocapsules, nanospheres, nano-emulsions, and nano-sized vesicular carriers [196].
Polymeric NPs are effective carriers for the oral administration of flavonoids. They enhance physicochemical stability, increase solubilization and bioavailability by improving absorption at the enterocyte level, and maintain therapeutic levels in blood and plasma with a significant increase in mean residence time [199,200]. Polymeric NPs protect against degradation, provide controlled release of therapeutic agents, and enhance specific transport [201].
Poly(D,L-lactic-co-glycolic acid) (PLGA) is a biopolymer used in the preparation of NPs for various therapeutic applications. It protects compounds from degradation and offers sustained drug release [202,203]. The US Food and Drug Administration (FDA) has approved PLGA due to its properties, including biocompatibility, biodistribution, and biodegradability [204,205]. PLGA has been used in numerous drug delivery systems, both targeted and non-targeted [206,207].
PLGA nanoparticles are taken up by endocytosis, releasing the drug in intracellular locations, thereby improving therapeutic action and reducing side effects [207,208]. The rapid absorption of PLGA nanoparticles by the reticuloendothelial system can be significantly reduced by modifying their surface with poly(ethylene glycol) (PEG). This modification extends the circulation time of nanosystems in the blood, allows targeting to tissues, and prevents opsonization [209,210]. These nanoparticles with hydrophilic surfaces have shown improved permeability, presumably due to the prolonged residence time of the carrier in the blood [211,212]. The surface modification of nanoparticles with polyethylene glycol (PEG) extends circulation time, reduces non-specific interactions, and favors accumulation in tumors due to increased permeability and retention [212,213].
Polycaprolactone (PCL), a biodegradable polymer, is suitable for controlled drug release due to its high permeability to many drugs and lack of toxicity [214]. PCL can also form blends with other polymers [215].
Table 9 highlights the effectiveness of polyphenol-based drug delivery systems in combating liver fibrosis by leveraging advanced formulations that enhance the bioavailability, targeting, and therapeutic efficacy of active compounds. These formulations, including nanoparticles, nanocomplexes, liposomes, and exosomes, significantly improve the delivery of polyphenols like galangin, quercetin, chrysin, luteolin, hesperidin, naringenin, silibinin, silymarin, and curcumin. The enhanced delivery systems allow these compounds to more effectively reduce oxidative stress, inflammation, and fibrotic markers such as ALT, AST, TGF-β1, and collagen deposition. Additionally, these systems improve the modulation of key signaling pathways like TGF-β/Smad, NF-κB, and Nrf2, leading to a better preservation of liver architecture and function in various experimental models of liver fibrosis.

4. Conclusions and Perspectives

The integration of plant-based therapeutics with advanced nanoparticulate drug delivery systems offers a promising approach to treating hepatic fibrosis. Polyphenols, phenolic acids, and flavonoids have shown significant potential in mitigating hepatic fibrosis by targeting multiple signaling pathways involved in the disease’s progression. These natural compounds exert anti-fibrotic effects through mechanisms such as the inhibition of hepatic stellate cell activation, reduction in extracellular matrix deposition, and modulation of inflammatory responses. Overall, the signaling pathways modulated by these natural compounds in the context of hepatic fibrosis, which lead to the inhibition of hepatic fibrosis, reduction in extracellular matrix (ECM) deposition, and overall liver protection, are the TGF-β/Smad pathway, AMPK/mTOR pathway, Wnt/β-catenin pathway, NF-κB pathway, PI3K/AKT/mTOR pathway, and hedgehog pathway (Figure 1).
However, the modulation of immune cells and immune responses mediated by polyphenols (Figure 2) are key events in the resolution of liver fibrosis: (a). the modulation of macrophage polarization from the pro-fibrotic M2 phenotype to the anti-fibrotic M1 phenotype, which subsequently produces pro-inflammatory cytokines that help clear apoptotic cells and degrade the extracellular matrix; (b). inhibition of hepatic stellate cell activation, through reducing the secretion of pro-fibrotic cytokines like TGF-β from immune cells; (c). regulation of T-cell responses, particularly influencing the balance between regulatory T-cells (Tregs) and effector T-cells; (d). reduction in oxidative stress and inflammation, by scavenging reactive oxygen species (ROS) and inhibiting the NF-κB signaling pathway, which are key contributors to liver fibrosis; (e). enhancement in autophagy in immune cells and hepatic stellate cells, a process that facilitates the clearance of damaged cells and reduces the deposition of extracellular matrix proteins; (f). modulation of the Kupffer cell activity, by reducing their pro-inflammatory cytokine production and promoting their role in clearing fibrotic tissue; (g). enhancing the cytotoxic activity of NK cells against activated hepatic stellate cells, leading to their apoptosis and reducing fibrogenesis.
Nanoparticulate drug delivery systems further enhance the therapeutic efficacy of these bioactive compounds by improving their solubility, bioavailability, and pharmacokinetics. Polymeric nanoparticles, solid lipid nanoparticles, polymeric micelles, and other novel drug delivery systems have demonstrated the ability to deliver therapeutic agents with high precision and reduced side effects.
Future research should focus on translating these findings into clinical applications. Rigorous clinical trials are needed to validate the safety and efficacy of plant-based therapeutics and nanoparticulate drug delivery systems in human subjects.
In conclusion, the convergence of plant-based therapeutics and innovative drug delivery systems represents a promising frontier in the fight against hepatic fibrosis. Continued research and development in this area hold the potential to significantly improve the management and treatment of this debilitating condition.

Author Contributions

Conceptualization, A.C. and A.H.; methodology, S.A.; investigation, A.C., F.F., S.D. and M.P.; resources, A.C.; writing—original draft preparation, A.C., F.F., A.H., S.A., S.D. and M.P.; writing—review and editing, A.C., A.H., S.A., S.D. and M.P.; supervision, F.F.; project administration, A.C.; funding acquisition, A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Romanian Ministry of Research and Innovation, PN-III-P1-1.1-PD-2021-0327 (PD 94/2022).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Shan, L.; Wang, F.; Zhai, D.; Meng, X.; Liu, J.; Lv, X. New Drugs for Hepatic Fibrosis. Front. Pharmacol. 2022, 13, 874408. [Google Scholar] [CrossRef]
  2. Chang, J.; Huang, C.; Li, S.; Jiang, X.; Chang, H.; Li, M. Research Progress Regarding the Effect and Mechanism of Dietary Polyphenols in Liver Fibrosis. Molecules 2023, 29, 127. [Google Scholar] [CrossRef] [PubMed]
  3. Tan, Z.; Sun, H.; Xue, T.; Gan, C.; Liu, H.; Xie, Y.; Yao, Y.; Ye, T. Liver Fibrosis: Therapeutic Targets and Advances in Drug Therapy. Front. Cell Dev. Biol. 2021, 9, 730176. [Google Scholar] [CrossRef] [PubMed]
  4. Altamirano-Barrera, A.; Barranco-Fragoso, B.; Méndez-Sánchez, N. Management Strategies for Liver Fibrosis. Ann. Hepatol. 2017, 16, 48–56. [Google Scholar] [CrossRef] [PubMed]
  5. Duval, F.; Moreno-Cuevas, J.E.; González-Garza, M.T.; Rodríguez-Montalvo, C.; Cruz-Vega, D.E. Protective Mechanisms of Medicinal Plants Targeting Hepatic Stellate Cell Activation and Extracellular Matrix Deposition in Liver Fibrosis. Chin. Med. 2014, 9, 27. [Google Scholar] [CrossRef] [PubMed]
  6. Latief, U.; Ahmad, R. Herbal Remedies for Liver Fibrosis: A Review on the Mode of Action of Fifty Herbs. J. Tradit. Complement. Med. 2018, 8, 352–360. [Google Scholar] [CrossRef]
  7. Zhao, Q.; Luan, X.; Zheng, M.; Tian, X.-H.; Zhao, J.; Zhang, W.-D.; Ma, B.-L. Synergistic Mechanisms of Constituents in Herbal Extracts during Intestinal Absorption: Focus on Natural Occurring Nanoparticles. Pharmaceutics 2020, 12, 128. [Google Scholar] [CrossRef]
  8. Hermenean, A.; Smeu, C.; Gharbia, S.; Krizbai, I.A.; Ardelean, A. Plant-Derived Biomolecules and Drug Delivery Systems in the Treatment of Liver and Kidney Diseases. Curr. Pharm. Des. 2016, 22, 5415–5441. [Google Scholar] [CrossRef]
  9. Del Prete, A.; Scalera, A.; Iadevaia, M.D.; Miranda, A.; Zulli, C.; Gaeta, L.; Tuccillo, C.; Federico, A.; Loguercio, C. Herbal Products: Benefits, Limits, and Applications in Chronic Liver Disease. Evid.-Based Complement. Altern. Med. ECAM 2012, 2012, 837939. [Google Scholar] [CrossRef]
  10. Singh, S.; Sharma, N.; Shukla, S.; Behl, T.; Gupta, S.; Anwer, M.K.; Vargas-De-La-Cruz, C.; Bungau, S.G.; Brisc, C. Understanding the Potential Role of Nanotechnology in Liver Fibrosis: A Paradigm in Therapeutics. Molecules 2023, 28, 2811. [Google Scholar] [CrossRef]
  11. El-Tantawy, W.H.; Temraz, A. Anti-Fibrotic Activity of Natural Products, Herbal Extracts and Nutritional Components for Prevention of Liver Fibrosis: Review. Arch. Physiol. Biochem. 2022, 128, 382–393. [Google Scholar] [CrossRef]
  12. Rezaeiamiri, E.; Bahramsoltani, R.; Rahimi, R. Plant-Derived Natural Agents as Dietary Supplements for the Regulation of Glycosylated Hemoglobin: A Review of Clinical Trials. Clin. Nutr. 2020, 39, 331–342. [Google Scholar] [CrossRef]
  13. Kesarwani, K.; Gupta, R. Bioavailability Enhancers of Herbal Origin: An Overview. Asian Pac. J. Trop. Biomed. 2013, 3, 253–266. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, D.; Zhang, Y.; Sun, B. The Molecular Mechanisms of Liver Fibrosis and Its Potential Therapy in Application. Int. J. Mol. Sci. 2022, 23, 12572. [Google Scholar] [CrossRef]
  15. Laddomada, B.; Blanco, A.; Mita, G.; D’Amico, L.; Singh, R.P.; Ammar, K.; Crossa, J.; Guzmán, C. Drought and Heat Stress Impacts on Phenolic Acids Accumulation in Durum Wheat Cultivars. Foods 2021, 10, 2142. [Google Scholar] [CrossRef] [PubMed]
  16. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant Polyphenols: Chemical Properties, Biological Activities, and Synthesis. Angew. Chem. Int. Ed. Engl. 2011, 50, 586–621. [Google Scholar] [CrossRef]
  17. Scarano, A.; Chieppa, M.; Santino, A. Plant Polyphenols-Biofortified Foods as a Novel Tool for the Prevention of Human Gut Diseases. Antioxidants 2020, 9, 1225. [Google Scholar] [CrossRef]
  18. Scarano, A.; Chieppa, M.; Santino, A. Looking at Flavonoid Biodiversity in Horticultural Crops: A Colored Mine with Nutritional Benefits. Plants 2018, 7, 98. [Google Scholar] [CrossRef] [PubMed]
  19. Tsao, R. Chemistry and Biochemistry of Dietary Polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef]
  20. Cháirez-Ramírez, M.H.; de la Cruz-López, K.G.; García-Carrancá, A. Polyphenols as Antitumor Agents Targeting Key Players in Cancer-Driving Signaling Pathways. Front. Pharmacol. 2021, 12, 710304. [Google Scholar] [CrossRef]
  21. Al Mamari, H. Phenolic Compounds: Classification, Chemistry, and Updated Techniques of Analysis and Synthesis. In Phenolic Compounds—Chemistry, Synthesis, Diversity, Non-Conventional Industrial, Pharmaceutical and Therapeutic Applications; IntechOpen: London, UK, 2022; Volume 26. [Google Scholar] [CrossRef]
  22. El-Seedi, H.R.; El-Said, A.M.A.; Khalifa, S.A.M.; Göransson, U.; Bohlin, L.; Borg-Karlson, A.-K.; Verpoorte, R. Biosynthesis, Natural Sources, Dietary Intake, Pharmacokinetic Properties, and Biological Activities of Hydroxycinnamic Acids. J. Agric. Food Chem. 2012, 60, 10877–10895. [Google Scholar] [CrossRef] [PubMed]
  23. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food Sources and Bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
  24. Rocha, L.D.; Monteiro, M.C.; Teodoro, A.J. Anticancer Properties of Hydroxycinnamic Acids—A Review. Cancer Clin. Oncol. 2012, 1, 109. [Google Scholar] [CrossRef]
  25. Yang, F.; Luo, L.; Zhu, Z.-D.; Zhou, X.; Wang, Y.; Xue, J.; Zhang, J.; Cai, X.; Chen, Z.-L.; Ma, Q.; et al. Chlorogenic Acid Inhibits Liver Fibrosis by Blocking the miR-21-Regulated TGF-Β1/Smad7 Signaling Pathway in Vitro and in Vivo. Front. Pharmacol. 2017, 8, 929. [Google Scholar] [CrossRef]
  26. Shi, H.; Dong, L.; Jiang, J.; Zhao, J.; Zhao, G.; Dang, X.; Lu, X.; Jia, M. Chlorogenic Acid Reduces Liver Inflammation and Fibrosis through Inhibition of Toll-like Receptor 4 Signaling Pathway. Toxicology 2013, 303, 107–114. [Google Scholar] [CrossRef] [PubMed]
  27. Wu, J.; Xue, X.; Fan, G.; Gu, Y.; Zhou, F.; Zheng, Q.; Liu, R.; Li, Y.; Ma, B.; Li, S.; et al. Ferulic Acid Ameliorates Hepatic Inflammation and Fibrotic Liver Injury by Inhibiting PTP1B Activity and Subsequent Promoting AMPK Phosphorylation. Front. Pharmacol. 2021, 12, 754976. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, X.; Huang, K.; Niu, Z.; Mei, D.; Zhang, B. Protective Effect of Isochlorogenic Acid B on Liver Fibrosis in Non-Alcoholic Steatohepatitis of Mice. Basic Clin. Pharmacol. Toxicol. 2019, 124, 144–153. [Google Scholar] [CrossRef] [PubMed]
  29. Truong, T.M.T.; Seo, S.H.; Chung, S.; Kang, I. Attenuation of Hepatic Fibrosis by P-Coumaric Acid via Modulation of NLRP3 Inflammasome Activation in C57BL/6 Mice. J. Nutr. Biochem. 2023, 112, 109204. [Google Scholar] [CrossRef]
  30. Li, G.-S.; Jiang, W.-L.; Tian, J.-W.; Qu, G.-W.; Zhu, H.-B.; Fu, F.-H. In Vitro and in Vivo Antifibrotic Effects of Rosmarinic Acid on Experimental Liver Fibrosis. Phytomedicine Int. J. Phytother. Phytopharm. 2010, 17, 282–288. [Google Scholar] [CrossRef]
  31. Wang, R.; Song, F.; Li, S.; Wu, B.; Gu, Y.; Yuan, Y. Salvianolic Acid A Attenuates CCl4-Induced Liver Fibrosis by Regulating the PI3K/AKT/mTOR, Bcl-2/Bax and Caspase-3/Cleaved Caspase-3 Signaling Pathways. Drug Des. Devel. Ther. 2019, 13, 1889–1900. [Google Scholar] [CrossRef]
  32. Fu, Y.; Zhou, X.; Wang, L.; Fan, W.; Gao, S.; Zhang, D.; Ling, Z.; Zhang, Y.; Ma, L.; Bai, F.; et al. Salvianolic Acid B Attenuates Liver Fibrosis by Targeting Ecm1 and Inhibiting Hepatocyte Ferroptosis. Redox Biol. 2024, 69, 103029. [Google Scholar] [CrossRef] [PubMed]
  33. Li, Z.-B.; Jiang, L.; Ni, J.-D.; Xu, Y.-H.; Liu, F.; Liu, W.-M.; Wang, S.-G.; Liu, Z.-Q.; Wang, C.-Y. Salvianolic Acid B Suppresses Hepatic Fibrosis by Inhibiting Ceramide Glucosyltransferase in Hepatic Stellate Cells. Acta Pharmacol. Sin. 2023, 44, 1191–1205. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, F.; Li, S.; Chen, P.; Gu, Y.; Wang, S.; Wang, L.; Chen, C.; Wang, R.; Yuan, Y. Salvianolic Acid B Inhibits Hepatic Stellate Cell Activation and Liver Fibrosis by Targeting PDGFRβ. Int. Immunopharmacol. 2023, 122, 110550. [Google Scholar] [CrossRef] [PubMed]
  35. Jiang, N.; Zhang, J.; Ping, J.; Xu, L. Salvianolic Acid B Inhibits Autophagy and Activation of Hepatic Stellate Cells Induced by TGF-Β1 by Downregulating the MAPK Pathway. Front. Pharmacol. 2022, 13, 938856. [Google Scholar] [CrossRef]
  36. Tao, S.; Duan, R.; Xu, T.; Hong, J.; Gu, W.; Lin, A.; Lian, L.; Huang, H.; Lu, J.; Li, T. Salvianolic Acid B Inhibits the Progression of Liver Fibrosis in Rats via Modulation of the Hedgehog Signaling Pathway. Exp. Ther. Med. 2022, 23, 116. [Google Scholar] [CrossRef]
  37. Wang, R.; Li, S.; Chen, P.; Yue, X.; Wang, S.; Gu, Y.; Yuan, Y. Salvianolic Acid B Suppresses Hepatic Stellate Cell Activation and Liver Fibrosis by Inhibiting the NF-κB Signaling Pathway via miR-6499-3p/LncRNA-ROR. Phytomedicine Int. J. Phytother. Phytopharm. 2022, 107, 154435. [Google Scholar] [CrossRef]
  38. Tian, S.; Chen, M.; Wang, B.; Han, Y.; Shang, H.; Chen, J. Salvianolic Acid B Blocks Hepatic Stellate Cell Activation via FGF19/FGFR4 Signaling. Ann. Hepatol. 2021, 20, 100259. [Google Scholar] [CrossRef]
  39. Wu, C.; Chen, W.; Ding, H.; Li, D.; Wen, G.; Zhang, C.; Lu, W.; Chen, M.; Yang, Y. Salvianolic Acid B Exerts Anti-Liver Fibrosis Effects via Inhibition of MAPK-Mediated Phospho-Smad2/3 at Linker Regions in Vivo and in Vitro. Life Sci. 2019, 239, 116881. [Google Scholar] [CrossRef]
  40. Shin, D.-S.; Kim, K.W.; Chung, H.Y.; Yoon, S.; Moon, J.-O. Effect of Sinapic Acid against Dimethylnitrosamine-Induced Hepatic Fibrosis in Rats. Arch. Pharm. Res. 2013, 36, 608–618. [Google Scholar] [CrossRef]
  41. Perazzoli, M.R.A.; Perondi, C.K.; Baratto, C.M.; Winter, E.; Creczynski-Pasa, T.B.; Locatelli, C. Gallic Acid and Dodecyl Gallate Prevents Carbon Tetrachloride-Induced Acute and Chronic Hepatotoxicity by Enhancing Hepatic Antioxidant Status and Increasing P53 Expression. Biol. Pharm. Bull. 2017, 40, 425–434. [Google Scholar] [CrossRef]
  42. Cui, B.; Yang, Z.; Wang, S.; Guo, M.; Li, Q.; Zhang, Q.; Bi, X. The Protective Role of Protocatechuic Acid against Chemically Induced Liver Fibrosis in Vitro and in Vivo. Die Pharm. 2021, 76, 232–238. [Google Scholar] [CrossRef]
  43. Qin, L.; Tan, J.; Lv, X.; Zhang, J. Vanillic Acid Alleviates Liver Fibrosis through Inhibiting Autophagy in Hepatic Stellate Cells via the MIF/CD74 Signaling Pathway. Biomed. Pharmacother. Biomed. Pharmacother. 2023, 168, 115673. [Google Scholar] [CrossRef] [PubMed]
  44. Beecher, G.R. Overview of Dietary Flavonoids: Nomenclature, Occurrence and Intake. J. Nutr. 2003, 133, 3248S–3254S. [Google Scholar] [CrossRef] [PubMed]
  45. Cook, N.C.; Samman, S. Flavonoids—Chemistry, Metabolism, Cardioprotective Effects, and Dietary Sources. J. Nutr. Biochem. 1996, 7, 66–76. [Google Scholar] [CrossRef]
  46. Ratnam, D.V.; Ankola, D.D.; Bhardwaj, V.; Sahana, D.K.; Kumar, M.N.V.R. Role of Antioxidants in Prophylaxis and Therapy: A Pharmaceutical Perspective. J. Control. Release Off. J. Control. Release Soc. 2006, 113, 189–207. [Google Scholar] [CrossRef]
  47. Middleton, E.; Kandaswami, C.; Theoharides, T.C. The Effects of Plant Flavonoids on Mammalian Cells: Implications for Inflammation, Heart Disease, and Cancer. Pharmacol. Rev. 2000, 52, 673–751. [Google Scholar]
  48. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An Overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef]
  49. Kubina, R.; Krzykawski, K.; Kabała-Dzik, A.; Wojtyczka, R.D.; Chodurek, E.; Dziedzic, A. Fisetin, a Potent Anticancer Flavonol Exhibiting Cytotoxic Activity against Neoplastic Malignant Cells and Cancerous Conditions: A Scoping, Comprehensive Review. Nutrients 2022, 14, 2604. [Google Scholar] [CrossRef]
  50. Forbes, A.M.; Meier, G.P.; Haendiges, S.; Taylor, L.P. Structure-Activity Relationship Studies of Flavonol Analogues on Pollen Germination. J. Agric. Food Chem. 2014, 62, 2175–2181. [Google Scholar] [CrossRef]
  51. George, J.; Tsuchishima, M.; Tsutsumi, M. Epigallocatechin-3-Gallate Inhibits Osteopontin Expression and Prevents Experimentally Induced Hepatic Fibrosis. Biomed. Pharmacother. Biomed. Pharmacother. 2022, 151, 113111. [Google Scholar] [CrossRef]
  52. Arffa, M.L.; Zapf, M.A.; Kothari, A.N.; Chang, V.; Gupta, G.N.; Ding, X.; Al-Gayyar, M.M.; Syn, W.; Elsherbiny, N.M.; Kuo, P.C.; et al. Epigallocatechin-3-Gallate Upregulates miR-221 to Inhibit Osteopontin-Dependent Hepatic Fibrosis. PLoS ONE 2016, 11, e0167435. [Google Scholar] [CrossRef]
  53. Yu, D.; Zhang, C.; Zhao, S.; Zhang, S.; Zhang, H.; Cai, S.; Shao, R.; He, H. The Anti-Fibrotic Effects of Epigallocatechin-3-Gallate in Bile Duct-Ligated Cholestatic Rats and Human Hepatic Stellate LX-2 Cells Are Mediated by the PI3K/Akt/Smad Pathway. Acta Pharmacol. Sin. 2015, 36, 473–482. [Google Scholar] [CrossRef] [PubMed]
  54. Tipoe, G.L.; Leung, T.M.; Liong, E.C.; Lau, T.Y.H.; Fung, M.L.; Nanji, A.A. Epigallocatechin-3-Gallate (EGCG) Reduces Liver Inflammation, Oxidative Stress and Fibrosis in Carbon Tetrachloride (CCl4)-Induced Liver Injury in Mice. Toxicology 2010, 273, 45–52. [Google Scholar] [CrossRef] [PubMed]
  55. Yasuda, Y.; Shimizu, M.; Sakai, H.; Iwasa, J.; Kubota, M.; Adachi, S.; Osawa, Y.; Tsurumi, H.; Hara, Y.; Moriwaki, H. (−)-Epigallocatechin Gallate Prevents Carbon Tetrachloride-Induced Rat Hepatic Fibrosis by Inhibiting the Expression of the PDGFRbeta and IGF-1R. Chem. Biol. Interact. 2009, 182, 159–164. [Google Scholar] [CrossRef] [PubMed]
  56. Zhen, M.-C.; Wang, Q.; Huang, X.-H.; Cao, L.-Q.; Chen, X.-L.; Sun, K.; Liu, Y.-J.; Li, W.; Zhang, L.-J. Green Tea Polyphenol Epigallocatechin-3-Gallate Inhibits Oxidative Damage and Preventive Effects on Carbon Tetrachloride-Induced Hepatic Fibrosis. J. Nutr. Biochem. 2007, 18, 795–805. [Google Scholar] [CrossRef] [PubMed]
  57. Zhao, Y.; Liu, X.; Ding, C.; Gu, Y.; Liu, W. Dihydromyricetin Reverses Thioacetamide-Induced Liver Fibrosis Through Inhibiting NF-κB-Mediated Inflammation and TGF-Β1-Regulated of PI3K/Akt Signaling Pathway. Front. Pharmacol. 2021, 12, 783886. [Google Scholar] [CrossRef]
  58. Zhou, X.; Yu, L.; Zhou, M.; Hou, P.; Yi, L.; Mi, M. Dihydromyricetin Ameliorates Liver Fibrosis via Inhibition of Hepatic Stellate Cells by Inducing Autophagy and Natural Killer Cell-Mediated Killing Effect. Nutr. Metab. 2021, 18, 64. [Google Scholar] [CrossRef]
  59. Kong, R.; Wang, N.; Luo, H.; Lu, J. Hesperetin Mitigates Bile Duct Ligation-Induced Liver Fibrosis by Inhibiting Extracellular Matrix and Cell Apoptosis via the TGF-Β1/Smad Pathway. Curr. Mol. Med. 2018, 18, 15–24. [Google Scholar] [CrossRef]
  60. Chen, X.; Li, X.-F.; Chen, Y.; Zhu, S.; Li, H.-D.; Chen, S.-Y.; Wang, J.-N.; Pan, X.-Y.; Bu, F.-T.; Huang, C.; et al. Hesperetin Derivative Attenuates CCl4-Induced Hepatic Fibrosis and Inflammation by Gli-1-Dependent Mechanisms. Int. Immunopharmacol. 2019, 76, 105838. [Google Scholar] [CrossRef]
  61. Zhu, S.; Chen, X.; Chen, S.-Y.; Wang, A.; Wu, S.; Wu, Y.-Y.; Cheng, M.; Xu, J.-J.; Li, X.-F.; Huang, C.; et al. Hesperetin Derivative Decreases CCl4 -Induced Hepatic Fibrosis by Ptch1-Dependent Mechanisms. J. Biochem. Mol. Toxicol. 2022, 36, e23149. [Google Scholar] [CrossRef]
  62. Lin, X.; Kong, L.-N.; Huang, C.; Ma, T.-T.; Meng, X.-M.; He, Y.; Wang, Q.-Q.; Li, J. Hesperetin Derivative-7 Inhibits PDGF-BB-Induced Hepatic Stellate Cell Activation and Proliferation by Targeting Wnt/β-Catenin Pathway. Int. Immunopharmacol. 2015, 25, 311–320. [Google Scholar] [CrossRef] [PubMed]
  63. Li, W.-X.; Chen, X.; Yang, Y.; Huang, H.-M.; Li, H.; Huang, C.; Meng, X.-M.; Li, J. Hesperitin Derivative-11 Suppress Hepatic Stellate Cell Activation and Proliferation by Targeting PTEN/AKT Pathway. Toxicology 2017, 381, 75–86. [Google Scholar] [CrossRef] [PubMed]
  64. Li, J.-J.; Jiang, H.-C.; Wang, A.; Bu, F.-T.; Jia, P.-C.; Zhu, S.; Zhu, L.; Huang, C.; Li, J. Hesperetin Derivative-16 Attenuates CCl4-Induced Inflammation and Liver Fibrosis by Activating AMPK/SIRT3 Pathway. Eur. J. Pharmacol. 2022, 915, 174530. [Google Scholar] [CrossRef] [PubMed]
  65. Nasehi, Z.; Kheiripour, N.; Taheri, M.A.; Ardjmand, A.; Jozi, F.; Shahaboddin, M.E. Efficiency of Hesperidin against Liver Fibrosis Induced by Bile Duct Ligation in Rats. BioMed Res. Int. 2023, 2023, 5444301. [Google Scholar] [CrossRef]
  66. Pérez-Vargas, J.E.; Zarco, N.; Shibayama, M.; Segovia, J.; Tsutsumi, V.; Muriel, P. Hesperidin Prevents Liver Fibrosis in Rats by Decreasing the Expression of Nuclear Factor-κB, Transforming Growth Factor-β and Connective Tissue Growth Factor. Pharmacology 2014, 94, 80–89. [Google Scholar] [CrossRef] [PubMed]
  67. Geng, W.; Zhou, G.; Zhao, B.; Xiao, Q.; Li, C.; Fan, S.; Dong, P.; Zheng, J. Liquiritigenin Suppresses the Activation of Hepatic Stellate Cells via Targeting miR-181b/PTEN Axis. Phytomedicine Int. J. Phytother. Phytopharm. 2019, 66, 153108. [Google Scholar] [CrossRef]
  68. Lee, E.H.; Park, K.-I.; Kim, K.-Y.; Lee, J.-H.; Jang, E.J.; Ku, S.K.; Kim, S.C.; Suk, H.Y.; Park, J.Y.; Baek, S.Y.; et al. Liquiritigenin Inhibits Hepatic Fibrogenesis and TGF-Β1/Smad with Hippo/YAP Signal. Phytomed. Int. J. Phytother. Phytopharm. 2019, 62, 152780. [Google Scholar] [CrossRef]
  69. Chen, L.; Xia, S.; Wang, S.; Zhou, Y.; Wang, F.; Li, Z.; Li, Y.; Kong, D.; Zhang, Z.; Shao, J.; et al. Naringenin Is a Potential Immunomodulator for Inhibiting Liver Fibrosis by Inhibiting the cGAS-STING Pathway. J. Clin. Transl. Hepatol. 2023, 11, 26–37. [Google Scholar] [CrossRef]
  70. Hernández-Aquino, E.; Quezada-Ramírez, M.A.; Silva-Olivares, A.; Casas-Grajales, S.; Ramos-Tovar, E.; Flores-Beltrán, R.E.; Segovia, J.; Shibayama, M.; Muriel, P. Naringenin Attenuates the Progression of Liver Fibrosis via Inactivation of Hepatic Stellate Cells and Profibrogenic Pathways. Eur. J. Pharmacol. 2019, 865, 172730. [Google Scholar] [CrossRef]
  71. Hernández-Aquino, E.; Zarco, N.; Casas-Grajales, S.; Ramos-Tovar, E.; Flores-Beltrán, R.E.; Arauz, J.; Shibayama, M.; Favari, L.; Tsutsumi, V.; Segovia, J.; et al. Naringenin Prevents Experimental Liver Fibrosis by Blocking TGFβ-Smad3 and JNK-Smad3 Pathways. World J. Gastroenterol. 2017, 23, 4354–4368. [Google Scholar] [CrossRef]
  72. El-Mihi, K.A.; Kenawy, H.I.; El-Karef, A.; Elsherbiny, N.M.; Eissa, L.A. Naringin Attenuates Thioacetamide-Induced Liver Fibrosis in Rats through Modulation of the PI3K/Akt Pathway. Life Sci. 2017, 187, 50–57. [Google Scholar] [CrossRef]
  73. Gajender; Mazumder, A.; Sharma, A.; Azad, M.A.K. A Comprehensive Review of the Pharmacological Importance of Dietary Flavonoids as Hepatoprotective Agents. Evid.-Based Complement. Altern. Med. ECAM 2023, 2023, 4139117. [Google Scholar] [CrossRef]
  74. Spagnuolo, C.; Moccia, S.; Russo, G.L. Anti-Inflammatory Effects of Flavonoids in Neurodegenerative Disorders. Eur. J. Med. Chem. 2018, 153, 105–115. [Google Scholar] [CrossRef]
  75. Tahir, M.S.; Almezgagi, M.; Zhang, Y.; Bashir, A.; Abdullah, H.M.; Gamah, M.; Wang, X.; Zhu, Q.; Shen, X.; Ma, Q.; et al. Mechanistic New Insights of Flavonols on Neurodegenerative Diseases. Biomed. Pharmacother. 2021, 137, 111253. [Google Scholar] [CrossRef] [PubMed]
  76. Daryanavard, H.; Postiglione, A.E.; Mühlemann, J.K.; Muday, G.K. Flavonols Modulate Plant Development, Signaling, and Stress Responses. Curr. Opin. Plant Biol. 2023, 72, 102350. [Google Scholar] [CrossRef] [PubMed]
  77. Falcone Ferreyra, M.L.; Rius, S.P.; Casati, P. Flavonoids: Biosynthesis, Biological Functions, and Biotechnological Applications. Front. Plant Sci. 2012, 3, 222. [Google Scholar] [CrossRef]
  78. Popiolek-Kalisz, J.; Fornal, E. The Impact of Flavonols on Cardiovascular Risk. Nutrients 2022, 14, 1973. [Google Scholar] [CrossRef] [PubMed]
  79. Sharma, A.; Sharma, P.; Singh Tuli, H.; Sharma, A.K. Phytochemical and Pharmacological Properties of Flavonols. In eLS; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2018; pp. 1–12. ISBN 978-0-470-01590-2. [Google Scholar]
  80. El-Fadaly, A.A.; Afifi, N.A.; El-Eraky, W.; Salama, A.; Abdelhameed, M.F.; El-Rahman, S.S.A.; Ramadan, A. Fisetin Alleviates Thioacetamide-Induced Hepatic Fibrosis in Rats by Inhibiting Wnt/β-Catenin Signaling Pathway. Immunopharmacol. Immunotoxicol. 2022, 44, 355–366. [Google Scholar] [CrossRef] [PubMed]
  81. Xiong, Y.; Lu, H.; Xu, H. Galangin Reverses Hepatic Fibrosis by Inducing HSCs Apoptosis via the PI3K/Akt, Bax/Bcl-2, and Wnt/β-Catenin Pathway in LX-2 Cells. Biol. Pharm. Bull. 2020, 43, 1634–1642. [Google Scholar] [CrossRef]
  82. Wang, X.; Gong, G.; Yang, W.; Li, Y.; Jiang, M.; Li, L. Antifibrotic Activity of Galangin, a Novel Function Evaluated in Animal Liver Fibrosis Model. Environ. Toxicol. Pharmacol. 2013, 36, 288–295. [Google Scholar] [CrossRef]
  83. Rashidi, M.; Matour, E.; Beheshti Nasab, H.; Cheraghzadeh, M.; Shakerian, E. Isorhamnetin Exerts Antifibrotic Effects by Attenuating Platelet-Derived Growth Factor-BB-Induced HSC-T6 Cells Activation via Suppressing PI3K-AKT Signaling Pathway. Iran. Biomed. J. 2023, 27, 199–204. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, N.; Feng, J.; Lu, X.; Yao, Z.; Liu, Q.; Lv, Y.; Han, Y.; Deng, J.; Zhou, Y. Isorhamnetin Inhibits Liver Fibrosis by Reducing Autophagy and Inhibiting Extracellular Matrix Formation via the TGF-Β1/Smad3 and TGF-Β1/P38 MAPK Pathways. Mediat. Inflamm. 2019, 2019, 6175091. [Google Scholar] [CrossRef] [PubMed]
  85. Yang, J.H.; Kim, S.C.; Kim, K.M.; Jang, C.H.; Cho, S.S.; Kim, S.J.; Ku, S.K.; Cho, I.J.; Ki, S.H. Isorhamnetin Attenuates Liver Fibrosis by Inhibiting TGF-β/Smad Signaling and Relieving Oxidative Stress. Eur. J. Pharmacol. 2016, 783, 92–102. [Google Scholar] [CrossRef] [PubMed]
  86. Cao, R.; Cao, C.; Hu, X.; Du, K.; Zhang, J.; Li, M.; Li, B.; Lin, H.; Zhang, A.; Li, Y.; et al. Kaempferol Attenuates Carbon Tetrachloride (CCl4)-Induced Hepatic Fibrosis by Promoting ASIC1a Degradation and Suppression of the ASIC1a-Mediated ERS. Phytomed. Int. J. Phytother. Phytopharm. 2023, 121, 155125. [Google Scholar] [CrossRef]
  87. Zhou, G.; Li, C.; Zhang, R.; Zhan, Y.; Lin, L.; Lang, Z.; Tao, Q.; Zheng, J. Kaempferol Inhibits Hepatic Stellate Cell Activation by Regulating miR-26b-5p/Jag1 Axis and Notch Pathway. Front. Pharmacol. 2022, 13, 881855. [Google Scholar] [CrossRef]
  88. Xu, T.; Huang, S.; Huang, Q.; Ming, Z.; Wang, M.; Li, R.; Zhao, Y. Kaempferol Attenuates Liver Fibrosis by Inhibiting Activin Receptor-like Kinase 5. J. Cell. Mol. Med. 2019, 23, 6403–6410. [Google Scholar] [CrossRef]
  89. Huang, H.; Wei, S.; Wu, X.; Zhang, M.; Zhou, B.; Huang, D.; Dong, W. Dihydrokaempferol Attenuates CCl4-Induced Hepatic Fibrosis by Inhibiting PARP-1 to Affect Multiple Downstream Pathways and Cytokines. Toxicol. Appl. Pharmacol. 2023, 464, 116438. [Google Scholar] [CrossRef] [PubMed]
  90. Perumal, N.; Perumal, M.; Halagowder, D.; Sivasithamparam, N. Morin Attenuates Diethylnitrosamine-Induced Rat Liver Fibrosis and Hepatic Stellate Cell Activation by Co-Ordinated Regulation of Hippo/Yap and TGF-Β1/Smad Signaling. Biochimie 2017, 140, 10–19. [Google Scholar] [CrossRef]
  91. Sang, L.; Wang, X.-M.; Xu, D.-Y.; Sang, L.-X.; Han, Y.; Jiang, L.-Y. Morin Enhances Hepatic Nrf2 Expression in a Liver Fibrosis Rat Model. World J. Gastroenterol. 2017, 23, 8334–8344. [Google Scholar] [CrossRef]
  92. Yuan, W.; Ahmad, S.; Najar, A. Morin, a Plant Derived Flavonoid, Modulates the Expression of Peroxisome Proliferator-Activated Receptor-γ Coactivator-1α Mediated by AMPK Pathway in Hepatic Stellate Cells. Am. J. Transl. Res. 2017, 9, 5662–5670. [Google Scholar]
  93. MadanKumar, P.; NaveenKumar, P.; Devaraj, H.; NiranjaliDevaraj, S. Morin, a Dietary Flavonoid, Exhibits Anti-Fibrotic Effect and Induces Apoptosis of Activated Hepatic Stellate Cells by Suppressing Canonical NF-κB Signaling. Biochimie 2015, 110, 107–118. [Google Scholar] [CrossRef] [PubMed]
  94. Heeba, G.H.; Mahmoud, M.E. Therapeutic Potential of Morin against Liver Fibrosis in Rats: Modulation of Oxidative Stress, Cytokine Production and Nuclear Factor Kappa B. Environ. Toxicol. Pharmacol. 2014, 37, 662–671. [Google Scholar] [CrossRef] [PubMed]
  95. MadanKumar, P.; NaveenKumar, P.; Manikandan, S.; Devaraj, H.; NiranjaliDevaraj, S. Morin Ameliorates Chemically Induced Liver Fibrosis in Vivo and Inhibits Stellate Cell Proliferation in Vitro by Suppressing Wnt/β-Catenin Signaling. Toxicol. Appl. Pharmacol. 2014, 277, 210–220. [Google Scholar] [CrossRef] [PubMed]
  96. Lee, H.-S.; Jung, K.H.; Park, I.-S.; Kwon, S.W.; Lee, D.-H.; Hong, S.-S. Protective Effect of Morin on Dimethylnitrosamine-Induced Hepatic Fibrosis in Rats. Dig. Dis. Sci. 2009, 54, 782–788. [Google Scholar] [CrossRef] [PubMed]
  97. Geng, Y.; Sun, Q.; Li, W.; Lu, Z.-M.; Xu, H.-Y.; Shi, J.-S.; Xu, Z.-H. The Common Dietary Flavonoid Myricetin Attenuates Liver Fibrosis in Carbon Tetrachloride Treated Mice. Mol. Nutr. Food Res. 2017, 61, 1600392. [Google Scholar] [CrossRef]
  98. Domitrović, R.; Rashed, K.; Cvijanović, O.; Vladimir-Knežević, S.; Škoda, M.; Višnić, A. Myricitrin Exhibits Antioxidant, Anti-Inflammatory and Antifibrotic Activity in Carbon Tetrachloride-Intoxicated Mice. Chem. Biol. Interact. 2015, 230, 21–29. [Google Scholar] [CrossRef]
  99. Salama, Y.A.; Hassan, H.M.; El-Gayar, A.M.; Abdel-Rahman, N. Combined Quercetin and Simvastatin Attenuate Hepatic Fibrosis in Rats by Modulating SphK1/NLRP3 Pathways. Life Sci. 2024, 337, 122349. [Google Scholar] [CrossRef]
  100. Aslam, A.; Sheikh, N.; Shahzad, M.; Saeed, G.; Fatima, N.; Akhtar, T. Quercetin Ameliorates Thioacetamide-Induced Hepatic Fibrosis and Oxidative Stress by Antagonizing the Hedgehog Signaling Pathway. J. Cell. Biochem. 2022, 123, 1356–1365. [Google Scholar] [CrossRef]
  101. Li, X.; Jin, Q.; Yao, Q.; Xu, B.; Li, L.; Zhang, S.; Tu, C. The Flavonoid Quercetin Ameliorates Liver Inflammation and Fibrosis by Regulating Hepatic Macrophages Activation and Polarization in Mice. Front. Pharmacol. 2018, 9, 72. [Google Scholar] [CrossRef]
  102. Wu, L.; Zhang, Q.; Mo, W.; Feng, J.; Li, S.; Li, J.; Liu, T.; Xu, S.; Wang, W.; Lu, X.; et al. Quercetin Prevents Hepatic Fibrosis by Inhibiting Hepatic Stellate Cell Activation and Reducing Autophagy via the TGF-Β1/Smads and PI3K/Akt Pathways. Sci. Rep. 2017, 7, 9289. [Google Scholar] [CrossRef]
  103. Li, X.; Jin, Q.; Yao, Q.; Xu, B.; Li, Z.; Tu, C. Quercetin Attenuates the Activation of Hepatic Stellate Cells and Liver Fibrosis in Mice through Modulation of HMGB1-TLR2/4-NF-κB Signaling Pathways. Toxicol. Lett. 2016, 261, 1–12. [Google Scholar] [CrossRef]
  104. Hernández-Ortega, L.D.; Alcántar-Díaz, B.E.; Ruiz-Corro, L.A.; Sandoval-Rodriguez, A.; Bueno-Topete, M.; Armendariz-Borunda, J.; Salazar-Montes, A.M. Quercetin Improves Hepatic Fibrosis Reducing Hepatic Stellate Cells and Regulating Pro-Fibrogenic/Anti-Fibrogenic Molecules Balance. J. Gastroenterol. Hepatol. 2012, 27, 1865–1872. [Google Scholar] [CrossRef] [PubMed]
  105. Gürler, S.; Kiraz, Y.; Baran, Y. Chapter 21. Flavonoids in Cancer Therapy: Current and Future Trends. In Biodiversity and Biomedicine; Ozturk, M., Egamberdieva, D., Pešić, M., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 403–440. [Google Scholar]
  106. Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. Bethesda Md 2017, 8, 423–435. [Google Scholar] [CrossRef] [PubMed]
  107. Hano, C.; Tungmunnithum, D. Plant Polyphenols, More than Just Simple Natural Antioxidants: Oxidative Stress, Aging and Age-Related Diseases. Medicines 2020, 7, 26. [Google Scholar] [CrossRef]
  108. Zhu, Z.; Hu, R.; Li, J.; Xing, X.; Chen, J.; Zhou, Q.; Sun, J. Alpinetin Exerts Anti-Inflammatory, Anti-Oxidative and Anti-Angiogenic Effects through Activating the Nrf2 Pathway and Inhibiting NLRP3 Pathway in Carbon Tetrachloride-Induced Liver Fibrosis. Int. Immunopharmacol. 2021, 96, 107660. [Google Scholar] [CrossRef] [PubMed]
  109. Melaibari, M.; Alkreathy, H.M.; Esmat, A.; Rajeh, N.A.; Shaik, R.A.; Alghamdi, A.A.; Ahmad, A. Anti-Fibrotic Efficacy of Apigenin in a Mice Model of Carbon Tetrachloride-Induced Hepatic Fibrosis by Modulation of Oxidative Stress, Inflammation, and Fibrogenesis: A Preclinical Study. Biomedicines 2023, 11, 1342. [Google Scholar] [CrossRef]
  110. Ji, J.; Yu, Q.; Dai, W.; Wu, L.; Feng, J.; Zheng, Y.; Li, Y.; Guo, C. Apigenin Alleviates Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation and Autophagy via TGF-Β1/Smad3 and P38/PPARα Pathways. PPAR Res. 2021, 2021, 6651839. [Google Scholar] [CrossRef]
  111. Qiao, M.; Yang, J.; Zhu, Y.; Zhao, Y.; Hu, J. Transcriptomics and Proteomics Analysis of System-Level Mechanisms in the Liver of Apigenin-Treated Fibrotic Rats. Life Sci. 2020, 248, 117475. [Google Scholar] [CrossRef]
  112. Wu, X.; Zhi, F.; Lun, W.; Deng, Q.; Zhang, W. Baicalin Inhibits PDGF-BB-Induced Hepatic Stellate Cell Proliferation, Apoptosis, Invasion, Migration and Activation via the miR-3595/ACSL4 Axis. Int. J. Mol. Med. 2018, 41, 1992–2002. [Google Scholar] [CrossRef]
  113. Qiao, H.; Han, H.; Hong, D.; Ren, Z.; Chen, Y.; Zhou, C. Protective Effects of Baicalin on Carbon Tetrachloride Induced Liver Injury by Activating PPARγ and Inhibiting TGFβ1. Pharm. Biol. 2011, 49, 38–45. [Google Scholar] [CrossRef]
  114. Sun, H.; Che, Q.-M.; Zhao, X.; Pu, X.-P. Antifibrotic Effects of Chronic Baicalein Administration in a CCl4 Liver Fibrosis Model in Rats. Eur. J. Pharmacol. 2010, 631, 53–60. [Google Scholar] [CrossRef] [PubMed]
  115. Balta, C.; Ciceu, A.; Herman, H.; Rosu, M.; Boldura, O.M.; Hermenean, A. Dose-Dependent Antifibrotic Effect of Chrysin on Regression of Liver Fibrosis: The Role in Extracellular Matrix Remodeling. Dose-Response Publ. Int. Hormesis Soc. 2018, 16, 1559325818789835. [Google Scholar] [CrossRef] [PubMed]
  116. Balta, C.; Herman, H.; Boldura, O.M.; Gasca, I.; Rosu, M.; Ardelean, A.; Hermenean, A. Chrysin Attenuates Liver Fibrosis and Hepatic Stellate Cell Activation through TGF-β/Smad Signaling Pathway. Chem. Biol. Interact. 2015, 240, 94–101. [Google Scholar] [CrossRef]
  117. Hasan, H.F.; Abdel-Rafei, M.K.; Galal, S.M. Diosmin Attenuates Radiation-Induced Hepatic Fibrosis by Boosting PPAR-γ Expression and Hampering miR-17-5p-Activated Canonical Wnt-β-Catenin Signaling. Biochem. Cell Biol. Biochim. Biol. Cell. 2017, 95, 400–414. [Google Scholar] [CrossRef]
  118. Hu, J.; Liu, Y.; Pan, Z.; Huang, X.; Wang, J.; Cao, W.; Chen, Z. Eupatilin Ameliorates Hepatic Fibrosis and Hepatic Stellate Cell Activation by Suppressing β-Catenin/PAI-1 Pathway. Int. J. Mol. Sci. 2023, 24, 5933. [Google Scholar] [CrossRef] [PubMed]
  119. Chen, Y.-X.; Huang, Q.-F.; Lin, X.; Wei, J.-B. [Protective effect of isoorientin on alcohol-induced hepatic fibrosis in rats]. Zhongguo Zhong Yao Za Zhi Zhongguo Zhongyao Zazhi China J. Chin. Mater. Medica 2013, 38, 3726–3730. [Google Scholar]
  120. Huang, Q.F.; Zhang, S.J.; Zheng, L.; Liao, M.; He, M.; Huang, R.; Zhuo, L.; Lin, X. Protective Effect of Isoorientin-2″-O-α-L-Arabinopyranosyl Isolated from Gypsophila Elegans on Alcohol Induced Hepatic Fibrosis in Rats. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2012, 50, 1992–2001. [Google Scholar] [CrossRef]
  121. Huang, Y.; Luo, W.; Chen, S.; Su, H.; Zhu, W.; Wei, Y.; Qiu, Y.; Long, Y.; Shi, Y.; Wei, J. Isovitexin Alleviates Hepatic Fibrosis by Regulating miR-21-Mediated PI3K/Akt Signaling and Glutathione Metabolic Pathway: Based on Transcriptomics and Metabolomics. Phytomed. Int. J. Phytother. Phytopharm. 2023, 121, 155117. [Google Scholar] [CrossRef]
  122. Kang, R.; Tian, W.; Cao, W.; Sun, Y.; Zhang, H.-N.; Feng, Y.-D.; Li, C.; Li, Z.-Z.; Li, X.-Q. Ligustroflavone Ameliorates CCl4-Induced Liver Fibrosis through down-Regulating the TGF-β/Smad Signaling Pathway. Chin. J. Nat. Med. 2021, 19, 170–180. [Google Scholar] [CrossRef]
  123. Batudeligen; Han, Z.; Chen, H.; Narisu; Xu, Y.; Anda; Han, G. Luteolin Alleviates Liver Fibrosis in Rat Hepatic Stellate Cell HSC-T6: A Proteomic Analysis. Drug Des. Devel. Ther. 2023, 17, 1819–1829. [Google Scholar] [CrossRef]
  124. Li, J.; Li, X.; Xu, W.; Wang, S.; Hu, Z.; Zhang, Q.; Deng, X.; Wang, J.; Zhang, J.; Guo, C. Antifibrotic Effects of Luteolin on Hepatic Stellate Cells and Liver Fibrosis by Targeting AKT/mTOR/p70S6K and TGFβ/Smad Signalling Pathways. Liver Int. Off. J. Int. Assoc. Study Liver 2014, 35, 1222–1233. [Google Scholar] [CrossRef] [PubMed]
  125. Domitrović, R.; Jakovac, H.; Tomac, J.; Sain, I. Liver Fibrosis in Mice Induced by Carbon Tetrachloride and Its Reversion by Luteolin. Toxicol. Appl. Pharmacol. 2009, 241, 311–321. [Google Scholar] [CrossRef]
  126. Xiu, M.; Zhao, Y.; Wang, X.; Yuan, S.; Qin, B.; Sun, J.; Cui, L.; Song, J. Regulation of SIRT1-TLR2/TLR4 Pathway in Cell Communication from Macrophages to Hepatic Stellate Cells Contribute to Alleviates Hepatic Fibrosis by Luteoloside. Acta Histochem. 2023, 125, 151989. [Google Scholar] [CrossRef]
  127. Hao, Y.; Feng, D.; Ye, H.; Liao, W. Nobiletin Alleviated Epithelial-Mesenchymal Transition of Hepatocytes in Liver Fibrosis Based on Autophagy-Hippo/YAP Pathway. Mol. Nutr. Food Res. 2024, 68, e2300529. [Google Scholar] [CrossRef] [PubMed]
  128. Sun, Y.; Weng, J.; Chen, X.; Ma, S.; Zhang, Y.; Zhang, F.; Zhang, Z.; Wang, F.; Shao, J.; Zheng, S. Oroxylin A Activates Ferritinophagy to Induce Hepatic Stellate Cell Senescence against Hepatic Fibrosis by Regulating cGAS-STING Pathway. Biomed. Pharmacother. Biomed. Pharmacother. 2023, 162, 114653. [Google Scholar] [CrossRef]
  129. Shen, M.; Guo, M.; Wang, Z.; Li, Y.; Kong, D.; Shao, J.; Tan, S.; Chen, A.; Zhang, F.; Zhang, Z.; et al. ROS-Dependent Inhibition of the PI3K/Akt/mTOR Signaling Is Required for Oroxylin A to Exert Anti-Inflammatory Activity in Liver Fibrosis. Int. Immunopharmacol. 2020, 85, 106637. [Google Scholar] [CrossRef]
  130. Bian, M.; He, J.; Jin, H.; Lian, N.; Shao, J.; Guo, Q.; Wang, S.; Zhang, F.; Zheng, S. Oroxylin A Induces Apoptosis of Activated Hepatic Stellate Cells through Endoplasmic Reticulum Stress. Apoptosis Int. J. Program. Cell Death 2019, 24, 905–920. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, F.; Jia, Y.; Li, M.; Wang, L.; Shao, J.; Guo, Q.; Tan, S.; Ding, H.; Chen, A.; Zhang, F.; et al. Blockade of Glycolysis-Dependent Contraction by Oroxylin a via Inhibition of Lactate Dehydrogenase-a in Hepatic Stellate Cells. Cell Commun. Signal. CCS 2019, 17, 11. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, W.; Zhang, Z.; Yao, Z.; Wang, L.; Zhang, F.; Shao, J.; Chen, A.; Zheng, S. Activation of Autophagy Is Required for Oroxylin A to Alleviate Carbon Tetrachloride-Induced Liver Fibrosis and Hepatic Stellate Cell Activation. Int. Immunopharmacol. 2018, 56, 148–155. [Google Scholar] [CrossRef]
  133. Seki, N.; Toh, U.; Kawaguchi, K.; Ninomiya, M.; Koketsu, M.; Watanabe, K.; Aoki, M.; Fujii, T.; Nakamura, A.; Akagi, Y.; et al. Tricin Inhibits Proliferation of Human Hepatic Stellate Cells in Vitro by Blocking Tyrosine Phosphorylation of PDGF Receptor and Its Signaling Pathways. J. Cell. Biochem. 2012, 113, 2346–2355. [Google Scholar] [CrossRef]
  134. Du, X.-S.; Li, H.-D.; Yang, X.-J.; Li, J.-J.; Xu, J.-J.; Chen, Y.; Xu, Q.-Q.; Yang, L.; He, C.-S.; Huang, C.; et al. Wogonin Attenuates Liver Fibrosis via Regulating Hepatic Stellate Cell Activation and Apoptosis. Int. Immunopharmacol. 2019, 75, 105671. [Google Scholar] [CrossRef]
  135. Ma, J.-Q.; Sun, Y.-Z.; Ming, Q.-L.; Tian, Z.-K.; Yang, H.-X.; Liu, C.-M. Ampelopsin Attenuates Carbon Tetrachloride-Induced Mouse Liver Fibrosis and Hepatic Stellate Cell Activation Associated with the SIRT1/TGF-Β1/Smad3 and Autophagy Pathway. Int. Immunopharmacol. 2019, 77, 105984. [Google Scholar] [CrossRef] [PubMed]
  136. Shi, H.; Shi, H.; Ren, F.; Chen, D.; Chen, Y.; Duan, Z. Naringin in Ganshuang Granule Suppresses Activation of Hepatic Stellate Cells for Anti-Fibrosis Effect by Inhibition of Mammalian Target of Rapamycin. J. Cell. Mol. Med. 2017, 21, 500–509. [Google Scholar] [CrossRef] [PubMed]
  137. Said, M.M.; Azab, S.S.; Saeed, N.M.; El-Demerdash, E. Antifibrotic Mechanism of Pinocembrin: Impact on Oxidative Stress, Inflammation and TGF-β /Smad Inhibition in Rats. Ann. Hepatol. 2018, 17, 307–317. [Google Scholar] [CrossRef] [PubMed]
  138. Zhou, F.; Wang, A.; Li, D.; Wang, Y.; Lin, L. Pinocembrin from Penthorum Chinense Pursh Suppresses Hepatic Stellate Cells Activation through a Unified SIRT3-TGF-β-Smad Signaling Pathway. Toxicol. Appl. Pharmacol. 2018, 341, 38–50. [Google Scholar] [CrossRef]
  139. Zhang, M.; Wang, Y.; Zhu, G.; Sun, C.; Wang, J. Hepatoprotective Effect and Possible Mechanism of Phytoestrogen Calycosin on Carbon Tetrachloride-Induced Liver Fibrosis in Mice. Naunyn. Schmiedebergs Arch. Pharmacol. 2020, 394, 189–204. [Google Scholar] [CrossRef] [PubMed]
  140. Xu, Y.; Zhang, D.; Yang, H.; Liu, Y.; Zhang, L.; Zhang, C.; Chen, G.; Hu, Y.; Chen, J.; Zhang, H.; et al. Hepatoprotective Effect of Genistein against Dimethylnitrosamine-Induced Liver Fibrosis in Rats by Regulating Macrophage Functional Properties and Inhibiting the JAK2/STAT3/SOCS3 Signaling Pathway. Front. Biosci. Landmark Ed. 2021, 26, 1572–1584. [Google Scholar] [CrossRef]
  141. Ganai, A.A.; Husain, M. Genistein Attenuates D-GalN Induced Liver Fibrosis/Chronic Liver Damage in Rats by Blocking the TGF-β/Smad Signaling Pathways. Chem. Biol. Interact. 2017, 261, 80–85. [Google Scholar] [CrossRef] [PubMed]
  142. Huang, Q.; Huang, R.; Zhang, S.; Lin, J.; Wei, L.; He, M.; Zhuo, L.; Lin, X. Protective Effect of Genistein Isolated from Hydrocotyle Sibthorpioides on Hepatic Injury and Fibrosis Induced by Chronic Alcohol in Rats. Toxicol. Lett. 2013, 217, 102–110. [Google Scholar] [CrossRef]
  143. Salas, A.L.; Montezuma, T.D.; Fariña, G.G.; Reyes-Esparza, J.; Rodríguez-Fragoso, L. Genistein Modifies Liver Fibrosis and Improves Liver Function by Inducing uPA Expression and Proteolytic Activity in CCl4-Treated Rats. Pharmacology 2008, 81, 41–49. [Google Scholar] [CrossRef]
  144. Zhang, L.; Zhang, H.; Gu, J.; Xu, W.; Yuan, N.; Sun, J.; Li, H. Glabridin Inhibits Liver Fibrosis and Hepatic Stellate Cells Activation through Suppression of Inflammation and Oxidative Stress by Activating PPARγ in Carbon Tetrachloride-Treated Mice. Int. Immunopharmacol. 2022, 113, 109433. [Google Scholar] [CrossRef]
  145. Li, X.; Zhang, H.; Pan, L.; Zou, H.; Miao, X.; Cheng, J.; Wu, Y. Puerarin Alleviates Liver Fibrosis via Inhibition of the ERK1/2 Signaling Pathway in Thioacetamide-Induced Hepatic Fibrosis in Rats. Exp. Ther. Med. 2019, 18, 133–138. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, G.-R.; Wei, S.-J.; Huang, Y.-Q.; Xing, W.; Wang, L.-Y.; Liang, L.-L. Mechanism of Combined Use of Vitamin D and Puerarin in Anti-Hepatic Fibrosis by Regulating the Wnt/β-Catenin Signalling Pathway. World J. Gastroenterol. 2018, 24, 4178–4185. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, S.; Shi, X.-L.; Feng, M.; Wang, X.; Zhang, Z.-H.; Zhao, X.; Han, B.; Ma, H.-C.; Dai, B.; Ding, Y.-T. Puerarin Protects against CCl4-Induced Liver Fibrosis in Mice: Possible Role of PARP-1 Inhibition. Int. Immunopharmacol. 2016, 38, 238–245. [Google Scholar] [CrossRef] [PubMed]
  148. Guo, C.; Xu, L.; He, Q.; Liang, T.; Duan, X.; Li, R. Anti-Fibrotic Effects of Puerarin on CCl4-Induced Hepatic Fibrosis in Rats Possibly through the Regulation of PPAR-γ Expression and Inhibition of PI3K/Akt Pathway. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2013, 56, 436–442. [Google Scholar] [CrossRef] [PubMed]
  149. Li, R.; Xu, L.; Liang, T.; Li, Y.; Zhang, S.; Duan, X. Puerarin Mediates Hepatoprotection against CCl4-Induced Hepatic Fibrosis Rats through Attenuation of Inflammation Response and Amelioration of Metabolic Function. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2013, 52, 69–75. [Google Scholar] [CrossRef]
  150. Xu, L.; Zheng, N.; He, Q.; Li, R.; Zhang, K.; Liang, T. Puerarin, Isolated from Pueraria Lobata (Willd.), Protects against Hepatotoxicity via Specific Inhibition of the TGF-Β1/Smad Signaling Pathway, Thereby Leading to Anti-Fibrotic Effect. Phytomed. Int. J. Phytother. Phytopharm. 2013, 20, 1172–1179. [Google Scholar] [CrossRef]
  151. Zhang, S.; Ji, G.; Liu, J. Reversal of Chemical-Induced Liver Fibrosis in Wistar Rats by Puerarin. J. Nutr. Biochem. 2006, 17, 485–491. [Google Scholar] [CrossRef]
  152. Li, J.-F.; Chen, B.-C.; Lai, D.-D.; Jia, Z.-R.; Andersson, R.; Zhang, B.; Yao, J.-G.; Yu, Z. Soy Isoflavone Delays the Progression of Thioacetamide-Induced Liver Fibrosis in Rats. Scand. J. Gastroenterol. 2011, 46, 341–349. [Google Scholar] [CrossRef]
  153. Gao, X.-X.; Shi, D.-H.; Chen, Y.-X.; Cui, J.-T.; Wang, Y.-R.; Jiang, C.-P.; Wu, J.-H. The Therapeutic Effects of Tectorigenin on Chemically Induced Liver Fibrosis in Rats and an Associated Metabonomic Investigation. Arch. Pharm. Res. 2012, 35, 1479–1493. [Google Scholar] [CrossRef]
  154. Zhan, W.; Liao, X.; Xie, R.-J.; Tian, T.; Yu, L.; Liu, X.; Liu, J.; Li, P.; Han, B.; Yang, T.; et al. The Effects of Blueberry Anthocyanins on Histone Acetylation in Rat Liver Fibrosis. Oncotarget 2017, 8, 96761–96773. [Google Scholar] [CrossRef]
  155. Zhan, W.; Liao, X.; Tian, T.; Yu, L.; Liu, X.; Li, B.; Liu, J.; Han, B.; Xie, R.J.; Ji, Q.H.; et al. Study on the Effects of Blueberry Treatment on Histone Acetylation Modification of CCl4-Induced Liver Disease in Rats. Genet. Mol. Res. GMR 2017, 16, 16019188. [Google Scholar] [CrossRef]
  156. Zhan, W.; Liao, X.; Yu, L.; Tian, T.; Liu, X.; Liu, J.; Cai, L.-J.; Xiao, X.; Xie, R.-J.; Yang, Q. Effects of Blueberries on Migration, Invasion, Proliferation, the Cell Cycle and Apoptosis in Hepatocellular Carcinoma Cells. Biomed. Rep. 2016, 5, 579–584. [Google Scholar] [CrossRef] [PubMed]
  157. Du, J.; Liu, L.; Fan, H.; Yu, Y.; Luo, Y.; Gu, F.; Yu, H.; Liao, X. Anthocyanins Improve Liver Fibrosis in Mice by Regulating the Autophagic Flux Level of Hepatic Stellate Cells by Mmu_circ_0000623. Food Sci. Nutr. 2023, 11, 3002–3018. [Google Scholar] [CrossRef]
  158. Hao, R.; Gao, J.; Liu, H.; Zhang, C.; Huang, J.; Fan, J.; Wei, J. Preventive Effect and Mechanism of Anthocyanins from Aronia Melanocarpa Elliot on Hepatic Fibrosis Through TGF-β/Smad Signaling Pathway. Cell Biochem. Biophys. 2022, 80, 737–745. [Google Scholar] [CrossRef] [PubMed]
  159. Sun, J.; Wu, Y.; Long, C.; He, P.; Gu, J.; Yang, L.; Liang, Y.; Wang, Y. Anthocyanins Isolated from Blueberry Ameliorates CCl4 Induced Liver Fibrosis by Modulation of Oxidative Stress, Inflammation and Stellate Cell Activation in Mice. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2018, 120, 491–499. [Google Scholar] [CrossRef] [PubMed]
  160. Jiang, X.; Guo, H.; Shen, T.; Tang, X.; Yang, Y.; Ling, W. Cyanidin-3-O-β-Glucoside Purified from Black Rice Protects Mice against Hepatic Fibrosis Induced by Carbon Tetrachloride via Inhibiting Hepatic Stellate Cell Activation. J. Agric. Food Chem. 2015, 63, 6221–6230. [Google Scholar] [CrossRef]
  161. Domitrović, R.; Jakovac, H. Antifibrotic Activity of Anthocyanidin Delphinidin in Carbon Tetrachloride-Induced Hepatotoxicity in Mice. Toxicology 2010, 272, 1–10. [Google Scholar] [CrossRef]
  162. Ma, Y.; Li, Y.; Zhang, H.; Wang, Y.; Wu, C.; Huang, W. Malvidin Induces Hepatic Stellate Cell Apoptosis via the Endoplasmic Reticulum Stress Pathway and Mitochondrial Pathway. Food Sci. Nutr. 2020, 8, 5095–5106. [Google Scholar] [CrossRef]
  163. Shi, Y.-S.; Li, X.-X.; Li, H.-T.; Zhang, Y. Pelargonidin Ameliorates CCl4-Induced Liver Fibrosis by Suppressing the ROS-NLRP3-IL-1β Axis via Activating the Nrf2 Pathway. Food Funct. 2020, 11, 5156–5165. [Google Scholar] [CrossRef]
  164. Lee, S.H.; Nan, J.-X.; Zhao, Y.Z.; Woo, S.W.; Park, E.-J.; Kang, T.-H.; Seo, G.S.; Kim, Y.-C.; Sohn, D.H. The Chalcone Butein from Rhus Verniciflua Shows Antifibrogenic Activity. Planta Med. 2003, 69, 990–994. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, X.; Tian, H.; Chen, J.; Huang, D.; Ding, F.; Ma, T.; Xi, J.; Wu, C.; Zhang, Y. Isobavachalcone Attenuates Liver Fibrosis via Activation of the Nrf2/HO-1 Pathway in Rats. Int. Immunopharmacol. 2024, 128, 111398. [Google Scholar] [CrossRef] [PubMed]
  166. Singh, H.; Sidhu, S.; Chopra, K.; Khan, M.U. Hepatoprotective Effect of Trans-Chalcone on Experimentally Induced Hepatic Injury in Rats: Inhibition of Hepatic Inflammation and Fibrosis. Can. J. Physiol. Pharmacol. 2016, 94, 879–887. [Google Scholar] [CrossRef] [PubMed]
  167. Dorn, C.; Heilmann, J.; Hellerbrand, C. Protective Effect of Xanthohumol on Toxin-Induced Liver Inflammation and Fibrosis. Int. J. Clin. Exp. Pathol. 2012, 5, 29–36. [Google Scholar] [CrossRef]
  168. Dorn, C.; Kraus, B.; Motyl, M.; Weiss, T.S.; Gehrig, M.; Schölmerich, J.; Heilmann, J.; Hellerbrand, C. Xanthohumol, a Chalcon Derived from Hops, Inhibits Hepatic Inflammation and Fibrosis. Mol. Nutr. Food Res. 2010, 54 (Suppl. S2), S205–S213. [Google Scholar] [CrossRef]
  169. Le, C.T.; Nguyen, G.; Park, S.Y.; Dong, H.N.; Cho, Y.K.; Lee, J.-H.; Im, S.-S.; Choi, D.-H.; Cho, E.-H. Phloretin Ameliorates Succinate-Induced Liver Fibrosis by Regulating Hepatic Stellate Cells. Endocrinol. Metab. 2023, 38, 395–405. [Google Scholar] [CrossRef]
  170. Ye, L.; Yu, Y.; Zhao, Y. Icariin-Induced miR-875-5p Attenuates Epithelial-Mesenchymal Transition by Targeting Hedgehog Signaling in Liver Fibrosis. J. Gastroenterol. Hepatol. 2020, 35, 482–491. [Google Scholar] [CrossRef]
  171. Algandaby, M.M.; Breikaa, R.M.; Eid, B.G.; Neamatallah, T.A.; Abdel-Naim, A.B.; Ashour, O.M. Icariin Protects against Thioacetamide-Induced Liver Fibrosis in Rats: Implication of Anti-Angiogenic and Anti-Autophagic Properties. Pharmacol. Rep. PR 2017, 69, 616–624. [Google Scholar] [CrossRef]
  172. Li, J.; Liu, P.; Zhang, R.; Cao, L.; Qian, H.; Liao, J.; Xu, W.; Wu, M.; Yin, Z. Icaritin Induces Cell Death in Activated Hepatic Stellate Cells through Mitochondrial Activated Apoptosis and Ameliorates the Development of Liver Fibrosis in Rats. J. Ethnopharmacol. 2011, 137, 714–723. [Google Scholar] [CrossRef]
  173. Izzo, C.; Annunziata, M.; Melara, G.; Sciorio, R.; Dallio, M.; Masarone, M.; Federico, A.; Persico, M. The Role of Resveratrol in Liver Disease: A Comprehensive Review from In Vitro to Clinical Trials. Nutrients 2021, 13, 933. [Google Scholar] [CrossRef]
  174. Di Pascoli, M.; Diví, M.; Rodríguez-Vilarrupla, A.; Rosado, E.; Gracia-Sancho, J.; Vilaseca, M.; Bosch, J.; García-Pagán, J.C. Resveratrol Improves Intrahepatic Endothelial Dysfunction and Reduces Hepatic Fibrosis and Portal Pressure in Cirrhotic Rats. J. Hepatol. 2013, 58, 904–910. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, D.-Q.; Sun, P.; Jin, Q.; Li, X.; Zhang, Y.; Zhang, Y.-J.; Wu, Y.-L.; Nan, J.-X.; Lian, L.-H. Resveratrol Regulates Activated Hepatic Stellate Cells by Modulating NF-κB and the PI3K/Akt Signaling Pathway. J. Food Sci. 2016, 81, H240–H245. [Google Scholar] [CrossRef] [PubMed]
  176. Zhu, L.; Mou, Q.; Wang, Y.; Zhu, Z.; Cheng, M. Resveratrol Contributes to the Inhibition of Liver Fibrosis by Inducing Autophagy via the microRNA-20a-mediated Activation of the PTEN/PI3K/AKT Signaling Pathway. Int. J. Mol. Med. 2020, 46, 2035–2046. [Google Scholar] [CrossRef] [PubMed]
  177. Abdu, S.B.; Al-Bogami, F.M. Influence of Resveratrol on Liver Fibrosis Induced by Dimethylnitrosamine in Male Rats. Saudi J. Biol. Sci. 2019, 26, 201–209. [Google Scholar] [CrossRef]
  178. Lee, E.-S.; Shin, M.-O.; Yoon, S.; Moon, J.-O. Resveratrol Inhibits Dimethylnitrosamine-Induced Hepatic Fibrosis in Rats. Arch. Pharm. Res. 2010, 33, 925–932. [Google Scholar] [CrossRef] [PubMed]
  179. Hong, S.-W.; Jung, K.H.; Zheng, H.-M.; Lee, H.-S.; Suh, J.-K.; Park, I.-S.; Lee, D.-H.; Hong, S.-S. The Protective Effect of Resveratrol on Dimethylnitrosamine-Induced Liver Fibrosis in Rats. Arch. Pharm. Res. 2010, 33, 601–609. [Google Scholar] [CrossRef]
  180. Li, S.; Han, B.; Li, J.; Lv, Z.; Jiang, H.; Liu, Y.; Yang, X.; Lu, J.; Zhang, Z. Resveratrol Alleviates Liver Fibrosis Induced by Long-Term Inorganic Mercury Exposure through Activating the Sirt1/PGC-1α Signaling Pathway. J. Agric. Food Chem. 2024, 72, 15985–15997. [Google Scholar] [CrossRef]
  181. Chai, Y.-L.; Zhang, H.-N.; Gao, Y.; Li, D.-H.; Zhang, H. A Study on the Protective Impact of Resveratrol on Liver Damage in Rats with Obstructive Jaundice. Comb. Chem. High Throughput Screen. 2024, 27, 1–9. [Google Scholar] [CrossRef]
  182. Hung, W.-L.; Hsiao, Y.-T.; Chiou, Y.-S.; Nagabhushanam, K.; Ho, C.-T.; Pan, M.-H. Hepatoprotective Effect of Piceatannol against Carbon Tetrachloride-Induced Liver Fibrosis in Mice. Food Funct. 2021, 12, 11229–11240. [Google Scholar] [CrossRef]
  183. Lee, M.-F.; Liu, M.-L.; Cheng, A.-C.; Tsai, M.-L.; Ho, C.-T.; Liou, W.-S.; Pan, M.-H. Pterostilbene Inhibits Dimethylnitrosamine-Induced Liver Fibrosis in Rats. Food Chem. 2013, 138, 802–807. [Google Scholar] [CrossRef]
  184. Shi, B.; Qian, J.; Miao, H.; Zhang, S.; Hu, Y.; Liu, P.; Xu, L. Mulberroside A Ameliorates CCl4-Induced Liver Fibrosis in Mice via Inhibiting pro-Inflammatory Response. Food Sci. Nutr. 2023, 11, 3433–3441. [Google Scholar] [CrossRef] [PubMed]
  185. Chauhan, N.; Dilbaghi, N.; Gopal, M.; Kumar, R.; Kim, K.-H.; Kumar, S. Development of Chitosan Nanocapsules for the Controlled Release of Hexaconazole. Int. J. Biol. Macromol. 2017, 97, 616–624. [Google Scholar] [CrossRef] [PubMed]
  186. Kumar, S.; Bhanjana, G.; Verma, R.K.; Dhingra, D.; Dilbaghi, N.; Kim, K.-H. Metformin-Loaded Alginate Nanoparticles as an Effective Antidiabetic Agent for Controlled Drug Release. J. Pharm. Pharmacol. 2017, 69, 143–150. [Google Scholar] [CrossRef]
  187. Kumar, S.; Bhanjana, G.; Kumar, A.; Taneja, K.; Dilbaghi, N.; Kim, K.-H. Synthesis and Optimization of Ceftriaxone-Loaded Solid Lipid Nanocarriers. Chem. Phys. Lipids 2016, 200, 126–132. [Google Scholar] [CrossRef]
  188. Kumar, S.; Dilbaghi, N.; Saharan, R.; Bhanjana, G. Nanotechnology as Emerging Tool for Enhancing Solubility of Poorly Water-Soluble Drugs. BioNanoScience 2012, 2, 227–250. [Google Scholar] [CrossRef]
  189. De Jong, W.H.; Borm, P.J.A. Drug Delivery and Nanoparticles:Applications and Hazards. Int. J. Nanomed. 2008, 3, 133–149. [Google Scholar] [CrossRef]
  190. Hashemi, M.; Ebrahimian, M. Recent Advances in Nanoformulations for Co-Delivery of Curcumin and Chemotherapeutic Drugs. Nanomedi. J. 2017, 4, 1–7. [Google Scholar] [CrossRef]
  191. Patel, T.; Zhou, J.; Piepmeier, J.M.; Saltzman, W.M. Polymeric Nanoparticles for Drug Delivery to the Central Nervous System. Adv. Drug Deliv. Rev. 2012, 64, 701–705. [Google Scholar] [CrossRef]
  192. Kang, L.; Gao, Z.; Huang, W.; Jin, M.; Wang, Q. Nanocarrier-Mediated Co-Delivery of Chemotherapeutic Drugs and Gene Agents for Cancer Treatment. Acta Pharm. Sin. B 2015, 5, 169–175. [Google Scholar] [CrossRef]
  193. Afsharzadeh, M.; Hashemi, M.; Mokhtarzadeh, A.; Abnous, K.; Ramezani, M. Recent Advances in Co-Delivery Systems Based on Polymeric Nanoparticle for Cancer Treatment. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1095–1110. [Google Scholar] [CrossRef]
  194. Pawar, A.; Rajalakshmi, S.; Mehta, P.; Shaikh, K.; Bothiraja, C. Strategies for Formulation Development of Andrographolide. RSC Adv. 2016, 6, 69282–69300. [Google Scholar] [CrossRef]
  195. Rajalakshmi, S.; Vyawahare, N.; Pawar, A.; Mahaparale, P.; Chellampillai, B. Current Development in Novel Drug Delivery Systems of Bioactive Molecule Plumbagin. Artif. Cells Nanomed. Biotechnol. 2018, 46, 209–218. [Google Scholar] [CrossRef] [PubMed]
  196. Jain, A.K.; Thareja, S. In Vitro and in Vivo Characterization of Pharmaceutical Nanocarriers Used for Drug Delivery. Artif. Cells Nanomed. Biotechnol. 2019, 47, 524–539. [Google Scholar] [CrossRef] [PubMed]
  197. Oroojan, A.A.; Ahangarpour, A.; Paknejad, B.; Zareian, P.; Hami, Z.; Abtahi, S.R. Effects of Myricitrin and Solid Lipid Nanoparticle-Containing Myricitrin on Reproductive System Disorders Induced by Diabetes in Male Mouse. World J. Mens Health 2021, 39, 147–157. [Google Scholar] [CrossRef]
  198. Jain, N.; Jain, R.; Thakur, N.; Gupta, B.; Jain, D.; Banveer, J.; Jain, S. Nanotechnology: A Seif and Effective Drug Delivery System. Asian J. Pharm. Clin. Res. 2010, 3, 159–165. [Google Scholar]
  199. Bilia, A.R.; Isacchi, B.; Righeschi, C.; Guccione, C.; Bergonzi, M.C. Flavonoids Loaded in Nanocarriers: An Opportunity to Increase Oral Bioavailability and Bioefficacy. Food Nutr. Sci. 2014, 5, 1212–1327. [Google Scholar] [CrossRef]
  200. Leonarduzzi, G.; Testa, G.; Sottero, B.; Gamba, P.; Poli, G. Design and Development of Nanovehicle-Based Delivery Systems for Preventive or Therapeutic Supplementation with Flavonoids. Curr. Med. Chem. 2010, 17, 74–95. [Google Scholar] [CrossRef]
  201. Liechty, W.B.; Kryscio, D.R.; Slaughter, B.V.; Peppas, N.A. Polymers for Drug Delivery Systems. Annu. Rev. Chem. Biomol. Eng. 2010, 1, 149–173. [Google Scholar] [CrossRef]
  202. Danhier, F.; Ansorena, E.; Silva, J.M.; Coco, R.; Le Breton, A.; Préat, V. PLGA-Based Nanoparticles: An Overview of Biomedical Applications. J. Control. Release Off. J. Control. Release Soc. 2012, 161, 505–522. [Google Scholar] [CrossRef]
  203. Sanna, V.; Roggio, A.M.; Posadino, A.M.; Cossu, A.; Marceddu, S.; Mariani, A.; Alzari, V.; Uzzau, S.; Pintus, G.; Sechi, M. Novel Docetaxel-Loaded Nanoparticles Based on Poly(Lactide-Co-Caprolactone) and Poly(Lactide-Co-Glycolide-Co-Caprolactone) for Prostate Cancer Treatment: Formulation, Characterization, and Cytotoxicity Studies. Nanoscale Res. Lett. 2011, 6, 260. [Google Scholar] [CrossRef]
  204. Avgoustakis, K. Pegylated Poly(Lactide) and Poly(Lactide-Co-Glycolide) Nanoparticles: Preparation, Properties and Possible Applications in Drug Delivery. Curr. Drug Deliv. 2004, 1, 321–333. [Google Scholar] [CrossRef] [PubMed]
  205. Langroodi, F.A.; Hafezi Ghahestani, Z.; Alibolandi, M.; Ebrahimian, M.; Hashemi, M. Evaluation of the Effect of Crocetin on Antitumor Activity of Doxorubicin Encapsulated in PLGA Nanoparticles. Nanomed. J. 2016, 3, 23–34. [Google Scholar] [CrossRef]
  206. Cheng, J.; Teply, B.A.; Sherifi, I.; Sung, J.; Luther, G.; Gu, F.X.; Levy-Nissenbaum, E.; Radovic-Moreno, A.F.; Langer, R.; Farokhzad, O.C. Formulation of Functionalized PLGA-PEG Nanoparticles for in Vivo Targeted Drug Delivery. Biomaterials 2007, 28, 869–876. [Google Scholar] [CrossRef] [PubMed]
  207. Panyam, J.; Sahoo, S.K.; Prabha, S.; Bargar, T.; Labhasetwar, V. Fluorescence and Electron Microscopy Probes for Cellular and Tissue Uptake of Poly(D,L-Lactide-Co-Glycolide) Nanoparticles. Int. J. Pharm. 2003, 262, 1–11. [Google Scholar] [CrossRef] [PubMed]
  208. Sah, H.; Thoma, L.A.; Desu, H.R.; Sah, E.; Wood, G.C. Concepts and Practices Used to Develop Functional PLGA-Based Nanoparticulate Systems. Int. J. Nanomed. 2013, 8, 747–765. [Google Scholar] [CrossRef] [PubMed]
  209. Gref, R.; Minamitake, Y.; Peracchia, M.T.; Trubetskoy, V.; Torchilin, V.; Langer, R. Biodegradable Long-Circulating Polymeric Nanospheres. Science 1994, 263, 1600–1603. [Google Scholar] [CrossRef]
  210. Panyam, J.; Zhou, W.-Z.; Prabha, S.; Sahoo, S.K.; Labhasetwar, V. Rapid Endo-Lysosomal Escape of Poly(DL-Lactide-Co-Glycolide) Nanoparticles: Implications for Drug and Gene Delivery. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2002, 16, 1217–1226. [Google Scholar] [CrossRef]
  211. Acharya, S.; Sahoo, S.K. PLGA Nanoparticles Containing Various Anticancer Agents and Tumour Delivery by EPR Effect. Adv. Drug Deliv. Rev. 2011, 63, 170–183. [Google Scholar] [CrossRef]
  212. Owens, D.E.; Peppas, N.A. Opsonization, Biodistribution, and Pharmacokinetics of Polymeric Nanoparticles. Int. J. Pharm. 2006, 307, 93–102. [Google Scholar] [CrossRef]
  213. Betancourt, T.; Byrne, J.D.; Sunaryo, N.; Crowder, S.W.; Kadapakkam, M.; Patel, S.; Casciato, S.; Brannon-Peppas, L. PEGylation Strategies for Active Targeting of PLA/PLGA Nanoparticles. J. Biomed. Mater. Res. A 2009, 91, 263–276. [Google Scholar] [CrossRef]
  214. Sinha, V.R.; Bansal, K.; Kaushik, R.; Kumria, R.; Trehan, A. Poly-Epsilon-Caprolactone Microspheres and Nanospheres: An Overview. Int. J. Pharm. 2004, 278, 1–23. [Google Scholar] [CrossRef]
  215. Dash, T.K.; Konkimalla, V.B. Polymeric Modification and Its Implication in Drug Delivery: Poly-ε-Caprolactone (PCL) as a Model Polymer. Mol. Pharm. 2012, 9, 2365–2379. [Google Scholar] [CrossRef]
  216. Xiong, Y.; Wu, B.; Guo, X.; Shi, D.; Xia, H.; Xu, H.; Liu, X. Galangin Delivered by Retinoic Acid-Modified Nanoparticles Targeted Hepatic Stellate Cells for the Treatment of Hepatic Fibrosis. RSC Adv. 2023, 13, 10987–11001. [Google Scholar] [CrossRef]
  217. Zhang, Q.; Xu, D.; Guo, Q.; Shan, W.; Yang, J.; Lin, T.; Ye, S.; Zhou, X.; Ge, Y.; Bi, S.; et al. Theranostic Quercetin Nanoparticle for Treatment of Hepatic Fibrosis. Bioconjug. Chem. 2019, 30, 2939–2946. [Google Scholar] [CrossRef] [PubMed]
  218. Ciceu, A.; Balta, C.; Herman, H.; Gharbia, S.; Ignat, S.-R.; Dinescu, S.; Váradi, J.; Fenyvesi, F.; Gyöngyösi, S.; Hermenean, A.; et al. Complexation with Random Methyl-β-Cyclodextrin and (2-Hidroxypropyl)-β-Cyclodextrin Enhances In Vivo Anti-Fibrotic and Anti-Inflammatory Effects of Chrysin via the Inhibition of NF-κB and TGF-Β1/Smad Signaling Pathways and Modulation of Hepatic Pro/Anti-Fibrotic miRNA. Int. J. Mol. Sci. 2021, 22, 1869. [Google Scholar] [CrossRef] [PubMed]
  219. Ignat, S.-R.; Dinescu, S.; Váradi, J.; Fenyvesi, F.; Nguyen, T.L.P.; Ciceu, A.; Hermenean, A.; Costache, M. Complexation with Random Methyl-β-Cyclodextrin and (2-Hydroxypropyl)-β-Cyclodextrin Promotes Chrysin Effect and Potential for Liver Fibrosis Therapy. Materials 2020, 13, 5003. [Google Scholar] [CrossRef] [PubMed]
  220. Ashour, A.A.; El-Kamel, A.H.; Mehanna, R.A.; Mourad, G.; Heikal, L.A. Luteolin-Loaded Exosomes Derived from Bone Marrow Mesenchymal Stem Cells: A Promising Therapy for Liver Fibrosis. Drug Deliv. 2022, 29, 3270–3280. [Google Scholar] [CrossRef]
  221. Morsy, M.A.; Nair, A.B. Prevention of Rat Liver Fibrosis by Selective Targeting of Hepatic Stellate Cells Using Hesperidin Carriers. Int. J. Pharm. 2018, 552, 241–250. [Google Scholar] [CrossRef]
  222. Yang, F.; Hu, S.; Sheng, X.; Liu, Y. Naringenin Loaded Multifunctional Nanoparticles to Enhance the Chemotherapeutic Efficacy in Hepatic Fibrosis. Biomed. Microdevices 2020, 22, 68. [Google Scholar] [CrossRef]
  223. Hermenean, A.; Ardelean, A.; Stan, M.; Hadaruga, N.; Mihali, C.-V.; Costache, M.; Dinischiotu, A. Antioxidant and Hepatoprotective Effects of Naringenin and Its β-Cyclodextrin Formulation in Mice Intoxicated with Carbon Tetrachloride: A Comparative Study. J. Med. Food 2014, 17, 670–677. [Google Scholar] [CrossRef]
  224. Luo, J.; Zhang, Z.; Zeng, Y.; Dong, Y.; Ma, L. Co-Encapsulation of Collagenase Type I and Silibinin in Chondroitin Sulfate Coated Multilayered Nanoparticles for Targeted Treatment of Liver Fibrosis. Carbohydr. Polym. 2021, 263, 117964. [Google Scholar] [CrossRef] [PubMed]
  225. Yang, Q.; Tan, T.; He, Q.; Guo, C.; Chen, D.; Tan, Y.; Feng, J.; Song, X.; Gong, T.; Li, J. Combined Amphiphilic Silybin Meglumine Nanosuspension Effective Against Hepatic Fibrosis in Mice Model. Int. J. Nanomed. 2023, 18, 5197–5211. [Google Scholar] [CrossRef]
  226. Abdullah, A.S.; Sayed, I.E.T.E.; El-Torgoman, A.M.A.; Kalam, A.; Wageh, S.; Kamel, M.A. Green Synthesis of Silymarin-Chitosan Nanoparticles as a New Nano Formulation with Enhanced Anti-Fibrotic Effects against Liver Fibrosis. Int. J. Mol. Sci. 2022, 23, 5420. [Google Scholar] [CrossRef]
  227. Gharbia, S.; Balta, C.; Herman, H.; Rosu, M.; Váradi, J.; Bácskay, I.; Vecsernyés, M.; Gyöngyösi, S.; Fenyvesi, F.; Voicu, S.N.; et al. Enhancement of Silymarin Anti-Fibrotic Effects by Complexation With Hydroxypropyl (HPBCD) and Randomly Methylated (RAMEB) β-Cyclodextrins in a Mouse Model of Liver Fibrosis. Front. Pharmacol. 2018, 9, 883. [Google Scholar] [CrossRef]
  228. Younis, N.; Shaheen, M.A.; Abdallah, M.H. Silymarin-Loaded Eudragit(®) RS100 Nanoparticles Improved the Ability of Silymarin to Resolve Hepatic Fibrosis in Bile Duct Ligated Rats. Biomed. Pharmacother. Biomed. Pharmacother. 2016, 81, 93–103. [Google Scholar] [CrossRef] [PubMed]
  229. Elzoheiry, A.; Ayad, E.; Omar, N.; Elbakry, K.; Hyder, A. Anti-Liver Fibrosis Activity of Curcumin/Chitosan-Coated Green Silver Nanoparticles. Sci. Rep. 2022, 12, 18403. [Google Scholar] [CrossRef] [PubMed]
  230. Negm, M.; Ahmed, N.; Barakat, L. The Role of Curcumin—Chitosan Nanoparticles in the Prevention and Treatment of Liver Fibrosis in Mice. Alfarama J. Basic Appl. Sci. 2022, 3, 8–28. [Google Scholar] [CrossRef]
  231. Zhang, L.; Pan, X.; Xu, L.; Zhang, L.; Huang, H. Mitochondria-Targeted Curcumin Loaded CTPP-PEG-PCL Self-Assembled Micelles for Improving Liver Fibrosis Therapy. RSC Adv. 2021, 11, 5348–5360. [Google Scholar] [CrossRef]
  232. Wang, J.; Pan, W.; Wang, Y.; Lei, W.; Feng, B.; Du, C.; Wang, X.-J. Enhanced Efficacy of Curcumin with Phosphatidylserine-Decorated Nanoparticles in the Treatment of Hepatic Fibrosis. Drug Deliv. 2018, 25, 1–11. [Google Scholar] [CrossRef]
  233. Algandaby, M.M.; Al-Sawahli, M.M.; Ahmed, O.A.A.; Fahmy, U.A.; Abdallah, H.M.; Hattori, M.; Ashour, O.M.; Abdel-Naim, A.B. Curcumin-Zein Nanospheres Improve Liver Targeting and Antifibrotic Activity of Curcumin in Carbon Tetrachloride-Induced Mice Liver Fibrosis. J. Biomed. Nanotechnol. 2016, 12, 1746–1757. [Google Scholar] [CrossRef]
  234. Singh, N.; Khullar, N.; Kakkar, V.; Kaur, I.P. Attenuation of Carbon Tetrachloride-Induced Hepatic Injury with Curcumin-Loaded Solid Lipid Nanoparticles. BioDrugs Clin. Immunother. Biopharm. Gene Ther. 2014, 28, 297–312. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A diagram of signaling pathways modulated by polyphenols and their drug delivery systems in hepatic fibrosis. This figure was created with BioRender.com.
Figure 1. A diagram of signaling pathways modulated by polyphenols and their drug delivery systems in hepatic fibrosis. This figure was created with BioRender.com.
Ijms 25 09346 g001
Figure 2. A diagram illustrating the role of immune cells and immune responses mediated by polyphenols in the resolution of liver fibrosis. This figure was created with BioRender.com.
Figure 2. A diagram illustrating the role of immune cells and immune responses mediated by polyphenols in the resolution of liver fibrosis. This figure was created with BioRender.com.
Ijms 25 09346 g002
Table 2. Pharmacological effects of flavanols in liver fibrosis.
Table 2. Pharmacological effects of flavanols in liver fibrosis.
Bioactive CompoundsCell Lines/
Animal Model
Pharmacological EffectsReference
Epigallocatechin-3-gallate
(EGCG)
Wistar rats
-
AST and AST serum levels
-
prevented deposition of collagen in the hepatic parenchyma
-
↓ total collagen content
-
↓ oxidative stress and lipid peroxidation
-
↓ MDA levels in serum
-
↓ HA and collagen type IV in serum
-
↓ serum OPN levels
-
inhibited activation of HSC cells
-
↓ the expression of α-SMA
-
↓ production of 4-HNE
-
inhibited protein expression of OPN
-
↓ mRNA levels of α-SMA, OPN, Col-1, and Col-3
[51]
HepG2 cells
Sprague-Dawley
rats
-
↑ the expression of miRNAs 221, 181a, and 10b in vitro
-
miRNAs 221, 181a, and 10b ↓ OPN mRNA when administered alone
-
EGCG required the expression of miR-221 to down-regulate OPN protein and its associated fibrogenic properties
-
in vivo, EGCG prevented toxin-induced fibrosis
-
↓ OPN expression
-
↑ miR-221, miR-181a, and miR-10b
[52]
Human hepatic
stellate LX-2 cells
Sprague-Dawley rats
-
ameliorated liver necrosis, inflammation, and fibrosis
-
suppressed the gene expression associated with liver inflammation and fibrogenesis: TNF-α, IL-1β, TGF-β1, MMP-9, α-SMA, and Col1α1
-
suppressed TGF-β1-stimulated expression of Col1α1, MMP-2, MMP-9, TGF-β1, TIMP-1, and α-SMA in LX-2 cells
-
suppressed the phosphorylation of Smad2/3 and Akt in the livers of bile duct ligation (BDL) rats and in TGF-β1-stimulated LX-2 cells
[53]
C57BL/b6 mice
-
↓ CCl4-induced necrosis, cellular injury, and formation of nitrotyrosine
-
serum ALT
-
↓ CCl4-induced collagen accumulation in the liver
-
↓ the expression levels of pro-inflammatory mediators: iNOS, TNF-α, and COX-2
-
attenuated CCl4-modified activity of NF-κB
-
↓ the mRNA expression levels of α-SMA, TGF-β1, pro-collagen-I, MMP-2, and TIMP-1
[54]
Wistar rats
-
↓ serum levels of AST and ALT
-
attenuated hepatic fibrosis
-
↓ HYP in the liver
-
↓ the expression of PDGFRβ and IGF-1R mRNAs
-
inhibited HSC activation
-
↓ the expression of PDGFRβ and α-SMA
[55]
Sprague-Dawley rats
Rat HSCs
-
ameliorated liver injury
-
↓ AST and ALT
-
↓ liver fibrosis and HSC activation in CCl4-treated rats
-
↓ the fibrotic areas
-
↓ HYP level
-
↓ α-SMA
-
↑ hepatic GSH levels
-
↓ the hepatic level of TBARS
-
↓ MMP-2
-
inhibited pro-MMP-2 activation in vitro
-
↓ concanavalin A induced MMP-2 activation through direct inhibition of MT1-MMP activity
-
↓ mRNA expression and protein level of MMP-2
[56]
DihydromyricetinICR mice
-
alleviated TAA-induced chronic hepatic injury
-
↓ the levels of ALT and AST in serum
-
improved TAA-induced liver histopathological damage
-
↓ collagen deposition
-
alleviated TAA-induced hepatotoxicity
-
↑ GSH and SOD
-
↓ MDA
-
↓ TGF-β1 and α-SMA
-
attenuated liver fibrosis by inhibiting TAA-induced inflammatory injury
-
↓ NF-κB
-
inhibited the expression of pro-inflammatory factors TNF-α and IL-1β
-
inhibited hepatocyte apoptosis by regulating the PI3K/Akt signaling pathway
-
↑ p-PI3K and p-AKT
-
↓ Bax, cleaved caspase-9, and cleaved caspase-3
-
↑ Bcl-2 and Bcl-XL
[57]
LX2 and NK92 cells
C57BL/6 J mice
-
ameliorated CCl4-induced liver fibrosis and HSC activation in vivo
-
↓ the serum levels of ALT and AST
-
↓ the expressions of Col1α1, Col1α2, TIMP-1, α-SMA, and desmin
-
↑ MMP-1
-
inhibited TGF-β1-induced HSC activation in vitro
-
suppressed the TGF-β1-induced increase in the viability of LX2 cells
-
↓ the expression of Col-1 and α-SMA proteins in TGF-β1-treated LX2 cells
-
inhibited HSC activation by inducing autophagy
-
↑ the expression of MAP1LC3B2 (LC3B-II) in the liver from CCl4-treated mice
-
↓ SQSTM1
-
↑ Beclin1 and Atg3
-
promoted the activation of hepatic NK cells in mice
-
↑ the frequency of hepatic NK cells
-
↑ the frequency of cells expressing NKG2D
-
↑ the frequency of IFN-γ expression
-
enhanced NK cell-mediated killing of HSCs via IFN-γ in vitro
-
suppressed the TGF-β1-induced decrease in NK92 cell viability
-
↑ the percentage of apoptotic and necrotic cells
-
induced IFN-γ production in NK92 cells through the AhR-NF-κB/STAT3 signaling pathway
-
↑ mRNA expression levels of AhR and CYP1A1
-
↑ P-STAT3/STAT3 ratio
[58]
HesperetinC57BL/6J
mice
HSC-T6 cells
-
alleviated hepatic injury and fibrosis
-
↓ the serum levels of ALT and AST
-
↓ the gene and serum levels of IL-6 and TNF-α
-
↓ histological scores
-
↓ collagen deposition
-
suppressed the formation of ECM
-
suppressed the serum level of LN and HA
-
↓ HYP
-
inhibited the liver fibrosis through down-regulation of the TGF-β1/Smad pathway
-
↓ the protein levels of TGF-β1
-
↓ the phosphorylation of Smad2 and Smad3
-
inhibited the activation and proliferation of HSC-T6 cells, and promoted apoptosis
-
↓ caspase 3 and Bax
-
↑ Bcl-2
-
inhibited the protein level of α-SMA and Col-1
[59]
Hesperetin derivativeLX-2 cells
C57BL/6J
mice
-
exhibited hepatoprotective and anti-fibrotic effect in vivo
-
↓ serum levels of ALT, AST, and ALP
-
inhibited collagen deposition
-
↓ serum levels of TGF-β1 and HA
-
↓ HYP in liver tissues
-
inhibited the up-regulation of liver fibrogenesis markers α-SMA, Col1α1, Col3α1, and TIMP-1 in primary HSCs and suppressed inflammatory responses in primary liver macrophages from hepatic fibrosis mice
-
protected against CCl4-induced inflammatory responses in vivo
-
↓ F4/80+ macrophage infiltration
-
↓ mRNA levels of inflammatory genes (MCP-1, TNF-α, IL-1β, and IL-6)
-
↓ the serum levels of TNF-α and IL-1β
-
suppressed TNF-α and IL-1β protein expression
-
inhibited the phosphorylation and activation of NF-κB-P65
-
suppressed the protein expression of α-SMA and Col1α1 and inhibited mRNA levels of α-SMA, Col1α1, Col3α1, TIMP-1, and PAI-1 in TGF-β1-activated LX-2 cells
-
attenuated CCl4-induced mouse liver fibrogenesis and TGF-β1-induced fibrotic responses in LX-2 cells via Gli-1-dependent mechanisms
-
inhibited mRNA levels and protein expression of Gli-1 and Shh
-
↓ expression of α-SMA and Gli-1 in TGF-β1-activated LX-2 cells
-
promoted apoptosis response in TGF-β1-activated LX-2 cells
-
↑ the level of Bax and cleaved-caspase3
[60]
Hesperetin derivativeC57BL/6J mice
HSC-T6 cells
-
exhibited hepatoprotective and anti-fibrotic effect in vivo
-
↓ the serum level of AST and ALT
-
↓ mRNA and the protein levels of Col1α1 and α-SMA
-
higher concentrations of HD exerted anti-fibrotic effect in TGF-β1-stimulated HSC-T6 cells in vitro
-
↑ Ptch1 expression in TGF-β1-stimulated HSC-T6 cells and CCl4-induced mice
-
↓ the activation of HSCs by targeting Ptch1
-
↓ TGF-β1-activated HSC-T6 cell proliferation
-
induced cell cycle arrest in the S phase by reducing the ratio of the G2/M phase in HSC-T6 cells stimulated with TGF-β1
[61]
Hesperetin derivative-7KunMing mice
HSC-T6 cells
-
attenuates liver fibrosis
-
↓ the adipose degeneration of hepatocytes
-
↓ the immigration of inflammatory cells
-
↓ collagen fibers
-
↓ the mRNA and protein levels of α-SMA and Col1α1
-
↓ the viability of PDGF-BB-induced HSC-T6 cells
-
inhibited the proliferation and the activation of PDGF-BB-treated HSC-T6 cells
-
inhibited the activation and proliferation of PDGF-BB-induced HSC-T6 cells by targeting Wnt/β-catenin signaling pathway
-
↓ the mRNA and protein levels of β-catenin, c-myc, and cyclin D 1
[62]
Hesperetin derivative
(HD-11)
Sprague-Dawley rats
HSC-T6 cells
-
exhibited hepatoprotective effect in vivo
-
↓ the protein levels of α-SMA
-
↓ the levels of ALT and AST
-
alleviated ECM deposition in rats with liver fibrosis
-
↓ the expression of α-SMA and Col1α1 in TGF-β1-induced HSC-T6 cells
-
promoted the expression of PTEN in vivo and in vitro
-
inhibited activated HSC-T6 cell proliferation induced by TGF-β1 via promoting cell cycle arrest
-
suppressed cell proliferation through PTEN/AKT pathway in TGF-β1-induced HSC-T6 cells
-
inhibited the expression of p-AKT in vivo and in vitro
[63]
Hesperetin derivative-16
(HD-16)
LX-2 cells
C57BL/6J
mice
-
alleviated CCl4-induced liver injury and fibrosis in mice
-
↓ the collagen deposition
-
↓ mRNA and protein levels of fibrogenic α-SMA and Col1α1
-
protected against CCl4-induced inflammatory responses in vivo
-
suppressed the levels of TNF-α and IL-1β
-
promoted the levels of IL-10 and IL-13 in serum and liver tissue homogenates
-
attenuated activation of HSCs in vitro
-
inhibited the viability of LX-2 cells
-
suppressed the protein expression of α-SMA and Col1α1 in TGF-β1-activated LX-2 cells
-
↓ the mRNA levels of α-SMA, Col1α1, Col3α1, and TIMP-1
-
suppressed the release of pro-inflammatory factors TNF-α and IL-1β
-
facilitated the release of anti-inflammatory factors IL-10 and IL-13 in TGF-β1-activated LX-2 cells
-
promoted SIRT3 expression and activity in vivo and in vitro
-
↑ the mRNA level of SIRT3, but not SIRT1
-
↑ SIRT3 deacetylase activity
-
↑ SIRT3 protein expression
-
SIRT3 depletion attenuated the anti-fibrotic effects of HD-16
-
↑ the expression of SIRT3 via activation of AMPK/SIRT3 pathway
-
attenuated inflammation and fibrosis through AMPK/SIRT3 pathway in TGF-β1-activated LX-2 cells
[64]
Hesperidin Wistar
rats
-
improved the histological morphology and structure of the liver parenchyma
-
↓ the levels of liver enzymes (AST, ALT, ALP, LDH) and total and direct bilirubin
-
↓ NO, MDA, PC
-
↓ inflammatory gene expression: TGF-β1, iNOS, caspase-3, and α-SMA
-
↑ the levels of total antioxidant capacity and GSH, SOD, and CAT enzyme activity
[65]
Wistar rats
-
prevented serum markers of liver damage
-
↓ ALT and γ-GT
-
exhibited antioxidant activity
-
↓ MDA
-
↑ GSH, the GSH/GSSG ratio, and total glutathione
-
preserved glycogen content
-
↓ collagen deposits
-
exhibited immunomodulatory activity
-
↓ the protein expression of NF-κB p65, TGF-β, CTGF, and IL-1β
[66]
LiquiritigeninC57BL/6J mice
Primary HSCs
-
ameliorated CCl4-induced liver fibrosis
-
↓ collagen deposition and α-SMA level
-
presented an inhibitory role in HSC proliferation
-
suppressed HSC transdifferentiation
-
α-SMA mRNA
-
↓ actin fibers in α-SMA protein
-
inhibited HSC activation
-
↓ the mRNA expression level of Col1α1
-
inhibited liver fibrosis via regulation of miR-181b and PTEN
-
↑ PTEN in vivo and in vitro
-
↓ miR-181b expression via Sp1
[67]
C57BL/6 mice
LX-2 cells
-
protected the liver from CCl4-induced toxicity
-
↓ ALT
-
ameliorated CCl4-induced liver fibrosis
-
↓ the regions of hepatic degeneration and inflammatory cell infiltration
-
↓ α-SMA
-
↓ collagen fibers
-
↓ HNE
-
suppressed TGF-β1/Smad signaling and HSC activation in vitro
-
↓ α-SMA
-
suppressed the TGF-β1-induced gene expression of PAI-1 and MMP-2
-
↓ Smad3 phosphorylation
-
inhibited dysregulation of Smad4 and 7 induced by TGF-β1
-
activated Hippo signaling
-
↑ activation of LATS1 with the induction of YAP phosphorylation
-
ameliorated experimental liver fibrosis by acting on the TGF-β1/Smad and Hippo/Yap pathways
[68]
NaringeninC57BL/6J mice
Human LX2 and L02 cells
-
↓ liver fibrosis in mice
-
↓ the levels of ALT, AST, ALP, ALT, LN, HA, PC-III, and IV-C
-
↓ the expression of inflammatory factors, such as IL-1β, IL-6, IL-18, and TNF-α
-
targeted cGAS in HSCs
-
↓ α-SMA and cGAS
-
inhibited the activation of HSCs in vitro
-
↓ mRNA levels of α-SMA and α1-procollagen
-
↓ α-SMA and α-SMA and Col-1 protein expression
-
↓ the secretion of inflammatory factors in LX2 cells by inhibiting the cGAS-STING pathway
-
↓ the expression of cGAS and STING mRNA
-
inhibited the mRNA levels of IL1-β, IL-6, IL-8, and NF-κB
-
↓ the expression of IRF3 protein
[69]
Wistar rats
-
reversed liver damage, biochemical and oxidative stress marker elevation, and fibrosis
-
↓ NF-κB, IL-6, IL-10, IL-1β
-
reversed collagen accumulation by modulating the synthesis and degradation of ECM
-
↓ HYP and Col-1
-
↓ CTGF, MMP-2, MMP-9, MMP-13, and TIMP-1 protein levels
-
inhibited TGF-β pathway
-
↓ TGF-β, α-SMA
-
↑ Smad7
-
inhibited the pro-fibrogenic JNK-Smad3 pathway
-
↑ JNK activation and Smad3 phosphorylation
[70]
Wistar rats
-
prevented necrosis and cholestasis and improved liver biosynthetic capacity in CCl4-treated rats
-
↓ ALT, AP, γ-GT
-
prevented the depletion of hepatic glycogen
-
prevented the oxidative stress caused by chronic liver damage
-
↓ MDA levels
-
↑ GSH level
-
↑ GPx enzymatic activity
-
prevented inflammation and necrosis in CCl4-treated rats by maintaining normal NF-κB, IL-1, and IL-10 levels
-
↓ collagen deposition
-
preserved the normal activity of MMP-9 and MMP-2 in experimental liver cirrhosis
-
blocked HSC transdifferentiation and Col-1 synthesis by inhibiting pro-fibrogenic proteins
-
↓ TGF-β, α-SMA, CTGF, Col-1, and MMP-13 protein expression
-
↑ Smad7 protein levels
-
inhibited HSC proliferation by blocking the JNK-pSmad3L pathway
-
prevented JNK activation, the elevation of Smad3 protein levels, and phosphorylation in the linker region
[71]
NaringinSprague-Dawley rats
-
protected rat liver from TAA-induced hepatic injury and inflammatory necrosis
-
↓ ALT, AST, TBIL
-
↑ albumin and total protein
-
restored the normal liver architecture
-
suppressed TAA-induced oxidative stress in rat liver
-
↓ MDA
-
↑ CAT, GPx, SOD
-
modulated the cytokine expression
-
↓ IL-6, IL-1β, IFN-γ
-
↑ IL-10
-
attenuated TAA-induced hepatic fibrogenesis
-
↓ collagen deposition
-
↓ α-SMA, TGF-β1, fibronectin
-
induced HSC apoptosis through disruption of p-Akt/Akt pathway
-
suppressed Akt phosphorylation
-
↓ the ratio of p-Akt/Akt
-
↑ caspase-3
[72]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; 4-HNE, 4-hydroxy-2-nonenal; AhR, aryl hydrocarbon receptor; Akt, protein kinase B; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AMPK, AMP-activated protein kinase; AP, alkaline phosphatase; AST, aspartate aminotransferase; Atg3, autophagy-related gene 3; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma-2; Bcl-XL, B-cell lymphoma-extra large; BDL, bile duct-ligated; CAT, catalase; CCl4, carbon tetrachloride; cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase; Col-1, collagen 1; Col1α1, collagen type I alpha 1; Col1α2, collagen type I alpha 2; Col-3, collagen 3; Col3α1, collagen type 3 alpha 1; CTGF, connective tissue growth factor; ECM, extracellular matrix; EGCG, epigallocatechin-3-gallate; eTIMP-1, tissue inhibitor of metalloproteinases 1; Gli-1, glioma-associated oncogene-1; GPx, glutathione peroxidase; GSH, glutathione; GSSG, oxidized glutathione; HA, hyaluronic acid; HD, hesperetin derivative; HD-11, hesperidin derivative; HD-16, hesperetin derivative 16; HNE, hydroxynonenal; HSCs, hepatic stellate cells; HYP, hydroxyproline; IFN-γ, interferon-gamma; IGF-1R, insulin-like growth factor (IGF)-1 receptor; IL-1, interleukin-1; IL-10, interleukin-10; IL-18, interleukin-18; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-8, interleukin-8; iNOS, inducible nitric oxide synthase; IRF3, interferon regulatory factor 3; IV-C, collagen type-IV; JNK, c-Jun N-terminal kinase; LATS1, large tumor suppressor kinase 1; LN, laminin; LX-2, human hepatic stellate cell line; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; miRNAs, microRNAs; MMP-1, matrix metalloproteinase 1; MMP-13, matrix metalloproteinase 13; MMP-2, matrix metalloproteinase 2; MMP-9, matrix metalloproteinase 9; MT1-MMP, membrane type 1-matrix metalloproteinase; NF-κB, nuclear factor kappa B; NK, natural killer; NO, nitric oxide; OPN, osteopontin; PAI-1, plasminogen activator inhibitor-1; p-Akt, phosphorylated protein kinase B; PC, protein carbonyl; PC-III, procollagen type-III; PDGF-BB, platelet-derived growth factor; PDGFRβ, platelet-derived growth factor receptor beta; PI3K, phosphatidylinositol 3-kinase; p-PI3K, phosphorylated phosphatidylinositol-3 kinase; pro-MMP-2, pro-matrix metalloproteinase-2; Ptch1, patched1; PTEN, phosphatase and tension homologue deleted on chromosome ten; Shh, Sonic hedgehog; SIRT3, sirtuin 3; Smad2/3, small mothers against decapentaplegic 2/3; SOD, superoxide dismutase; Sp1, specificity protein 1; SQSTM1, sequestosome 1; STAT3, signal transducer and activator of transcription 3; STING, stimulator of interferon genes; TAA, thioacetamide; TAC, total antioxidant capacity; TBARS, thiobarbituratic acid-reactive substances; TGF-β1, transforming growth factor beta 1; TNF-α, tumor necrosis factor alpha; YAP, Yes-associated protein; α-SMA, alpha smooth muscle actin; γ-GT, γ-glutamyl transpeptidase; TBIL, total bilirubin.
Table 3. Pharmacological effects of flavonols in liver fibrosis.
Table 3. Pharmacological effects of flavonols in liver fibrosis.
Bioactive
Compounds
Cell Lines/
Animal Model
Pharmacological EffectsReference
FisetinAlbino Wistar rats
-
↓ serum levels of ALT, AST, ALP, TBIL, and liver index
-
↑ the levels of ALB and total protein
-
↑ GSH
-
↓ MDA
-
↓ inflammatory biomarkers: TNF-α, IL-6
-
↓ TGF-β1, Col-1, TIMP-1
-
↑ MMP-9
-
suppressed Wnt3a gene expression associated with decreased β-catenin
-
↑ GSK-3β levels
-
↓ the progress of histologic hepatic fibroplasia
-
↓ α-SMA and cyclin D1
[80]
GalanginLX-2 cells
-
inhibited the proliferation of LX-2 cells
-
induced the apoptosis of LX-2 cells
-
↓ the mRNA and protein expression of α-SMA and Col-1
-
inhibited PI3K/Akt signaling
-
↓ the expression of proteins p-PI3K and p-Akt
-
triggered the mitochondrial apoptotic pathway by regulating the expression of the Bcl-2 family of proteins
-
↑ Bax
-
↓ Bcl-2
-
↑ Bax/Bcl-2 ratio
-
inhibited the Wnt/β-catenin pathway
-
↓ the expression of phospho-GSK-3β, total β-catenin, non-phospho(active) β-catenin
[81]
Sprague-Dawley rats
-
↓ hepatic index
-
↓ serum HA and LN levels
-
↓ serum ALT and AST levels
-
↑ serum level of ALB
-
↓ MDA
-
↑ SOD and GSH-Px activities
-
↓ HYP
-
alleviated liver damage
-
↓ steatosis and hepatic lesions
-
↓ α-SMA
-
↓ TGF-β
[82]
IsorhamnetinHSC-T6 cells
-
↓ HSC-T6 activation
-
↓ mRNA expression of ColA1 and α-SMA in the activated HSC-T6 cells
-
suppressed the p-Akt
[83]
C57 mice
-
inhibited liver fibrosis induced by CCl4 and BDL in vivo
-
↓ ALT and AST
-
liver HYP levels
-
↓ collagen deposition
-
inhibited massive macrophage recruitment in liver tissues
-
↓ the mRNA and protein expression of F4/80
-
inhibited HSC activation and ECM deposition
-
↓ α-SMA, Col-1
-
↑ PPAR-γ
-
↑ MMP-2
-
↓ TIMP-1
-
inhibited autophagy in both liver fibrosis mouse models
-
↓ Beclin-1 and LC3
-
↓ TGF-β1-activated Smad3 and p38 MAPK signaling pathways
-
↓ TGF-β
-
↓ p-Smad3 and p-p38 MAPK proteins
[84]
ICR mice
LX-2 cells
-
inhibited HSC activation in vitro
-
↓ PAI-1, α-SMA, Col1α1
-
inhibited TGF-β/Smad signaling pathway
-
inhibited SBE reporter activity
-
↓ Smad3-dependent transcription of SBE reporter genes
-
blocked TGF-β1-induced phosphorylation of Smad2 and Smad3
-
↑ nuclear Nrf2 levels
-
↑ the expression of antioxidant enzymes: GCL and HO-1
-
prevented ROS production in LX-2 cells
-
inhibited liver fibrosis in vivo
-
↓ ALT and AST
-
hepatic degeneration, inflammatory cell infiltration, and collagen accumulation
-
↓ the expression of phosphorylated Smad3, TGF-β1, α-SMA, and PAI-1
-
↓ 4-HNE, nitrotyrosine-positive cells
-
↑ GSH
[85]
KaempferolSprague-Dawley rats
HSC-T6 cells
-
ameliorated CCl4-induced hepatic fibrosis in rats
-
↓ ALT, AST
-
↓ α-SMA, Col-1, ASIC1a, VEGF
-
inhibited the activation and VEGF release of HSCs
-
↓ VEGF, α-SMA, Col-1
-
inhibited ASIC1a protein expression
-
promoted the degradation of ASIC1a protein via the ubiquitination pathway
-
hindered Ca2+ influx
-
inhibited the level of ERS in vitro
-
↓ p-eIF2, ATF-4, VEGF
-
inhibited ERS by targeting ASIC1a in vitro
-
inhibited HSC activation and ERS by suppressing ASIC1a expression
[86]
C57BL/6 J mice
Primary HSCs
-
ameliorated CCl4-induced liver fibrosis in vivo
-
collagen deposition
-
↓ α-SMA
-
↓ ALT, AST, HYP
-
suppressed HSC activation in vitro
-
↓ α-SMA, Col-1
-
promoted HSC activation inhibition via down-regulating Jag1
-
↓ TGF-β, Notch, NF-κB, MAPK/JNK, MAPK/ERK, Wnt
-
↓ Hes1, Hes5
-
↓ Jag1
-
suppressed HSC activation through miR-26b-5p-mediated Jag1 axis
[87]
C57BL/6 mice
Primary HSCs
-
attenuated CCl4-induced liver injury and inflammation
-
↓ the necroinflammatory scores and collagen deposition in the liver tissue
-
↓ ALT, AST
-
↓ LN, HA
-
suppressed HSC collagen synthesis both in vitro and in vivo
-
↓ Col1α1
-
↓ collagen deposition
-
inhibited HSC activation both in vivo and in vitro
-
↓ α-SMA
-
↓ the phosphorylation of Smad2 and Smad3
-
bonded to the ATP-binding site of activin receptor-like kinase 5
[88]
DihydrokaempferolC57BL/6 mice
HepG2 and LX-2 cells
-
attenuated CCl4-induced liver injury and hepatic fibrosis in vivo
-
↓ ALT, AST
-
↓ hepatic index
-
↓ α-SMA, Col-1, Col-3
-
exhibited antioxidant and anti-inflammatory activity and alleviated hepatocyte DNA damage
-
↓ HYP, MDA, H2O2
-
↑ SOD
-
↓ IL-6, IL-1β, TNF-α
-
suppressed apoptosis of hepatocytes in mice and HepG-2 cells
-
inhibited the TGF-β1-Smad2/3 and ERK1/2 signaling pathway
-
↓ TGF-β1, p-Smad2/3, p-ERK1/2, α-SMA, Col-1, collagen 3
-
↓ the phosphorylation levels of NF-κB P65, ASK1, and JNK
-
↓ IL-6, IL-1β, and TNF-α
-
inhibited PARP-1 activity in hepatocytes by binding to Glu-988 and his-862 residues
[89]
MorinLX-2 cells
Wistar rats
-
activated Hippo signaling in cultured LX-2 cells
-
↑ Mst1, Lats1
-
↓ Yap, TAZ
-
suppressed exacerbated TGF-β/Smad signaling in cultured LX-2 cells
-
↓ TGF-β1, p-Smad2/3
-
↓ MMP-2, MMP-9, TIMP-1
-
↑ MMP-1
-
activated Hippo signaling in vivo
-
↑ Mst1, Lats1
-
↓ Yap, TAZ
-
↓ TGF-β signaling and attenuated fibrillar collagen deposition in fibrotic rats
-
↓ TGF-β1, p-Smad2/3
-
↓ MMP-2, MMP-9, TIMP-1
-
↑ MMP-1
-
↑ HYP
-
↓ Col-1, Col-3
[90]
Sprague-Dawley
rats
-
↓ ALT, AST, ALP
-
↓ hyperplasia of fiber tissue
-
↓ inflammatory cells
-
↓ α-SMA, Col-1, Col-3
-
↑ Nrf2, HO-1, NQO1
[91]
Sprague-Dawley rats
Cultured HSCs
-
enhanced the expression of PGC-1α
-
the effect of morin on PGC-1α expression was mediated by AMPK activation
-
enhanced the activity and the expression of SOD2 via AMP/PGC-1α axis
-
PGC-1α inhibited the expression of α1 (I) collagen
[92]
LX-2 cells
Wistar rats
-
inhibited canonical NF-κB signaling in cultured LX-2 cells
-
↓ α-SMA, NF-κB p65
-
induced apoptosis in cultured LX-2 cells
-
↓ Bcl-2
-
↑ Bax, cyt c
-
activated caspase-9 and cleaved caspase-3
-
induced apoptosis
-
attenuated liver fibrosis by suppressing NF-κB signaling in vivo
-
↓ NF-κBp65
-
↑ IκBα
-
↓ α-SMA
-
↓ collagen deposition
-
induced apoptosis in vivo
-
↓ Bcl-2
-
↑ Bax, cyt c
-
activated caspase-9 and cleaved caspase-3
[93]
Albino rats
-
↓ liver index
-
↓ serum levels of ALT, AST, ALP, and TBIL
-
↓ oxidative stress
-
↓ MDA, NO
-
↑ GSH
-
prevented abnormal collagen deposition
-
↓ HYP
-
attenuated the inflammatory and fibrogenic markers
-
↓ TNF-α, iNOS, NF-κB p65
[94]
LX-2 cells
Albino rats
-
↓ the viability of cultured LX-2 cells
-
induced G1 cell cycle arrest in cultured LX-2 cells
-
↓ cyclin D1
-
inhibited Wnt signaling in cultured LX-2 cells
-
↓ GFAP
-
↓ serum levels of AST, ALT, ALP, LDH, and γ-GT in vivo
-
↓ MDA, TBARS
-
↑ SOD, CAT, GPx, GR
-
↓ collagen accumulation
-
inhibited HSC activation in DEN-induced rats
-
↓ GFAP
-
alleviated DEN-induced liver fibrosis through suppression of Wnt signaling
-
↓ GSK-3β, β-catenin, cyclin D1
[95]
Sprague-Dawley rats
-
↓ fibrosis score
-
↓ serum levels of ALT, AST, and bilirubin
-
↓ mRNA and protein levels of Col-1, TGF-β1, and α-SMA
[96]
MyricetinBALB/c mice
CFSC-8B cells
-
inhibited TGF-β1 or PDGF-BB-induced cell proliferation and migration in CFSC-8B cells
-
↓ ECM accumulation induced by TGF-β1 or PDGF-BB in CFSC-8B cells
-
↓ α-SMA, Col-1
-
suppressed TGF-β1-stimulated phosphorylation of Smad2, AKT, ERK, and P38 in CFSC-8B cells
-
↓ PDGF-BB-induced ERK and Akt phosphorylation in CFSC-8B cells
-
attenuated liver fibrosis induced by CCl4 in mice
-
↓ degree of liver fibrosis
-
↓ ALT, AST
-
↓ α-SMA, Col-1
-
inhibited the phosphorylation of Smad2, MAPK, Akt, ERK, JNK, and P38
[97]
MyricitrinBALB/c mice
-
↓ ALT, AST
-
ameliorated centrilobular necrosis
-
↓ hepatic oxidative stress
-
↑ GSH, TAC
-
↓ 4-HNE, TBARS
-
↑ CYP2E1
-
suppressed inflammation
-
↓ COX-2, TNF-α
-
inhibited the pro-fibrotic response
-
↓ TGF-β1, α-SMA
-
improved the regeneration of hepatic tissue
-
↑ PCNA
[98]
QuercetinSprague-Dawley rats
-
ameliorated hepatic dysfunction
-
altered parameters of sphingolipid and pyroptosis pathways
-
↓ serum levels of ALT, AST, and ALP
-
↑ ALB and total protein levels
-
↓ fibrosis and inflammation
-
↓ α-SMA, IL-1β, PPAR-γ, TGF-β1, caspase-1, caspase-3
-
↓ SphK1 and NLRP3
-
↓ MDA
-
↑ TAC, GSH, Nrf2
[99]
Wistar rats
-
↓ HDL cholesterol, LDL cholesterol
-
regulated oxidative stress
-
↑ SOD, GSH
-
↓ the mediators of the Hh signaling and inflammation: Shh, Ihh, Ptch-1, Smo, Hhip, Gli-3, TNF-α, NF-κβ, and Socs-3
-
↓ hepatic lobule degeneration, the intralobular occurrence of inflammatory cells, and hepatocytic necrosis
[100]
BALB/c mice
Raw 264.7 cells
-
attenuated liver inflammation and fibrogenesis in CCl4-treated mice
-
↓ Col-3 or Col-4 protein expression
-
↓ gene expression of Col3α1, Col4α1, CTGF, and TIMP-1
-
inhibited HSC activation
-
inhibited massive macrophage recruitment into the fibrotic livers of CCl4-induced mice
-
↓ F4/80 and CD68
-
inhibited M1 polarization and M1-related inflammatory cytokines in fibrotic livers
-
↓ TNF-α, IL-1β, IL-6, MCP-1
-
attenuated M2 macrophage polarization and expression of immunosuppressive genes in fibrotic livers
-
inhibited macrophage activation and M1 polarization in vitro
-
↓ the mRNA expression of M1 macrophage markers: TNF-α, IL-1β, IL-6, and NOS2
-
inhibited hepatic macrophage activation and suppressed M1 polarization through regulating the expression of Notch1 on macrophages in vivo and in vitro
[101]
C57 mice
-
improved liver fibrosis induced by BDL or CCl4 in vivo
-
↓ ALT, AST
-
↓ HYP
-
inhibited ECM formation and regulated MMP-9 and TIMP-1 expression
-
↓ the levels of serum HA, LN, Col-1, and α-SMA
-
↓ Col-1, α-SMA, and TIMP-1
-
↑ MMP-9
-
attenuated liver damage by suppressing the TGF-β1/Smads signaling pathway
-
↓ TGF-β1
-
↓ p-Smad2 and p-Smad3
-
attenuated liver damage via the PI3K/Akt signaling pathway
-
↑ PI3K, p-Akt
-
inhibited autophagy process in BDL- or CCl4-induced liver fibrosis
-
↓ Beclin-1, LC3
-
↑ P62
[102]
BALB/c mice
RHSteC cells
-
ameliorated liver injury, inflammation, and hepatic fibrogenesis induced by CCl4
-
↓ ALT, AST
-
↓ the mean fibrosis score
-
↓ Col-1
-
inhibited the activation of HSCs in vivo and in vitro
-
↓ α-SMA protein expression
-
↓ the expressions of mRNA encoding collagen-α1 (I), TGF-β1, and α-SMA
-
reduced fibrosis
-
↓ HMGB1, TLR2, TLR4
-
inhibited the cytoplasmic translocation of HMGB1 in hepatocytes of fibrotic livers
-
attenuated CCl4-induced nuclear translocation of NF-κB p65 and inhibited degradation of IκBα
[103]
Wistar rats
-
↓ hepatic markers in serum: AST, ALT, GGT
-
↓ fibrosis index
-
↓ fibrotic area
-
↓ collagen
-
improved gene expression of antioxidant enzymes
-
↑ CAT and SOD
-
inhibited inflammatory markers
-
↓ TNF-α, IL-6, NF-κB
-
↓ mRNA expression of pro-fibrogenic molecules
-
↓ TGF-β1, Col-1, CTGF, TIMP-1
-
↑ anti-fibrogenic molecule gene expression
-
↑ MMP-2, MMP-9
-
α-SMA
-
induced HSC apoptosis
-
↑ the number of apoptotic cells
[104]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; LDH, lactate dehydrogenase; GFAP, glial fibrillary acidic protein; 4-HNE, 4-hydroxy-2-nonenal; Akt, protein kinase B; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AMPK, adenosine monophosphate-activated protein kinase; ASK1, apoptosis signal-regulating kinase-1; AST, aspartate aminotransferase; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma-2; BDL, bile duct-ligated; CAT, catalase; CCl4, carbon tetrachloride; Col-1, collagen 1; Col1α1, collagen type I alpha 1 chain; Col-3, collagen 3; Col3α1, collagen type 3 alpha 1; Col4α1, collagen type 4 alpha 1; COX-2, cyclooxygenase-2; CTGF, connective tissue growth factor; CYP2E1, cytochrome P450 2E1; DEN, diethylnitosamine; ECM, extracellular matrix; ERK, extracellular signal-regulated protein kinase; ERK1/2, extracellular signal-regulated kinases 1/2; GCL, glutamate-cysteineligase; Gli-3, glioma-associated oncogene-3; GPx, glutathione peroxidase; GR, glutathione reductase; GSH, reduced glutathione; GSH-Px, glutathione peroxidase; GSK-3β, glycogen synthase kinase-3 beta; HA, hyaluronic acid; Hes1, hes family bHLH transcription factor 1; Hes5, hes family bHLH transcription factor 5; Hhip, Smo, hedgehog-interacting protein; HMGB1, high-mobility group box 1; HO-1, heme oxygenase-1; HSCs, hepatic stellate cells; HYP, hydroxyproline; Ihh, Indian hedgehog; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; Jag1, Jagged 1; JNK, c-Jun N-terminal kinase; LN, laminin; LX-2, human hepatic stellate cell line; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemotactic protein-1; MDA, malondialdehyde; miRNAs, microRNAs; MMP-1, matrix metalloproteinase 1; MMP-2, matrix metalloproteinase 2; MMP-9, matrix metalloproteinase 9; NF-κB, nuclear factor kappa B; NO, nitric oxide; NOS2, nitric oxide synthase 2; NQO1, quinone oxidoreductase 1; Nrf2, nuclear factor erythroid 2-related factor 2; PAI-1, plasminogen activator inhibitor-1; p-Akt, phosphorylated protein kinase B; PARP-1, poly ADP-ribose polymerase-1; PCNA, proliferating cell nuclear antigen; PDGF-BB, platelet-derived growth factor; PGC-1α, peroxisome proliferator-activated receptor-γ coactivator-1α; PI3K, phosphatidylinositol-3 kinase; PPAR-γ, peroxisome proliferator-activated receptor-γ; p-Akt, phosphorylated protein kinase B; p-PI3K, phosphorylated phosphatidylinositol-3 kinase; p-Smad3, phosphorylated Smad3; Ptch-1, patched homolog 1; RHSteC, primary rat hepatic stellate cell line; ROS, reactive oxygen species; SBE, Smad binding element; Shh, Sonic hedgehog; Smad2/3, small mothers against decapentaplegic 2/3; Socs-3, suppressor of cytokine signaling 3; SOD, superoxide dismutase; SOD-2, superoxide dimutase-2; SphK1, sphingosine kinase-1; TAC, total antioxidant capacity; TBARS, thiobarbituratic acid-reactive substances; TGF-β1, transforming growth factor beta 1; TIMP-1, tissue inhibitor of metalloproteinases 1; TLR2, toll-like receptor 2; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor alpha; VEGF, vascular endothelial growth factor; Yap, Yes-associated protein; α-SMA, alpha smooth muscle actin; γ-GT, γ-glutamyl transpeptidase; TBIL, total bilirubin.
Table 4. Pharmacological effects of flavones in liver fibrosis.
Table 4. Pharmacological effects of flavones in liver fibrosis.
Bioactive CompoundsCell Lines/
Animal Model
Pharmacological EffectsReference
AlpinetinC57BL/6 mice
-
ameliorated the liver injury in mice induced by CCl4 administration
-
↓ HYP, collagen deposition
-
↓ α-SMA, fibronectin, and α1(I) procollagen both at mRNA and protein levels
-
inhibited the expression of COX-2 and iNOS in vivo
-
inhibited ROS
-
↓ MDA
-
↑ GSH, CAT, GSH-Px, SOD
-
inhibited sinusoidal angiogenesis
-
↓ VEGF, VEGFR2, PDGF-βR, HIF-1α
-
inhibited the activation of the NLRP3 inflammasome
-
↓ NLRP3, caspase-1 p20, ASC, mature IL-1β, mature IL-18
-
had protective effects against CCl4-induced liver fibrosis in mice through the activation of Nrf2 signaling
-
↑ protein expression of HO-1, NQO1, GCLM, and GCLC
[108]
ApigeninSwiss albino mice
-
improved hepatic biomarkers
-
↓ ALT, AST, TBIL
-
enhanced oxidative biomarkers
-
↑ GSH, MDA, CAT
-
↓ IL-1β
-
, IL-6, TNF-α
-
↓ VEGF, CD34
[109]
LX-2 cells
C57 mice
-
inhibited proliferation and decreased the viability of LX2 cells
-
protected the liver against fibrosis induced by CCl4 and BDL in mice
-
↓ ALT, AST
-
normalized the structure of portal area
-
↓ fibrous tissue hyperplasia and inflammatory cell infiltration
-
inhibited the activation of HSCs and regulated the balance of TIMP-1 and MMP-2
-
↓ Col-1, α-SMA, IL-1β
-
↑ MMP-2, TIMP-1
-
alleviated autophagy during liver fibrosis
-
↓ Beclin-1 and LC3II/LC3I
-
↑ p62
-
relieved hepatic fibrosis induced by CCl4 and BDL via down-regulating the TGF-β1/Smad3 and p38/PPAR-α pathways
-
↓ TGF-β1, p-Smad3, p-p38
-
↑ PPAR-α
[110]
Wistar rats
-
alleviated CCl4-induced liver fibrosis
-
↓ AST, ALT, ALP, LDH, HYP, TP, TBIL, DB, HA, LN, PCIII, and IV-C
-
↓ the mRNA and protein expression of TGF-β1, α-SMA, HIF-1α, FAK/p-FAK, VEGF, iNOS, and p38 MAPK/p-p38 MAPK
-
↑ ALB, SOD, and GSH-PX
-
↓ MDA
[111]
BaicalinHSC-T6 cells
-
inhibited the proliferation, activation, apoptosis, and cell cycle progression of activated HSC-T6 cells induced by PDGF-BB
-
↓ α-SMA
-
↑ the number of apoptotic cells
-
↑ the number of cells in S phase
-
↓ the number of cells in the G0/G1 phase
-
↓ EMT of activated HSC-T6 cells induced by PDGF-BB
-
inhibited the motility and invasive ability of the induced HST-T6 cells
-
↑ miR-3595
-
miR-3595 regulated the anti-fibrotic effect of baicalin
-
induced miR-3595 expression that modulated the expression levels of ACSL4
-
↓ ACSL4
[112]
Sprague-Dawley
-
exhibited hepatoprotective effects
-
↓ ALT, AST, ALP
-
suppressed hepatic fibrosis
-
↓ HYP
-
↓ the area of hepatic fibrosis
-
suppressed release of inflammatory factors
-
↓ TNF-α, TGF-β1
-
abrogated the expression of TGF-β1-related proteins and increased the expression of PPARγ mRNA and protein
-
↓ the mRNA levels of TGF-β1, Tβ-RI, Tβ-RII, Smad3, α-SMA, and αI(I) collagen
-
↑ the expression of PPARγ mRNA and protein
[113]
Baicalein Sprague-Dawley rats
-
↓ AST, ALT
-
↓ HA, LN, PCIII levels
-
↓ MMP-2, MMP-9
-
↓ PDGF-β receptor protein levels
-
↓ HYP
-
Col-1, Col-3
-
↓ liver fibrosis
[114]
ChrysinCD1 mice
-
↓ fibrotic scores
-
↓ alleviated the ultrastructure of livers
-
↓ Col-1 mRNA expression
-
restored TIMP-1/MMP balance
-
↓ TIMP-1, MMP-2, MMP-3, and MMP-9 mRNA expression
-
↑ MMP-1 mRNA expression
[115]
CD1 mice
-
ameliorated hepatic lesions in vivo
-
inhibited the activation of HSC cells
-
↓ α-SMA, TGF-β1
-
↓ Smad 2, Smad 3
[116]
Diosmin Albino rats
-
improved hepatotoxicity biomarkers of fibrotic rats
-
↓ AST, ALT γ-GT, TBIL
-
↑ ALB, TP serum level
-
mitigated oxidative stress, enhanced antioxidant defense
-
↑ MDA, AOPP, NO levels
-
↑ GPx, CAT, GSH
-
alleviated hepatic inflammatory response
-
↓ iNOS, COX-2, NF-κB p65
-
abrogated pro-fibrogenic cytokines and stimulated PPAR-γ expression
-
↓ α-SMA, TGF-β
-
↑ PPAR-γ
-
↑ CD36, Arg-1
-
represses miR-17-5p/canonical Wnt/β-catenin signaling axis
-
↓ miR-17-5p
-
↓ Wnt genes (Wnt3a, Wnt10b)
-
↓ frizzled receptors (Fzd-1, Fzd-4)
-
depleted β-arrestin 2, prevented GSK-3β phosphorylation, and destabilized cytoplasmic β-catenin in radiation-induced liver fibrosis
-
↓ the protein expression of β-arrestin 2 and p-GSK-3β
-
↓ the protein expression of β-catenin, TCF-4, and cyclin-D1
[117]
EupatilinLX-2 cells
c57BL/6J mice
-
represses the activation of HSCs in vitro
-
↓ the expression of collagens and α-SMA
-
inhibited the proliferation of HSCs
-
↓ cell viability
-
↓ c-Myc, cyclin B1, cyclin D1, CDK6
-
inhibited the EMT of HSCs
-
↓ N-cadherin
-
PAI-1 regulated the EMT and activation of HSCs
-
↓ PAI-1
-
knockdown of PAI-1 using PAI-1-specific shRNA suppressed the levels of Col1α1, α-SMA, and N-cadherin in LX-2 cells
-
inactivated the
-
β-catenin signaling pathway to slow down the EMT progression of HSCs in hepatic fibrosis
-
↓ the protein level of β
-
-catenin and its nuclear translocation, while the transcript level of
-
β-catenin was not affected in LX-2 cells
-
ameliorated hepatic fibrosis in vivo
-
↓ AST
-
↓ Col1α1, fibronectin, α-SMA, PAI-1, N-cadherin,
-
β-catenin
[118]
Isoorientin Wistar rats
-
↓ ALT, AST, IL-6, TNF-α, MDA, MPO, HA, LN, PCIII, HYP
-
↑ SOD, GSH-Px
-
↓ α-SMA, TGF-β1
[119]
Isoorientin-2″-O-α-L-arabinopyranosylWistar rats
-
improved the liver function
-
↓ ALT, AST, ALP, γ-GT
-
↓ IL-6, TNF-α, MPO
-
attenuated oxidative stress
-
↓ MDA
-
↑ SOD, GSH-Px
-
inhibited collagen deposition
-
↓ serum levels of HA, LN, PCIII, and HYP
-
induced HSC apoptosis
-
↓ Bcl-2 mRNA
-
↓ α-SMA, TGF-β1
[120]
IsovitexinC57BL/6 mice
LX2 and JS-1 cells
-
ameliorated CCl4-induced hepatic fibrosis in vivo
-
↓ ALT, AST
-
↑ ALB
-
↓ hepatic inflammation and alleviated liver injury in vivo
-
↓ IL-6, TNF-α
-
↑ IL-10
-
attenuated oxidative stress in vivo
-
↓ ROS level
-
↑ SOD, CAT
-
inhibited HSC activation and ameliorated hepatic fibrosis in vitro
-
↓ the cell viability and colony count in LX2 cells
-
↓ cell proliferation
-
↑ the apoptotic rate of LX2 and JS-1 cells
-
↓ collagen deposition in vitro
-
inhibited the gene and protein expressions of Col-1, Col-3, and α-SMA in LX2 cells
-
inhibited the PI3K-Akt pathway in vivo and in vitro
-
↑ PTEN gene expression
-
↓ PI3K, Akt, and mTOR levels
-
↑ p-PTEN/PTEN ratio
-
↓ p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR
-
activated autophagy
-
↑ the mRNA and protein expression levels of Atg5, Atg7, Beclin-1, and LC3 in vivo
-
enhanced Atg5 and Atg7 protein expression and the LC3 II/I ratio and inhibited P62 protein levels in LX2 and JS-1 cells
-
inhibited miR-21 expression by down-regulating m6A modification
-
↑ pri-miR-21 level
-
↓ the m6A enrichment of pri-miR-21
-
attenuated liver fibrosis by modulating the GSH metabolic pathway
-
↑ GSH and GSH/GSSG levels
-
↓ GSSG levels
[121]
LigustroflavoneC57BL/6J mice
LX-2 cells
-
protected against CCl4-induced liver injury
-
ameliorated CCl4-induced hepatic dysfunction
-
↓ ALT, AST
-
↑ ALB
-
alleviated oxidative injury
-
↓ MDA
-
↑ GSH-Px, SOD
-
ameliorated CCl4-induced histological damage and liver fibrosis in liver injury
-
↓ necrosis of HSCs
-
↓ collagen deposition
-
↓ HYP
-
attenuated the expressions of α-SMA and Col-1 in vivo and in vitro
-
↓ vimentin and ↑ E-cadherin in vivo
-
↓ TGF-β/Smad signaling pathway in vitro
-
↓ Tβ-RI, Tβ-RII, p-Smad2, p-Smad3, and Smad4 in LX-2 cells
[122]
LuteolinHSC-T6 cells
Wistar rats
-
attenuated liver fibrosis in HSC-T6 cells
-
↓ α-SMA and Col1α1
-
↓ the liver fibrosis index level in vivo
-
↓ number of inflammatory cells
-
↓ collagen deposition
-
↓ ALT, AST, ALP
-
↑ CCR1, CD59, NAGA
-
↓ ITIH3, MKI67, KIF23, DNMT1, P4HA3, CCDC80, APOB, FBLN2
[123]
HSC-T6 cells
Sprague-Dawley rats
-
suppressed HSC activation
-
↓ α-SMA
-
↑ E-cadherin
-
inhibited proliferation, migration, collagen synthesis, and the expression of fibrosis-related genes in the activated HSCs and HSC-T6 cells stimulated with or without TGFβ1 or PDGF
-
induced apoptosis of HSC-T6 cells
-
↑ caspase 3 activity
-
↑ p53
-
↓ Bcl-2, cyclin E, and p-CDK-2
-
induced G1 arrest
-
ameliorated liver fibrosis in vivo
-
improved the histological changes
-
↓ ECM accumulation
-
↓ ALT, AST
-
↓ α-SMA, Col-1, Col-3, vimentin, and snail
-
↑ E-cadherin
-
induces HSC apoptosis in liver fibrosis induced by BDL
-
inhibited PDGF- and TGFβ1-simulated phosphorylation of AKT and Smad2/3 pathway in HSCT6 cells
-
↓ PDGF-induced phosphorylation of AKT (Ser 473)
-
↓ mTOR
-
↓ the mTOR substrate p70S6K
-
↓ TGF-β1-induced AKT signaling pathway
-
inhibited TGF-β1-induced Smad2 and Smad3 phosphorylation
-
suppressed the expression of phosphorylated AKT and Smad2 in vivo
-
↓ p-Smad2 and p-AKT
[124]
Balb/c mice
-
ameliorated CCl4-induced hepatic fibrosis
-
↓ ALT, AST, ALP
-
↓ HYP
-
↑ GSH
-
↓ Cu/Zn SOD activity
-
↑ the liver total retinol concentration
-
↓ hepatic lesions and hyaline deposits
-
inhibited HSC activation
-
↓ α-SMA, GFAP
-
↑ MT I/II
-
↑ MMP-9
[125]
LuteolosideHSC-T6 cells
-
regulated the balance of ECM in activated HSC-T6
-
↓ α-SMA, Col-1, TIMP-1/MMP-13 ratio
-
↑ protein expression levels of SIRT1 and ERRα
-
inhibited TLR2/TLR4 pathway
-
↓ TLR2 and TLR4 in activated HSC-T6
-
↓ the protein expressions of MyD88, IRAK1, and IRAK4
-
alleviated the production of inflammatory cytokines in activated HSC-T6
-
↓ NLRP3, ASC, caspase-1, IL-1β
-
regulated TLR2/TLR4 and ECM expressions by targeting SIRT1
-
↑ SIRT1
-
inhibited the protein expression levels of TLR4, TLR2, NLRP3, caspase-1, IL-1β, and α-SMA
-
↓ TLR2, α-SMA
-
attenuated inflammatory response in HSCs induced by inflammatory factors from activated macrophages
-
↓ TLR4, IRAK4, NLRP3, caspase-1, IL-1β, α-SMA
[126]
NobiletinC57/BL6J mice
-
ameliorated fiber deposition and liver injury in CCl4-induced liver fibrosis
-
↓ ALT, AST
-
↓ α-SMA, fibronectin 1, Col1α1
-
↓ the expression of inflammatory cytokines: TNF-α, IL-6, IL-1β
-
↓ TGF-β1
-
↓ NLRP3 and its downstream inflammatory factors IL-18 and IL-1β
-
alleviated hepatocyte EMT in liver fibrosis
-
↓ N-cadherin, vimentin, TIMP-1, TIMP-2
-
↑ MMP-2
-
↓ α-SMA
-
alleviated inflammatory status and reduced the production of ROS
-
induced Hippo/YAP pathway to regulate hepatocyte EMT in liver fibrosis
-
↓ YAP and inhibited its nuclear translocation
-
↓ TEAD2
-
↓ CCN1 and CCN2
-
↑ autophagy and enhanced autophagy flux to ameliorate hepatocyte EMT
-
↑ LC3-II expression
-
↓ p62
-
inhibited EMT in hepatocytes through promoting autophagy flux and thereby facilitating YAP degradation
[127]
Oroxylin ALX-2 cells
ICR mice
-
ameliorated CCl4-induced liver fibrosis in vivo
-
↓ formation of fibrous septa
-
↓ reduced collagen deposition
-
↓ α-SMA, fibronectin, Col-1
-
activated ferritinophagy in HSCs
-
ferritinophagy was mediated by NCOA4
-
↑ NCOA4
-
↓ FTH1
-
↑ LC3 and Beclin1
-
↓ p62
-
promoted iron levels in HSCs and induced lipid ROS accumulation
-
regulated HSC senescence through ferritinophagy
-
↓ p16 and p21
-
↓ cyclin D1, cyclin E1, CDK4, and CDK6 in the presence of siNCOA4
-
inhibited telomerase activity in HSCs to a reduced extent in the presence of NCOA4 interference
-
↓ α-SMA and Col-1
-
the cGAS-STING pathway played an important role in oroxylin A-induced HSC senescence
-
↑ SA-β-Gal
-
inhibited telomerase activity in HSCs to a lesser extent after disrupting the cGAS-STING pathway
-
promoted the secretion of cytokines like IFN-β by the cGAS-STING pathway to regulate ferritinophagy
-
↑ p16 and p21 in LX2 cells
-
cGAS siRNA
-
↓ the expression of NCOA4
-
↓ the expression level of autophagy-related phenotype
-
↓ the content of ROS and iron ions in HSCs
[128]
C57BL/6 mice
Primary HSCs
-
inhibited inflammation in liver fibrosis in vivo
-
↓ NF-κB, α-SMA, IL-1β, IL-6, IL-18, TNF-α, IFN-γ
-
↓ the release of inflammatory factors in HSCs
-
↓ NF-κB, NLRP3, TNF-α, IL-1β, IL-18
-
exerted anti-inflammatory effect by activating autophagy through PI3K/Akt/mTOR signaling
-
inhibited the phosphorylation of PI3K, AKT, and mTOR
-
↑ the conversion of LC3-I to LC3-II
-
↑ the protein expression of Atg5-Atg12 and Beclin1/Atg6
-
↓ the expression of P62
-
the induction of PI3K/Akt/mTOR signaling impaired the oroxylin-A-mediated anti-inflammatory effect in HSCs
-
↓ the expression of TGF-βR and PDGF-βR at mRNA and protein level
-
ROS accumulation played a pivotal function in the inhibition of PI3K/Akt/mTOR signaling and anti-inflammatory effect of oroxylin A in HSCs
-
↓ IL-6, IL-10, IL-18, IL-1β, TNF-α, IFN-γ
[129]
HSC-T6 cells
ICR mice
-
protected mouse livers from CCl4-induced injury and inflammation accompanied by ERS activation
-
↓ ALT, AST, ALP, LDH
-
↓ serum levels of IL-6, IL-8, TNF-α
-
improved morphological changes in liver tissue
-
↓ collagen deposition
-
↑ HA, LN, PCIII, and CIV in serum
-
↓ α-SMA, α1(I) procollagen, fibronectin
-
inhibited HSC proliferation in vitro and induced cell cycle arrest in S phase
-
↓ cyclin A, cyclin E, CDK
-
↑ p15, p21, and p27
-
↓ PDGF-β, TGF-β, EGF receptors
-
induced apoptosis of activated HSCs through caspase activation
-
↓ Bcl-2
-
↑ Bax
-
↑ cleaved-caspase-9, cleaved-caspase-3, the cleaved form of PARP
-
↑ caspase-7 and caspase-8
-
inhibited collagen synthesis and induced collagen degradation in activated HSCs
-
↓ fibronectin, α-SMA, Col-1
-
↑ MMP-9
-
↓ TIMP-2
-
activated the ERS pathway in activated HSCs
-
↑ CHOP and calnexin
-
triggered apoptosis through activation of the ERS pathway
-
activation of ERS signaling inhibited the accumulation of collagen and alleviated inflammatory reactions in vivo
[130]
LX2 and LO2 cells
ICR mice
-
inhibited HSC contraction
-
blocked aerobic glycolysis in HSCs
-
↓ glucose uptake and consumption
-
↓ lactate production
-
↓ the mRNA and protein expression of HK2, PFK1, and PKM2
-
↓ the intracellular ATP levels
-
inhibited LDH-A in HSCs
-
↓ liver injury and fibrosis, and inhibited HSC glycolysis and contraction in vivo
-
↓ the liver/body weight ratio
-
↓ ALT, AST, TBIL, IBIL
-
↓ HA, LN, PC-III, HYP
-
↓ α-SMA, fibronectin, α1(I) procollagen
-
inhibited sinusoidal capillarization and restored the fenestrae of liver sinusoidal endothelial cells in fibrotic mice
-
↓ the mRNA and protein expression of HK2, PFK1, PKM2, and LDH-A
-
↓ α-SMA, MLC2 phosphorylation
[131]
ICR mice
Primary mouse HSCs
-
alleviated CCl4-induced liver injury and fibrogenesis in the mouse model
-
restored the normal morphology of hepatocytes and blocked the formation of fibrous nodules
-
↓ ALT, AST, ALP
-
↓ the collagen accumulation
-
↓ α-SMA, α1 (I) collagen, fibronectin
-
inhibited the expression of PDGF-βR and TGFβ-R1
-
inhibited PGDF-BB-induced HSC activation in vitro
-
↓ α-SMA, desmin, α1(I) collagen, fibronectin
-
↓ TGF-β and TNF-α in activated HSCs
-
promoted autophagy in CCl4-induced mouse liver fibrosis
-
↑ the expression of autophagy makers: LC3-A/B, Atg3, Atg4, Atg5, Beclin1/Atg6, Atg7, Atg9, Atg12, Atg14
-
↓ the expression of autophagy substrate p62
-
promoted autophagy in activated HSCs
-
↑ LC3-B, Atg3, Atg5, Beclin1/Atg6, Atg12, Atg14
-
↓ the expression of autophagy substrate p62
-
↑ autophagic vacuoles
-
inhibition of autophagy by specific inhibitor 3-MA completely abolished oroxylin A-induced anti-fibrosis effect
[132]
TricinLI90 cells
-
inhibited PDGF-BB-induced cell proliferation by blocking cell cycle progression and cell migration in the human HSC line LI90 and culture-activated HSCs
-
inhibited PDGF-BB-induced phosphorylation of p38 MAPK
-
↓ PDGF-BB-stimulated cell proliferation
-
inhibited PDGF-BB-induced phosphorylation of PDGF-Rβ, ERK, and Akt in HSCs
[133]
WogoninC57BL/6 mice
T6, LX-2 cells
-
attenuated liver injury in CCl4-induced mice
-
↓ ALT, AST
-
↓ CCl4-induced liver fibrosis in vivo
-
α-SMA and Col1α1
-
promoted apoptosis of HSCs in vivo
-
↑ cle-caspase3/9
-
attenuated liver fibrosis by promoting apoptosis of HSC-T6 cells and LX-2 cells induced by TGF-β1 in vitro
-
enhanced cle-caspase3 and cle-caspase9 expression and the ratio of Bax/Bcl-2 in T6 cells
[134]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; 3-MA, 3-methyladenine; ACSL4, long-chain-fatty-acid-CoA ligase 4; Akt, protein kinase B; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AOPP, advanced oxidative protein product; AST, aspartate aminotransferase; Atg12, autophagy-related gene 12; Atg14, autophagy-related gene 14; Atg3, autophagy-related gene 3; Atg4, autophagy-related gene 4; Atg5, autophagy-related gene 5; Atg6, autophagy-related gene 6; Atg7, autophagy-related gene 7; Atg9, autophagy-related gene 9; Bcl-2, B-cell lymphoma-2; BDL, bile duct-ligated; CAT, catalase; CCl4, carbon tetrachloride; CD34, vascular endothelial cell antigen; CDK4, cyclin-dependent kinase 4; CDK6, cyclin-dependent kinase 6; cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase; CHOP, CCAAT/enhancer-binding protein (C/EBP) homologous protein; CIV, type IV collagen; cle-caspase 3, cleaved caspase-3; cle-caspase 9, cleaved caspase-9; Col-1, collagen 1; Col1α1, collagen type I alpha 1; Col-3, collagen 3; COX-2, cyclooxygenase-2; DB, direct bilirubin; ECM, extracellular matrix; EGF, epidermal growth factor; EMT, epithelial–mesenchymal transition; ERK, extracellular signal-regulated protein kinase; ERRα, estrogen-related receptor alpha; ERS, endoplasmic reticulum stress; FTH1, ferritin heavy chain 1; GCLC, glutamate-cysteine ligase catalytic subunit; GFAP glial fibrillary acidic protein; GPx, glutathione peroxidase; GSH, glutathione; GSH-Px, glutathione peroxidase; GSK-3β, glycogen synthase kinase-3 beta; GSSG, oxidized glutathione; HA, hyaluronic acid; HIF-1α, hypoxia-inducible factor 1-alpha; HK2, hexose kinase 2; HO-1, heme oxygenase-1; HSCs, hepatic stellate cells; HYP, hydroxyproline; IBIL, indirect bilirubin; IFN-β, interferon-beta; IFN-γ, interferon-gamma; IL-18, interleukin-18; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; IV-C, collagen type-IV; LC3-I, microtubule-associated protein 1 light chain 3 beta-I; LC3-II, microtubule-associated protein 1 light chain 3 beta-II; LDH, lactate dehydrogenase; LDH-A, lactate dehydrogenase-A; LN, laminin; MAPK, mitogen-activated protein kinase; MDA, malondialdehyde; MMP-1, matrix metalloproteinase 1; MMP-13, matrix metalloproteinase 13; MMP-2, matrix metalloproteinase 2; MMP-3, matrix metalloproteinase 3; MMP-9, matrix metalloproteinase 9; MPO, myeloperoxidase; MT I/II, metallothionein; mTOR, mammalian target of rapamycin; NCOA4, nuclear receptor coactivator 4; NLRP3, NLR family pyrin domain containing 3; NO, nitric oxide; NQO1, quinone oxidoreductase 1; Nrf2, nuclear factor erythroid 2-related factor 2; PAI-1, plasminogen activator inhibitor-1; p-Akt, phosphorylated protein kinase B; PARP, poly (ADP-ribose) polymerase; p-CDK-2, phosphorylated cyclin-dependent kinase 2; PCIII, procollagen type III; PDGF, platelet-derived growth factor; PDGF-BB, platelet-derived growth factor; PDGF-β, platelet-derived growth factor beta; PDGF-βR, platelet-derived growth factor receptor beta; PFK1, phosphofructokinase 1; p-GSK-3β, phosphorylated glycogen synthase kinase-3 beta; PI3K, phosphatidylinositol 3-kinase; PKM2, pyruvate kinase type M2; p-mTOR, phosphorylated mammalian target of rapamycin; p-p38, phosphorylated p38; PPAR-α, peroxisome proliferator-activated receptor alpha; PPARγ, peroxisome proliferator-activated receptors γ; p-PI3K, phosphorylated phosphatidylinositol 3-kinase; p-PTEN, phosphorylated phosphatase and tension homologue deleted on chromosome ten; p-Smad2, phosphorylated Smad2; p-Smad3, phosphorylated Smad3; PTEN, phosphatase and tension homologue deleted on chromosome ten; ROS, reactive oxygen species; SIRT1, sirtuin 1; SOD, superoxide dismutase; STING, stimulator of interferon genes; TBIL, total bilirubin; TGF-β1, transforming growth factor beta 1; TGF-βR, transforming growth factor β receptor; TIMP-1, tissue inhibitor of metalloproteinases 1; TIMP-2, tissue inhibitor of metalloproteinases 2; TLR2, toll-like receptor 2; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor alpha; TP, total protein; Tβ-RI, type I receptors for TGF-β; Tβ-RII, type II receptors for TGF-β; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; VEGFR2, vascular endothelial growth factor receptor 2; YAP, Yes-associated protein; α-SMA, alpha smooth muscle actin; γ-GT, γ-glutamyl transpeptidase.
Table 5. Pharmacological effects exerted by flavonones in liver fibrosis.
Table 5. Pharmacological effects exerted by flavonones in liver fibrosis.
Bioactive CompoundsCell Lines/
Animal Model
Pharmacological EffectsReference
AmpelopsinICR mice
Primary mouse HSC cells
-
attenuated CCl4-induced liver dysfunction
-
↓ ALT, AST
-
suppressed CCl4-induced hepatic fibrosis
-
↓ collagen deposition, ECM deposition, and α-SMA
-
inhibited the HSC activation in vitro
-
↓ α-SMA, MMP-9
-
↑ TIMP-1
-
regulated SIRT1/TGF-β1/Smad3 signaling pathway in liver fibrosis
-
↑ SIRT1
-
↓ TGF-β1, p-Smad3
-
regulated SIRT1/TGF-β1/Smad3 signaling pathway in cultured HSCs
-
↑ SIRT1
-
↓ TGF-β1, p-Smad3
-
↓ TGF-β1, p-Smad3, Col-1, and α-SMA in cultured HSCs
-
promoted CCl4-induced autophagy in the livers of mice
-
↑ LC3-II and Beclin-1 in cultured HSCs
-
↓ α-SMA and Col-1 in cultured HSCs
-
PI3K/AKT/mTOR pathway was involved in the anti-fibrotic effect of ampelopsin
-
↓ the phosphorylation of AKT and mTOR in cultured HSCs
[135]
NaringinBalb/c mice
Primary mouse HSCs
-
suppressed activation of primary mouse HSCs
-
reversed activated HSCs to quiescent cells
-
↓ Col-1 and α-SMA
-
the suppressive effect of naringin on HSC activation was enhanced by inhibition of autophagy
-
↑ Beclin1, LC3-II
-
↓ p62
-
↓ Col-1 and α-SMA
-
suppressed activation of HSCs through mTOR pathway
-
↓ phosphor-mTOR (Ser2481, Ser2448), phosphor-p70S6K (Thr389), and phosphor-S6 (Ser235/236)
[136]
Pinocembrin Wistar rats
-
restored liver transaminases and total cholesterol to normal levels
-
ameliorated oxidative stress injury
-
↑ GSH, SOD
-
↓ MDA
-
↑ Nrf2 and HO-1
-
alleviated pro-inflammatory cytokines
-
↓ TNF-α, NF-κB
-
↓ the markers of fibrosis
-
↓ Col-1, α-SMA, TGF-β, p-Smad 2/3
[137]
Human immortalized HSC LX-2 cells
Rat immortalized HSCs
HSC-T6 cells
-
inhibited HSC activation
-
↓ Col-1 and α-SMA in both LX-2 and HSC-T6 cells
-
suppressed HSC activation through inhibiting TGF-β-Smad signaling pathway
-
↓ TGF-β1 production and secretion
-
suppressed Smad phosphorylation and activation
-
PI3K-Akt signaling was involved in the anti-fibrotic effect of PIN
-
↓ the phosphorylation levels of PI3K and Akt in both LX-2 and HSC-T6 cells
-
↑ SIRT3 expression, which deacetylated and activated SOD-2 to enhance ROS clearance, resulting in inactivation of HSCs
-
activated GSK-3β to promote Smad degradation in HSCs
-
↓ p-GSK-3β
-
suppressed HSC activation through SIRT3-TGF-β-Smad signaling pathway
[138]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; Akt, protein kinase B; ALT, alanine aminotransferase; AST, aspartate aminotransferase; CCl4, carbon tetrachloride; Col-1, collagen 1; ECM, extracellular matrix; GSH, glutathione; GSK-3β, glycogen synthase kinase-3 beta; HO-1, heme oxygenase-1; HSCs, hepatic stellate cells; LC3-II, autophagy-related protein microtubule-associated protein light chain three II; MDA, malondialdehyde; MMP-9, matrix metalloproteinase 9; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor kappa B; Nrf2, nuclear factor erythroid 2-related factor 2; p-GSK-3β, phosphorylated glycogen synthase kinase-3 beta; PI3K, phosphatidylinositol-3 kinase; p-Smad 2, phosphorylated Smad 2; p-Smad 2/3, phosphorylated Smad 2/3; p-Smad3, phosphorylated Smad3; ROS, reactive oxygen species; SIRT1, silent mating-type information regulation 2 homolog 1; SIRT3, silent mating-type information regulation 2 homolog 3; SOD, superoxide dismutase; SOD-2, superoxide dimutase-2; TGF-β1, transforming growth factor beta 1; TIMP-1, tissue inhibitor of metalloproteinases 1; TNF-α, tumor necrosis factor alpha; α-SMA, alpha smooth muscle actin.
Table 6. Pharmacological effects of isoflavones in liver fibrosis.
Table 6. Pharmacological effects of isoflavones in liver fibrosis.
Bioactive CompoundsCell Lines/
Animal Model
Pharmacological EffectsReference
CalycosinC57BL/6 mice
-
suppressed the liver index in CCl4-induced hepatic fibrosis mice
-
inhibited oxidative stress
-
↓ AST, ALT, MDA
-
↑ SOD
-
↓ the scores of collagen deposition and liver fibrosis
-
inhibited collagen synthesis
-
↓ α-SMA, Col-1, HYP
-
regulated the expressions of MMP-1 and TIMP-1 in CCl4-induced hepatic fibrosis mice
-
↑ MMP-1
-
↓ TIMP-1
-
↑ the MMP-1/TIMP-1 ratio
-
regulated the expression of estrogen receptors in CCl4-induced hepatic fibrosis mice
-
↑ Erβ
-
activated JAK2-STAT3 pathway
-
↑ p-JAK2/JAK2, p-STAT3/STAT3
[139]
GenisteinLX2 cells
Wistar rats
-
inhibited cell viability and proliferation and induced cell cycle arrest at G0/G1 phase in LX2 cells
-
ameliorated liver injury and the collagen deposition in rats with DMN-induced fibrosis model
-
↓ AST, ALT, ALP, TBA
-
↓ HYP
-
suppressed the expression levels of HSC activation marker α-SMA and Col1A1 in vivo and in vitro
-
↓ the mRNA expression levels of MMP-2/9 and TIMP-1 in vivo
-
regulated inflammatory infiltration and macrophage functional properties in vivo
-
↓ the mRNA expression levels of IL-1β, IL-6, TNF-α, and MCP-1
-
↓ CD68
-
↑ CD163, CD206
-
inhibited the JAK2/STAT3/Socs-3 signaling pathway
-
↓ p-JAK2/JAK2, p-STAT3/STAT3, and Socs-3 protein in vivo and in vitro
[140]
Wistar rats
-
ameliorates D-GalN-induced functional and histological damage
-
inhibited the activation of HSCs
-
↓ α-SMA
-
↓ the accumulation of collagen matrix
-
↓ TGF-β, Col-1, Col-3, HYP
-
↓ nitrotyrosine
-
inhibited TGF-β/Smad signaling
-
↓ TGF-β, Smad 2/3
[141]
SPF-Wistar rats
-
↓ the plasma alcohol concentration
-
attenuated the activity of hepatic enzymes ADH and ALDH
-
↓ AST, ALT
-
↓ the levels of inflammatory mediators: IL-6, TNF-α, MPO, NF-κB
-
↑ SOD, GSH-Px, GSH-Rd, CAT
-
↓ MDA
-
inhibited collagen deposition and ↓ pathological tissue damage: HA, LN, and PCIII
-
attenuated the degree of liver fibrogenesis and the formation of pseudo-lobulus
-
↓ inflammatory cell infiltration
[142]
Wistar rats
-
↓ hepatic collagen
-
improved liver function
-
↓ the amount of collagen fibers and the extent of necrotic areas
-
↑ the capacity of the liver to degrade Col-1 and Matrigel
-
↑ the number of uPA-immunoreactive cells
-
↓ the number of fiber septa in pericentral and perisinusoidal areas
-
↓ activation of HSCs
-
↓ the number of collagen fibers
-
↓ AST, ALT, TBIL
[143]
GlabridinJS1 cells
C57BL/6 mice
-
ameliorated the liver injury in CCl4-treated mouse livers
-
↓ HA, ALT, AST
-
↓ the ratio of liver/body weight of mice
-
inhibited the liver fibrosis in vivo
-
↓ collagen deposition
-
↓ HYP, mRNA, and protein expression of α-SMA, fibronectin, and α1(I)procollagen
-
↑ mRNA and protein expression of PPARγ in vivo
-
inhibited the inflammation and oxidative stress in vivo
-
↓ IL-6, IL-1β, TNF-α
-
↑ IL-10
-
↓ iNOS
-
↓ MDA
-
↑ GSH, T-AOC
-
↓ the cell viability of PDGF-BB-stimulated JS-1 cells
-
inhibited the protein expression of α-SMA, fibronectin, and α1(I)procollagen
-
↑ the expression of PPARγ in stimulated JS-1 cells
-
disruption of PPARγ attenuated the anti-inflammatory and antioxidative stress effects of glabridin in stimulated JS-1 cells
[144]
PuerarinSprague-Dawley rats
-
alleviated inflammation and fibrosis in TAA-induced liver fibrosis in rats
-
↓ histopathological changes and collagen fibers
-
↓ HYP
-
↓ the protein expression levels of Col-1 and fibronectin
-
alleviated TGF-β1 expression and HSC activation
-
↓ TGF-β1, α-SMA
-
inhibited the ERK1/2 signaling pathway
-
↓ p-ERK1/2
-
↓ the ratio of p-ERK1/2 to ERK1/2
[145]
Wistar rats
-
↓ HA level in blood and the HYP level in liver
-
↓ the areas of liver fibrosis in rats
-
↓ the mRNA levels of Col-1, Col-3, Wnt, and β-catenin
-
↓ the protein levels of Wnt1 and β-catenin
[146]
C57BL/6J mice
-
protected against CCl4-induced chronic liver injury
-
↓ the serum ALT, AST, ALB, and TBIL levels
-
attenuates CCl4-induced chronic liver fibrosis
-
↓ α-SMA, Col-1, TGF-β, CTGF
-
inhibited the NF-κB signaling pathway
-
inhibited ROS production and mitochondrial dysfunction in vivo
-
↓ 4-HNE
-
↑ mtDNA number
-
↑ complex I and II activities
-
↓ protein expression of cytosolic cytochrome C
-
↓ caspase 3
-
protected against CCl4-induced liver lesion via modulation on PARP-1
-
↓ PARP-1
[147]
Wistar rats
-
ameliorated the liver metabolic function
-
↓ ALT, AST, TBIL
-
↑ ALB, TP
-
↓ ECM deposition
-
↓ Col-3, LN, HA
-
alleviated the degree of fibrogenesis and ameliorated hepatocellular injury
-
attenuated the accumulation of collagen
-
↓ HYP, PCIII, Col-1
-
regulated the expression of TIMP-1 and MMP-2 at the mRNA level
-
↑ MMP-2
-
↓ TIMP-1
-
↑ PPAR-γ
-
↓ p-PI3K, p-Akt
[148]
Wistar rats
-
↓ ALT, AST, ALB, TP
-
restored the hepatic morphology
-
↓ TNF-α and NF-κB expressions at protein level
-
↑ SOD
-
↓ MDA
-
↓ TGF-βl
-
the mRNA level of iNOS
[149]
Sprague-Dawley rats
HSC-T6 cells
-
enhanced the survival rate of DMN-treated rats
-
↓ the activity of functional liver enzymes
-
↓ ALT, AST
-
reversed the effect of increased collagenic accumulation
-
↓ the serum levels of HA, LN, PCIII, and CIV
-
↓ visceral indices
-
↓ liver index and thymus/spleen indices
-
alleviated liver fibrosis
-
↓ the area of the collagen fibers and the infiltration of inflammatory cells
-
restored the hepatic parenchyma
-
↑ number of hepatocytes
-
↓ collagen deposition
-
↓ HYP, Col-1
-
↓ protein expression levels of TGF-β1, Smad2, Smad3, α-SMA, and TIMP-1
-
↑ protein expression levels of Smad7 and MMP-1
-
inhibited the proliferation of HSC-T6 cells
-
↓ the protein levels of TβRI, Smad2, and Smad3
-
↑ the protein levels of Smad7
[150]
Wistar rats
-
↓ serum ALT, AST
-
↑ apoptosis of activated HSCs
-
↓ Bcl-2 mRNA
[151]
Soy isoflavoneSprague-Dawley rats
-
↓ the amount of collagen fibers and the extent of necrotic areas
-
↓ collagen fibers around the HSCs following high-dose soy isoflavone administration
-
↓ PDGF-BB, TIMP-1
-
↓ α-SMA and TGF-β1
[152]
TectorigeninSprague-Dawley rats
-
improved the histological scores
-
↓ the average severity scores for liver fibrosis
-
↓ ALT, AST
-
↑ ALB, the ratio of albumin to globulin (A/G)
-
↓ HA, LN, and PIIIP
-
↓ HYP
-
↓ MDA
-
↑ SOD and GSH-Px
[153]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; 4-HNE, 4-hydroxy-2-nonenal; A/G, the ratio of albumin to globulin; ADH, alcohol dehydrogenase; ALB, albumin; ALDH, aldehyde dehydrogenase; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; Bcl-2, B-cell lymphoma-2; CAT, catalase; CCl4, carbon tetrachloride; CIV, type IV collagen; Col-1, collagen 1; Col1A1, collagen type I alpha 1; Col-3, collagen 3; CTGF, connective tissue growth factor; D-GalN, D-Galactosamine; DMN, dimethylnitrosamine; ERK1/2, extracellular signal-regulated kinases 1/2; ERα, estrogen receptor α; ERβ, estrogen receptor β; GSH-Px, glutathione peroxidase; GSH-Rd, glutathione reductase; HA, hyaluronic acid; HSCs, hepatic stellate cells; HYP, hydroxyproline; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; JAK2, Janus kinase 2; LN, laminin; MCP-1, monocyte chemotactic protein-1; MDA, malondialdehyde; MMP-1, matrix metalloproteinase 1; MMP-2, matrix metalloproteinase 2; MMP-9, matrix metalloproteinase 9; MPO, myeloperoxidase; mtDNA, mitochondrial DNA; NF-κB, nuclear factor kappa B; p-Akt, phosphorylated protein kinase B; PARP-1, poly (ADP-ribose) polymerase 1; PCIII, type III precollagen; PDGF-BB, platelet-derived growth factor; p-ERK1/2, phosphorylated extracellular signal-regulated kinases 1/2; PI3K, phosphorylated phosphoinositide 3-kinase; PIIIP, procollagen III N-terminal peptide; p-JAK2, phosphorylated Janus kinase 2; PPARγ, peroxisome proliferator-activated receptors γ; p-STAT3, phosphorylated signal transducer and activator of transcription 3; ROS, reactive oxygen species; Socs-3, suppressor of cytokine signaling 3; SOD, superoxide dismutase; STAT3, signal transducer and activator of transcription 3; TAA, thioacetamide; T-AOC, total antioxidant capacity; TBA, total bile acid; TBIL, total bilirubin TGF-β, transforming growth factor beta; TGF-β1, transforming growth factor beta 1; TIMP-1, tissue inhibitor of metalloproteinase-1; TNF-α, tumor necrosis factor alpha; TP, total protein; TβRI, TGF-β receptor type I; uPA, urokinase-type plasminogen activator; α-SMA, alpha smooth muscle actin.
Table 7. Pharmacological effects of anthocyanidins in liver fibrosis.
Table 7. Pharmacological effects of anthocyanidins in liver fibrosis.
Bioactive CompoundsCell Lines/
Animal Model
Pharmacological Effects Reference
Anthocyanins C57BL/6J mice
Mice HSC cell line
-
attenuated liver fibrosis progression in mice
-
↓ ALT and AST
-
inhibited the hepatic inflammatory response and macrophage polarization
-
inhibited the activation and migration of HSCs
-
↓ Col-1 and α-SMA
-
reversed the blocked autophagic flux induced by PDGF or CCl4
-
↑ TFEB
[157]
Anthocyanins from Aronia Melanocarpa ElliotHSC-T6 cells
-
inhibited the proliferation cells and reduced the activation of HSC-T6 cells
-
inhibit the expressions of AST, ALT, ALP, and TBIL
-
improved the expressions of TP and ALB
-
inhibited the secretion of inflammatory cytokines IL-1, IL-6, TNF-α, and COX-2
-
inhibited the expression of TGF-β1, p-Smad2, α-SMA, and Col-1
[158]
Anthocyanins from blueberryC57BL/6J mice
-
prevented CCl4-induced liver damage
-
↓ ALT, AST
-
protected against hepatic oxidative stress induced by CCl4
-
↓ MDA
-
attenuated CCl4-induced hepatic inflammation
-
↓ CXCL2, MIP-2, MCP-1, and IL-1β
-
modulated macrophage subsets in CCl4-induced liver injury
-
attenuated CCl4-induced hepatic fibrosis
-
↓ α-SMA
-
↑ MMP-9
-
↓ PCNA, TIMP-1
-
attenuated HSC activation
-
↓ α-SMA, Col-1, TGFβ-1, and CTGF
[159]
HSC-T6 cells
SD rats
-
inhibited the proliferation of HSC-T6 cells
-
↑ the apoptosis of HSC-T6
-
↓ the protein expression of α-SMA Col-1 and TIMP-1 in vitro
-
↑ acH3K9, acH3K14, and acH3K18
-
↓ ALT, AST
-
↓ HA, CIV
-
↓ the grade of liver fibrosis
[154]
Cyanidin-3-O-β-glucosideC57BL mice
-
exhibited a protective effect on liver injury and fibrosis
-
suppressed the activation of HSCs
-
↓ α-SMA, desmin, MMP-2, and MMP-9
-
regulated oxidative stress and apoptosis in liver
-
↓ MDA
-
↑ GSH, SOD
-
ameliorated apoptosis of hepatic cells
-
↓ ALT, AST
-
suppressed leukocyte recruitment but had no significant effect on the activation of Kupffer cells
-
ameliorated the infiltration of inflammatory cells such as neutrophils and leukocytes
-
suppressed the production of pro-inflammatory cytokines
-
↓ TNF-α, IL-17
-
↑ the level of anti-inflammation cytokine IL-10
-
↓ liver growth factors and MCP-1
-
↓ TGF-β1 and PDGF
[160]
Delphinidin Balb/C mice
-
↓ ALT, AST
-
attenuated oxidative stress
-
↓ HYP
-
↓ hepatic lesions, hepatocyte ballooning, and fatty degeneration
-
↓ collagen deposits
-
↓ α-SMA, TNF-α, TGF-β1
-
↑ MT I/II
-
↑ MMP-9
[161]
MalvidinHSC-T6 cells
-
inhibited HSC-T6 cell proliferation
-
destroyed HSC-T6 cell morphology
-
induced HSC-T6 cell apoptosis
-
triggered HSC-T6 cell ROS generation
-
↑ MDA
-
↓ SOD, GSH/GSSG ratio
-
induced apoptosis through an ERS pathway and a mitochondrial pathway
-
↑ caspase-12, GRP78, CHOP
-
↓ Bcl-2
-
↑ Bax, caspase-3
[162]
PelargonidinC57BL/6J mice
LX-2 cells
-
mitigated the liver weights and relative liver weights compared to the CCl4-treated group in mice
-
ameliorated the hepatic injury markers in vivo
-
↓ ALT, AST, ALP
-
attenuated liver damage and collagen deposition in vivo
-
suppressed α-SMA, Col-1, and TIMP-1 expression in vivo
-
inhibited NLRP3 activation and inflammation in CCl4-induced mice
-
↓ TNF-α, IL-1β, IL-6
-
attenuated oxidative stress through Nrf2 activation in vivo
-
↓ MDA
-
↑ GSH, SOD
-
↑ Nrf2 in the nucleus
-
suppressed the expression of fibrosis markers in TGF-β-activated LX-2 cells
-
↓ α-SMA, COL1A1, TIMP-1, Serpin E1/PAL-1
-
attenuated cellular oxidative stress and inflammation through Nrf2 activation in TGF-β-activated LX-2 cells
-
↑ the Nrf2 protein level in the nucleus
-
↓ the mRNA levels of NLRP3, caspase 1, and IL-1β
[163]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma-2; CCl4, carbon tetrachloride; CHOP, CCAAT/enhancer-binding protein (C/EBP) homologous protein; CIV, type IV collagen; Col-1, collagen 1; Col1A1, collagen type I alpha 1; COX-2, cyclooxygenase-2; CTGF, connective tissue growth factor; CXCL2, chemokine (C-X-C motif) ligand 2; ERS, endoplasmic reticulum stress; GRP78, glucose-regulated protein 78; GSH, glutathione; GSSG, oxidized glutathione; HA, hyaluronic acid; HSCs, hepatic stellate cells; HYP, hydroxyproline; IL-1, interleukin-1; IL-10, interleukin-10; IL-17, interleukin-17; IL-1β, interleukin-1 β; IL-6, interleukin-6; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; MIP-2, macrophage inflammatory protein 2; MMP-2, matrix metalloproteinase 2; MMP-9, matrix metalloproteinase 9; MT I/II, metallothionein; NLRP3, NLR family pyrin domain containing 3; Nrf2, nuclear factor erythroid 2-related factor 2; PCNA, proliferating cell nuclear antigen; PDGF, platelet-derived growth factor; p-Smad2, phosphorylated Smad2; SOD, superoxide dismutase; TBIL, total bilirubin; TFEB, transcription factor EB; TGF-β1, transforming growth factor beta 1; TIMP-1, tissue inhibitor of metalloproteinases 1; TNF-α, tumor necrosis factor alpha; TP, total protein; α-SMA, alpha smooth muscle actin.
Table 8. Pharmacological effects of chalcones in liver fibrosis.
Table 8. Pharmacological effects of chalcones in liver fibrosis.
Class of ChalconesBioactive CompoundsCell Lines/
Animal Model
Pharmacological EffectsReference
ChalconesButeinSprague-Dawley rats
-
↓ ALT, AST
-
↓ HYP
-
↓ MDA
-
↓ alpha1(I) collagen and TIMP-1 mRNA
[164]
IsobavachalconeSPF-grade SD rats
HSC-T6 cells
-
attenuated CCl4-induced liver fibrosis
-
↓ HYP
-
↓ ALT, AST
-
inhibited CCl4-induced HSC activation
-
↓ α-SMA and Col-1
-
attenuated CCl4-induced inflammatory response and oxidative stress
-
↓ TNF-α, IL-1β, IL-6
-
↑ SOD and GSH
-
↓ MDA
-
activated Nrf2/HO-1 pathway in vivo
-
↓ HO-1, NQO-1
-
↓ NF-κB, TNF-α, IL-1β, IL-6
-
inhibited TGF-β1-induced activation of HSC-T6 cells
-
↓ α-SMA, Col-1
-
attenuated inflammatory response and oxidative stress in vitro
-
↓ TNF-α, IL-1β, IL-6
-
↑ SOD, GSH
-
↓ MDA
-
the anti-fibrogenic effects were mediated through the Nrf2/HO-1 pathway
-
↓ NF-κB
-
↓ α-SMA, Col-1
-
the Nrf2 inhibitor (ML385) attenuated the effect of IBC on inhibiting the activation of quiescent HSCs
[165]
Trans-chalconeWistar rats
-
↓ ALT, AST, ALP, IBIL, TBIL
-
↓ tissue collagen level
-
↓ TGF-β1
-
↓ TBARS, hepatic nitrite/nitrate
-
↑ GSH
-
↓ TNF-α
-
↓ inflammatory cell infiltration
-
reverted hepatic necrosis
-
induced regenerative changes in hepatocytes
[166]
Prenylated chalconesXanthohumolBALB/c mice
-
↓ ALT, AST
-
inhibited the pro-inflammatory and pro-fibrogenic hepatic gene expression
-
↓ TNF-α, IL-1α, MCP-1, ICAM-1, NF-κB
-
↓ TGF-β, Col-1, TIMP-1
-
inhibited the activation of HSCs
-
↓ α-SMA
[167]
Primary human hepatocytes (PHHs)
and HSCS
BALB/c mice
-
inhibited the activation of HSCs in vitro
-
↓ α-SMA, Col-1
-
induced apoptosis in activated HSCs in vitro
-
inhibited NF-κB activity and pro-inflammatory gene expression of activated HSCs in vitro
-
↓ NF-κB, MCP-1
-
inhibited pro-inflammatory gene expression of hepatocytes
-
↓ IL-8
-
↓ hepatic inflammation and expression of pro-fibrogenic genes in a murine model of NASH
[168]
DihydrochalconesPhloretin LX-2 cells
C57BJ6 mice
-
↓ the fibrogenic marker expression
-
↓ α-SMA, Col-1, TGFβ-1
-
inhibited cell proliferation, increased apoptosis expression induced by succinate in HSCs
-
inhibited succinate-induced migration and contraction of HSCs
-
↓ TIMP-1, p-MLC2
-
↓ succinate-induced aerobic glycolysis in activated HSCs
-
↓ GLUT-1, LDHA, HK II
-
improved liver fibrosis induced by a sodium succinate diet in vivo
-
↓ ALT, triglycerides
-
↓ α-SMA, Col-1
-
↓ expression of glycolytic markers in the livers of mice with sodium succinate diet-induced liver fibrosis
-
↓ GLUT-1, LDHA
[169]
IcariinMouse primary HSCs
C57BL/6 J mice
-
inhibited EMT, fibrogenesis, cell growth, and migration of HSCs, and promoted the apoptosis of HSCs
-
↑ E-cadherin, GFAP
-
↓ desmin, vimentin, α-SMA, Col1A1
-
↓ the cell growth
-
enhanced the caspase-3 activity of HSCs
-
exerted hepatoprotective effects on CCl4 model in mice
-
↓ collagen, α-SMA
-
↑ E-cadherin, GFAP
-
↓ desmin, vimentin
-
exerted protective effects on liver fibrosis through miR-875-5p
-
↑ miR-875-5p
[170]
Wistar rats
-
↓ ALT, AST
-
↑ ALB
-
↓ the number of inflammatory cells
-
↓ hepatic collagen deposition
-
↓ Col-1α mRNA, HYP
-
presented anti-angiogenic effect
-
↓ mRNA of Ang-1
-
↓ protein expression of VEGF, PDGF-β, and CTGF
-
↓ HMGB1, TGF-β, Beclin-1 mRNA
-
↑ BAMBI mRNA
-
exhibited anti-autophagic activity
-
↑ mTOR and p70S6 kinase expression
-
↓ TLR4, NF-κB, IL-1β, COX-2
[171]
IcaritinWistar rats
HSC-T6 and LX-2
-
inhibited the growth of activated HSCs
-
induced the apoptosis of activated HSCs
-
↑ Bak-1, Bmf, Bax
-
↓ Bcl-2 expression
-
↓ AST, ALT, TP, and A/G in vivo
-
ameliorated the development of liver fibrosis in rats
-
↓ HYP, Col-1
-
↓ number of activated HSCs in vivo
-
↓ α-SMA
[172]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; A/G, the ratio of albumin to globulin; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; Ang-1, angiopoietin-1; AST, aspartate aminotransferase; BAMBI, “bone morphogenetic protein” activin membrane-bound inhibitor; Bax, Bcl-2-associated X protein; CCl4, carbon tetrachloride; Col-1, collagen 1; Col1A1, collagen type I alpha 1; Col-1α, collagen-1 alpha; COX-2, cyclooxygenase-2; CTGF, connective tissue growth factor; EMT, epithelial–mesenchymal transition; GFAP, glial fibrillary acidic protein; GLUT-1, glucose transporter type 1; GSH, glutathione; HK II, mitochondrial-bound hexokinase (HK) II; HMGB1, high-mobility group box 1; HO-1, heme oxygenase-1; HSCs, hepatic stellate cells; HYP, hydroxyproline; IBC, isobavachalcone; IBIL, indirect bilirubin; ICAM-1, intercellular adhesion molecule-1; IL-1α, interleukin-1α; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-8, interleukin-8; LDHA, lactate dehydrogenase A; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; mTOR, mammalian target of rapamycin; NASH, non-alcoholic steatohepatitis; NF-κB, nuclear factor kappa B; NQO-1, NAD(P)H quinone dehydrogenase 1; Nrf2, nuclear factor erythroid 2-related factor 2; PDGF-β, platelet-derived growth factor beta; SOD, superoxide dismutase; TBARS, thiobarbituratic acid-reactive substances; TBIL, total bilirubin; TGF-β1, transforming growth factor beta 1; TIMP-1, tissue inhibitor of metalloproteinases 1; TLR4, toll-like receptors 4; TNF-α, tumor necrosis factor alpha; TP, total protein; VEGF, vascular endothelial growth factor; α-SMA, alpha smooth muscle actin; p-MLC2, phosphorylated myosin light chain 2.
Table 9. The main pharmacological effects exerted by polyphenol-based drug delivery systems in liver fibrosis.
Table 9. The main pharmacological effects exerted by polyphenol-based drug delivery systems in liver fibrosis.
Polyphenolic CompoundsActive IngredientFormulationsExperimental ModelPharmacological EffectsReference
FlavonolsGalanginGalangin delivered by retinoic acid-modified nanoparticlesC57BL/6 mice
-
↓ ALT, AST
-
↓ HYP
-
↓ HA, LN, PCIII, Col IV
-
↓ fibrosis index
[216]
QuercetinTheranostic quercetin nanoparticleBABL-c mice
HSC cells
-
inhibited proliferation of activated HSCs
-
attenuated the fibrotic level of the fibrotic tissue, the appearance of degenerated hepatocytes, and inflammatory cell infiltration
-
↓ HYP
-
↓ ALT, AST
-
↓ the protein expression of α-SMA
[217]
FlavonesChrysinCHR-HPBCD, CHR-RAMEB nanocomplexesCD1 mice
-
alleviated CCl4-induced liver fibrosis collagen deposition, and ultrastructural changes
-
↓ Col-1
-
↓ the score of liver fibrosis
-
inhibited the activation of HSCs
-
↓ α-SMA
-
↓ the TGF-β1/Smad signaling pathway
-
↓ TGF-β1, Smad2, Smad3
-
↑ Smad7
-
↓ the NF-κB-mediated inflammatory pathway
-
↓ NF-κB, TNF-α, IL-6
-
modulated ECM by TIMP-1/MMP balance
-
↓ MMP-2, MMP-3, MMP-9, TIMP-1
-
↑ MMP-1
-
modulated pro-fibrotic and anti-fibrotic miRNA expression
[218]
CHR-HPBCD, CHR-RAMEB complexesHuh7 and LX2 cells
-
the 1:1 CHR-RAMEB pretreatment avoided p65 translocation
-
the 1:2 CHR-RAMEB complex
-
↑ ORAC values
-
improved SOD activity
-
produced the highest stimulation of GPx activity
-
↓ α-SMA expression at lower concentration than CHR-HPBCD
[219]
LuteolinLuteolin-loaded exosomes derived from bone marrow MSCsSprague-Dawley rats
-
exhibited anti-fibrotic activity
-
↓ the % relative liver weight
-
↓ ALT, AST, ALP
-
↓ TNF-α
-
↓ HYP, TGF-β, MMP-2
-
restored the liver architecture
-
↓ collagen deposition
[220]
FlavanonesHesperidinHesperidin-loaded liposomesWistar rats
-
↓ ALT, AST, and ALP
-
↑ ALB
-
improved the liver histological architecture, with restoration of normal hepatocytes and central veins
-
↓ TGF-β1
[221]
NaringeninNaringenin-loaded multifunctional nanoparticlesWistar albino rats
-
↓ CCl4-induced liver fibrosis
-
reversed liver damage
-
↓ ALP, AST, TBIL
-
↓ the pro-inflammatory cytokines TNF-α, IL-1b, and IL-6
-
improved pro-MMP-2 and -MMP-2 activation in the hepatic cells
[222]
Naringenin and its β-cyclodextrin formulationSwiss mice
-
↓ AST, ALT
-
↑ CAT, SOD, GPx
-
↓ MDA
-
improved centrilobular necrosis, steatosis, fibrosis, GSH, and the altered ultrastructure of hepatocytes
[223]
FlavonolignansSilibininCollagenase type I and silibinin in chondroitin sulfate-coated multilayered nanoparticlesKunming mice
-
inhibited the development of hepatic fibrosis
-
↓ the number of highly proliferative HSCs, the extent of collagen fiber deposition, and the number of inflammatory lesions
-
↓ ALT, AST, TBIL
-
↑ ALB
-
↓ HYP
-
↑ GSH-Px
[224]
SilybinCombined amphiphilic silybin meglumine nanosuspensionHepG2, LO2, LX-2, and RAW264.7 cells
Kunming mice
-
inhibited Col-1 secretion in LX-2 cells
-
exerted strong anti-fibrotic effects, by reducing deposition of collagen fibers and ECM along the central venous or portal area
-
prevented hepatic fibrosis by reversing HSC activation
-
↓ α-SMA, Col-1, TGF-β
[225]
SylimarinSilymarin-chitosan nanoparticlesSprague-Dawley rats
-
↑ body weight
-
↓ liver weight and liver index values
-
↓ AST, ALT, ALP, TBIL
-
↑ ALB
-
↓ MDA, TGF-β
-
↑ Nrf2
-
↑ the hepatic expression of protective miRNAs: miR-22, miR-29c, and miR-219a expression
-
↓ TGFβ
-
R1, TGFβ
-
R2, Col3A1
-
improved hepatic architecture
-
↓ the degree of liver fibrosis
-
↓ α-SMA
[226]
Sylimarin–HPBCD and Sylimarin–RAMEB complexesCD1 mice
-
↓ oxidative damage and ↑ antioxidant enzyme activities
-
alleviated CCl4-induced structural changes in liver
-
↓ the score of liver fibrosis
-
↓ NF-κB signaling and inflammatory cytokines: NF-κB p50, NF-κB p65, TNF-α, and IL-6
-
inhibited activation and proliferation of HSCs
-
↓ α-SMA
-
↓ TGF-β1/Smad signaling pathway
-
↓ TGF-β1, Smad 2 and Smad 3
-
↑ Smad 7
-
↓ Col-1 and the deposition of collagen in hepatic tissue
-
↓ MMP-2, MMP-9, TIMP-1
-
↑ MMP-1
[227]
Silymarin-loaded Eudragit® RS100 nanoparticlesWistar albino rats
-
↓ AST, ALT, TBIL
-
↓ MDA
-
↑ GSH
-
↓ α-SMA
-
↓ HYP, TGF-β1, TNF-α
-
↓ the hepatic expression of TIMP-1 and CK-19
-
↑ HGF
-
↑ the hepatic expression of MMP-2
-
↑ MMP-2/TIMP-1 ratio
-
↓ collagen
-
restored hepatic architecture
[228]
CurcuminoidsCurcuminCurcumin/chitosan-coated green-silver nanoparticlesMice
-
↓ hepatic architectural lesions
-
restored the normal amount of collagenous connective tissue
-
modulated Col1A1, α-SMA, PDGFRB, TIMP-1, and ACTB gene expression
-
exerted its inhibitory role through the direct binding to fibrosis mediating proteins such as PDGFRB, TIMP-1, TLR-9, and TGF-β
[229]
Curcumin–chitosan nanoparticlesAlbino mice
-
↓ ALT, AST, ALP
-
↓ AFP, caspase-3
-
↓ MDA
-
↑ GSH, CAT
[230]
Curcumin-loaded CTPP-PEG-PCL self-assembled micellesSprague-Dawley rats and Balb/c mice
-
inhibited the liver fibrosis
-
↓ ALT, AST
-
↓ the relative positive area of liver fibrosis
-
enhanced the bioavailability of curcumin and extended the retention time of curcumin in vivo
-
enhanced the anti-fibrosis effect
[231]
Curcumin with phosphatidylserine-decorated nanoparticlesSprague-Dawley rats
-
↓ ALT, AST
-
↓ the serum levels of pro-inflammatory cytokines TNF-α, IL-1b, and IL-6
-
↓ the degree of liver fibrogenesis
-
↓ Col-1
-
↓ α-SMA
-
↑ HGF expression and activated MMP-2 secretion in the liver
[232]
Curcumin-zein nanospheresSWR
mice
-
↓ AST, ALT, ALP
-
↑ ALB
-
↓ pericentral and periportal fibrosis, inflammatory cells, ballooning degeneration, and steatosis
-
↓ MDA
-
↑ SOD, CAT, GSH-Px
-
↓ Col-1, TIMP-2, TGF-β
-
↑ MMP-2
-
↓ HYP
[233]
Curcumin-loaded solid lipid nanoparticlesWistar rats
-
attenuated histopathological changes
-
improved the state of steatosis, ameliorated inflammation
-
↓ AST, ALT
-
attenuated oxidative stress:
-
↓ MDA
-
↑ SOD
-
↓ TNF-α
[234]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; ACTB, housekeeping beta-actin; AFP, alpha fetoprotein; ALB, albumin; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; CAT, catalase; CCl4, carbon tetrachloride; CHR, chrysin; CK-19, cytokeratin-19; Col IV, collagen IV; Col-1, collagen 1; Col1A1, collagen type I alpha 1; Col3A1, collagen type 3 alpha 1; CTPP-PEG-PCL, 3-carboxypropyl-triphenylphosphonium bromide-poly(ethylene glycol)-poly(3-caprolactone); ECM, extracellular matrix; GPx, glutathione peroxidase; GSH-Px, glutathione peroxidase; HA, hyaluronic acid; HGF, growth factor; HPBCD, hydroxypropyl β-cyclodextrin; HSCs, hepatic stellate cells; HYP, hydroxyproline; IL-1b, interleukin-1b; IL-6, interleukin-6; LN, laminin; LX-2, human hepatic stellate cell line; MDA, malondialdehyde; miRNAs, microRNAs; MMP-1, matrix metalloproteinase 1; MMP-2, matrix metalloproteinase 2; MMP-3, matrix metalloproteinase 3; MMP-9, matrix metalloproteinase 9; MSCs, mesenchymal stem cells; NF-κB, nuclear factor kappa B; Nrf2, nuclear factor erythroid 2-related factor 2; ORAC, oxygen radical absorbance capacity; PCIII, procollagen type III; PDGFRB, platelet-derived growth factor receptor beta; PLGA, poly lactic-co-glycolic acid; pro-MMP-2, pro-matrix metalloproteinase-2; RAMEB, randomly methylated β-cyclodextrin; SOD, superoxide dismutase; TBIL, total bilirubin; TGF-β1, transforming growth factor beta 1; TGFβR1, transforming growth factor beta-receptor 1; TGFβR2, transforming growth factor beta-receptor 2; TIMP-1, tissue inhibitor of metalloproteinases 1; TIMP-2, tissue inhibitor of metalloproteinases 2; TLR-9, toll-like receptor 9; TNF-α, tumor necrosis factor alpha; α-SMA, alpha smooth muscle actin.
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

Ciceu, A.; Fenyvesi, F.; Hermenean, A.; Ardelean, S.; Dumitra, S.; Puticiu, M. Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems. Int. J. Mol. Sci. 2024, 25, 9346. https://doi.org/10.3390/ijms25179346

AMA Style

Ciceu A, Fenyvesi F, Hermenean A, Ardelean S, Dumitra S, Puticiu M. Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems. International Journal of Molecular Sciences. 2024; 25(17):9346. https://doi.org/10.3390/ijms25179346

Chicago/Turabian Style

Ciceu, Alina, Ferenc Fenyvesi, Anca Hermenean, Simona Ardelean, Simona Dumitra, and Monica Puticiu. 2024. "Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems" International Journal of Molecular Sciences 25, no. 17: 9346. https://doi.org/10.3390/ijms25179346

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop