Next Article in Journal
Discovering Potential Compounds for Venous Disease Treatment through Virtual Screening and Network Pharmacology Approach
Next Article in Special Issue
Medicinal Chemistry Strategies for the Modification of Bioactive Natural Products
Previous Article in Journal
Research Progress on Effects of Ginsenoside Rg2 and Rh1 on Nervous System and Related Mechanisms
Previous Article in Special Issue
Profiling the Antidiabetic Potential of Compounds Identified from Fractionated Extracts of Entada africana toward Glucokinase Stimulation: Computational Insight
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanism and Progress of Natural Products in the Treatment of NAFLD-Related Fibrosis

by
Jin-Zhong Li
1,†,
Ning Chen
2,†,
Nan Ma
3,4,* and
Min-Ran Li
1,*
1
Division of Infectious Disease, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
2
General Medicine, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
3
Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
4
JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2023, 28(23), 7936; https://doi.org/10.3390/molecules28237936
Submission received: 30 October 2023 / Revised: 22 November 2023 / Accepted: 29 November 2023 / Published: 4 December 2023

Abstract

:
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most prevalent chronic liver disorder worldwide, with liver fibrosis (LF) serving as a pivotal juncture in NAFLD progression. Natural products have demonstrated substantial antifibrotic properties, ushering in novel avenues for NAFLD treatment. This study provides a comprehensive review of the potential of natural products as antifibrotic agents, including flavonoids, polyphenol compounds, and terpenoids, with specific emphasis on the role of Baicalin in NAFLD-associated fibrosis. Mechanistically, these natural products have exhibited the capacity to target a multitude of signaling pathways, including Hedgehog, Wnt/β-catenin, TGF-β1, and NF-κB. Moreover, they can augment the activities of antioxidant enzymes, inhibit pro-fibrotic factors, and diminish fibrosis markers. In conclusion, this review underscores the considerable potential of natural products in addressing NAFLD-related liver fibrosis through multifaceted mechanisms. Nonetheless, it underscores the imperative need for further clinical investigation to authenticate their effectiveness, offering invaluable insights for future therapeutic advancements in this domain.

1. Introduction

Nonalcoholic fatty liver disease (NAFLD) has become the leading chronic liver disease worldwide and is characterized by the accumulation of more than 5% of fat in liver cells [1]. Nonalcoholic steatohepatitis (NASH) is characterized by inflammation, hepatocyte ballooning, and necrosis and gradually progresses to fibrosis, which is dominated by hepatic stellate cells (HSCs) and the excess accumulation of extracellular matrix (ECM) proteins [2]. In large-scale biopsy-confirmed NAFLD studies, the presence of NASH does not increase the risk of liver-specific incidence or overall mortality. However, the risk of liver-related mortality increases exponentially with the progression of fibrosis, suggesting that liver fibrosis (LF) is the only independent correlate of total mortality in NAFLD patients [3,4].

2. Mechanisms of LF in Fatty Liver Disease

Liver tissue repair involves the concerted actions of various cell types (HSCs, hepatocytes, liver progenitor cells, endothelial cells, and immune cells) [5]. Persistent activation of HSCs and abnormal reprogramming of liver progenitor cells lead to excessive collagen deposition and accumulation during chronic liver injury [6]. The result of fibrosis is the continued and even amplified production of fibrotic cells through preferential recruitment rather than the decomposition of the fibrotic subpopulation [7].
Activated HSCs are the major precursors of activated myofibroblasts, which are the primary source of ECM. Transforming growth factor-beta (TGF-β) and platelet-derived growth factor are two major cytokines that promote the activation and proliferation of HSCs [8]. The activation of myofibroblasts occurs through a common mechanism called epithelial–mesenchymal transition (EMT), wherein quiescent hepatic stellate cells (Q-HSCs) with an epithelial phenotype, liver progenitor cells, bile duct epithelial cells, and sinusoidal endothelial cells transform into mature myofibroblasts with a mesenchymal phenotype [9]. Many other cellular factors, intracellular signaling pathways, and transcription factors are involved in this process. Thus, inhibiting HSC activation is a key factor in preventing the development of LF. HSCs participate in the development of LF through multiple signaling pathways, including TGF-β/Smad, PI3K/Akt, Notch, RAS/ERK, Wnt, Hedgehog, and P38MAPA [10]. Different mechanisms and the integration of multiple signals from hepatocytes, immune cells, and extracellular tissues generate a coherent reparative response.

2.1. Hedgehog (Hh) Signaling

Hedgehog (Hh) is a classic morphogen secreted by ligand-producing cells that diffuses into the extracellular space to regulate Hh-responsive target cells [11]. Hh regulates various biological processes, including proliferation, differentiation, vitality and adult liver regeneration, in Hh receptor-expressing cells [12]. Hh activation stimulates Hh-responsive cells to produce other factors that regulate injury repair. For example, Hh signaling induces HSCs to express TGF-β, CTGF, amphiregulin, jagged and Wnt ligands [13]. Hh stimulates the production of vascular endothelial growth factor by hepatic sinusoidal endothelial cells and induces the expression of osteopontin and chemokines by catheter cells, which recruit various types of immune cells to the damaged liver [14]. These immune cells further secrete multiple cytokines, such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α), exacerbating liver inflammation and damage. Additionally, Hh regulates macrophage polarization, thereby modulating the local balance of inflammatory, anti-inflammatory and fibrotic cytokines [15]. Studies have shown a relationship between Hh signaling pathway activation and liver progenitor cell generation in NAFLD, and the Hh pathway promotes liver progenitor cell proliferation to replace damaged hepatocytes with newly regenerated healthy hepatocytes [16,17]. While Hh signaling is necessary for injured adult livers to regenerate, chronic inflammation and fibrosis are caused when the pathway activation is excessive and/or prolonged [16]. Research has demonstrated that Q-HSCs express high levels of the Hedgehog-interaction protein (Hhip) [18]. After 24 h of culture in a serum-containing matrix, the expression of Hhip decreased by 90%, which was accompanied by the production of the sonic hedgehog (Shh) ligand and activation of the Hh signaling pathway. Activation of the Hh pathway also occurs in the methionine–choline deficient (MCD) diet-induced model of NASH fibrosis, and transgenic mice exhibit greater Hh pathway activation than wild-type mice, resulting in more severe fibrosis [19]. Inhibiting the Hh pathway via drug intervention (such as cyclopamine or GDC-0449, which are both Smo antagonists) can prevent the progression of LF (Figure 1) [12,20].

2.2. TGF-β1

TGF-β is the strongest profibrotic cytokine that is upregulated during LF and a strong inducer of EMT [21,22]. This factor can induce fibrosis by activating HSCs and liver progenitor cells, stimulating ECM synthesis, and inhibiting matrix degradation through the production of tissue inhibitors of metalloproteinases (TIMP)-1 [23]. The TGF-β family contains five subtypes. Generally, TGF-β1 is the most widely and deeply studied subtype in LF [24,25]. Shortly after liver injury, liver parenchymal cells and activated HSCs produce a large amount of TGF-β1. When TGF-β1 binds to the TGF-β Ⅱ receptor in the cell membrane of HSCs, the Ⅱ receptor phosphorylates the Ⅰ receptor, and the activated Ⅰ receptor induces the phosphorylation of Smad2 and Smad3. After being phosphorylated, Smad2, Smad3 and Smad4 form a complex, which is transferred into the nucleus. Smad3 binds to the promoter region of collagen, stimulating its transcription and producing a large amount of ECM [26]. However, TGF-β1 also plays an important physiological role in many aspects of cell proliferation, development, apoptosis and other biological processes. It is not feasible to widely target TGF-β1 as an antifibrotic strategy because of its functional diversity and pleiotropic effects. In a recent study, the inhibition of TGF-β type I receptor (ALK5) was coupled to mannose 6-phosphate human serum albumin (M6PHSA), and M6PHSA specifically delivered the ALK5 inhibitor to HSCs [27]. And, HSC αv integrin depletion inhibits fibrosis by reducing TGF-β activation (Figure 2) [28].

2.3. Wnt/β-Catenin

The Wnt/β-catenin signaling pathway is responsible for normal development, regeneration, metabolic partitioning and hepatobiliary development in the liver and maintains liver homeostasis [29]. The results show that the Wnt signaling pathway is closely related to the activation and proliferation of HSCs and LF [30]. Canonical Wnt signaling depends on β-catenin. The binding of Wnt ligands induces spatial interactions between cell surface receptors and their coreceptors, forming ternary complexes [31]. Ligand binding causes receptor conformational changes and then activates the downstream Wnt signaling pathway. After being activated, β-catenin is transferred to the nucleus, triggering the expression of Wnt target genes (Figure 3) [32]. A study involving rat HSC lines proved that β-catenin was highly expressed in the nucleus of activated HSCs, and siRNA-mediated knockout of β-catenin could inhibit HSC proliferation, increase apoptosis and inhibit the synthesis of type I and type III collagen [33], which indicated that β-catenin siRNA alleviated LF by controlling the activation of HSCs [34].

3. Natural Products with Potential Activity

At present, nearly half of the drugs used in liver treatment are natural products or derivatives of natural products [35,36,37]. Many natural products, which are mainly derived from plants, contain many active ingredients [38,39]. Because of their relative applicability, effectiveness and safety, natural drugs are now growing globally [40]. Recent investigations on functional foods show that many natural preparations have protective and therapeutic effects on the liver. Herbs and nutritional supplements also make them beneficial to the liver [41]. The natural product exerts anti-fibrosis effects by blocking signaling pathways such as Hedgehog, Wnt/β-catenin, TGF-β1, and NF-κB (Table 1).

3.1. Flavonoids

Flavonoids are polyphenols with a C3-C6-C3 core structure. Because phenolic hydroxyl groups are connected to different functional groups, they exhibit different biological activities. Most flavonoids protect the liver, inhibit oxidation, inflammation, diabetes, and cardiovascular disease and have immunomodulatory effects [83,84].

3.1.1. Baicalin

Baicalin (Figure 4A) is a kind of flavonoid compound extracted from the dried root of Scutellaria baicalensis. A large number of in vitro and in vivo studies show that baicalin has different pharmacological properties, including antioxidant, anti-inflammatory and hepatoprotective properties. These biological properties can be attributed to the fact that baicalin can target multiple pathways and bind to multiple signaling molecules [85]. A mouse model of NAFLD induced by an MCD diet showed that baicalin treatment significantly inhibited liver inflammation induced by MCD. This outcome was also related to decreases in serum TNF-α, IL-1β and monocyte chemoattractant protein-1 (MCP-1) levels, the inhibition of macrophage influx, and the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In addition, baicalin inhibits hepatic fibrosis by inhibiting α-smooth muscle actin (α-SMA), TGF-β1 and COL1A1 production [42]. A recent study showed that baicalin (200 mg/kg) could reduce the expression of fibrosis-related genes such as α-SMA, connective tissue growth factor and inflammatory factors such as TNF-α, macrophage inflammatory protein-1α, IL-1β and macrophage inflammatory protein-2, thus effectively inhibiting LF. In vitro studies also showed that baicalin could inhibit the activation of HSCs and downregulate the expression of α-SMA, fibronectin, TIMP1 and collagen 1 [43].
Many mechanisms of these therapeutic effects have been revealed. For example, baicalin decreased the expression of miR-3595, increased the activity of long-chain fatty acid coenzyme A ligase 4, and significantly inhibited the activity of HSCs, resulting in a decrease in fibrosis in HSC-T6 hepatocytes caused by platelet-derived growth factor [86]. In addition, baicalin inhibits PPAR-γ through Wnt signaling, which can reduce the activity of HSCs [87,88]. Baicalin alleviates LF induced by carbon tetrachloride (CCl4) in mice by regulating TGF-β1, hydroxyproline, procollagen type III, laminin (LN) and hyaluronic acid (HA). Baicalin can also reduce LF by inhibiting the activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx) [44].

3.1.2. Galangin

Galangin (GA, Figure 4B) (3,5,7-trihydroxyflavone) is a natural polyphenol compound extracted from the rhizome of Alpinia officinarum. Studies have reported various pharmacological properties of GA, such as the inhibition of inflammation, oxidation, tumors, allergy and Alzheimer’s disease [89,90]. LX-2 cells were selected as the LF model in vitro; GA effectively inhibited the proliferation of LX-2 cells and induced apoptosis in a dose-dependent manner, and the mRNA and protein expression levels of α-SMA and collagen I were significantly downregulated. Further studies showed that GA significantly reversed LF and induced apoptosis in HSCs by blocking the PI3K/Akt, Bax/Bcl-2 and Wnt pathways [45,91]. After 12 weeks of GA treatment by gavage, the levels of HA, adhesion protein, serum total protein, albumin, alanine aminotransferase and aspartate aminotransferase were significantly reduced in a CCl4-induced rat model, which indicated that the reduction in oxidative stress levels could improve the state of LF. A study on the pathological mechanism showed that GA could significantly reduce the levels of malondialdehyde (MDA) and hydroxyproline and increase the activities of SOD and catalase in hepatic tissue [46]. GA can improve LF by scavenging free radicals, reducing lipid peroxidation, and inhibiting the activation and proliferation of HSCs. However, oral GA administration is associated with low bioavailability due to its water solubility and hydrophobicity, which limits its clinical use. Retinoic acid-modified acrylic nanoparticles were used to encapsulate GA, which significantly controlled its release and HSC targeting to improve the antifibrotic effect of GA on the liver [92].

3.1.3. Silymarin

Silymarin (Figure 4C) is a polyphenol flavonoid antioxidant derived from plants that are mainly composed of flavonoid lignans, flavonoids and polyphenol molecules, and silybin is the most common and bioactive [93,94]. Related research shows that silymarin can protect the liver by reducing free radicals and lipid peroxidation [95]. After 10 days of 100 mg/kg silymarin treatment by gavage in a rat model induced by CCl4, MDA levels decreased and glutathione levels increased, indicating that silymarin has a significant antioxidant capacity and can protect the liver from damage. Studies have shown that silymarin can protect against NASH induced by an MCD diet by interfering with the inflammatory cytokine TNF-α, inhibiting the activation of HSCs, and reducing the expression of α1-procollagen in HSCs [47]. In addition, silymarin ameliorated LF by reducing the level of connective tissue growth factor in rats [48]. In an in vitro model of human LF, silybin dose-dependently inhibited the production of procollagen induced by growth factors in activated HSCs, resulting in antifibrotic effects [96]. The antifibrotic effect of silymarin has also been confirmed in humans. In a randomized, double-blind, placebo-controlled trial, compared with those in the placebo group, more patients in the silymarin 2100 mg/day group had measurable improvements in fibrosis. Noninvasive fibrosis indices (AST/platelet ratio index, fibrosis-4 score and NAFLD fibrosis score) in the silymarin group were significantly improved. In addition, there were more patients with fibrosis improvement or remission in the silymarin group, and silymarin changed liver stiffness favorably (the change in liver hardness was −0.7 vs. 6.0 kPa), but there was no significant difference between the two groups [97].

3.2. Polyphenol Compounds

Natural polyphenols are secondary metabolites of plants and have important roles in the prevention and treatment of many diseases, including cancer, cardiovascular disease, diabetes, aging and neurodegenerative diseases [98]. Studies have shown that polyphenols have a variety of pharmacological effects on oxidative stress, lipid metabolism, insulin resistance and inflammation, which are the most important pathological processes in the etiology of liver disease [98,99].

3.2.1. Curcumin

Curcumin (Figure 5A) is a polyphenol compound isolated from Curcuma longa that contains many functional antioxidant groups, including β-diketone groups, carbon–carbon double bonds and phenyl rings. Due to its ability to eliminate lipid free radicals in cell membranes and convert them to phenoxyl free radicals, curcumin is considered to be a strong fat-soluble antioxidant [100]. It was found that curcumin (200 mg/kg/day for 3 weeks) protected against NASH induced by CCl4, and decreases in lipid accumulation and MDA deposition in histopathology were observed [49]. Curcumin also inhibited the occurrence and progression of LF in NASH mice induced by an MCD diet, which was characterized by a decrease in the secretion of TIMP-1 and the inhibition of 8-OH-deoxyguanosine-mediated liver oxidative stress in HSCs [50]. In addition, the protein expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) in curcumin-treated rats increased significantly, suggesting that the prevention/improvement of NASH may be related to the activation of NRF2 [51]. An innovative mouse model of NASH and hepatocellular carcinoma (HCC) was used to study the potential mechanism by which curcumin can treat NASH. The results showed that curcumin improved hepatic steatosis and fibrosis in mice and caused a significant decrease in fibrosis biomarkers. The most important discovery was that curcumin inhibited the translocation of high mobility group protein B1 (HMGB1)-NF-κB, thus preventing NASH progression and hepatic injury [52].
Curcumin has been proven to have antifibrotic effects in various LF models, and its mechanism includes (1) inhibiting TGF-β/Smad signal transduction by activating autophagy, effectively reducing the occurrence of EMT in hepatocytes and inhibiting the production of ECM [101]; (2) reducing the phosphorylation of JNK and Smad3, inhibiting the activation of HSCs and inducing their apoptosis [102,103]; (3) decreasing the expression of HIF-1α through the ERK pathway [104]; (4) reversing LF by downregulating DNMT1, α-SMA and COL1A1 and demethylating key genes [105]; (5) inhibiting the activation of Kupffer cells (KCs) and reducing the secretion of chemokines to reduce the infiltration of monocytes [106]; and (6) targeting HSCs through a PPAR-γ activation-dependent mechanism to weaken sinus angiogenesis in LF [107].

3.2.2. Resveratrol

Resveratrol (3,5,4′-trihydroxy-trans-stilbene, Figure 5B), which is a nonflavonoid phenol first isolated from Veratrum grandiflorum, has antiaging, anticancer, anti-inflammatory and antioxidant effects [108]. Resveratrol plays an interesting role in regulating the formation and deposition of new fibers. Resveratrol treatment by gavage reduced portal vein pressure and improved hepatic endothelial function in cirrhotic rats [109,110]. After resveratrol (10 mg/kg/day and 20 mg/kg/day) administration for 2 weeks, portal vein pressure decreased in cirrhotic rats, which was related to a decrease in thromboxane A2 and an increase in endothelial NO synthesis, which in turn was associated with a significant decrease in LF [53,54]. Resveratrol can prevent LF in various animal models, and the thickening and deposition of collagen fibers are significantly reduced in rats that are pretreated with resveratrol. Supplementation with resveratrol before dimethylnitrosamine (DMN) induction can significantly improve fibrosis, vasodilation, congestion, wall thickening, duct proliferation and necrosis [55]. The mechanism may involve decreasing the levels of MDA and the quantity of reduced glutathione (GSH), increasing the levels of GPx and SOD, and inhibiting the mRNA expression of inflammatory mediators, including inducible NO, TNF-α and IL-1β, and hypoxia-inducible factor-1α (HIF-1α) [111,112,113,114]. A recent study showed that resveratrol activated the PTEN/PI3K/AKT axis to alleviate LF in rats, and autophagy was enhanced after RSV treatment. In addition, resveratrol reversed the inhibitory effect of miR-20a on PTEN expression, decreased the expression of miR-20a, and promoted the protein expression of PTEN, PI3K and p-AKT, thus weakening LF [54].

3.2.3. Kaempferol

Kaempferol (Figure 5C) is the most common glycoside flavonoid widely distributed in foods, beverages and the plant kingdom [115]. Kaempferol and its glycosylated derivatives have cardioprotective, neuroprotective, anti-inflammatory, antidiabetic, antioxidant, antitumor and anticancer effects [116,117]. Kaempferol has recently attracted much attention because of its multitarget characteristics and its potential for preventing and treating NAFLD [118]. In oleic acid-induced HepG2 cells and HFD-induced rats, kaempferol inhibited the NF-κB pathway and significantly reduced the levels of TNF-α and IL-6, thus significantly improving LF [119]. Some studies have shown that kaempferol significantly improves the number of inflammatory cells in the necrotic area of the hepatic lobule and central venules and reduces the levels of LN and HA. Protein analysis showed that kaempferol inhibited the development of LF by inhibiting the activation of HSCs. The Western blot results showed that kaempferol downregulated TGF-β1-induced α-SMA and the phosphorylation of Smad2/3 in a dose-dependent manner. In addition, kaempferol selectively binds to ALK5 and further downregulates the TGF-β1/Smad pathway [56]. Xing Wan et al. conducted similar research and reached a similar conclusion, but the new discovery here was that kaempferol reduced liver inflammation and fibrosis by inhibiting the TNF-α/NF-κB pathway [57].

3.3. Terpenoids

There is increasing evidence that terpenoids can effectively inhibit the progression of NAFLD and play a therapeutic role in different stages of the disease, including improving lipid metabolism, inhibiting oxidative stress, inhibiting inflammation and preventing fibrosis [120].

3.3.1. Geraniol

Geraniol (Figure 6A) is an acyclic isoprene monoterpene isolated from the essential oils of aromatic plants. In recent years, increasing evidence has shown that geraniol has an important antioxidant effect [121]. It has been reported that geraniol was effective in lowering the risk of hyperlipidemia in atherogenic diet-fed hamsters by improving endothelial function and preventing LF [122,123]. It was found that geraniol reduced the activity of myeloperoxidase and the protein expression of TNF-α and IL-6 in the livers of MCD-fed rats and significantly reduced the levels of COL1A1 and α-SMA [58]. In addition, geraniol increased the activities of GSH, SOD, catalase, glutathione reductase, glutathione-S-transferase (GST) and GSH-Px in the livers of rats and exerted antioxidant and anti-inflammatory effects [59].

3.3.2. Acanthoic Acid

Acantholic acid (AA, Figure 6B) is a diterpene isolated from the root of Eleutherococcus senticosus. The treatment of liver diseases is an important aspect of the use of AA. The value of AA in liver diseases has been widely explored. For example, AA regulated LF and lipid deposition in HSC-T4 cells stimulated by ethanol combined with LPS by reducing lipoprotein2/4 through the TLR6 and IRAK1 signaling pathways [60]. AA also increased antioxidant enzymes and significantly reduced histopathological changes and the expression of caspase-3 and HIF-1α [61]. AA may be an attractive candidate for the treatment of NAFLD. Studies have shown that AA activates the farnesoid X receptor (FXR) and liver X receptor (LXR) signaling pathways and promotes the expression of the AMPK-SIRT1 signaling pathway, which plays a role in regulating fat metabolism and improving fibrosis [62].

3.3.3. Ginsenoside

Ginsenosides (Figure 6C) are a series of glycosylated triterpenoids isolated and identified from the dry root and rhizome of Panax ginseng. Ginsenosides Rb1, Rb2, Rg1, Rg2, Rh1 and Mc1 have been proven to have protective effects on the liver [124]. Ginsenoside Rg1 is a phytochemical with biological activity, and it is the most commonly reported ginsenoside in the treatment of NAFLD [125]. Hou et al. showed that ginsenoside Rb1 alleviated LF by inhibiting fat deposition and the secretion of prostaglandin E2 and TIMP-1 [126]. Han et al. proposed that ginsenoside 25-OCH3-PPD could protect against LF and inflammation by activating the LXR signaling pathway in thioacetamide-induced mice. Compound K (CK) and ginsenoside Rh1 are the main metabolites of Panax notoginseng saponins (PNS) [63]. Previous studies have shown that PNS inhibits the activation of HSCs and LF by downregulating the expression of TIMP-1, collagen (PC)-I, PC-III and TGF-β1 [64]. A recent study showed that CK or Rh1 alone or in combination significantly improved liver damage caused by an HFD. Histologically, CK and Rh1 significantly reversed hepatocyte injury and LF induced by the HFD. In vitro, CK or Rh1 alone or in combination significantly induced apoptosis in HSC-T6 cells and inhibited cell proliferation and activation. In addition, CK and Rh1 alone or in combination inhibited the expression of TIMP-1, PC-I and PC-III. These results showed that CK and Rh1 had positive effects on NAFLD through antifibrotic and hepatoprotective activities [65].

3.3.4. Corosolic Acid

Corosolic acid (CA, Figure 6D), a natural pentacyclic triterpenoid extracted from Lagerstroemia speciosa L. leaf, has efficacy in producing antidiabetic, anti-obesity, anti-inflammatory, antihyperlipidemic and antiviral effects [127,128]. In mouse models of NASH induced by HFD and CCl4, CA inhibits the transcription of profibrotic markers (including α-SMA, PC-1 and TIMP-1) and proinflammatory cytokines (including TNF-α, IL-1β, caspase-1 and IL-6) related to LF. CA also inhibits NF-κB translocation and the TGF-β1/Smad2 and AMPK pathways. In addition, CA decreased the expression of α-SMA and PC-1 and the phosphorylation level of Smad2 in LX2 cells treated with TGF-β1. The results showed that CA could improve fibrosis associated with NASH by regulating the TGF-β1/Smad2, NF-κB and AMPK signaling pathways [66].

3.3.5. Lycopene

Lycopene (Figure 6E) is a lipophilic carotenoid hydrocarbon pigment found in red, pink, and orange fruit and vegetables [129]. Lycopene prevented the development of NASH induced by lipotoxicity by reducing oxidative stress in mice. Lycopene reduced the activity of peritoneal macrophages induced by LPS-/IFN-γ-/TNF-α and the expression of fibrotic genes in HSCs induced by TGF-β1 [67]. Lycopene has hepatoprotective and antioxidant effects in the context of NAFLD, and downregulating the expression of TNF-α and CYP2E1 may be one of the mechanisms [68]. Kitade et al. showed that lycopene improved LF by inhibiting the activity of HSCs [69]. In the same context, lycopene inhibited the activation of HSCs and regulated cell lipid storage by promoting the expression of PPAR-α and retinoid X receptor-β and -γ [130].

3.3.6. Astaxanthin

Astaxanthin (ASTX, Figure 6F) is a kind of ketocarotene belonging to the tetraterpenes and has the strongest ability to absorb oxygen free radicals. Its antioxidant activity is higher than that of carotene, which is 1000 times that of vitamin E [131]. Natural astaxanthin is extracted from the green algae Haematococcus pluvialis, the red yeast Phaffia rhodozyma as well as crustacean byproducts [70]. Studies have shown that ASTX plays an important role in the prevention and treatment of LF, NAFLD, liver cancer and liver injury caused by drugs and ischemia and has therapeutic potential in both healthy and diseased livers [71]. ASTX inhibited the activation of the Smad3 pathway in HSCs by blocking the TGF-β1 signaling pathway [132]. In addition, ASTX decreased the activation of KCs and HSCs and increased the ratio of M1 macrophages to KCs in a mouse model of MCD-induced NASH. In addition, ASTX inhibited the expression of the fibrosis-related genes TGF-β1, Col1A1 and PAI-1 and alleviated liver inflammation and fibrosis [133]. These results indicate that ASTX may be a new and promising treatment for NASH.

3.3.7. Glycyrrhizic Acid and Glycyrrhetinic Acid

Glycyrrhizic acid (GL, Figure 6G) and glycyrrhetinic acid (GA, Figure 6H) are the main bioactive compounds extracted from Glycyrrhiza uralensis Fisch and have been widely used for antitumor, anti-inflammatory, antiviral, and hepatoprotective purposes and for portal hypertension relief [72,73]. A mouse model of NASH induced by the MCD diet showed that glycyrrhizic acid and glycyrrhetinic acid inhibited deoxycholic acid-induced NLRP3 inflammasome-associated inflammation and blocked the mutual FXR-NLRP3 inflammasome pathways, significantly improving collagen deposition and decreasing the expression of α-SMA. Glycyrrhizic acid also significantly inhibited the mRNA expression of TGF-β1, TIMP1 and 2, collagen 1 and 2 and other fibrotic genes [74]. It was found that glycyrrhizic acid and its metabolite glycyrrhetinic acid inhibited the transcription of PC-I mediated by Smad3 and the activation of Q-HSCs in primary cultures and LF [75,134]. Glycyrrhizic acid also regulates the CD4+ T-cell response during liver fibrogenesis via the JNK, ERK and PI3K/AKT pathways [135].

4. Others

4.1. Calycosin

Calycosin (Figure 7A) is a phytoestrogen with a similar structure to mammalian estrogen that is extracted from the root of Astragalus membranaceus. Pharmacological research and clinical practice have proven that calycosin improves triglyceride metabolism and antioxidant free radicals, inhibits liver injury, regulates glucose uptake disorders in hepatocytes, and inhibits HCC [76,77,136,137,138]. Overexpression of ERβ or calycosin alone inhibited the proliferation and migration of LX-2 cells induced by TGF-β1, downregulated α-SMA, PC-I, TIMP-1, and p-STAT3 and upregulated the protein expression of matrix metalloproteinase (MMP)-1. There was positive feedback between ERβ and calycosin. ERβ may inhibit the main functions of LX-3 cells by inhibiting the phosphorylation of STAT2, which is an important way for calycosin to inhibit liver fibrosis [78]. In addition, calycosin inhibited LF by balancing the MMP-1/TIMP-1 system, increasing the expression of Erβ and activating the JAK2-STAT3 pathway [139]. In addition, a rat model of MCD-induced NASH showed that calycosin inhibited the activation of HSCs by activating FXR and promoted the expression of PPARa, CPT1, Syndecan-1 and LPL, which are involved in the β-oxidation of free fatty acids, thereby reducing triglyceride accumulation and LF [140].

4.2. Emodin

Emodin (Figure 7B) is a compound extracted from rhubarb. Emodin is widely used to treat cardiovascular diseases, asthma, cancer, diabetes and organ fibrosis [79,80,141]. In an MCD-induced mouse model, emodin improved hepatic function, serum inflammation, histopathological inflammation scores and LF by inhibiting the expression of NLRP3 and the assembly of NLRP3 inflammatory bodies [81]. It was found that emodin inhibited the activation of HSCs by inhibiting the mRNA expression of TGF-β1, Smad4 and α-SMA [82]. In addition, emodin induced HSC apoptosis through the p53/ERK/p38 axis [142]. Oxidative stress is one of the pathological factors of LF. YAP1 is the main downstream target mediating oxidative stress. Emodin inhibited the phosphorylation of YAP1 and the generation of oxidative stress by reducing the expression of YES1 and AMPK, thus alleviating liver injury and slowing the occurrence of LF [143].

5. Conclusions and Perspectives

LF, which is the prepathological state of various liver diseases, such as cirrhosis and HCC, has become the key to effectively preventing and treating liver diseases. Natural products have stable curative effects and high safety and tolerance. Therefore, natural products with the ability to improve LF are gradually being discovered and studied. In this paper, natural products that inhibit hepatic fibrosis were summarized, and their mechanisms were analyzed in detail. The results showed that the natural products inhibited hepatic fibrosis mainly by blocking the Hedgehog, Wnt/β-catenin, TGF-β1 and NF-κB signaling pathways, enhancing the activities of SOD and GSH-Px, inhibiting the activities of TGF-β1, IL-1β, PPAR-γ and TNF-α, and decreasing the levels of MDA and TIMP1. At present, research on natural products to improve LF is mainly based on animal models. Therefore, their clinical application value should be explored in follow-up research to provide a reference for the clinical use of natural products to treat LF.

Author Contributions

Conceptualization, N.M. and M.-R.L.; methodology, J.-Z.L. and N.C.; software, J.-Z.L.; writing—original draft preparation, J.-Z.L.; writing—review and editing, N.C., N.M. and M.-R.L.; visualization, J.-Z.L.; funding acquisition, M.-R.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Youth Program of the National Nature Science Foundation of China (grant number 82000556), the Fundamental Research Funds for the Central Universities (grant number 21623109), and the Guangzhou Basic and Applied Basic Research Project (grant number SL2022A04J00501).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Eslam, M.; Sarin, S.K.; Wong, V.W.; Fan, J.-G.; Kawaguchi, T.; Ahn, S.H.; Zheng, M.-H.; Shiha, G.; Yilmaz, Y.; Gani, R.; et al. The Asian Pacific Association for the Study of the Liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatol. Int. 2020, 14, 889–919. [Google Scholar] [CrossRef] [PubMed]
  2. Wong, V.W.; Chitturi, S.; Wong, G.L.; Yu, J.; Chan, H.L.; Farrell, G.C. Pathogenesis and novel treatment options for non-alcoholic steatohepatitis. Lancet Gastroenterol. Hepatol. 2016, 1, 56–67. [Google Scholar] [CrossRef] [PubMed]
  3. Hagström, H.; Nasr, P.; Ekstedt, M.; Hammar, U.; Stål, P.; Hultcrantz, R.; Kechagias, S. Fibrosis stage but not NASH predicts mortality and time to development of severe liver disease in biopsy-proven NAFLD. J. Hepatol. 2017, 67, 1265–1273. [Google Scholar] [CrossRef] [PubMed]
  4. Dulai, P.S.; Singh, S.; Patel, J.; Soni, M.; Prokop, L.J.; Younossi, Z.; Sebastiani, G.; Ekstedt, M.; Hagstrom, H.; Nasr, P.; et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology 2017, 65, 1557–1565. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, L.; Kong, D.; Xia, S.; Wang, F.; Li, Z.; Zhang, F.; Zheng, S. Crosstalk Between Autophagy and Innate Immunity: A Pivotal Role in Hepatic Fibrosis. Front. Pharmacol. 2022, 13, 891069. [Google Scholar] [CrossRef] [PubMed]
  6. Carter, J.K.; Friedman, S.L. Hepatic Stellate Cell-Immune Interactions in NASH. Front. Endocrinol. 2022, 13, 867940. [Google Scholar] [CrossRef] [PubMed]
  7. Holt, A.P.; Salmon, M.; Buckley, C.D.; Adams, D.H. Immune Interactions in LF. Clin. Liver Dis. 2008, 12, 861–882. [Google Scholar] [CrossRef]
  8. Parola, M.; Pinzani, M. LF: Pathophysiology, pathogenetic targets and clinical issues. Mol. Asp. Med. 2018, 65, 37–55. [Google Scholar] [CrossRef]
  9. Roehlen, N.; Crouchet, E.; Baumert, T.F. LF: Mechanistic Concepts and Therapeutic Perspectives. Cells 2020, 9, 875. [Google Scholar] [CrossRef]
  10. Higashi, T.; Friedman, S.L.; Hoshida, Y. Hepatic stellate cells as key target in LF. Adv. Drug Deliv. Rev. 2017, 121, 27–42. [Google Scholar] [CrossRef]
  11. Briscoe, J.; Thérond, P.P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 2013, 14, 416–429. [Google Scholar] [CrossRef]
  12. Swiderska-Syn, M.; Syn, W.K.; Xie, G.; Krüger, L.; Machado, M.V.; Karaca, G.A.; Michelotti, G.; Choi, S.S.; Premont, R.T.; Diehl, A.M. Myofibroblastic cells function as progenitors to regenerate murine livers after partial hepatectomy. Gut 2014, 63, 1333–1344. [Google Scholar] [CrossRef]
  13. Schwabe, R.F.; Tabas, I.; Pajvani, U.B. Mechanisms of Fibrosis Development in Nonalcoholic Steatohepatitis. Gastroenterology 2020, 158, 1913–1928. [Google Scholar] [CrossRef] [PubMed]
  14. Xie, G.; Choi, S.S.; Syn, W.-K.A.; Michelotti, G.; Swiderska, M.; Karaca, G.; Chan, I.S.; Chen, Y.; Diehl, A.M. Hedgehog signalling regulates liver sinusoidal endothelial cell capillarisation. Gut 2013, 62, 299–309. [Google Scholar] [CrossRef] [PubMed]
  15. Pereira, T.A.; Xie, G.; Choi, S.S.; Syn, W.-K.; Voieta, I.; Lu, J.; Chan, I.S.; Swiderska, M.; Amaral, K.B.; Antunes, C.M.; et al. Macrophage-derived Hedgehog ligands promotes fibrogenic and angiogenic responses in human schistosomiasis mansoni. Liver Int. 2013, 33, 149–161. [Google Scholar] [CrossRef] [PubMed]
  16. Swiderska-Syn, M.; Suzuki, A.; Guy, C.D.; Schwimmer, J.B.; Abdelmalek, M.F.; Lavine, J.E.; Diehl, A.M. Hedgehog pathway and pediatric nonalcoholic fatty liver disease. Hepatology 2013, 57, 1814–1825. [Google Scholar] [CrossRef] [PubMed]
  17. Pazzaglia, S.; Cifaldi, L.; Saran, A.; Nobili, V.; Fruci, D.; Alisi, A. Hedgehog/hyaluronic acid interaction network in nonalcoholic fatty liver disease, fibrosis, and hepatocellular carcinoma. Hepatology 2012, 56, 1589. [Google Scholar] [CrossRef]
  18. Choi, S.S.; Omenetti, A.; Witek, R.P.; Moylan, C.A.; Syn, W.-K.; Jung, Y.; Yang, L.; Sudan, D.L.; Sicklick, J.K.; Michelotti, G.A.; et al. Hedgehog pathway activation and epithelial-to-mesenchymal transitions during myofibroblastic transformation of rat hepatic cells in culture and cirrhosis. Am. J. Physiol. Gastrointes. Liver Physiol. 2009, 297, G1093–G1106. [Google Scholar] [CrossRef]
  19. Syn, W.; Jung, Y.; Omenetti, A.; Abdelmalek, M.; Guy, C.D.; Yang, L.; Wang, J.; Witek, R.P.; Fearing, C.M.; Pereira, T.A.; et al. Hedgehog-mediated epithelial-to-mesenchymal transition and fibrogenic repair in nonalcoholic fatty liver disease. Gastroenterology 2009, 137, 1478–1488.e8. [Google Scholar] [CrossRef]
  20. Michelotti, G.A.; Xie, G.; Swiderska, M.; Choi, S.S.; Karaca, G.; Krüger, L.; Premont, R.; Yang, L.; Syn, W.-K.; Metzger, D.; et al. Smoothened is a master regulator of adult liver repair. J. Clin. Investig. 2013, 123, 2380–2394. [Google Scholar] [CrossRef]
  21. Gong, Y.; Yang, Y. Activation of Nrf2/AREs-mediated antioxidant signalling, and suppression of profibrotic TGF-β1/Smad3 pathway: A promising therapeutic strategy for LF—A review. Life Sci. 2020, 256, 117909. [Google Scholar] [CrossRef] [PubMed]
  22. Gressner, A.M.; Weiskirchen, R.; Breitkopf, K.; Dooley, S. Roles of TGF-beta in LF. Front. Biosci. 2002, 7, d793–d807. [Google Scholar] [CrossRef] [PubMed]
  23. Dooley, S.; ten Dijke, P. TGF-β in progression of liver disease. Cell Tissue Res. 2012, 347, 245–256. [Google Scholar] [CrossRef] [PubMed]
  24. Dewidar, B.; Meyer, C.; Dooley, S.; Beinker, M. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis—Updated 2019. Cells 2019, 8, 1419. [Google Scholar] [CrossRef] [PubMed]
  25. Fabregat, I.; Moreno-Càceres, J.; Sánchez, A.; Dooley, S.; Dewidar, B.; Giannelli, G.; Ten Dijke, P. IT-LIVER Consortium. TGF-β signalling and liver disease. FEBS J. 2016, 283, 2219–2232. [Google Scholar] [CrossRef] [PubMed]
  26. Hermansyah, D.; Putra, A.; Muhar, A.M.; Retnaningsih Wirastuti, K.; Dirja, B.T. Mesenchymal Stem Cells Suppress TGF-β Release to Decrease α-SMA Expression in Ameliorating CCl4-Induced LF. Med. Arch. 2021, 75, 16–22. [Google Scholar] [CrossRef] [PubMed]
  27. van Beuge, M.M.; Prakash, J.; Lacombe, M.; Post, E.; Reker-Smit, C.; Beljaars, L.; Poelstra, K. Enhanced effectivity of an ALK5-Inhibitor after cell-specific delivery to hepatic stellate cells in mice with liver injury. PLoS ONE 2013, 8, e56442. [Google Scholar] [CrossRef]
  28. Henderson, N.C.; Arnold, T.D.; Katamura, Y.; Giacomini, M.M.; Rodriguez, J.D.; McCarty, J.H.; Pellicoro, A.; Raschperger, E.; Betsholtz, C.; Ruminski, P.G.; et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat. Med. 2013, 19, 1617–1624. [Google Scholar] [CrossRef]
  29. Shree Harini, K.; Ezhilarasan, D. Wnt/beta-catenin signaling and its modulators in nonalcoholic fatty liver diseases. Hepatobiliary Pancreat. Dis. Int. 2022, 22, S1499–S3872. [Google Scholar] [CrossRef]
  30. Miao, C.G.; Yang, Y.Y.; He, X.; Huang, C.; Huang, Y.; Zhang, L.; Lv, X.W.; Jin, Y.; Li, J. Wnt signaling in LF: Progress, challenges and potential directions. Biochimie 2013, 95, 2326–2335. [Google Scholar] [CrossRef]
  31. Pinson, K.I.; Brennan, J.; Monkley, S.; Avery, B.J.; Skarnes, W.C. An LDL-receptor-related protein mediates Wnt signalling in mice. Nature 2000, 407, 535–538. [Google Scholar] [CrossRef]
  32. Barrott, J.J.; Cash, G.M.; Smith, A.P.; Barrow, J.R.; Murtaugh, L.C. Deletion of mouse Porcn blocks Wnt ligand secretion and reveals an ectodermal etiology of human focal dermal hypoplasia/Goltz syndrome. Proc. Natl. Acad. Sci. USA 2011, 108, 12752–12757. [Google Scholar] [CrossRef] [PubMed]
  33. Ge, W.S.; Wang, Y.J.; Wu, J.X.; Fan, J.G.; Chen, Y.W.; Zhu, L. β-catenin is overexpressed in LF and blockage of Wnt/β -catenin signaling inhibits hepatic stellate cell activation. Mol. Med. Rep. 2014, 9, 2145–2151. [Google Scholar] [CrossRef] [PubMed]
  34. Guo, Y.; Xiao, L.; Sun, L.; Liu, F. Wnt/beta-catenin signaling: A promising new target for fibrosis diseases. Physiol. Res. 2012, 61, 337–346. [Google Scholar] [CrossRef] [PubMed]
  35. Paterson, I.; Anderson, E.A. Chemistry. The renaissance of natural products as drug candidates. Science 2005, 310, 451–453. [Google Scholar] [CrossRef] [PubMed]
  36. Teuten, E.L.; Xu, L.; Reddy, C.M. Two abundant bioaccumulated halogenated compounds are natural products. Science 2005, 307, 917–920. [Google Scholar] [CrossRef] [PubMed]
  37. Rollinger, J.M.; Langer, T.; Stuppner, H. Strategies for Efficient Lead Structure Discovery from Natural Products. Curr. Med. Chem. 2006, 13, 1491–1507. [Google Scholar] [CrossRef]
  38. Majeed, R.; Reddy, M.V.; Chinthakindi, P.K.; Sangwan, P.L.; Hamid, A.; Chashoo, G.; Saxena, A.K.; Koul, S. Bakuchiol derivatives as novel and potent cytotoxic agents: A report. Eur. J. Med. Chem. 2012, 49, 55–67. [Google Scholar] [CrossRef]
  39. Rašković, A.; Milanović, I.; Pavlović, N.; Ćebović, T.; Vukmirović, S.; Mikov, M. Antioxidant activity of rosemary (Rosmarinus officinalis L.) essential oil and its hepatoprotective potential. BMC Complement. Altern. Med. 2014, 14, 225. [Google Scholar] [CrossRef]
  40. Zhang, A.; Sun, H.; Wang, X. Recent advances in natural products from plants for treatment of liver diseases. Eur. J. Med. Chem. 2013, 63, 570–577. [Google Scholar] [CrossRef]
  41. Ahmad, A.; Ginnebaugh, K.R.; Li, Y.; Padhye, S.B.; Sarkar, F.H. Molecular targets of naturopathy in cancer research: Bridge to modern medicine. Nutrients 2015, 7, 321–334. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, J.; Zhang, H.; Deng, X.; Zhang, N.; Liu, B.; Xin, S.; Li, G.; Xu, K. Baicalin attenuates non-alcoholic steatohepatitis by suppressing key regulators of lipid metabolism, inflammation and fibrosis in mice. Life Sci. 2018, 192, 46–54. [Google Scholar] [CrossRef] [PubMed]
  43. Hu, Q.; Zhang, W.; Wu, Z.; Tian, X.; Xiang, J.; Li, L.; Li, Z.; Peng, X.; Wei, S.; Ma, X.; et al. Baicalin and the liver-gut system: Pharmacological bases explaining its therapeutic effects. Pharmacol. Res. 2021, 165, 105444. [Google Scholar] [CrossRef]
  44. Wei, X.L.; Fang, R.T.; Yang, Y.H.; Bi, X.Y.; Ren, G.X.; Luo, A.L.; Zhao, M.; Zang, W.J. Protective effects of extracts from Pomegranate peels and seeds on LF induced by carbon tetrachloride in rats. BMC Complement. Altern. Med. 2015, 15, 389. [Google Scholar] [CrossRef] [PubMed]
  45. Xiong, Y.; Lu, H.; Xu, H. Galangin Reverses LF 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] [PubMed]
  46. Wang, X.; Gong, G.; Yang, W.; Li, Y.; Jiang, M.; Li, L. Antifibrotic activity of galangin, a novel function evaluated in animal LF model. Environ. Toxicol. Pharmacol. 2013, 36, 288–295. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, M.; Yang, S.-G.; Kim, J.M.; Lee, J.-W.; Kim, Y.S.; Lee, J.I. Silymarin suppresses hepatic stellate cell activation in a dietary rat model of non-alcoholic steatohepatitis: Analysis of isolated hepatic stellate cells. Int. J. Mol. Med. 2012, 30, 473–479. [Google Scholar] [CrossRef]
  48. Crocenzi, F.A.; Roma, M.G. Silymarin as a New hepatoprotective agent in experimental cholestasis: New possibilities for an ancient medication. Curr. Med. Chem. 2006, 13, 1055–1074. [Google Scholar] [CrossRef]
  49. Hismiogullari, S.E.; Hismiogullari, A.A.; Sunay, F.B.; Paksoy, S.; Can, M.; Aksit, H.; Karaca, O.; Yavuz, O. The protective effect of curcumin on carbon tetrachloride induced liver damage. Revue Méd. Vét. 2014, 165, 194–200. [Google Scholar]
  50. Vizzutti, F.; Provenzano, A.; Galastri, S.; Milani, S.; Delogu, W.; Novo, E.; Caligiuri, A.; Zamara, E.; Arena, U.; Laffi, G.; et al. Curcumin limits the fibrogenic evolution of experimental steatohepatitis. Lab. Investig. 2010, 90, 104–115. [Google Scholar] [CrossRef]
  51. Li, B.; Wang, L.; Lu, Q.; Da, W. Liver injury attenuation by curcumin in a rat NASH model: An Nrf2 activation-mediated effect? Ir. J. Med. Sci. 2016, 185, 93–100. [Google Scholar] [CrossRef] [PubMed]
  52. Afrin, R.; Arumugam, S.; Rahman, A.; Wahed, M.I.I.; Karuppagounder, V.; Harima, M.; Suzuki, H.; Miyashita, S.; Suzuki, K.; Yoneyama, H.; et al. Curcumin ameliorates liver damage and progression of NASH in NASH-HCC mouse model possibly by modulating HMGB1-NF-κB translocation. Int. Immunopharmacol. 2017, 44, 174–182. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. 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]
  55. 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] [PubMed]
  56. 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] [PubMed]
  57. Wan, X.; Huang, H.C.; Wang, X.P.; Hu, Z.H.; Liu, K.Y.; Huang, D.B. Euonymus alatus and its monomers alleviate LF both in mice and LX2 cells by blocking TβR1-Smad2/3 and TNF-α-NF-κB pathways. Am. J. Transl. Res. 2019, 11, 106–119. [Google Scholar]
  58. Chen, J.; Fan, X.; Zhou, L.; Gao, X. Treatment with geraniol ameliorates methionine-choline-deficient diet-induced non-alcoholic steatohepatitis in rats. J. Gastroenterol. Hepatol. 2016, 31, 1357–1365. [Google Scholar] [CrossRef]
  59. El Azab, E.F.; Elguindy, N.M.; Yacout, G.A.; Elgamal, D.A. Hepatoprotective Impact of Geraniol Against CCl4-Induced LF in Rats. Pak. J. Biol. Sci. 2020, 23, 1650–1658. [Google Scholar] [CrossRef]
  60. Song, J.; Han, X.; Yao, Y.-L.; Li, Y.-M.; Zhang, J.; Shao, D.-Y.; Hou, L.-S.; Fan, Y.; Song, S.-Z.; Lian, L.-H.; et al. Acanthoic acid suppresses lipin1/2 via TLR4 and IRAK4 signalling pathways in EtOH- and lipopolysaccharide-induced hepatic lipogenesis. J. Pharm. Pharmacol. 2018, 70, 393–403. [Google Scholar] [CrossRef]
  61. Wu, Y.-L.; Jiang, Y.-Z.; Jin, X.-J.; Lian, L.-H.; Piao, J.-Y.; Wan, Y.; Jin, H.-R.; Lee, J.J.; Nan, J.-X. Acanthoic acid, a diterpene in Acanthopanax koreanum, protects acetaminophen-induced hepatic toxicity in mice. Phytomedicine 2010, 17, 475–479. [Google Scholar] [CrossRef] [PubMed]
  62. Bai, T.; Yao, Y.-L.; Jin, X.-J.; Lian, L.-H.; Li, Q.; Yang, N.; Jin, Q.; Wu, Y.-L.; Nan, J.-X. Acanthoic acid, a diterpene in Acanthopanax koreanum, ameliorates the development of LF via LXRs signals. Chem. Biol. Interact. 2014, 218, 63–70. [Google Scholar] [CrossRef] [PubMed]
  63. Han, X.; Song, J.; Lian, L.H.; Yao, Y.L.; Shao, D.Y.; Fan, Y.; Hou, L.S.; Wang, G.; Zheng, S.; Wu, Y.L. Ginsenoside 25-OCH3-PPD Promotes Activity of LXRs To Ameliorate P2X7R-Mediated NLRP3 Inflammasome in the Development of LF. J. Agric. Food Chem. 2018, 66, 7023–7035. [Google Scholar] [CrossRef] [PubMed]
  64. Lo, Y.-T.; Tsai, Y.-H.; Wu, S.-J.; Chen, J.-R.; Chao, J.C.-J.; Yada, K.; Ishibashi, H.; Mori, H.; Morine, Y.; Zhu, C.; et al. Ginsenoside Rb1 inhibits cell activation and LF in rat hepatic stellate cells. J. Med. Food 2011, 14, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, X.-J.; Liu, W.-J.; Wen, M.-L.; Liang, H.; Wu, S.-M.; Zhu, Y.-Z.; Zhao, J.-Y.; Dong, X.-Q.; Li, M.-G.; Bian, L.; et al. Ameliorative effects of Compound K and ginsenoside Rh1 on non-alcoholic fatty liver disease in rats. Sci. Rep. 2017, 7, 41144. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, G.; Cui, Z.; Gao, X.; Liu, H.; Wang, L.; Gong, J.; Wang, A.; Zhang, J.; Ma, Q.; Huang, Y.; et al. Corosolic acid ameliorates non-alcoholic steatohepatitis induced by high-fat diet and carbon tetrachloride by regulating TGF-β1/Smad2, NF-κB, and AMPK signaling pathways. Phytother. Res. 2021, 35, 5214–5226. [Google Scholar] [CrossRef] [PubMed]
  67. Ni, Y.; Zhuge, F.; Nagashimada, M.; Nagata, N.; Xu, L.; Yamamoto, S.; Fuke, N.; Ushida, Y.; Suganuma, H.; Kaneko, S.; et al. Lycopene prevents the progression of lipotoxicity-induced nonalcoholic steatohepatitis by decreasing oxidative stress in mice. Free. Radic. Biol. Med. 2020, 152, 571–582. [Google Scholar] [CrossRef]
  68. Jiang, W.; Guo, M.-H.; Hai, X. Hepatoprotective and antioxidant effects of lycopene on non-alcoholic fatty liver disease in rat. World J. Gastroenterol. 2016, 22, 10180–10188. [Google Scholar] [CrossRef]
  69. Kitade, Y.; Watanabe, S.; Masaki, T.; Nishioka, M.; Nishino, H. Inhibition of LF in LEC rats by a carotenoid, lycopene, or a herbal medicine, Sho-saiko-to. Hepatol. Res. 2002, 22, 196–205. [Google Scholar] [CrossRef]
  70. Higuera-Ciapara, I.; Félix-Valenzuela, L.; Goycoolea, F.M. Astaxanthin: A review of its chemistry and applications. Crit. Rev. Food Sci. Nutr. 2006, 46, 185–196. [Google Scholar] [CrossRef]
  71. Li, J.; Guo, C.; Wu, J. Astaxanthin in Liver Health and Disease: A Potential Therapeutic Agent. Drug Des. Dev. Ther. 2020, 14, 2275–2285. [Google Scholar] [CrossRef] [PubMed]
  72. Asl, M.N.; Hosseinzadeh, H. Review of pharmacological effects of Glycyrrhiza sp. and its bioactive compounds. Phytother. Res. 2008, 22, 709–724. [Google Scholar] [CrossRef] [PubMed]
  73. Pun, C.K.; Huang, H.-C.; Chang, C.-C.; Chuang, C.-L.; Yen, C.-H.; Hsu, S.-J.; Lee, F.-Y.; Hou, M.-C.; Huang, Y.-H. Glycyrrhizin Attenuates Portal Hypertension and Collateral Shunting via Inhibition of Extrahepatic Angiogenesis in Cirrhotic Rats. Int. J. Mol. Sci. 2021, 22, 7662. [Google Scholar] [CrossRef] [PubMed]
  74. Yan, T.; Wang, H.; Cao, L.; Wang, Q.; Takahashi, S.; Yagai, T.; Li, G.; Krausz, K.W.; Wang, G.; Gonzalez, F.J.; et al. Glycyrrhizin Alleviates Nonalcoholic Steatohepatitis via Modulating Bile Acids and Meta-Inflammation. Drug Metab. Dispos. 2018, 46, 1310–1319. [Google Scholar] [CrossRef]
  75. Moro, T.; Shimoyama, Y.; Kushida, M.; Hong, Y.Y.; Nakao, S.; Higashiyama, R.; Sugioka, Y.; Inoue, H.; Okazaki, I.; Inagaki, Y. Glycyrrhizin and its metabolite inhibit Smad3-mediated type I collagen gene transcription and suppress experimental murine liver fibrosis. Life Sci. 2008, 83, 531–539. [Google Scholar] [CrossRef] [PubMed]
  76. Xu, Y.; Xiong, J.; Zhao, Y.; He, B.; Zheng, Z.; Chu, G.; Zhu, Q. Calycosin rebalances advanced glycation end products-induced glucose uptake dysfunction of hepatocyte in vitro. Am. J. Chin. Med. 2015, 43, 1191–1210. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, D.; Wang, S.; Zhu, L.; Tian, Y.; Wang, H.; Zhuang, Y.; Li, Y.; Wang, D. Profiling of hepatocellular carcinoma cell cycle regulating genes targeted by calycosin. BioMed Res. Int. 2013, 2013, 317926. [Google Scholar] [CrossRef]
  78. Wang, Y.; Wu, C.; Zhou, J.; Fang, H.; Wang, J. Overexpression of estrogen receptor β inhibits cellular functions of human hepatic stellate cells and promotes the anti-fibrosis effect of calycosin via inhibiting STAT3 phosphorylation. BMC Pharmacol. Toxicol. 2022, 23, 77. [Google Scholar] [CrossRef]
  79. Lin, L.; Liu, Y.; Fu, S.; Qu, C.; Li, H.; Ni, J. Inhibition of Mitochondrial Complex Function—The Hepatotoxicity Mechanism of Emodin Based on Quantitative Proteomic Analyses. Cells 2019, 8, 263. [Google Scholar] [CrossRef]
  80. Shang, H.; Jia, X.; Liu, H.; Zhang, X.; Shao, Y. A comprehensive review of emodin in fibrosis treatment. Fitoterapia 2023, 165, 105358. [Google Scholar] [CrossRef]
  81. Wu, C.; Bian, Y.; Lu, B.; Wang, D.; Azami, N.L.B.; Wei, G.; Ma, F.; Sun, M. Rhubarb free anthraquinones improved mice nonalcoholic fatty liver disease by inhibiting NLRP3 inflammasome. J. Transl. Med. 2022, 20, 294. [Google Scholar] [CrossRef] [PubMed]
  82. Dong, M.-X.; Jia, Y.; Zhang, Y.-B.; Li, C.-C.; Geng, Y.-T.; Zhou, L.; Li, X.-Y.; Liu, J.-C.; Niu, Y.-C. Emodin protects rat liver from CCl4-induced fibrogenesis via inhibition of hepatic stellate cells activation. World J. Gastroenterol. 2009, 15, 4753–4762. [Google Scholar] [CrossRef]
  83. Wang, A.; Lin, L.; Wang, Y. Traditional Chinese herbal medicine penthorum Chinense pursh: A phytochemical and pharmacological review. Am. J. Chin. Med. 2015, 43, 601–620. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, A.; Li, M.; Huang, H.; Xiao, Z.; Shen, J.; Zhao, Y.; Yin, J.; Kaboli, P.J.; Cao, J.; Cho, C.H.; et al. A review of Penthorum chinense Pursh for hepatoprotection: Traditional use, phytochemistry, pharmacology, toxicology and clinical trials. J. Ethnopharmacol. 2020, 251, 112569. [Google Scholar] [CrossRef] [PubMed]
  85. Zhong, X.; Liu, H. Baicalin attenuates diet induced nonalcoholic steatohepatitis by inhibiting inflammation and oxidative stress via suppressing JNK signaling pathways. Biomed. Pharmacother. 2018, 98, 111–117. [Google Scholar] [CrossRef] [PubMed]
  86. 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] [PubMed]
  87. Yang, M.D.; Chiang, Y.-M.; Higashiyama, R.; Asahina, K.; Mann, D.A.; Mann, J.; Wang, C.C.; Tsukamoto, H. Rosmarinic acid and baicalin epigenetically derepress peroxisomal proliferator-activated receptor γ in hepatic stellate cells for their antifibrotic effect. Hepatology 2012, 55, 1271–1281. [Google Scholar] [CrossRef] [PubMed]
  88. 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]
  89. Huang, Y.C.; Tsai, M.S.; Hsieh, P.C.; Shih, J.H.; Wang, T.S.; Wang, Y.C.; Lin, T.H.; Wang, S.H. Galangin ameliorates cisplatin-induced nephrotoxicity by attenuating oxidative stress, inflammation and cell death in mice through inhibition of ERK and NF-kappaB signaling. Toxicol. Appl. Pharmacol. 2017, 329, 128–139. [Google Scholar] [CrossRef]
  90. Kim, Y.-R.; Kim, M.-A.; Cho, H.-J.; Oh, S.-K.; Lee, I.-K.; Kim, U.-K.; Lee, K.-Y. Galangin prevents aminoglycoside-induced ototoxicity by decreasing mitochondrial production of reactive oxygen species in mouse cochlear cultures. Toxicol. Lett. 2016, 245, 78–85. [Google Scholar] [CrossRef]
  91. Ou, R.; Lin, L.; Zhao, M.; Xie, Z. Action mechanisms and interaction of two key xanthine oxidase inhibitors in galangal: Combination of in vitro and in silico molecular docking studies. Int. J. Biol. Macromol. 2020, 162, 1526–1535. [Google Scholar] [CrossRef] [PubMed]
  92. 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 LF. RSC Adv. 2023, 13, 10987–11001. [Google Scholar] [CrossRef] [PubMed]
  93. Gillessen, A.; Schmidt, H.H.-J. Silymarin as Supportive Treatment in Liver Diseases: A Narrative Review. Adv. Ther. 2020, 37, 1279–1301. [Google Scholar] [CrossRef] [PubMed]
  94. Aghemo, A.; Alekseeva, O.P.; Angelico, F.; Bakulin, I.G.; Bakulina, N.V.; Bordin, D.; Bueverov, A.O.; Drapkina, O.M.; Gillessen, A.; Kagarmanova, E.M.; et al. Role of silymarin as antioxidant in clinical management of chronic liver diseases: A narrative review. Ann. Med. 2022, 54, 1548–1560. [Google Scholar] [CrossRef] [PubMed]
  95. Gazák, R.; Walterova, D.; Kren, V. Silybin and silymarin—New and emerging applications in medicine. Curr. Med. Chem. 2007, 14, 315–338. [Google Scholar] [CrossRef] [PubMed]
  96. Trappoliere, M.; Caligiuri, A.; Schmid, M.; Bertolani, C.; Failli, P.; Vizzutti, F.; Novo, E.; di Manzano, C.; Marra, F.; Loguercio, C.; et al. Silybin, a component of sylimarin, exerts antiinflammatory and antifibrogenic effects on human hepatic stellate cells. J. Hepatol. 2009, 50, 1102–1111. [Google Scholar] [CrossRef] [PubMed]
  97. Wah Kheong, C.; Nik Mustapha, N.R.; Mahadeva, S. A randomized trial of silymarin for the treatment of nonalcoholic steatohepatitis. Clin. Gastroenterol. Hepatol. 2017, 15, 1940–1949. [Google Scholar] [CrossRef]
  98. Li, A.-N.; Li, S.; Zhang, Y.-J.; Xu, X.-R.; Chen, Y.-M.; Li, H.-B. Resources and biological activities of natural polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef]
  99. Li, S.; Tan, H.Y.; Wang, N.; Cheung, F.; Hong, M.; Feng, Y. The Potential and Action Mechanism of Polyphenols in the Treatment of Liver Diseases. Oxid. Med. Cell. Longev. 2018, 2018, 8394818. [Google Scholar] [CrossRef]
  100. Farzaei, M.H.; Zobeiri, M.; Parvizi, F.; El-Senduny, F.F.; Marmouzi, I.; Coy-Barrera, E.; Naseri, R.; Nabavi, S.M.; Rahimi, R.; Abdollahi, M. Curcumin in Liver Diseases: A Systematic Review of the Cellular Mechanisms of Oxidative Stress and Clinical Perspective. Nutrients 2018, 10, 855. [Google Scholar] [CrossRef]
  101. 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]
  102. Hernández-Aquino, E.; Quezada-Ramírez, M.A.; Silva-Olivares, A.; Ramos-Tovar, E.; Flores-Beltrán, R.E.; Segovia, J.; Shibayama, M.; Muriel, P. Curcumin downregulates Smad pathways and reduces hepatic stellate cells activation in experimental fibrosis. Ann. Hepatol. 2020, 19, 497–506. [Google Scholar] [CrossRef]
  103. Shu, J.-C.; He, Y.-J.; Lv, X.; Ye, G.-R.; Wang, L.-X. Curcumin prevents liver fibrosis by inducing apoptosis and suppressing activation of hepatic stellate cells. J. Nat. Med. 2009, 63, 415–420. [Google Scholar] [CrossRef]
  104. Zhao, Y.; Ma, X.; Wang, J.; He, X.; Hu, Y.; Zhang, P.; Wang, R.; Li, R.; Gong, M.; Luo, S.; et al. Curcumin protects against CCl4-induced LF in rats by inhibiting HIF-1α through an ERK-dependent pathway. Molecules 2014, 19, 18767–18780. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, P.; Huang, R.; Xiong, Y.-L.; Wu, C. Protective effects of curcumin against LF through modulating DNA methylation. Chin. J. Nat. Med. 2016, 14, 255–264. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, X.-A.; Chen, G.; Liu, Y.; Chen, Y.; Wu, H.; Xiong, Y.; Wang, G.; Jia, B.; Li, Y.; Xia, J.; et al. Curcumin reduces Ly6Chi monocyte infiltration to protect against LF by inhibiting Kupffer cells activation to reduce chemokines secretion. Biomed. Pharmacother. 2018, 106, 868–878. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, F.; Zhang, Z.; Chen, L.; Kong, D.; Zhang, X.; Lu, C.; Lu, Y.; Zheng, S. Curcumin attenuates angiogenesis in LF and inhibits angiogenic properties of hepatic stellate cells. J. Cell. Mol. Med. 2014, 18, 1392–1406. [Google Scholar] [CrossRef]
  108. 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]
  109. 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 LF and Portal Pressure in Cirrhotic Rats. J. Hepatol. 2013, 58, 904–910. [Google Scholar] [CrossRef]
  110. de Oliveira, C.M.; Martins, L.A.M.; de Sousa, A.C.; Moraes, K.d.S.; Costa, B.P.; Vieira, M.Q.; Coelho, B.P.; Borojevic, R.; de Oliveira, J.R.; Guma, F.C.R. Resveratrol Increases the Activation Markers and Changes the Release of Inflammatory Cytokines of Hepatic Stellate Cells. Mol. Cell. Biochem. 2020, 476, 649–661. [Google Scholar] [CrossRef]
  111. Lee, E.S.; Shin, M.O.; Yoon, S.; Moon, J.O. Resveratrol Inhibits Dimethylnitrosamine-induced LF in Rats. Arch. Pharm. Res. 2010, 33, 925–932. [Google Scholar] [CrossRef] [PubMed]
  112. 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 LF in Rats. Arch. Pharm. Res. 2010, 33, 601–609. [Google Scholar] [CrossRef] [PubMed]
  113. Hessin, A.F.; Hegazy, R.R.; Hassan, A.A.; Yassin, N.Z.; Kenawy, S.A.B. Resveratrol prevents LF via two possible pathways: Modulation of alpha fetoprotein transcriptional levels and normalization of protein kinase C responses. Indian. J. Pharm. 2017, 49, 282–289. [Google Scholar] [CrossRef] [PubMed]
  114. Zhao, W.; Li, A.; Feng, X.; Hou, T.; Liu, K.; Liu, B.; Zhang, N. Metformin and resveratrol ameliorate muscle insulin resistance through preventing lipolysis and inflammation in hypoxic adipose tissue. Cell. Signal. 2016, 28, 1401–1411. [Google Scholar] [CrossRef] [PubMed]
  115. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Gondal, T.A.; Saeed, F.; Imran, A.; Shahbaz, M.; Fokou, P.V.T.; Arshad, M.U.; Khan, H.; et al. Kaempferol: A key emphasis to its anticancer potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed]
  116. Calderon-Montaño, J.M.; Burgos-Morón, E.; Perez-Guerrero, C.; Lopez-Lazaro, M. A review on the dietary flavonoid kaempferol. Mini-Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  117. Santos JS, D.; Gonçalves Cirino, J.P.; de Oliveira Carvalho, P.; Ortega, M.M. The pharmacological action of kaempferol in central nervous system diseases: A review. Front. Pharmacol. 2021, 11, 565700. [Google Scholar] [CrossRef]
  118. Alkandahri, M.Y.; Pamungkas, B.T.; Oktoba, Z.; Shafirany, M.Z.; Sulastri, L.; Arfania, M.; Anggraeny, E.N.; Pratiwi, A.; Astuti, F.D.; Indriyani; et al. Hepatoprotective Effect of Kaempferol: A Review of the Dietary Sources, Bioavailability, Mechanisms of Action, and Safety. Adv. Pharmacol. Pharm. Sci. 2023, 2023, 1387665. [Google Scholar] [CrossRef]
  119. Liu, P.; Wu, P.; Yang, B.; Wang, T.; Li, J.; Song, X.; Sun, W. Kaempferol prevents the progression from simple steatosis to non-alcoholic steatohepatitis by inhibiting the NF-κB pathway in oleic acid-induced HepG2 cells and high-fat diet-induced rats. J. Funct. Foods 2021, 85, 10. [Google Scholar] [CrossRef]
  120. Yao, P.; Liu, Y. Terpenoids: Natural Compounds for Non-Alcoholic Fatty Liver Disease (NAFLD) Therapy. Molecules 2022, 28, 272. [Google Scholar] [CrossRef]
  121. Jayachandran, M.; Chandrasekaran, B.; Namasivayam, N. Geraniol attenuates oxidative stress by Nrf2 activation in diet-induced experimental atherosclerosis. J. Basic. Clin. Physiol. Pharmacol. 2014, 26, 335–346. [Google Scholar] [CrossRef] [PubMed]
  122. Jayachandran, M.; Chandrasekaran, B.; Namasivayam, N. Effect of geraniol, a plant derived monoterpene on lipids and lipid metabolizing enzymes in experimental hyperlipidemic hamsters. Mol. Cell. Biochem. 2015, 398, 39–53. [Google Scholar] [CrossRef] [PubMed]
  123. Jayachandran, M.; Chandrasekaran, B.; Namasivayam, N. Geraniol attenuates fibrosis and exerts anti-inflammatory effects on diet induced atherogenesis by NF-κB signaling pathway. Eur. J. Pharmacol. 2015, 762, 102–111. [Google Scholar] [CrossRef] [PubMed]
  124. Hou, M.; Wang, R.; Zhao, S.; Wang, Z. Ginsenosides in Panax genus and their biosynthesis. Acta Pharm. Sin. B 2021, 11, 1813–1834. [Google Scholar] [CrossRef] [PubMed]
  125. Lim, W.; Mudge, K.W.; Vermeylen, F. Effects of population, age, and cultivation methods on ginsenoside content of wild American ginseng (Panax quinquefolium). J. Agric. Food Chem. 2005, 53, 8498–8505. [Google Scholar] [CrossRef] [PubMed]
  126. Hou, Y.L.; Tsai, Y.H.; Lin, Y.H.; Chao, J.C. Ginseng extract and ginsenoside Rb1 attenuate carbon tetrachloride-induced LF in rats. BMC Complement. Altern. Med. 2014, 14, 415. [Google Scholar] [CrossRef] [PubMed]
  127. Zhao, J.; Zhou, H.; An, Y.; Shen, K.; Yu, L. Biological effects of corosolic acid as an anti-inflammatory, anti-metabolic syndrome and anti-neoplasic natural compound. Oncol. Lett. 2021, 21, 84. [Google Scholar] [CrossRef]
  128. Yang, J.; Leng, J.; Li, J.-J.; Tang, J.-F.; Li, Y.; Liu, B.-L.; Wen, X.-D. Corosolic acid inhibits adipose tissue inflammation and ameliorates insulin resistance via AMPK activation in high-fat fed mice. Phytomedicine 2016, 23, 181–190. [Google Scholar] [CrossRef]
  129. Li, N.; Wu, X.; Zhuang, W.; Xia, L.; Chen, Y.; Wu, C.; Rao, Z.; Du, L.; Zhao, R.; Yi, M.; et al. Tomato and lycopene and multiple health outcomes: Umbrella review. Food Chem. 2021, 343, 128396. [Google Scholar] [CrossRef]
  130. Elias, M.d.B.; Oliveira, F.L.; Guma, F.C.R.; Martucci, R.B.; Borojevic, R.; Teodoro, A.J. Lycopene inhibits hepatic stellate cell activation and modulates cellular lipid storage and signaling. Food Funct. 2019, 10, 1974–1984. [Google Scholar] [CrossRef]
  131. Zhang, C.; Chen, X.; Too, H.-P. Microbial astaxanthin biosynthesis: Recent achievements, challenges, and commercialization outlook. Appl. Microbiol. Biotechnol. 2020, 104, 5725–5737. [Google Scholar] [CrossRef] [PubMed]
  132. Yang, Y.; Kim, B.; Park, Y.-K.; Koo, S.I.; Lee, J.-Y. Astaxanthin prevents TGFβ1-induced pro-fibrogenic gene expression by inhibiting Smad3 activation in hepatic stellate cells. Biochim. Biophys. Acta 2015, 1850, 178–185. [Google Scholar] [CrossRef] [PubMed]
  133. Ni, Y.; Nagashimada, M.; Zhuge, F.; Zhan, L.; Nagata, N.; Tsutsui, A.; Nakanuma, Y.; Kaneko, S.; Ota, T. Astaxanthin prevents and reverses diet-induced insulin resistance and steatohepatitis in mice: A comparison with vitamin E. Sci. Rep. 2015, 5, 17192. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, C.; Duan, X.; Sun, X.; Liu, Z.; Sun, P.; Yang, X.; Sun, H.; Liu, K.; Meng, Q. Protective effects of glycyrrhizic acid from edible botanical glycyrrhiza glabra against non-alcoholic steatohepatitis in mice. Food Funct. 2016, 7, 3716–3723. [Google Scholar] [CrossRef] [PubMed]
  135. Tu, C.-T.; Li, J.; Wang, F.-P.; Li, L.; Wang, J.-Y.; Jiang, W. Glycyrrhizin regulates CD4+T cell response during liver fibrogenesis via JNK, ERK and PI3K/AKT pathway. Int. Immunopharmacol. 2012, 14, 410–421. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, S.; Zhai, C.; Liu, Q.; Wang, X.; Ren, Z.; Zhang, Y.; Zhang, Y.; Wu, Q.; Sun, S.; Li, S.; et al. Cycloastragenol, a triterpene aglycone derived from Radix astragali, suppresses the accumulation of cytoplasmic lipid droplet in 3T3-L1 adipocytes. Biochem. Biophys. Res. Commun. 2014, 450, 306–311. [Google Scholar] [CrossRef] [PubMed]
  137. Chen, X.L.; Meng, Q.; Wang, C.Y.; Liu, Q.; Sun, H.; Huo, X.K.; Sun, P.Y.; Yang, X.B.; Peng, J.; Liu, K. Protective effects of calycosin against CCl4-Induced liver injury with activation of FXR and STAT3 in mice. Pharm. Res. 2015, 32, 538–548. [Google Scholar] [CrossRef]
  138. Li, J.; Lin, X.; Ma, Y.F.; Huang, Y.F. Protective effect of calycosin-7-O-β-D-Glucopyranoside Against Oxidative Stress of BRL-3A Cells Induced by Thioacetamide. Pharmacogn. Mag. 2015, 11, 524–532. [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-Schmiedeberg’s Arch. Pharmacol. 2021, 394, 189–204. [Google Scholar] [CrossRef]
  140. Duan, X.; Meng, Q.; Wang, C.; Liu, Z.; Liu, Q.; Sun, H.; Sun, P.; Yang, X.; Huo, X.; Peng, J.; et al. Calycosin attenuates triglyceride accumulation and LF in murine model of non-alcoholic steatohepatitis via activating farnesoid X receptor. Phytomedicine 2017, 25, 83–92. [Google Scholar] [CrossRef]
  141. Dong, X.; Fu, J.; Yin, X.; Cao, S.; Li, X.; Lin, L.; Ni, J. Huyiligeqi Emodin: A Review of its Pharmacology, Toxicity and Pharmacokinetics. Phytother. Res. 2016, 30, 1207–1218. [Google Scholar] [CrossRef]
  142. Liang, B.; Gao, L.; Wang, F.; Li, Z.; Li, Y.; Tan, S.; Chen, A.; Shao, J.; Zhang, Z.; Sun, L.; et al. The mechanism research on the anti-liver fibrosis of emodin based on network pharmacology. IUBMB Life 2021, 73, 1166–1179. [Google Scholar] [CrossRef]
  143. Lee, E.H.; Baek, S.Y.; Park, J.Y.; Kim, Y.W. Emodin in Rheum. undulatum inhibits oxidative stress in the liver via AMPK with Hippo/Yap signalling pathway. Pharm. Biol. 2020, 58, 333–341. [Google Scholar] [CrossRef]
Figure 1. Hedgehog (Hh) signaling pathway and target of natural compounds.
Figure 1. Hedgehog (Hh) signaling pathway and target of natural compounds.
Molecules 28 07936 g001
Figure 2. TGF-β1 signaling pathway and target of natural compounds.
Figure 2. TGF-β1 signaling pathway and target of natural compounds.
Molecules 28 07936 g002
Figure 3. Wnt/β-catenin signaling pathway and target of natural compounds.
Figure 3. Wnt/β-catenin signaling pathway and target of natural compounds.
Molecules 28 07936 g003
Figure 4. The chemical structures of (A) Baicalin; (B) Galangin; (C) Silymarin.
Figure 4. The chemical structures of (A) Baicalin; (B) Galangin; (C) Silymarin.
Molecules 28 07936 g004
Figure 5. The chemical structures of (A) Curcumin; (B) Resveratrol; (C) Kaempferol.
Figure 5. The chemical structures of (A) Curcumin; (B) Resveratrol; (C) Kaempferol.
Molecules 28 07936 g005
Figure 6. The chemical structures of (A) Geraniol; (B) Acanthoic Acid; (C) Ginsenoside; (D) Corosolic Acid; (E) Lycopene; (F) Astaxanthin; (G) Glycyrrhizic Acid; (H) Glycyrrhetinic Acid.
Figure 6. The chemical structures of (A) Geraniol; (B) Acanthoic Acid; (C) Ginsenoside; (D) Corosolic Acid; (E) Lycopene; (F) Astaxanthin; (G) Glycyrrhizic Acid; (H) Glycyrrhetinic Acid.
Molecules 28 07936 g006
Figure 7. The chemical structures of (A) Calycosin; (B) Emodin.
Figure 7. The chemical structures of (A) Calycosin; (B) Emodin.
Molecules 28 07936 g007
Table 1. Pharmacological effects of natural products with anti-fibrotic activity in NAFLD.
Table 1. Pharmacological effects of natural products with anti-fibrotic activity in NAFLD.
Natural
Products
ResourceInterfering
Mechanism
ModelPharmacological EffectsRefs
BaicalinFlavonoidsNF-κB, Wnt, PPAR-γMiceAntioxidant, anti-inflammatory and hepatoprotective[42,43,44]
GalanginFlavonoidsPI3K/Akt, Bax/Bcl-2, WntLX-2 cellScavenges free radicals, reduces lipid peroxidation, inhibits the activation and proliferation of HSCs[45,46]
SilymarinFlavonoidsTNF-α, connective tissue growth factorRatsReduces free radicals and lipid peroxidation[47,48]
CurcuminPolyphenol compoundsTGF-β/Smad, JNK/Smad3, ERK, PPAR-γMiceAntioxidant and antifibrotic[49,50,51,52]
ResveratrolPolyphenol compoundsGPx/SOD, PTEN/PI3K/AKTRatsAnti-inflammatory and antioxidant[53,54,55]
KaempferolPolyphenol compoundsTGF-β1/Smad2/3, TNF-α/NF-κBRatsAnti-inflammatory, antioxidant,[56,57]
GeraniolTerpenoidsTNF-α, IL-6, GPx/SODRatsAntioxidant and anti-inflammatory[58,59]
Acanthoic AcidTerpenoidsFXR/LXR-AMPK-SIRT1Mice and HSC-T6 cellsAntifibrotic[60,61,62]
GinsenosideTerpenoidsLXR, TGF-β1Mice and HSC-T6 cellsAnti-inflammatory and antifibrotic[63,64,65]
Corosolic AcidTerpenoidsNF-κ, TGF-β1/Smad2, AMPKMiceAnti-obesity, anti-inflammatory, antihyperlipidemic[66]
LycopeneTerpenoidsTNF-α, PPAR-α and RXR-β/γRatsAntioxidant and antifibrotic[67,68,69]
AstaxanthinTerpenoidsTGF-β1/Smad3,MiceAntioxidant[70,71]
Glycyrrhizic AcidTerpenoidsFXR-NLRP3, JNK, ERK, PI3K/AKTRatsAnti-inflammatory[72,73,74,75]
Glycyrrhetinic AcidTerpenoidsFXR-NLRP3RatsAnti-inflammatory, hepatoprotective[72]
CalycosinIsoflavoneTGF-β1, Erβ, JAK2-STAT3, FXRLX-2 cells, ratsImproves triglyceride metabolism and antioxidant free radicals, inhibits liver injury[76,77,78]
EmodinIsoflavoneTGF-β1, p53/ERK/p38, YAP1MiceAntioxidant and antifibrotic[79,80,81,82]
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

Li, J.-Z.; Chen, N.; Ma, N.; Li, M.-R. Mechanism and Progress of Natural Products in the Treatment of NAFLD-Related Fibrosis. Molecules 2023, 28, 7936. https://doi.org/10.3390/molecules28237936

AMA Style

Li J-Z, Chen N, Ma N, Li M-R. Mechanism and Progress of Natural Products in the Treatment of NAFLD-Related Fibrosis. Molecules. 2023; 28(23):7936. https://doi.org/10.3390/molecules28237936

Chicago/Turabian Style

Li, Jin-Zhong, Ning Chen, Nan Ma, and Min-Ran Li. 2023. "Mechanism and Progress of Natural Products in the Treatment of NAFLD-Related Fibrosis" Molecules 28, no. 23: 7936. https://doi.org/10.3390/molecules28237936

Article Metrics

Back to TopTop