Skip to main content

Pharmacological and therapeutic effects of natural products on liver regeneration-a comprehensive research

Abstract

Liver regeneration (LR) refers to the physiological process by which hepatocytes undergo cellular proliferation to restore the structure and function of the liver following significant hepatocyte loss due to injury or partial hepatectomy (PH). While the liver possesses a remarkable regenerative capacity, this process is tightly regulated to ensure appropriate cessation once homeostasis is reestablished. Various strategies, including technological interventions and pharmacological agents, have been explored to enhance LR. Among these, natural products have emerged as promising candidates for promoting LR. For instance, quercetin, a natural compound, has been shown to enhance LR following PH by maintaining redox homeostasis and stimulating hepatocyte proliferation. However, natural products present certain limitations, such as poor solubility and low bioavailability, which may hinder their clinical application. Modifications in the formulation and mode of administration have demonstrated potential in overcoming these challenges and optimizing their pharmacological effects. Recent advancements in research have further highlighted the growing relevance of natural products, including traditional Chinese medicine (TCM), in the context of LR. Despite this progress, a comprehensive and systematic review of their roles, mechanisms, and therapeutic potential remains lacking. This review aims to bridge this gap by summarizing natural products with demonstrated potential to promote LR. Drawing on data from PubMed, Web of Science, and CNKI databases, it elucidates their pharmacological effects and regulatory mechanisms, providing a valuable reference for future research and clinical application in the field of LR.

Introduction

Liver regeneration (LR) refers to the liver's ability to restore its structure and function following partial hepatectomy (PH) or chemical injury. This process is primarily driven by hepatocyte proliferation, supported by other cell types such as biliary epithelial cells, which can differentiate into hepatocytes and contribute to tissue repair. Promoting LR is clinically significant; for instance, effective recovery from liver transplantation (LT) relies on robust regenerative capacity. According to 2023 statistics, about 32.4% of the world's population is affected by non-alcoholic fatty liver disease (NAFLD), and the number of deaths has doubled in the last three decades (9,3757 in 1990 and 16,8969 in 2019), has become an important reason of LT in Europe (8.4% of liver transplants in 2016) and United States (21% of liver transplants in 2018) [1], and after LT the transplanted liver rapidly starts the regeneration process to adapt to the needs in the recipient's body, so excellent liver regenerative capacity helps the transplanted liver to regain its function more quickly. Liver regenerative capacity can be enhanced by traditional pharmacological interventions or novel mesenchymal stem cell transplantation techniques [2] and the establishment of decellularized liver scaffolds [3], etc. Among these, natural product-derived drugs have garnered significant attention due to their favorable safety profile and abundant availability, offering promising potential in the field of LR research.

Natural products refer to organic compounds with specific chemical structures and biological activities produced by plants, animals, and microorganisms, which can be divided into flavonoids, alkaloids, polysaccharides, saponins, etc., and have the advantages of wide sources and low toxicity. In addition, their excellent pharmacological effects have also been under close scrutiny by scientists, such as anti-inflammatory, antioxidant [4, 5], antimicrobial [6, 7], antiviral [8, 9], and immunomodulatory effects [10, 11], these outstanding pharmacological effects provide the feasibility of natural products in treating cancer [12], viral diseases [13] and cardiovascular diseases [14]. In addition, natural products gerberellin [15] and silymarin [16], which are natural hepato-protective agents, have shown therapeutic effects on liver diseases. Meanwhile, Geraniol (Ger) and dioscin are also able to promote LR by promoting hepatocyte proliferation and reducing liver injury. Notably, in addition to natural product monomers, Chinese herbal compound preparations also showed excellent effects in promoting LR. For example, SiNiSan (SNS), a traditional formula, was able to promote hepatocyte proliferation by resisting oxidative stress and facilitated LR in mice after 70% PH [17]. All these results suggest that natural products have great potential in promoting LR.

Collectively, accumulated evidence indicates that natural products exert a promotive effect on LR. This review provides a comprehensive summary of natural product monomers with the potential to enhance LR, along with their underlying regulatory mechanisms, aiming to support drug discovery efforts in this domain.

Overview of LR

LR refers to the process that after PH or liver damage, the number of hepatocytes decreases dramatically, and a variety of feedback signals can promote the proliferation of hepatocytes in the G0 phase, and the residual hepatocytes compensate for the loss of and damage to liver tissues and restore the liver's physiological functions through cell proliferation. The organism possesses the ability to precisely detect the size of the regenerating liver and halt LR at the appropriate time.

LR consists of three phases, including priming phase (the first stage), proliferative phase (the second stage), and termination phase (the third stage) (Fig. 1). The first (I) stage is priming phase, in which interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) are key factors that stimulate the transition of hepatocytes from G0 phase to the G1 phase of the cell cycle [18]. Subsequently, the second (II) stage is proliferative phase, in which complete mitogens and auxiliary mitogens play an important role. Complete mitogens such as epidermal growth factor (EGF), hepatocyte growth factor (HGF), and transforming growth factor-α (TGF-α), can directly stimulate DNA synthesis and cell proliferation. Moreover, accessory mitogens include bile acids (BA), norepinephrine (NE), estrogen, and vascular endothelial growth factor (VEGF), which can promote hepatocyte proliferation by amplifying or accelerating the effects of full mitogens and may also lead to delayed LR in their absence [19]. Stimulated by these cytokines, hepatocytes complete mitosis. Finally, the third (III) stage is termination phase, transforming growth factor-β (TGF-β) is a negative regulatory factor, which inhibits hepatocyte mitosis and proliferation, stopping LR [20]. After going through these three stages, the liver completes regeneration.

Fig. 1
figure 1

The outline of the LR process. After PH, the remaining hepatocytes undergo three phases, including priming phase, proliferative phase and termination phase, and a number of factors including IL-6, TNF-α, EGF, HGF, TGF-α, BA, NE, oestrogen, VEGF, and TGF-β are involved in this process, which collectively help in the recovery of the liver. LR liver regeneration, PH partial hepatectomy, IL-6 interleukin-6, TNF-α tumor necrosis factor-α, EGF epidermal growth factor, HGF hepatocyte growth factor, TGF-α transforming growth factor-α, BA bile acids, NE norepinephrine, VEGF vascular endothelial growth factor, TGF-β transforming growth factor-β

Several factors influencing the process of LR warrant attention. First, hepatic portal artery hemodynamics play a pivotal role. Studies have demonstrated that increased portal blood flow and pressure can stimulate LR by activating HGF, promoting hepatocyte hypertrophy, and inhibiting apoptosis [21]. Second, the accumulation of acetylcholine has been found to enhance LR and protect the liver from surgery-induced injury via the action of M3 muscarinic acetylcholine receptor antagonists [22]. Finally, autophagy, a process of breaking down intracellular components through lysosomes, maintains and regulates homeostasis within eukaryotic cells [23]. Reduced autophagic activity significantly impairs LR after PH, whereas enhancing autophagy through mTOR-independent inducers markedly improves LR in aging livers [24]. Collectively, these findings have guided the identification of several potential therapeutic agents targeting LR, offering promising avenues for the treatment of liver diseases.

Currently, while various drugs have been identified to promote LR, natural products have demonstrated distinct advantages in recent years, highlighting the need for further exploration of their potential in facilitating LR.

Single-component natural products for LR

Currently, studies have proved that natural product monomers have excellent performance in treating liver diseases, such as resveratrol (RESV) for NAFLD [25] and silymarin for hepatic fibrosis [26]. In addition, the effects of natural product monomers on LR have been well-documented, including those derived from saponins, flavonoids, phenols, and terpenoids (Table 1).

Table 1 The role of single components of natural products in promoting LR

Saponins

Dioscin, a natural steroidal saponin, mainly presenting in andrographis paniculata of the dioscoreaceae family, exhibits various pharmacological effects such as anti-inflammatory, antioxidant [27], and anti-apoptosis [28], and has been shown to treat NAFLD [29], relieve acetaminophen (APAP) -induced liver injury [30], and improve hepatic ischemia–reperfusion (I/R) injury [31]. In addition, dioscin had a positive effect on LR, as evidenced by the ability of dioscin to promote recovery and regeneration of the liver in the 70% PH mice model [32]. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are well-recognized biomarkers for assessing liver injury, with elevated serum levels indicating hepatic damage. Recent studies have demonstrated that dioscin effectively reduces serum ALT and AST levels, thereby mitigating liver injury in mice subjected to 70% PH [33]. Furthermore, proliferating cell nuclear antigen (PCNA) is an important cofactor for DNA replication [34], and the cell cycle proteins (Cyclins), as well as cell cycle protein-dependent kinases (CDKs), are at the core of cell cycle regulation [35], all of which are crucial indicators for evaluating the proliferative state of cells. It is well-documented that dioscin significantly enhances the expression of PCNA, cyclin-dependent kinase 4 (CDK4), cyclin-dependent kinase 2 (CDK2), cell cycle protein D1 (Cyclin D1), and cell cycle protein E1 (Cyclin E1) in primary hepatocytes and AML-12 cells in mice, thereby promoting cell proliferation and LR [32]. In summary, dioscin is widely used as a highly potent and low toxicity natural hepatoprotective agent for the treatment of various liver diseases, in which its effects on LR are also outstanding, not only promoting LR through direct stimulation of hepatocyte proliferation but also reducing liver injury and providing favorable conditions for LR. However, dioscin has low solubility, poor stability [36], and poor oral bioavailability [37] limiting its clinical application. Researchers have designed hybrid micelles consisting of D-α-Tocopherol polyethylene glycol 1000 succinate (TPGS) and Soluplus® copolymers encapsulating insoluble dioscin to improve its solubility and stability [36], it provides more possibilities for the clinical use of dioscin for LR.

Panax notoginseng saponin (PNS) is a kind of saponin natural product extracted from the traditional Chinese herb Panax notoginseng, which possesses anti-inflammatory [38] and anti-oxidative stress [39] effects. PNS has shown efficacy in treating NAFLD [40], preventing hepatic fibrosis [41], and alleviating alcoholic liver disease (ALD) [42]. Recent studies have indicated that PNS has the potential to promote LR. In the 70% PH mouse model, PNS was able to reduce the levels of serum ALT and AST, alleviate liver injury, and increase the expression of PCNA, which significantly promotes the proliferation of primary mouse hepatocytes. Moreover, PNS can reduce apoptosis in primary mouse hepatocytes and facilitate the regeneration of damaged livers [43]. In summary, PNS contributes to LR by promoting hepatocyte proliferation and inhibiting hepatocyte apoptosis. However, careful consideration should be given to dosage and individual patient characteristics to minimize the risk of adverse effects.

Flavonoids

Baicalin, a flavonoid natural product derived from scutellaria baicalensis, exhibits remarkable pharmacological effects such as antioxidant [44], anti-inflammatory [45], and lipid metabolism regulation [46]. As the study has progressed, baicalin has shown hepatoprotective effects in hepatic fibrosis [47], ALD [48], and NAFLD [49]. Meanwhile, recent studies have shown that in the APAP-induced liver injury mice model, baicalin significantly decreased serum ALT and AST levels, and increased the expression of PCNA and Cyclin D1, promoting hepatocyte proliferation [50]. In addition, interleukin-18 (IL-18) is a cytokine that can promote the proliferation of hepatocytes after hepatectomy and is beneficial for LR [51], NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, a multimeric protein complex that stimulates the generation of interleukin-1β (IL-1β) and IL-18 and is a target of natural products to alleviate liver injury [52]. Notably, it has been reported that NLRP3 is closely associated with the regeneration of a variety of organs or tissues, including epithelial regeneration of lung tissue [53], bone regeneration [54], skin regeneration [55], nerve regeneration [56]. Additionally, its role in liver tissue regeneration has also been confirmed [57]. Existing literature has thoroughly demonstrated that baicalin can activate NLRP3, leading to increased levels of IL-1β and IL-18 in the liver, thereby promoting hepatocyte proliferation [50]. These findings have confirmed that baicalein can be used to promote LR, and its wide range of sources, and low toxicity [58] give it great potential and advantages in the field of liver disease treatment.

Dihydromyricetin (DMY) is a flavonoid mainly found in Garcinia cambogia, a vine plant, and is a natural antioxidant [59] with anti-inflammatory effects [60]. DMY has been used in treating NAFLD [61] and APAP-induced liver injury [62], and it also promotes LR. In the CCl4-induced acute liver injury mice model, DMY reduced inflammation and oxidative stress in the liver by attenuating the inflammatory response, lowering serum levels of ALT and AST, and enhancing superoxide dismutase (SOD) activity [63]. Additionally, apoptosis-related proteins Caspase family and cytochrome c, are essential for the initiation and execution of apoptosis [64, 65], and the literature has amply demonstrated that DMY can reduce hepatocyte apoptosis by inhibiting the expression of cytochrome c and Caspase -8, 3, 6 and 9. At the same time, DMY increased the expression of PCNA, which promotes hepatocyte proliferation [63]. These findings suggest that DMY is characterized by a synergistic effect of anti-inflammatory, antioxidant, hepatocyte proliferation, and apoptosis reduction to promote LR after liver injury.

Quercetin (Que) is a polyphenolic flavonoid found in the stem bark and fruit skin of plants, and its anti-inflammatory [66], antioxidant [67], and anti-apoptotic effects [68] have been demonstrated in studies. Que, as a natural hepatoprotective agent, its efficacy in liver disease is prominent, such as NAFLD [69], hepatic fibrosis [70], and alcohol-induced liver injury [71]. In addition, it was shown that Que also has a prominent effect on LR, in the 70% PH rats model, Que can increase the expression of PCNA, promote hepatocyte proliferation, and significantly reduce hepatocyte apoptosis after PH [72]. In addition, maintaining redox balance in the liver can promote LR [17], Malondialdehyde (MDA) is an important biomarker for assessing lipid peroxidation [73]. Additionally, SOD [74] plays a key role in antioxidant defense, while glutathione (GSH) [75] is an important antioxidant in the body. GSH helps regulate reactive oxygen species (ROS) levels in vivo, thereby reducing cellular damage associated with lipid peroxidation. The study clearly demonstrates that Que can reduce MDA levels in the liver while increasing GSH levels and SOD activity, thereby exerting antioxidant effects and promoting LR [72]. In summary, Que has been demonstrated as a natural hepatoprotective agent and can promote LR after PH.

Phenols

Curcumin (CUR), a phenolic natural product found in turmeric, with biological activities such as anti-inflammatory [76], antioxidant [77], and antibacterial [78], and has been shown to alleviate various liver diseases, such as hepatic fibrosis [79], NAFLD [80], and drug-induced liver injury [81]. Meanwhile, in the 70% PH rat model, CUR was able to promote LR. CUR exhibited antioxidant activity that can reduce the level of MDA and increase the levels of SOD activity and GSH in liver tissues. Meanwhile, CUR also increased the expression of PCNA to promote hepatocyte proliferation and reduced apoptosis in hepatocytes [82]. Currently, there are limited reports on the mechanisms through which CUR promotes LR, highlighting the need for further investigation into its potential for enhancing LR. However, the phenolic structure of CUR results in very low solubility, poor stability, and low bioavailability, which greatly hampers the development of its clinical applications, to ameliorate these limitations, some improvements can be made to its delivery format to facilitate its clinical application in LR.

RESV, a phenolic compound found mainly in grapes, exhibits anti-inflammatory [83], anti-tumor [84], and other pharmacological effects. Furthermore, RESV has been reported to be effective in alcoholic fatty liver disease (AFLD) [85], NAFLD [25], and hepatic fibrosis [86]. In addition, RESV has shown the potential to promote LR. Firstly, RESV can reduce liver injury, it significantly suppressed cytochrome P450 (CYP450) activity, especially CYP2E1, CYP3A11, and CYP1A2, thus reducing the production of hepatotoxic N-acetyl-p-benzoquinone imine (NAPQI), the serum ALT and AST levels were also reduced, and the use of RESV pretreatment increased GSH levels and reduced oxidative stress, all of which indicated that APAP-induced liver injury was attenuated with RESV. Secondly, RESV increased the levels of PCNA, as well as the cell cycle regulatory proteins CyclinD1 and CDK4, which promoted the proliferation of hepatocytes [87]. In a word, RESV’s high safety profile and wide availability underscore its potential to promote LR in future applications.

Schisandrin B (SolB) is a phenolic natural compound derived from the herb Schisandra chinensis. It exhibits significant pharmacological properties, including anti-aging and anti-inflammatory effects [88]. As a natural hepatoprotective agent, SolB is able to treat acute hepatotoxicity caused by APAP [89], as well as cholestatic liver injury [90]. After liver injury, Kupffer cells secrete IL-6, an inflammatory cytokine that induces hepatocyte dedifferentiation and thus promotes LR [91], in addition, HGF and EGF [92] are factors that can promote LR. Experiments have shown that in the 2/3 PH mice model, SolB increased the levels of IL-6, HGF, and EGF, induced the expression of Cyclin D1 and PCNA, and promoted LR [93]. Moreover, p53 is an important tumor suppressor gene and its activation leads to cell cycle arrest, cellular senescence, and even cell death [94]. P21, a known cell cycle protein-dependent kinase inhibitor, is mainly transcriptionally regulated by p53 [95], and both are capable of adversely affecting LR. It should be mentioned that in the APAP-induced liver injury model, SolB was able to reduce the expression of p53 and p21 [96]. In addition, B-cell lymphoma-2 (Bcl-2) family proteins are able to influence apoptosis by controlling the permeability of the mitochondrial outer membrane, and this family can be categorized into pro-apoptotic proteins and anti-apoptotic proteins (e.g. Bcl-2) [97]. Notably, it is well documented that SolB can increase the expression of Bcl-2, decrease hepatocyte apoptosis, and have positive effects on LR [96]. Therefore, SolB represents a promising therapeutic option for enhancing LR, particularly in the context of liver recovery following partial hepatectomy (PH) or liver transplantation.

Rosmarinic acid (RA), a phenolic acid ingredient primarily found in rosemary, with anti-inflammatory, antioxidant, anti-apoptotic [98], and antimicrobial [99] effects, has been shown to treat APAP-induced liver injury [100] and nonalcoholic steatohepatitis (NASH) [101]. In addition, RA has the potential to promote LR. In the 2/3 PH mouse model, RA treatment reduced serum levels of alanine ALT, AST, and alkaline phosphatase (ALP). Furthermore, RA increased the expression of PCNA, promoted hepatocyte proliferation, and alleviated liver injury [102]. These results suggest that RA is a promising natural product that enhances LR. However, further investigation is needed to fully understand the role of RA in LR, and the safety of its clinical application requires additional research.

Terpenoids

Ursolic acid (UA), a terpenoid found mainly in bearberry, exhibits strong pharmacological activities such as anti-inflammatory [103], antibacterial, and antioxidant [104], and UA is effective in a number of liver diseases, such as NAFLD [105] and hepatic fibrosis [106]. Meanwhile, UA exhibited a remarkable ability to promote LR. In the 70% PH mouse model, UA significantly enhanced DNA synthesis in hepatocytes. Additionally, the expression of Cyclin D1, cyclin E, and CCAAT/enhancer-binding protein β (C/EBPβ) was elevated. C/EBPβ is a protein known to facilitate LR following activation, suggesting that UA may promote hepatocyte proliferation and support LR after 70% PH [107]. However, UA has shown shortcomings such as low bioavailability and solubility, poor targeting, and rapid metabolism, and has certain limitations in clinical application. Therefore, researchers must implement strategies to address these deficiencies, thereby enhancing the effectiveness of their application in promoting LR.

Oleanolic acid (OA) is a terpenoid mainly found in the leaves of Olea europaea and the fruit of Ligustrum officinale, with anti-inflammatory, antioxidant [108], cardioprotective [109], and hypoglycemic effects [110], and research has shown that OA is a hepatoprotective agent that can alleviate acute liver injury [111], cholestatic liver injury [112], and promote LR. The promotional effects on LR were demonstrated by the ability to promote liver recovery after 70% PH. It is well documented that in the 70% PH mouse model, OA can increase the expression of PCNA in vivo and in vitro [113]. In addition, forkhead box protein O1 (FOXO1), a transcription factor that can regulate cellular processes, and inhibition of its expression promotes cell proliferation [114]. Retinoblastoma-like protein 2 (RBL2) is capable of interacting with other substances to affect cell proliferation, such as the transcription factor E2F, and the inhibition of the RBL2/E2F4 complex can promote cell proliferation [115]. Moreover, cyclin-dependent kinase inhibitor 1B (CDKN1B) is a known cell cycle-dependent kinase inhibitor that inhibits cell proliferation [116], and it has been reported that OA can inhibit the expression of FOXO1, RBL2, and CDKN1B, and promote the proliferation of hepatocytes [113]. Therefore, OA can promote LR following 70% PH through various mechanisms. Ger is a terpenoid natural compound that exists in plants of the Brassicaceae and Geranylgeranyl family, with many important pharmacological activities such as anti-inflammatory [117], antioxidant, and antimicrobial [118]. Studies have demonstrated that Ger has hepatoprotective properties. For example, Ger prevents cyclophosphamide-induced hepatotoxicity [119], improves bisphenol-A-induced liver injury [120], and promotes LR. As we all know, inflammatory cytokines IL-6 and TNF-α are crucial for LR [121], in the 70% PH rat model, Ger can reduce serum ALT levels, increase the expression of IL-6 and TNF-α, as well as the mitotic activity of hepatocytes, and promote LR [122]. In conclusion, the role of Ger in inducing LR after PH has been confirmed, but the specific molecular mechanisms need to be further investigated.

In summary, all these natural products have great potential in promoting LR, and it is interesting to note that different natural products may promote LR through the same mechanism of action, most of them, such as dioscin, PNS, Baicalin, RESV, SolB, UA, and OA, can up-regulate the expression of Cyclins (such as PCNA, CDK2, CDK4, Cyclin D1 and Cyclin E1) to directly promote the proliferation of hepatocytes, and in addition the antioxidant effect of Que, CUR, the regulation of inflammatory factors by DMY, Ger, and the anti-apoptotic effect of DMY, SolB all create favorable conditions for LR.

Regulatory mechanisms of herbal medicines on LR

In summary, natural product monomers are promotive for LR and can repair damaged liver, and their regulatory mechanisms on LR have received increasing attention. In recent years, several signaling pathways have been identified as being closely associated with LR (Fig. 2).

Fig. 2
figure 2

Signaling pathways of natural products for LR. Activation of Wnt/β-catenin, PI3K/Akt, Notch and Nrf2 signalling pathways promotes cell proliferation and is strongly associated with LR. LR liver regeneration. (by FigDraw)

Wnt/β-catenin signaling pathway

The Wnt/β-catenin signaling pathway, comprising the Wnt ligand, Frizzled receptor, LRP5/LRP6 co-receptor, Dishevelled (Dvl) protein, glycogen synthase kinase-3β (GSK3β), β-catenin, TCF/LEF transcription factors, Axin protein, and CK1 kinase, regulates cell proliferation, differentiation, and tissue development. This is achieved through the modulation of β-catenin's stability and its transcriptional activity in the nucleus. Notably, activating the Wnt/β-catenin signaling pathway includes the following steps. Firstly, Wnt ligands bind specifically to Frizzled receptors and LRP5/6 co-receptors on the cell membrane, and this binding triggers phosphorylation and dephosphorylation reactions of proteins in the cytoplasm, which can activate β-catenin. Subsequently, β-catenin translocates to the nucleus, where it interacts with TCF/LEF transcription factors and activates the expression of target genes, such as Matrix Metalloproteinases (MMPs) and cellular myelocytomatosis oncogene (c-Myc), which play a role in the regulation of cell proliferation and can thus promote cell proliferation [123]. Therefore, the Wnt/β-catenin signaling pathway is closely associated with LR. Additionally, several natural products have been demonstrated to enhance LR through the modulation of the Wnt/β-catenin signaling pathway. For example, tanshinone IIA (TSA) is a phenanthrenequinone compound extracted from traditional Chinese medicine (TCM) Danshen and β-catenin is a key component of the classic Wnt signaling pathway and has been shown to regulate hepatocyte proliferation, it is well documented that moderate concentrations of TSA can increase the expression of β-catenin, activate the Wnt/β-catenin signaling pathway, and stimulate the proliferation of hepatic oval cells, thus enhancing the repair and regeneration of injured livers and improving LR after orthotopic LT [124]. Furthermore, SNS is a traditional Chinese medicine formula, which can promote liver stem cell differentiation through the Wnt/β-catenin signaling pathway, thereby alleviating liver injury. The study has shown that 14 days after treatment with SNS in PH rats, phosphorylation of GSK3β was significantly increased. GSK3β is a key component of the Wnt signaling pathway, and when the Wnt signaling pathway is inactivated, GSK3β degrades β-catenin, resulting in low intracellular levels of β-catenin. However, when the Wnt signaling pathway is activated, GSK3β is inactivated by phosphorylation, and the level of β-catenin is increased. As a result, the levels of β-catenin and Dvl2 increased, after which β-catenin was transferred to the nucleus and bound to Lymphoid enhancer-binding factor 1 (LEF1) receptor to activate the expression of c-Myc and Cyclin D1, which are related to cell proliferation and differentiation, and activated the Wnt/β-catenin signaling pathway, promoting LR in rats after hepatectomy [125]. Although SNS has shown potential efficacy in promoting LR, its side effects and contraindications must be carefully considered, and its use should adhere strictly to medical guidance. In conclusion, the Wnt/β-catenin signaling pathway represents a highly promising target for enhancing LR, warranting further investigation to fully explore its therapeutic potential.

PI3K/Akt signaling pathway

The PI3K/Akt signaling pathway is a complex network consisting of the following components: cell surface receptors (e.g., tyrosine kinase receptor), phosphatidylinositol 3 kinase (PI3K) second messenger phosphatidylinositol 3,4,5-trisphosphate (PIP3), protein kinase B (Akt), phosphatidylinositol-dependent protein kinase 1 (PDK1), mechanistic targets of rapamycin complexes 1, 2 (mTORC1, mTORC2), and Phosphatase and Tensin Homolog Deleted on Chromosome 10 (PTEN). Notably, the activation of the PI3K/Akt signaling pathway can affect cell proliferation and involves the following steps. Firstly, growth factors or cytokines bind to cell membrane surface receptors, initiating the activation of PI3K, which phosphorylates phosphatidylinositol 4,5-bisphosphate (PIP2) to form PIP3. PIP3 plays a critical role in recruiting Akt and PDK1 to the plasma membrane. Subsequently, Akt is fully activated through phosphorylation by PDK1 and mTORC2. The activated Akt then regulates various cellular processes, including survival, proliferation, metabolism, and apoptosis, by phosphorylating key substrate proteins such as Bcl2-Associated X (BAX), GSK3β, and cell cycle inhibitors, p21 and p27 [126]. Activation of the PI3K/Akt signaling pathway plays a key role in the early regenerative response after PH. Early studies have shown that inhibition of the PI3K/Akt signaling pathway inhibits hepatocyte replication on the one hand, and leads to structural abnormalities of vacuolization, lipid deposition, and glycogen accumulation of regenerating hepatocytes on the other hand, which attenuates LR [127]. Additionally, it has been reported in studies that some natural products can promote LR through PI3K/Akt signaling pathway. For example, salidroside, a natural product found in the roots of rhodiola rosea, is well documented that salidroside can act on the PI3K/AKT/Gsk3β signaling pathway, significantly reduce ROS production and lipid accumulation, inhibit apoptosis of hepatocytes and promote the proliferation of hepatocytes, which can promote LR [128]. PNS is a kind of saponin natural product extracted from the traditional Chinese herb Panax notoginseng. In the mice model of PH, PNS significantly increased the phosphorylation level of PI3K and Akt, and the activation of Akt initiated the phosphorylation of the downstream target mTOR on the one hand, playing a role in promoting hepatocyte proliferation; on the other hand, Akt initiated the phosphorylation of another target Bcl-2-associated agonist of cell death (Bad), one of the pro-apoptotic proteins of the Bcl-2 family, and in the phosphorylated state, Bad promoted cell survival, in the dephosphorylated state, it exerts the opposite effect to promote apoptosis [129]. Therefore, PNS promotes LR by up-regulating the PI3K/Akt/mTOR and Akt/Bad signaling pathways to promote hepatocyte proliferation and transplantation of hepatocytes for apoptosis [43]. All in all, the therapeutic effects of herbal ingredients through the PI3K/Akt signaling pathway provide more strategies and ideas to promote LR.

Notch signaling pathway

The Notch signaling pathway plays a crucial role in regulating cell proliferation and apoptosis. It involves several key components, including the Notch receptors (Notch1, 2, 3, and 4), ligands (Jagged-1, Jagged-2, and Delta-1, Delta-3, Delta-4), the DNA-binding protein CSL, and a range of downstream effectors. Normally, activation of the classical Notch signaling pathway involves the following process. Firstly, the Notch protein is transported to the endoplasmic reticulum as a single-chain precursor, where it undergoes glycosylation and is transported to the Golgi for cleavage at the S1 site to form the mature Notch receptor which is transported to the surface of the cell membrane, where it binds to ligands (Jagged/Delta) on the neighboring cell membrane, and this binding triggers the cleavage of the Notch receptor at the S2 site, and the intermediate formed is cleaved by γ-secretase to undergo S3 site cleavage, releasing theNotch1 intracellular domain (NICD). Then, NICD binds to the transcription factor CSL to recruit coactivators that activate the transcription of downstream target genes [130]. Activating the Notch/Jagged signaling pathway is important for LR. On the one hand, activation of the Notch signaling pathway can initiate the transcription of downstream target genes of the Hes and Hey families, which are associated with cell proliferation, and promote hepatocyte proliferation. On the other hand, its activation contributes to the formation of bile ducts, which is an important link in the LR [131]. Furthermore, the researchers demonstrated that in the 2/3 PH rat model, the Notch signaling pathway regulates the cell cycle of proliferating hepatocytes involved in LR and is important for LR processes. After hepatectomy in rats, the Notch signaling pathway was significantly activated, in contrast to the addition of inhibitors of γ-secretase (Notch receptor S3 cleavage essential enzyme for conversion to NICD, a key enzyme in Notch signaling pathway activation) revealed that abnormalities in the S and M phases of the hepatocyte proliferation cycle resulted in delayed LR after PH in rats [132], which indicates that the normal function of the Notch signaling pathway is crucial for the proper progression of LR. Notably, studies have found that natural products can promote LR by activating the Notch signaling pathway. For instance, dioscin, a saponin-like natural product, can significantly increase Notch1 and Jagged1 levels in the 70% PH mice model, upregulate the expression of PS1, and increase PS1-dependent γ-secretase activity, leads to the nuclear translocation of NICD1, and activate the Notch1/Jagged1 signaling pathway, leading to the promotion of the expression of downstream targets associated with cell proliferation: (Hey1, Hes1, EGFR, VEGF) and cell cycle regulatory proteins (CyclinD1, CyclinE1, CDK4, and CDK2) to promote hepatocyte proliferation and LR [32]. Notably, however, whether other natural products can promote LR through the Notch signaling pathway needs to be further explored by researchers.

Nrf2 signaling pathway

The nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway consists of the Nrf2, the kelch-like ECH-associated protein 1 (keap1), the antioxidant response element (ARE), and a series of target genes. The activation of the Nrf2 signaling pathway involves several key processes. Under normal conditions, the Nrf2 protein is tightly bound to Keap1. However, upon oxidative stress or other stimuli, this binding is disrupted. Subsequently, Nrf2 translocates to the nucleus, where it binds to the ARE and activates the transcription and translation of target genes. It has been shown that activation of Nrf2 can advance the mitotic process of cell proliferation through the Cyclin A2 and Wee1/Cdc2/Cyclin B1 pathways, whereas inhibition of Nrf2 delays mitosis in hepatocytes and affects the proliferative process [133].

The Nrf2 signaling pathway has demonstrated potential in promoting LR. Furthermore, natural products have been shown to enhance LR through modulation of Nrf2 activity. For example, CUR and Que, both natural activators of Nrf2, exhibit beneficial effects on LR by promoting cell proliferation, reducing apoptosis, and exerting antioxidant effects [134]. In conclusion, future studies will further elucidate the mechanism of the Nrf2 signaling pathway and provide new insights into LR.

Self-assembled nanodrug delivery systems

The self-assembled nanodrug delivery system (SANDDS) is an organized nanostructure characterized by a well-defined structure and stability, which is formed through the spontaneous aggregation of molecules via non-covalent interactions, without the need for external intervention. Notably, SANDDS offers several advantages, including simple synthesis, reduced toxicity, and enhanced bioavailability [135].

SANDDS has shown great potential in the biomedical field to enable drugs to perform their optimal roles. For example, compounds such as alkaloids, flavonoids, organic acids, and terpenoids possess multiple functional groups and action sites within their structures, which allow them to self-assemble into self-assembled nanodrug delivery systems (SANDDS). This self-assembly process enhances the bioavailability and therapeutic efficacy of these natural products [136]. For example, CUR by green reprecipitation method synthesized pure CUR nanoparticles (CNPs) without carrier and loaded with cardiolipin LP (liposomes) can improve the poor solubility of curcumin and have more prominent efficacy in Alzheimer's disease (AD) [137].

More importantly, SANDDS can improve the shortcomings of solubility and bioavailability of some of the above-mentioned natural products that promote LR (Table 2), such as DMY, Que, CUR, RESV, RA, and UA, which is very important for the future treatment of liver diseases. DMY, its low solubility, poor permeability, unstable nature, and rapid metabolism in vivo lead to poor bioavailability and unsatisfactory pharmacodynamics [138] [139]. DMY-MS self-assembled polymeric micelles formed by self-assembly of DMY with Polyethylene glycol-15-hydroxystearate (Solutol®HS15) as a carrier significantly improved the solubility, oral bioavailability, and anti-alcohol resistance of DMY and had a slow-release [140]. CUR also has the disadvantages of poor solubility and stability, it can be self-assembled and encapsulated in H. pluvialis protein (HPP)-galactose (GAL) nano complex (HPP-GAL) nanoparticles by hydrogen bonding to synthesize HPP-GAL-CUR nano preparations, which improves the stability and bioavailability of CUR, and in the ALD mice model, HPP-GAL-CUR exhibits good liver-targeting properties due to the ability of GAL itself to accumulate in the liver, and is more effective than free CUR for the treatment of ALD in mice [141]. SANDDS can also solve the challenges of RESV applications, RESV has poor water solubility and low chemical stability and is susceptible to chemical degradation stimulated by exogenous environmental stresses [142, 143]. Notably, the solubility and stability of RESV were improved by encapsulating RESV in nanoparticulate Res NPs formed by self-assembly of α-lipoic acid (α-LA), lactobionic acid (LA), and glycogen (Gly) into nanocarriers. In addition, in the NAFLD mice model, Res NPs were able to target the liver with better therapeutic efficacy compared to the free form of RESV [144] and facilitate the future use of RESV in the treatment of clinical diseases, including LR. Furthermore, RA as a key polyphenolic antioxidant can promote LR, but its poor stability as well as low bioavailability have greatly limited its translation into clinical applications. Nanoparticles of PRA NPs synthesized by self-assembled RA, 1,4-phenylene diboronic acid (PBA), and glycerol monooleate (GMO), improved the stability of RA and were able to be targeted to the liver with increased antioxidant activity relative to free RA in the mouse model of acute liver injury [145]. UA is also one of the natural products mentioned above that promotes LR, researchers developed an amphiphilic self-assembled nanomedicine consisting of UA, LA, and low-polyamidoamine (low-PAMAM) dendrimer: UA-G0/G1-LA, which solved the problem of low solubility of UA, and in the H22 mice model, the positive electric charge and nano size of UA-G0/G1-LA helped to be targeted to the tumor site and effectively inhibited the growth of liver tumors, which significantly improved the retention time and bioavailability of UA in vivo [146]. In addition, TSA is a natural product that can promote LR [124], but some limitations exist in its clinical use, such as poor solubility, poor photothermal stability, short elimination half-life, poor hepatic targeting, and strong hepatic first-pass effect [147], which greatly reduces its bioavailability. mPEG-DCA is a drug carrier that can control the release of the drug, prolong the circulating time, and improve the drug accumulation (EPR effect) in the tumor. Therefore, mPEG-DCA was used as a carrier for co-loading TSA and GA to synthesize nano preparations, which improved the solubility and chemical stability of TSA and GA. More importantly, it showed good tumor targeting and biocompatibility in H22 tumor-bearing mice, prolonged the mean residence time (MRT) of TSA in mice, and had a better therapeutic efficacy [148].

Table 2 Application of different types of SANDDS to natural products that promote LR

Taken together, the application of SANDDS has the potential to address the limitations of natural products and enhance their therapeutic efficacy, offering promising prospects for the medical field.

Future prospects

Altogether, natural products including ingredients of TCM are rich resources for the discovery of new medicines, and natural products exhibit obvious anti-inflammatory, antioxidant, and hepatoprotective effects, garnering substantial research interest. This review summarizes the research progress on natural products, including TCM, in promoting LR. Numerous studies have demonstrated that natural products can enhance LR by modulating various signaling pathways. Although studies have confirmed the ability of natural products to promote LR, this ability has only been verified in animal experiments, and no substantial progress has been made in relevant clinical trials (Fig. 3), natural products have the advantages of low toxicity, wide sources, and diverse structures, and some marketed natural product formulations can promote hepatocyte repair and regeneration, such as the Five Spirit pill (TCM) and Huganning Tablets, they can also be transformed into lead compounds [149] for the development of new drugs to promote LR based on their diverse structural properties, thus these natural products have potential for clinical applications. Moreover, the exploration of the targets of action of natural products on LR is still limited, and the dynamic changes of gene expression, protein changes, and metabolic pathways in the process of LR promotion by natural products can be comprehensively analyzed by multi-omics technology [150, 151] to uncover new targets and molecular mechanisms. Therefore, it remains a challenge in the future to translate the LR-promoting effects of these natural products from animal studies to human applications and to exploit their novel targets for LR.

Fig. 3
figure 3

Experimental progress of natural products on LR. This timeline illustrates the experimental progress of multiple natural products in LR research, covering cell, animal, and human (no human experiments are currently in progress) studies from 2012 to 2024. LR: liver regeneration

However, challenges related to improving their bioavailability and minimizing potential side effects remain unresolved and need to be addressed in both current and future research. One approach to enhancing the bioavailability of natural products while minimizing side effects is the use of targeted drug delivery systems (DDS) [152, 153]. These systems allow for precise delivery of drugs to the intended site of action. Among DDS, nanoparticle-based drug carriers are commonly employed. By encapsulating active ingredients within these nano carriers, their solubility and bioavailability are improved, which in turn enhances their therapeutic efficacy [154]. SANDDS have garnered increasing attention from scientists due to their advantageous properties, these spontaneous nano-formulations, formed through non-covalent interactions, are relatively simple to prepare and exhibit superior biodegradability and biocompatibility compared to traditional drug nano-formulations [136]. As a result, they hold the potential for enhancing the efficacy of drug treatments for various diseases, including liver disease. For example, Ger is an effective natural product against hepatocellular carcinoma (HCC), but its poor solubility has greatly hindered its clinical application. The multiple bioresponsive self-assembled nano drug delivery system (HSSG) developed by using self-assembly technology to couple Ger with hyaluronic acid (HA) improves the therapeutic efficacy of Ger against HCC, realizes the targeted and controlled release of the drug, and reduces its toxicity [155]. In conclusion, SANDDS is anticipated to enhance the bioavailability and hepatic targeting of natural products that promote LR, with the goal of minimizing adverse effects and improving therapeutic efficacy.

In addition, the use of artificial intelligence (AI) technology to directly screen for drugs that can promote LR from a broad range of natural product molecules holds significant potential for advancing LR research. As AI technology becomes increasingly widespread [156], it is important to note that AI can analyze large volumes of biomedical data to identify new therapeutic targets, including those related to cancer [157], and this approach not only accelerates the target identification process but also improves the accuracy and effectiveness of drug design. PandaOmics is a platform that integrates artificial intelligence and bioinformatics technologies to identify therapeutic targets and biomarkers for various diseases. It utilizes a range of disease-specific models to rank potential targets, aiding in the screening of therapeutic targets. This platform has previously been used to identify targets for the treatment of amyotrophic lateral sclerosis [158]. Taken together, AI demonstrates significant potential in screening drug targets. Moving forward, we anticipate utilizing various AI platforms to identify targets and natural products that can promote LR.

In a word, natural products including TCM have a promising future in LR, and corresponding research and development are in progress, but the acceleration of the clinical translation of natural products to LR, the discovery of natural products that can act on new targets of LR and the reduction of toxic side effects of natural products in LR remain an important task.

Conclusion

In summary, natural products including TCM have shown great potential and unique advantages in promoting LR As research progresses, natural products are expected to offer more effective therapeutic options for enhancing LR. However, improving the targeting capabilities of these products remains a critical challenge. We believe that loading natural products onto SANDDS can be utilized to localize their targeting effects on hepatocytes and promote LR, and that screening for targets and drugs to promote LR using AI and multi-omics technology has unlimited potential in the future. Consequently, it is essential for researchers to engage in sustained and comprehensive studies in this field to drive substantial progress.

Availability of data and materials

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

Abbreviations

α-LA:

α-Lipoic acid

Aa:

Aurantiamide acetate

AD:

Alzheimer's disease

AFLD:

Alcoholic fatty liver disease

AI:

Artificial intelligence

Akt:

Protein kinase B

ALD:

Alcoholic liver disease

ALP:

Alkaline phosphatase

ALT:

Alanine aminotransferase

APAP:

Acetaminophen

ARE:

Antioxidant response element

AST:

Aspartate aminotransferase

BA:

Bile acids

Bad:

Bcl-2-associated agonist of cell death

BAX:

Bcl2-Associated X

Bcl-2:

B-cell lymphoma-2

CDKs:

Cell cycle protein-dependent kinases

CDK2:

Cyclin-dependent kinase 2

CDK4:

Cyclin-dependent kinase 4

CDKN1B:

Cyclin-dependent kinase inhibitor 1B

C/EBPβ:

CCAAT/enhancer-binding protein β

c-Myc:

Cellular myelocytomatosis oncogene

CNPs:

Curcumin nanoparticles

CUR:

Curcumin

Cyclins:

Cell cycle proteins

Cyclin D1:

Cell cycle protein D1

Cyclin E1:

Cell cycle protein E1

CYP450:

Cytochrome P450

DDS:

Drug delivery systems

DMY:

Dihydromyricetin

Dvl:

Dishevelled

EGF:

Epidermal growth factor

FOXO1:

Forkhead Box protein O1

GAL:

Galactose

Ger: Geraniol Gly:

Glycogen

GMO:

Glycerol monooleate

GSH:

Glutathione

GSK3β:

Glycogen synthase kinase 3β

HA:

Hyaluronic acid

HCC:

Hepatocellular carcinoma

HGF:

Hepatocyte growth factor

Hif-1α:

Hypoxia inducible factor-1-α

HPP:

H. pluvialis protein

HSSG:

Multiple bioresponsive self-assembled nano drug delivery system

IL-1β:

Interleukin-1β

IL-18:

Interleukin-18

IL-6:

Interleukin-6

I/R:

Ischemia–reperfusion

keap1:

Kelch-like ECH-associated protein 1

LA:

Lactobionic acid

LEF1:

Lymphoid enhancer-binding factor 1

Low-PAMAM:

Low-Polyamidoamine

LR:

Liver regeneration

LT:

Liver transplantation

LP:

Liposomes

MDA:

Malondialdehyde

MMPs:

Matrix Metalloproteinases

MRT:

Mean residence time

mTORC1:

Mechanistic targets of rapamycin complexes 1

mTORC2:

Mechanistic targets of rapamycin complexes2

NAFLD:

Non-alcoholic fatty liver disease

NAPQI:

N-acetyl-p-benzoquinone imine

NASH:

Nonalcoholic steatohepatitis

NE:

Norepinephrine

NICD:

Notch1 intracellular domain

NLRP3:

NOD-like receptor pyrin domain containing 3

Nrf2:

Nuclear factor erythroid 2-related factor 2

OA:

Oleanolic acid

P:

Palmitin

PBA:

1,4-Phenylene diboronic acid

PCNA:

Proliferating cell nuclear antigen

PDK1:

Phosphatidylinositol-dependent protein kinase 1

PH:

Partial hepatectomy

PIP2:

Phosphatidylinositol 4,5-bisphosphate

PIP3:

Phosphatidylinositol 3,4,5-trisphosphate

PI3K:

Phosphatidylinositol 3 kinase

PNS:

Panax notoginseng saponin

PTEN:

Phosphatase and Tensin Homolog Deleted on Chromosome 10

Que:

Quercetin

RA:

Rosmarinic acid

RBL2:

Retinoblastoma-like protein 2

RESV:

Resveratrol

ROS:

Reactive oxygen species

SA:

Scutebarbatine A

SANDDS:

Self-assembled nanodrug delivery systems

SNS:

SiNiSan

SOD:

Superoxide dismutase

SolB:

Schisandrin B

TCM:

Traditional Chinese medicine

TGF-α:

Transforming growth factor-α

TGF-β:

Transforming growth factor-β

TNF-α:

Tumor necrosis factor-α

TPGS:

D-α-Tocopherol polyethylene glycol 1000 succinate

TSA:

Tanshinone IIA

UA:

Ursolic acid

VEGF:

Vascular endothelial growth factor

References

  1. Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol. 2023;79(2):516–37.

    Article  PubMed  Google Scholar 

  2. Hu C, Zhao L, Wu Z, Li L. Transplantation of mesenchymal stem cells and their derivatives effectively promotes liver regeneration to attenuate acetaminophen-induced liver injury. Stem Cell Res Ther. 2020;11(1):88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dias ML, Paranhos BA, Goldenberg R. Liver scaffolds obtained by decellularization: a transplant perspective in liver bioengineering. J Tissue Eng. 2022;13:20417314221105304.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Chen GL, Fan MX, Wu JL, Li N, Guo MQ. Antioxidant and anti-inflammatory properties of flavonoids from lotus plumule. Food Chem. 2019;277:706–12.

    Article  CAS  PubMed  Google Scholar 

  5. Kou RW, Xia B, Wang ZJ, Li JN, Yang JR, Gao YQ, et al. Triterpenoids and meroterpenoids from the edible Ganoderma resinaceum and their potential anti-inflammatory, antioxidant and anti-apoptosis activities. Bioorg Chem. 2022;121:105689.

    Article  CAS  PubMed  Google Scholar 

  6. Shamsudin NF, Ahmed QU, Mahmood S, Ali Shah SA, Khatib A, Mukhtar S, et al. Antibacterial effects of flavonoids and their structure-activity relationship study: a comparative interpretation. Mol. 2022;27(4):1149.

    Article  CAS  PubMed  Google Scholar 

  7. Dowgiallo MG, Miller BC, Kassu M, Smith KP, Fetigan AD, Guo JJ, et al. The convergent total synthesis and antibacterial profile of the natural product streptothricin F. Chem Sci. 2022;13(12):3447–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Teng YF, Xu L, Wei MY, Wang CY, Gu YC, Shao CL. Recent progresses in marine microbial-derived antiviral natural products. Arch Pharmacal Res. 2020;43(12):1215–29.

    Article  CAS  Google Scholar 

  9. Bergamo A, Sava G. Lysozyme: a natural product with multiple and useful antiviral properties. Mol. 2024;29(3):652.

    Article  CAS  PubMed  Google Scholar 

  10. Wang S, Yang Y, Wang Q, Wu Z, Liu X, Chen S, et al. Structural characterization and immunomodulatory activity of a polysaccharide from finger citron extracted by continuous phase-transition extraction. Int J Biol Macromol. 2023;240:124491.

    Article  CAS  PubMed  Google Scholar 

  11. Zhong Z, Vong CT, Chen F, Tan H, Zhang C, Wang N, et al. Immunomodulatory potential of natural products from herbal medicines as immune checkpoints inhibitors: helping to fight against cancer via multiple targets. Med Res Rev. 2022;42(3):1246–79.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Zhang B, Tian L, Xie J, Chen G, Wang F. Targeting miRNAs by natural products: a new way for cancer therapy. Biomed Pharmacother. 2020;130: 110546.

    Article  CAS  PubMed  Google Scholar 

  13. Khwaza V, Oyedeji OO, Aderibigbe BA. Antiviral activities of oleanolic acid and its analogues. Mol. 2018;23(9):2300.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Singhai H, Rathee S, Jain SK, Patil UK. The potential of natural products in the management of cardiovascular disease. Curr Pharm Des. 2024;30(8):624–38.

    Article  CAS  PubMed  Google Scholar 

  15. Zhao L, Wang Y, Liu J, Wang K, Guo X, Ji B, et al. Protective effects of genistein and puerarin against chronic alcohol-induced liver injury in mice via antioxidant, anti-inflammatory, and anti-apoptotic mechanisms. J Agric Food Chem. 2016;64(38):7291–7.

    Article  CAS  PubMed  Google Scholar 

  16. Polyak SJ, Ferenci P, Pawlotsky JM. Hepatoprotective and antiviral functions of silymarin components in hepatitis C virus infection. Hepatology. 2013;57(3):1262–71.

    Article  CAS  PubMed  Google Scholar 

  17. Yang X, Zhang J, Li Y, Hu H, Li X, Ma T, et al. Si-Ni-San promotes liver regeneration by maintaining hepatic oxidative equilibrium and glucose/lipid metabolism homeostasis. J Ethnopharmacol. 2024;326: 117918.

    Article  CAS  PubMed  Google Scholar 

  18. Ma J-T, Xia S, Zhang B-K, Luo F, Guo L, Yang Y, et al. The pharmacology and mechanisms of traditional Chinese medicine in promoting liver regeneration: a new therapeutic option. Phytomed. 2023;116: 154893.

    Article  CAS  PubMed  Google Scholar 

  19. Tao Y, Wang M, Chen E, Tang H. Liver regeneration: analysis of the main relevant signaling molecules. Mediators Inflamm. 2017;2017:4256352.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kang L-I, Mars WM, Michalopoulos GK. Signals and cells involved in regulating liver regeneration. Cells. 2012;1(4):1261–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Marubashi S, Sakon M, Nagano H, Gotoh K, Hashimoto K, Kubota M, et al. Effect of portal hemodynamics on liver regeneration studied in a novel portohepatic shunt rat model. Surgery. 2004;136(5):1028–37.

    Article  PubMed  Google Scholar 

  22. Ormaechea EB, Cornide-Petronio ME, Negrete-Sanchez E, De León CGÁ, Alvarez-Mercado AI, Gulfo J, et al. Effects of cortisol-induced acetylcholine accumulation on tissue damage and regeneration in steatotic livers in the context of partial hepatectomy under vascular occlusion. Transpl. 2018;102:S699.

    Article  Google Scholar 

  23. Ichimiya T, Yamakawa T, Hirano T, Yokoyama Y, Hayashi Y, Hirayama D, et al. Autophagy and autophagy-related diseases: a review. Int J Mol Sci. 2020;21(23):8974.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Xu F, Hua C, Tautenhahn HM, Dirsch O, Dahmen U. The role of autophagy for the regeneration of the aging liver. Int J Mol Sci. 2020;21(10):3606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Teng W, Zhao L, Yang S, Zhang C, Liu M, Luo J, et al. The hepatic-targeted, resveratrol loaded nanoparticles for relief of high fat diet-induced nonalcoholic fatty liver disease. J Control Release. 2019;307:139–49.

    Article  CAS  PubMed  Google Scholar 

  26. Luo S, Yang Y, Zhao T, Zhang R, Fang C, Li Y, et al. Albumin-based Silibinin nanocrystals targeting activated hepatic stellate cells for liver fibrosis therapy. ACS Appl Mater Interfaces. 2023;15(6):7747–58.

    Article  CAS  PubMed  Google Scholar 

  27. Zhang Z, Zhao X, Gao M, Xu L, Qi Y, Wang J, et al. Dioscin alleviates myocardial infarction injury via regulating BMP4/NOX1-mediated oxidative stress and inflammation. Phytomed. 2022;103:154222.

    Article  CAS  PubMed  Google Scholar 

  28. Wang S, Zheng Y, Jin S, Fu Y, Liu Y. Dioscin protects against cisplatin-induced acute kidney injury by reducing ferroptosis and apoptosis through activating Nrf2/HO-1 Signaling. Antioxidants. 2022;11(12):2443.

    CAS  PubMed  Google Scholar 

  29. Zhang X, Yin G, Chen S, Meng D, Yu W, Liu H, et al. Diosgenin ameliorating non-alcoholic fatty liver disease via Nrf2-mediated regulation of oxidative stress and ferroptosis. Diabetes Obes Metab. 2024;26(12):5745–56.

    Article  CAS  PubMed  Google Scholar 

  30. Zhao X, Cong X, Zheng L, Xu L, Yin L, Peng J. Dioscin, a natural steroid saponin, shows remarkable protective effect against acetaminophen-induced liver damage in vitro and in vivo. Toxicol Lett. 2012;214(1):69–80.

    Article  CAS  PubMed  Google Scholar 

  31. Tao X, Wan X, Xu Y, Xu L, Qi Y, Yin L, et al. Dioscin attenuates hepatic ischemia-reperfusion injury in rats through inhibition of oxidative-nitrative stress, inflammation and apoptosis. Transplantation. 2014;98(6):604–11.

    Article  CAS  PubMed  Google Scholar 

  32. Xu L, Gu L, Tao X, Xu Y, Qi Y, Yin L, et al. Effect of dioscin on promoting liver regeneration via activating Notch1/Jagged1 signal pathway. Phytomedicine. 2018;38:107–17.

    Article  CAS  PubMed  Google Scholar 

  33. Chinnappan R, Mir TA, Alsalameh S, Makhzoum T, Adeeb S, Al-Kattan K, et al. Aptasensors are conjectured as promising ALT and AST diagnostic tools for the early diagnosis of acute liver injury. Life. 2023;13(6):1273.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wilson RH, Biasutto AJ, Wang L, Fischer R, Baple EL, Crosby AH, et al. PCNA dependent cellular activities tolerate dramatic perturbations in PCNA client interactions. DNA Repair. 2017;50:22–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Grison A, Atanasoski S. Cyclins, cyclin-dependent kinases, and cyclin-dependent kinase inhibitors in the mouse nervous system. Mol Neurobiol. 2020;57(7):3206–18.

    Article  CAS  PubMed  Google Scholar 

  36. Zhao J, Xu Y, Wang C, Ding Y, Chen M, Wang Y, et al. Soluplus/TPGS mixed micelles for dioscin delivery in cancer therapy. Drug Dev Ind Pharm. 2017;43(7):1197–204.

    Article  CAS  PubMed  Google Scholar 

  37. Li K, Tang Y, Fawcett JP, Gu J, Zhong DJS. Characterization of the pharmacokinetics of dioscin in rat. Steroids. 2005;70(8):525–30.

    Article  CAS  PubMed  Google Scholar 

  38. Li J, Wang RF, Zhou Y, Hu HJ, Yang YB, Yang L, et al. Dammarane-type triterpene oligoglycosides from the leaves and stems of Panax notoginseng and their antiinflammatory activities. J Ginseng Res. 2019;43(3):377–84.

    Article  PubMed  Google Scholar 

  39. Wang J, Zeng L, Zhang Y, Qi W, Wang Z, Tian L, et al. Pharmacological properties, molecular mechanisms and therapeutic potential of ginsenoside Rg3 as an antioxidant and anti-inflammatory agent. Front Pharmacol. 2022;13:975784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xu Y, Wang N, Tan HY, Li S, Zhang C, Feng Y. Gut-liver axis modulation of Panax notoginseng saponins in nonalcoholic fatty liver disease. Hep Intl. 2021;15(2):350–65.

    Article  Google Scholar 

  41. Yuan D, Xiang T, Huo Y, Liu C, Wang T, Zhou Z, et al. Preventive effects of total saponins of Panax japonicus on fatty liver fibrosis in mice. Arch Med Sci. 2018;14(2):396–406.

    Article  CAS  PubMed  Google Scholar 

  42. Liu F, Bai X, Ding RB, Hu YJ, Su H, Wan JB. UPLC/Q-TOFMS-based metabolomics studies on the protective effect of Panax Notoginseng Saponins on alcoholic liver injury. Am J Chin Med. 2015;43(4):695–714.

    Article  CAS  PubMed  Google Scholar 

  43. Zhong H, Wu H, Bai H, Wang M, Wen J, Gong J, et al. Panax notoginseng saponins promote liver regeneration through activation of the PI3K/AKT/mTOR cell proliferation pathway and upregulation of the AKT/Bad cell survival pathway in mice. BMC Complement Altern Med. 2019;19(1):122.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Peng-Fei L, Fu-Gen H, Bin-Bin D, Tian-Sheng D, Xiang-Lin H, Ming-Qin Z. Purification and antioxidant activities of baicalin isolated from the root of huangqin (Scutellaria baicalensis gcorsi). J Food Sci Technol. 2013;50(3):615–9.

    Article  PubMed  Google Scholar 

  45. Dinda B, Dinda M, Dinda S, De UC. An overview of anti-SARS-CoV-2 and anti-inflammatory potential of baicalein and its metabolite baicalin: Insights into molecular mechanisms. Eur J Med Chem. 2023;258:115629.

    Article  CAS  PubMed  Google Scholar 

  46. Ge PY, Qi YY, Qu SY, Zhao X, Ni SJ, Yao ZY, et al. Potential mechanism of S. baicalensis on Lipid metabolism explored via network pharmacology and untargeted lipidomics. Drug Des Dev Therapy. 2021;15:1915–30.

    Article  Google Scholar 

  47. Sun J, Chen Y, Wang T, Ali W, Ma Y, Yuan Y, et al. Baicalin and N-acetylcysteine regulate choline metabolism via TFAM to attenuate cadmium-induced liver fibrosis. Phytomed. 2024;125:155337.

    Article  CAS  PubMed  Google Scholar 

  48. Fang L, Wang HF, Chen YM, Bai RX, Du SY. Baicalin confers hepatoprotective effect against alcohol-associated liver disease by upregulating microRNA-205. Int Immunopharmacol. 2022;107:108553.

    Article  CAS  PubMed  Google Scholar 

  49. Shi H, Qiao F, Lu W, Huang K, Wen Y, Ye L, et al. Baicalin improved hepatic injury of NASH by regulating NRF2/HO-1/NRLP3 pathway. Eur J Pharmacol. 2022;934:75270.

    Article  CAS  PubMed  Google Scholar 

  50. Shi L, Zhang S, Huang Z, Hu F, Zhang T, Wei M, et al. Baicalin promotes liver regeneration after acetaminophen-induced liver injury by inducing NLRP3 inflammasome activation. Free Radical Biol Med. 2020;160:163–77.

    Article  CAS  Google Scholar 

  51. Zhang J, Ma C, Liu Y, Yang G, Jiang Y, Xu C. Interleukin 18 accelerates the hepatic cell proliferation in rat liver regeneration after partial hepatectomy. Gene. 2014;537(2):230–7.

    Article  CAS  PubMed  Google Scholar 

  52. Liu Y, Guo ZW, Li J, Li AH, Huo TG. Insight into the regulation of NLRP3 inflammasome activation by mitochondria in liver injury and the protective role of natural products. Biomed Pharmacother. 2022;156:113968.

    Article  CAS  PubMed  Google Scholar 

  53. Zhou H, Zhang Q, Liu C, Fan J, Huang W, Li N, et al. NLRP3 inflammasome mediates abnormal epithelial regeneration and distal lung remodeling in silica-induced lung fibrosis. Int J Mol Med. 2024;53(3):25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chen Y, Aili M, Chen F, Gong Y, Chen X, Zhang L. Drugs targeting NLRP3 inflammasome in the treatment of diabetic bone disorders. Endocr Metab Immune Disord Drug Targets. 2023;23(10):1265–77.

    Article  CAS  PubMed  Google Scholar 

  55. Vinaik R, Abdullahi A, Barayan D, Jeschke MG. NLRP3 inflammasome activity is required for wound healing after burns. Transl Res. 2020;217:47–60.

    Article  CAS  PubMed  Google Scholar 

  56. Du X, Amin N, Xu L, Botchway BOA, Zhang B, Fang M. Pharmacological intervention of curcumin via the NLRP3 inflammasome in ischemic stroke. Front Pharmacol. 2023;14:1249644.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ando T, Ito H, Kanbe A, Hara A, Seishima M. Deficiency of NALP3 signaling impairs liver regeneration after partial hepatectomy. Inflamm. 2017;40(5):1717–25.

    Article  CAS  PubMed  Google Scholar 

  58. Wang L, Feng T, Su Z, Pi C, Wei Y, Zhao L. Latest research progress on anticancer effect of baicalin and its aglycone baicalein. Arch Pharmacal Res. 2022;45(8):535–57.

    Article  CAS  Google Scholar 

  59. Ml X, Ma YJ, Xu K, Chen MC. Structure–activity relationship for dihydromyricetin as a new natural antioxidant in polymer. J Appl Polymer Sci. 2013;128(3):1436–42.

    Article  Google Scholar 

  60. Sun Y, Liu S, Yang S, Chen C, Yang Y, Lin M, et al. Mechanism of Dihydromyricetin on inflammatory diseases. Front Pharmacol. 2021;12:794563.

    Article  CAS  PubMed  Google Scholar 

  61. Gong H, Xu H, Li M, Zhang D. Molecular mechanism and therapeutic significance of dihydromyricetin in nonalcoholic fatty liver disease. Eur J Pharmacol. 2022;935: 175325.

    Article  CAS  PubMed  Google Scholar 

  62. Dong S, Ji J, Hu L, Wang H. Dihydromyricetin alleviates acetaminophen-induced liver injury via the regulation of transformation, lipid homeostasis, cell death and regeneration. Life Sci. 2019;227:20–9.

    Article  CAS  PubMed  Google Scholar 

  63. Xie J, Liu J, Chen TM, Lan Q, Zhang QY, Liu B, et al. Dihydromyricetin alleviates carbon tetrachloride-induced acute liver injury via JNK-dependent mechanism in mice. World J Gastroenterol. 2015;21(18):5473–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Boice A, Bouchier-Hayes L. Targeting apoptotic caspases in cancer. Biochim Biophys Acta. 2020;1867(6): 118688.

    Article  CAS  Google Scholar 

  65. Santucci R, Sinibaldi F, Cozza P, Polticelli F, Fiorucci L. Cytochrome c: an extreme multifunctional protein with a key role in cell fate. Int J Biol Macromol. 2019;136:1237–46.

    Article  CAS  PubMed  Google Scholar 

  66. Saeedi-Boroujeni A, Mahmoudian-Sani MR. Anti-inflammatory potential of quercetin in COVID-19 treatment. J Inflam. 2021;18(1):3.

    Article  CAS  Google Scholar 

  67. Xu D, Hu MJ, Wang YQ, Cui YL. Antioxidant activities of quercetin and its complexes for medicinal application. Mol. 2019;24(6):1123.

    Article  CAS  Google Scholar 

  68. Ishikawa Y, Kitamura M. Anti-apoptotic effect of quercetin: intervention in the JNK- and ERK-mediated apoptotic pathways. Kidney Int. 2000;58(3):1078–87.

    Article  CAS  PubMed  Google Scholar 

  69. Cao P, Wang Y, Zhang C, Sullivan MA, Chen W, Jing X, et al. Quercetin ameliorates nonalcoholic fatty liver disease (NAFLD) via the promotion of AMPK-mediated hepatic mitophagy. J Nutr Biochem. 2023;120:109414.

    Article  CAS  PubMed  Google Scholar 

  70. Zhang Q, Xu D, Guo Q, Shan W, Yang J, Lin T, et al. Theranostic quercetin nanoparticle for treatment of hepatic fibrosis. Bioconjug Chem. 2019;30(11):2939–46.

    Article  CAS  PubMed  Google Scholar 

  71. Lee S, Lee J, Lee H, Sung J. Relative protective activities of quercetin, quercetin-3-glucoside, and rutin in alcohol-induced liver injury. J Food Biochem. 2019;43(11):e13002.

    Article  PubMed  Google Scholar 

  72. Kanter M, Tuncer I, Erboga M, Atanassova P, Takir M, Kostek OJFM. The effects of quercetin on liver regeneration after liver resection in rats. Folia Morphol. 2016;75(2):179–87.

    Article  CAS  Google Scholar 

  73. Mas-Bargues C, Escrivá C, Dromant M, Borrás C, Viña J. Lipid peroxidation as measured by chromatographic determination of malondialdehyde Human plasma reference values in health and disease. Arch Biochem Biophys. 2021;709:108941.

    Article  CAS  PubMed  Google Scholar 

  74. Saxena P, Selvaraj K, Khare SK, Chaudhary N. Superoxide dismutase as multipotent therapeutic antioxidant enzyme: role in human diseases. Biotech Lett. 2022;44(1):1–22.

    Article  CAS  Google Scholar 

  75. Kennedy L, Sandhu JK, Harper ME, Cuperlovic-Culf M. Role of glutathione in cancer: from mechanisms to therapies. Biomolecules. 2020;10(10):1429.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Peng Y, Ao M, Dong B, Jiang Y, Yu L, Chen Z, et al. Anti-inflammatory effects of curcumin in the inflammatory diseases: status, limitations and countermeasures. Drug Des Dev Ther. 2021;15:4503–25.

    Article  CAS  Google Scholar 

  77. Llano S, Gómez S, Londoño J, Restrepo A. Antioxidant activity of curcuminoids. Phys Chem Chem Phys. 2019;21(7):3752–60.

    Article  CAS  PubMed  Google Scholar 

  78. Hussain Y, Alam W, Ullah H, Dacrema M, Daglia M, Khan H, et al. Antimicrobial potential of curcumin: therapeutic potential and challenges to clinical applications. Antibiot. 2022;11(3):322.

    CAS  PubMed  Google Scholar 

  79. Wu P, Huang R, Xiong YL, Wu C. Protective effects of curcumin against liver fibrosis through modulating DNA methylation. Chin J Nat Med. 2016;14(4):255–64.

    CAS  PubMed  Google Scholar 

  80. Feng WW, Kuang SY, Tu C, Ma ZJ, Pang JY, Wang YH, et al. Natural products berberine and curcumin exhibited better ameliorative effects on rats with non-alcohol fatty liver disease than lovastatin. Biomed Pharmacother. 2018;99:325–33.

    Article  CAS  PubMed  Google Scholar 

  81. Sunoqrot S, Abu Shalhoob M, Jarrar Y, Hammad AM, Al-Ameer HJ, Al-Awaida W. Nanoencapsulated curcumin mitigates liver injury and drug-metabolizing enzymes induction in diclofenac-treated mice. ACS Omega. 2024;9(7):7881–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Toydemir T, Kanter M, Erboga M, Oguz S, Erenoglu C. Antioxidative, antiapoptotic, and proliferative effect of curcumin on liver regeneration after partial hepatectomy in rats. Toxicol Ind Health. 2015;31(2):162–72.

    Article  PubMed  Google Scholar 

  83. Meng T, Xiao D, Muhammed A, Deng J, Chen L, He J. Anti-inflammatory action and mechanisms of resveratrol. Mol. 2021;26(1):229.

    Article  CAS  PubMed  Google Scholar 

  84. Varoni EM, Lo Faro AF, Sharifi-Rad J, Iriti M. Anticancer molecular mechanisms of resveratrol. Front Nutr. 2016;3:8.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Tang L, Yang F, Fang Z, Hu C. Resveratrol ameliorates alcoholic fatty liver by inducing autophagy. Am J Chin Med. 2016;44(6):1207–20.

    Article  CAS  PubMed  Google Scholar 

  86. Jing Z, Jian P, Na J, Lieming X. Resveratrol inhibits hepatic stellate cell activation by regulating autophagy and apoptosis through the SIRT1 and JNK signaling pathways. J Food Biochem. 2022;46(12):e14463–563.

    Google Scholar 

  87. Wang Y, Jiang Y, Fan X, Tan H, Zeng H, Wang Y, et al. Hepato-protective effect of resveratrol against acetaminophen-induced liver injury is associated with inhibition of CYP-mediated bioactivation and regulation of SIRT1-p53 signaling pathways. Toxicol Lett. 2015;236(2):82–9.

    Article  CAS  PubMed  Google Scholar 

  88. Liu X, Wang K, Zheng Q, Liu X, Yang Y, Xie C, et al. Schisandrol B inhibits calcification of aortic valve by targeting p53 related inflammatory and senescence. Biomed Pharmacother. 2024;178:117241.

    Article  CAS  PubMed  Google Scholar 

  89. Jiang YM, Wang Y, Tan HS, Yu T, Fan XM, Chen P, et al. Schisandrol B protects against acetaminophen-induced acute hepatotoxicity in mice via activation of the NRF2/ARE signaling pathway. Acta Pharmacol Sin. 2016;37(3):382–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Liang H, Yang X, Li H, Wang X, Su H, Li X, et al. Schisandrol B protects against cholestatic liver injury by inhibiting pyroptosis through pregnane X receptor. Biochem Pharmacol. 2022;204:115222.

    Article  CAS  PubMed  Google Scholar 

  91. Li L, Cui L, Lin P, Liu Z, Bao S, Ma X, et al. Kupffer-cell-derived IL-6 is repurposed for hepatocyte dedifferentiation via activating progenitor genes from injury-specific enhancers. Cell Stem Cell. 2023;30(3):283–99.

    Article  CAS  PubMed  Google Scholar 

  92. Adas G, Koc B, Adas M, Duruksu G, Subasi C, Kemik O, et al. Effects of mesenchymal stem cells and VEGF on liver regeneration following major resection. Langenbecks Arch Surg. 2016;401(5):725–40.

    Article  PubMed  Google Scholar 

  93. Li X, Sun J, Fan X, Guan L, Li D, Zhou Y, et al. Schisandrol B promotes liver regeneration after partial hepatectomy in mice. Eur J Pharmacol. 2018;818:96–102.

    Article  CAS  PubMed  Google Scholar 

  94. Lazo PA. Reverting p53 activation after recovery of cellular stress to resume with cell cycle progression. Cell Signal. 2017;33:49–58.

    Article  CAS  PubMed  Google Scholar 

  95. Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair. 2016;42:63–71.

    Article  CAS  PubMed  Google Scholar 

  96. Jiang Y, Fan X, Wang Y, Chen P, Zeng H, Tan H, et al. Schisandrol B protects against acetaminophen-induced hepatotoxicity by inhibition of CYP-mediated bioactivation and regulation of liver regeneration. Toxicol Sci. 2015;143(1):107–15.

    Article  CAS  PubMed  Google Scholar 

  97. Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. The BCL-2 family reunion. Mol Cell. 2010;37(3):299–310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Thingore C, Kshirsagar VV, Gursahani M, Juvekar A. P2–215: rosmarinic acid attenuates oxidative stress, neuroinflammation and neurodegeneration against lipopolysaccharide-induced alzheimer’s disease via jnk-3 and caspase-3 inhibition in mice. Alzheimers Dement. 2019;15(7S):P660–760.

    Google Scholar 

  99. Zhang J, Cui X, Zhang M, Bai B, Yang Y, Fan S. The antibacterial mechanism of perilla rosmarinic acid. Biotechnol Appl Biochem. 2022;69(4):1757–64.

    Article  CAS  PubMed  Google Scholar 

  100. Yao Y, Li R, Liu D, Long L, He N. Rosmarinic acid alleviates acetaminophen-induced hepatotoxicity by targeting Nrf2 and NEK7-NLRP3 signaling pathway. Ecotoxicol Environ Saf. 2022;241:113773.

    Article  CAS  PubMed  Google Scholar 

  101. Kim M, Yoo G, Randy A, Son YJ, Hong CR, Kim SM, et al. Lemon balm and its constituent, rosmarinic acid, alleviate liver damage in an animal model of nonalcoholic steatohepatitis. Nutrients. 2020;12(4):1166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Lou K, Yang M, Duan E, Zhao J, Yu C, Zhang R, et al. Rosmarinic acid stimulates liver regeneration through the mTOR pathway. Phytomed. 2016;23(13):1574–82.

    Article  CAS  PubMed  Google Scholar 

  103. Zhao J, Zheng H, Sui Z, Jing F, Quan X, Zhao W, et al. Ursolic acid exhibits anti-inflammatory effects through blocking TLR4-MyD88 pathway mediated by autophagy. Cytokine. 2019;123:154726.

    Article  CAS  PubMed  Google Scholar 

  104. do Nascimento PG, Lemos TL, Bizerra AM, Arriaga AM, Ferreira DA, Santiago GM, et al. Antibacterial and antioxidant activities of ursolic acid and derivatives. Mol. 2014;19(1):1317–27.

    Article  Google Scholar 

  105. Dong M, Cui Q, Li Y, Li Y, Chang Q, Bai R, et al. Ursolic acid suppresses fatty liver-associated hepatocellular carcinoma by regulating lipid metabolism. Food Biosci. 2024;60:104460.

    Article  CAS  Google Scholar 

  106. Yuan N, Qi L, Wang Z, Yipeng W, Chenkai H, Xuan Z. Ursolic acid reverses liver fibrosis by inhibiting NOX4/NLRP3 inflammasome pathways and bacterial dysbiosis. Gut microbes. 2021;13(1):1972746–846.

    Article  Google Scholar 

  107. Jin YR, Jin JL, Li CH, Piao XX, Jin NG. Ursolic acid enhances mouse liver regeneration after partial hepatectomy. Pharm Biol. 2012;50(4):523–8.

    Article  CAS  PubMed  Google Scholar 

  108. Tsai SJ, Yin MC. Antioxidative and anti-inflammatory protection of oleanolic acid and ursolic acid in PC12 cells. J Food Sci. 2008;73(7):H174–8.

    Article  CAS  PubMed  Google Scholar 

  109. Li C, He J, Li Y, Zhang C, Wang Z, Wu X, et al. Cardioprotective role of oleanolic acid in patients with type 2 diabetes mellitus. Heliyon. 2024;10(11):e31303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Harley BK, Amponsah IK, Ben IO, Mireku-Gyimah NA, Anokwah D, Neglo D, et al. Hypoglycaemic activity of Oleanonic acid, a 3-oxotriterpenoid isolated from Aidia Genipiflora (DC.) Dandy, involves inhibition of carbohydrate metabolic enzymes and promotion of glucose uptake. Biomed Pharmacotherapy. 2022;149:112833.

    Article  CAS  Google Scholar 

  111. Wan XL, Lu YF, Xu SF, Wu Q, Liu J. Oeanolic acid protects against the hepatotoxicity of D-galactosame plus endotoxin in mice. Biomed Pharmacother. 2017;93:1040–6.

    Article  CAS  PubMed  Google Scholar 

  112. Liu J, Liu J, Meng C, Huang C, Liu F, Xia C. Oleanolic acid alleviates ANIT-induced cholestatic liver injury by activating Fxr and Nrf2 pathways to ameliorate disordered bile acids homeostasis. Phytomed. 2022;102:154173.

    Article  CAS  PubMed  Google Scholar 

  113. Song S, Peng H, Li Y, Zhao T, Cao R, Zheng L, et al. Oleanolic acid promotes liver regeneration after partial hepatectomy via regulating pregnane X receptor signaling pathway in mice. Chem Biol Interact. 2024;393:110970.

    Article  CAS  PubMed  Google Scholar 

  114. Mao XP, Zhang LS, Huang B, Zhou SY, Liao J, Chen LW, et al. Mir-135a enhances cellular proliferation through post-transcriptionally regulating PHLPP2 and FOXO1 in human bladder cancer. J Transl Med. 2015;13:86.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Zhu Y, Gu J, Li Y, Peng C, Shi M, Wang X, et al. MiR-17-5p enhances pancreatic cancer proliferation by altering cell cycle profiles via disruption of RBL2/E2F4-repressing complexes. Cancer Lett. 2018;412:59–68.

    Article  CAS  PubMed  Google Scholar 

  116. Chen G, Sun W, Li Y, Li M, Jia X, Wang J, et al. miR-196a promotes proliferation of mammary epithelial cells by targeting CDKN1B. Animals. 2023;13(23):3682.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Ye CJ, Li SA, Zhang Y, Lee WH. Geraniol targets K(V)1.3 ion channel and exhibits anti-inflammatory activity in vitro and in vivo. Fitoterapia. 2019;139:104394.

    Article  CAS  PubMed  Google Scholar 

  118. Lin L, Long N, Qiu M, Liu Y, Sun F, Dai M. The inhibitory efficiencies of geraniol as an anti-inflammatory, antioxidant, and antibacterial, natural agent against methicillin-resistant staphylococcus aureus infection in vivo. Infect Drug Resist. 2021;14:2991–3000.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Mohammed MJ, Tadros MG, Michel HE. Geraniol protects against cyclophosphamide-induced hepatotoxicity in rats: possible role of MAPK and PPAR-γ signaling pathways. Food Chem Toxicol. 2020;139:111251.

    Article  CAS  PubMed  Google Scholar 

  120. Zhou B, Niu Y, Wang D, Liu L, Guo JJFS. Biotechnology geraniol alleviates liver injury induced by bisphenol A via modulating NLRP3/caspase-1 pathway and gut microbiota in mice model. Food Sci Biotechnol. 2024;33(13):3119–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Selzner N, Selzner M, Odermatt B, Tian Y, Van Rooijen N, Clavien PA. ICAM-1 triggers liver regeneration through leukocyte recruitment and Kupffer cell-dependent release of TNF-alpha/IL-6 in mice. Gastroenterology. 2003;124(3):692–700.

    Article  CAS  PubMed  Google Scholar 

  122. Canbek M, Uyanoglu M, Canbek S, Ceyhan E, Ozen A, Durmus B, et al. The effect of geraniol on liver regeneration after hepatectomy in rats. In Vivo. 2017;31(2):209–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, et al. Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther. 2022;7(1):3.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Ze X, Jia J, Li X, You H, Zhao X, Zhang D, et al. Tanshinone IIA promotes the proliferation of WB-F344 hepatic oval cells via Wnt/β-catenin signaling. Mol Med Rep. 2016;13(2):1501–8.

    Article  CAS  PubMed  Google Scholar 

  125. Xu W, Du X, Li J, Zhang Z, Ma X, Luo D, et al. SiNiSan alleviates liver injury by promoting hepatic stem cell differentiation via Wnt/β-catenin signaling pathway. Phytomedicine. 2022;99:153969.

    Article  CAS  PubMed  Google Scholar 

  126. Zhang C, Sun C, Zhao Y, Ye B, Yu G. Signaling pathways of liver regeneration: Biological mechanisms and implications. IScience. 2024;27(1):108683.

    Article  CAS  PubMed  Google Scholar 

  127. Jackson LN, Larson SD, Silva SR, Rychahou PG, Chen LA, Qiu S, et al. PI3K/Akt activation is critical for early hepatic regeneration after partial hepatectomy. Am J Physiol Gastrointest Liver Physiol. 2008;294(6):G1401–10.

    Article  CAS  PubMed  Google Scholar 

  128. Cui Z, Jin N, Amevor FK, Shu G, Du X, Kang X, et al. Dietary supplementation of salidroside alleviates liver lipid metabolism disorder and inflammatory response to promote hepatocyte regeneration via PI3K/AKT/Gsk3-β pathway. Poult Sci. 2022;101(9):102034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Sastry KS, Ibrahim WN, Chouchane AI. Multiple signaling pathways converge on proapoptotic protein BAD to promote survival of melanocytes. FASEB J. 2020;34(11):14602–14.

    Article  CAS  PubMed  Google Scholar 

  130. Shi Q, Xue C, Zeng Y, Yuan X, Chu Q, Jiang S, et al. Notch signaling pathway in cancer: from mechanistic insights to targeted therapies. Signal Transduct Target Ther. 2024;9(1):128.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Xu HY, Li BJ, Wang RF, Andersson R. Alterations of Notch/Jagged mRNA and protein expression after partial hepatectomy in rats. Scand J Gastroenterol. 2008;43(12):1522–8.

    Article  CAS  PubMed  Google Scholar 

  132. Zhang F, Zhang J, Li X, Li B, Tao K, Yue S. Notch signaling pathway regulates cell cycle in proliferating hepatocytes involved in liver regeneration. J Gastroenterol Hepatol. 2018;33(8):1538–47.

    Article  CAS  PubMed  Google Scholar 

  133. Zou Y, Hu M, Lee J, Nambiar SM, Garcia V, Bao Q, et al. Nrf2 is essential for timely M phase entry of replicating hepatocytes during liver regeneration. Am J Physiol Gastrointest Liver Physiol. 2015;308(4):G262–8.

    Article  CAS  PubMed  Google Scholar 

  134. Jayasuriya R, Dhamodharan U, Ali D, Ganesan K, Xu B, Ramkumar KM. Targeting Nrf2/Keap1 signaling pathway by bioactive natural agents: possible therapeutic strategy to combat liver disease. Phytomed. 2021;92:153755.

    Article  CAS  PubMed  Google Scholar 

  135. Qiao L, Yang H, Gao S, Li L, Fu X, Wei Q. Research progress on self-assembled nanodrug delivery systems. J Mater Chem B. 2022;10(12):1908–22.

    Article  CAS  PubMed  Google Scholar 

  136. Huang J, Zhu Y, Xiao H, Liu J, Li S, Zheng Q, et al. Formation of a traditional Chinese medicine self-assembly nanostrategy and its application in cancer: a promising treatment. Chinese Med. 2023;18(1):66.

    Article  CAS  Google Scholar 

  137. Feng Q, Zhang X, Zhao X, Liu J, Wang Q, Yao Y, et al. Intranasal delivery of pure nanodrug loaded liposomes for Alzheimer’s disease treatment by efficiently regulating microglial polarization. Small. 2024;20(50):e2405781.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Liu H, Gan C, Shi H, Qu K, Jing L, Lu M, et al. Gastric floating pill enhances the bioavailability and drug efficacy of dihydromyricetin in vivo. J Drug Delivery Sci Technol. 2021;61:102279.

    Article  CAS  Google Scholar 

  139. Liu L, Yin X, Wang X, Li X. Determination of dihydromyricetin in rat plasma by LC-MS/MS and its application to a pharmacokinetic study. Pharm Biol. 2017;55(1):657–62.

    Article  CAS  PubMed  Google Scholar 

  140. Ye J, Bao S, Zhao S, Zhu Y, Ren Q, Li R, et al. Self-assembled micelles improve the oral bioavailability of dihydromyricetin and anti-acute alcoholism activity. AAPS PharmSciTech. 2021;22(3):111.

    Article  CAS  PubMed  Google Scholar 

  141. Liu X, Song Y, Cheng S, Tan M. Curcumin delivery nanoparticles based on Maillard reaction of Haematococcus pluvialis protein/galactose for alleviating acute alcoholic liver damage. Food Sci Human Wellness. 2024;13(5):2629–41.

    Article  CAS  Google Scholar 

  142. Allan KE, Lenehan CE, Ellis AV. UV light stability of α-cyclodextrin/resveratrol host–guest complexes and isomer stability at varying pH. Aust J Chem. 2009;62(8):921–6.

    Article  CAS  Google Scholar 

  143. Pinto MdC, García-Barrado JA, Macías P. Oxidation of resveratrol catalyzed by soybean lipoxygenase. J Agri Food Chem. 2003;51(6):1653–7.

    Article  CAS  Google Scholar 

  144. Li X, Chen XX, Xu Y, Xu XB, Wu WF, Zhao Q, et al. Construction of glycogen-based nanoparticles loaded with resveratrol for the alleviation of high-fat diet-induced nonalcoholic fatty liver disease. Biomacromol. 2022;23(1):409–23.

    Article  CAS  Google Scholar 

  145. Tang P, Wu H, Wang M, Zhang R, Liang B, Li X, et al. Self-assembled poly (rosmarinic acid) nanoantioxidant for enhanced acute liver injury treatment. ACS Applied Nano Materials. 2023;6(18):16392–9.

    Article  CAS  Google Scholar 

  146. Shen Z, Li B, Liu Y, Zheng G, Guo Y, Zhao R, et al. A self-assembly nanodrug delivery system based on amphiphilic low generations of PAMAM dendrimers-ursolic acid conjugate modified by lactobionic acid for HCC targeting therapy. Nanomed Nanotechnol Biol Med. 2018;14(2):227–36.

    Article  CAS  Google Scholar 

  147. Ashour AA, El-Kamel AH, Abdelmonsif DA, Khalifa HM, Ramadan AA. Modified lipid nanocapsules for targeted tanshinone IIA delivery in liver fibrosis. Int J Nanomed. 2021;16:8013–33.

    Article  CAS  Google Scholar 

  148. Zong L, Wang Y, Song S, Zhang H, Mu S, Liu W, et al. Formulation and evaluation on synergetic anti-hepatoma effect of a chemically stable and release-controlled nanoself-assembly with natural monomers. Int J Nanomed. 2023;18:3407–28.

    Article  CAS  Google Scholar 

  149. Liu H, Guo X, Yang J, Xia C, Yao Y, Li X, et al. Structure optimization of natural product catalpol to obtain novel and potent analogs against heart failure. J Med Chem. 2025;68(4):4540–60.

    Article  CAS  PubMed  Google Scholar 

  150. Matchett KP, Wilson-Kanamori JR, Portman JR, Kapourani CA, Fercoq F, May S, et al. Multimodal decoding of human liver regeneration. Nature. 2024;630(8015):158–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Cross A, Harris JM, Arbe-Barnes E, Nixon C, Dhairyawan R, Hall A, et al. Characterisation of HBV and co-infection with HDV and HIV through spatial transcriptomics. EGastroenterology. 2024;2(3):e100067.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Li J, Wang W, Zhang Y, Yang Z. Research progress on vesicles from Chinese medicinal herbs. Chinese Herbal Med. 2023;52(3):349–60.

    Google Scholar 

  153. Wang RN, Zhao HC, Huang JY, Wang HL, Li JS, Lu Y, et al. Challenges and strategies in progress of drug delivery system for traditional Chinese medicine Salviae Miltiorrhizae Radix et Rhizoma (Danshen). Chinese Herbal Med. 2021;13(1):78–89.

    Article  Google Scholar 

  154. Huang L, Luo S, Tong S, Lv Z, Wu J. The development of nanocarriers for natural products. Wiley Interdisc Rev Nanomed Nanobiotechnol. 2024;16(3):e1967.

    Article  CAS  Google Scholar 

  155. Duan S, Xia Y, Tian X, Cui J, Zhang X, Yang Q, et al. A multi-bioresponsive self-assembled nano drug delivery system based on hyaluronic acid and geraniol against liver cancer. Carbohyd Polym. 2023;310:120695.

    Article  CAS  Google Scholar 

  156. Koh B, Danpanichkul P, Wang M, Tan DJH, Ng CH. Application of artificial intelligence in the diagnosis of hepatocellular carcinoma. EGastroenterology. 2023;1(2):e100002.

    Article  PubMed  PubMed Central  Google Scholar 

  157. You Y, Lai X, Pan Y, Zheng H, Vera J, Liu S, et al. Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Ther. 2022;7(1):156.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Pun FW, Liu BHM, Long X, Leung HW, Leung GHD, Mewborne QT, et al. Identification of therapeutic targets for amyotrophic lateral sclerosis using pandaomics - an AI-enabled biological target discovery platform. Front Aging Neurosc. 2022;14:914017.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Over the course of my researching and writing this paper, I would like to express my thanks to all those who have helped me. The figures were created with FigDraw.

Chemical compounds studied in this article

Dioscin (PubChem CID:119245).

Panax notoginseng saponins (PubChem CID:441154).

Baicalin (PubChem CID: 64982).

Dihydromyricetin (PubChem CID:161557).

Quercetin (PubChem CID: 5280343).

Resveratrol (PubChem CID: 445154).

Schisandrol B (PubChem CID: 634470).

Curcumin (PubChem CID: 969516).

Rosmarinic acid (PubChem CID: 5281792).

Ursolic acid (PubChem CID: 64945).

Oleanolic acid (PubChem CID:10494).

Ger (PubChem CID: 637566).

Funding

National Natural Science Foundation of China (82373932); Natural Science Foundation of Anhui Province (2208085MH203); Department of Education of Anhui Province Outstanding Young Teacher Training Project (YQZD2023023); Anhui Medical University Youth Shuangpei Program (2024); Anhui Province University outstanding youth research project (2024AH020006); Anhui Translational Medicine Research Institute Project (2022-zhyx-C09).

Author information

Authors and Affiliations

Authors

Contributions

Chang Tian: Conceptualization; writing—original draft; writing—review and editing; visualization; Yuhan Wang: Conceptualization; writing—original draft; writing—review and editing; Ran Wang: Conceptualization; writing—original draft; writing—review and editing; Tao Xu: Conceptualization; supervision; Funding acquisition; writing—original draft; writing—review and editing; Linxin Pan: Conceptualization; supervision; writing—original draft; writing—review and editing.

Corresponding authors

Correspondence to Linxin Pan or Tao Xu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they do not have any competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tian, C., Wang, Y., Wang, R. et al. Pharmacological and therapeutic effects of natural products on liver regeneration-a comprehensive research. Chin Med 20, 57 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13020-025-01108-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13020-025-01108-y

Keywords