Next Article in Journal
The Sigma Factor AsbI Is Required for the Expression of Acinetobactin Siderophore Transport Genes in Aeromonas salmonicida
Previous Article in Journal
Gated Calcium Ion Channel and Mutation Mechanisms in Multidrug-Resistant Tuberculosis
Previous Article in Special Issue
Expression and Function of BMP and Activin Membrane-Bound Inhibitor (BAMBI) in Chronic Liver Diseases and Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liver Fibrosis Resolution: From Molecular Mechanisms to Therapeutic Opportunities

1
Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
2
Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9671; https://doi.org/10.3390/ijms24119671
Submission received: 28 April 2023 / Revised: 25 May 2023 / Accepted: 30 May 2023 / Published: 2 June 2023

Abstract

:
The liver is a critical system for metabolism in human beings, which plays an essential role in an abundance of physiological processes and is vulnerable to endogenous or exogenous injuries. After the damage to the liver, a type of aberrant wound healing response known as liver fibrosis may happen, which can result in an excessive accumulation of extracellular matrix (ECM) and then cause cirrhosis or hepatocellular carcinoma (HCC), seriously endangering human health and causing a great economic burden. However, few effective anti-fibrotic medications are clinically available to treat liver fibrosis. The most efficient approach to liver fibrosis prevention and treatment currently is to eliminate its causes, but this approach’s efficiency is too slow, or some causes cannot be fully eliminated, which causes liver fibrosis to worsen. In cases of advanced fibrosis, the only available treatment is liver transplantation. Therefore, new treatments or therapeutic agents need to be explored to stop the further development of early liver fibrosis or to reverse the fibrosis process to achieve liver fibrosis resolution. Understanding the mechanisms that lead to the development of liver fibrosis is necessary to find new therapeutic targets and drugs. The complex process of liver fibrosis is regulated by a variety of cells and cytokines, among which hepatic stellate cells (HSCs) are the essential cells, and their continued activation will lead to further progression of liver fibrosis. It has been found that inhibiting HSC activation, or inducing apoptosis, and inactivating activated hepatic stellate cells (aHSCs) can reverse fibrosis and thus achieve liver fibrosis regression. Hence, this review will concentrate on how HSCs become activated during liver fibrosis, including intercellular interactions and related signaling pathways, as well as targeting HSCs or liver fibrosis signaling pathways to achieve the resolution of liver fibrosis. Finally, new therapeutic compounds targeting liver fibrosis are summarized to provide more options for the therapy of liver fibrosis.

1. Introduction

The liver, the main organ in the body, is involved in many physiological activities, including the elimination of infections, xenobiotics, medicines, and other toxic chemicals; the metabolism of glucose, protein, and lipids; and immunological functions [1,2]. Despite the liver’s considerable capacity for regeneration [3], ongoing damage can bring about fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC) [2]. Liver fibrosis, which results from an inappropriate response to liver injury’s wound-healing process, is a pathological condition [4,5], and the main pathological characteristics are the excessive accumulation of extracellular matrix (ECM) proteins and the loss of the normal structure of the liver tissue [6,7,8]. Additionally, various etiological factors can lead to liver fibrosis, such as viral infections, alcohol, metabolic diseases, biliary diseases, hepatic toxins [9], autoimmune disease [5], and prolonged use of some medications, including methotrexate, methyldopa, chlorpromazine, tolbutamide, etc. [8,10] (Figure 1).
Chronic liver illnesses, including viral hepatitis and fatty liver deteriorate in part due to liver fibrosis. Without adequate treatment, cirrhosis, which poses a major threat to human health, can arise in 75–80% of these conditions [11,12]. Patients with chronic liver disorders who progress to liver fibrosis and cirrhosis suffer high morbidity and mortality rates, placing a significant economic burden on society [8,13]. Liver fibrosis is typically a slowly progressing condition with no clinical symptoms. Although liver fibrosis originally happens as part of the liver repair process, if it is not controlled, it develops into a pathogenic condition [14]. With ongoing damage and ECM deposition, the liver is gradually hardening and stiffening over time, followed by a slow loss of function. Liver fibrosis may then progress into cirrhosis or even liver cancer, with a variety of other consequences involving portal hypertension and liver failure [15,16,17]. Currently, the only effective treatment for advanced liver disease is liver transplantation [15,18], but it has its limitations. In all etiologies of chronic liver illnesses, the main factor causing liver failure is liver fibrosis, and the degree of fibrosis is the most reliable indicator of mortality from liver-related causes [19,20,21,22]. Accordingly, discovering suitable strategies to prevent or reverse liver fibrosis is crucial to preventing further liver fibrosis deterioration.
The process of liver fibrosis is regulated by a variety of cells and cytokines [23]. When the liver is continuously damaged, the damaged cells release pro-inflammatory or pro-fibrotic factors, which in turn activate relevant signaling pathways and promote hepatic stellate cells (HSCs) activation, resulting in the production of large amounts of ECM, leading to further deterioration of liver fibrosis [16]. Most of the therapeutic drugs for liver fibrosis are still in clinical trials and research stages [23]. Hence, this review will concentrate on the activation of HSCs in liver fibrosis and the cellular and molecular pathways of liver fibrosis resolution. Furthermore, the new therapeutic compounds for liver fibrosis in recent years are summarized.

2. The Mechanisms of Liver Fibrosis

The continuous deposition of ECM [24], which harms the liver’s physiological structure [13] and impairs its capacity for normal activity [7], is the hallmark of liver fibrosis. Additionally, it is caused by highly active crosstalk among various cells, including hepatocellular parenchymal cells (hepatocytes), non-parenchymal cells (like Kupffer and endothelial cells), and immune cells [23,25,26]. Myofibroblasts (MFs) contribute to the production of ECM and are regarded as the primary effector cells in the development of fibrosis [4,27]. Although the source of activated MFs may vary depending on the different etiologies, HSCs can be considered their main origin [3]. Thus quiescent hepatic stellate cells (qHSCs) activation and subsequent conversion into MFs are essential occurrences in the pathogenesis of liver fibrosis [7,26,28]. Consequently, treating liver fibrosis requires limiting the activation of HSCs [7].

2.1. Hepatic Stellate Cells

HSCs are a unique group of pre-hepatic cells that are situated in the Disse space next to sinusoidal endothelial cells and hepatocytes [29]. HSCs in a healthy liver display a quiescent state, and their physiological roles involve fat storage and the metabolism of vitamin A [12]. The secretion of sufficient amounts of ECM proteins, including type III collagen, type IV collagen, and laminin, to maintain the homeostasis of ECM is another duty performed by the qHSCs [23].
In the process of developing liver fibrosis, HSCs are crucial [5,30]. MFs, which secrete matrix proteins and are the primary cause of liver fibrogenesis, are mostly derived from activated hepatic stellate cells (aHSCs) [6]. Several important cells and inflammatory mediators, such as inflammatory stimuli and fibrogenic cytokines like transforming growth factor-β (TGF-β), reactive oxygen species (ROS), etc., released by activated macrophages, platelets, and damaged hepatocytes, activate the qHSCs when the liver is injured [23,27]. Furthermore, non-coding RNAs (ncRNAs) can regulate the development of liver fibrosis by acting on HSCs. For example, circular RNAs (circRNAs) frequently function as miRNA sponges that control HSC activation and proliferation and hence participate in the development of liver fibrosis [31]. In addition, it has been found that exosomes, a type of extracellular vesicles (EVs), from various cell sources can also stimulate HSCs and play an important role in the process of liver fibrosis [32,33]. Exosomes can carry various cellular components, including nucleic acids, proteins, lipids, and other molecules involved in the transmission of information between cells [32,34]. It has been found that exosomal cellular communication network factor 2 (CCN2), derived from HSCs, contains connective tissue growth factors that promote the development of liver fibrosis and can amplify or attenuate fiber formation signals by binding to other exosomal components [35].
HSCs also perform a transdifferentiation process that changes them to a proliferative and contractile myofibroblast phenotype from a quiescent phenotype [13,36]. Additionally, in conditions of chronic injury, the continued activation of HSCs causes an imbalance between the deposition and dissolution of the ECM, which in turn causes liver fibrosis to proceed [13]. MFs may continue to be triggered by excessive cytokines, which result in the formation of massive ECM aggregates and excessive ECM deposition [23]. The abnormal deposition of ECM results in aberrant wound healing responses and then increases the formation and growth of liver fibrosis [27,36]. In conclusion, liver fibrosis is caused by, develops into, and can be reversed by HSCs. Moreover, a range of cells, including Kupffer cells (KCs), hepatocytes, and numerous signaling pathways, can regulate HSC activation [12].

2.2. The Intercellular Crosstalk of HSC Activation

For the preservation of healthy liver function and cell survival, intercellular interaction in the hepatic microenvironment is crucial [6]. Additionally, the progression of liver fibrosis, which is an intricate process of liver self-repair, is related to the highly active crosstalk among several cell types [25]. Although HSCs are thought to be the primary cell of action in liver fibrosis, other types of liver cells also contribute significantly to the condition. In fact, interaction with other hepatic cells, such as hepatocytes, liver sinusoidal endothelial cells (LSECs), and inflammatory cells, is necessary for HSC activation (Figure 1). Through the release of cytokines and other signaling molecules, these cells communicate with one another and either encourage or suppress the activation of HSCs [6].

2.2.1. Hepatocytes

More than 80% of all liver cells are hepatocytes, the most significant parenchymal cells in the liver [6]. Hepatocytes perform a variety of tasks in physiological environments, including detoxification and the release of bile, proteins, and lipids [37]. Hepatocytes that have suffered injury generate substances that stimulate HSCs and inflammatory cells, which bring about the development of fibrosis [6].
One of the key initial stages in the pathophysiology of all liver illnesses and their etiologies is hepatocyte death. Damage-associated molecular patterns (DAMPs), which are intracellular substances that can be released by dying hepatocytes and act as warning signals to neighboring cells like KCs and HSCs, are essential for inflammation and the formation of fibrosis [13]. High-mobility group box-1 (HMGB1), a DNA-binding non-histone nuclear protein, is typically produced in eukaryotic cells and is one of the most researched DAMPs in liver disease [38]. Furthermore, it has been demonstrated that HMGB1 could directly activate HSCs by controlling HSC autophagy in liver fibrosis development models linked to the hepatitis B virus (HBV) [39]. Furthermore, hepatocytes can also enhance the activation of HSCs by secreting exosomes, thus promoting the progression of liver fibrosis [34]. Lipotoxic hepatocyte-derived exosomal miR-27a was found to promote the development of liver fibrosis in metabolically associated fatty liver disease (MAFLD) by enhancing HSC proliferation and activation and inhibiting aHSC autophagy [40].

2.2.2. Liver Sinusoidal Endothelial Cells

LSECs mostly exhibit anti-inflammatory, anti-fibrotic, and regeneration-promoting actions in healthy liver tissue [41]. In addition, the interaction among HSCs, hepatocytes, and LSECs could maintain HSC quiescence and regenerate hepatocytes [6], which is essential for fibrosis development.
Studies have demonstrated that LSECs can maintain HSCs quiescence through a vascular endothelial growth factor (VEGF)-stimulated pathway [42]. When the liver is injured, the capillary production of LSECs both decreases the synthesis of vasodilators (such as NO and cyclooxygenase) and increases the synthesis of vasoconstrictors (such as endothelin 1 and thromboxane A2) [23]. Although changing the phenotype of LSECs, this imbalance also helps to activate HSCs, which in turn encourages inflammation and liver fibrosis [43,44]. Moreover, LSECs can release signaling molecules including TGF-β and platelet-derived growth factor (PDGF) or activate signaling pathways like Wnt/β-catenin, which in turn activate HSCs [23].

2.2.3. Inflammatory Cells

Inflammation brought on by persistent liver injury can be triggered by the release of ROS and other signals from damaged cells, which in turn activate inflammatory cells like macrophages, lymphocytes, and others [45]. Additionally, liver fibrosis worsens due to the mutual activation of inflammatory cells and HSCs [13].
The predominant macrophage populations in the initial stages of the damage are pro-inflammatory [46]. Among them, hepatic macrophages (KCs), which are located within the lumen of the liver sinusoids [17], play crucial roles in liver fibrosis development [47]. When the liver is injured, the KCs become active and quickly secrete substances that promote inflammation and fibrosis, such as interleukin (IL)-1β, tumor necrosis factor-alpha (TNF-α), chemokine (C-C motif) ligand 2 (CCL2), and CCL5. This causes HSC activation and exacerbates liver fibrosis [6,13,48]. Additionally, the activation of KCs can boost HSCs’ nuclear factor kappa-B (NF-κB), which in turn encourages the release of pro-inflammatory cytokines [49].

2.3. The Molecular Mechanisms of HSC Activation

Multiple signaling pathways can modulate the activation of HSCs at the molecular level. Growth factors and ligand-receptor signaling pathways, profibrogenic response pathways, cell death signaling, immune-related signaling, metabolically regulated pathways, nuclear receptors, and epigenetic changes are the main signaling pathways involved in HSC activation [6]. The TGF-β signal transduction pathway, PDGF signal transduction pathway, Hippo signaling pathway, and ROS that act on the activation of HSCs are mostly summarized in this part (Figure 1).

2.3.1. TGF-β Signaling Pathway

TGF-β, a crucial pro-fibrotic cytokine in the liver [50,51], includes TGF-β1, TGF-β2, and TGF-β3, among which TGF-β1 is the most potent of these proteins and the main regulator of chronic liver disease, increasing the illness’s progression from early liver damage to inflammation, fibrosis, and cirrhosis, even HCC [52,53]. Additionally, TGF-β1 is widely implicated in HSC activation and proliferation [13,23]. The production of laminin, α-smooth muscle actin (α-SMA), fibronectin, and fibrous collagen (mostly types I and III) is increased as a consequence of the most potent cytokine, TGF-β1. Additionally, its expression is relatively low in normal liver tissue, but when liver injury occurs, KCs and LSECs can produce enormous amounts of TGF-β1 to encourage the activation of HSCs, and then the aHSCs subsequently secrete TGF-β1 from themselves to act on themselves or other cells [16,51]. Both canonical (Smad) and non-canonical (non-Smad) pathways can be used by TGF-β to cause fibrosis [54].
A crucial mechanism in the induction of ECM formation during liver fibrosis is the TGF-β/Smads pathway [5,11,53]. Additionally, the two primary downstream mediators are Smad2 and Smad3 [55,56]. Intracellular signal transducers that specifically react to the modification of the TGF-β receptor make up the Smad superfamily [6]. According to their specific functions, the Smad protein family can be divided into three categories. Smad2 and Smad3 are Smads that are receptor-regulated (R-Smads) [13]. In TGF-β/Smad pathways, TGF-β will bind to the TGF-β receptor type II, which then phosphorylates TGF-β receptor type I, recruits and phosphorylates R-Smads (Smad2/3) [53], and then forms a complex with Smad4, which translocates into the nucleus and stimulates target gene transcription (like collagen-I) [53,57,58].
TGF-β may stimulate HSCs through non-Smad pathways as well as the Smad pathway, including mitogen-activated protein kinase (MAPK), extracellular regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), etc. For instance, kindlin-2 overexpression was related positively to the TGF-β signaling pathway, while activated TGF-β could promote kindlin-2 expression through the p38 and MAPK signaling pathways [59,60].
In summary, both Smad and non-Smad pathways have the potential to activate HSCs, which would then favor the formation and progression of liver fibrosis.

2.3.2. PDGF Signaling Pathway

PDGF belongs to a group of growth factors whose physiological functions include controlling cell proliferation and migration and promoting the synthesis of vital components of the connective tissue matrix [23,61]. In normal conditions, platelets produce PDGF. Additionally, upon liver injury, KCs might facilitate the intrahepatic recruitment of platelets [13]. Additionally, aHSCs can also express PDGF, and these cells also express the PDGF receptor (PDGFR) on their surface. The activation of HSCs is then stimulated by PDGF using an autocrine pathway [62,63].
In addition, the phospholipase Cγ(PLCγ), the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway, and the signal transducer and activator of transcription (STAT) pathway can be triggered when PDGFR is stimulated. Once these downstream components control the expression levels of pro-fibrosis target genes, liver fibrosis is facilitated [10,64].

2.3.3. Hippo Signaling Pathway

HSC activation can be regulated by the Hippo pathway [65]. Hippo is a signaling mechanism that modulates the size of cells and organs. Additionally, it is crucial for tissue homeostasis, carcinogenesis, and liver growth regulation during development [53,66]. Yes-associated protein 1 (YAP1), a Hippo pathway downstream effector, is essential for regulating HSC activation in response to chronic damage [53,66]. Yes-associated protein (Yap) is translocated into the nucleus upon hippo pathway kinase inactivation to encourage the transcription of genes like connective tissue growth factor (CTGF) and TGF-β [53]. Moreover, it has been demonstrated that Hippo pathway elements, including YAP1 and the protein kinases macrophage stimulation 1 (MST1) and MST2, are essential for the initial activation of HSCs [67].

2.3.4. Reactive Oxygen Species

ROS can function as signal molecules in numerous signaling pathways, including transcriptional regulation, proliferation, and differentiation [68]. Excessive production of ROS can result in oxidative stress, which promotes a sustained inflammatory response as well as HSC activation and proliferation, which then leads to the appearance and growth of liver fibrosis [68]. ROS are produced as part of a cell’s regular metabolic activity [13]. Additionally, at low concentrations, ROS can function as secondary messengers to trigger various cellular reactions, which can alter cell growth, encourage apoptotic pathways, and aid in the removal of pathogens [13,67]. High ROS concentrations, however, have the potential to damage proteins and DNA, trigger hepatocyte necrosis and apoptosis, release mediators like TGF-β and TNF-α, and eventually lead to HSC activation [67]. In addition to encouraging the recruitment of circulating inflammatory cells into the liver, ROS can encourage KCs to release pro-fibrotic mediators, which in turn activate HSCs [10]. In summary, high levels of ROS are important for HSC activation.

3. Regression of Liver Fibrosis

Although previous studies have made great progress in understanding the pathogenesis and progression of fibrosis, the complicated pathogenesis of fibrosis has presented some challenges in developing anti-hepatic fibrosis medications. Additionally, earlier research supported the belief that fibrosis was an unreversible process, but a growing number of clinical investigations have demonstrated that fibrosis will have regression at least partially when the underlying cause is eliminated [3,17,69]. The liver has a great capacity for regeneration when compared to organs like the lung and kidney, and liver fibrosis has a greater possibility to regress and return to its normal structure than other tissues, which may make the therapy of liver fibrosis easier [3,14].
Recent research has demonstrated that liver fibrosis is typically reversible. Liver fibrosis is not a one-way, irreversible process, even when it is advanced. When the stimulation of liver injury is removed, the liver can activate repair mechanisms that inhibit MF activation and change immune cells, particularly macrophages, from being pro-inflammatory to a repair state [4,28]. Antiviral therapy, for instance, exhibited regression of fibrosis in patients with viral hepatitis-related liver fibrosis, indicating that the underlying cause of the fibrosis can be treated to reverse liver fibrosis [14]. It has been demonstrated that removing the primary cause of chronic inflammation might cause severe liver fibrosis brought on by chronic HBV and hepatitis C virus (HCV) infection to regress [13].
Consequently, it is necessary to develop more effective therapies to delay fibrosis progression or promote regression of liver fibrosis, which can help prevent the advancement of cirrhosis or cancer [12,69]. New treatment targets for liver fibrosis will be identified if the processes of liver fibrosis regression are understood. The main ways to achieve liver fibrosis regression [3] are to eliminate the factors causing liver damage [70], reduce the number of aHSCs [71], inhibit the signal pathways linked to liver fibrosis [72], and promote the degradation of the ECM [73] (Figure 1).

3.1. Reducing the Number of Activated HSCs

The modulation of cell levels, cytokines, and their molecular levels are all part of the intricate mechanism behind liver fibrosis [11]. Notably, stimulation of HSCs is a typical feature of liver fibrosis [11,68]. Eliminating the chronic liver injury causes liver fibrosis to regress along with a decrease or disappearance of aHSCs [74]. Moreover, clinical and experimental studies have both demonstrated that the disappearance of aHSCs by apoptosis [75], inactivation into a quiescent-like state [76,77], or senescence [78] may lead to the regression of liver fibrosis. HSCs may be the primary target of liver fibrosis regression due to their crucial role in the condition [79]. Consequently, reducing the number of aHSCs, including inhibiting their proliferation or activation [7,24], promoting HSC apoptosis or autophagy [11,68,80], and deactivating the activated HSCs [76,77] to reverse liver fibrosis, may be an anti-fibrosis strategy [81].

3.1.1. Limitation of HSC Activation

The phenotype of qHSC can change from static to active when the liver is injured by inflammation or mechanical stimulation [82]. After that, aHSCs can express α-SMA and produce ECM components like collagen and laminin [83]. Moreover, aHSCs can release pro-inflammatory and pro-fibrotic molecules that aid in the development of liver fibrosis. The resulting aHSCs are a crucial part of the fibrosis response that is driven by numerous pathways and is one of the primary effector cells in liver fibrosis [58]. Hence, liver fibrosis can be prevented or reversed by preventing the proliferation or activation of HSCs.
Changes in the cell microenvironment, including soluble mediators, ECM composition, matrix hardness, and interactions with adjacent cells (e.g., damaged hepatocytes and immune cells), can significantly affect the activation of HSCs [3]. A metabolic enzyme called alcohol dehydrogenase (ADH) is mostly expressed in the liver, and ADHI is the classical liver ADH among these family members [84]. Research [85] has shown that the activity of ADHI in human fibrotic liver is significantly higher than that in normal liver. ADHI might encourage the proliferation, motility, adhesion, and invasion of HSC-T6 cells, a rat HSC line. Additionally, the liver fibrosis in mice caused by carbon tetrachloride (CCl4) can be attenuated by the ADH inhibitor 4-methylpyrazole (4-MP), which might be connected to the suppression of the activity of ADHI [86]. This suggests that inhibiting HSC activation can alleviate liver fibrosis. Amydrium hainanense (AH) is a plant drug, and its aqueous extract (AHWE) has an antifibrotic effect on mice with liver injury. The primary mechanism through which AHWE had this impact was by preventing HSC activation and proliferation [87]. The inhibition of circASPH, which is derived from the aspartate β-hydroxylase (ASPH) gene locus, can suppress the activation and proliferation of HSCs, while its upregulation promoted the occurrence of these events in a CCl4-induced mouse model of liver fibrosis [88]. CircDIDO1 (death-inducer obliterator 1) inhibits HSC activation by inhibiting the Akt pathway, and overexpression of circDIDO1 can induce cell cycle arrest and inhibit HSC proliferation and activation in the human HSC cell line LX-2 [89]. All of these provide a new approach to reversing liver fibrosis by inhibiting HSC proliferation or activation.

3.1.2. Apoptosis of HSCs

HSCs’ aberrant activation and proliferation during the progression of liver fibrosis may accelerate the death of other healthy liver cells, leading to further deterioration of the disease [90,91]. A type of programmed cell death called apoptosis helps control the HSCs’ balance between growth and death during fibrosis [3]. Clinical trials showed that the number of apoptotic HSCs decreased with the fibrosis stage increasing, independent of HSC activation or proliferation [92]. Studies on animal models of liver injury induced by different methods have shown that reducing the number of HSCs by inducing apoptosis is directly related to the regression of fibrosis, suggesting that HSC apoptosis is also a major determinant for liver fibrosis regression [75,92]. Therefore, the reversal of liver fibrosis can be achieved by accelerating the death of aHSCs and reducing the total number of aHSCs [93,94].
The authors of [95] have shown that liver fibrosis treatment could rely on endoplasmic reticulum stress (ERS)-mediated apoptosis. Additionally, the ERS inducer tunicamycin (TM) can cause the apoptosis of HSCs through the calpain-2/Ca2+-dependent ERS pathway in the HSC line HSC-T6 [95]. An antiviral medication named tenofovir disoproxil fumarate (TDF) is frequently used to treat chronic hepatitis B (CHB). Additionally, recent clinical studies have shown that TDF treatment can not only suppress the virus but also make liver fibrosis regress. In a mouse model of liver fibrosis, TDF leads to aHSC apoptosis by downregulating the PI3K/Akt/mammalian target of rapamycin (mTOR) signaling pathway and ultimately improving liver fibrosis [96]. In the hunt for medications to treat or reverse liver fibrosis, it is crucial to investigate the process causing HSC apoptosis.

3.1.3. Inactivation of HSCs

Studies have shown that over half of mouse and human HSCs return to a deactivated or inactive state during the reversal of liver fibrosis and avoid apoptosis or cell senescence [76,97]. Consequently, targeting the deactivation of aHSCs has become a novel and effective method for liver fibrosis resolution [76,97].
The inactivation of the zinc finger transcription factor GATA4 in HSCs has been demonstrated to result in aberrant ECM component formation and HSC activation in the embryonic liver [97]. Research has discovered that reactivation of GATA4 could change aHSCs into a quiescent phenotype and then reverse liver fibrosis [83]. Thus, GATA4 may be an appropriate target for the resolution of liver fibrosis. Peroxisome proliferator-activated receptor-γ (PPAR-γ), which is a major regulator of the adipogenesis process, has been found to associate with the activation of HSCs [77]. PPAR-γ levels are noticeably reduced during HSC activation [98], and overexpression of PPAR-γ in aHSCs induces a transition to a quieter phenotype [99,100]. Transcription factor 21 (Tcf21) has been found to be a deactivation factor of fibrogenic HSCs [101]. In vitro and in vivo tests, overexpression of Tcf21 in aHSCs not only suppressed fibrosis-related gene expression but also restored the cells to a quiescent phenotype, at least partially. These phenotypic alterations were accompanied by the regression of liver fibrosis and improved liver function [101]. Thus, Tcf21 may be an option for therapy for the reversal of liver fibrosis.
Our understanding of the underlying molecular processes that lead to aHSC inactivation remains limited. Therefore, the molecular mechanism of aHSC inactivation still needs to be investigated to discover additional candidates for reversing liver fibrosis.

3.2. Inhibiting the Liver Fibrosis-Related Signal Pathways

Liver fibrosis, including the activation of HSCs, involves multiple molecular mechanisms and signaling pathways, and disruption of these pathways helps to reverse the liver fibrosis process. Consequently, the regression of liver fibrosis can be achieved by interfering with these signaling pathways [79,102]. Many signals are crucial to the progression of liver fibrosis, including TGF-β [103], Wnt/β-catenin [104], Notch [105], Hedgehog [106], Hippo [93,107], and inflammasome signaling pathways [108].
The most potent fibrogenic cytokine, TGF-β, has an influence on all stages of disease progression, from primary liver injury to fibrosis and cirrhosis [14,109]. During liver fibrosis, TGF-β is upregulated, and its main role is to activate HSCs. It can also enhance the synthesis of tissue inhibitors of matrix metalloproteinases (TIMPs) and directly promote the production of interstitial fibrillar collagens [110]. Therefore, liver fibrosis can be efficiently prevented by inhibiting TGF-β or disrupting its downstream signaling pathway [51]. Over time, various tactics to inhibit TGF-signaling have been investigated, including targeting TGF-β isomers or suppressing TGF-β receptor activation [110]. In addition, many drugs have been discovered in recent years to regulate fibrosis progression by affecting the TGF-β signaling pathway. For example, Jageum–Jung (JGJ), a possible medication for treating liver fibrosis, can inhibit HSC activation by regulating the TGF-β1/Smad signaling pathway [109]. GNS561 is a novel oral anticancer drug with high hepatotropic properties, and it prevents the activation of HSCs and decreases the deposition of ECM by interfering with TGF-β1 maturation and TGF-β1/Smad signaling [16]. Due to its several crucial functions in immunological control, wound healing, cell differentiation, and proliferation regulation, targeting TGF-β is likely to have unintended harmful effects [14]. Therefore, there is a need to target specific steps of TGF-β activation in a more localized manner to lessen general toxicity. In addition, more targets need to be found to provide more options for liver fibrosis reversal.
Liver fibrosis can also be alleviated by targeting the Notch signaling pathway, so the Notch signaling pathway may be one of the targets for reversing hepatic fibrosis [6]. For example, costunolide (COS), which is isolated from Saussurea lappa, is involved in the WW domain-containing protein 2 (WWP2)-mediated notch homologue protein 3 (Notch3) degradation through ubiquitin-dependent lysosome pathways and subsequent restriction of the Notch3-hairy/enhancer of split-1 (HES1) pathway, thereby alleviating liver fibrosis [111]. In addition, PDGF and its receptors, ROS, YAP/transcriptional co-activator with PDZ-binding motif (TAZ), and other signaling pathways can be used as therapeutic targets for hepatic fibrosis regression.
In addition to the above pathways, the reversal of liver fibrosis can also be achieved through the direct promotion of ECM degradation, immune cells and their signaling pathways, stem cell transplantation, and other methods. However, more clinical trials are needed to reverse liver fibrosis.

4. The Therapeutic Compounds of Liver Fibrosis

Liver fibrosis directly damages liver function, is an essential stage before cirrhosis and is closely related to liver cancer. Numerous cell types, signaling pathways, and chemicals can be thought of as potential therapeutic targets because they all have a significant impact on the emergence, development, and resolution of liver fibrosis [8]. However, there is currently no clinically approved effective anti-fibrosis therapy for slowing or reversing liver fibrosis [7,112]. Treatment targeting the cause is thought to be the most effective method to prevent and treat liver fibrosis [57]. Though liver fibrosis may be slowed or reversed by eliminating the pathogen, it often happens too slowly or too little to avoid life-threatening complications. It is feasible to treat liver fibrosis with medicinal intervention, as some anti-fibrosis medications have demonstrated a potential function in inhibiting the advancement of anti-fibrosis in animal studies [12]. Certain compounds currently in clinical development have limited clinical efficacy, so the medical need to develop more effective and safer anti-fibrosis drugs remains high [102]. Therefore, we summarize the compounds that have been found to have therapeutic effects on hepatic fibrosis in recent years (Figure 2).

4.1. Natural Compounds

Natural compounds have gained prominence globally in recent years as one of the most significant sources of anti-fibrosis medications due to their affordability and accessibility [8,57]. Several studies have reported that natural products, like terpenoids, phenols, alkaloids, etc., as well as crude extracts or isolated molecules from plants, can alleviate various fibrosis diseases in vitro and in vivo [57]. Here is a summary of natural compounds that have been suggested to reduce liver fibrosis and their primary mechanisms of action (Table 1).
A variety of cancers, inflammation, and viral infections can be frequently managed with herbal plants. 18β-Glycyrrhetinic acid (18β-GA), a triterpenoid aglycone, is one of the chemical components of the natural product Glycyrrhizic acid [19,113,114]. Additionally, in mice with liver fibrosis brought on by bile duct ligation (BDL), 18β-GA can reduce liver fibrosis by causing ROS-mediated apoptosis in aHSCs [19]. In traditional Chinese medicine, Gan Huang Cao is a Miao plant used for medicinal and edible purposes [115]. Additionally, 88 substances, like flavonoids, organic acids, and coumarins, were extracted and identified from it by the researchers. The primary component of flavonoids, kaempferol (KA), has a variety of anti-inflammatory, antioxidant, and anti-apoptotic properties [115,116]. Studies have demonstrated that kaempferol could effectively attenuate the development of liver fibrosis [117,118]. The findings showed that kaempferol could inhibit HSC activation and suppress HSC collagen synthesis via downregulating the levels of phosphorylated Smad2 and Smad3, as well as the TGF-β1/Smads pathway [117]. Kaempferol can also inhibit HSC activation through the miR-26b-5p/Jag1 axis and the Notch pathway [118]. Therefore, kaempferol may be a novel natural compound to treat liver fibrosis. Ferulic acid (FA) is a polyphenolic compound that is abundant in grains, vegetables, and plants. FA is also among the primary active ingredients in various Chinese herbs like Angelica sinensis. FA exhibits a broad spectrum of biological functions, including antioxidant, enhancement of immune function, anti-inflammatory, and hepatoprotective properties [25,119]. FA has been found to alleviate liver fibrosis in rats by CCl4-induced liver fibrosis. In terms of molecular mechanisms, FA can reduce the activation of HSCs that is brought on by TGF-β1 through the inhibition of Smad2/3 phosphorylation and subsequent Smad4 signal transduction. Blocking these pathways could contribute to reversing the liver fibrosis process [120].
Table 1. Anti-fibrotic natural compounds.
Table 1. Anti-fibrotic natural compounds.
CompoundsAnimal ModelTargets/Pathways/MechanismsReference
18beta-glycyrrhetinic acid (18β-GA)BDL-induced liver fibrosis in miceTargeting PRDX1/2[19]
Resulting in the accumulation of cellular ROS
Inducing apoptosis in activated HSCs
Kaempferol (KA)CCl4-induced liver fibrosis in female C57BL/6 miceInhibiting Notch pathway via miR-26b-5p/Jag1[116,117,118]
Downregulation of TGF-β1/Smad2/3 signaling
Inhibiting HSC activation
Swertia purpurascens Wall extract (SPE)CCl4-induced liver fibrosis in male Wistar ratsInhibition of TGF-β/Smad/NF-κB signaling[9]
Inhibiting oxidative stress and inflammation
Demethylzeylasteral (T-96)CCl4-induced liver fibrosis in male C57BL/6J miceSuppressing AGAP2-mediated FAK/Akt signaling [121]
Inhibition of downstream TGF-β1 induced fibrosis-related gene expression
Ferulic acid (FA)CCl4-induced fibrotic models in male C57BL/6J miceInhibiting hepatic oxidative stress, macrophage activation, and HSC activation[25,120]
CCl4-induced liver fibrosis in male Wistar ratsThrough PTP1B and AMPK signaling pathways.
Inhibition of the TGF-β1/Smad pathway
Methoxyeugenol CCl4-induced liver fibrosis in male BALB/c miceModulating the activated phenotype of HSCs by activating PPAR-γ[57]
A protective effect against oxidative stress damage in hepatocytes
Benzoquinone derivatives TAA-induced liver fibrosis in male Balb/C mice Ameliorating oxidative stress and inflammation [68]
Inducing apoptosis in activated LX-2 cells
Attenuating liver fibrosis in TAA-induced mice
Carnosol (CS)CCl4-induced liver fibrosis in male SD ratsActivation of SIRT1/EZH2[28]
Inhibiting HSC activation and reversing EMT
Carnosic acid (CA)BDL-induced liver fibrosis in male SD ratsModulating the miR-29b-3p/HMGB1/TLR4 signaling pathway[7]
Attenuating BDL-induced liver fibrosis
Stachydrine (STA)CCl4-induced liver fibrosis in male SD ratsInhibiting inflammation and oxidative stress [122]
Upregulating the ratio of MMPs/TIMPs to promote the degradation of ECM
CatalpolCCl4-induced liver fibrosis in male SD ratsActivating autophagy to exert anti-inflammatory effects[8]
Inhibiting the activation of HSCs
Mogroside IVE (MGIVE)CCl4-induced liver fibrosis in male C57BL/6 miceInhibiting the TLR4 signaling pathway[112]
Reducing inflammatory responses
Inhibiting TGF-β1 induced HSC activation
MorinDEN-induced liver fibrosis in male Wistar ratsActivating HIPPO signaling[53]
Downregulating TGF-β signaling
Attenuating fibrillar collagen deposition
Preventing HSC activation
S-allyl cysteine (SAC)CCl4-induced liver fibrosis in male Wistar ratsInhibiting the expression of TGF-β[123]
A decrease in the profibrogenic cytokine, TGF-β
Correcting cytokine abnormality
SMND-309CCl4-induced liver fibrosis in male SD ratsSuppressing the expression of CTGF[51]
Scavenging lipid peroxidation products
Increasing endogenous anti-oxidation enzyme activity
Quercetin (QE)CCl4-induced liver fibrosis in male C57 miceSuppressing the TGF-β1/Smads signaling pathway[124]
BDL-induced liver fibrosis in male C57 miceActivating the PI3K/Akt signaling pathway to inhibit autophagy
Attenuating HSC activation
Tormentic acid (TA)CCl4-induced liver fibrosis in male SD ratsSuppressing HSC activation[125]
Suppressing the PI3K/Akt/mTOR signaling pathway
Inhibiting the NF-κB signaling pathway
Gomisin DCCl4-induced liver fibrosis in male Balb/C miceInhibiting HSC proliferation and activation[82]
Promoting HSC apoptosis
Regulating the PDGF-BB/PDGFRβ signaling pathway
Costunolide (COS)CCl4-induced liver fibrosis in male Balb/C miceRegulating the Notch3–HES1 pathway[111]
BDL-induced liver fibrosis in male SD ratsDisturbing the PPM1G/WWP2 complex
Blocking the inhibitory effect of PPM1G on WWP2
5-Methoxytryptophan (5-MTP)CCl4-induced liver fibrosis in male SD ratsRegulation of the FOXO3a/miR-21/ATG5 pathway[126]
Inducing autophagy and suppressing HSCs’ activation
Physalin BCCl4-induced liver fibrosis in male C57BL/6J miceDisrupting LAP2α/HDAC1 complexes[127]
BDL-induced liver fibrosis in male C57BL/6J miceInhibiting HDAC1-mediated GLI1 deacetylation
Inhibiting HSC activation
Ginsenoside Rg1 (G-Rg1)CCl4-induced liver fibrosis male C57BL/6J miceAttenuating IDO1-mediated inhibition of maturation of DCs[128]
Inhibiting the proliferation of HSCs
Germarcone (GER)MCS/MCD diet induction in male C57BL/6 miceReducing the release of ROS[129]
Regulating TGF-β/Smad and apoptosis pathways
Inhibiting the activation and survival of HSCs
SwerosideCCl4-induced liver fibrosis in male C57BL/6 miceRegulation of FXR-miR-29a signaling pathway[130]
Inhibition of HSC proliferation
ArtesunateCCl4-induced liver fibrosis in male ICR miceActivating HSC ferroptosis[91]
Inhibiting HSC activation
Linderalactone (LIN)CCl4-induced liver fibrosis in C57BL/6 miceInhibiting TGF-β/Smad signaling[131]
Suppressing HSC activation
In recent decades, Chinese herbal formulas, including more than two kinds of Traditional Chinese Medicine (TCM), have been extensively utilized to treat diseases of the liver due to their synergistic therapeutic effect. Hence, Chinese herbal formulations have been researched for their anti-liver fibrosis effects [7,69,132]. Fuzheng Huayu (FZHY) formula, a formulation from TCM, is often utilized to treat chronic liver diseases in China [133,134]. Additionally, it is composed of Salvia miltiorrhiza, Prunus davidiana, Schisandra chinensis, Pinus massoniana, and Gynostemma pentaphyllum, etc. Additionally, FZHY has been demonstrated to improve liver fibrosis by targeting more than one molecule, like the TGF-β/Smad signaling pathways and TNF-α-induced apoptosis of hepatocytes [135,136]. A new TCM formula, named JY5, was obtained by the analysis of the FZHY’s primary active components, including salvianolic acid B, schisantherin A, and amygdalin [137]. Additionally, this study also demonstrated that JY5 blocked HSC activation by suppressing the Notch signaling pathway to alleviate liver fibrosis in animal models of liver fibrosis. Table 2 summarizes more Chinese herbal formulations and their main anti-fibrosis mechanisms.

4.2. Synthetic Compounds

To meet the needs of liver fibrosis treatment, a variety of highly effective compounds or improved derivatives can also be synthesized by chemical means, such as small molecular compounds and pyrazole derivatives.
PI3K is an essential signaling molecule that regulates numerous cellular functions [141]. When stimulated, PI3K is activated and phosphorylated to promote Akt phosphorylation, which can regulate multiple apoptosis-related signal transduction pathways [125]. Moreover, the PI3K/Akt pathway is also tightly linked to the incidence and development of liver fibrosis [139]. In CCl4-induced liver fibrosis mice, PI3K/Akt signaling inhibition can ameliorate liver fibrosis [139]. HS-173 [142] is a PI3K inhibitor. Additionally, the study has demonstrated that it could block the PI3K/Akt signaling pathway to induce HSC apoptosis and inhibit proliferation, which can attenuate the progression of liver fibrosis [142]. Consequently, HS-173 may be a useful medicinal compound for treating liver fibrosis. The derivatives of barbituric acids, a class of aromatic hydrocarbons, are crucial in biology and medicine [143]. Antioxidant, anti-inflammatory, and other biological activities are a few of the biological effects of barbituric acids and their derivatives [143,144]. Novel barbituric acid (BA) derivatives have been found to be able to prevent rats from developing non-alcoholic fatty liver disease (NAFLD) [145]. BA derivatives may therefore be a potential medicinal compound for treating liver fibrosis. BA-5 was screened for six distinct barbiturate derivatives’ antifibrotic effects on activating HSCs induced by TGF-β1 [102]. Additionally, further research revealed that BA-5 could block the TGF-β1 signaling pathways and the NF-κB signaling pathways induced by lipopolysaccharide (LPS) to inhibit HSC activation and liver fibrosis. Moreover, BA-5 can also inhibit macrophage recruitment and activation [102]. Altogether, BA-5 may be an efficient synthetic compound for liver fibrosis due to its dual therapeutic potential. Table 3 summarizes more synthetic compounds and their main anti-fibrosis mechanisms.

4.3. Drug Delivery System

Liver fibrosis, a serious disease, is a major threat to human health. With continuous, in-depth research, numerous anti-fibrosis medications have been found and show potent anti-fibrosis actions in vitro. However, there is no effective drug to treat liver fibrosis in the clinic. The reason is that most therapeutic medicines lack specificity, have limited solubility, and have poor liver accumulation effects. Accordingly, therapeutic drugs cannot reach an effective therapeutic concentration in vivo and are easily absorbed by non-target organ cells. For example, silymarin and curcumin are very effective against liver fibrosis. However, their application is restricted due to their low bioavailability, poor water solubility, lack of specific targeting, and additional liver toxicity [15,154]. To improve the efficacy of drug therapy, drug delivery systems, especially nanoparticles (NPs), have attracted extensive attention.
Exosomes are nanosized extracellular vesicles, usually between 30 and 150 nm in size, and some exosomes derived from stem cells can reduce liver fibrosis by carrying components that regulate HSC activation [155,156]. Exosomes can cross biological barriers to transfer nucleic acids, proteins, and other bioactive molecules [157]. Therefore, exosomes can also be used as delivery carriers for the treatment of hepatic fibrosis [34]. Osteopontin (OPN) is a matrix-bound protein that plays a significant role in liver fibrosis [158]. The exosomes carry small interfering RNA (siRNA) targeting OPN (ExosiRNA-OPN) and can deliver siRNA safely and effectively into the liver to reduce OPN expression. ExosiRNA−OPN was found to inhibit TGF-β1 signaling by reducing HMGB1, then to attenuate the progression of liver fibrosis in a CCl4-induced mouse model of liver fibrosis [157].
With the development of nanotechnology, NPs provide new therapeutic opportunities for drug delivery to treat liver fibrosis. The nanomedicine delivery system can be employed to specifically target therapeutic agents to key cell types to alleviate or reverse liver fibrosis and achieve combination therapy with little systemic toxicity [1,159]. NPs have advantages such as adjustable dimensions and shapes, various compositions, and changeable surface characteristics, which can control drug release, improve drug pharmacokinetics, reduce adverse reactions, and promote targeted drug delivery and combination treatment of liver fibrosis, so as to solve the problem that the anti-fibrosis effect of traditional drugs is limited in vivo [15,159,160]. Nano-drug delivery systems are frequently divided into inorganic and organic NPs based on their natural properties. Metal ion NPs, such as Ag NPs and inorganic oxide NPs, are instances of inorganic NPs [1]. However, the biodegradability of inorganic NPs in vivo is poor, and the leftover NPs that accumulate there can harm the liver. Additionally, organic polymer materials like polyethylene glycol (PEG), polylactic acid (PLA), etc. can be used to synthesize organic NPs. The majority of organic NPs exhibit strong in vivo biodegradability, low toxicity, and few side effects [1]. Organic NPs, including lipid nanoparticles (LNPs), polymer NPs, and protein NPs, have been widely studied in treating liver fibrosis. These NPs can target the delivery of therapeutic drugs to the key cells of liver fibrosis, especially HSCs, thereby reducing the adverse reactions of drugs and improving the therapeutic effect of drugs in vivo.
TNF-stimulated gene 6 (TSG-6) was identified as a pivotal antifibrosis cytokine of mesenchymal stem cells that showed strong antifibrotic activity. Studies have shown specific liver targeting and high TSG-6 loading efficacy of calcium phosphate (CaP) @ bovine serum albumin (BSA) NPs. The CCl4-induced fibrosis in mice therapeutic study further demonstrated the improved therapeutic effects of TSG-6-loaded CaP@BSA [161]. LNPs include stable nucleic acid LNPs (SNALPs), phospholipids, steroid-based drug coupling liposomes, nanostructured lipid carriers (NLCs), etc. [162]. For example, the main peptide component found in honeybee venom, melittin, has been shown to be useful in lowering aHSC populations in fibrotic liver tissues [163]. However, the systemic toxicity caused by its nonspecific action limits its clinical application in fibrosis. To reduce the nonspecific effects of melittin, targeted drug delivery systems can be used to enable it to work in just fibrotic tissue. Fibroblast activation protein (FAP) is specifically overexpressed on the surfaces of aHSCs in the fibrotic liver [164]. This study [163] designed a delivery system to release melittin into the fibrotic liver surroundings by cleaving promelittin exclusively in liver areas that express FAP. Additionally, promelittin was attached to the PEGylated liposomes’ surfaces to improve liver transport and bloodstream stability. The results showed that promelittin-modified liposomes (PRL) are selectively activated in the fibrotic liver, and the FAP-specific liberation of melittin can decrease the number of aberrant HSCs and reduce the excessive production of collagen fibers to alleviate fibrosis. Moreover, PRL may be designed to increase antifibrotic efficacy by encasing additional antifibrotic chemical compounds in their core. In addition, the vitamin A-coupled liposomal Rho/Rho-associated protein kinase (ROCK) inhibitor may reduce liver fibrosis without causing systemic side effects while inhibiting HSC activation by targeting HSCs for drug delivery [165]. The SB431542-loaded liposomes showed better efficacy for treating liver fibrosis by inhibiting TGF-β signaling. Loading SB431542 into liposomes solves the limitations of poor water solubility and low bioavailability of this small-molecule drug [166].
However, the clinical transformation of nanomedicine for anti-fibrosis therapy is still complex in terms of specific targeting, safety, etc. More understanding of potential targets for hepatic fibrosis is therefore needed to identify promising anti-fibrosis drug candidates. Hence, the clinical application of nano-drug delivery systems must overcome many challenges.

5. Conclusions

Liver fibrosis is the organism’s attempt to repair chronic liver damage brought on by various pathogenic agents. With the in-depth study of liver damage and fibrosis mechanisms in the past few decades, it has been discovered that liver fibrosis is in a reversible stage and the liver has a strong regenerative ability. If timely treatment is carried out during this period, liver fibrosis reversal can be achieved and the liver can recover to a mostly healthy state. Therefore, it is important to diagnose and treat liver fibrosis early and promptly for the prevention of cirrhosis and liver cancer. However, since various cells in the body, including immune cells and signaling pathways, play dual roles in different liver microenvironments, it is crucial to further investigate the molecular mechanism of hepatic fibrosis regression to find more drug targets and efficient anti-fibrosis drugs, which are crucial for the effective treatment of hepatic fibrosis.

Author Contributions

Conceptualization, Q.P. and L.T.; writing—original draft preparation, Q.P.; writing—review and editing, L.T. and Q.Y.; funding acquisition, L.T. and Q.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (No. 32271363), the Luzhou Science and Technology Program (No. 2021-JYJ-71), and the Sichuan Science and Technology Program (No. 2022NSFSC1368).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Peng, W.; Cheng, S.; Bao, Z.; Wang, Y.; Zhou, W.; Wang, J.; Yang, Q.; Chen, C.; Wang, W. Advances in the research of nanodrug delivery system for targeted treatment of liver fibrosis. Biomed. Pharmacother. 2021, 137, 111342. [Google Scholar] [CrossRef] [PubMed]
  2. Hadi, F.; Awan, S.J.; Tayyeb, A.; Maqbool, T.; Shehzadi, S.; Malik, S.; Kausar, H.; Malik, A. Hepato-protective role of itraconazole mediated cytochrome p450 pathway inhibition in liver fibrosis. Pak. J. Pharm. Sci. 2020, 33, 2751–2758. [Google Scholar] [CrossRef] [PubMed]
  3. Caligiuri, A.; Gentilini, A.; Pastore, M.; Gitto, S.; Marra, F. Cellular and Molecular Mechanisms Underlying Liver Fibrosis Regression. Cells 2021, 10, 2759. [Google Scholar] [CrossRef] [PubMed]
  4. Gao, C.C.; Bai, J.; Han, H.; Qin, H.Y. The versatility of macrophage heterogeneity in liver fibrosis. Front. Immunol. 2022, 13, 968879. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, H.; Chen, Q.S.; Dahan, A.; Xue, J.Y.; Wei, L.W.; Tan, W.F.; Zhang, G.Q. Transcriptomic analyses reveal the molecular mechanisms of schisandrin B alleviates CCl4-induced liver fibrosis in rats by RNA-sequencing. Chem. Biol. Interact. 2019, 309, 108675. [Google Scholar] [CrossRef]
  6. Zhang, J.; Liu, Q.; He, J.; Li, Y. Novel Therapeutic Targets in Liver Fibrosis. Front. Mol. Biosci. 2021, 8, 766855. [Google Scholar] [CrossRef]
  7. Zhang, S.; Wang, Z.C.; Zhu, J.; Xu, T.; Zhao, Y.; Zhao, H.Y.; Tang, F.; Li, Z.L.; Zhou, J.J.; Gao, D.Y.; et al. Carnosic Acid Alleviates BDL-Induced Liver Fibrosis through miR-29b-3p-Mediated Inhibition of the High-Mobility Group Box 1/Toll-Like Receptor 4 Signaling Pathway in Rats. Front. Pharmacol. 2018, 8, 976. [Google Scholar] [CrossRef] [Green Version]
  8. Liu, Z.; Zhu, P.; Zhang, L.; Xiong, B.; Tao, J.; Guan, W.; Li, C.; Chen, C.; Gu, J.; Duanmu, J.; et al. Autophagy inhibition attenuates the induction of anti-inflammatory effect of catalpol in liver fibrosis. Biomed. Pharmacother. 2018, 103, 1262–1271. [Google Scholar] [CrossRef]
  9. Raj, D.; Sharma, V.; Upadhyaya, A.; Kumar, N.; Joshi, R.; Acharya, V.; Kumar, D.; Patial, V. Swertia purpurascens Wall ethanolic extract mitigates hepatic fibrosis and restores hepatic hepcidin levels via inhibition of TGF beta/SMAD/NF kappa B signaling in rats. J. Ethnopharmacol. 2022, 284, 114741. [Google Scholar] [CrossRef]
  10. Milito, A.; Brancaccio, M.; D’Argenio, G.; Castellano, I. Natural Sulfur-Containing Compounds: An Alternative Therapeutic Strategy against Liver Fibrosis. Cells 2019, 8, 1356. [Google Scholar] [CrossRef] [Green Version]
  11. Li, J.J.; Guo, C.Y.; Wu, J.Y. Astaxanthin in Liver Health and Disease: A Potential Therapeutic Agent. Drug. Des. Dev. Ther. 2020, 14, 2275–2285. [Google Scholar] [CrossRef]
  12. Kaftanovskaya, E.M.; Ng, H.H.; Soula, M.; Rivas, B.; Myhr, C.; Ho, B.A.; Cervantes, B.A.; Shupe, T.D.; Devarasetty, M.; Hu, X.; et al. Therapeutic effects of a small molecule agonist of the relaxin receptor ML290 in liver fibrosis. Faseb J. 2019, 33, 12435–12446. [Google Scholar] [CrossRef] [Green Version]
  13. Roehlen, N.; Crouchet, E.; Baumert, T.F. Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives. Cells 2020, 9, 875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rahman, S.R.; Roper, J.A.; Grove, J.I.; Aithal, G.P.; Pun, K.T.; Bennett, A.J. Integrins as a drug target in liver fibrosis. Liver Int. 2022, 42, 507–521. [Google Scholar] [CrossRef] [PubMed]
  15. Gu, L.; Zhang, F.; Wu, J.; Zhuge, Y. Nanotechnology in Drug Delivery for Liver Fibrosis. Front. Mol. Biosci. 2021, 8, 804396. [Google Scholar] [CrossRef] [PubMed]
  16. Bestion, E.; Jilkova, Z.M.; Mege, J.L.; Novello, M.; Kurma, K.; Pour, S.T.A.; Lalmanach, G.; Vanderlynden, L.; Fizanne, L.; Bassissi, F.; et al. GNS561 acts as a potent anti-fibrotic and pro-fibrolytic agent in liver fibrosis through TGF-beta 1 inhibition. Ther. Adv. Chronic Dis. 2020, 11, 2040622320942042. [Google Scholar] [CrossRef]
  17. Feng, M.; Ding, J.; Wang, M.; Zhang, J.; Zhu, X.; Guan, W. Kupffer-derived matrix metalloproteinase-9 contributes to liver fibrosis resolution. Int. J. Biol. Sci. 2018, 14, 1033–1040. [Google Scholar] [CrossRef]
  18. Itaba, N.; Kono, Y.; Watanabe, K.; Yokobata, T.; Oka, H.; Osaki, M.; Kakuta, H.; Morimoto, M.; Shiota, G. Reversal of established liver fibrosis by IC-2-engineered mesenchymal stem cell sheets. Sci. Rep. 2019, 9, 6841. [Google Scholar] [CrossRef] [Green Version]
  19. Zhang, Q.; Luo, P.; Zheng, L.H.; Chen, J.Y.; Zhang, J.Z.; Tang, H.; Liu, D.D.; He, X.L.; Shi, Q.L.; Gu, L.W.; et al. 18beta-glycyrrhetinic acid induces ROS-mediated apoptosis to ameliorate hepatic fibrosis by targeting PRDX1/2 in activated HSCs. J. Pharm. Anal. 2022, 12, 570–582. [Google Scholar] [CrossRef]
  20. Li, W.Y.; Chen, J.Y.; Sun, C.; Sparks, R.P.; Pantano, L.; Rahman, R.U.; Moran, S.P.; Pondick, J.V.; Kirchner, R.; Wrobel, D.; et al. Nanchangmycin regulates FYN, PTK2, and MAPK1/3 to control the fibrotic activity of human hepatic stellate cells. eLife 2022, 11, e74513. [Google Scholar] [CrossRef]
  21. Wu, K.K. Control of Tissue Fibrosis by 5-Methoxytryptophan, an Innate Anti-Inflammatory Metabolite. Front. Pharmacol. 2021, 12, 759199. [Google Scholar] [CrossRef]
  22. Haak, A.J.; Kostallari, E.; Sicard, D.; Ligresti, G.; Choi, K.M.; Caporarello, N.; Jones, D.L.; Tan, Q.; Meridew, J.; Diaz Espinosa, A.M.; et al. Selective YAP/TAZ inhibition in fibroblasts via dopamine receptor D1 agonism reverses fibrosis. Sci. Transl. Med. 2019, 11, eaau6296. [Google Scholar] [CrossRef]
  23. Wang, F.D.; Zhou, J.; Chen, E.Q. Molecular Mechanisms and Potential New Therapeutic Drugs for Liver Fibrosis. Front. Pharmacol. 2022, 13, 787748. [Google Scholar] [CrossRef]
  24. Zhang, Y.; Yu, X.; Wang, Z.; Yin, M.; Zhao, Z.; Li, Y.; Li, W. Pokeweed antiviral protein attenuates liver fibrosis in mice through regulating Wnt/Jnk mediated glucose metabolism. Saudi J. Gastroenterol. 2018, 24, 157–164. [Google Scholar] [CrossRef]
  25. Wu, J.Z.; Xue, X.Y.; Fan, G.F.; Gu, Y.Q.; Zhou, F.; Zheng, Q.; Liu, R.P.; Li, Y.J.; Ma, B.N.; Li, S.; et al. Ferulic Acid Ameliorates Hepatic Inflammation and Fibrotic Liver Injury by Inhibiting PTP1B Activity and Subsequent Promoting AMPK Phosphorylation. Front. Pharmacol. 2021, 12, 754976. [Google Scholar] [CrossRef]
  26. Karimi, J.; Mohammadalipour, A.; Sheikh, N.; Khodadadi, I.; Hashemnia, M.; Goudarzi, F.; Khanjarsim, V.; Solgi, G.; Hajilooi, M.; Bahabadi, M.; et al. Protective effects of combined Losartan and Nilotinib on carbon tetrachloride (CCl4)-induced liver fibrosis in rats. Drug. Chem. Toxicol. 2020, 43, 468–478. [Google Scholar] [CrossRef]
  27. Ding, N.; Wei, B.; Fu, X.H.; Wang, C.; Wu, Y.M. Natural Products that Target the NLRP3 Inflammasome to Treat Fibrosis. Front. Pharmacol. 2020, 11, 591393. [Google Scholar] [CrossRef]
  28. Zhao, H.Y.; Wang, Z.C.; Tang, F.; Zhao, Y.; Feng, D.C.; Li, Y.; Hu, Y.; Wang, C.; Zhou, J.J.; Tian, X.F.; et al. Carnosol-mediated Sirtuin 1 activation inhibits Enhancer of Zeste Homolog 2 to attenuate liver fibrosis. Pharmacol. Res. 2018, 128, 327–337. [Google Scholar] [CrossRef]
  29. Wei, M.J.; Zhang, Y.; Zhang, H.; Huang, Z.L.; Miao, H.; Zhang, T.Y.; Lu, B.; Ji, L.L. HMGB1 induced endothelial to mesenchymal transition in liver fibrosis: The key regulation of early growth response factor 1. Biochim. Et Biophys. Acta-Gen. Subj. 2022, 1866, 130202. [Google Scholar] [CrossRef]
  30. Garbuzenko, D.V. Pathophysiological mechanisms of hepatic stellate cells activation in liver fibrosis. World J. Clin. Cases 2022, 10, 3662–3676. [Google Scholar] [CrossRef]
  31. Nokkeaw, A.; Thamjamrassri, P.; Tangkijvanich, P.; Ariyachet, C. Regulatory Functions and Mechanisms of Circular RNAs in Hepatic Stellate Cell Activation and Liver Fibrosis. Cells 2023, 12, 378. [Google Scholar] [CrossRef] [PubMed]
  32. Hu, X.; Ge, Q.; Zhang, Y.; Li, B.; Cheng, E.; Wang, Y.; Huang, Y. A review of the effect of exosomes from different cells on liver fibrosis. Biomed. Pharmacother. 2023, 161, 114415. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, L.; Wang, Y.; Quan, J. Exosomes derived from natural killer cells inhibit hepatic stellate cell activation and liver fibrosis. Hum. Cell 2020, 33, 582–589. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, Y.; Zheng, Y.; Yang, Y.; Liu, K.; Wu, J.; Gao, P.; Zhang, C. Exosomes in liver fibrosis: The role of modulating hepatic stellate cells and immune cells, and prospects for clinical applications. Front. Immunol. 2023, 14, 1133297. [Google Scholar] [CrossRef] [PubMed]
  35. Pi, L.; Sun, C.; Jn-Simon, N.; Basha, S.; Thomas, H.; Figueroa, V.; Zarrinpar, A.; Cao, Q.; Petersen, B. CCN2/CTGF promotes liver fibrosis through crosstalk with the Slit2/Robo signaling. J. Cell. Commun. Signal. 2023, 17, 137–150. [Google Scholar] [CrossRef] [PubMed]
  36. Zhao, J.; Qi, Y.F.; Yu, Y.R. STAT3: A key regulator in liver fibrosis. Ann. Hepatol. 2021, 21, 100224. [Google Scholar] [CrossRef]
  37. Schulze, R.J.; Schott, M.B.; Casey, C.A.; Tuma, P.L.; McNiven, M.A. The cell biology of the hepatocyte: A membrane trafficking machine. J. Cell. Biol. 2019, 218, 2096–2112. [Google Scholar] [CrossRef]
  38. Ni, Y.A.; Chen, H.; Nie, H.; Zheng, B.; Gong, Q. HMGB1: An overview of its roles in the pathogenesis of liver disease. J. Leukoc. Biol. 2021, 110, 987–998. [Google Scholar] [CrossRef]
  39. Li, J.; Zeng, C.; Zheng, B.; Liu, C.; Tang, M.; Jiang, Y.; Chang, Y.; Song, W.; Wang, Y.; Yang, C. HMGB1-induced autophagy facilitates hepatic stellate cells activation: A new pathway in liver fibrosis. Clin. Sci. 2018, 132, 1645–1667. [Google Scholar] [CrossRef]
  40. Luo, X.; Xu, Z.X.; Wu, J.C.; Luo, S.Z.; Xu, M.Y. Hepatocyte-derived exosomal miR-27a activateshepatic stellate cells through the inhibitionof PINK1-mediated mitophagy in MAFLD. Mol. Ther. Nucleic Acids 2021, 26, 1241–1254. [Google Scholar] [CrossRef]
  41. Ding, B.S.; Nolan, D.J.; Butler, J.M.; James, D.; Babazadeh, A.O.; Rosenwaks, Z.; Mittal, V.; Kobayashi, H.; Shido, K.; Lyden, D.; et al. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature 2010, 468, 310–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. DeLeve, L.D. Liver sinusoidal endothelial cells in hepatic fibrosis. Hepatology 2015, 61, 1740–1746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Poisson, J.; Lemoinne, S.; Boulanger, C.; Durand, F.; Moreau, R.; Valla, D.; Rautou, P.E. Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J. Hepatol. 2017, 66, 212–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Xie, G.; Wang, X.; Wang, L.; Wang, L.; Atkinson, R.D.; Kanel, G.C.; Gaarde, W.A.; Deleve, L.D. Role of differentiation of liver sinusoidal endothelial cells in progression and regression of hepatic fibrosis in rats. Gastroenterology 2012, 142, 918–927.e916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Jaeschke, H. Reactive oxygen and mechanisms of inflammatory liver injury: Present concepts. J. Gastroenterol. Hepatol. 2011, 26 (Suppl. S1), 173–179. [Google Scholar] [CrossRef] [PubMed]
  46. Tacke, F.; Zimmermann, H.W. Macrophage heterogeneity in liver injury and fibrosis. J. Hepatol. 2014, 60, 1090–1096. [Google Scholar] [CrossRef] [Green Version]
  47. Wynn, T.A.; Barron, L. Macrophages: Master regulators of inflammation and fibrosis. Semin. Liver Dis. 2010, 30, 245–257. [Google Scholar] [CrossRef] [Green Version]
  48. Abdelghffar, E.A.; Obaid, W.A.; Alamoudi, M.O.; Mohammedsaleh, Z.M.; Annaz, H.; Abdelfattah, M.A.O.; Sobeh, M. Thymus fontanesii attenuates CCl4-induced oxidative stress and inflammation in mild liver fibrosis. Biomed. Pharmacother. 2022, 148, 112738. [Google Scholar] [CrossRef]
  49. Liu, C.; Tao, Q.; Sun, M.; Wu, J.Z.; Yang, W.; Jian, P.; Peng, J.; Hu, Y.; Liu, C.; Liu, P. Kupffer cells are associated with apoptosis, inflammation and fibrotic effects in hepatic fibrosis in rats. Lab. Invest. 2010, 90, 1805–1816. [Google Scholar] [CrossRef] [Green Version]
  50. Dewidar, B.; Meyer, C.; Dooley, S.; Meindl-Beinker, A.N. TGF-beta in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019, 8, 1419. [Google Scholar] [CrossRef] [Green Version]
  51. Hou, J.; Tian, J.; Jiang, W.; Gao, Y.; Fu, F. Therapeutic effects of SMND-309, a new metabolite of salvianolic acid B, on experimental liver fibrosis. Eur. J. Pharmacol. 2011, 650, 390–395. [Google Scholar] [CrossRef]
  52. Ghafoory, S.; Varshney, R.; Robison, T.; Kouzbari, K.; Woolington, S.; Murphy, B.; Xia, L.; Ahamed, J. Platelet TGF-beta1 deficiency decreases liver fibrosis in a mouse model of liver injury. Blood Adv. 2018, 2, 470–480. [Google Scholar] [CrossRef] [Green Version]
  53. Perumal, N.; Perumal, M.; Halagowder, D.; Sivasithamparam, N. Morin attenuates diethylnitrosamine-induced rat liver fibrosis and hepatic stellate cell activation by co-ordinated regulation of Hippo/Yap and TGF-beta1/Smad signaling. Biochimie 2017, 140, 10–19. [Google Scholar] [CrossRef]
  54. Finnson, K.W.; Almadani, Y.; Philip, A. Non-canonical (non-SMAD2/3) TGF-beta signaling in fibrosis: Mechanisms and targets. Semin. Cell. Dev. Biol. 2020, 101, 115–122. [Google Scholar] [CrossRef]
  55. Zhang, J.H.; Li, Y.P.; Liu, Q.H.; Huang, Y.; Li, R.; Wu, T.; Zhang, Z.J.; Zhou, J.; Huang, H.; Tang, Q.; et al. Sirt6 Alleviated Liver Fibrosis by Deacetylating Conserved Lysine 54 on Smad2 in Hepatic Stellate Cells. Hepatology 2021, 73, 1140–1157. [Google Scholar] [CrossRef]
  56. Wang, Y.; Tu, K.; Liu, D.; Guo, L.; Chen, Y.; Li, Q.; Maiers, J.L.; Liu, Z.; Shah, V.H.; Dou, C.; et al. p300 Acetyltransferase Is a Cytoplasm-to-Nucleus Shuttle for SMAD2/3 and TAZ Nuclear Transport in Transforming Growth Factor beta-Stimulated Hepatic Stellate Cells. Hepatology 2019, 70, 1409–1423. [Google Scholar] [CrossRef] [Green Version]
  57. Basso, B.D.; Haute, G.V.; Ortega-Ribera, M.; Luft, C.; Antunes, G.L.; Bastos, M.S.; Carlessi, L.P.; Levorse, V.G.; Cassel, E.; Donadio, M.V.F.; et al. Methoxyeugenol deactivates hepatic stellate cells and attenuates liver fibrosis and inflammation through a PPAR-gamma and NF-kB mechanism. J. Ethnopharmacol. 2021, 280, 114433. [Google Scholar] [CrossRef]
  58. Campana, L.; Iredale, J.P. Regression of Liver Fibrosis. Semin. Liver Dis. 2017, 37, 1–10. [Google Scholar] [CrossRef]
  59. Zhang, Z.; Mu, Y.; Zhang, J.; Zhou, Y.; Cattaneo, P.; Veevers, J.; Peter, A.K.; Manso, A.M.; Knowlton, K.U.; Zhou, X.; et al. Kindlin-2 Is Essential for Preserving Integrity of the Developing Heart and Preventing Ventricular Rupture. Circulation 2019, 139, 1554–1556. [Google Scholar] [CrossRef]
  60. Yu, J.; Hu, Y.; Gao, Y.; Li, Q.; Zeng, Z.; Li, Y.; Chen, H. Kindlin-2 regulates hepatic stellate cells activation and liver fibrogenesis. Cell. Death Discov. 2018, 4, 34. [Google Scholar] [CrossRef]
  61. Heldin, C.H.; Westermark, B. Mechanism of Action and In Vivo Role of Platelet-Derived Growth Factor. Physiol. Rev. 1999, 79, 1283–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kocabayoglu, P.; Lade, A.; Lee, Y.A.; Dragomir, A.C.; Sun, X.; Fiel, M.I.; Thung, S.; Aloman, C.; Soriano, P.; Hoshida, Y.; et al. beta-PDGF receptor expressed by hepatic stellate cells regulates fibrosis in murine liver injury, but not carcinogenesis. J. Hepatol. 2015, 63, 141–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Hayes, B.J.; Riehle, K.J.; Shimizu-Albergine, M.; Bauer, R.L.; Hudkins, K.L.; Johansson, F.; Yeh, M.M.; Mahoney, W.M., Jr.; Yeung, R.S.; Campbell, J.S. Activation of platelet-derived growth factor receptor alpha contributes to liver fibrosis. PLoS ONE 2014, 9, e92925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ying, H.Z.; Chen, Q.; Zhang, W.Y.; Zhang, H.H.; Ma, Y.; Zhang, S.Z.; Fang, J.; Yu, C.H. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol. Med. Rep. 2017, 16, 7879–7889. [Google Scholar] [CrossRef] [Green Version]
  65. Tsuchida, T.; Friedman, S.L. Mechanisms of hepatic stellate cell activation. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 397–411. [Google Scholar] [CrossRef]
  66. Russell, J.O.; Camargo, F.D. Hippo signalling in the liver: Role in development, regeneration and disease. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 297–312. [Google Scholar] [CrossRef]
  67. Acharya, P.; Chouhan, K.; Weiskirchen, S.; Weiskirchen, R. Cellular Mechanisms of Liver Fibrosis. Front. Pharmacol. 2021, 12, 671640. [Google Scholar] [CrossRef]
  68. Miao, Y.; Wu, Y.L.; Jin, Y.J.; Lei, M.Z.; Nan, J.X.; Wu, X. Benzoquinone derivatives with antioxidant activity inhibit activated hepatic stellate cells and attenuate liver fibrosis in TAA-induced mice. Chem.-Biol. Interact. 2020, 317, 108945. [Google Scholar] [CrossRef]
  69. Zheng, M.; Li, Y.Y.; Wang, G.F.; Jin, J.Y.; Wang, Y.H.; Wang, T.M.; Yang, L.; Liu, S.Y.; Wu, J.S.; Wang, Z.T.; et al. Protective effect of cultured bear bile powder against dimethylnitrosamine-induced hepatic fibrosis in rats. Biomed. Pharmacother. 2019, 112, 108701. [Google Scholar] [CrossRef]
  70. Cordero-Espinoza, L.; Huch, M. The balancing act of the liver: Tissue regeneration versus fibrosis. J. Clin. Investig. 2018, 128, 85–96. [Google Scholar] [CrossRef] [Green Version]
  71. Jun, J.I.; Lau, L.F. Resolution of organ fibrosis. J. Clin. Investig. 2018, 128, 97–107. [Google Scholar] [CrossRef] [Green Version]
  72. Krenkel, O.; Puengel, T.; Govaere, O.; Abdallah, A.T.; Mossanen, J.C.; Kohlhepp, M.; Liepelt, A.; Lefebvre, E.; Luedde, T.; Hellerbrand, C.; et al. Therapeutic inhibition of inflammatory monocyte recruitment reduces steatohepatitis and liver fibrosis. Hepatology 2018, 67, 1270–1283. [Google Scholar] [CrossRef] [Green Version]
  73. Karsdal, M.A.; Nielsen, S.H.; Leeming, D.J.; Langholm, L.L.; Nielsen, M.J.; Manon-Jensen, T.; Siebuhr, A.; Gudmann, N.S.; Ronnow, S.; Sand, J.M.; et al. The good and the bad collagens of fibrosis—Their role in signaling and organ function. Adv. Drug Deliv. Rev. 2017, 121, 43–56. [Google Scholar] [CrossRef]
  74. Lee, Y.A.; Wallace, M.C.; Friedman, S.L. Pathobiology of liver fibrosis: A translational success story. Gut 2015, 64, 830–841. [Google Scholar] [CrossRef] [Green Version]
  75. Issa, R.; Williams, E.; Trim, N.; Kendall, T.; Arthur, M.J.P.; Reichen, J.; Benyon, R.C.; Iredale, J.P. Apoptosis of hepatic stellate cells: Involvement in resolution of biliary fibrosis and regulation by soluble growth factors. Gut 2001, 48, 548–557. [Google Scholar] [CrossRef] [Green Version]
  76. Kisseleva, T.; Cong, M.; Paik, Y.; Scholten, D.; Jiang, C.; Benner, C.; Iwaisako, K.; Moore-Morris, T.; Scott, B.; Tsukamoto, H.; et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc. Natl. Acad. Sci. USA 2012, 109, 9448–9453. [Google Scholar] [CrossRef] [Green Version]
  77. El Taghdouini, A.; Najimi, M.; Sancho-Bru, P.; Sokal, E.; van Grunsven, L.A. In vitro reversion of activated primary human hepatic stellate cells. Fibrogenesis Tissue Repair. 2015, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  78. Nishizawa, H.; Iguchi, G.; Fukuoka, H.; Takahashi, M.; Suda, K.; Bando, H.; Matsumoto, R.; Yoshida, K.; Odake, Y.; Ogawa, W.; et al. IGF-I induces senescence of hepatic stellate cells and limits fibrosis in a p53-dependent manner. Sci. Rep. 2016, 6, 34605. [Google Scholar] [CrossRef] [Green Version]
  79. Tan, S.W.; Liu, H.L.; Ke, B.L.; Jiang, J.; Wu, B. The peripheral CB1 receptor antagonist JD5037 attenuates liver fibrosis via a CB1 receptor/beta-arrestin1/Akt pathway. Br. J. Pharmacol. 2020, 177, 2830–2847. [Google Scholar] [CrossRef]
  80. Zhang, Y.Q.; Hua, L.P.; Lin, C.F.; Yuan, M.Z.; Xu, W.; Raj, A.D.; Venkidasamy, B.; Cespedes-Acuna, C.L.; Nile, S.H.; Yan, G.H.; et al. Pien-Tze-Huang alleviates CCl4-induced liver fibrosis through the inhibition of HSC autophagy and the TGF-beta 1/Smad2 pathway. Front. Pharmacol. 2022, 13, 937484. [Google Scholar] [CrossRef]
  81. Higashi, T.; Friedman, S.L.; Hoshida, Y. Hepatic stellate cells as key target in liver fibrosis. Adv. Drug Deliv. Rev. 2017, 121, 27–42. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, R.; Liu, F.; Chen, P.; Li, S.; Gu, Y.; Wang, L.; Chen, C.; Yuan, Y. Gomisin D alleviates liver fibrosis through targeting PDGFRbeta in hepatic stellate cells. Int. J. Biol. Macromol. 2023, 235, 123639. [Google Scholar] [CrossRef] [PubMed]
  83. Arroyo, N.; Villamayor, L.; Diaz, I.; Carmona, R.; Ramos-Rodriguez, M.; Munoz-Chapuli, R.; Pasquali, L.; Toscano, M.G.; Martin, F.; Cano, D.A.; et al. GATA4 induces liver fibrosis regression by deactivating hepatic stellate cells. JCI Insight 2021, 6, e150059. [Google Scholar] [CrossRef]
  84. Jelski, W.; Szmitkowski, M. Alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) in the cancer diseases. Clin. Chim. Acta 2008, 395, 1–5. [Google Scholar] [CrossRef] [PubMed]
  85. Gao, N.; Li, J.; Li, M.R.; Qi, B.; Wang, Z.; Wang, G.J.; Gao, J.; Qiao, H.L. Higher Activity of Alcohol Dehydrogenase Is Correlated with Hepatic Fibrogenesis. J. Pharmacol. Exp. Ther. 2018, 367, 473–482. [Google Scholar] [CrossRef]
  86. Zhao, T.; Han, Z.; Zhang, J.; Ding, Y.; Chen, J.; Qiao, H.; Gao, N. Effect of ADHI on hepatic stellate cell activation and liver fibrosis in mice. Biochem. Biophys. Res. Commun. 2023, 651, 98–106. [Google Scholar] [CrossRef]
  87. Wu, B.; Huang, L.; Wang, Y.; Zeng, L.; Lin, Y.; Li, J.; Wang, S.; Zhang, G.; An, L. Yao medicine Amydrium hainanense suppresses hepatic fibrosis by repressing hepatic stellate cell activation via STAT3 signaling. Front. Pharmacol. 2022, 13, 1043022. [Google Scholar] [CrossRef]
  88. Meng, H.; Jiang, L.; Jia, P.; Niu, R.; Bu, F.; Zhu, Y.; Pan, X.; Li, J.; Liu, J.; Zhang, Y.; et al. Inhibition of circular RNA ASPH reduces the proliferation and promotes the apoptosis of hepatic stellate cells in hepatic fibrosis. Biochem. Pharmacol. 2023, 210, 115451. [Google Scholar] [CrossRef]
  89. Ma, L.; Wei, J.F.; Zeng, Y.L.; Liu, J.P.; Xiao, E.H.; Kang, Y.H.; Kang, Y. Mesenchymal stem cell-originated exosomal circDIDO1 suppresses hepatic stellate cell activation by miR-141-3p/PTEN/AKT pathway in human liver fibrosis. Drug. Deliv. 2022, 29, 440–453. [Google Scholar] [CrossRef]
  90. An, P.; Wei, L.L.; Zhao, S.; Sverdlov, D.Y.; Vaid, K.A.; Miyamoto, M.; Kuramitsu, K.; Lai, M.; Popov, Y.V. Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nat. Commun. 2020, 11, 2362. [Google Scholar] [CrossRef]
  91. Kong, Z.; Liu, R.; Cheng, Y. Artesunate alleviates liver fibrosis by regulating ferroptosis signaling pathway. Biomed. Pharmacother. 2019, 109, 2043–2053. [Google Scholar] [CrossRef]
  92. Gonzalez, S.A.; Fiel, M.I.; Sauk, J.; Canchis, P.W.; Liu, R.C.; Chiriboga, L.; Yee, H.T.; Jacobson, I.M.; Talal, A.H. Inverse association between hepatic stellate cell apoptosis and fibrosis in chronic hepatitis C virus infection. J. Viral Hepat. 2009, 16, 141–148. [Google Scholar] [CrossRef]
  93. Koda, Y.; Teratani, T.; Chu, P.S.; Hagihara, Y.; Mikami, Y.; Harada, Y.; Tsujikawa, H.; Miyamoto, K.; Suzuki, T.; Taniki, N.; et al. CD8(+) tissue-resident memory T cells promote liver fibrosis resolution by inducing apoptosis of hepatic stellate cells. Nat. Commun. 2021, 12, 4474. [Google Scholar] [CrossRef]
  94. Xu, T.; Ni, M.M.; Xing, L.; Li, X.F.; Meng, X.M.; Huang, C.; Li, J. NLRC5 regulates TGF-beta1-induced proliferation and activation of hepatic stellate cells during hepatic fibrosis. Int. J. Biochem. Cell. Biol. 2016, 70, 92–104. [Google Scholar] [CrossRef]
  95. Liu, H.; Dai, L.; Wang, M.; Feng, F.; Xiao, Y. Tunicamycin Induces Hepatic Stellate Cell Apoptosis Through Calpain-2/Ca(2+)-Dependent Endoplasmic Reticulum Stress Pathway. Front. Cell. Dev. Biol. 2021, 9, 684857. [Google Scholar] [CrossRef]
  96. Lee, S.W.; Kim, S.M.; Hur, W.; Kang, B.Y.; Lee, H.L.; Nam, H.; Yoo, S.H.; Sung, P.S.; Kwon, J.H.; Jang, J.W.; et al. Tenofovir disoproxil fumarate directly ameliorates liver fibrosis by inducing hepatic stellate cell apoptosis via downregulation of PI3K/Akt/mTOR signaling pathway. PLoS ONE 2021, 16, e0261067. [Google Scholar] [CrossRef]
  97. Liu, X.; Xu, J.; Rosenthal, S.; Zhang, L.J.; McCubbin, R.; Meshgin, N.; Shang, L.; Koyama, Y.; Ma, H.Y.; Sharma, S.; et al. Identification of Lineage-Specific Transcription Factors That Prevent Activation of Hepatic Stellate Cells and Promote Fibrosis Resolution. Gastroenterology 2020, 158, 1728–1744.e1714. [Google Scholar] [CrossRef] [Green Version]
  98. Kim, N.; Choi, S.; Lim, C.; Lee, H.; Oh, J. Albumin mediates PPAR-gamma or C/EBP-alpha-induced phenotypic changes in pancreatic stellate cells. Biochem. Biophys. Res. Commun. 2010, 391, 640–644. [Google Scholar] [CrossRef]
  99. Hazra, S.; Xiong, S.; Wang, J.; Rippe, R.A.; Krishna, V.; Chatterjee, K.; Tsukamoto, H. Peroxisome proliferator-activated receptor gamma induces a phenotypic switch from activated to quiescent hepatic stellate cells. J. Biol. Chem. 2004, 279, 11392–11401. [Google Scholar] [CrossRef] [Green Version]
  100. Miyahara, T.; Schrum, L.; Rippe, R.; Xiong, S.; Yee, H.F., Jr.; Motomura, K.; Anania, F.A.; Willson, T.M.; Tsukamoto, H. Peroxisome proliferator-activated receptors and hepatic stellate cell activation. J. Biol. Chem. 2000, 275, 35715–35722. [Google Scholar] [CrossRef] [Green Version]
  101. Nakano, Y.; Kamiya, A.; Sumiyoshi, H.; Tsuruya, K.; Kagawa, T.; Inagaki, Y. A Deactivation Factor of Fibrogenic Hepatic Stellate Cells Induces Regression of Liver Fibrosis in Mice. Hepatology 2020, 71, 1437–1452. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, Y.H.; Suk, F.M.; Liu, C.L.; Chen, T.L.; Twu, Y.C.; Hsu, M.H.; Liao, Y.J. Antifibrotic Effects of a Barbituric Acid Derivative on Liver Fibrosis by Blocking the NF-kappa B Signaling Pathway in Hepatic Stellate Cells. Front. Pharmacol. 2020, 11, 388. [Google Scholar] [CrossRef] [Green Version]
  103. Xu, F.; Liu, C.; Zhou, D.; Zhang, L. TGF-beta/SMAD Pathway and Its Regulation in Hepatic Fibrosis. J. Histochem. Cytochem. 2016, 64, 157–167. [Google Scholar] [CrossRef] [Green Version]
  104. Nishikawa, K.; Osawa, Y.; Kimura, K. Wnt/beta-Catenin Signaling as a Potential Target for the Treatment of Liver Cirrhosis Using Antifibrotic Drugs. Int. J. Mol. Sci. 2018, 19, 3103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Bansal, R.; van Baarlen, J.; Storm, G.; Prakash, J. The interplay of the Notch signaling in hepatic stellate cells and macrophages determines the fate of liver fibrogenesis. Sci. Rep. 2015, 5, 18272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Xie, G.; Karaca, G.; Swiderska-Syn, M.; Michelotti, G.A.; Kruger, L.; Chen, Y.; Premont, R.T.; Choi, S.S.; Diehl, A.M. Cross-talk between Notch and Hedgehog regulates hepatic stellate cell fate in mice. Hepatology 2013, 58, 1801–1813. [Google Scholar] [CrossRef] [Green Version]
  107. Mannaerts, I.; Leite, S.B.; Verhulst, S.; Claerhout, S.; Eysackers, N.; Thoen, L.F.; Hoorens, A.; Reynaert, H.; Halder, G.; van Grunsven, L.A. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J. Hepatol. 2015, 63, 679–688. [Google Scholar] [CrossRef]
  108. Tao, Y.; Wang, N.; Qiu, T.; Sun, X. The Role of Autophagy and NLRP3 Inflammasome in Liver Fibrosis. Biomed. Res. Int. 2020, 2020, 7269150. [Google Scholar] [CrossRef]
  109. Song, Y.R.; Jang, M.H.; Jang, B.; Bae, S.J.; Bak, S.B.; Lee, S.M.; Yun, U.-J.; Lee, J.H.; Park, S.M.; Jung, D.H.; et al. Jageum-Jung, the herbal pharmaceuticals, inhibits the hepatic fibrogenesis as mediated with TGF-β1/smad signaling. Mol. Cell. Toxicol. 2022, 18, 243–251. [Google Scholar] [CrossRef]
  110. Dewidar, B.; Soukupova, J.; Fabregat, I.; Dooley, S. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis: Updated. Curr. Pathobiol. Rep. 2015, 3, 291–305. [Google Scholar] [CrossRef] [Green Version]
  111. Ge, M.X.; Liu, H.T.; Zhang, N.; Niu, W.X.; Lu, Z.N.; Bao, Y.Y.; Huang, R.; Yu, D.K.; Shao, R.G.; He, H.W. Costunolide represses hepatic fibrosis through WW domain-containing protein 2-mediated Notch3 degradation. Br. J. Pharmacol. 2020, 177, 372–387. [Google Scholar] [CrossRef]
  112. Cao, F.Y.; Zhang, Y.F.; Li, W.G.; Shimizu, K.S.; Xie, H.F.; Zhang, C.F. Mogroside IVE attenuates experimental liver fibrosis in mice and inhibits HSC activation through downregulating TLR4-mediated pathways. Int. Immunopharmacol. 2018, 55, 183–192. [Google Scholar] [CrossRef]
  113. Zhao, Y.; Su, X. Antibacterial activity of 18beta-glycyrrhetinic acid against Neisseria gonorrhoeae in vitro. Biochem. Biophys. Rep. 2023, 33, 101427. [Google Scholar] [CrossRef]
  114. Kumar, A.; Archo, S.; Singh, C.P.; Naikoo, S.H.; Singh, B.; Kaur, S.; Tasduq, S.A. Photoprotective effect of 18beta-glycyrrhetinic acid derivatives against ultra violet (UV)-B-Induced skin aging. Bioorg Med. Chem. Lett. 2022, 76, 128984. [Google Scholar] [CrossRef]
  115. Du, Y.C.; Lai, L.; Zhang, H.; Zhong, F.R.; Cheng, H.L.; Qian, B.L.; Tan, P.; Xia, X.M.; Fu, W.G. Kaempferol from Penthorum chinense Pursh suppresses HMGB1/TLR4/NF-kappaB signaling and NLRP3 inflammasome activation in acetaminophen-induced hepatotoxicity. Food Funct. 2020, 11, 7925–7934. [Google Scholar] [CrossRef]
  116. Xiao, X.L.; Hu, Q.C.; Deng, X.Y.; Shi, K.Y.; Zhang, W.W.; Jiang, Y.X.; Ma, X.; Zeng, J.H.; Wang, X.Y. Old wine in new bottles: Kaempferol is a promising agent for treating the trilogy of liver diseases. Pharmacol. Res. 2022, 175, 106005. [Google Scholar] [CrossRef]
  117. Xu, T.; Huang, S.; Huang, Q.; Ming, Z.; Wang, M.; Li, R.; Zhao, Y. Kaempferol attenuates liver fibrosis by inhibiting activin receptor-like kinase 5. J. Cell. Mol. Med. 2019, 23, 6403–6410. [Google Scholar] [CrossRef]
  118. Zhou, G.; Li, C.; Zhang, R.; Zhan, Y.; Lin, L.; Lang, Z.; Tao, Q.; Zheng, J. Kaempferol Inhibits Hepatic Stellate Cell Activation by Regulating miR-26b-5p/Jag1 Axis and Notch Pathway. Front. Pharmacol. 2022, 13, 881855. [Google Scholar] [CrossRef]
  119. Wei, Z.; Xue, Y.; Xue, Y.; Cheng, J.; Lv, G.; Chu, L.; Ma, Z.; Guan, S. Ferulic acid attenuates non-alcoholic steatohepatitis by reducing oxidative stress and inflammation through inhibition of the ROCK/NF-kappaB signaling pathways. J. Pharmacol. Sci. 2021, 147, 72–80. [Google Scholar] [CrossRef]
  120. Mu, M.; Zuo, S.; Wu, R.M.; Deng, K.S.; Lu, S.; Zhu, J.J.; Zou, G.L.; Yang, J.; Cheng, M.L.; Zhao, X.K. Ferulic acid attenuates liver fibrosis and hepatic stellate cell activation via inhibition of TGF-beta/Smad signaling pathway. Drug. Des. Dev. Ther. 2018, 12, 4107–4115. [Google Scholar] [CrossRef] [Green Version]
  121. Chen, K.; Guo, W.R.; Li, R.X.; Han, Y.Q.; Gao, Q.; Wang, S.Z. Demethylzeylasteral attenuates hepatic stellate cell activation and liver fibrosis by inhibiting AGAP2 mediated signaling. Phytomedicine 2022, 105, 154349. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, J.L.; Yang, A.H.; Wu, Y.; Guan, W.; Xiong, B.; Peng, X.Q.; Wei, X.J.; Chen, C.; Liu, Z.G. Stachydrine ameliorates carbon tetrachloride-induced hepatic fibrosis by inhibiting inflammation, oxidative stress and regulating MMPs/TIMPs system in rats. Biomed. Pharmacother. 2018, 97, 1586–1594. [Google Scholar] [CrossRef] [PubMed]
  123. Kodai, S.; Takemura, S.; Kubo, S.; Azuma, H.; Minamiyama, Y. Therapeutic administration of an ingredient of aged-garlic extracts, S-allyl cysteine resolves liver fibrosis established by carbon tetrachloride in rats. J. Clin. Biochem. Nutr. 2015, 56, 179–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wu, L.; Zhang, Q.; Mo, W.; Feng, J.; Li, S.; Li, J.; Liu, T.; Xu, S.; Wang, W.; Lu, X.; et al. Quercetin prevents hepatic fibrosis by inhibiting hepatic stellate cell activation and reducing autophagy via the TGF-β1/Smads and PI3K/Akt pathways. Sci. Rep. 2017, 7, 9289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lin, X.; Wei, Y.; Li, Y.; Xiong, Y.; Fang, B.; Li, C.; Huang, Q.; Huang, R.; Wei, J. Tormentic Acid Ameliorates Hepatic Fibrosis in vivo by Inhibiting Glycerophospholipids Metabolism and PI3K/Akt/mTOR and NF-kappaB Pathways: Based on Transcriptomics and Metabolomics. Front. Pharmacol. 2022, 13, 801982. [Google Scholar] [CrossRef]
  126. Tong, M.; Zheng, Q.; Liu, M.; Chen, L.; Lin, Y.H.; Tang, S.G.; Zhu, Y.M. 5-methoxytryptophan alleviates liver fibrosis by modulating FOXO3a/miR-21/ATG5 signaling pathway mediated autophagy. Cell. Cycle 2021, 20, 676–688. [Google Scholar] [CrossRef]
  127. Zhu, X.Y.; Ye, S.T.; Yu, D.K.; Zhang, Y.Q.; Li, J.; Zhang, M.H.; Leng, Y.R.; Yang, T.; Luo, J.G.; Chen, X.L.; et al. Physalin B attenuates liver fibrosis via suppressing LAP2 alpha-HDAC1-mediated deacetylation of the transcription factor GLI1 and hepatic stellate cell activation. Br. J. Pharmacol. 2021, 178, 3428–3447. [Google Scholar] [CrossRef]
  128. Mo, C.; Xie, S.W.; Zeng, T.; Lai, Y.Q.; Huang, S.; Zhou, C.Y.; Yan, W.X.; Huang, S.H.; Gao, L.; Lv, Z.P. Ginsenoside-Rg1 acts as an IDO1 inhibitor, protects against liver fibrosis via alleviating IDO1-mediated the inhibition of DCs maturation. Phytomedicine 2021, 84, 153524. [Google Scholar] [CrossRef]
  129. Li, Z.Y.; Wang, Z.L.; Dong, F.; Shi, W.; Dai, W.Z.; Zhao, J.; Li, Q.; Fang, Z.E.; Ren, L.T.; Liu, T.T.; et al. Germacrone Attenuates Hepatic Stellate Cells Activation and Liver Fibrosis via Regulating Multiple Signaling Pathways. Front. Pharmacol. 2021, 12, 745561. [Google Scholar] [CrossRef]
  130. Gong, J.T.; Yang, F.; Yang, Q.L.; Tang, X.W.; Shu, F.F.; Xu, L.M.; Wang, Z.T.; Yang, L. Sweroside ameliorated carbon tetrachloride (CCl4)-induced liver fibrosis through FXR-miR-29a signaling pathway. J. Nat. Med. 2020, 74, 17–25. [Google Scholar] [CrossRef]
  131. Zhang, Y.; Cai, B.; Li, Y.; Xu, Y.; Wang, Y.; Zheng, L.; Zheng, X.; Yin, L.; Chen, G.; Wang, Y.; et al. Identification of linderalactone as a natural inhibitor of SHP2 to ameliorate CCl(4)-induced liver fibrosis. Front. Pharmacol. 2023, 14, 1098463. [Google Scholar] [CrossRef]
  132. Wei, S.; Niu, M.; Wang, J.; Wang, J.; Su, H.; Luo, S.; Zhang, X.; Guo, Y.; Liu, L.; Liu, F.; et al. A network pharmacology approach to discover active compounds and action mechanisms of San-Cao Granule for treatment of liver fibrosis. Drug. Des. Devel Ther. 2016, 10, 733–743. [Google Scholar] [CrossRef] [Green Version]
  133. Chang, Q.Q.; Pan, Y.F.; Yang, J.Y.; Li, R.S.; Yuan, C.L.; Liao, Y.F.; Zhang, D.D.; Liu, C.H. Antiliver Fibrosis Formula of Fuzheng Huayu Alleviates Inflammatory Response. Evid. Based Complement. Altern. Med. 2022, 2022, 5752803. [Google Scholar] [CrossRef]
  134. Chen, J.; Hu, Y.; Chen, L.; Liu, W.; Mu, Y.; Liu, P. The effect and mechanisms of Fuzheng Huayu formula against chronic liver diseases. Biomed. Pharmacother. 2019, 114, 108846. [Google Scholar] [CrossRef]
  135. Hu, X.Q.; Song, Y.N.; Wu, R.; Cai, F.F.; Zhang, Y.; Peng, J.H.; Hu, Y.Y.; Su, S.B. Metabolic mechanisms of Fuzheng-Huayu formula against liver fibrosis in rats. J. Ethnopharmacol. 2019, 238, 111888. [Google Scholar] [CrossRef]
  136. Li, X.M.; Peng, J.H.; Sun, Z.L.; Tian, H.J.; Duan, X.H.; Liu, L.; Ma, X.; Feng, Q.; Liu, P.; Hu, Y.Y. Chinese medicine CGA formula ameliorates DMN-induced liver fibrosis in rats via inhibiting MMP2/9, TIMP1/2 and the TGF-beta/Smad signaling pathways. Acta Pharmacol. Sin. 2016, 37, 783–793. [Google Scholar] [CrossRef] [Green Version]
  137. Fu, Y.D.; Xiao, Z.; Tian, X.T.; Liu, W.; Xu, Z.; Yang, T.; Hu, Y.H.; Zhou, X.X.; Fang, J.; Gao, S.Q.; et al. The Novel Chinese Medicine JY5 Formula Alleviates Hepatic Fibrosis by Inhibiting the Notch Signaling Pathway. Front. Pharmacol. 2021, 12, 671152. [Google Scholar] [CrossRef]
  138. Yang, Y.; Sun, M.Y.; Li, W.D.; Liu, C.B.; Jiang, Z.S.; Gu, P.F.; Li, J.Q.; Wang, W.; You, R.L.; Ba, Q.; et al. Rebalancing TGF-beta/Smad7 signaling via Compound kushen injection in hepatic stellate cells protects against liver fibrosis and hepatocarcinogenesis. Clin. Transl. Med. 2021, 11, e410. [Google Scholar] [CrossRef]
  139. Gong, Z.; Lin, J.; Zheng, J.; Wei, L.; Liu, L.; Peng, Y.; Liang, W.; Hu, G. Dahuang Zhechong pill attenuates CCl4-induced rat liver fibrosis via the PI3K-Akt signaling pathway. J. Cell. Biochem. 2020, 121, 1431–1440. [Google Scholar] [CrossRef]
  140. Ke, C.; Gao, J.L.; Tu, J.Y.; Wang, Y.; Xiao, Y.X.; Wu, Y.; Liu, Y.J.; Zhou, Z.S. Ganfule capsule alleviates bile duct ligation-induced liver fibrosis in mice by inhibiting glutamine metabolism. Front. Pharmacol. 2022, 13, 930785. [Google Scholar] [CrossRef]
  141. Bai, T.; Lian, L.H.; Wu, Y.L.; Wan, Y.; Nan, J.X. Thymoquinone attenuates liver fibrosis via PI3K and TLR4 signaling pathways in activated hepatic stellate cells. Int. Immunopharmacol. 2013, 15, 275–281. [Google Scholar] [CrossRef] [PubMed]
  142. Son, M.K.; Ryu, Y.L.; Jung, K.H.; Lee, H.; Lee, H.S.; Yan, H.H.; Park, H.J.; Ryu, J.K.; Suh, J.K.; Hong, S.; et al. HS-173, a novel PI3K inhibitor, attenuates the activation of hepatic stellate cells in liver fibrosis. Sci. Rep. 2013, 3, 3470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Liao, Y.J.; Hsu, S.M.; Chien, C.Y.; Wang, Y.H.; Hsu, M.H.; Suk, F.M. Treatment with a New Barbituric Acid Derivative Exerts Antiproliferative and Antimigratory Effects against Sorafenib Resistance in Hepatocellular Carcinoma. Molecules 2020, 25, 2856. [Google Scholar] [CrossRef] [PubMed]
  144. Sokmen, B.B.; Ugras, S.; Sarikaya, H.Y.; Ugras, H.I.; Yanardag, R. Antibacterial, antiurease, and antioxidant activities of some arylidene barbiturates. Appl. Biochem. Biotechnol. 2013, 171, 2030–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zheng, H.; Li, S.; Ma, L.; Cheng, L.; Deng, C.; Chen, Z.; Xie, C.; Xiang, M.; Jiang, W.; Chen, L. A novel agonist of PPAR-gamma based on barbituric acid alleviates the development of non-alcoholic fatty liver disease by regulating adipocytokine expression and preventing insulin resistance. Eur. J. Pharmacol. 2011, 659, 244–251. [Google Scholar] [CrossRef]
  146. Shen, D.F.; Cheng, H.; Cai, B.Z.; Cai, W.F.; Wang, B.; Zhu, Q.; Wu, Y.B.; Liu, M.; Chen, R.J.; Gao, F.F.; et al. N-n-Butyl haloperidol iodide ameliorates liver fibrosis and hepatic stellate cell activation in mice. Acta Pharmacol. Sin. 2022, 43, 133–145. [Google Scholar] [CrossRef]
  147. Jiao, W.X.; Bai, M.; Yin, H.W.; Liu, J.Y.; Sun, J.; Su, X.X.; Zeng, H.H.; Wen, J.H. Therapeutic Effects of an Inhibitor of Thioredoxin Reductase on Liver Fibrosis by Inhibiting the Transforming Growth Factor-beta 1/Smads Pathway. Front. Mol. Biosci. 2021, 8, 690170. [Google Scholar] [CrossRef]
  148. Kong, L.J.; Li, H.; Du, Y.J.; Pei, F.H.; Hu, Y.; Zhao, L.L.; Chen, J. Vatalanib, a tyrosine kinase inhibitor, decreases hepatic fibrosis and sinusoidal capillarization in CCl4-induced fibrotic mice. Mol. Med. Rep. 2017, 15, 2604–2610. [Google Scholar] [CrossRef] [Green Version]
  149. Chen, J.; Liu, D.G.; Yang, G.; Kong, L.J.; Du, Y.J.; Wang, H.Y.; Li, F.D.; Pei, F.H.; Song, J.T.; Fan, Y.J.; et al. Endostar, a novel human recombinant endostatin, attenuates liver fibrosis in CCl4-induced mice. Exp. Biol. Med. (Maywood) 2014, 239, 998–1006. [Google Scholar] [CrossRef]
  150. Song, Z.H.; Liu, X.H.; Zhang, W.; Luo, Y.; Xiao, H.; Liu, Y.; Dai, G.Q.; Hong, J.; Li, A.M. Ruxolitinib suppresses liver fibrosis progression and accelerates fibrosis reversal via selectively targeting Janus kinase 1/2. J. Transl. Med. 2022, 20, 157. [Google Scholar] [CrossRef]
  151. Lu, Z.N.; Niu, W.X.; Zhang, N.; Ge, M.X.; Bao, Y.Y.; Ren, Y.; Guo, X.L.; He, H.W. Pantoprazole ameliorates liver fibrosis and suppresses hepatic stellate cell activation in bile duct ligation rats by promoting YAP degradation. Acta Pharmacol. Sin. 2021, 42, 1808–1820. [Google Scholar] [CrossRef]
  152. Yoon, Y.C.; Fang, Z.H.; Lee, J.E.; Park, J.H.; Ryu, J.K.; Jung, K.H.; Hong, S.S. Selonsertib Inhibits Liver Fibrosis via Downregulation of ASK1/MAPK Pathway of Hepatic Stellate Cells. Biomol. Ther. 2020, 28, 527–536. [Google Scholar] [CrossRef]
  153. Marti-Rodrigo, A.; Alegre, F.; Moragrega, A.B.; Garcia-Garcia, F.; Marti-Rodrigo, P.; Fernandez-Iglesias, A.; Gracia-Sancho, J.; Apostolova, N.; Esplugues, J.V.; Blas-Garcia, A. Rilpivirine attenuates liver fibrosis through selective STAT1-mediated apoptosis in hepatic stellate cells. Gut 2020, 69, 920–932. [Google Scholar] [CrossRef]
  154. Bisht, S.; Khan, M.A.; Bekhit, M.; Bai, H.; Cornish, T.; Mizuma, M.; Rudek, M.A.; Zhao, M.; Maitra, A.; Ray, B.; et al. A polymeric nanoparticle formulation of curcumin (NanoCurc) ameliorates CCl4-induced hepatic injury and fibrosis through reduction of pro-inflammatory cytokines and stellate cell activation. Lab. Investig. 2011, 91, 1383–1395. [Google Scholar] [CrossRef] [Green Version]
  155. Shen, J.; Cao, J.; Chen, M.; Zhang, Y. Recent advances in the role of exosomes in liver fibrosis. J. Gastroenterol. Hepatol. 2023. [Google Scholar] [CrossRef]
  156. Wang, N.; Li, X.; Zhong, Z.; Qiu, Y.; Liu, S.; Wu, H.; Tang, X.; Chen, C.; Fu, Y.; Chen, Q.; et al. 3D hESC exosomes enriched with miR-6766-3p ameliorates liver fibrosis by attenuating activated stellate cells through targeting the TGFbetaRII-SMADS pathway. J. Nanobiotechnol. 2021, 19, 437. [Google Scholar] [CrossRef]
  157. Tang, M.; Guo, C.; Sun, M.; Zhou, H.; Peng, X.; Dai, J.; Ding, Q.; Wang, Y.; Yang, C. Effective delivery of osteopontin small interference RNA using exosomes suppresses liver fibrosis via TGF-beta1 signaling. Front. Pharmacol. 2022, 13, 882243. [Google Scholar] [CrossRef]
  158. Song, Z.; Chen, W.; Athavale, D.; Ge, X.; Desert, R.; Das, S.; Han, H.; Nieto, N. Osteopontin Takes Center Stage in Chronic Liver Disease. Hepatology 2021, 73, 1594–1608. [Google Scholar] [CrossRef]
  159. Poilil Surendran, S.; George Thomas, R.; Moon, M.J.; Jeong, Y.Y. Nanoparticles for the treatment of liver fibrosis. Int. J. Nanomed. 2017, 12, 6997–7006. [Google Scholar] [CrossRef] [Green Version]
  160. Petros, R.A.; DeSimone, J.M. Strategies in the design of nanoparticles for therapeutic applications. Nat. Rev. Drug. Discov. 2010, 9, 615–627. [Google Scholar] [CrossRef]
  161. Wang, M.; Zhang, M.; Fu, L.H.; Lin, J.; Zhou, X.M.; Zhou, P.H.; Huang, P.; Hu, H.; Han, Y. Liver-targeted delivery of TSG-6 by calcium phosphate nanoparticles for the management of liver fibrosis. Theranostics 2020, 10, 36–49. [Google Scholar] [CrossRef] [PubMed]
  162. Salunkhe, S.A.; Chitkara, D.; Mahato, R.I.; Mittal, A. Lipid based nanocarriers for effective drug delivery and treatment of diabetes associated liver fibrosis. Adv. Drug Deliv. Rev. 2021, 173, 394–415. [Google Scholar] [CrossRef] [PubMed]
  163. Lee, J.; Byun, J.; Shim, G.; Oh, Y.K. Fibroblast activation protein activated antifibrotic peptide delivery attenuates fibrosis in mouse models of liver fibrosis. Nat. Commun. 2022, 13, 1516. [Google Scholar] [CrossRef] [PubMed]
  164. Schuppan, D.; Ashfaq-Khan, M.; Yang, A.T.; Kim, Y.O. Liver fibrosis: Direct antifibrotic agents and targeted therapies. Matrix Biol. 2018, 68–69, 435–451. [Google Scholar] [CrossRef]
  165. Okimoto, S.; Kuroda, S.; Tashiro, H.; Kobayashi, T.; Taogoshi, T.; Matsuo, H.; Ohdan, H. Vitamin A-coupled liposomal Rho-kinase inhibitor ameliorates liver fibrosis without systemic adverse effects. Hepatol. Res. 2019, 49, 663–675. [Google Scholar] [CrossRef] [Green Version]
  166. Zhang, J.; Li, R.; Liu, Q.; Zhou, J.; Huang, H.; Huang, Y.; Zhang, Z.; Wu, T.; Tang, Q.; Huang, C.; et al. SB431542-Loaded Liposomes Alleviate Liver Fibrosis by Suppressing TGF-beta Signaling. Mol. Pharm. 2020, 17, 4152–4162. [Google Scholar] [CrossRef]
Figure 1. Hepatic stellate cells in the progression and regression of liver fibrosis. Quiescent hepatic stellate cells are activated when the liver is damaged. Many cells, including hepatocytes, liver sinusoidal endothelial cells, macrophages, TGF-β, and PDGF signal pathways, are involved in this process, which leads to the proliferation of aHSCs, and excessive secretion of ECM proteins, and eventually cirrhosis. Inhibition of HSC proliferation, induction of HSC death, or inactivation of HSCs can reverse hepatic fibrosis. qHSCs: quiescent hepatic stellate cells. TGF-β: transforming growth factor-β. PDGF: platelet-derived growth factor. DAMPs: damage-associated molecular patterns. ROS: reactive oxygen species. aHSCs: activated hepatic stellate cells. LSEC: liver sinusoidal endothelial cell. MMPs: matrix metalloproteinases. TIMPs: tissue inhibitors of matrix metalloproteinases. ECM: extracellular matrix. iHSC: inactivated hepatic stellate cell.
Figure 1. Hepatic stellate cells in the progression and regression of liver fibrosis. Quiescent hepatic stellate cells are activated when the liver is damaged. Many cells, including hepatocytes, liver sinusoidal endothelial cells, macrophages, TGF-β, and PDGF signal pathways, are involved in this process, which leads to the proliferation of aHSCs, and excessive secretion of ECM proteins, and eventually cirrhosis. Inhibition of HSC proliferation, induction of HSC death, or inactivation of HSCs can reverse hepatic fibrosis. qHSCs: quiescent hepatic stellate cells. TGF-β: transforming growth factor-β. PDGF: platelet-derived growth factor. DAMPs: damage-associated molecular patterns. ROS: reactive oxygen species. aHSCs: activated hepatic stellate cells. LSEC: liver sinusoidal endothelial cell. MMPs: matrix metalloproteinases. TIMPs: tissue inhibitors of matrix metalloproteinases. ECM: extracellular matrix. iHSC: inactivated hepatic stellate cell.
Ijms 24 09671 g001
Figure 2. Therapeutic compounds for liver fibrosis resolution. Inhibition of HSC proliferation, induction of HSC death, or inactivation of HSCs and inhibition of liver fibrosis-related signal pathways can reverse liver fibrosis. Various compounds, including natural and synthetic compounds, have been found to have therapeutic effects in various animal models of liver fibrosis. Meanwhile, in order to improve the utilization rate of therapeutic drugs, a variety of nano-drug delivery systems are also used in the treatment of liver fibrosis. aHSC: activated hepatic stellate cell. HSC: hepatic stellate cell. TGF-β: transforming growth factor-β. TAA: thioacetamide. CCl4: carbon tetrachloride. BDL: bile duct ligation. DEN: diethylnitrosamine. DMN: dimethylnitrosamine.
Figure 2. Therapeutic compounds for liver fibrosis resolution. Inhibition of HSC proliferation, induction of HSC death, or inactivation of HSCs and inhibition of liver fibrosis-related signal pathways can reverse liver fibrosis. Various compounds, including natural and synthetic compounds, have been found to have therapeutic effects in various animal models of liver fibrosis. Meanwhile, in order to improve the utilization rate of therapeutic drugs, a variety of nano-drug delivery systems are also used in the treatment of liver fibrosis. aHSC: activated hepatic stellate cell. HSC: hepatic stellate cell. TGF-β: transforming growth factor-β. TAA: thioacetamide. CCl4: carbon tetrachloride. BDL: bile duct ligation. DEN: diethylnitrosamine. DMN: dimethylnitrosamine.
Ijms 24 09671 g002
Table 2. Anti-fibrotic Chinese herbal formulations.
Table 2. Anti-fibrotic Chinese herbal formulations.
CompoundsAnimal ModelTargets/Pathways/MechanismsReference
Pien-Tze-Huang (PZH)CCl4-induced liver fibrosis in male SD ratsAutophagy and TGF-β1/Smad2 signaling pathways[80]
Inhibiting HSC activation and diminishing fibrogenesis
Compound Kushen injection (CKI)CCl4-induced chronic liver fibrosis in C57BL/6 miceInhibiting HSC activation[138]
MCD diet-induced NASH model in C57BL/6 miceRebalancing TGF-β/Smad7 signaling
CCl4-induced HCC models in C57BL/6 mice
JY5CCl4-induced liver fibrosis in male Wistar or SD ratsInhibition of the Notch signaling pathway[137]
BDL-induced liver fibrosis in male Wistar or SD ratsInhibiting the activation of HSCs
CCl4-induced liver fibrosis in male C57/BL6 mice
Pokeweed antiviral protein (PAP)CCl4-induced liver fibrosis in male C57/BL6 miceInhibition of glycolysis through regulation of the Wnt/JNK pathway[24]
Suppressing HSC activation
San-Cao Granule (SCG)TAA-induced liver fibrosis in SD ratsInhibiting the TGF-β1/Smad pathway[132]
CGADMN-induced liver fibrosis in male Wistar ratsDownregulation of MMP2/9 activities and TIMP1/2 protein expression[136]
Inhibition of the TGF-β1/Smad signaling pathway
Inhibiting the activation of HSCs
Dahuang Zhechong pill (DHZCP)CCl4-induced liver fibrosis in male SD ratsInactivating the PI3K/Akt pathway[139]
Suppressing HSC proliferation
Jageum–Jung (JGJ)CCl4-induced liver fibrosis in male C57/BL6 miceInhibiting the activation and translocation of STAT-1 and NF-κB[109]
Regulating TGF-β1/Smad signaling.
Inhibiting HSC activation
Ganfule capsule (GFL)BDL-induced liver fibrosis in male C57/BL6 miceInhibiting glutamine metabolism[140]
Restricting the activation of the NF-κB pathway
Table 3. Anti-fibrotic synthetic compounds.
Table 3. Anti-fibrotic synthetic compounds.
CompoundsAnimal ModelTargets/Pathways/MechanismsReference
N-n-Butyl haloperidol iodide (F2)TAA-induced liver fibrosis in C57BL/6J miceReducing responsiveness of HSCs to TGF-β1[146]
CCl4-induced liver fibrosis in C57BL/6J miceInhibiting activated HSCs
Butaselen (BS)CCl4-induced liver fibrosis in male BALB/c miceInducing the apoptosis of activated HSCs[147]
Inhibiting the production of α-SMA and collagens by HSCs
Downregulating TGF-β1 expression and blocking the TGF-β1/Smads pathway
barbituric acid (BA)-5CCl4-induced liver fibrosis in male C57BL/6 miceBlocking both the TGF-β1 and LPS-induced NF-κB signaling pathways[102]
Inhibiting HSC activation and liver fibrosis
Inhibiting macrophages recruitment and activation
JD5037CCl4-induced liver fibrosis in male miceBlocking the CB1 receptor/β-arrestin1/Akt signaling pathway[79]
BDL-induced liver fibrosis in male miceBlocking the activation of HSCs
GNS561DEN-induced liver fibrosis in male ratsDisrupting TGF-β1 maturation and TGF-β1/Smad and MAPK signaling[16]
Inducing the apoptosis of HSCs
Preventing HSC activation and decreasing ECM deposition
Cultured bear bile powder (CBBP)DMN-induced liver fibrosis in wild-type Wistar ratsElevating the expression of PPAR-α and PPAR-γ[69]
Improving β-FAO and inhibiting inflammation
VatalanibCCl4-induced liver fibrosis in male BALB/c miceReducing liver inflammation[148]
EndostarCCl4-induced liver fibrosis in male BALB/c miceImproving liver function and reducing liver inflammation[149]
Inhibiting a-SMA protein expression
Reducing collagen accumulation
HS-173CCl4-induced liver fibrosis in male BALB/c mice Blocking the PI3K/Akt pathway[142]
Promoting HSC apoptosis
Inhibiting the expression of fibrotic mediators
RuxolitinibCCl4-induced liver fibrosis in male C57BL/6 miceBlocking JAK1/2 pathway[150]
TAA-induced liver fibrosis in male C57BL/6 miceInhibiting the activation of HSCs
Pantoprazole (PPZ)BDL-induced liver fibrosis in male SD ratsPromoting the proteasome-dependent degradation and ubiquitination of YAP[151]
Inhibiting HSC activation
Disrupting the interaction between OTUB2 and YAP
Tenofovir disoproxil fumarate (TDF)TAA-induced liver fibrosis in male C57BL/6 miceDownregulating the PI3K/Akt/mTOR signaling pathway[96]
The apoptosis of activated HSCs
SelonsertibDMN-induced liver fibrosis in male SD ratsBlocking ASK1/MAPK signaling[152]
Inducing apoptosis of HSCs
Rilpivirine (RPV)CCl4-induced fibrosis in female C57BL/6J miceSelective STAT1-dependent induction of apoptosis in HSCs[153]
BDL-induced fibrosis in female C57BL/6J micePromoting liver regeneration
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pei, Q.; Yi, Q.; Tang, L. Liver Fibrosis Resolution: From Molecular Mechanisms to Therapeutic Opportunities. Int. J. Mol. Sci. 2023, 24, 9671. https://doi.org/10.3390/ijms24119671

AMA Style

Pei Q, Yi Q, Tang L. Liver Fibrosis Resolution: From Molecular Mechanisms to Therapeutic Opportunities. International Journal of Molecular Sciences. 2023; 24(11):9671. https://doi.org/10.3390/ijms24119671

Chicago/Turabian Style

Pei, Qiying, Qian Yi, and Liling Tang. 2023. "Liver Fibrosis Resolution: From Molecular Mechanisms to Therapeutic Opportunities" International Journal of Molecular Sciences 24, no. 11: 9671. https://doi.org/10.3390/ijms24119671

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

Article Metrics

Back to TopTop