Next Article in Journal
Crocin Inhibits Angiogenesis and Metastasis in Colon Cancer via TNF-α/NF-kB/VEGF Pathways
Next Article in Special Issue
Focal Liver Lesion MRI Feature Identification Using Efficientnet and MONAI: A Feasibility Study
Previous Article in Journal
Perirhinal Cortex LTP Does Not Require Astrocyte BDNF-TrkB Signaling
Previous Article in Special Issue
Optimized Isolation and Characterization of C57BL/6 Mouse Hepatic Stellate Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advancements in Antifibrotic Therapies for Regression of Liver Fibrosis

1
Department of Research and Development, Cure Therapeutics Inc., Suwon 16506, Korea
2
Department of Microbiology, All India Institute of Medical Sciences, Rishikesh 249203, India
3
Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
4
Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh 249203, India
*
Author to whom correspondence should be addressed.
Cells 2022, 11(9), 1500; https://doi.org/10.3390/cells11091500
Submission received: 28 February 2022 / Revised: 21 April 2022 / Accepted: 26 April 2022 / Published: 29 April 2022
(This article belongs to the Special Issue Hepatology: From Natural History to Therapeutics)

Abstract

:
Cirrhosis is a severe form of liver fibrosis that results in the irreversible replacement of liver tissue with scar tissue in the liver. Environmental toxicity, infections, metabolic causes, or other genetic factors including autoimmune hepatitis can lead to chronic liver injury and can result in inflammation and fibrosis. This activates myofibroblasts to secrete ECM proteins, resulting in the formation of fibrous scars on the liver. Fibrosis regression is possible through the removal of pathophysiological causes as well as the elimination of activated myofibroblasts, resulting in the reabsorption of the scar tissue. To date, a wide range of antifibrotic therapies has been tried and tested, with varying degrees of success. These therapies include the use of growth factors, cytokines, miRNAs, monoclonal antibodies, stem-cell-based approaches, and other approaches that target the ECM. The positive results of preclinical and clinical studies raise the prospect of a viable alternative to liver transplantation in the near future. The present review provides a synopsis of recent antifibrotic treatment modalities for the treatment of liver cirrhosis, as well as a brief summary of clinical trials that have been conducted to date.

Graphical Abstract

1. Background

Chronic liver diseases are becoming a global health burden; they are responsible for approximately 2 million deaths per year, with approximately 1 million deaths resulting from complications associated with cirrhosis [1]. The mortality rate varies significantly across different geographical regions. In places such as Central Asia, Central Europe, Eastern Europe, and Central America, it is regarded as a public health priority. Furthermore, only about one-third of countries have accurate mortality data [1]. Increased mortality may be linked to viral hepatitis, but other factors—such as alcoholic and non-alcoholic fatty liver diseases, which are currently the most common in developed countries—may also be contributing factors [2]. Because of the increasing morbidity and mortality associated with chronic liver diseases, it is imperative that action be taken immediately to prioritize the identification and treatment of patients [3].
Over the last decade, the number of studies on organ fibrosis has increased, and common characteristics of fibrosis across different tissues have been identified. While tissue fibroblasts are considered to be heterogeneous, myofibroblasts exhibit similar phenotypes and molecular characteristics in fibrotic organs such as the lungs, kidneys, and liver, indicating that a conserved pathogenic pathway is common in fibrotic organs of different origins [4]. Initially, the development of advanced fibrosis was thought to be associated with specific signaling pathways. However, the signaling networks in fibrotic diseases have been overlooked, and a comprehensive understanding of molecular pathways is required to understand the disease. Because of the activation of the myofibroblast cell type, the pathogenesis of fibrosis shares its etiology with altered epithelial–mesenchymal cell interactions, inflammation, and fibroblast proliferation [5]. The TGFβ-SMAD2/3 signaling pathway, which is involved in cell transdifferentiation into myofibroblasts and abnormal extracellular matrix deposition, is another common pathway [6].
The progression of fibrosis into cirrhosis is determined by the development of portal hypertension, the activation of systemic inflammation, and the development of hyperdynamic circulatory dysfunction [7]. Portal hypertension is a detrimental complication that can obstruct portal blood flow [8]. In the case of cirrhosis, increased intrahepatic vascular resistance to the portal flow can elevate portal pressure. In patients with chronic liver diseases, portal hypertension can be driven by progressive fibrosis and intrahepatic vasoconstriction [9,10]. Because these diseases do not predictably manifest themselves, they are associated with varying mortality risks, as previously stated. As a result, a multistate approach to describing the clinical course has been considered. This necessitates the evaluation of the probabilities of various outcomes. This review aims to summarize relevant clinical states, possible regression methodologies, and design frameworks that may apply to multistate models of fibrosis.

1.1. Hepatic Scarring and Extracellular Matrix Leading to Fibrosis

Fibrogenesis is regarded as a normal wound-healing mechanism that occurs in response to any type of tissue damage. The activation of fibrogenic pathways by hepatocellular injuries results in the secretion of fibrogenic components into the extracellular matrix which, in turn, results in the formation of scar tissue [11,12]. In a normal liver, the ECM regulates a balance between synthesis and degradation [13]. Regulated amounts of glycoproteins—such as fibronectin and laminin—type IV collagen (non-fibrogenic), and proteoglycans, such as heparan sulfate, are also found in the collagen fibers [14,15]. However, during chronic injury, the ECM production exceeds that of its degradation by 6–8-fold, leading to the thickening of fibrotic septa and the crosslinking of collagen [12]. As illustrated in Figure 1, the non-fibrogenic type IV collagen is gradually replaced by the fibrogenic type I and II collagen [12].
The involvement of parenchymal and non-parenchymal liver cells, as well as infiltrating immune cells, complicates the process of the progression and resolution of fibrosis [16,17]. Additionally, smooth-muscle actin, fibronectin, and hyaluronic acid are secreted into the ECM [18]. Cirrhosis develops as a result of disease progression and sustained fibrogenesis. It is defined by the end-stage accumulation of scarring as well as the distortion of liver cells and their vasculature [19]. While the response to tissue injury is rapid, persistent or repeated injury results in the death of hepatocytes via apoptosis and necrosis, which is a critical step in activating inflammatory and pro-fibrogenic pathways, thereby initiating the progression of fibrosis [12,16].
Most chronic liver diseases, such as viral hepatitis B and C or alcoholic (ASH) and non-alcoholic steatohepatitis (NASH), can progress to the formation of fibrotic tissue and scarring [20,21,22]. Persistent liver injury can activate HSCs, which are one of the primary sources of hepatic scarring [23]. HSCs are classified into four distinct phenotypes—quiescent, activated, inactivated, and senescent—each of which plays a critical role in liver fibrosis [24]; their phenotypes are highlighted in Table 1. Activated HSCs can undergo apoptosis or revert to an inactivated phenotype that is similar to but distinct from quiescent HSCs during regression [25]. Immune cells can aid in fibrogenesis and fibrosis regression by facilitating the reabsorption of the fibrous scar [26].
KCs, NK cells, NKT cells, and dendritic cells coordinate with antigen-presenting cells—T and B cells—to connect the innate and adaptive immunity in the liver. An imbalanced inflammatory milieu following persistent injury to the liver gradually advances to liver fibrosis [30]. Recently, various novel immune cells—including T-helper 17 (Th17), regulatory T cells (Tregs), mucosa-associated invariant T cells (MAITs), and their related cytokines have been reported to regulate liver fibrosis [31,32,33]. Th17 cells—a subset of CD4+ T show fibrogenic properties owing to their interleukin (IL)-17, IL-22, and IL-23 production [34,35]. High levels of intrahepatic Th17 and IL-17 are commonly observed in liver fibrosis caused by various etiologies [36,37]. IL-17A promotes the transformation of HSCs into myofibroblasts and the production of collagen through the STAT3 signaling pathway [35]. The Tregs/Th17 ratio is found to be altered during liver fibrosis progression. The JAK inhibitor “tofacitinib” has been shown to restore the Tregs/Th17 balance, thereby alleviating liver fibrosis [38,39]. The MAIT cells account for approximately 30% of all CD3+ T cells present in the liver, and can be stimulated by IL-12 and IL-18 to secrete IFN-γ and granzyme [40,41]. MAIT cells are also found to promote the production of pro-inflammatory cytokines, such as IL-6 and IL-8, in mono-derived macrophages in animal models [42]. High levels of CXCR6+ T cells (auto-aggressive) have been detected in the blood of hepatitis-C–infected patients compared with healthy controls, and > 60% of intrahepatic human T cells expressed CXCR6, including CD4, CD8, and CD56+ (NK) T cells [43]. CXCL16 is a ligand for CXCR6+, and has been detected in the hepatocytes and bile ducts of patients with liver disease [43], as well as in murine liver sinusoidal endothelial cells [44] and in the macrophages or dendritic cells of other organs. Functionally, CXCR6+ CD8 T cells express granzyme, TNF-α, IFN-γ, and programmed cell death protein 1 (PD-1), suggesting an activated exhausted phenotype during NASH. In a sequential process triggered by IL-15, metabolic signals such as acetate and extracellular ATP activate CXCR6+ CD8 [45] T cells that promote non-specific killing of hepatocytes and instigate disease progression [45].
Single-cell RNA sequencing (scRNA-Seq) has uncovered the complex cell-to-cell interactions of non-parenchymal cells in chronic liver diseases and cirrhosis. scRNA-Seq analysis of healthy and cirrhotic human livers unveiled heterogeneity in fibrosis-associated non-parenchymal cells [46]. Ramachandran et al. showed the presence of a specific macrophage subpopulation—i.e., scar-associated macrophages (SAMs)—to be more prevalent in cirrhotic tissue in comparison to the healthy liver. These SAMs can activate HSCs, and promote mesenchymal cell activation and fibrogenesis. scRNA-Seq analysis also unveiled the role of platelet-derived growth factor receptor-α (PDGFRα) mesenchymal cells in scar-associated mesenchymal cells and two previously unknown subpopulations of scar-associated endothelial cells [46]. Understanding the molecular mechanism underlying hepatic fibrogenesis is critical for developing novel therapeutic strategies [25].

1.2. Fibrosis Reversibility and Possible Regression

Recent evidence refutes the long-held belief that cirrhosis (a more advanced form of fibrosis) is always irreversible [47,48]. In certain animal experiments, removing the causative agent resulted in cirrhosis regression [49]. For instance, in the carbon tetrachloride (CCl4) and bile duct ligation (BDL) models of cirrhosis, cessation of the injury results in resolution of the cirrhosis [47]. Regardless of the initial cause of injury, fibrosis marks a common pathway for chronic hepatic inflammation. However, since the wound-healing response is a dynamic process, it has the potential to resolve without scarring. The removal of the injurious stimulus has been shown to improve fibrosis in viral hepatitis [50,51], alcoholic liver disease [52], biliary obstruction [50], and autoimmune hepatitis [51]. Thus, the aspect common to all cases of cirrhosis improvement is the eradication of the underlying causative agent. Effective fibrolysis requires the targeting of several mechanisms, including (a) ECM degradation, (b) myofibroblast deactivation, (c) hepatocyte regeneration, and (d) vascular and parenchymal remodeling.
Although the precise point at which cirrhosis becomes irreversible is unknown, chronic damage results in an increase in the production of acellular and thick fibrotic septa that are resistant to degradation [53]. Collagen—the most abundant ECM scaffolding protein, is crosslinked and wrapped in elastin filaments, which results in decreased matrix metalloprotein (MMP) expression/activation [54]. Additionally, ECM crosslinking affects myofibroblast behavior, and an insoluble ECM prevents myofibroblasts from deactivating [18]. The efficiency of fibrolysis is determined by several factors—most notably the HSC population and is modulated by the ability of hepatocytes to regenerate and the response of inflammatory cells to repeated injury. The immune system plays a critical role in the progression and regression of fibrosis, and macrophages play a critical role in wound healing [26]. Apart from matrix degradation, fibrosis resolution requires the deactivation/eradication of activated HSCs.
The role of platelets in improving liver fibrosis has been proven. Studies have shown that platelets can decrease collagen production by inactivating HSCs [55]. Growth factors such as platelet-derived growth factor (PDGF) and hepatocyte growth factor (HGF) promote liver regeneration [55]. The regenerative effect of platelets in the liver involves a direct effect on hepatocytes, a cooperative effect with liver sinusoidal endothelial cells, and a collaborative effect with KCs [55].

1.3. Potential Targets for Cellular Therapy

Antifibrotic therapy primarily targets activated HSCs, (portal) myofibroblasts, and the ECM. However, HSCs and myofibroblasts have the ability to communicate with a wide variety of cell types, and can initiate fibrogenesis, induce their quiescence and apoptosis, or even remove excess ECM via the release of fibrotic enzymes and phagocytosis [16]. These pathways may collectively contribute to the development of additional or complementary pharmacological targets. Direct-action antifibrotics are those that target HSCs, myofibroblasts, and the ECM, whereas indirect-action antifibrotics target other cell types and their pathways [16].
When developing antifibrotic treatment strategies, it may be beneficial to consider two major multicellular functional units that can contribute to fibrosis, depending on the etiology and stage of liver disease: (1) perisinusoidal/vascular—consisting of pericytes such as HSCs, sinusoidal endothelial cells, macrophages/KCs, NKs, and other inflammatory cells, as well as hepatocytes; (2) portal/periportal—consisting of cholangiocytes/ductular cells, portal fibroblasts, and myofibroblasts, as well as various inflammatory cells such as T and B cells and macrophages/dendritic cells [16,56,57]. Other cellular interactions occurring within these units include various growth factors, cytokines, and proteases that are potentially useful in the treatment of fibrosis. During fibrosis, the cellular interaction within and between these multicellular units is distorted as a result of the chronic wound-healing response, which is characterized by the deposition of excessive scar tissue and the remodeling of the blood vessels [58]. As previously stated, even in patients with advanced fibrosis/cirrhosis, when the injury is eliminated, there can be a reversal in liver architecture, as seen in patients treated for chronic hepatitis B and C infections [59,60]. Although there have been reversals, it is a slow process—particularly for patients with advanced fibrosis. As a result, there has been little progress in the development of antifibrotic therapies that are both effective and free of major side effects [16]. Furthermore, these therapies should carefully modulate multicellular units toward fibrolysis while simultaneously directing them toward non-fibrotic tissue maintenance. As a result, combination therapies that target two or more key cellular molecules or units will be required.

2. Designing the Framework for Developing Antifibrotic Therapies

2.1. Role of Non-Invasive Biomarkers

In recent years, there has been an increase in the need for and interest in identifying liver fibrosis through the use of non-invasive surrogate markers [61]. Both clinicians and patients may find serum markers of liver fibrosis to be an appealing and cost-effective alternative to liver biopsy in certain situations [56]. The non-invasive markers of liver fibrosis are split into two approaches: one is a physical approximation to measure liver stiffness, and the other used blood-based biomarkers. The stiffness and decreased elasticity of fibrotic liver can be assessed using transient elastography (TE) and magnetic resonance elastography (MRE), along with many other radiological techniques. Serum-based liver fibrosis markers are extensively evaluated and are typically divided into direct and indirect markers. Direct markers of fibrosis are smaller or larger fragments of the ECM released into the circulation during ECM turnover. They are further classified into matrix deposition (fibrogenesis-linked) and matrix degradation (fibrolysis-linked) markers. Indirect markers are routine laboratory tests reflecting hepatic alteration. Some algorithms use radiological, direct, or indirect panels of tests in different combinations. These combinational biomarkers improve the ability to correctly assess the degree of liver fibrosis [57]. Non-invasive biomarkers have previously been comprehensively reviewed by many studies and can be referred to by other research groups [62,63,64]. This field continues to evolve, and the search for ideal biomarkers is ongoing. Recent advances in the -omics approach have generated many clinically significant biomarkers for liver fibrosis; however, these newly identified biomarker candidates need validation in terms of performance characteristics.

2.1.1. Evolving Biomarker Candidates for Liver Fibrosis

Next-generation sequencing has identified mutations in genes that are highly expressed in hepatic cells—such as ABCB4, ALDOB, GBE1, FAH, ASL, SLC25A13, and SERPINA1—which predispose the liver to fibrosis [65]. Genetic variants in non-parenchymal cells, as well as in the inflammatory cytokines, have also been shown to increase susceptibility to fibrosis [66,67]. Seven genomic loci on chromosomes 4, 5, 7, 12, and 17 that impact fibrosis phenotypes have been identified based on quantitative trait locus analysis [63]. A summary of several genetic polymorphisms implicated in the occurrence of liver fibrosis was provided by Acharya et al. [12]. Wang et al. utilized single-cell RNA-sequencing-derived data from fibrotic/cirrhotic human livers and identified 61 liver-fibrosis-associated genes that may serve as a catalog of translatable drug target candidates. Furthermore, the gene regulatory network analysis identified CREB3L1 as a master regulator of many of these fibrosis-associated genes [64]. In the context of epigenetics, abnormal DNA methylation patterns have been found to be associated with inappropriate gene repression in liver fibrosis. Mild and severe liver fibrosis may show differential DNA methylation patterns at peroxisome proliferator-activated receptor-γ promoters in cell-free DNA [68]. Metabolomics comparison has identified serum metabolite signatures of liver fibrosis progression in chronic hepatitis C patients. Four serum metabolites were found to be significantly elevated in HCV patients with more advanced liver fibrosis severity. The choline–uric acid ratio was found to optimally differentiate between the early and late stages of liver fibrosis [66].
Numerous studies published in the last few years have established the role of urokinase plasminogen activator receptor (uPAR) and soluble urokinase-type plasminogen activator (suPAR) as biomarkers for liver fibrosis [67]. suPAR is a non-specific biomarker of inflammation. Elevated levels of it can be found in the bloodstream, and this is a strong indicator of chronic inflammation and underlying pathologies [69,70]. suPAR has also been shown to be elevated in chronic liver dysfunction, such as progressive liver fibrosis/cirrhosis [71,72,73]. Plasma levels of uPAR have been found to be closely related to the fibrosis stage in chronic hepatitis B and C [69]. Elevated suPAR concentration in cirrhotic patients is correlated significantly with the degree of cirrhosis and liver failure. Thus, serum suPAR is a potential novel biomarker for the diagnosis of cirrhosis, and indicative of an adverse prognosis (Figure 2) [74,75].
Using a cutoff level of > 9 ng/mL, suPAR predicted a poor prognosis, with a sensitivity and specificity of 70.7% and 77.8%, respectively [70]. In patients with decompensated cirrhosis, the suPAR level was significantly higher (median 12.9 ng/mL) than in patients with compensated cirrhosis (8.9 ng/mL) [76]. A strong correlation between suPAR and declining liver function (increasing AST/ALT and INR), independent of the etiology, was found [72]. suPAR has been evaluated as a prognostic marker of the severity of acute alcoholic pancreatitis. Using a cutoff value of 5 ng/mL, the sensitivity and specificity for predicting moderate or severe pancreatitis were 79% and 78%, respectively [77].
Another receptor, known as the Fas receptor or apoptosis antigen (APO-1), is a death receptor found on the surface of cells that, when bound to its ligand (the Fas ligand), induces programmed cell death (apoptosis) (FasL) [78]. Additionally, it has a soluble form (sFas) that is generated via alternative mRNA splicing. Both sFas and sFasL are non-invasive serum cell death biomarkers. sFasL levels have been found to be elevated in hepatitis, acute liver failure, and sepsis [74]. Additionally, sFasL levels are increased in NASH and steatosis [75,79]. As a result, the Fas/FasL signaling pathway is associated with a variety of diseases, including hepatitis, alcoholic liver disease (ALD), and fatty liver/steatosis associated with obesity [75].

2.1.2. Role of Gut Microbiota as a Biomarker in Liver Fibrosis

Recently, many studies have disclosed the relationship between liver fibrosis and alteration of the gut microbiota and have successfully identified the gut microbiota as a biomarker for predicting liver fibrosis. Li et al. reported a lower community richness in rats with liver fibrosis in comparison to rats with normal livers. A significant difference in bacterial community diversity was found between different fibrosis stages [80]. Metagenomic phylogenetic analysis of stool samples revealed a universal gut-microbiome-derived signature that accurately identifies cirrhosis across geographically separated cohorts regardless of etiology. The study also suggested that the key microbial species within the signature might play causal roles in the pathophysiology of cirrhosis [81]. Another study reported a panel of 30 features, including 27 bacterial features, with a discriminatory ability to detect cirrhosis in patients with non-alcoholic fatty liver disease [82]. Bacteroides and Ruminococcus have been proven to be associated with NASH and the severity of fibrosis [83]. Loomba et al. demonstrated a differential gut microbiome composition of NAFLD patients with or without fibrosis [84]. Although the gut microbiome signature shows promising results to improve disease diagnosis, to realize its full potential, multicentric human studies with a large sample size are required

2.2. Role of Metabolic Agents

2.2.1. Farnesoid X Receptor Agonist

The farnesoid X nuclear receptor (FXR), also known as the bile acid receptor, is involved in the secretion and reabsorption of bile acids. Its activation implies decrease in gluconeogenic gene expression, improved hyperglycemia, peripheral insulin resistance as well as reducing circulating triglycerides [85,86]. Hence, it is a potential target for NASH and related liver fibrosis.
Obeticholic acid (OCA) is an agonist of FXR that decreases bile acid synthesis and exerts anti-inflammatory and antifibrotic effects. In a multicenter, randomized, placebo-controlled trial [87,88] patients with NASH exhibited improvements in liver histological features after treatment.

2.2.2. PPAR Agonist

PPAR is a key regulator of lipid metabolism, and is approved by the Food and Drug Administration (FDA) as a molecular target for dyslipidemia [89]. Isoforms of PPAR include PPARα, which regulates cholesterol and bile acid homeostasis, and PPARγ, which contributes to inhibiting the activation of HSCs and reduces collagen production [90]. (Lanifibranor) IVA337 is a next-generation pan-PPAR agonist that has shown preventive and curative effects on fibrosis in a CCL4 model [91], and was tested clinically in patients without worsening the fibrosis at a dosage of 1200 mg, decreasing the SAF-A score by at least 2 points [87].

2.2.3. Insulin-Based Targets

The phase 3 ARMOR study (NCT04104321) inhibits SCD1 (stearoyl-CoA desaturase-1), which promotes the synthesis of fatty acids and reduces insulin resistance. The drug has been seen to be well tolerated in animal studies as well as in phase 2 trials [88]. There was a statistically significant decrease in fat percentage in patients receiving 300 mg of Aramchol vs. placebo. The details of various targets and their modes of action are shown in Table 2, which primarily highlights some current studies affecting NASH- and NAFLD-related liver fibrosis.

2.2.4. Renin–Angiotensin System Inhibitor

The renin–angiotensin system (RAS) is a crucial regulator of liver fibrosis as well as portal hypertension [98]. Activated HSCs can secrete angiotensin II, which can promote fibrosis via the angiotensin receptors [90]. Similarly, angiotensin receptor blockades may also attenuate liver fibrosis. Losartan (50 mg daily for 48 weeks) can decrease serum aminotransferase levels and promote improvements in NASH, with no adverse events [99,100]. Recently a phase 3 trial has also been posted to compare the efficacy of candesartan and ramipril in hepatitis-C-virus-related liver fibrosis [101,102]. Ras has also been shown to be associated with hypertension; therefore, candesartan—a widely used therapy—has shown promising results in clinical trials as well. It has demonstrated significant improvement in treatment outcomes with a reduction in fibrosis scores and α-SMA-positive fibrotic areas [12]. Long-term treatment with irbesartan in severe fibrosis with chronic hepatitis C showed no improvement in fibrosis score, but was well tolerated and considered to be a safe treatment [103].

2.2.5. Inhibition of HMG-CoA Reductase

Statins are HMG-CoA reductase inhibitors, which reduce serum cholesterol levels by inhibiting the activity of HMG-CoA reductase [104]. The effects of statins on reducing liver inflammation, oxidative stress, and fibrosis have been reported in many animal model studies [105]. However, the safety of statins needs to be evaluated further. Moreover, a study reported that 3% of cirrhotic patients who were administered statins had severe rhabdomyolysis [106]. Currently, placebo-controlled trials examining statins, including simvastatin, are underway to check the safety of the drugs [107]. Various clinical trial details are shown in Table 3, highlighting studies associated with liver fibrosis.

2.3. Cellular Target-Specific Fibrosis Resolution

A targeted approach is directed towards known or established molecular targets or pathways that are critical for fibrogenesis or fibrolysis and, more importantly, do not overlap significantly with unrelated pathways, so as to avoid potential side effects [114]. These agents may be enzyme inhibitors or small-molecule inhibitors.
This review article discusses a variety of lesser-known transcription factors (Table 4). Numerous antifibrotic strategies have been developed to inhibit the pro-fibrogenic TGF signaling pathway, including the use of soluble TGF receptor type II [115,116], TGF-blocking antibodies [117], and TGF antisense oligonucleotides, or molecules [118] that disrupt downstream signal transduction. Lerdelimumab and metelimumab—two monoclonal antibodies against TGFβ—are currently in phase 3 and phase 1/2 clinical trials for reducing scarring after glaucoma surgery and systemic sclerosis, respectively [119,120,121]. However, systemic targeting of the TGF pathway is limited because, in addition to stimulating wound healing and fibrosis, it acts as a central inhibitor of inflammation, and is required for epithelial differentiation and apoptosis. Another promising strategy is to inhibit tissue inhibitor of metalloproteinase-1 (TIMP-1) [122]—a major mediator of liver fibrosis—using a recombinant mutant protein derived from its ligand MMP-9 [123].
An additional intriguing novel target is integrin v6—a cell surface receptor expressed on activated epithelia during wound healing, tumorigenesis, and embryogenesis [124,125]. It is expressed exclusively on activated cholangiocytes in the liver, which act as potent promoters of liver fibrogenesis [126]. Inhibition of v6 has been shown to reduce collagen deposition, improve liver function, and slow the progression of fibrosis [127]. Hepatocyte nuclear factor 4 (HNF4) is another transcriptional factor involved in the differentiation and function of hepatocytes. Forced expression of HNF4 has been shown to ameliorate hepatic fibrosis, improve liver function, and inhibit EMT in a fibrosis model [128]. Additionally, siRNA-mediated inhibition of HNF exacerbated hepatic fibrosis and decreased E-cadherin, vimentin, and fibroblast-specific protein-1 expression [129].
Table 4. Molecular targets for antifibrotic therapies and their associated clinical trials.
Table 4. Molecular targets for antifibrotic therapies and their associated clinical trials.
TargetCellsDrugClinical
Trial Stage
Clinical Trial DetailsReferences
TGFβHSCs, cholangiocytes, inflammatory cells, endotheliaSoluble type II, anti-TGFβ antibody;Phase 2FG-3019 for HBV infection, anti-CTGF monoclonal antibody;[119,120,121]
Lerdelimumab;Phase 3Monoclonal antibody, Cambridge antibody technology;
MetelimumabPhase 1/2Monoclonal antibody, Cambridge antibody technology
TIMP1HSCs, human endothelial cells, lymphoma, and breast carcinomaMMP antagonist Serum levels of TIMP-I in 268 patients with liver diseases[122]
Integrin αvβ6Activated epitheliaSmall-molecule antagonist, blocking ABPhase 2Monoclonal antibody (STX-100) for idiopathic pulmonary fibrosis[125]
TLR-4Macrophages, HSCSmall-molecule antagonists and downstream targetsPreclinical trialTLR4-deficient mice protected against hepatic injury;
vaccine development for hepatitis B (GlaxoSmithKline/Dynavax)
[130,131]
HNF4αHepatocytes, pancreatic beta cellsHNF4α agonistsPreclinical trial(CureVac) restoration of HNF4α via mRNA delivery using paraoxonase 1 as a therapeutic target[132]
LPAHSCLPA receptor and small-molecule antagonistPhase 2Idiopathic pulmonary fibrosis treatment using the LPA1 pathway[133]
Lysophosphatidic acid (LPA) is a lipid mediator that has been implicated in a variety of functions, including apoptosis, migration, proliferation, and cancer cell invasion [134]. The expression of LPA and LPA1R (lysophosphatidic acid receptor) is elevated in a variety of inflammatory states [135]. While LPA has a variety of physiological effects on the receptors of parenchymal cells, LPA1R antagonists have been shown to have an antifibrotic effect on liver fibrosis and lung fibrosis [133,136,137]. Table 4 summarizes the molecular targets, as well as the clinical trials that have been conducted and their current statuses.

2.4. Role of Growth Factors in Liver Fibrosis

HGF was identified as a mitogen for hepatocytes; it is produced by stromal cells, and stimulates epithelial cell proliferation, motility, morphogenesis, and angiogenesis in a variety of organs [138]. TGF-β plays a critical role in tissue fibrosis during chronic organ injury by converting HGF-producing fibroblasts to ECM-producing myofibroblasts [139,140]. HGF inhibits TGF-β production in myofibroblast cultures [141,142], and also blocks the TGF-mediated signaling pathway by inhibiting nuclear Smad2/3 activation [143], which can result in antifibrotic effects in vivo. Additionally, HGF inhibits the function of other fibrotic cytokines such as PDGF and CTGF/CCN2 and acts as a pro-fibrogenic factor, the details of which are mentioned below. MMPs were also induced in myofibroblasts by HGF [144,145]. MMPs must be induced by HGF not only for ECM degradation, but also for myofibroblast elimination, both of which contribute to fibrosis resolution. For example, HGF-induced MMP 9 degrades fibronectin—a critical cellular anchor [144]; this results in the apoptosis of myofibroblasts, which is required for the resolution of fibrosis. The resolution of fibrosis creates an opening for epithelial and endothelial repair, which may result in organ recovery. HGF is also required for tissue protection during inflammatory diseases, either directly on macrophages, dendritic cells, or lymphocytes [138] (immunogenic-cell-based mechanism), or indirectly on epithelial cells (epithelial-cell-based mechanism). Both mechanisms have the potential to be beneficial in inflammatory states. Given that HGF is required for organ protection and tissue regeneration, it is reasonable to assume that HGF-based therapy, variants, or fragments—in combination with activation of HGF/c-Met signaling, which decreases TGF- mediators—may show promise in treating a variety of inflammatory or fibrotic diseases [146]. Figure 3 shows a balance between MMPs and TIMPs for potential fibrosis resolution.
CTGF/CCN2 is a pro-fibrogenic molecule and a multifunctional matricellular protein produced by a variety of cell types. Of the many functions of CTGF/CCN2, it also has the ability to promote fibrosis, and can be seen to be overexpressed in many fibrotic lesions, including in the liver [147]. CTGF/CCN2 is activated by TGF-β, and mediates ECM-inducing properties previously attributed to TGF-β. In the fibrotic liver, CTGF/CCN2 mRNA and protein are produced by fibroblasts, myofibroblasts, HSCs, endothelial cells, and bile duct epithelial cells [148]. CTGF/CCN2 expression in cultured HSCs is also enhanced following their activation by TGF-β, while exogenous CTGF/CCN2 promotes HSC adhesion, proliferation, and collagen production. CCN2’s action has also been confirmed in vivo, with a transgenic FVB mouse [148]. Production of human CCN2 mRNA and its elevated levels were found in transgenic livers. It is believed that during the initiation of downstream fibrogenic events in the liver, the production of CTGF/CCN2 is regulated primarily by TGF-β, and CTGF/CCN2 plays an important role in HSC activation and the progression of fibrosis [147].
Another growth factor that works via the PDGF-α receptor and is considered to be a potent mitogen for human fibroblasts, as well as vascular smooth muscle cells in vitro, is the PDGF [149]. Studies suggest that this also plays an important role in the regulation of fibrosis. Initial experiments included a murine model derived from the transgenic overexpression of PDGF in the heart-induced fibroblast proliferation, which ultimately resulted in cardiac fibrosis, hypertrophy, and eventually cardiomyopathy [150,151]. Transgenic mice with liver-specific PDGF overexpression experienced liver fibrosis and hepatocellular carcinoma [152]. Blocking PDGF signaling has also been known to inhibit HSC proliferation and ameliorate liver fibrogenesis [153]. Clinical studies have also shown that excessive activation of PDGF and its downstream molecules is associated with necroinflammation and fibrosis in patients with hepatic damage [154,155]. Hence, PDGF and its signaling pathway play an important role in the development and prognosis of hepatic fibrosis.

2.5. Reduction in Inflammation and Immune Response

Inflammation is a critical and complex component of liver fibrosis; following liver injury, an accumulation of recruited inflammatory cells occurs at the site of the injury [20]. Platelets, neutrophils, macrophages, mast cells, and NK cells from the innate immune response, as well as T and B cells from the adaptive immune response, also participate in the fibrogenesis process [156]. The inflammatory response of immune cells during the process of fibrosis is mediated by a cocktail of pro- and anti-inflammatory compounds, including cytokines, chemokines, and growth factors [157,158]. HSCs also actively participate in the inflammatory process through their interactions with various immune cells. Additionally, HSCs are converted from a dormant state to an activated state via the myofibroblast-like phenotype, which is involved in proliferation and extracellular matrix deposition [159].
Tumor necrosis factor-alpha (TNF-α)-stimulated gene 6 (TSG-6) was identified as a cDNA derived from TNF-treated human fibroblasts, which maps to chromosome 2q23.3 [160,161,162,163]. This 35 kDa glycoprotein is not found in healthy adults, but is produced in response to inflammatory mediators, and is detected in a variety of inflammatory diseases, including those that cause liver fibrosis [164]. However, it has been demonstrated that increased TSG-6 expression during an inflammatory process contributes negatively to the inflammatory response [165]. TSG-6 was recently identified as a critical immune modulator secreted by human mesenchymal stem cells (hMSCs), and is responsible for hMSC-based therapeutic effects such as improved wound healing and cardiac function [166] but, more importantly, its effect on liver regeneration using conditioned media and organoids derived from TSG-6-treated HSCs in an acute liver injury model has been demonstrated [167]. In recent years, studies have documented that signal transducer and activator of transcription 3 (STAT 3) is closely related to the development and occurrence of liver fibrosis. TSG-6 was found to be effective at inhibiting hepatic oxidative stress and inducing hepatic M2 macrophage polarization by suppressing STAT3 activation [168]. Additionally, this study suggests that TSG-6 may play a critical role in liver regeneration, and may act as a protective factor against liver damage caused by inflammation and fibrosis [168], as illustrated in Figure 4. This could aid in the development of more novel therapeutic strategies, either alone or in combination.

2.6. Immune-Mediated Role of NK Cells

Cell senescence is a state of terminal growth arrest that is triggered by stress signals and cellular damage, and ultimately results in cell apoptosis if not corrected [169]. Senescent cells have an enlarged morphology and distinct metabolic and gene expression patterns in comparison to proliferating cells. Additionally, they exhibit a senescence-associated secretory phenotype, which is characterized by the production of pro-inflammatory factors, angiogenesis factors, and MMPs, which alter tissue function by promoting angiogenesis, attracting immune cells, and remodeling the ECM [170,171,172]. Due to mounting evidence that senescent cells have a detrimental effect on age-related declines and inflammatory diseases such as fibrosis, a major goal in this field is to develop an intervention capable of selectively identifying and eliminating senescent cells. Because these cells express a wide variety of proteins—particularly those on their cell surface—the concept of identifying the presence of a unique cell surface protein on senescent cells is critical in this strategy. Dipeptidyl peptidase 4 (DPP4 or CD26) was identified as a surface protein expressed more abundantly in senescent cells using mass spectroscopy [171]. RNA isolation from proliferating and senescent cells, followed by RT-qPCR analysis, also revealed that senescent cells expressed significantly more DPP4 mRNA, implying that DPP4 may promote cellular senescence [171].
NK cells are innate immune system lymphocytes that can rapidly eliminate stressed or apoptotic cells [173]. NK cells kill senescent cells via exocytosis of granules containing perforin and granzyme, and produce IFN-γ in response to senescent cell interaction [174]. Another preferential method of eradicating senescent cells is through antibody-dependent cytotoxicity (ADCC) [175]. The ADCC assay employs antibodies to recognize specific antigens on the cell surface, and directs natural killer cells to selectively destroy antibody-labeled cells [176]. The presence of DPP4 on the surface of these cells makes them suitable targets for NK cells that recognize anti-DPP4 antibodies, and the dual action of DPP-induced cellular senescence and NK cells can be seen in action in Figure 5 [171]. Additionally, it was observed that using an anti-PD1 antibody (CT-011) enhanced NK cell function [177]. It has been demonstrated that inhibiting the PD-1 pathway enhances NK cells’ IFN-γ release.

2.7. Role of Cytokines in Liver Fibrosis

Suppressive cytokines are critical for orchestrating the shape of numerous immune cells, along with fibrotic tissue reduction. Interleukins (IL) are a family of immunomodulatory cytokines or small signaling proteins that play a critical role in immune response regulation [178]. They are produced by a variety of cell types during inflammatory responses, and the balance of these cytokines dictates the outcome of the immune response. As a result, they are a critical therapeutic tool in the treatment of patients with liver diseases. In chronic liver diseases, interleukins may have both pro- and anti-inflammatory functions; some even express both, and are dependent on the inflammatory stimulus [178]. For example, IL-17 can promote hepatic fibrogenesis by activating hepatic stellate cells, whereas IL-22 protects against the development of fibrosis or steatohepatitis [179]. Similarly, IL-13 and IL-33 are related to Th2 and innate lymphoid cells, respectively, and contribute to the fibrotic response to liver injury, whereas IL-10 is a model anti-inflammatory interleukin with tissue-protective properties during chronic liver injury and fibrogenesis [178]. Table 5 summarizes the functions of various interleukins.
Several cytokines can affect the number as well as the function of myofibroblasts. Both inhibitory and stimulatory effects on myofibroblasts have been described [191]. TGF-β, PDGF and IL-6 are key cytokines for the formation and activity of myofibroblasts. Additionally, IL-4, IL-13, and IL-22 show pro-fibrotic activity by enhancing the production of collagen type I in normal fibroblasts [192,193,194]. TNF-α induces proliferation and collagen synthesis of atrial and intestinal myofibroblasts [195]. Apart from these stimulatory cytokines, several signaling molecules inhibit myofibroblast formation and activity. For instance, interferon-γ (IFN-γ) inhibits collagen synthesis, sensitizes skin fibroblasts to Fas-mediated apoptosis, and inhibits the effects of IL-4 [196,197]. A time- and dose-dependent induction of αSMA expression in human lung fibroblasts has been demonstrated, suggesting that myofibroblasts can be partially activated and enhance the production of collagen type I in normal fibroblasts [194,198]. Additionally, cytokines have the potential to enhance immune-cell-mediated immunity, particularly in NK cells. Due to its ability to promote T- and NK-cell proliferation, homeostasis, and cytotoxicity [199], IL-2 was the first cytokine to be used in clinics to enhance immune responses, including a novel IL-2 variant called super-2 with a higher binding affinity for IL-2R [200]. This modified IL-2 induced an increase in cytotoxic T-cell expansion and a decrease in Treg-cell activation. Another cytokine, IL-15, stimulates CD8+ T cells and non-differentiated NK cells, suggesting that it could be a significant immunotherapeutic agent [201]. IL-12 is another cytokine that can be administered to enhance NK cells’ cytolytic activity. It facilitates the release of IFN-γ, migration, and NK-mediated ADCC (as mentioned in Section 2.6) [202].
Chemokines are a family of small heparin-binding molecules that assist leukocytes in infiltrating the liver following acute or chronic injury [203]. Chemokines are produced by resident cells of the liver, including hepatocytes, HSCs, leukocytes, and platelets. Recently, numerous significant effects of specific chemokines and their receptors have been discovered. The C-X-C chemokine receptor type 3 (CXCR3) is a critical chemokine receptor that binds to the CXC chemokine ligands CXCL9, CXCL10, and CXCL11 [204]. CXCL9 is secreted directly by hepatocytes, and can bind to HSCs via the CXCR3 receptor. Unlike other chemokines, CXCL9 does not stimulate HSCs, but inhibits collagen secretion and mRNA expression. Given the critical role of stellate cells in liver fibrogenesis, it is reasonable to assume that CXCL9 has antifibrotic properties [203,205].

2.8. Role of miRNA Family

The mechanism of hepatic fibrosis is a highly complex process involving numerous cellular and molecular events. As a result of exogenous factors, dormant HSCs become activated and transform into myofibroblasts, resulting in the formation of ECM and, ultimately, liver fibrosis. The progression of hepatic fibrosis is associated with a variety of integrated signaling pathways, including the MAPK, Wnt, PI3K/AKT, and Hedgehog/Gli pathways [206]. The use of miRNAs to induce RNA interference and thus regulate gene transcription levels, is generating considerable interest in this field. miR-29 has been an extensively studied and referenced microRNA in the development of liver fibrosis [207]. It is a miRNA that inhibits fibrogenesis and is also required for HSC activation. The important fibrogenic cytokine TGF-β is a potent activator of HSCs [208]. It inhibits miR-29 expression and promotes a fibrogenic environment by activating HSCs and increasing ECM deposition [209,210]. Additionally, by modulating the PI3K/AKT pathway, the miR-29 family induces cell apoptosis [206]. By inhibiting alpha-1 type I collagen, miR-29 overexpression results in decreased collagen deposition (Col1A1) [209,211]. Additionally, this family is involved in post-translational ECM and fibril formation processing. The miR-29a subgroup has been shown to inhibit the activation of qHSCs by targeting HDAC4, whereas miR-29b induces cell apoptosis by targeting AKT3 and PI3KR1 [212]. Zhang et al. established an inverse correlation between miR-29b expression and heat shock protein (HSP47) expression, which are critical regulators of ECM maturation, and demonstrated that miR-29b overexpression results in abnormal collagen formation [213].
Similarly, it has been demonstrated that the miR-15 family promotes cell proliferation and induces apoptosis [214,215]. As illustrated in Figure 6, liver cells take and release exosomal miRNAs, and the extracellular vesicles (EVs) that are derived from adipose tissue, HSCs, and neutrophils [216]—containing miR-155, 214, and 223, respectively—are taken up by the liver cells, which can lead to suppression of PPARγ, leading to insulin resistance [217]. The subgroup miR-16 targets HGF and Smad7 to promote the TGF-/Smad signaling pathway, and inhibits cell proliferation by targeting CD1, whereas miR-15b and miR-16b promote apoptosis by targeting Bcl-2 [206]. Yan et al. identified a novel miRNA (miR-34a) that promoted HSC activation by targeting acyl-CoA synthetase long-chain family member 1 (ACSL1). Inhibiting miR-34a increased ACSL1 levels and decreased α-SMA levels [218]. Yuan et al. investigated the therapeutic potential of miR-155 in mice following liver injury caused by N-acetyl-p-aminophenol (APAP). It was found to be upregulated in liver tissue and blood samples following APAP injury. miR155-/- mice had elevated AST and ALT levels, as well as inflammatory mediators such as TNF-α and IL-6. Its deficiency may result in increased p65 and IKK expression, activating NF-κB [219]. Additionally, connective tissue growth factor (CTGF/CCN2) has been shown to be associated with fibrogenesis in HSCs. CCN2 upregulation is associated with miR-214 downregulation in fibrotic or steatotic livers or culture-activated primary murine HSCs [220]. miR223 was also found to protect against liver fibrosis via its regulation of multiple hepatic cell targets. It inhibits the transcriptional coactivator with PDZ-binding motif (TAZ) expression in hepatocytes, resulting in a decrease in hepatocyte-derived Indian hedgehog (IHH) secretion, which acts as a ligand for hedgehog signaling in HSCs [221].

2.9. Role of Mesenchymal Stromal Cells in Liver Tissues

MSCs are one of the main candidates for cellular therapies. Most of the time, these cells come from adult bone marrow (BM-MSCs), but umbilical cord blood (UCB) has also been shown to be a good source of hematopoietic stem cells, with evidence that MSCs do exist in CB [222]. Since they can change into mesodermal cells, they have a wide range of immunomodulatory properties. Many other immune cells can also be taken from the cord blood mononuclear cells (CBMCs), including the NK cell population [223], but the number of CB NK cells is very low, and they are known to be incompletely formed and need to be activated to keep working properly [224]. There are many ways to increase the number of these cells, which could help the immune system return to normal after a direct cell transplant, as well as in other cell-therapy-based research areas.
MSCs are also considered to be immune-privileged because they lack class II major histocompatibility complex (MHC) molecules, and have a very low number of class I MHC molecules, which enables their allogeneic use [225]. Numerous studies in clinical trials indicate that transferred MSCs have an antifibrotic effect (Table 6), but the timing of the therapy is critical, as the effect on fibrosis is mediated by a reduction in inflammation rather than directly promoting degradation. Numerous studies have been published demonstrating a decrease in TGFβ-1 and α-SMA gene expression in liver cells following MSC treatment [226,227,228,229]. Administration of BM-derived MSCs alleviated fibrosis and improved the hypoxic liver microenvironment in a CCL4-induced animal model. These cells exhibit a direct relationship with the TGF2/SMAD signaling pathway in liver cells [230,231]. Several studies also support the idea of MSCs being engrafted/differentiated directly into damaged tissue. The engrafted cells were detected long after transplantation in a liver intoxication model caused by a lethal dose of APAP [232].
MSCs can also exert pro-regenerative and antifibrotic effects in liver tissues by inducing the proliferation of resident mature hepatocytes or progenitor cells via the secretion of paracrine factors. They can inhibit the activation of HSCs in vitro via the production of IL-10, VEGF-A, and HGF, as demonstrated by gene expression analysis [243]. Additionally, the MSC secretome has been shown to be less invasive and effective in liver regeneration [244]. Numerous studies demonstrate that the secretome obtained from UC-MSCs differentiated or committed to hepatocyte-like cells enhances hepatic function both in vitro and in vivo. UC-MSC enriched with milk factor globule EGF8 (MFGE-8)—an antifibrotic protein—suppressed the expression of fibrosis by downregulating the α-SMA and TGF-β pathways [245]. TGF-activated HSCs using conditioned media from amniotic MSCs (AMSCs) [246] and BM-MSCs [247] also exhibited antifibrotic activity
EVs have been studied recently for their potential role in disease. MSC-derived EVs have been shown to promote tissue regeneration in a variety of tissues, including the heart, lungs, brain, kidneys, and liver. In a variety of preclinical models [248,249], MSC-EVs have been shown to possess therapeutic properties. In terms of fibrosis, they act on hepatocytes, activated HSCs, and immune cells by modulating their signaling pathways. Li et al. [250] provided one of the first pieces of evidence demonstrating that EVs from UC-MSCs alleviate hepatic inflammation and collagen deposition in a CCL4 liver fibrosis model. TNF-β, IL-1, and IFN-γ mRNA expression in liver tissue was decreased by UC-MSC-EVs [251]. Embryonic stem cells (ESCs) have also been identified as a potential alternative source of MSCs, with ESC-MSC-EVs increasing hepatocyte viability and decreasing apoptosis and pro-fibrotic cytokine expression in a thioacetamide (TAA) animal model [252]. Thus, MSC treatment has been shown to reduce fibrosis by downregulating fibrosis-related gene expression levels, including α-SMA, TGF-β, and collagens, with promising results obtained in preclinical studies using MSC-EVs.

2.10. Current Challenges in Clinical Trials

The current progress and research being conducted to better understand the mechanisms of hepatic fibrosis and its therapeutic targets emphasizes the critical nature of developing clinical trial designs that can evaluate the efficacy of antifibrotic drugs. It is also critical to understand the dynamics of fibrosis regression and the current inaccuracies of standard fibrosis staging systems (e.g., Ishak, Brunt, Metavir) [253], such as collagen proportionate area quantification [254]. Sustained suppression of hepatitis B and C results in significant improvements in inflammation and necrosis, as well as regression of fibrosis. Challenges are also directly related to drug efficacy. Selecting the appropriate endpoint and duration of therapy are also essential for assessing the efficacy of the drug in clinical trials. The reversal of NAFLD/NASH (without worsening fibrosis) or the improvement of fibrosis (with no further deterioration of NASH) are the endpoints for pre-cirrhosis patients. Moreover, adequate stratification is essential to ensure reliability in clinical trials. In trials for NAFLD, similar drugs are often given to patients with different underlying comorbidities, which can lead to a variety of treatment responses that must be considered and managed with specific strategies.
At the moment, many new antifibrotic agents being tested in clinical trials focus on NASH as an etiology [255]. This increased awareness and focus on NASH has resulted in remarkable advancements in specific therapies, as well as a growing understanding of obesity- and fatty-liver-related diseases, which affect 10 times as many people as HCV in the United States and Europe [256]. Another factor that could be considered is the stage of the disease—specifically, drugs that target inflammation and cell injury. They may be effective at the disease’s early and intermediate stages. Innovative trial designs should be considered, as they may help to address current pitfalls associated with liver fibrosis trials, including NASH and NAFLD. Ultimately, the approval of antifibrotic drugs will be based on endpoints that are either directly related to or reasonably predict specific clinical outcomes.

3. Conclusions

We continue to gain a better understanding of the pathophysiology of liver fibrosis. It can be triggered by genetic and metabolic disorders, chronic viral hepatitis, infections, drugs, cholestasis, and other environmental factors. Medical complications can occur because of the accumulation of ECM, disruption of lobular structure, and deterioration of hepatocellular function during this process. Numerous cellular and extracellular agents have been identified that can be activated or transformed into ECM-synthesizing phenotypes. Numerous experimental studies have demonstrated that a variety of agents may have antifibrotic potential. Despite this, only a small number of candidates have progressed successfully to the clinical trial stage. Numerous potential therapies have emerged in the current scenario that demonstrate promise—either individually, or as part of a combination study that targets multiple cells or pathways in order to provide a more holistic solution to the current fibrosis issues. However, while numerous therapies have demonstrated promise, additional research is necessary to determine whether these therapies can be translated into clinical practice. At the moment, removing fibrosis-causing agents and factors affecting stellate-cell activation remains a critical strategy for reducing and preventing the disease.

Author Contributions

A.J., K.K., A.K., P.S., B.M. and B.J.O. contributed to the draft conceptualization, data curation, formal analysis, validation, visualization, and writing of the original draft. All authors contributed to editing and reviewing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The conception, design, writing, and editing of this review article did not utilize any funding sources.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

All financial assistance during this review paper writing was provided by Cure Therapeutics Inc. Suwon, South Korea- 16506. The graphical abstract and Figure 1, Figure 4, Figure 5, and Figure 6 were created with BioRender.com, (Accessed on 29 April 2022).

Conflicts of Interest

The authors declare no conflict of interest that pertain to this work.

Abbreviations

AbbreviationMeaning
ABCB4ATP-binding cassette subfamily B member 4
ACSL1Acyl-CoA synthetase long-chain family member
ADCCAntibody-dependent cytotoxicity
AKTAKT serine/threonine kinase 3
ALDAlcoholic liver disease
ALDOBAldolase, fructose-bisphosphate B
APAPN-acetyl-p-aminophenol
APO-1Apoptosis antigen 1
ASHAlcoholic steatohepatitis
ASLArgininosuccinate lyase
BDLBile duct ligation
CCL4Carbon tetrachloride
CCN2Cellular communication network factor 2
CD1Cluster of differentiation 1
CREB3L1CAMP-responsive element-binding protein 3-like 1
CTGFConnective tissue growth factor
DPP4Dipeptidyl peptidase 4
ECMExtracellular matrix
EVsExtracellular vesicles
FAHFumarylacetoacetate hydrolase
FXR Farnesoid X receptor
GBE11,4-alpha-glucan branching enzyme 1
HDAC4Histone deacetylase 4
HGFHepatocyte growth factor
hMSCsHuman mesenchymal stem cells
HNF4Hepatocyte nuclear factor 4
HSCs, qHSCsHepatic stellate cells, quiescent hepatic stellate cells
HSP47Heat shock protein 47
IGFInsulin-like growth factor
IHHIndian Hedgehog
IKK, NF-κBInhibitor of nuclear factor-κB (IκB) kinase, nuclear factor-kappa B
ILsInterleukins
KCsKupffer cells
LPALysophosphatidic acid
LSECsLiver sinusoidal endothelium cells
MAITsMucosa-associated invariant T cells
MAPKMitogen-activated protein kinase
MFGE-8Milk factor globule EGF8
MHCMajor histocompatibility complex
miRNAsmicroRNAs
MMPMatrix metalloprotein
MREMagnetic resonance elastography
NASHNon-alcoholic steatohepatitis
NKsNatural killer cells
PD-1 Programmed cell death protein 1
PDGFPlatelet-derived growth factor
PI3kPhosphatidylinositol 3-kinase
PPARϒPeroxisome proliferators–activated receptor γ
SAMScar-associated macrophages
SERPINASerpin family A member 1
SLC25A13Solute carrier family 25-member 13
SMADFusion of Caenorhabditis elegans Sma genes and the Drosophila Mad, mothers against decapentaplegic
STAT3Signal transducer and activator of transcription 3
TAAThioacetamide
TAZTranscriptional coactivator with PDZ-binding motif
TETransient elastography
TGF-βTransforming growth factor β
TIMPTissue inhibitor of metalloproteinase
TLR4Toll-like receptor-4
TNFTumor necrosis factor
TNF-αTumor Necrosis Factor α
TregsRegulatory T cells
TSG-6TNF-α-stimulated gene 6
UCBUmbilical cord blood
uPA, uPARUrokinase plasminogen activator, urokinase plasminogen activator receptor
WntWingless-related integration site
WPBsWeibel–Palade bodies

References

  1. Asrani, S.K.; Devarbhavi, H.; Eaton, J.; Kamath, P.S. Burden of liver diseases in the world. J. Hepatol. 2019, 70, 151–171. [Google Scholar] [CrossRef] [PubMed]
  2. Mokdad, A.A.; Lopez, A.D.; Shahraz, S.; Lozano, R.; Mokdad, A.H.; Stanaway, J.; Murray, C.J.; Naghavi, M. Liver cirrhosis mortality in 187 countries between 1980 and 2010: A systematic analysis. BMC Med. 2014, 12, 1–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mitra, S.; De, A.; Chowdhury, A. Epidemiology of non-alcoholic and alcoholic fatty liver diseases. Transl. Gastroenterol. Hepatol. 2020, 5, 16. [Google Scholar] [CrossRef] [PubMed]
  4. Marcellin, P.; Kutala, B.K. Liver diseases: A major, neglected global public health problem requiring urgent actions and large-scale screening. Liver Int. 2018, 38, 2–6. [Google Scholar] [CrossRef] [Green Version]
  5. Makarev, E.; Izumchenko, E.; Aihara, F.; Wysocki, P.T.; Zhu, Q.; Buzdin, A.; Sidransky, D.; Zhavoronkov, A.; Atala, A. Common pathway signature in lung and liver fibrosis. Cell Cycle 2016, 15, 1667–1673. [Google Scholar] [CrossRef] [Green Version]
  6. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef] [Green Version]
  7. Pardali, E.; Sanchez-Duffhues, G.; Gomez-Puerto, M.C.; Ten Dijke, P. TGF-β-induced endothelial-mesenchymal transition in fibrotic diseases. Int. J. Mol. Sci. 2017, 18, 2157. [Google Scholar] [CrossRef] [Green Version]
  8. D’Amico, G.; Morabito, A.; D’Amico, M.; Pasta, L.; Malizia, G.; Rebora, P.; Valsecchi, M.G. Clinical states of cirrhosis and competing risks. J. Hepatol. 2018, 68, 563–576. [Google Scholar] [CrossRef] [Green Version]
  9. Iwakiri, Y.; Groszmann, R.J. Pathophysiology of portal hypertension. Variceal Hemorrhage 2014, 18, 281–291. [Google Scholar]
  10. Sauerbruch, T.; Trebicka, J. Future therapy of portal hypertension in liver cirrhosis–a guess. F1000prime Rep. 2014, 6, 95. [Google Scholar] [CrossRef] [Green Version]
  11. Khanam, A.; Saleeb, P.G.; Kottilil, S. Pathophysiology and Treatment Options for Hepatic Fibrosis: Can It Be Completely Cured? Cells 2021, 10, 1097. [Google Scholar] [CrossRef] [PubMed]
  12. Acharya, P.; Chouhan, K.; Weiskirchen, S.; Weiskirchen, R. Cellular Mechanisms of Liver Fibrosis. Front. Pharmacol. 2021, 12, 1072. [Google Scholar] [CrossRef] [PubMed]
  13. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef] [PubMed]
  14. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  15. Tátrai, P.; Egedi, K.; Somorácz, Á.; Van Kuppevelt, T.H.; Dam, G.t.; Lyon, M.; Deakin, J.A.; Kiss, A.; Schaff, Z.; Kovalszky, I. Quantitative and qualitative alterations of heparan sulfate in fibrogenic liver diseases and hepatocellular cancer. J. Histochem. Cytochem. 2010, 58, 429–441. [Google Scholar] [CrossRef] [Green Version]
  16. Trautwein, C.; Friedman, S.L.; Schuppan, D.; Pinzani, M. Hepatic fibrosis: Concept to treatment. J. Hepatol. 2015, 62, S15–S24. [Google Scholar] [CrossRef] [Green Version]
  17. Magee, N.; Zou, A.; Zhang, Y. Pathogenesis of nonalcoholic steatohepatitis: Interactions between liver parenchymal and nonparenchymal cells. BioMed Res. Int. 2016, 2016, 5170402. [Google Scholar] [CrossRef] [Green Version]
  18. Herrera, J.; Henke, C.A.; Bitterman, P.B. Extracellular matrix as a driver of progressive fibrosis. J. Clin. Investig. 2018, 128, 45–53. [Google Scholar] [CrossRef] [Green Version]
  19. Lee, U.E.; Friedman, S.L. Mechanisms of hepatic fibrogenesis. Best Pract. Res. Clin. Gastroenterol. 2011, 25, 195–206. [Google Scholar] [CrossRef]
  20. Tanwar, S.; Rhodes, F.; Srivastava, A.; Trembling, P.M.; Rosenberg, W.M. Inflammation and fibrosis in chronic liver diseases including non-alcoholic fatty liver disease and hepatitis C. World J. Gastroenterol. 2020, 26, 109. [Google Scholar] [CrossRef]
  21. Peng, C.-Y.; Chien, R.-N.; Liaw, Y.-F. Hepatitis B virus-related decompensated liver cirrhosis: Benefits of antiviral therapy. J. Hepatol. 2012, 57, 442–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Lackner, C.; Tiniakos, D. Fibrosis and alcohol-related liver disease. J. Hepatol. 2019, 70, 294–304. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, C.-Y.; Yuan, W.-G.; He, P.; Lei, J.-H.; Wang, C.-X. Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J. Gastroenterol. 2016, 22, 10512. [Google Scholar] [CrossRef] [PubMed]
  24. Sanz-García, C.; Fernández-Iglesias, A.; Gracia-Sancho, J.; Arráez-Aybar, L.A.; Nevzorova, Y.A.; Cubero, F.J. The Space of Disse: The Liver Hub in Health and Disease. Livers 2021, 1, 3–26. [Google Scholar] [CrossRef]
  25. Kisseleva, T.; Brenner, D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 151–166. [Google Scholar] [CrossRef] [PubMed]
  26. Wynn, T.A.; Vannella, K.M. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 2016, 44, 450–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Delgado, M.E.; Cárdenas, B.I.; Farran, N.; Fernandez, M. Metabolic Reprogramming of Liver Fibrosis. Cells 2021, 10, 3604. [Google Scholar] [CrossRef] [PubMed]
  28. Fujita, T.; Narumiya, S. Roles of hepatic stellate cells in liver inflammation: A new perspective. Inflamm. Regen. 2016, 36, 1–6. [Google Scholar] [CrossRef] [Green Version]
  29. Zhang, M.; Serna-Salas, S.; Damba, T.; Borghesan, M.; Demaria, M.; Moshage, H. Hepatic stellate cell senescence in liver fibrosis: Characteristics, mechanisms and perspectives. Mech. Ageing Dev. 2021, 199, 111572. [Google Scholar] [CrossRef]
  30. Wan, M.; Han, J.; Ding, L.; Hu, F.; Gao, P. Novel Immune Subsets and Related Cytokines: Emerging Players in the Progression of Liver Fibrosis. Front. Med. 2021, 8, 604894. [Google Scholar] [CrossRef]
  31. Kong, X.; Feng, D.; Wang, H.; Hong, F.; Bertola, A.; Wang, F.S.; Gao, B. Interleukin-22 induces hepatic stellate cell senescence and restricts liver fibrosis in mice. Hepatology 2012, 56, 1150–1159. [Google Scholar] [CrossRef] [PubMed]
  32. Zhan, T.; Ma, H.; Jiang, S.; Zhong, Z.; Wang, X.; Li, C.; Yu, D.; Liu, L.; Xu, J.; Xia, C. Interleukin-9 blockage reduces early hepatic granuloma formation and fibrosis during Schistosoma japonicum infection in mice. Immunology 2019, 158, 296–303. [Google Scholar] [CrossRef] [PubMed]
  33. Böttcher, K.; Rombouts, K.; Saffioti, F.; Roccarina, D.; Rosselli, M.; Hall, A.; Luong, T.; Tsochatzis, E.A.; Thorburn, D.; Pinzani, M. MAIT cells are chronically activated in patients with autoimmune liver disease and promote profibrogenic hepatic stellate cell activation. Hepatology 2018, 68, 172–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Tan, Z.; Qian, X.; Jiang, R.; Liu, Q.; Wang, Y.; Chen, C.; Wang, X.; Ryffel, B.; Sun, B. IL-17A plays a critical role in the pathogenesis of liver fibrosis through hepatic stellate cell activation. J. Immunol. 2013, 191, 1835–1844. [Google Scholar] [CrossRef] [Green Version]
  35. Meng, F.; Wang, K.; Aoyama, T.; Grivennikov, S.I.; Paik, Y.; Scholten, D.; Cong, M.; Iwaisako, K.; Liu, X.; Zhang, M. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 2012, 143, 765–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sun, H.; Zhang, J.; Zhang, H.; Zou, Z.; Wang, F.; Jia, J. Increased Th17 cells contribute to disease progression in patients with HBV-associated liver cirrhosis. J. Viral Hepat. 2012, 19, 396–403. [Google Scholar] [CrossRef] [PubMed]
  37. Hara, M.; Kono, H.; Furuya, S.; Hirayama, K.; Tsuchiya, M.; Fujii, H. Interleukin-17A plays a pivotal role in cholestatic liver fibrosis in mice. J. Surg. Res. 2013, 183, 574–582. [Google Scholar] [CrossRef]
  38. Gu, L.; Deng, W.S.; Sun, X.F.; Zhou, H.; Xu, Q. Rapamycin ameliorates CCl4-induced liver fibrosis in mice through reciprocal regulation of the Th17/Treg cell balance. Mol. Med. Rep. 2016, 14, 1153–1161. [Google Scholar] [CrossRef] [Green Version]
  39. Wang, H.; Feng, X.; Han, P.; Lei, Y.; Xia, Y.; Tian, D.; Yan, W. The JAK inhibitor tofacitinib ameliorates immune-mediated liver injury in mice. Mol. Med. Rep. 2019, 20, 4883–4892. [Google Scholar] [CrossRef]
  40. Ussher, J.E.; Klenerman, P.; Willberg, C.B. Mucosal-associated invariant T-cells: New players in anti-bacterial immunity. Front. Immunol. 2014, 5, 450. [Google Scholar] [CrossRef]
  41. Dusseaux, M.; Martin, E.; Serriari, N.; Péguillet, I.; Premel, V.; Louis, D.; Milder, M.; Le Bourhis, L.; Soudais, C.; Treiner, E. Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17–secreting T cells. Blood J. Am. Soc. Hematol. 2011, 117, 1250–1259. [Google Scholar] [CrossRef] [PubMed]
  42. Hegde, P.; Weiss, E.; Paradis, V.; Wan, J.; Mabire, M.; Sukriti, S.; Rautou, P.-E.; Albuquerque, M.; Picq, O.; Gupta, A.C.; et al. Mucosal-associated invariant T cells are a profibrogenic immune cell population in the liver. Nat. Commun. 2018, 9, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Heydtmann, M.; Lalor, P.F.; Eksteen, J.A.; Hubscher, S.G.; Briskin, M.; Adams, D.H. CXC chemokine ligand 16 promotes integrin-mediated adhesion of liver-infiltrating lymphocytes to cholangiocytes and hepatocytes within the inflamed human liver. J. Immunol. 2005, 174, 1055–1062. [Google Scholar] [CrossRef] [Green Version]
  44. Geissmann, F.; Cameron, T.O.; Sidobre, S.; Manlongat, N.; Kronenberg, M.; Briskin, M.J.; Dustin, M.L.; Littman, D.R. Intravascular immune surveillance by CXCR6+ NKT cells patrolling liver sinusoids. PLoS Biol. 2005, 3, e113. [Google Scholar] [CrossRef] [Green Version]
  45. Shimaoka, T.; Kume, N.; Minami, M.; Hayashida, K.; Kataoka, H.; Kita, T.; Yonehara, S. Molecular cloning of a novel scavenger receptor for oxidized low density lipoprotein, SR-PSOX, on macrophages. J. Biol. Chem. 2000, 275, 40663–40666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ramachandran, P.; Dobie, R.; Wilson-Kanamori, J.R.; Dora, E.F.; Henderson, B.E.P.; Luu, N.T.; Portman, J.R.; Matchett, K.P.; Brice, M.; Marwick, J.A.; et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 2019, 575, 512–518. [Google Scholar] [CrossRef]
  47. Issa, R.; Williams, E.; Trim, N.; Kendall, T.; Arthur, M.; Reichen, J.; Benyon, R.; Iredale, J. 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]
  48. Dienstag, J.L.; Goldin, R.D.; Heathcote, E.J.; Hann, H.; Woessner, M.; Stephenson, S.L.; Gardner, S.; Gray, D.F.; Schiff, E.R. Histological outcome during long-term lamivudine therapy. Gastroenterology 2003, 124, 105–117. [Google Scholar] [CrossRef]
  49. Ramachandran, P.; Iredale, J.P. Reversibility of liver fibrosis. Ann. Hepatol. 2009, 8, 283–291. [Google Scholar] [CrossRef]
  50. Hammel, P.; Couvelard, A.; O’Toole, D.; Ratouis, A.; Sauvanet, A.; Fléjou, J.F.; Degott, C.; Belghiti, J.; Bernades, P.; Valla, D. Regression of liver fibrosis after biliary drainage in patients with chronic pancreatitis and stenosis of the common bile duct. New Engl. J. Med. 2001, 344, 418–423. [Google Scholar] [CrossRef]
  51. Dufour, J.-F.; DeLellis, R.; Kaplan, M.M. Reversibility of hepatic fibrosis in autoimmune hepatitis. Ann. Intern. Med. 1997, 127, 981–985. [Google Scholar] [CrossRef] [PubMed]
  52. Parés, A.; Caballería, J.; Bruguera, M.; Torres, M.; Rodés, J. Histological course of alcoholic hepatitis: Influence of abstinence, sex and extent of hepatic damage. J. Hepatol. 1986, 2, 33–42. [Google Scholar] [CrossRef]
  53. Ismail, M.H.; Pinzani, M. Reversal of hepatic fibrosis: Pathophysiological basis of antifibrotic therapies. Hepatic Med. Evid. Res. 2011, 3, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hernández-Gea, V. Liver Fibrosis: What Is Reversible and What Not? How to Assess Regression. In Portal Hypertension VI; Springer: Cham, Switzerland, 2016; pp. 111–115. [Google Scholar]
  55. Kurokawa, T.; Ohkohchi, N. Platelets in liver disease, cancer and regeneration. World J. Gastroenterol. 2017, 23, 3228–3239. [Google Scholar] [CrossRef] [PubMed]
  56. Baranova, A.; Lal, P.; Birerdinc, A.; Younossi, Z.M. Non-invasive markers for hepatic fibrosis. BMC Gastroenterol. 2011, 11, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Chin, J.L.; Pavlides, M.; Moolla, A.; Ryan, J.D. Non-invasive Markers of Liver Fibrosis: Adjuncts or Alternatives to Liver Biopsy? Front. Pharmacol. 2016, 7, 159. [Google Scholar] [CrossRef] [Green Version]
  58. Schuppan, D.; Kim, Y.O. Evolving therapies for liver fibrosis. J. Clin. Investig. 2013, 123, 1887–1901. [Google Scholar] [CrossRef] [Green Version]
  59. Nallagangula, K.S.; Nagaraj, S.K.; Venkataswamy, L.; Chandrappa, M. Liver fibrosis: A compilation on the biomarkers status and their significance during disease progression. Future Sci. OA 2018, 4, FSO250. [Google Scholar] [CrossRef] [Green Version]
  60. Joseph, J. Serum Marker Panels for Predicting Liver Fibrosis–An Update. Clin. Biochem. Rev. 2020, 41, 67. [Google Scholar]
  61. Papastergiou, V.; Tsochatzis, E.; Burroughs, A.K. Non-invasive assessment of liver fibrosis. Ann. Gastroenterol. 2012, 25, 218. [Google Scholar]
  62. Bocsan, I.C.; Milaciu, M.V.; Pop, R.M.; Vesa, S.C.; Ciumarnean, L.; Matei, D.M.; Buzoianu, A.D. Cytokines genotype-phenotype correlation in nonalcoholic steatohepatitis. Oxidative Med. Cell. Longev. 2017, 2017, 4297206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Hall, R.A.; Liebe, R.; Hochrath, K.; Kazakov, A.; Alberts, R.; Laufs, U.; Bohm, M.; Fischer, H.P.; Williams, R.W.; Schughart, K.; et al. Systems genetics of liver fibrosis: Identification of fibrogenic and expression quantitative trait loci in the BXD murine reference population. PLoS ONE 2014, 9, e89279. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, Z.Y.; Keogh, A.; Waldt, A.; Cuttat, R.; Neri, M.; Zhu, S.; Schuierer, S.; Ruchti, A.; Crochemore, C.; Knehr, J.; et al. Single-cell and bulk transcriptomics of the liver reveals potential targets of NASH with fibrosis. Sci. Rep. 2021, 11, 19396. [Google Scholar] [CrossRef] [PubMed]
  65. Krenkel, O.; Hundertmark, J.; Ritz, T.P.; Weiskirchen, R.; Tacke, F. Single Cell RNA Sequencing Identifies Subsets of Hepatic Stellate Cells and Myofibroblasts in Liver Fibrosis. Cells 2019, 8, 503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Shanmuganathan, M.; Sarfaraz, M.O.; Kroezen, Z.; Philbrick, H.; Poon, R.; Don-Wauchope, A.; Puglia, M.; Wishart, D.; Britz-McKibbin, P. A Cross-Platform Metabolomics Comparison Identifies Serum Metabolite Signatures of Liver Fibrosis Progression in Chronic Hepatitis C Patients. Front. Mol. Biosci. 2021, 8, 676349. [Google Scholar] [CrossRef]
  67. Mahmood, N.; Mihalcioiu, C.; Rabbani, S.A. Multifaceted role of the urokinase-type plasminogen activator (uPA) and its receptor (uPAR): Diagnostic, prognostic, and therapeutic applications. Front. Oncol. 2018, 8, 24. [Google Scholar] [CrossRef] [Green Version]
  68. Moran-Salvador, E.; Mann, J. Epigenetics and Liver Fibrosis. Cell Mol. Gastroenterol. Hepatol. 2017, 4, 125–134. [Google Scholar] [CrossRef] [Green Version]
  69. Akdogan, O.; Atak Yucel, A.; Gok Sargin, Z.; Sonmez, C.; Esendagli Yilmaz, G.; Ozenirler, S. Evaluation of Plasma Urokinase-Type Plasminogen Activator Receptor (UPAR) in Patients With Chronic Hepatitis B, C and Non-Alcoholic Fatty Liver Disease (NAFLD) as Serological Fibrosis Marker. J. Clin. Exp. Hepatol. 2019, 9, 29–33. [Google Scholar] [CrossRef]
  70. Zimmermann, H.W.; Koch, A.; Seidler, S.; Trautwein, C.; Tacke, F. Circulating soluble urokinase plasminogen activator is elevated in patients with chronic liver disease, discriminates stage and aetiology of cirrhosis and predicts prognosis. Liver Int. 2012, 32, 500–509. [Google Scholar] [CrossRef]
  71. Garnaes, E.; Mortensen, C.; Hobolth, L.; Andersen, O.; Nehlin, J.; Moller, S. Kinetics of the soluble urokinase plasminogen activator receptor (suPAR) in cirrhosis. PLoS ONE 2019, 14, e0220697. [Google Scholar] [CrossRef] [Green Version]
  72. Nikkola, A.; Aittoniemi, J.-J.; Huttunen, R.; Sand, J.; Laukkarinen, J. Mo1351 Systemic Levels of Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR) Predict the Severity of Acute Alcohol Pancreatitis. Gastroenterology 2015, 148, S-680. [Google Scholar] [CrossRef]
  73. Chounta, A.; Ellinas, C.; Tzanetakou, V.; Pliarhopoulou, F.; Mplani, V.; Oikonomou, A.; Leventogiannis, K.; Giamarellos-Bourboulis, E.J. Serum soluble urokinase plasminogen activator receptor as a screening test for the early diagnosis of hepatocellular carcinoma. Liver Int. 2015, 35, 601–607. [Google Scholar] [CrossRef] [PubMed]
  74. Radu-Ionita, F.; Pyrsopoulos, N.T.; Jinga, M.; Tintoiu, I.C.; Sun, Z.; Bontas, E. Liver Diseases: A Multidisciplinary Textbook; Springer Nature: Cham, Switzerland, 2020. [Google Scholar]
  75. Eguchi, A.; Wree, A.; Feldstein, A.E. Biomarkers of liver cell death. J. Hepatol. 2014, 60, 1063–1074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zimmermann, H.; Reuken, P.; Koch, A.; Bartneck, M.; Adams, D.; Trautwein, C.; Stallmach, A.; Tacke, F.; Bruns, T. Soluble urokinase plasminogen activator receptor is compartmentally regulated in decompensated cirrhosis and indicates immune activation and short-term mortality. J. Intern. Med. 2013, 274, 86–100. [Google Scholar] [CrossRef] [PubMed]
  77. Nikkola, A.; Aittoniemi, J.; Huttunen, R.; Rajala, L.; Nordback, I.; Sand, J.; Laukkarinen, J. Plasma Level of Soluble Urokinase-type Plasminogen Activator Receptor Predicts the Severity of Acute Alcohol Pancreatitis. Pancreas 2017, 46, 77–82. [Google Scholar] [CrossRef] [Green Version]
  78. Yamada, A.; Arakaki, R.; Saito, M.; Kudo, Y.; Ishimaru, N. Dual role of Fas/FasL-mediated signal in peripheral immune tolerance. Front. Immunol. 2017, 8, 403. [Google Scholar] [CrossRef] [Green Version]
  79. Rada, P.; González-Rodríguez, Á.; García-Monzón, C.; Valverde, Á.M. Understanding lipotoxicity in NAFLD pathogenesis: Is CD36 a key driver? Cell Death Dis. 2020, 11, 1–15. [Google Scholar] [CrossRef]
  80. Li, Z.; Ni, M.; Yu, H.; Wang, L.; Zhou, X.; Chen, T.; Liu, G.; Gao, Y. Gut Microbiota and Liver Fibrosis: One Potential Biomarker for Predicting Liver Fibrosis. Biomed. Res. Int. 2020, 2020, 3905130. [Google Scholar] [CrossRef]
  81. Oh, T.G.; Kim, S.M.; Caussy, C.; Fu, T.; Guo, J.; Bassirian, S.; Singh, S.; Madamba, E.V.; Bettencourt, R.; Richards, L.; et al. A Universal Gut-Microbiome-Derived Signature Predicts Cirrhosis. Cell Metab. 2020, 32, 878–888. [Google Scholar] [CrossRef]
  82. Caussy, C.; Tripathi, A.; Humphrey, G.; Bassirian, S.; Singh, S.; Faulkner, C.; Bettencourt, R.; Rizo, E.; Richards, L.; Xu, Z.Z.; et al. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat. Commun. 2019, 10, 1406. [Google Scholar] [CrossRef]
  83. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Loomba, R.; Seguritan, V.; Li, W.; Long, T.; Klitgord, N.; Bhatt, A.; Dulai, P.S.; Caussy, C.; Bettencourt, R.; Highlander, S.K.; et al. Gut Microbiome-Based Metagenomic Signature for Non-invasive Detection of Advanced Fibrosis in Human Nonalcoholic Fatty Liver Disease. Cell Metab. 2017, 25, 1054–1062. [Google Scholar] [CrossRef] [PubMed]
  85. Porez, G.; Prawitt, J.; Gross, B.; Staels, B. Bile acid receptors as targets for the treatment of dyslipidemia and cardiovascular disease: Thematic review series: New lipid and lipoprotein targets for the treatment of cardiometabolic diseases. J. Lipid Res. 2012, 53, 1723–1737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jiao, Y.; Lu, Y.; Li, X.-y. Farnesoid X receptor: A master regulator of hepatic triglyceride and glucose homeostasis. Acta Pharmacol. Sin. 2015, 36, 44–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. New Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef]
  88. Ratziu, V.; de Guevara, L.; Safadi, R.; Poordad, F.; Fuster, F.; Flores-Figueroa, J.; Arrese, M.; Fracanzani, A.L.; Ben Bashat, D.; Lackner, K. Aramchol in patients with nonalcoholic steatohepatitis: A randomized, double-blind, placebo-controlled phase 2b trial. Nat. Med. 2021, 27, 1825–1835. [Google Scholar] [CrossRef]
  89. Ghonem, N.S.; Assis, D.N.; Boyer, J.L. Fibrates and cholestasis. Hepatology 2015, 62, 635–643. [Google Scholar] [CrossRef] [Green Version]
  90. Guo, Y.-C.; Lu, L.-G. Antihepatic fibrosis drugs in clinical trials. J. Clin. Transl. Hepatol. 2020, 8, 304. [Google Scholar] [CrossRef]
  91. Wettstein, G.; Luccarini, J.M.; Poekes, L.; Faye, P.; Kupkowski, F.; Adarbes, V.; Defrêne, E.; Estivalet, C.; Gawronski, X.; Jantzen, I. The new-generation pan-peroxisome proliferator-activated receptor agonist IVA337 protects the liver from metabolic disorders and fibrosis. Hepatol. Commun. 2017, 1, 524–537. [Google Scholar] [CrossRef]
  92. University Hospital Southampton NHS Foundation Trust; National Institute for Health Research, UK. Treatment of Non Alcoholic Fatty Liver Disease With n-3 Fatty Acids. Available online: https://ClinicalTrials.gov/show/NCT00760513 (accessed on 25 January 2022).
  93. Faghihzadeh, F.; Adibi, P.; Rafiei, R.; Hekmatdoost, A. Resveratrol supplementation improves inflammatory biomarkers in patients with nonalcoholic fatty liver disease. Nutr. Res. 2014, 34, 837–843. [Google Scholar] [CrossRef]
  94. The Effects of Resveratrol Supplement on Biochemical Factors and Hepatic Fibrosis in Patients With Nonalcoholic Steatohepatitis. Available online: https://ClinicalTrials.gov/show/NCT02030977 (accessed on 24 January 2022).
  95. Rinella, M.E.; Dufour, J.-F.; Anstee, Q.M.; Goodman, Z.; Younossi, Z.; Harrison, S.A.; Loomba, R.; Sanyal, A.J.; Bonacci, M.; Trylesinski, A. Non-invasive evaluation of response to obeticholic acid in patients with NASH: Results from the REGENERATE study. J. Hepatol. 2022, 76, 536–548. [Google Scholar] [CrossRef] [PubMed]
  96. Ratziu, V.; Sanyal, A.J.; Loomba, R.; Rinella, M.; Harrison, S.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; MacConell, L.; Shringarpure, R. REGENERATE: Design of a pivotal, randomised, phase 3 study evaluating the safety and efficacy of obeticholic acid in patients with fibrosis due to nonalcoholic steatohepatitis. Contemp. Clin. Trials 2019, 84, 105803. [Google Scholar] [CrossRef] [Green Version]
  97. Flint, A.; Andersen, G.; Hockings, P.; Johansson, L.; Morsing, A.; Sundby Palle, M.; Vogl, T.; Loomba, R.; Plum-Mörschel, L. Randomised clinical trial: Semaglutide versus placebo reduced liver steatosis but not liver stiffness in subjects with non-alcoholic fatty liver disease assessed by magnetic resonance imaging. Aliment. Pharmacol. Ther. 2021, 54, 1150–1161. [Google Scholar] [CrossRef] [PubMed]
  98. Shim, K.Y.; Eom, Y.W.; Kim, M.Y.; Kang, S.H.; Baik, S.K. Role of the renin-angiotensin system in hepatic fibrosis and portal hypertension. Korean J. Intern. Med. 2018, 33, 453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Colmenero, J.; Bataller, R.; Sancho-Bru, P.; Domínguez, M.; Moreno, M.; Forns, X.; Bruguera, M.; Arroyo, V.; Brenner, D.A.; Ginès, P. Effects of losartan on hepatic expression of nonphagocytic NADPH oxidase and fibrogenic genes in patients with chronic hepatitis C. Am. J. Physiol. -Gastrointest. Liver Physiol. 2009, 297, G726–G734. [Google Scholar] [CrossRef] [Green Version]
  100. Effects of Losartan on Hepatic Fibrogenesis in Chronic Hepatitis C. Available online: https://ClinicalTrials.gov/show/NCT00298714 (accessed on 13 February 2022).
  101. Yonsei, U. Effect of Candesartan in Alcoholic Liver Fibrosis. Available online: https://ClinicalTrials.gov/show/NCT00990639 (accessed on 13 February 2022).
  102. Sherief, A.-E.; Tanta, U. Effect of Some Drugs on Liver Fibrosis. Available online: https://ClinicalTrials.gov/show/NCT03770936 (accessed on 13 February 2022).
  103. Evaluation of Irbesartan on Hepatic Fibrosis in Chronic Hepatitis C. Available online: https://ClinicalTrials.gov/show/NCT00265642 (accessed on 22 February 2022).
  104. Tan, Z.; Sun, H.; Xue, T.; Gan, C.; Liu, H.; Xie, Y.; Yao, Y.; Ye, T. Liver Fibrosis: Therapeutic Targets and Advances in Drug Therapy. Front. Cell Dev. Biol. 2021, 9, 730176. [Google Scholar] [CrossRef] [PubMed]
  105. Jang, Y.O.; Kim, S.H.; Cho, M.-Y.; Kim, K.S.; Park, K.-S.; Cha, S.-K.; Kim, M.Y.; Chang, S.J.; Baik, S.K. Synergistic effects of simvastatin and bone marrow-derived mesenchymal stem cells on hepatic fibrosis. Biochem. Biophys. Res. Commun. 2018, 497, 264–271. [Google Scholar] [CrossRef]
  106. Abraldes, J.G.; Albillos, A.; Bañares, R.; Turnes, J.; González, R.; García–Pagán, J.C.; Bosch, J. Simvastatin lowers portal pressure in patients with cirrhosis and portal hypertension: A randomized controlled trial. Gastroenterology 2009, 136, 1651–1658. [Google Scholar] [CrossRef]
  107. Efficacy of Simvastatin in Alcoholic Liver Fibrosis. Available online: https://ClinicalTrials.gov/show/NCT04971577 (accessed on 13 February 2022).
  108. Merck, S.; Dohme, C. Prevention of Disease Progress in Chronic Hepatitis C Patients With Liver Fibrosis (Study P02570AM2)(COMPLETED). Available online: https://ClinicalTrials.gov/show/NCT00049842 (accessed on 13 February 2022).
  109. Merck, S.; Dohme, C. PEG-Intron Plus Rebetol Treatment of Chronic Hepatitis C Subjects Who Failed Response to Alpha-Interferon Plus Ribavirin (Study P02370). 2007. Available online: https://clinicaltrials.gov/ct2/show/NCT00039871 (accessed on 13 January 2022).
  110. RWTH Aachen University; Hannover Medical School. Induction of Fibrosis Regression on Patients With Chronic Hepatitis B Infection. Available online: https://ClinicalTrials.gov/show/NCT01341106 (accessed on 13 January 2022).
  111. Chang, T.T.; Liaw, Y.F.; Wu, S.S.; Schiff, E.; Han, K.H.; Lai, C.L.; Safadi, R.; Lee, S.S.; Halota, W.; Goodman, Z. Long-term entecavir therapy results in the reversal of fibrosis/cirrhosis and continued histological improvement in patients with chronic hepatitis B. Hepatology 2010, 52, 886–893. [Google Scholar] [CrossRef]
  112. Peking University People’s Hospital; RenJi Hospital; Peking University; Shanghai Zhongshan Hospital; Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Public Health Clinical Center; Nanfang Hospital of Southern Medical University; Sir Run Run Shaw Hospital; Beijing YouAn Hospital; Peking University First Hospital; et al. Optimized Treatment and Regression of HBV-induced Liver Fibrosis. Available online: https://ClinicalTrials.gov/show/NCT01938781 (accessed on 13 February 2022).
  113. Judit Pich, Institut d’Investigacions Biomèdiques August Pi i Sunyer. Efficacy of the Combination of Simvastatin Plus Rifaximin in Patients With Decompensated Cirrhosis to Prevent ACLF Development. Available online: https://ClinicalTrials.gov/show/NCT03780673 (accessed on 22 February 2022).
  114. Popov, Y.; Schuppan, D. Targeting liver fibrosis: Strategies for development and validation of antifibrotic therapies. Hepatology 2009, 50, 1294–1306. [Google Scholar] [CrossRef]
  115. George, J.; Roulot, D.; Koteliansky, V.E.; Bissell, D.M. In vivo inhibition of rat stellate cell activation by soluble transforming growth factor β type II receptor: A potential new therapy for hepatic fibrosis. Proc. Natl. Acad. Sci. USA 1999, 96, 12719–12724. [Google Scholar] [CrossRef] [Green Version]
  116. Zhang, S.; Gong, Y.; Xiao, J.; Chai, Y.; Lei, J.; Huang, H.; Xiang, T.; Shen, W. A COL1A1 promoter-controlled expression of TGF-β soluble receptor inhibits hepatic fibrosis without triggering autoimmune responses. Dig. Dis. Sci. 2018, 63, 2662–2672. [Google Scholar] [CrossRef] [PubMed]
  117. Biswas, S.; Guix, M.; Rinehart, C.; Dugger, T.C.; Chytil, A.; Moses, H.L.; Freeman, M.L.; Arteaga, C.L. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Investig. 2007, 117, 1305–1313. [Google Scholar] [CrossRef] [Green Version]
  118. Kemaladewi, D.U.; Pasteuning, S.; Van Der Meulen, J.W.; Van Heiningen, S.H.; van Ommen, G.-J.; Ten Dijke, P.; Aartsma-Rus, A.; Ac’t Hoen, P.; Hoogaars, W.M. Targeting TGF-β signaling by antisense oligonucleotide-mediated knockdown of TGF-β type I receptor. Mol. Ther. Nucleic Acids 2014, 3, e156. [Google Scholar] [CrossRef] [PubMed]
  119. Akhurst, R.J.; Hata, A. Targeting the TGFβ signalling pathway in disease. Nat. Rev. Drug Discov. 2012, 11, 790–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Denton, C.P.; Merkel, P.A.; Furst, D.E.; Khanna, D.; Emery, P.; Hsu, V.M.; Silliman, N.; Streisand, J.; Powell, J.; Åkesson, A. Recombinant human anti–transforming growth factor β1 antibody therapy in systemic sclerosis: A multicenter, randomized, placebo-controlled phase I/II trial of CAT-192. Arthritis Rheum. 2007, 56, 323–333. [Google Scholar] [CrossRef] [PubMed]
  121. Zhang, X.; Yun, J.S.; Han, D.; Yook, J.I.; Kim, H.S.; Cho, E.S. TGF-β pathway in salivary gland fibrosis. Int. J. Mol. Sci. 2020, 21, 9138. [Google Scholar] [CrossRef] [PubMed]
  122. Muzzillo, D.; Imoto, M.; Fukuda, Y.; Koyama, Y.; Saga, S.; Nagai, Y.; Hayakawa, T. Clinical evaluation of serum tissue inhibitor of metalloproteinases-1 levels in patients with liver diseases. J. Gastroenterol. Hepatol. 1993, 8, 437–441. [Google Scholar] [CrossRef]
  123. Roderfeld, M.; Weiskirchen, R.; Wagner, S.; Berres, M.L.; Henkel, C.; Grötzinger, J.; Gressner, A.M.; Matern, S.; Roeb, E. Inhibition of hepatic fibrogenesis by matrix metalloproteinase-9 mutants in mice. FASEB J. 2006, 20, 444–454. [Google Scholar] [CrossRef]
  124. Breuss, J.; Gallo, J.; DeLisser, H.; Klimanskaya, I.; Folkesson, H.; Pittet, J.; Nishimura, S.; Aldape, K.; Landers, D.; Carpenter, W. Expression of the beta 6 integrin subunit in development, neoplasia and tissue repair suggests a role in epithelial remodeling. J. Cell Sci. 1995, 108, 2241–2251. [Google Scholar] [CrossRef]
  125. Biogen STX-100 in Patients With Idiopathic Pulmonary Fibrosis (IPF). Available online: https://ClinicalTrials.gov/show/NCT01371305 (accessed on 22 February 2022).
  126. Patsenker, E.; Popov, Y.; Stickel, F.; Jonczyk, A.; Goodman, S.L.; Schuppan, D. Inhibition of integrin αvβ6 on cholangiocytes blocks transforming growth factor-β activation and retards biliary fibrosis progression. Gastroenterology 2008, 135, 660–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Chen, G.; Zhang, L.; Chen, L.; Wang, H.; Zhang, Y.; Bie, P. Role of integrin αvβ6 in the pathogenesis of ischemia-related biliary fibrosis after liver transplantation. Transplantation 2013, 95, 1092–1099. [Google Scholar] [CrossRef] [PubMed]
  128. Ning, B.-F.; Ding, J.; Yin, C.; Zhong, W.; Wu, K.; Zeng, X.; Yang, W.; Chen, Y.-X.; Zhang, J.-P.; Zhang, X. Hepatocyte nuclear factor 4α suppresses the development of hepatocellular carcinoma. Cancer Res. 2010, 70, 7640–7651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Yue, H.-Y.; Yin, C.; Hou, J.-L.; Zeng, X.; Chen, Y.-X.; Zhong, W.; Hu, P.-F.; Deng, X.; Tan, Y.; Zhang, J. Hepatocyte nuclear factor 4α attenuates hepatic fibrosis in rats. Gut 2010, 59, 236–246. [Google Scholar] [CrossRef] [PubMed]
  130. Weber, S.N.; Bohner, A.; Dapito, D.H.; Schwabe, R.F.; Lammert, F. TLR4 deficiency protects against hepatic fibrosis and diethylnitrosamine-induced pre-carcinogenic liver injury in fibrotic liver. PLoS ONE 2016, 11, e0158819. [Google Scholar] [CrossRef]
  131. Kanzler, H.; Barrat, F.J.; Hessel, E.M.; Coffman, R.L. Therapeutic targeting of innate immunity with Toll-like receptor agonists and antagonists. Nat. Med. 2007, 13, 552–559. [Google Scholar] [CrossRef]
  132. Yang, T.; Poenisch, M.; Khanal, R.; Hu, Q.; Dai, Z.; Li, R.; Song, G.; Yuan, Q.; Yao, Q.; Shen, X. Therapeutic HNF4A mRNA attenuates liver fibrosis in a preclinical model. J. Hepatol. 2021, 75, 1420–1433. [Google Scholar] [CrossRef]
  133. Palmer, S.M.; Snyder, L.; Todd, J.L.; Soule, B.; Christian, R.; Anstrom, K.; Luo, Y.; Gagnon, R.; Rosen, G. Randomized, double-blind, placebo-controlled, phase 2 trial of BMS-986020, a lysophosphatidic acid receptor antagonist for the treatment of idiopathic pulmonary fibrosis. Chest 2018, 154, 1061–1069. [Google Scholar] [CrossRef] [Green Version]
  134. Mills, G.B.; Moolenaar, W.H. The emerging role of lysophosphatidic acid in cancer. Nat. Rev. Cancer 2003, 3, 582–591. [Google Scholar] [CrossRef]
  135. Xiang, H.; Lu, Y.; Shao, M.; Wu, T. Lysophosphatidic acid receptors: Biochemical and clinical implications in different diseases. J. Cancer 2020, 11, 3519. [Google Scholar] [CrossRef] [Green Version]
  136. Swaney, J.; Chapman, C.; Correa, L.; Stebbins, K.; Bundey, R.; Prodanovich, P.; Fagan, P.; Baccei, C.; Santini, A.; Hutchinson, J. A novel, orally active LPA1 receptor antagonist inhibits lung fibrosis in the mouse bleomycin model. Br. J. Pharmacol. 2010, 160, 1699–1713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Mazzocca, A.; Dituri, F.; Lupo, L.; Quaranta, M.; Antonaci, S.; Giannelli, G. Tumor-secreted lysophostatidic acid accelerates hepatocellular carcinoma progression by promoting differentiation of peritumoral fibroblasts in myofibroblasts. Hepatology 2011, 54, 920–930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Nakamura, T.; Mizuno, S. The discovery of hepatocyte growth factor (HGF) and its significance for cell biology, life sciences and clinical medicine. Proc. Jpn. Acad. Ser. B 2010, 86, 588–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Mizuno, S.; Matsumoto, K.; Nakamura, T. HGF as a renotrophic and anti-fibrotic regulator in chronic renal disease. Front. Biosci. 2008, 13, 7072–7086. [Google Scholar] [CrossRef] [Green Version]
  140. Mizuno, S.; Nakamura, T. Hepatocyte growth factor: A regenerative drug for acute hepatitis and liver cirrhosis. Regen. Med. 2007, 2, 161–170. [Google Scholar] [CrossRef]
  141. Mizuno, S.; Nakamura, T. Suppressions of chronic glomerular injuries and TGF-β1 production by HGF in attenuation of murine diabetic nephropathy. Am. J. Physiol. -Ren. Physiol. 2004, 286, F134–F143. [Google Scholar] [CrossRef] [Green Version]
  142. Nakamura, T.; Matsumoto, K.; Mizuno, S.; Sawa, Y.; Matsuda, H.; Nakamura, T. Hepatocyte growth factor prevents tissue fibrosis, remodeling, and dysfunction in cardiomyopathic hamster hearts. Am. J. Physiol. -Heart Circ. Physiol. 2005, 288, H2131–H2139. [Google Scholar] [CrossRef] [Green Version]
  143. Xu, F.; Liu, C.; Zhou, D.; Zhang, L. TGF-β/SMAD pathway and its regulation in hepatic fibrosis. J. Histochem. Cytochem. 2016, 64, 157–167. [Google Scholar] [CrossRef]
  144. Mizuno, S.; Matsumoto, K.; Li, M.-Y.; Nakamura, T. HGF reduces advancing lung fibrosis in mice: A potential role for MMP-dependent myofibroblast apoptosis. FASEB J. 2005, 19, 1–18. [Google Scholar] [CrossRef]
  145. Kim, W.-H.; Matsumoto, K.; Bessho, K.; Nakamura, T. Growth inhibition and apoptosis in liver myofibroblasts promoted by hepatocyte growth factor leads to resolution from liver cirrhosis. Am. J. Pathol. 2005, 166, 1017–1028. [Google Scholar] [CrossRef] [Green Version]
  146. Ishikawa, T.; Factor, V.M.; Marquardt, J.U.; Raggi, C.; Seo, D.; Kitade, M.; Conner, E.A.; Thorgeirsson, S.S. Hepatocyte growth factor/c-met signaling is required for stem-cell–mediated liver regeneration in mice. Hepatology 2012, 55, 1215–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Rachfal, A.W.; Brigstock, D.R. Connective tissue growth factor (CTGF/CCN2) in hepatic fibrosis. Hepatol. Res. 2003, 26, 1–9. [Google Scholar] [CrossRef]
  148. Tong, Z.; Chen, R.; Alt, D.S.; Kemper, S.; Perbal, B.; Brigstock, D.R. Susceptibility to liver fibrosis in mice expressing a connective tissue growth factor transgene in hepatocytes. Hepatology 2009, 50, 939–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Eitner, F.; Bücher, E.; van Roeyen, C.; Kunter, U.; Rong, S.; Seikrit, C.; Villa, L.; Boor, P.; Fredriksson, L.; Bäckström, G. PDGF-C is a proinflammatory cytokine that mediates renal interstitial fibrosis. J. Am. Soc. Nephrol. 2008, 19, 281–289. [Google Scholar] [CrossRef] [Green Version]
  150. Pontén, A.; Li, X.; Thorén, P.; Aase, K.; Sjöblom, T.; Östman, A.; Eriksson, U. Transgenic overexpression of platelet-derived growth factor-C in the mouse heart induces cardiac fibrosis, hypertrophy, and dilated cardiomyopathy. Am. J. Pathol. 2003, 163, 673–682. [Google Scholar] [CrossRef] [Green Version]
  151. Li, X.; Pontén, A.; Aase, K.; Karlsson, L.; Abramsson, A.; Uutela, M.; Bäckström, G.; Hellström, M.; Boström, H.; Li, H. PDGF-C is a new protease-activated ligand for the PDGF α-receptor. Nat. Cell Biol. 2000, 2, 302–309. [Google Scholar] [CrossRef]
  152. Campbell, J.S.; Hughes, S.D.; Gilbertson, D.G.; Palmer, T.E.; Holdren, M.S.; Haran, A.C.; Odell, M.M.; Bauer, R.L.; Ren, H.-P.; Haugen, H.S. Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2005, 102, 3389–3394. [Google Scholar] [CrossRef] [Green Version]
  153. Tang, N.; Zhang, Y.-P.; Ying, W.; Yao, X.-X. Interleukin-1β upregulates matrix metalloproteinase-13 gene expression via c-Jun N-terminal kinase and p38 MAPK pathways in rat hepatic stellate cells. Mol. Med. Rep. 2013, 8, 1861–1865. [Google Scholar] [CrossRef]
  154. Jain, M.K.; Adams-Huet, B.; Terekhova, D.; Kushner, L.E.; Bedimo, R.; Li, X.; Holodniy, M. Acute and chronic immune biomarker changes during interferon/ribavirin treatment in HIV/HCV co-infected patients. J. Viral Hepat. 2015, 22, 25–36. [Google Scholar] [CrossRef] [Green Version]
  155. Okada, H.; Honda, M.; Campbell, J.S.; Sakai, Y.; Yamashita, T.; Takebuchi, Y.; Hada, K.; Shirasaki, T.; Takabatake, R.; Nakamura, M. Acyclic retinoid targets platelet-derived growth factor signaling in the prevention of hepatic fibrosis and hepatocellular carcinoma development. Cancer Res. 2012, 72, 4459–4471. [Google Scholar] [CrossRef] [Green Version]
  156. Wick, G.; Backovic, A.; Rabensteiner, E.; Plank, N.; Schwentner, C.; Sgonc, R. The immunology of fibrosis: Innate and adaptive responses. Trends Immunol. 2010, 31, 110–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. McQuitty, C.E.; Williams, R.; Chokshi, S.; Urbani, L. Immunomodulatory role of the extracellular matrix within the liver disease microenvironment. Front. Immunol. 2020, 11, 2903. [Google Scholar] [CrossRef] [PubMed]
  158. Seki, E.; Schwabe, R.F. Hepatic inflammation and fibrosis: Functional links and key pathways. Hepatology 2015, 61, 1066–1079. [Google Scholar] [CrossRef] [PubMed]
  159. Tsuchida, T.; Friedman, S.L. Mechanisms of hepatic stellate cell activation. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 397–411. [Google Scholar] [CrossRef]
  160. Lee, T.H.; Klampfer, L.; Shows, T.; Vilcek, J. Transcriptional regulation of TSG6, a tumor necrosis factor-and interleukin-1-inducible primary response gene coding for a secreted hyaluronan-binding protein. J. Biol. Chem. 1993, 268, 6154–6160. [Google Scholar] [CrossRef]
  161. Nentwich, H.A.; Mustafa, Z.; Rugg, M.S.; Marsden, B.D.; Cordell, M.R.; Mahoney, D.J.; Jenkins, S.C.; Dowling, B.; Fries, E.; Milner, C.M. A novel allelic variant of the human TSG-6 gene encoding an amino acid difference in the CUB module: Chromosomal localization, frequency analysis, modeling, and expression. J. Biol. Chem. 2002, 277, 15354–15362. [Google Scholar] [CrossRef]
  162. Lee, T.H.; Lee, G.W.; Ziff, E.B.; Vilcek, J. Isolation and characterization of eight tumor necrosis factor-induced gene sequences from human fibroblasts. Mol. Cell. Biol. 1990, 10, 1982–1988. [Google Scholar]
  163. Lee, T.H.; Wisniewski, H.-G.; Vilcek, J. A novel secretory tumor necrosis factor-inducible protein (TSG-6) is a member of the family of hyaluronate binding proteins, closely related to the adhesion receptor CD44. J. Cell Biol. 1992, 116, 545–557. [Google Scholar] [CrossRef] [Green Version]
  164. Glant, T.T.; Kamath, R.V.; Bárdos, T.; Gál, I.; Szántó, S.; Murad, Y.M.; Sandy, J.D.; Mort, J.S.; Roughley, P.J.; Mikecz, K. Cartilage-specific constitutive expression of TSG-6 protein (product of tumor necrosis factor α–stimulated gene 6) provides a chondroprotective, but not antiinflammatory, effect in antigen-induced arthritis. Arthritis Rheum. 2002, 46, 2207–2218. [Google Scholar] [CrossRef]
  165. Milner, C.M.; Day, A.J. TSG-6: A multifunctional protein associated with inflammation. J. Cell Sci. 2003, 116, 1863–1873. [Google Scholar] [CrossRef] [Green Version]
  166. Prockop, D.J.; Oh, J.Y. Mesenchymal stem/stromal cells (MSCs): Role as guardians of inflammation. Mol. Ther. 2012, 20, 14–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Wang, S.; Kim, J.; Lee, C.; Oh, D.; Han, J.; Kim, T.-J.; Kim, S.-W.; Seo, Y.-S.; Oh, S.-h.; Jung, Y. Tumor necrosis factor-inducible gene 6 reprograms hepatic stellate cells into stem-like cells, which ameliorates liver damage in mouse. Biomaterials 2019, 219, 119375. [Google Scholar] [CrossRef] [PubMed]
  168. Wan, Y.-M.; Wu, H.-M.; Li, Y.-H.; Xu, Z.-Y.; Yang, J.-H.; Liu, C.; He, Y.-F.; Wang, M.-J.; Wu, X.-N.; Zhang, Y. TSG-6 Inhibits Oxidative Stress and Induces M2 Polarization of Hepatic Macrophages in Mice With Alcoholic Hepatitis via Suppression of STAT3 Activation. Front. Pharmacol. 2020, 11, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Kumari, R.; Jat, P. Mechanisms of cellular senescence: Cell cycle arrest and senescence associated secretory phenotype. Front. Cell Dev. Biol. 2021, 9, 485. [Google Scholar] [CrossRef] [PubMed]
  170. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Kim, K.M.; Noh, J.H.; Bodogai, M.; Martindale, J.L.; Yang, X.; Indig, F.E.; Basu, S.K.; Ohnuma, K.; Morimoto, C.; Johnson, P.F. Identification of senescent cell surface targetable protein DPP4. Genes Dev. 2017, 31, 1529–1534. [Google Scholar] [CrossRef] [Green Version]
  172. Levi, N.; Papismadov, N.; Solomonov, I.; Sagi, I.; Krizhanovsky, V. The ECM path of senescence in aging: Components and modifiers. FEBS J. 2020, 287, 2636–2646. [Google Scholar] [CrossRef]
  173. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
  174. Antonangeli, F.; Zingoni, A.; Soriani, A.; Santoni, A. Senescent cells: Living or dying is a matter of NK cells. J. Leukoc. Biol. 2019, 105, 1275–1283. [Google Scholar] [CrossRef]
  175. Rossi, M.; Abdelmohsen, K. The Emergence of Senescent Surface Biomarkers as Senotherapeutic Targets. Cells 2021, 10, 1740. [Google Scholar] [CrossRef]
  176. Weiner, G.J. Building better monoclonal antibody-based therapeutics. Nat. Rev. Cancer 2015, 15, 361–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Benson, D.M.; Bakan, C.E.; Mishra, A.; Hofmeister, C.C.; Efebera, Y.; Becknell, B.; Baiocchi, R.A.; Zhang, J.; Yu, J.; Smith, M.K. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: A therapeutic target for CT-011, a novel monoclonal anti–PD-1 antibody. Blood 2010, 116, 2286–2294. [Google Scholar] [CrossRef] [PubMed]
  178. Hammerich, L.; Tacke, F. Interleukins in chronic liver disease: Lessons learned from experimental mouse models. Clin. Exp. Gastroenterol. 2014, 7, 297. [Google Scholar] [PubMed] [Green Version]
  179. Narayanan, S.; Surette, F.A.; Hahn, Y.S. The immune landscape in nonalcoholic steatohepatitis. Immune Netw. 2016, 16, 147–158. [Google Scholar] [CrossRef] [Green Version]
  180. Wang, K.S.; Ritz, J.; Frank, D.A. IL-2 induces STAT4 activation in primary NK cells and NK cell lines, but not in T cells. J. Immunol. 1999, 162, 299–304. [Google Scholar]
  181. Kovalovich, K.; DeAngelis, R.A.; Li, W.; Furth, E.E.; Ciliberto, G.; Taub, R. Increased toxin-induced liver injury and fibrosis in interleukin-6–deficient mice. Hepatology 2000, 31, 149–159. [Google Scholar] [CrossRef]
  182. Thompson, K.; Maltby, J.; Fallowfield, J.; McAulay, M.; Millward-Sadler, H.; Sheron, N. Interleukin-10 expression and function in experimental murine liver inflammation and fibrosis. Hepatology 1998, 28, 1597–1606. [Google Scholar] [CrossRef]
  183. Louis, H.; Van Laethem, J.L.; Wu, W.; Quertinmont, E.; Degraef, C.; Van den Berg, K.; Demols, A.; Goldman, M.; Le Moine, O.; Geerts, A. Interleukin-10 controls neutrophilic infiltration, hepatocyte proliferation, and liver fibrosis induced by carbon tetrachloride in mice. Hepatology 1998, 28, 1607–1615. [Google Scholar] [CrossRef]
  184. Hoffmann, K.F.; Caspar, P.; Cheever, A.W.; Wynn, T.A. IFN-γ, IL-12, and TNF-α are required to maintain reduced liver pathology in mice vaccinated with Schistosoma mansoni eggs and IL-12. J. Immunol. 1998, 161, 4201–4210. [Google Scholar]
  185. Parihar, R.; Dierksheide, J.; Hu, Y.; Carson, W.E. IL-12 enhances the natural killer cell cytokine response to Ab-coated tumor cells. J. Clin. Investig. 2002, 110, 983–992. [Google Scholar] [CrossRef]
  186. Choi, S.S.; Chhabra, V.S.; Nguyen, Q.H.; Ank, B.J.; Stiehm, E.R.; Roberts, R.L. Interleukin-15 enhances cytotoxicity, receptor expression, and expansion of neonatal natural killer cells in long-term culture. Clin. Diagn. Lab. Immunol. 2004, 11, 879–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Ki, S.H.; Park, O.; Zheng, M.; Morales-Ibanez, O.; Kolls, J.K.; Bataller, R.; Gao, B. Interleukin-22 treatment ameliorates alcoholic liver injury in a murine model of chronic-binge ethanol feeding: Role of signal transducer and activator of transcription 3. Hepatology 2010, 52, 1291–1300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Yang, L.; Zhang, Y.; Wang, L.; Fan, F.; Zhu, L.; Li, Z.; Ruan, X.; Huang, H.; Wang, Z.; Huang, Z. Amelioration of high fat diet induced liver lipogenesis and hepatic steatosis by interleukin-22. J. Hepatol. 2010, 53, 339–347. [Google Scholar] [CrossRef] [PubMed]
  189. Fabre, T.; Molina, M.F.; Soucy, G.; Goulet, J.-P.; Willems, B.; Villeneuve, J.-P.; Bilodeau, M.; Shoukry, N.H. Type 3 cytokines IL-17A and IL-22 drive TGF-β–dependent liver fibrosis. Sci. Immunol. 2018, 3, eaar7754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Mitra, A.; Satelli, A.; Yan, J.; Xueqing, X.; Gagea, M.; Hunter, C.A.; Mishra, L.; Li, S. IL-30 (IL27p28) attenuates liver fibrosis through inducing NKG2D-rae1 interaction between NKT and activated hepatic stellate cells in mice. Hepatology 2014, 60, 2027–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Van Caam, A.; Vonk, M.; van den Hoogen, F.; van Lent, P.; van der Kraan, P. Unraveling SSc Pathophysiology; The Myofibroblast. Front. Immunol. 2018, 9, 2452. [Google Scholar] [CrossRef] [Green Version]
  192. Fedarko, N.S.; Pacocha, S.E.; Huang, S.-K.; Lichtenstein, L.M.; Essayan, D.M. Interleukin-13 modulates collagen homeostasis in human skin and keloid fibroblasts. J. Pharmacol. Exp. Ther. 2000, 292, 988–994. [Google Scholar]
  193. McGee, H.M.; Schmidt, B.A.; Booth, C.J.; Yancopoulos, G.D.; Valenzuela, D.M.; Murphy, A.J.; Stevens, S.; Flavell, R.A.; Horsley, V. IL-22 promotes fibroblast-mediated wound repair in the skin. J. Investig. Dermatol. 2013, 133, 1321–1329. [Google Scholar] [CrossRef] [Green Version]
  194. Hashimoto, S.; Gon, Y.; Takeshita, I.; Maruoka, S.; Horie, T. IL-4 and IL-13 induce myofibroblastic phenotype of human lung fibroblasts through c-Jun NH2-terminal kinase–dependent pathway. J. Allergy Clin. Immunol. 2001, 107, 1001–1008. [Google Scholar] [CrossRef]
  195. Theiss, A.L.; Simmons, J.G.; Jobin, C.; Lund, P.K. Tumor necrosis factor (TNF) alpha increases collagen accumulation and proliferation in intestinal myofibroblasts via TNF receptor 2. J. Biol. Chem. 2005, 280, 36099–36109. [Google Scholar] [CrossRef] [Green Version]
  196. Motz, K.; Samad, I.; Yin, L.X.; Murphy, M.K.; Duvvuri, M.; Ding, D.; Hillel, A.T. Interferon-γ treatment of human laryngotracheal stenosis–derived fibroblasts. JAMA Otolaryngol. Head Neck Surg. 2017, 143, 1134–1140. [Google Scholar] [CrossRef]
  197. Rosenbloom, J.; Feldman, G.; Freundlich, B.; Jimenez, S.A. Inhibition of excessive scleroderma fibroblast collagen production by recombinant γ-interferon: Association with a coordinate decrease in types I and III procollagen messenger RNA levels. Arthritis Rheum. Off. J. Am. Coll. Rheumatol. 1986, 29, 851–856. [Google Scholar] [CrossRef] [PubMed]
  198. Saito, A.; Okazaki, H.; Sugawara, I.; Yamamoto, K.; Takizawa, H. Potential action of IL-4 and IL-13 as fibrogenic factors on lung fibroblasts in vitro. Int. Arch. Allergy Immunol. 2003, 132, 168–176. [Google Scholar] [CrossRef] [PubMed]
  199. Berraondo, P.; Sanmamed, M.F.; Ochoa, M.C.; Etxeberria, I.; Aznar, M.A.; Pérez-Gracia, J.L.; Rodríguez-Ruiz, M.E.; Ponz-Sarvise, M.; Castañón, E.; Melero, I. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 2019, 120, 6–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Levin, A.M.; Bates, D.L.; Ring, A.M.; Krieg, C.; Lin, J.T.; Su, L.; Moraga, I.; Raeber, M.E.; Bowman, G.R.; Novick, P. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature 2012, 484, 529–533. [Google Scholar] [CrossRef] [Green Version]
  201. Dubois, S.; Patel, H.J.; Zhang, M.; Waldmann, T.A.; Müller, J.R. Preassociation of IL-15 with IL-15Rα-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44high T cells and its antitumor action. J. Immunol. 2008, 180, 2099–2106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Lehmann, D.; Spanholtz, J.; Sturtzel, C.; Tordoir, M.; Schlechta, B.; Groenewegen, D.; Hofer, E. IL-12 directs further maturation of ex vivo differentiated NK cells with improved therapeutic potential. PLoS ONE 2014, 9, e87131. [Google Scholar] [CrossRef] [Green Version]
  203. Wasmuth, H. Chemokines as inflammatory mediators of fibrosis in liver fibrosis. J. Transl. Med. 2010, 8, I13. [Google Scholar] [CrossRef] [Green Version]
  204. Lacotte, S.; Brun, S.; Muller, S.; Dumortier, H. CXCR3, Inflammation, and autoimmune diseases. Contemp. Challenge Autoimmun. 2009, 1173, 310. [Google Scholar] [CrossRef]
  205. Groover, M.K.; Richmond, J.M. Potential therapeutic manipulations of the CXCR3 chemokine axis for the treatment of inflammatory fibrosing diseases. F1000Research 2020, 9, 1197. [Google Scholar] [CrossRef]
  206. Jiang, X.-P.; Ai, W.-B.; Wan, L.-Y.; Zhang, Y.-Q.; Wu, J.-F. The roles of microRNA families in hepatic fibrosis. Cell Biosci. 2017, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Jiang, H.; Zhang, G.; Wu, J.-H.; Jiang, C.-P. Diverse roles of miR-29 in cancer. Oncol. Rep. 2014, 31, 1509–1516. [Google Scholar] [CrossRef] [Green Version]
  208. Kitano, M.; Bloomston, P.M. Hepatic stellate cells and microRNAs in pathogenesis of liver fibrosis. J. Clin. Med. 2016, 5, 38. [Google Scholar] [CrossRef] [PubMed]
  209. Bandyopadhyay, S.; Friedman, R.C.; Marquez, R.T.; Keck, K.; Kong, B.; Icardi, M.S.; Brown, K.E.; Burge, C.B.; Schmidt, W.N.; Wang, Y. Hepatitis C virus infection and hepatic stellate cell activation downregulate miR-29: miR-29 overexpression reduces hepatitis C viral abundance in culture. J. Infect. Dis. 2011, 203, 1753–1762. [Google Scholar] [CrossRef] [PubMed]
  210. Kwiecinski, M.; Noetel, A.; Elfimova, N.; Trebicka, J.; Schievenbusch, S.; Strack, I.; Molnar, L.; von Brandenstein, M.; Töx, U.; Nischt, R. Hepatocyte growth factor (HGF) inhibits collagen I and IV synthesis in hepatic stellate cells by miRNA-29 induction. PLoS ONE 2011, 6, e24568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Ogawa, T.; Iizuka, M.; Sekiya, Y.; Yoshizato, K.; Ikeda, K.; Kawada, N. Suppression of type I collagen production by microRNA-29b in cultured human stellate cells. Biochem. Biophys. Res. Commun. 2010, 391, 316–321. [Google Scholar] [CrossRef]
  212. Huang, Y.-H.; Tiao, M.-M.; Huang, L.-T.; Chuang, J.-H.; Kuo, K.-C.; Yang, Y.-L.; Wang, F.-S. Activation of Mir-29a in activated hepatic stellate cells modulates its profibrogenic phenotype through inhibition of histone deacetylases 4. PLoS ONE 2015, 10, e0136453. [Google Scholar] [CrossRef] [Green Version]
  213. Zhang, Y.; Ghazwani, M.; Li, J.; Sun, M.; Stolz, D.B.; He, F.; Fan, J.; Xie, W.; Li, S. MiR-29b inhibits collagen maturation in hepatic stellate cells through down-regulating the expression of HSP47 and lysyl oxidase. Biochem. Biophys. Res. Commun. 2014, 446, 940–944. [Google Scholar] [CrossRef] [Green Version]
  214. Mei, Z.; Su, T.; Ye, J.; Yang, C.; Zhang, S.; Xie, C. The miR-15 family enhances the radiosensitivity of breast cancer cells by targeting G2 checkpoints. Radiat. Res. 2015, 183, 196–207. [Google Scholar] [CrossRef]
  215. Zhu, K.; He, Y.; Xia, C.; Yan, J.; Hou, J.; Kong, D.; Yang, Y.; Zheng, G. MicroRNA-15a inhibits proliferation and induces apoptosis in CNE1 nasopharyngeal carcinoma cells. Oncol. Res. 2016, 24, 145. [Google Scholar] [CrossRef]
  216. Wang, X.; He, Y.; Mackowiak, B.; Gao, B. MicroRNAs as regulators, biomarkers and therapeutic targets in liver diseases. Gut 2021, 70, 784–795. [Google Scholar] [CrossRef] [PubMed]
  217. Johnson, C.; Drummer IV, C.; Virtue, A.; Gao, T.; Wu, S.; Hernandez, M.; Singh, L.; Wang, H.; Yang, X.-F. Increased expression of resistin in microRNA-155-deficient white adipose tissues may be a possible driver of metabolically healthy obesity transition to classical obesity. Front. Physiol. 2018, 9, 1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Yan, G.; Li, B.; Xin, X.; Xu, M.; Ji, G.; Yu, H. MicroRNA-34a promotes hepatic stellate cell activation via targeting ACSL1. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2015, 21, 3008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Yuan, K.; Zhang, X.; Lv, L.; Zhang, J.; Liang, W.; Wang, P. Fine-tuning the expression of microRNA-155 controls acetaminophen-induced liver inflammation. Int. Immunopharmacol. 2016, 40, 339–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Chen, L.; Charrier, A.; Zhou, Y.; Chen, R.; Yu, B.; Agarwal, K.; Tsukamoto, H.; Lee, L.J.; Paulaitis, M.E.; Brigstock, D.R. Epigenetic regulation of connective tissue growth factor by MicroRNA-214 delivery in exosomes from mouse or human hepatic stellate cells. Hepatology 2014, 59, 1118–1129. [Google Scholar] [CrossRef] [Green Version]
  221. Wang, X.; Seo, W.; Park, S.H.; Fu, Y.; Hwang, S.; Rodrigues, R.M.; Feng, D.; Gao, B.; He, Y. MicroRNA-223 restricts liver fibrosis by inhibiting the TAZ-IHH-GLI2 and PDGF signaling pathways via the crosstalk of multiple liver cell types. Int. J. Biol. Sci. 2021, 17, 1153. [Google Scholar] [CrossRef]
  222. Bieback, K.; Kluter, H. Mesenchymal stromal cells from umbilical cord blood. Curr. Stem Cell Res. Ther. 2007, 2, 310–323. [Google Scholar] [CrossRef]
  223. Gasper, M.A.; Kunwar, P.; Itaya, G.; Lejarcegui, N.; Bosire, R.; Maleche-Obimbo, E.; Wamalwa, D.; Slyker, J.; Overbaugh, J.; Horton, H. Natural killer cell and T-cell subset distributions and activation influence susceptibility to perinatal HIV-1 infection. AIDS 2014, 28, 1115. [Google Scholar] [CrossRef] [Green Version]
  224. Sarvaria, A.; Jawdat, D.; Madrigal, J.A.; Saudemont, A. Umbilical cord blood natural killer cells, their characteristics, and potential clinical applications. Front. Immunol. 2017, 8, 329. [Google Scholar] [CrossRef] [Green Version]
  225. Le Blanc, K.; Tammik, C.; Rosendahl, K.; Zetterberg, E.; Ringdén, O. HLA expression and immunologic propertiesof differentiated and undifferentiated mesenchymal stem cells. Exp. Hematol. 2003, 31, 890–896. [Google Scholar] [CrossRef]
  226. Rabani, V.; Shahsavani, M.; Gharavi, M.; Piryaei, A.; Azhdari, Z.; Baharvand, H. Mesenchymal stem cell infusion therapy in a carbon tetrachloride-induced liver fibrosis model affects matrix metalloproteinase expression. Cell Biol. Int. 2010, 34, 601–605. [Google Scholar] [CrossRef] [PubMed]
  227. Jang, Y.O.; Kim, M.Y.; Cho, M.Y.; Baik, S.K.; Cho, Y.Z.; Kwon, S.O. Effect of bone marrow-derived mesenchymal stem cells on hepatic fibrosis in a thioacetamide-induced cirrhotic rat model. BMC Gastroenterol. 2014, 14, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Fathy, M.; Okabe, M.; Saad Eldien, H.M.; Yoshida, T. AT-MSCs antifibrotic activity is improved by eugenol through modulation of TGF-β/Smad signaling pathway in rats. Molecules 2020, 25, 348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Sun, T.; Li, H.; Bai, Y.; Bai, M.; Gao, F.; Yu, J.; Wu, R.; Du, L.; Li, F. Ultrasound-targeted microbubble destruction optimized HGF-overexpressing bone marrow stem cells to repair fibrotic liver in rats. Stem Cell Res. Ther. 2020, 11, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Zhang, L.; Zhou, D.; Li, J.; Yan, X.; Zhu, J.; Xiao, P.; Chen, T.; Xie, X. Effects of bone marrow-derived mesenchymal stem cells on hypoxia and the transforming growth factor beta 1 (TGFβ-1) and SMADs pathway in a mouse model of cirrhosis. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 7182. [Google Scholar] [CrossRef] [PubMed]
  231. Zhang, L.T.; Fang, X.Q.; Chen, Q.F.; Chen, H.; Xiao, P.; Peng, X.B.; Zhang, S.X.; Li, J.F.; Mao, X.R. Bone marrow-derived mesenchymal stem cells inhibit the proliferation of hepatic stellate cells by inhibiting the transforming growth factor β pathway. Mol. Med. Rep. 2015, 12, 7227–7232. [Google Scholar] [CrossRef]
  232. Stock, P.; Brückner, S.; Winkler, S.; Dollinger, M.M.; Christ, B. Human bone marrow mesenchymal stem cell-derived hepatocytes improve the mouse liver after acute acetaminophen intoxication by preventing progress of injury. Int. J. Mol. Sci. 2014, 15, 7004–7028. [Google Scholar] [CrossRef] [Green Version]
  233. Asian Institute of Gastroenterology, India. Combination of Autologous MSC and HSC Infusion in Patients With Decompensated Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT04243681 (accessed on 22 February 2022).
  234. Ukraine Association of Biobank. Long Term Follow up Mesenchymal Stem Cell Therapy for Patients Virus-related Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT05080465 (accessed on 22 February 2022).
  235. Sun Yat-Sen University; Third Affiliated Hospital, Sun Yat-Sen University. Autologous Bone Marrow Mesenchymal Stem Cells Transplantation Via Hepatic Artery in Patients With Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT00976287 (accessed on 13 January 2022).
  236. S-Evans Biosciences Co., Ltd.; Zhejiang University; Zhejiang General Hospital of Armed Police; Zhenjiang First People’s Hospital; Wuhan General Hospital of Guangzhou Military Command. Human Menstrual Blood-derived Mesenchymal Stem Cells for Patients With Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT01483248 (accessed on 20 January 2022).
  237. Alliancells Bioscience Corporation Limited. Safety and Efficacy Study of Umbilical Mesenchymal Stem Cells for Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT01573923 (accessed on 13 January 2022).
  238. Chaitanya Hospital, Pune. A Clinical Study to Evaluate the Safety and Efficacy of Mesenchymal Stem Cells in Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT01877759 (accessed on 23 February 2022).
  239. Shenzhen Beike Bio-Technology Co., Ltd.; No.85 Hospital, Changning, Shanghai, China. Human Umbilical Cord Mesenchymal Stem Cells Transplantation for Patients With Decompensated Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT01342250 (accessed on 23 February 2022).
  240. Rohto Pharmaceutical Co., Ltd. A Study of ADR-001 in Patients With Liver Cirrhosis. Available online: https://ClinicalTrials.gov/show/NCT03254758 (accessed on 23 February 2022).
  241. Shahid Beheshti University of Medical Sciences; Tarbiat Modarres University. Improvement of Liver Function in Liver Cirrhosis Patients After Autologous Mesenchymal Stem Cell Injection:a Phase I-II Clinical Trial. Available online: https://ClinicalTrials.gov/show/NCT00420134 (accessed on 27 December 2021).
  242. Royan, I. Transplantation of Autologous Mesenchymal Stem Cell in Decompensate Cirrhotic Patients With Pioglitazone. Available online: https://ClinicalTrials.gov/show/NCT01454336 (accessed on 20 January 2022).
  243. Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
  244. Driscoll, J.; Patel, T. The mesenchymal stem cell secretome as an acellular regenerative therapy for liver disease. J. Gastroenterol. 2019, 54, 763–773. [Google Scholar] [CrossRef] [Green Version]
  245. An, S.Y.; Jang, Y.J.; Lim, H.-J.; Han, J.; Lee, J.; Lee, G.; Park, J.Y.; Park, S.-Y.; Kim, J.H.; Do, B.-R. Milk fat globule-EGF factor 8, secreted by mesenchymal stem cells, protects against liver fibrosis in mice. Gastroenterology 2017, 152, 1174–1186. [Google Scholar] [CrossRef] [Green Version]
  246. Fu, Q.; Ohnishi, S.; Sakamoto, N. Conditioned Medium from Human Amnion-Derived Mesenchymal Stem Cells Regulates Activation of Primary Hepatic Stellate Cells. Stem Cells Int. 2018, 2018, 4898152. [Google Scholar] [CrossRef] [PubMed]
  247. Huang, B.; Cheng, X.; Wang, H.; Huang, W.; Wang, D.; Zhang, K.; Zhang, H.; Xue, Z.; Da, Y.; Zhang, N. Mesenchymal stem cells and their secreted molecules predominantly ameliorate fulminant hepatic failure and chronic liver fibrosis in mice respectively. J. Transl. Med. 2016, 14, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Varderidou-Minasian, S.; Lorenowicz, M.J. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics 2020, 10, 5979. [Google Scholar] [CrossRef] [PubMed]
  249. Fiore, E.J.; Domínguez, L.M.; Bayo, J.; García, M.G.; Mazzolini, G.D. Taking advantage of the potential of mesenchymal stromal cells in liver regeneration: Cells and extracellular vesicles as therapeutic strategies. World J. Gastroenterol. 2018, 24, 2427. [Google Scholar] [CrossRef] [PubMed]
  250. Li, T.; Yan, Y.; Wang, B.; Qian, H.; Zhang, X.; Shen, L.; Wang, M.; Zhou, Y.; Zhu, W.; Li, W. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013, 22, 845–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Dong, L.; Pu, Y.; Chen, X.; Qi, X.; Zhang, L.; Xu, L.; Li, W.; Ma, Y.; Zhou, S.; Zhu, J. hUCMSC-extracellular vesicles downregulated hepatic stellate cell activation and reduced liver injury in S. japonicum-infected mice. Stem Cell Res. Ther. 2020, 11, 1–11. [Google Scholar] [CrossRef] [PubMed]
  252. Mardpour, S.; Hassani, S.N.; Mardpour, S.; Sayahpour, F.; Vosough, M.; Ai, J.; Aghdami, N.; Hamidieh, A.A.; Baharvand, H. Extracellular vesicles derived from human embryonic stem cell-MSCs ameliorate cirrhosis in thioacetamide-induced chronic liver injury. J. Cell. Physiol. 2018, 233, 9330–9344. [Google Scholar] [CrossRef]
  253. Goodman, Z.D. Grading and staging systems for inflammation and fibrosis in chronic liver diseases. J. Hepatol. 2007, 47, 598–607. [Google Scholar] [CrossRef]
  254. Huang, Y.; De Boer, W.B.; Adams, L.A.; MacQuillan, G.; Bulsara, M.K.; Jeffrey, G.P. Image analysis of liver biopsy samples measures fibrosis and predicts clinical outcome. J. Hepatol. 2014, 61, 22–27. [Google Scholar] [CrossRef]
  255. Drenth, J.P.; Schattenberg, J.M. The nonalcoholic steatohepatitis (NASH) drug development graveyard: Established hurdles and planning for future success. Expert Opin. Investig. Drugs 2020, 29, 1365–1375. [Google Scholar] [CrossRef]
  256. Younossi, Z.M. Non-alcoholic fatty liver disease—A global public health perspective. J. Hepatol. 2019, 70, 531–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Cellular alterations during hepatic fibrosis: Multiple factors can cause liver injury. These factors induce liver inflammation through various pathways and cellular systems. Normal liver parenchyma contains a hepatocyte layer with microvilli and layers of fenestrated liver sinusoidal endothelium cells (LSECs), and a higher number of quiescent hepatic stellate cells (qHSCs), Kupffer cells (KCs), and natural killer cells (NKs). It also contains a normal amount of basement-forming collagens (Types IV and VI). Upon injury, the HSCs become activated and secrete a large amount of ECM, which leads to the loss of both endothelial fenestrations and hepatocyte microvilli, resulting in impairment of bidirectional metabolic exchange of portal venous flow. TNF-α can mediate a dual and opposing effect by acting on TNF receptors expressed on the endothelial cells. The LSECs also promote vascular leakage of plasma proteins and initiate the exocytosis of Weibel–Palade bodies (WPBs, denoted as a yellow oval), bringing P-selectin to the cell surface, which initiates diapedesis. Replacement of fibrillary collagen occurs, consisting of collagen I, III, and fibronectin. Furthermore, there is infiltration of immune cells, such as neutrophils and monocytes, and the injured area recruits the NK-T cells and alters liver morphology.
Figure 1. Cellular alterations during hepatic fibrosis: Multiple factors can cause liver injury. These factors induce liver inflammation through various pathways and cellular systems. Normal liver parenchyma contains a hepatocyte layer with microvilli and layers of fenestrated liver sinusoidal endothelium cells (LSECs), and a higher number of quiescent hepatic stellate cells (qHSCs), Kupffer cells (KCs), and natural killer cells (NKs). It also contains a normal amount of basement-forming collagens (Types IV and VI). Upon injury, the HSCs become activated and secrete a large amount of ECM, which leads to the loss of both endothelial fenestrations and hepatocyte microvilli, resulting in impairment of bidirectional metabolic exchange of portal venous flow. TNF-α can mediate a dual and opposing effect by acting on TNF receptors expressed on the endothelial cells. The LSECs also promote vascular leakage of plasma proteins and initiate the exocytosis of Weibel–Palade bodies (WPBs, denoted as a yellow oval), bringing P-selectin to the cell surface, which initiates diapedesis. Replacement of fibrillary collagen occurs, consisting of collagen I, III, and fibronectin. Furthermore, there is infiltration of immune cells, such as neutrophils and monocytes, and the injured area recruits the NK-T cells and alters liver morphology.
Cells 11 01500 g001
Figure 2. suPAR as a serum biomarker: Upon inflammation and for controlling fibrin degradation, the uPAR is cleaved from the cell surface as Pro-uPA and activates in the uPA form. Plasminogen is converted to plasmin via either the plasminogen activator receptor or the urokinase receptor and helps in the degradation of the ECM by breaking fibrin strands during liver fibrosis. In patients with liver diseases, circulating suPAR levels increase with the increase in disease severity, and are indicative of an adverse prognosis.
Figure 2. suPAR as a serum biomarker: Upon inflammation and for controlling fibrin degradation, the uPAR is cleaved from the cell surface as Pro-uPA and activates in the uPA form. Plasminogen is converted to plasmin via either the plasminogen activator receptor or the urokinase receptor and helps in the degradation of the ECM by breaking fibrin strands during liver fibrosis. In patients with liver diseases, circulating suPAR levels increase with the increase in disease severity, and are indicative of an adverse prognosis.
Cells 11 01500 g002
Figure 3. The importance of balance between MMPs and TIMPs, and between HGF and TGF-β, as hepatoprotective and counteracting agents in liver fibrosis.
Figure 3. The importance of balance between MMPs and TIMPs, and between HGF and TGF-β, as hepatoprotective and counteracting agents in liver fibrosis.
Cells 11 01500 g003
Figure 4. The role of TSG-6 in cellular growth and proliferation in fibrotic liver cells: TSG-6 has the potential to improve liver injury; it can induce proliferation, stemness, and increase the immunomodulatory mechanism of MSCs. TSG-6 reduces inflammation and changes in tissue repair via mechanisms such as reducing neutrophil infiltration and activation, and inhibiting inflammatory M1–M2 polarization of monocytes. M2 macrophages produce complex cytokines, and have various functions; they can be further divided into M2a, M2b, M2c, and M2d subtypes. M2a cells can prevent fibrosis by inducing regulatory T cells.
Figure 4. The role of TSG-6 in cellular growth and proliferation in fibrotic liver cells: TSG-6 has the potential to improve liver injury; it can induce proliferation, stemness, and increase the immunomodulatory mechanism of MSCs. TSG-6 reduces inflammation and changes in tissue repair via mechanisms such as reducing neutrophil infiltration and activation, and inhibiting inflammatory M1–M2 polarization of monocytes. M2 macrophages produce complex cytokines, and have various functions; they can be further divided into M2a, M2b, M2c, and M2d subtypes. M2a cells can prevent fibrosis by inducing regulatory T cells.
Cells 11 01500 g004
Figure 5. NK cell responses and targeted therapy: (1) NK cells are the first responders in the immune system, and can directly recognize and begin the cell death mechanism. (2) NK cells release exosomes with cytotoxic capabilities, and can contain miRNAs, cytokines, and NK cell surface receptors. (3) Activated NK cells selectively kill early or activated HSCs, but not quiescent HSCs. IFN-γ-producing NK cells directly induce HSC death, but also further enhance NK cell cytotoxicity against HSCs. Quiescent cells do not express elevated NK-activating ligand, and are hence resistant. (4) In proliferating cells, DPP4 is expressed at low levels, but in senescent cells, DPP4 mRNA levels increase, leading to the production of DPP4, which localizes on the cell surface and is exposed to the extracellular space. The localization of DPP4 enables selective elimination by immune cells such as NKs via ADCC.
Figure 5. NK cell responses and targeted therapy: (1) NK cells are the first responders in the immune system, and can directly recognize and begin the cell death mechanism. (2) NK cells release exosomes with cytotoxic capabilities, and can contain miRNAs, cytokines, and NK cell surface receptors. (3) Activated NK cells selectively kill early or activated HSCs, but not quiescent HSCs. IFN-γ-producing NK cells directly induce HSC death, but also further enhance NK cell cytotoxicity against HSCs. Quiescent cells do not express elevated NK-activating ligand, and are hence resistant. (4) In proliferating cells, DPP4 is expressed at low levels, but in senescent cells, DPP4 mRNA levels increase, leading to the production of DPP4, which localizes on the cell surface and is exposed to the extracellular space. The localization of DPP4 enables selective elimination by immune cells such as NKs via ADCC.
Cells 11 01500 g005
Figure 6. The role of various miRNA families in liver fibrosis: Liver cells take and release exosomal microRNAs (miRNAs). Their role in liver fibrosis: Extracellular vesicles (EVs) derived from adipose tissue, HSCs, and neutrophils containing miR-155, -214, and -223, respectively, are taken up by liver cells, leading to increased insulin resistance by suppressing PPARγ. miR-214 in HSCs is shuttled by EVs to hepatocytes, resulting in inhibition of CCN2/Ccn2. Under a high-fat diet and alcohol consumption, miR-223 is elevated in hepatocytes, and attenuates NASH progression by targeting Cxcl10 and Taz. The miR-29 and miR-15 families regulate hepatic fibrosis in the following ways: (1) miR-29a targets AKT3 and PI3K, which helps to induce cell apoptosis through the caspase-9 cascade pathway; (2) PDGF and IGF receptors suppress the overall effect of the PI3K/AKT signaling pathway; (3) miR-16 targets HGF and SMAD7, and blocks the TGF-β/Smad signaling pathway; (4) miR-192 downregulates cyclin M1 and inhibits cell proliferation; (5) miR-1 promotes endothelial inflammation.
Figure 6. The role of various miRNA families in liver fibrosis: Liver cells take and release exosomal microRNAs (miRNAs). Their role in liver fibrosis: Extracellular vesicles (EVs) derived from adipose tissue, HSCs, and neutrophils containing miR-155, -214, and -223, respectively, are taken up by liver cells, leading to increased insulin resistance by suppressing PPARγ. miR-214 in HSCs is shuttled by EVs to hepatocytes, resulting in inhibition of CCN2/Ccn2. Under a high-fat diet and alcohol consumption, miR-223 is elevated in hepatocytes, and attenuates NASH progression by targeting Cxcl10 and Taz. The miR-29 and miR-15 families regulate hepatic fibrosis in the following ways: (1) miR-29a targets AKT3 and PI3K, which helps to induce cell apoptosis through the caspase-9 cascade pathway; (2) PDGF and IGF receptors suppress the overall effect of the PI3K/AKT signaling pathway; (3) miR-16 targets HGF and SMAD7, and blocks the TGF-β/Smad signaling pathway; (4) miR-192 downregulates cyclin M1 and inhibits cell proliferation; (5) miR-1 promotes endothelial inflammation.
Cells 11 01500 g006
Table 1. Hepatic stellate cell phenotypes.
Table 1. Hepatic stellate cell phenotypes.
Cell TypeFunctionsReferences
1Quiescent
  • Storage of vitamin A, which is found in numerous intracellular droplets.
  • Multiple thorn-like cytoplasmic extensions can protrude into the sinusoidal space or make direct contact with hepatocytes.
  • These extensions can also be used as sinusoidal sentinels, which can detect biochemical or mechanical alterations in hepatocytes.
[27]
2Activated
  • They lose lipid-rich granules and transdifferentiate into α-SMA-positive myofibroblasts.
  • They produce increased amounts of ECM and pro-inflammatory as well as pro-fibrogenic cytokines, and cause liver fibrosis.
[28]
3Inactivated
  • Reverted or inactivated HSCs present a restored expression of their pro-fibrogenic protein profile (including changes in collagen-1, α-SMA, TGF-beta receptor type-1 (TGFRI), and TIMP1 expression).
  • They do not express quiescent makers (such as perilipin 2 and adiponectin receptor 1).
[27]
4Senescent
  • Senescent hepatic stellate cells display decreased collagen production and proliferation.
  • Induction of senescence could be a protective mechanism against the progression of liver fibrosis.
  • The concept of therapy-induced senescence has been proposed to treat liver fibrosis.
[29]
Table 2. Clinical trials for metabolic targets affecting NASH/NAFLD-based liver fibrosis.
Table 2. Clinical trials for metabolic targets affecting NASH/NAFLD-based liver fibrosis.
CT No.Treatment/
Drug Name
Mechanism of ActionTarget
Diseases
Clinical Trial DesignClinical
Phase
EfficacyNumber of
Patients
References
NCT00760513OMACORReduce the synthesis of triglycerides (TGs)NAFLDTreatment with long-chain n-3 fatty acid for 18 months affects biomarkers of NAFLD.Phase 420% decrease in liver fat103 participants[92]
NCT02030977ResveratrolAntioxidantNAFLD, Liver
Fibrosis
Effect of liver enzymes,
inflammatory factors, and fibrosis in patients with NAFLD. Patients were steatosis grade 1.
Phase 3Study
completed/ no results reported
50 patients[93,94]
NCT02548351Obeticholic acid
REGENE-RATE
FXR agonistNASH with
fibrosis
Obeticholic acid treatment compared to placebo on histological improvement and liver-related clinical outcomes.Phase 3Active2480 participants[95,96]
NCT03008070IVA337PPAR agonistNASH with
fibrosis
A next-generation pan-PPAR agonist for the pathophysiology of NASH.Phase 2Not worsening fibrosis at higher dose of 1200 mg247 participants[91]
NCT02684591AramcholSCD1 inhibitorNASHTo test the efficacy of 400 mg and 600 mg of Aramchol. Phase 2No significant adverse effects, did not reduce hepatic fat247 participants[88]
NCT03357380Semaglutidereduces HbA1c,NAFLDComparing changes in early-stage scar tissue and fat deposition in the liver. Participants self-inject medicine once daily for 72 weeks.Phase 1No significant adverse
Effect, did not reduce
hepatic fat
67 participants[97]
Table 3. Clinical trials for metabolic targets affecting liver fibrosis.
Table 3. Clinical trials for metabolic targets affecting liver fibrosis.
NCT No.Treatment/Drug
Name
Mechanismof ActionTarget
Diseases
Clinical Trial DesignClinical
Phase
EfficacyNumber of
Patients
References
NCT00049842Peginterferon alph-2b
(SCH 54031)
Type 1
interferon
activator
Liver
fibrosis,
chronic
hepatitis C
Evaluate safety and efficacy of PEG-Intron vs. no treatmentPhase 3Lower
fibrosis progression
540
participants
[108,109]
NCT01938781Entecavir,
Peg-IFN
Inhibits HBV DNA
polymerase
Liver
fibrosis
For patients with F2/F3, one arm is entecavir for 2 years, and the other is entecavir for 0.5 years and entecavir plus peg-IFN for 1 yearPhase 4Study
completed/
no results
reported
400
participants
[110,111,112]
NCT00298714LosartanAngiotensin II type 1
receptor
antagonist
Liver
fibrosis,
chronic
hepatitis C
Administration of angiotensin II type 1 (AT1) receptor antagonists in HSCs (fibrosis F2-F3)Phase 4Study
completed/
no results
reported
20
participants
[99,100]
NCT00990639Candesartan andramiprilAngiotensin II type 1
receptor
antagonist
Liver
fibrosis
extent with chronichepatitis C
Evaluating drug action and changes in
FibroScan recording
Phase 3Pending45
participants
[101,102]
NCT00265642IrbesartanAngiotensin II type 1
receptor
antagonist
Liver
fibrosis,
chronic hepatitis C
AT1 receptor antagonists of angiotensin II have inhibitory effects on TGF-beta 1 production, and can limit the progression of liver fibrosisPhase 3No results reported200
participants
[103]
NCT04971577SimvastatinHMG-CoA
reductase
inhibitors (statins)
Liver
fibrosis
Simvastatin for reducing liver fibrosis in patients with advanced fibrosis due to alcoholPhase 2/3Active90
participants
[107,113]
Table 5. Review of different interleukin functions in liver fibrosis.
Table 5. Review of different interleukin functions in liver fibrosis.
Interleukin
Type
Produced byResponse CellFunctionReferences
IL-2CD4+ T cells, CD8+ T cells, dendritic cells, and thymic cellsT cells and NK cellsEnhances cytotoxicity in NK cells; activates STAT1, STAT3, and STAT5.[180]
IL-6Lymphocytes, monocytes, fibroblasts, vascular smooth muscle cells, and endothelial cellsNon-parenchymal cellsDeletion of IL-6 increases hepatocyte injury and apoptosis.[181]
IL-10Hepatic stellate cells, liver sinusoidal endothelial cells, Kupffer cells, lymphocytes, and Th cellsHSCsIL-10 inhibits HSC activation.[182,183]
IL-12Macrophages, dendritic cells, and B lymphocytesTh1
  • IL-12 shifts immune response to Th1;
  • Secretion of IFN-γ and augmentation of the cytolytic activity.
[35,184,185]
IL-15MonocytesNK cells
  • Secretion of IFN-γ, macrophage colony-stimulating factor, and TNF-α;
  • Work minimal synergism with IL-12.
[186]
IL-22αβ T-cell classes Th1, Th22, and Th17, along with γδ T cells, NKTs, ILC3, neutrophils, and macrophagesHSCs
  • Reduces fat accumulation and steatosis;
  • Induces senescence in HSCs.
[31,187,188,189]
IL-30Th2 cells upon activationNKT and HSCsAttenuates liver fibrosis through inducing NKG2D–rae1 interaction.[190]
Table 6. Studies of liver fibrosis using mesenchymal stem cells.
Table 6. Studies of liver fibrosis using mesenchymal stem cells.
NCT No.SponsorTarget
Diseases
Clinical Trial DesignClinical
Phase
StatusNumber of
Patients
References
NCT04243681Asian Institute of Gastroenterology, IndiaLiver cirrhosisCombination of autologous mesenchymal and hematopoietic stem cells infused in patientsPhase 4No results reported5
participants
[233]
NCT05080465Ukraine Association of BiobankLiver cirrhosisLong-term follow-up autologous MSC therapy for patients with virus-related liver cirrhosisPhase 3Active700
participants
[234]
NCT00976287 Sun Yat-Sen UniversityLiver fibrosis,
chronic hepatitis C
Liver function was monitored by serum examination. The levels of serum alanine aminotransferase (ALT), total bilirubin (TB), prothrombin time (PT), and albumin (ALB) were examined at pre-transplantation, and 3 days to 2 years post-transplantationPhase 2Results
not posted
50
participants
[235]
NCT01483248Zhejiang University, ChinaLiver cirrhosis,
fibrosis
Menstrual blood-derived stem cells can improve the disease conditions in patients with liver cirrhosis.Phase 1/2No results posted50
participants
[236]
NCT01573923 Allian cells Bioscience Corporation LimitedLiver cirrhosisIntravenous administration of umbilical MSCs for the treatment of patients with liver cirrhosis in the next three years.Phase 1/2No results reported320
participants
[237]
NCT01877759Chaitanya Hospital, IndiaLiver cirrhosisBone-marrow-derived autologous stem cells + human umbilical-cord-derived MSCs Phase 1/2No results reported20
participants
[238]
NCT01342250Shenzhen Beike Bio-Technology Co., Ltd.Liver cirrhosisSafety and efficacy of human umbilical cord (hUC)-MSC transplantation for patients with decompensated liver cirrhosisPhase 1/2No results reported20
participants
[239]
NCT03254758Rohto Pharmaceutical Co., Ltd., JapanDecompensated liver cirrhosisFirst-in-human study of ADR-001, adipose-derived mesenchymal stem cells (AD-MSCs)Phase 1/2Recruiting27
participants
[240]
NCT00420134Shahid Beheshti University of Medical Sciences, IranLiver failure,
cirrhosis
Investigators try to separate MSCs from end-stage liver disease, and then these cells are differentiated into the progenitors of hepatocytes; finally, the investigators inject these cells into the portal vein under ultrasound guidance.Phase 1/2No results reported30
Participants
[241]
NCT01454336Royan InstituteLiver fibrosisPioglitazone and autologous bone marrow MSC transplantation.Phase 1Completed3
participants
[242]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jangra, A.; Kothari, A.; Sarma, P.; Medhi, B.; Omar, B.J.; Kaushal, K. Recent Advancements in Antifibrotic Therapies for Regression of Liver Fibrosis. Cells 2022, 11, 1500. https://doi.org/10.3390/cells11091500

AMA Style

Jangra A, Kothari A, Sarma P, Medhi B, Omar BJ, Kaushal K. Recent Advancements in Antifibrotic Therapies for Regression of Liver Fibrosis. Cells. 2022; 11(9):1500. https://doi.org/10.3390/cells11091500

Chicago/Turabian Style

Jangra, Anshika, Ashish Kothari, Phulen Sarma, Bikash Medhi, Balram Ji Omar, and Karanvir Kaushal. 2022. "Recent Advancements in Antifibrotic Therapies for Regression of Liver Fibrosis" Cells 11, no. 9: 1500. https://doi.org/10.3390/cells11091500

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