2.1. Hepatic Stellate Cells and Myofibroblasts
Current technological advances such as single-cell RNA-sequencing [sc-RNA-seq], high dimensional multi-omics [proteomics, genomics] and spatial transcriptomics enabled comprehensive knowledge of cellular biological processes and cell types, which has generated novel insights into the tissue biology and disease mechanisms in liver fibrosis [
3]. HSCs are a nonparenchymal cell population that in the healthy organ accounts for around 5%–8% of all liver cells [
2,
3]. HSCs are localized in the space of Disse, between the basolateral region of hepatocytes and the anti-luminal surface of the liver sinusoidal endothelial cells [LSECs] [
2,
3,
4]. In the healthy liver, quiescent HSCs store retinoids [vitamin A and its metabolites] within their cytoplasmic lipid droplets, which are essential for the perpetuation of quiescent HSCs [
2,
3,
4,
5].
Upon liver injury, the amount of vitamin A stored by quiescent HSCs decreases progressively, and the cells transdifferentiate into activated HSCs that produce αSMA, collagen I, inhibitors of matrix degradation, and pro-inflammatory cytokines as well as chemokines that, by positive chemotaxis, attract immune cells to the region of inflammation [
2,
3,
4,
5]. Activated HSCs display a contractile, proliferative, and fibrogenic phenotype [
3,
4]. Ultrastructurally, they are characterized by a rough endoplasmic reticulum and a Golgi apparatus producing collagen [
2,
3]. Activated HSCs promote the recruitment and infiltration of immune cells in the liver by producing several proinflammatory cytokines, such as CCL2, CCL5, IL8, chemokine [C-X-C motif] ligand-12 [CXCL12], and by expressing adhesion molecules, such as intercellular adhesion molecule 1 [ICAM-1] and vascular cell adhesion molecule 1 [VCAM-1] [
2,
4,
5]. These cells migrate to the region of injury to contribute to the formation of a fibrous scar. HSCs, together with LSECs and hepatic macrophages, constitute the liver microcirculatory microenvironment having critical roles in governing the vascular tone and controlling inflammation [
5].
Experimental mouse models have revealed the existence of different subsets of HSCs that display distinct functions during chronic liver disease [
2,
4]. Single-cell technologies have shown that quiescent HSCs account for a relatively homogeneous population. However, activated HSCs can be discerned into several subsets [
2,
3,
4,
5,
19,
20]. Functionally, quiescent HSCs produce high levels of growth factors and can protect hepatocytes from injury, whereas myofibroblasts mainly express ECM proteins, such as collagen I, collagen III, VI, and XIV, cytokines and chemokines, which drive inflammation and fibrosis [
2,
3,
4,
5]. In cirrhotic livers, a population of quiescent HSC has been detected that expresses various growth factors and cytokines and suppresses liver inflammation and fibrosis [
2,
5]. In addition, activated HSCs also display immunoregulatory functions: in different mouse models they were found to promote T-cell apoptosis through PD-1 and PD-L1; affect B-cell function via immune checkpoint molecules; and contribute to immune tolerance through promoting immunosuppressive cells such as Tregs and myeloid-derived suppressor cells [MDSCs] [
3]. At present, it is not clearly understood under which conditions HSCs act proinflammatory or tolerogenic, respectively [
2,
3]. There is an extensive interplay between HSCs and immune cells, such as macrophages [
2,
3,
4,
5]. Experimental trials using HSCs and macrophages revealed that soluble mediators derived from activated HSCs promote the differentiation of macrophages to a proinflammatory phenotype [
2,
3] Additionally, novel data from scRNA-seq mouse studies reveal that HSCs communicate with surrounding endothelial cells immune cells via soluble mediators, many of which are upregulated during liver injury [
2].
Various factors, such as toxic lipids, lipid mediators, inflammation signals, growth factors, and distinct signals, including cell-cell contact with macrophages, cytokines derived from activated immune cells, and pathogen-associated molecular patterns [PAMPs] or danger-associated molecular patterns [DAMPs], mediate the activation of HSCs during liver injury [
2,
3,
4,
5,
14,
15]. Growth factors, including transforming growth factor β [TGFβ], platelet-derived growth factor [PDGF], and epidermal growth factor [EGF] are key drivers of HSC activation [
2,
4,
5,
15,
16,
17]. Once activated, HSCs can release autocrine signals that maintain their fibrogenic state [
16,
17]. TGFβ is the central mediator in the activation of HSCs, which is produced by infiltrating lymphocytes and monocytes, Kupffer cells [KCs], and damaged hepatocytes [
2,
4,
5,
19]. It is responsible for the imbalance between ECM synthesis and degradation in liver fibrosis [
20]. In chronic liver disease, hepatocyte death and consequent recruitment of macrophages play an important role in HSC activation and fibrosis via producing TGFβ [
16]. In addition, the engulfment of cell debris by macrophages fosters their expression of TGFβ [
16]. Macrophages remove apoptotic hepatocytes through the process of efferocytosis, which promotes the expression of growth factors, such as macrophage-derived TGFβ, resulting in the activation of HSCs [
21].
Figure 1. The transition of quiescent HSCs into their activated phenotype is an energy-consuming process that requires fundamental reprogramming of their metabolic pathways [
2,
5]. Activated HSCs oftentimes display high-level expression of ECM-related genes [
5]. Phosphorylation of SMAD proteins – e.g., SMAD3 – mediates TGFβ-related HSC activation, which eventually results in the upregulation of collagen I and III synthesis [
22,
23]. In addition, α-SMA and connective tissue growth factor [CTGF] genes are upregulated by SMAD proteins [
24]. TGFβ may promote HSC activation by other mechanisms besides the SMAD pathway, such as mitogen-activated protein kinase-1, p38, and c-jun N-terminal kinase-related mechanisms [
25,
26]. The type 3 cytokine, IL-17 derived from TH
17 cells and neutrophils exhibits an important role in activating the TGFβ signaling pathway [
27]. In addition to this direct impact, latent TGFβ accumulated in the ECM is activated by integrin-αv-mediated contraction of HSCs [
27]. Therefore, deletion of the αv subunit in HSC prevents TGFβ activation and protects mice from liver fibrosis [
2]. TGFβ signaling comprises various pathways such as the non-canonical TAK1/JNK and the RAS/RAF/MAPK pathways as well as SMAD pathway, resulting in ECM synthesis [
28,
29]. TGFβ induces autophagy by interacting with insulin-like growth factor-binding protein-associated protein 1, which activates the P13K/AKT/mTOR pathway [
30]. In addition, the epigenetic regulator TET3 is involved in the TGFβ/SMAD2/3 activation pathway [
2].
TGFβ-induced HSC activation is associated with an increased expression of other profibrogenic growth factors including platelet-derived growth factor [PDGF] and vascular endothelial growth factor [VEGF] [
20,
31]. PDGF is a mitogenic growth factor whose polypeptide chains A and B are arranged in AA, AB, or in BB combinations. Autocrine PDGF-BB stimulation is most potent to induce HSC proliferation [
32,
33]. As such, the autocrine stimulation of HSCs by TGFβ and PDGF-AA initiates HSC activation and perpetuates liver fibrosis [
20,
31,
34]. PDGF acts on HSCs through PDGF receptor-β [PDGFRβ], which can promote increased signaling and HSC activation [
34]. In HSCs, PDGFRβ activation induces their proliferation and migration, drives cell survival and promotes the expression of hedgehog pathway ligand – such as sonic hedgehog – which triggers HSC activation [
35,
36]. Interestingly, experimental PDGFRβ deletion resulted in the regression of liver fibrosis, whereas the activation of this receptor enhanced fibrogenesis [
37,
38]. In line with this finding, there is a correlation between the degree of fibrosis and the PDGFRβ level in patients with NAFLD [
39]. Under physiological conditions, PDGF is mainly expressed by platelets, while the factor is additionally expressed by endothelial cells, macrophages and activated HSCs in liver diseases [
2,
38,
39]. During liver injury, VEGF is produced by hepatocytes, which promotes HSC activation and proliferation, leading to increased production of ECM proteins and TGFβ. Similarly, the accumulation of cholesterol and fatty acids during metabolic liver injury leads to the release of hedgehog ligands and exosomes that promote HSC proliferation and ECM production [
5]. The phagocytosis of cholesterol-laden hepatocytes by KCs and macrophages differentiated
de novo from infiltrating monocytes causes inflammasome activation and the production of proinflammatory cytokines, which perpetuate liver inflammation, HSC activation and fibrosis [
5].
Figure 2.
Studies on the origin of hepatic myofibroblasts have documented their heterogenous derivation, which
inter alia includes HSCs, portal fibroblasts and bone marrow-derived cells such as fibrocytes and mesenchymal stem cells [
40,
41,
42]. In contrast, liver parenchymal cells do not serve as a source of myofibroblast generation [
4,
40]. Recent studies clearly identified both liver-resident activated HSCs and activated portal fibroblasts to be the main source of hepatic myofibroblasts in chronic liver disease [
43,
44]. This view is strongly supported by results from cell fate mapping and deep phenotyping trials in both hepatotoxin-induced liver fibrosis models and models of biliary fibrosis where activated HSCs and activated portal fibroblasts were found to constitute more than 90% of the collagen-producing cells [
43,
44]. However, the composition of myofibroblasts is highly variable depending on the underlying cause of liver fibrosis. HSCs are usually activated as a result of toxic liver injury affecting the centrilobular and perisinusoidal regions of the liver. However, both activated HSCs and portal fibroblasts promote cholestatic liver fibrosis caused by periportal injury [
43,
44]. In biliary fibrosis models, 70% of the myofibroblast population are derived from activated fibroblasts at the onset of cholestatic injury, while the majority of myofibroblasts in later stages of disease was found to originate from HSCs [
43]. In addition, fibrocytes constitute a small population of bone marrow-derived myofibroblasts in mice with cholestatic or toxic fibrosis [
45]. Potential other sources of hepatic myofibroblasts in the pathogenesis of liver fibrosis might include epithelial-to-mesenchymal transition [EMT] [
46], but – at least in mice – cell fate mapping studies excluded EMT from the pathogenesis of liver fibrosis [
47,
48,
49,
50,
51]. It has been reported that mesenchymal stem cells experimentally obtained from murine bone marrow proliferate in the liver and lead to an increase in tissue fibroblasts [
52].
Differentiation of quiescent HSCs into activated HSCs is a two-step process: The initiation phase encompasses the priming of HSCs which sensitizes them to fibrogenic and proliferative cytokines [
41,
47,
48,
53]. Initiation of liver fibrogenesis is accompanied by increased cell activity containing the proto-oncogene c-myc as well as cyclin E1 [
23]. Damaged and apoptotic hepatocytes induce HSC activation by degrading the regular ECM composition in the space of Disse, releasing reactive oxygen species and proinflammatory cytokines, and recruiting immune cells, which collectively perpetuates HSC activation [
54,
55,
56,
57]. Activation of the PDGF receptor, the occurrence of a contractile and fibrogenic phenotype, and alteration of growth factor signaling are the fundamental properties of the initiation phase [
53,
58]. During the initiation phase, quiescent HSCs transdifferentiate into activated HSCs that produce PDGF and PDGFRβ. PDGF promotes HSC proliferation and results in the increased production of profibrogenic cytokines such as TGFβ, which further activate HSCs to upregulate α-SMA expression and stimulate ECM secretion [
2,
3]. Accordingly, in animal models of experimental liver fibrosis, depletion of PDGFRβ causes a reduction in fibrosis, while auto-activation of this receptor increases fibrogenesis [
2,
3]. In addition, levels of circulating PDGFRβ can provide information about the extent of liver fibrosis [
2]. The perpetuation phase is characterized by specific phenotypic alterations, including proliferation, contractility, fibrogenesis, altered matrix degradation and inflammatory signaling [
2,
4,
53,
59]. The composition, distribution and amount of ECM proteins in fibrogenesis vary depending on the etiological factor. In the healthy liver, the ECM is in the space of Disse and predominantly consists of collagen IV and laminin [
2,
3,
58]. During chronic injury, fibrillar collagens such as collagen I and III become essential proteins of the ECM [
58]. Elimination of the underlying cause leads to resolution during which HSCs undergo apoptosis, become senescent or revert to an inactive HSC phenotype [
2,
4,
53].
2.2. Inflammatory Cell Species Driving Hepatic Fibrogenesis
Chronic inflammation plays a critical role in the pathogenesis of hepatic fibrosis [
2,
4,
5,
59]. The immune response in the acutely damaged liver can be initiated by endogenous molecules [
59]. Damaged and dying cells release various soluble molecules acting as DAMPs. Necrotic cells release high mobility group box 1 protein [HMGB1] into the microenvironment by necrotic cells. However, macrophages and dendritic cells can contribute as well [
59] – and in fact, HMGB1 recruits these myeloid cells to the damaged site. In the early stages of chronic liver disease, damaged parenchymal cells release inflammatory cytokines and soluble mediators that induce the activation of inflammatory cells, which include macrophages, lymphocytes and NK cells [
2,
5,
59]. Neutrophils are critical first responders of the innate immune system, and they can contribute to hepatic inflammation through the production of proinflammatory cytokines and neutrophil extracellular traps [NETs], KC activation and the recruitment of additional types of immune cells [
60,
61,
62]. Upon liver injury, neutrophils are rapidly recruited to the injured site to remove the apoptotic cells [
61]. These cells further serve as a source of cytokines, and they kill bacteria through ejecting nuclear and mitochondrial DNA nets and the release of anti-microbial enzymes [
4,
62]. In mice, the elimination of neutrophils or the deletion of neutrophil-derived soluble molecules [IL-8, IL-18, IL-17, CCL3, CCL4 and CCL2] attenuates the development of hepatic fibrosis [
63,
64]. Damaged and dying hepatocytes release P2Y
14 ligands, such as uridine 5’-diphosphate [UDP]-glucose and UDP-galactose, which on HSCs bind to the P2Y
14 receptor and promote HSC activation [
5,
65]. In mice, experimental P2Y
14 deficiency led to reduced fibrosis [
65]. In addition, damaged hepatocytes may release nuclear HMGB1 and directly activate HSCs [
66]. Mitochondria-derived danger signals [mito-DAMPs] are abundant in the liver, as hepatocytes comprise a high number of mitochondria due to their high metabolic activity [
67] [see also below]. Thus, mito-DAMPs are increased in patients with chronic liver disease and have been evidenced to stimulate HSC activation and scar tissue formation in experimental mouse models [
68]. Some liver cell populations – including HSCs, KCs and LSECs – express receptors for these danger signals [
68]. The formation of an inflammasome is a critical process that triggers the inflammatory response via IL-1β and IL-18, which eventually induces inflammatory cell death [
69].
Macrophages play central roles in the maintenance of liver homeostasis, tissue remodeling and the induction of immune responses [
2,
4,
70]. They play a critical role during inflammation, injury and fibrogenesis, but they can also promote fibrosis resolution [
5,
70]. Hepatic macrophages, which in the healthy liver consist exclusively of yolk sac-derived KCs, are increasingly displaced by bone marrow-derived macrophages in the diseased liver [
73]. They display high phenotypic and functional diversity and plasticity, which means that they change their transcriptional profiles and functions based on environmental, tissue-associated, and inflammatory stimuli [
5,
70,
71,
72]. KCs are the major cell population of the innate immune system; this particular cell population is able to sense hepatocyte stress and injury signals from other cells as well as to engulf cellular debris and release inflammatory signals [
5,
73]. The activation of TGFβ-expressing macrophages plays an important role in hepatic fibrogenesis [
2,
4,
5,
70]. Generally, liver injury activates liver macrophages that thereupon phagocytose bacteria and remove their metabolites from the portal circulation [
5,
70]. Activated hepatic macrophages produce various cytokines, including TGF-β1, PDGF, TNF-α, IL-1, IL-6, IL-10, CXCL1, CCL2, CCL5 [also known as RANTES] and mediators such as ROS that trigger HSC activation and drive the infiltration of bone marrow-derived monocytes and neutrophils into the damaged liver [
5,
70]. Monocyte-derived macrophages and KCs are the main sources of TGFβ.
Macrophages perpetuate the survival of myofibroblasts via IL-1β and TNF-dependent activation of NF-κB [
2,
3,
4,
5,
70]. CCL3 is a ligand for CCR1 and CCR5 that fosters the progression of hepatic fibrosis [
4,
70]. In mouse models of liver fibrosis, induced by CCl
4 or by a methionine- and choline-deficient diet, blocking CCL3 or CCL5 decreased HSC activation and reduced fibrosis [
2,
4,
5]. Genetic deletion or inhibition of myeloid TGFβ improved liver fibrosis in mice, whereas genetic overexpression of TGFβ promoted liver fibrosis, indicating that TGFβ is a key driver of fibrosis [
75,
76]. However, long-term depletion of TGFβ may cause impaired liver regeneration, delayed wound healing and a high carcinogenesis risk in mice [
77]. IL-6, TNF and IL-1β may act synergistically with TGFβ, and genetic deletion of these cytokines may weaken the development of liver fibrosis [
78,
79]. Lipopolysaccharide [LPS], IL-1β, and TNF can enhance TGFβ signaling by downregulating BMP activin membrane-bound inhibitor [BAMBI], which serves as a pseudo-receptor for the TGFβ type I receptor family and a negative modulator of TGFβ signaling [
80,
81]. TGFβ and IL-6 are key drivers in the differentiation of naïve T cells to T helper 17 [Th17] cells [
79,
82]. Murine Th17 cells express remarkably potent profibrogenic IL-17A and anti-inflammatory IL-22 [
5]. Studies using single-cell technologies revealed that monocyte-derived macrophages can replace KCs and acquire a phenotype of lipid-associated macrophages or scar-associated macrophages [SAMs], including TREM2, CD9, and osteopontin expression [
83,
84,
85]. Lipid-associated macrophages can be subdivided into a transitional CX3CR1
+CCR2
+ lipid-associated subpopulation, and classical trem2
+Cd3
+Cd9
+Gpnmb
+ lipid-associated macrophages [
70,
86,
87]. Their function is associated with the expression of triggering receptor [TREM2], which mediates lipid uptake and metabolism [
87].
Macrophages have a key role during inflammation, injury, and fibrogenesis, but they can also support the resolution of fibrosis [
88]. During the progression of fibrosis, inflammation accelerates macrophage trafficking and promotes their accumulation in the liver, where these cells express cytokines and chemokines that stimulate HSC activation [
88]. The monocyte influx into the liver is mainly regulated by the chemokine monocyte chemoattractant protein-1 [MCP-1] [CCL2] and its associated receptor C-C chemokine receptor 2 [CCR2] [
88]. The MCP-1/CCR2 pathway is particularly activated in NASH patients [
70]. KC- and HSC-derived CCL2 contributes to the increased differentiation of immature monocyte-derived LYC6C
hi macrophages in the liver [
89]. Macrophages play a critical role in the resolution of liver fibrosis, that are the main source of fibrinolytic matrix metalloproteinases [MMPs], including MMP12 and MMP13 [
70]. During the regression of liver fibrosis, macrophages differentiate into a LY6C
low phenotype, stop the production of proinflammatory and fibrogenic factors, and secrete MMPs [
90]. Macrophages also produce MMP9 and TNF-related apoptosis-inducing ligands [TRAIL] that promote HSC apoptosis [
74]. A CD11b
hi/F4/80
infLY6C
low macrophage subpopulation was identified during the resolution of liver fibrosis; these special cells do not produce fibrogenic and/or inflammatory factors, but they continue to secrete MMPs including MMP9 and MMP12 [
59,
74,
89], and they upregulate CX3CR1. Compared with wild-type mice, mice deficient in CX3CR1 are associated with increased inflammatory cell accumulation and fibrosis after CCl
4 treatment [
74]. Mechanistic studies, combining RNA sequencing, functional in-vivo studies and co-culture experiments have revealed that mucosal-associated invariant T cell [MAIT] monocyte/macrophage interplay promotes liver fibrosis regression via reprogramming the macrophage phenotype [
13]. MAIT cells recruited to the fibrotic septa of the diseased liver and exhibit an activated phenotype, which directly impacts hepatic myofibroblasts and can contribute to the progression of fibrosis by enhancing a local inflammatory response [
13].
Liver inflammation is remarkably driven by extrahepatic signals originating from other regions such as gut or adipose tissue [
5]. In chronic liver disease, the gut-liver axis – which refers to the anatomical and physiological connection between liver and gut – is influenced by dietary fat and protein, bacterial metabolites, PAMPs and intestinal and adipose tissue hormones; this may promote the progression of liver disease [
91]. Also, the intestinal microbiome influences hepatic bile acid metabolism and the translocation of gut-derived signals due to leaky gut; these parameters may be found to be predictive of clinical outcomes in patients with chronic liver disease [
92,
93,
94]. In NASH, the gut-liver axis is activated and the interaction between liver injury, liver regeneration and increased gut permeability may foster inflammatory, pro-fibrogenic, and pro-carcinogenic pathways [
93]. As a result of the gut’s permeability defect, the intestinal microbiome and the liver interact through bacteria and bacterial metabolites, which affects liver metabolism and further drives hepatic inflammation [
95]. The gut permeability defect leads to the translocation of microbial products such as LPS, which elevates their serum levels and promotes the proinflammatory response elicited by hepatic macrophages [
96]. In addition, dysfunctional gut permeability enables PAMPs and DAMPs to enter the liver where they activate immune cells and engage with hepatic Toll-like receptors [TLRs] to induce proinflammatory and fibrotic pathways [
97,
98]. Furthermore, the gut microbiome has a role in controlling the composition of bile acids and modifying secondary bile acids before being recycled to the liver [
99]. Bile acids, such as cholic and chenodeoxycholic acid [CDCA], are important signaling molecules and are synthesized from cholesterol in hepatocytes surrounding the hepatic central vein [
100]. Their synthesis is regulated by the farnesoid X receptor [FXR] mainly via downstream targets [
101]. If low levels of primary bile acids reach the small intestine, proinflammatory bacteria proliferate and the production of toxic bile acids increases, which promotes liver damage and inflammation [
102]. Conversely, hepatocellular FXR activation by different bile acids prevents upregulation of inflammatory response genes and induces cell survival [
103]. Therefore, the composition of the bile acid pool is an important determinant in the clinical outcomes of liver inflammation and fibrosis, which is closely related to gut microbiome dysbiosis [
103]. In metabolic liver disease, free fatty acids flowing from adipose tissue to the liver may promote lipotoxicity and inflammation, particularly in patients with insulin resistance [
104]. Furthermore, experimental trials have revealed that adipose tissue-derived leptin activates KCs and increases their responsiveness to endotoxin, which results in inflammation [
105,
106]. Free fatty acids and ethanol can also drive liver inflammation.
2.3. Hepatocytes
Hepatocytes represent approximately 80% of the liver’s total cell number [
4,
5]. Under physiological conditions, they have several functions, such as biotransformation/detoxification and the production of bile as well as numerous proteins and lipids [
4,
5]. Hepatocytes are also a primary target for toxic metabolites influencing the liver [
2,
4,
68]. Regardless of the underlying etiology, liver injury damages hepatocytes, which thereupon release intracellular molecules such as DAMPs that drive chronic inflammation and fibrogenesis [
68]. These molecules are recognized by the innate immune system via pattern recognition receptors that oftentimes are the same molecular sensors that detect pathogens [
68]. Due to their critical metabolic function in the body, hepatocytes are abundantly furnished with mitochondria, with a stunning number of 1,000-2,000 of these organelles per hepatocyte [
107]. Mito-DAMPs released from damaged hepatocytes, including adenosine triphosphate [ATP], DNA fragments, and fatty acids promote the recruitment of resident and infiltrated macrophages, neutrophils, and NK cells [
68,
108]. In fact, mito-DAMPs derived from damaged hepatocytes may be among the most abundant and potent danger signals perpetuating the innate immune response [
108]. Mitochondrially originated danger signals from damaged hepatocytes directly activate HSCs and drive the progression of liver fibrosis [
68]. Nucleotide-binding oligomerization domain-like receptors [NLRP3] are a fundamental component of the inflammasome and the downstream targets of DAMPs [
59,
107]. Therefore, DAMPs exhibit a pivotal role in fibrogenesis and inflammation [
4,
5,
107]. During hepatic fibrogenesis, hepatocytes alter their gene expression and secretion profile [
2,
4,
5]. In addition, certain hepatocyte-derived molecules including Notch, osteopontin, TGFβ, NADH oxidase 4 and Indian Hedgehog have fibrogenic properties [
109]. Moreover, damaged hepatocytes secrete exosomes that,
inter alia, encapsulate micro-RNAs that promote HSC activation [
2]. It is generally accepted that inflammation is a fundamental component in the process of liver fibrogenesis, while factors secreted from damaged hepatocytes alone cannot directly activate HSCs [
2,
68,
107]. Ballooned hepatocytes display profibrogenic features and promote HSC activation by producing sonic hedgehog [
2]. Hepatocytic cholesterol accumulation also results in HSC activation by stabilizing the transcriptional regulator TAZ in hepatocytes, which leads to the secretion of the profibrotic factor Indian Hedgehog [
2,
110]. Fatty acids, such as palmitic acid, promote the release of extracellular vesicles loaded with microRNAs [e.g., miR-128-3p]. These micro-RNAs, when released from the extracellular vesicles upon their uptake by HSCs inhibit the expression of peroxisome proliferator-activated receptor [PPAR]γ, which shifts quiescent HSCs to the activated HSC phenotype [
108].
2.4. Liver Sinusoidal Endothelial Cells
Liver sinusoidal endothelial cells [LSECs] are unique endothelial cells that form the boundary of the hepatic sinusoids [
110]. They are characterized by a lack of an organized basement membrane and the presence of open fenestrae that form a permeable barrier, which enables a direct connection between hepatocytes and access to oxygen, micronutrients and macronutrients from the vessels [
110,
111,
112]. In addition, they are permanently exposed to molecules translocated from the gut [
110,
111]. LSECs have an essential role in the maintenance of hepatic homeostasis, including regulation of the vascular tone, in inflammation and thrombosis, and in the control of hepatic immune responses [
110]. In the healthy liver, differentiated LSECs are gatekeepers of fibrogenesis by maintaining HSCs in their quiescent state [
111,
112]. They play an important role in the secretion of vasoactive molecules, such as nitric oxide [NO] [
113]. In addition, they act as regional antigen-presenting cells [APCs] that govern – in addition to hepatic dendritic cells and KCs – intrahepatic immune responses by secreting cytokines and by activating or inhibiting immune cell signaling pathways [
114,
115]. LSECs actively participate in the clearance of antigens reaching the liver sinusoids and contribute to the perpetuation of the tolerogenic state [
116,
117]]. They orchestrate the sinusoidal blood flow via their impact on HSCs and thus perpetuate a low portal pressure [
111]. Furthermore, LSECs drive intrahepatic coagulation by several mechanisms, such as the production of pro- and anticoagulant factors, the recruitment and activation of neutrophils, and the crosstalk with platelets [
116,
117]. The fenestrated LSEC pheno type is orchestrated by two VEGF-dependent pathways, i.e., the VEGF-eNOS-soluble guan ylate cyclase [sGC]-cGMP pathway and a VEGF-dependent, NO-independent pathway [
118].
LSECs play a critical role at all stages of chronic liver disease through four processes: sinusoid capillarization, angiogenesis, angiocrine signals and vasoconstriction [111, 114,115]. Chronic liver injury results in the capillarization of LSECs – also called dedifferentiation – which thus lose their vasoprotective features and assume pathologic vasoconstrictive, proinflammatory and prothrombotic properties [
111,
118]. Capillarization refers to a lack of fenestration and the development of a more continuous basal membrane [
110,
111,
118]. In-vitro studies with LSECs cocultured with HSCs revealed the role of capillarized LSECs on HSC activation; specifically, LSECs from normal rat liver prevented HSC activation and induced their reversion to a quiescent phenotype, whereas capillarized LSECs were not capable to induce this phenotype [
118,
119]. A study in cirrhotic rats revealed that the administration of an sGC activator that induces the VEGF-eNOS-sGC-cGMP pathway, which restored LSEC fenestration, resulting in HSC quiescence and cirrhosis regression [
120]. Oxidative stress related to NO signaling was implicated in LSEC dysfunction, resulting in fibrogenesis [
117]. For example, reduced NO bioavailability has been observed in LSECs isolated from cirrhotic rat liver [
117]. Capillarized LSECs secrete several cytokines and soluble molecules influencing neighboring cells, promoting their dedifferentiation and driving the development of chronic complications of liver disease, including portal hypertension [
120]. Preclinical models of liver fibrosis have revealed increased production of vaso constrictors by LSECs [
121]. Activation of the cyclooxygenase 1 [COX1]-TXA2 pathway and endothelin 1 in rat chronic injury participate in sinusoidal contraction and exacerbate microvascular dysfunction [
121,
122]. Besides increasing vasoconstriction, capillarized LSECs have a defective eNOS-NO pathway; this entails portal hypertension and endothelial dysfunction [
110]. Downregulation of eNOS and bioavailability have been shown in cirrhotic rat liver [
123]. Gracia-Sancho et al. demonstrated that elevated hepatic oxidative stress in preclinical cirrhosis and impaired activity of the transcription factor KLF2 further contribute to diminish NO availability and exacerbate sinusoidal vasoconstriction [
117]. KLF2 is a nuclear transcription factor sensitive to shear stress enabling endothelial vasoprotection [
110,
118]. In experimental models of liver cirrhosis, liver endothelial KLF2 is upregulated as a compensatory mechanism driving the transcription of its vasoprotective target genes [
124]. LSECs may also play a role in the pathogenesis of portal hypertension through a dysregulation of their antithrombotic capacity [
125,
126]. LSEC capillarization and the loss of the KLF2-dependent vasoprotective pathways promote the recruitment and activation of platelets [inducing microthrombosis and fibrin deposition within the hepatic sinusoids], which results in hypoxia and sinusoidal hypertension [
110,
125,
126].
LSECs can maintain HSCs quiescent as long as they are differentiated [
119,
120]. VEGF contributes to the maintenance of LSEC differentiation [
119]. There is a close relationship between liver fibrosis and angiogenesis in that fibrosis fosters angiogenesis and, in turn, liver angiogenesis exacerbates liver fibrosis [
127,
128]. Besides LSECs, endothelial progenitor cells [EPCs], i.e., bone marrow-derived endothelial cells from, also contribute to intrahepatic angiogenesis [
129,
130]. LSECs also orchestrate fibrosis through secreting angiocrine signals, paracrine molecules produced by endothelial cells that maintain organ homeostasis and regulate organ regeneration and tumor growth [
131]. A recent study revealed that LSECs produce distinct angiocrine signals that maintain the balance between liver regeneration and fibrosis. Following acute liver injury, activation of the CXCR7-Id1 pathway in LSECs triggers expression of hepatotropic angiocrine factors that drive liver regeneration. Conversely, chronic liver injury leads to persistent FGFR1 activation in LSECs, which inactivates the CXCR7-Id1 pathway and provides a CXCR4-driven pro-fibrogenic angiocrine response, thereby provoking liver fibrosis [
131]. Importantly, endothelial dysfunction occurs early in chronic liver disease, even before the onset of fibrosis and inflammation [
131].
Figure 2 summarizes the most important molecular pathways and cellular interactions related to the activation and deactivation of HSCs detailed above. Further influencing factors are described below this figure.
Figure 2.
Molecular pathways and cellular interactions involved in HSC activation and deactivation. Activated HSCs are the main effector cells during hepatic fibrosis. In the healthy liver, they metabolize and store retinoids. Upon activation by fibrogenic stimuli, quiescent HSCs transdifferentiate into myofibroblasts, lose their vitamin A, upregulate α-smooth muscle actin [αSMA] and produce collagen I. Various factors, including immune cell-derived fibrogenic molecules, growth factors, lipopolysaccharide as well as profibrotic lipid mediators such as lysophosphatidylinositol and lysophosphatidic acid induce HSC activation in the course of chronic liver disease. TGFβ is the most HSC potent activator, which is produced by infiltrating lymphocytes and monocytes, Kupffer cells [KCs], and damaged hepatocytes. IL-17, produced by neutrophils and TH17 cells, sensitizes HSCs to TGFβ by upregulating TGFβ receptor II [TGFβRII]. In addition, platelet-derived growth factor [PDGF], which is produced by endothelial cells and macrophages, further promotes HSC activation. During fibrosis resolution, HSCs either die or revert to an inactive state by upregulating transcription factors such as peroxisome proliferator-activated receptor-γ [PPARγ], GATA-binding factor 4 [GATA4], GATA6 and transcription factor 21 [TCF21]. NK and CD8+ T cells can eliminate activated HSCs by inducing apoptosis. [Further abbreviations: GM-CSF – granulocyte/macrophage colony-stimulating factor; HH – hedgehog ligands; IHH – Indian hedgehog; LPA – lysophosphatidic acid; LPI – lysophosphatidylinositol; LPS- lipopolysaccharide; miRNA – microRNA; MSR1 – macrophage scavenger receptor 1; NF-κB – nuclear factor κ-light chain-enhancer of activated B cells; OPN – osteopontin; oxLDL – oxidized low-density lipoprotein; ROS – reactive oxygen species; S1P – sphingosine-1-phosphate; SHH – sonic hedgehog; TLR4 – toll-like receptor 4.].
Figure 2.
Molecular pathways and cellular interactions involved in HSC activation and deactivation. Activated HSCs are the main effector cells during hepatic fibrosis. In the healthy liver, they metabolize and store retinoids. Upon activation by fibrogenic stimuli, quiescent HSCs transdifferentiate into myofibroblasts, lose their vitamin A, upregulate α-smooth muscle actin [αSMA] and produce collagen I. Various factors, including immune cell-derived fibrogenic molecules, growth factors, lipopolysaccharide as well as profibrotic lipid mediators such as lysophosphatidylinositol and lysophosphatidic acid induce HSC activation in the course of chronic liver disease. TGFβ is the most HSC potent activator, which is produced by infiltrating lymphocytes and monocytes, Kupffer cells [KCs], and damaged hepatocytes. IL-17, produced by neutrophils and TH17 cells, sensitizes HSCs to TGFβ by upregulating TGFβ receptor II [TGFβRII]. In addition, platelet-derived growth factor [PDGF], which is produced by endothelial cells and macrophages, further promotes HSC activation. During fibrosis resolution, HSCs either die or revert to an inactive state by upregulating transcription factors such as peroxisome proliferator-activated receptor-γ [PPARγ], GATA-binding factor 4 [GATA4], GATA6 and transcription factor 21 [TCF21]. NK and CD8+ T cells can eliminate activated HSCs by inducing apoptosis. [Further abbreviations: GM-CSF – granulocyte/macrophage colony-stimulating factor; HH – hedgehog ligands; IHH – Indian hedgehog; LPA – lysophosphatidic acid; LPI – lysophosphatidylinositol; LPS- lipopolysaccharide; miRNA – microRNA; MSR1 – macrophage scavenger receptor 1; NF-κB – nuclear factor κ-light chain-enhancer of activated B cells; OPN – osteopontin; oxLDL – oxidized low-density lipoprotein; ROS – reactive oxygen species; S1P – sphingosine-1-phosphate; SHH – sonic hedgehog; TLR4 – toll-like receptor 4.].
2.6. Other Immune Cells in the Pathogenesis of Liver Fibrosis
Immune cells, such as T lymphocytes, macrophages, DCs, granulocytes, and mast cells, play important roles in the development and progression of fibrosis [
132]. These immune cells produce many mediators driving inflammation, fibrogenesis as well as the activation of fibroblasts and T lymphocytes, such as CD4
+ and CD8
+ T cells [
132,
133]. In patients with chronic liver disease, the number of effector CD8
+ T cells increases [
132]. Innate lymphoid cells [ILCs] constitute a subset of innate immune cells with lymphoid phenotypes; however, they do not express rearranged antigen receptors [
132,
133,
134]. ILCs are classified into three groups based on the production of specific transcription factors, cell-surface markers, and signature cytokines [
134]. Group-1 ILCs [a.k.a. ILC1s] consist of IFN-γ-producing and T-bet-dependent ILCs; group-2 ILCs [a.k.a. ILC2s] cover a subset of cells expressing type-2 cytokines such as IL-5 and IL-13; and group-3 ILCs [a.k.a. ILC3s] produce IL-17 and IL-22 [
133,
134]. Recently, this classification has been revised, and two novel members have been added to the ILC family, which are conventional NK [NK] cells and lymphoid tissue-inducer cells [
134]. ILCs, particularly ILC1s and ILC3s, are involved in liver fibrogenesis [
82,
83]. Recent studies revealed that ILC1s are recruited in adipose tissue of type-2 diabetes patients, where inducing fibrogenesis via the TGF-β signaling pathway [
132,
133]. ILC3s expressing IL-17 are increased in CCL4-induced cirrhotic mice [
133,
134]. Adoptive transfer of ILC3s into ILC-depleted mice led to ECM accumulation and induced liver fibrosis. These findings indicated a pro-fibrogenic role of ILC3s in the progression of liver fibrosis [
134,
135]. Clinical and experimental studies have demonstrated that cNK cells are critically involved in the immune pathogenesis of chronic liver disease [CLD], and they display antifibrotic activity through the induction of apoptosis and/or killing of activated HSCs [
132,
133,
135]. The antifibrotic activity of NK cells is associated with the surface expression of activating NK cell receptors, including NKG2D, NKp46, and NKp30, which recognize specific molecules produced by activated HSCs [
134,
135,
136]. Forkel et al. revealed a link between the severity of liver fibrosis and the proportion of intrahepatic ILC2s, which may express IL-13 and exhibit pro-fibrotic activity [
137].
Myeloid dendritic cells [DCs] [in the DC-specific literature differentiating between three principal DC subclasses, this subclass is dubbed “classical” or “conventional”] assume a critical role in the development of liver fibrosis [
138]. This is due to the fact that DCs serve as key professional APCs that regulate and orchestrate innate and adaptive immune responses in infections, chronic inflammatory diseases, cancer, autoimmunity and, conversely, the induction of immune tolerance [
132,
138]. CCl
4-induced liver fibrosis is more pronounced in HBV-transgenic mice as opposed to wild-type mice [
139]. Depletion of NK cells and NKT cells or the blockade of CD1d inhibit α-SMA expression in the liver [
139]. The number of mucosal-associated invariant T cells [MAIT cells] decreases in cirrhotic patients [
140]. MAIT cells can promote HSC proliferation and the production of collagen and proinflammatory cytokines in vitro [
140]. Increasing evidence suggests that adaptive immune cells – including Th1, Th2, and Th17 cells – also play important roles in liver fibrogenesis [
141]. Novel studies revealed that Tregs have a relevant role in the development of liver fibrosis, and it has been shown that signaling via the mammalian target of rapamycin [mTOR] is involved in orchestrating the protective function of Tregs [
132,
141]. Finally, B cells also promote the development and progression of liver fibrosis [
132,
141]. It has thus become increasingly evident that the great majority of immune cell populations and subpopulations normally participating in the realization of innate and adaptive immune responses are recruited in the context of liver fibrosis. While this certainly is in part due to fibrosis-specific activation processes, it can be reasonably assumed that this inflationary development also is a result of the excessive amount of soluble and cell membrane signals expressed in the progression of fibrosis. If so, any therapeutic efforts towards downregulating detrimental immunological processes involved in liver fibrogenesis will be increasingly difficult the longer this process continues.
2.7. Metabolic Reprogramming of HSCs in Liver Fibrogenesis
HSCs regulate their energy expenditure to perpetuate their distinct functions during the development and progression of liver fibrosis [
142]. Recent studies revealed that cells involved in the development and regression of liver fibrosis undergo metabolic reprogramming to meet their energy requirements [
141,
142,
143]. The transition of quiescent to activated HSCs requires a high energy demand and therefore a reprogramming of cellular meta bolic pathways [
2,
118]. This prominently includes significant changes in carbohydrate cata bolism – such as the upregulation of glycolysis – for energy supply during the cells’ transition to myofibroblasts [
142]. HSCs transform glucose to lactate to avoid oxidative phosphorylation [aerobic glycolysis], which has a weaker effect than oxidative phosphorylation in generating ATP [
1,
2,
142]. Lactate reduces the extracellular pH and induces TGFβ1 activation [
1]. The combination of low extracellular pH with an increased lactic acid level promotes myofibroblast transformation through the activation of TGFβ1 [
1,
145,
146]. In the course of glycolysis, pyruvate is transformed to acetyl-CoA within the mitochondria [
146]. Recent data documented that lactate is a significant metabolite in the activation and perpetuation phase of HSCs, and HSC activation requires more metabolic demands than ATP generation only [
1,
142]. Activated HSCs require high levels of aerobic glycolysis for the transition into the myofibroblast phenotype. Oxidative phosphorylation is one of the significant energy sources of activated HSCs, an indication of which is the increased number and activity of mitochondria [
1,
142]. This effect can be explained by the fact that these cells depend on both oxidative phosphorylation-generated ATP and mitochondria-derived ROS [
142]. Excessive ROS production activates TGFβ signaling and stimulates inflammatory cells that, as detailed further above, contribute to hepatic fibrogenesis [
1,
142].
Preclinical trials using immortalized human activated HSCs or primary murine HSCs, respectively, demonstrated an increase in enzymes that process glucose intracellularly after activation, including hexokinase 2 [HK2], fructose-2,6-biphosphatase-3 [PFKFB3], and pyru vate kinase [PK] [
1,
58,
142]. HSCs upregulate their glycolytic pathway [
1]. In addition, activated HSCs downregulate the expression of proteins involved in gluconeogenesis, including phosphoenolpyruvate carboxykinase-1 [PCK1] and fructose bisphosphatase-1 [FBP1], and remove central carbon metabolites from the TCA cycle to facilitate lactate accumulation [
1,
43]. However, these findings require to be validated under physiological conditions as the data stem from in-vitro experiments using primary or immortalized HSCs [
1,
4,
53,
58]. The metabolic reprogramming of HSCs and the enzymes involved in aerobic glycolysis are regulated by the activation of the Hedgehog [Hh] pathway through the expression hypoxia-inducible factor 1-α [HIF-1α] together with TGF-β1 [
43]. In addition, glutaminyl and protein metabolism are also upregulated by some enzymes, such as glutaminase 1 [GLS-1] [
142]. During activation, HSCs have to master a significant bioenergetic challenge to fuel all their secretory functions, and their protein metabolism is also reprogrammed. Du et al. showed the expression of genes involved in protein metabolism during carbohydrate metabolism [
145]. They also reported that approximately 38% of genes differently expressed by quiescent HSCs and activated HSCs are involved in protein metabolism [
75]. Glutaminolysis is the transformation of glutamine into α-ketoglutarate, which is usually identified in cancer cells and enables the generation of ATP needed for cell anabolism [
75,
142,
145,
146]. As previously mentioned, the utilization of vitamin A depots for the catabolism of fatty acids to enable energy supply is one of the hallmarks of HSC activation [
1,
2,
4,
53,
58]. During this process, LRAT expression is substantially reduced in activated HSCs, causing a decrease in vitamin A storage and the progression of fibrosis [
142,
145,
146,
148].
The metabolism of lipid droplets during HSC activation generates fatty acids for β-oxi dation [
43,
147,
148,
149]. HSC activation is also promoted by transcriptional drivers of fatty acid content, including PPARγ and the sterol regulatory-element-binding protein-1 [SREBP-1c]. Increased glycolysis and glutaminolysis, together with enhanced fatty acid oxidation, have been considered to be relevant drivers of fibroblast activation [
150]. Activated HSCs have been subdivided into three classes based on their expression profiles, being pro-regenerative [as characterized by increased growth factor expressions], anti-regenerative, and a mixed phenotype [
142]. During activation, genes involved in retinol catabolism, including retinyl ester hydrolase [REH], are upregulated, while enzymes involved in retinol esterification, including LRAT, are downregulated [
142]. As a result, lipid droplets are lost and metabolized to activate the β-oxidation pathway. Enzymes involved in lipid metabolism, including the liver X receptors [LXRs], are upregulated, while adipogenic regulators, including sterol regulatory element-binding protein 1 [SEREB-1c], are downregulated [
142]. Activated HSCs have been shown to increase the rate of aerobic glycolysis and related enzymes while decreasing enzymes involved in gluconeogenesis [
151]. Basal levels of glycolysis and mitochondrial respiration are substantially higher in rat activated HSCs when compared with quiescent HSCs [
151]. This finding was associated with extensive mitochondrial fusion in rat and human activated HSCs, which evolved without changes in mitochondrial DNA content and electron transport chain [ETC] components [
151]. ROS accumulates intracellularly and in the oxidative phosphorylation pathway, while the tricarboxylic acid [TCA] pathway is downregulated [
151]. Experimental trials detected that RNA-binding proteins such as polyadenylation-element-binding protein 4 [CPEB4], human antigen R [HuR], and tristetraprolin [TTP] were identified to be critical regulators of processes in HSCs [
142].
2.8. Metabolic Regulation of Liver Fibrosis
Autophagy is a stress response mechanism that involves the degradation of cellular components and organelles through a lysosome-dependent pathway to generate energy and nutrients; this mechanism has been shown to also play a critical role in HSC activation and fibrogenesis [
2]. Autophagy is required to perpetuate an activated phenotype in HSCs [
2]. In mice with autophagy-defective HSCs, CCl
4-induced liver injury leads to the cessation of ECM deposition and development of fibrosis [
2,
5]. Several profibrogenic and proinflammatory molecules, such as TGFβ and lipopolysaccharide upregulate autophagy in HSCs [
152,
153]. TGFβ-induced autophagy displays a role in HSC activation through the c-jun
N-terminal kinase and extracellular signal-regulated kinase signaling pathways [
152]. Lipopolysaccharides-induced upregulation of autophagy modulates the suppression of the TGFβ pseudo receptor BAMBI [
153]. In addition, macrophage-derived PGE2 can drive HSC activation and fibrosis through promoting autophagy [
154]. In a mouse model of diet-induced fatty liver disease, M2 macrophages were found to induce HSC autophagy by expressing PGE
2, ultimately fostering HSC activation, ECM production, and fibrosis develop ment [
154]. Antagonization of the PGE/EP4 pathway suppresses HSC autophagy and regresses liver fibrosis [
154].
Autophagy drives HSC activation through fatty acids formed as a result of cleavage of retinyl esters within lipid droplets [
109,
155]. In experimental mouse models of CCl
4-induced liver fibrosis, autophagy-related protein 7 [ATG7], fibrogenesis, and ECM accumulation were found to be remarkably reduced, but this finding was not observed in cultured HSCs when exogenous fatty acid was added [
156]. Endoplasmic reticulum [ER] stress signals promote gene expression in HSCs by inducing autophagy and HSC activation [
118,
157,
158]. Targeted lentiviral delivery of GRP78 protein, an ER stress protein, can inhibit fibrogenesis following CCl
4 treatment. Inhibition of the Inositol-requiring enzyme 1 [IRE1] in HSCs decreases both their activation and autophagic activity and leads to reduced fibrogenic activity [
118,
157,
158]. Ectopic overexpression of X-box binding protein [XBP1], a transcription factor downstream of IRE1, promotes type 1 collagen production in HSCs, which is suppressed by the knockdown of ATG7 [
2,
118,
158]]. Another signaling pathway activated by ER stress is PKR-like endoplasmic reticulum kinase [PERK], which stimulates HSC activation. ER stress in HSCs induces hepatic fibrosis through dysregulation of miR-18, which is regulated by activation of PERK and destabilization of heterogeneous nuclear ribonucleoprotein A1 [HNRNPA1] [
118,
158]. Heat shock protein 47 [HSP47] is a collagen-specific molecular chaperone that has critical roles in the expression of type 1 collagen [
2,
4,
5,
155]. In HSP47-depleted HSCs, immature type 1 procollagen accumulates [
1,
2,
4,
5,
155]. When autophagy is inhibited in these cells, ER stress and apoptosis are increased compared with wild-type HSCs [
4]. JANK signaling, which merges downstream of ER stress, was associated with hepatocyte damage, NAFLD, and hepatic fibrosis [
1,
4]. JNK1 signaling in HSCs strongly promotes liver fibrosis; however, JNK1 signaling does not affect hepatocytes in a similar manner [
1,
4]. An experimental trial in mice has indicated that liver fibrosis is substantially diminished in JNK1-deficient mice compared with wild-type mice and in those with hepatocyte-specific JNK1 deficiency in response to CCl
4 or BDL [
159].
2.9. Epigenetic Regulation of HSCs
Recent data indicate that the transition of quiescent HSCs to activated HSCs requires an epigenetic mechanism to silence adipogenic differentiation factors and enhance the de-novo expression of genes related to novel phenotypes [
2,
4,
5]. There are three fundamental mechanisms of epigenetic regulation, i.e., noncoding RNA [ncRNA] expression, DNA methylation, and histone modification [
160]. Among several endogenous factors regulating gene expression, certain miRNAs are essential [
161]. In chronic liver disease, some signaling pathways are promoted by inflammatory molecules triggering liver fibrosis; these signaling pathways are orchestrated by miRNAs [
160,
161,
162]. MiRNAs are single-stranded noncoding RNAs containing about 18-25 nucleotides and drive post-transcriptional gene expression through altering mRNA degradation [
163]; they display either pro-fibrotic or antifibrotic effects [
162,
163]. Experimental studies identified several profibrogenic miRNAs, including miR-21, miR-221, miR-222, and miR-27 [
162,
163]. Conversely, miR-214, miR-378a, miR148a, miR-133a, and miR-195 display antifibrotic effects [
162,
163]. The miR-15 family induces cell proliferation and promotes apoptosis, while the miR-29 family orchestrates the accumulation of ECM and promotes apoptosis by modulating the P13/AKT signaling pathway [
162]. MiR-29 family members promote various signaling pathways, such as TGFβ, NF-κB, and P13K/AKT, promoting liver fibrosis [
144,
162]. The interaction between miR-29b and the TGFβ/Smad3 signaling pathway arises in activated HSCs. Smad3 negatively regulates miR-29b expression, which directly drives the TGFβ/Smad3 pathway and induces liver fibrosis [
164]. Additionally, miR-34 stimulates HSCs and promotes liver fibrosis in rats by inducing acyl-CoA synthetase, which exhibits a critical role in hepatic lipid metabolism [
144,
162,
164]. Furthermore, a recent trial revealed that a lack of miR-21 in NASH-associated chronic liver disease leads to decreased steatosis, inflammation, and lipoapoptosis, resulting in decreased fibrosis [
142]. Myocardin-related transcription factor A [MRTF-A] drives profibrogenic transcription by accumulating a histone methyltransferase complex to the promoters of fibrogenic genes [
1,
4,
5]. MRTF-A-deficient mice show resistance to CCl
4-induced fibrosis [
1,
109]. Long noncoding RNAs [lncRNAs] are a class of transcripts with more than 200 nucleotides in length, which function as RNAs but do not encode proteins [
165]. Emerging studies have revealed that lncRNAs are relevant drivers in fibrogenesis [
165]. LncRNAs extensively modulate gene expression at transcriptional, post-transcriptional, and epigenetic levels by interacting with proteins, DNA or mRNAs [
165]. Studies in multidrug resistance 2 knockout [Mdr2
-/-] mice clearly revealed that cholangiocyte-derived exosomal H19 is transferred to adjacent hepatocytes and HSCs, which propels the progression of cholestatic liver fibrosis by modulating bile acid accumulation in hepatocytes and promotes the activation and proliferation of HSCs [
165,
166]. Studies reported that lncRNAs can interplay with the enhancer of zeste homolog [EZH2] and orchestrate fibrosis progression [
165].
As mentioned above, activated HSCs alter their gene expression profile and transdifferentiate into a profibrogenic myofibroblast phenotype. The existence of various transcription factors was suggested to orchestrate this process, but suitable models on the reprogramming of the myofibroblast epigenome are still missing [
167]. Epigenetic mechanisms may participate in these signature transcriptional processes during HSC activation [
167,
168]. DNA CpG methylation exhibits a role in the global DNA alteration during HSC activation [
168]. Methylated CpG motifs within the promoter of PPARγ accumulate methyl-CpG-binding protein 2 that subsequently promotes H3K9 methylation and accumulates transcriptional repressor chromobox protein homolog 5, which inhibits PPARγ production and drives HSC activation [
169]. Page et al. investigated whether HSC activation may be accompanied by DNA methylation remodeling; they compared global changes of 5-metylcytosine and 5-hydroximethycytosine during HSC activation [
168]. Methylation of CpG plays an essential role in gene silencing and 5-mC may be additionally oxidized to secrete 5-hmC, which is generally detected in transcriptionally active genes [
167]. The researchers also investigated the production of DNA methyltransferases [DNMT1, 3a, and 3b] orchestrating annotation of 5-mC and methylcytosine dioxygenase [TET] that oxidize 5-mC to 5-hmC [
168]. In this study, the production of TET proteins has been found to be uniformly reduced in both animal and human models, with a concurrent decrease in global levels of 5-hmC in fibrotic livers compared with healthy controls [
168]. In contrast, the expression of DNMT proteins tended to increase in fibrotic liver, but the increase in DNMT level did not change the global amount of 5-mC, which may be due to changes in DNMT activity [
168]. The researchers suggest that DNA 5-mC/5-hcM is a pivotal step in HSC activation and fibrogenesis [
168]. Changes in DNA methylation during HSC activation may add novel perspectives to the molecular mechanisms underlying fibrogenesis and may lead to the discovery of potential novel drugs and biomarkers that might be used to prevent or delay the progression of liver fibrosis.
Liver fibrosis is further associated with alterations in DNA methylation patterns and the expression of epigenetic enzymes involved in local fibrogenesis [
161,
162]. A genome-wide study of DNA methylation patterns in CCl
4 mouse liver tissues revealed hypomethylation of fibrosis-related genes before the onset of liver fibrosis [
170]. Furthermore, on the third day of culture, a ~60% loss of original DNA methylation levels was detected in rat HSCs, a condition that triggered fibrogenic activity [
171]. Although the transition of HSCs to myofibroblasts is associated with a general loss of DNA methylation, the presence of gene-specific DNA hypomethylation and hypermethylation has been documented in genome-wide DNA methylation studies [
171,
172]. For instance, upregulation of profibrogenic genes such as Actg2, Loxl1, Loxl2, and Col4A1/2 is associated with a decrease in promoter methylation levels in activated HSCs [
173]. Conversely, downregulation of Smad7, and Pten genes is associated with DNA hypermethylation [
174]. DNA methylation is orchestrated through the attachment of methyl-binding proteins, which subsequently accumulate the transcriptional repressor complex [
175]. The expression of MeCP2, a methyl-binding protein, is promoted during the transition of HSCs to myofibroblasts [
176]. MeCP2-deficient mice displayed attenuation of the expression of fibrogenic markers such as collagen-1, TIMP-1, and α-SMA and regression of liver fibrosis after CCl
4 treatment [
177]. PPARγ must be silenced for HSCs to be activated and to transdifferentiate into the myofibroblast phenotype [
178]. MeCP2 silences PPARγ via different mechanisms [
178]: Specifically, MeCP2 represses PPARγ expression by binding to methyl-CpG residues in the PPARγ promoter or by enhancing EZH2 expression [
178]. CpG methylation is driven through DNA methyltransferases [DNMTs] [
42,
57]. DNMT-mediated hypermethylation of phosphatase and tensin homolog [PTEN] initiates the activation of HSCs [
39,
49]. Many studies revealed the key role of histone methylation in liver fibrosis. The MeCP2-mediated transdifferentiation of HSCs is, in part, regulated by two distinct histone methyltransferases, i.e., EZH2 and ASH1 [
179]. Their expression is promoted during HSC activation, leading to fostering H3K27me3 and H3K4me modifications, respectively [
162]. Overexpression of EZH2 promotes the expression of fibronectin, α-SMA, and collagen 1α1 in HSCs [
180]. Studies in in-vitro and in-vivo models of CCl
4 and BDL-induced liver fibrosis have demonstrated that EZH2 inhibitors such as 3-deazaneplanocin A [DZNep] and GSK-503 have antifibrotic properties [
181]. Methyl-CpG binding protein and histone-lysine N-methyltransferase enhancer of zest homolog 2 [EZH2] regulate epigenetic signaling by means of repressing PPARγ [
182]. The Wnt/β-catenin signaling pathway activates HSCs by increasing MECP2 protein levels, which in turn represses PPARγ [
183].
Before discussing the mechanisms of regression of liver fibrosis, we present a sketch of the change in hepatic macrophage populations on the path towards liver fibrosis [
Figure 3].