Skip to main content

REVIEW article

Front. Med., 05 October 2015
Sec. Pathology
Volume 2 - 2015 | https://doi.org/10.3389/fmed.2015.00072

Targeted therapies in liver fibrosis: combining the best parts of platelet-derived growth factor BB and interferon gamma

  • 1Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, Groningen, Netherlands
  • 2Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, Groningen, Netherlands

Cytokines, growth factors, and other locally produced mediators play key roles in the regulation of disease progression. During liver fibrosis, these mediators orchestrate the balance between pro- and antifibrotic activities as exerted by the hepatic cells. Two important players in this respect are the profibrotic mediator platelet-derived growth factor BB (PDGF-BB) and the antifibrotic cytokine interferon gamma (IFNγ). PDGF-BB, produced by many resident and infiltrating cells, causes extensive proliferation, migration, and contraction of hepatic stellate cells (HSCs) and myofibroblasts. These cells are the extracellular matrix-producing hepatic cells and they highly express the PDGFβ receptor. On the other hand, IFNγ is produced by natural killer cells in fibrotic livers and is endowed with proinflammatory, antiviral, and antifibrotic activities. This cytokine attracted much attention as a possible therapeutic compound in fibrosis. However, clinical trials yielded disappointing results because of low efficacy and adverse effects, most likely related to the dual role of IFNγ in fibrosis. In our studies, we targeted the antifibrotic IFNγ to the liver myofibroblasts. For that, we altered the cell binding properties of IFNγ, by delivery of the IFNγ-nuclear localization sequence to the highly expressed PDGFβ receptor using a PDGFβ receptor recognizing peptide, thereby creating a construct referred to as “Fibroferon” (i.e., fibroblast-targeted interferon γ). In recent years, we demonstrated that HSC-specific delivery of IFNγ increased its antifibrotic potency and improved its general safety profile in vivo, making Fibroferon highly suitable for the treatment of (fibrotic) diseases associated with elevated PDGFβ receptor expression. The present review summarizes the knowledge on these two key mediators, PDGF-BB and IFNγ, and outlines how we used this knowledge to create the cell-specific antifibrotic compound Fibroferon containing parts of both of these mediators.

Introduction

Nowadays the most prevalent causes of chronic injury to the liver are chronic hepatitis B and C viral (HBV and HCV, respectively) infections, alcohol abuse, and metabolic problems related to obesity. Irrespective of the etiology of the disease, all types of chronic damage can finally lead to the induction of a common pathological response in the liver. In this common pathological response, the same cells and mediators play a role in the progression from chronic injury to fibrosis, cirrhosis, and eventually to tumor formation. At the moment, hepatocellular carcinoma is among the top three of most lethal carcinomas and its incidence is still increasing (1, 2).

Fibrogenesis is characterized by excessive fibrotic matrix deposition and by activation of macrophages and myofibroblasts. Within the liver, the major pool of matrix-producing myofibroblasts originates from activated and transformed hepatic stellate cells (HSCs), although other fibroblast sources such as the portal fibroblast or fibrocytes also have been identified (35). Clearly not only myofibroblasts and resident macrophages (Kupffer cells) are involved, but all other resident cell types contribute each in their own way to the fibrotic pathology. Additionally, infiltrating immune cells, both of the innate and acquired immune system, such as B cells, T cells, natural killer (NK) cells, NKT cells, neutrophils, and monocytes contribute to the disease progression. Their specific roles in the disease are becoming more and more clear (4, 6, 7). Although multiple cells are involved in hepatic fibrosis, crucial players are the fibroblasts, i.e., in the liver the activated HSCs and myofibroblasts, and macrophages, i.e., the Kupffer cells together with infiltrated monocytes. The fibroblasts are considered the master producers of all the fibrotic matrix proteins, whereas the macrophages are the master regulators (4, 5, 8).

Fibroblasts and macrophages communicate with each other via production and release of various growth factors, cytokines, and chemokines. The two most prominent profibrotic growth factors are platelet-derived growth factor BB (PDGF-BB) and transforming growth factor beta (TGFβ). While PDGF-BB stimulates fibroblast proliferation and migration, TGFβ is more involved in activation, transformation, and matrix production. In our studies, we chose the PDGFβ receptor (PDGFβR) as our target receptor for the delivery of antifibrotic compounds (9). The PDGFβR was preferred since the TGFβ receptor was not only expressed on HSCs and myofibroblasts but also hepatocytes and macrophages express this receptor (1012). Next to the high production of profibrotic factors in diseased livers, various antifibrotic factors are produced and released as well (13). One of the major antifibrotic factors is interferon gamma (IFNγ). This cytokine is excreted by infiltrating T cells and NK(T) cells (6, 14). However, as disease progression continues, the antifibrotic factors cannot effectively counterbalance all profibrotic stimuli, leading to disturbance of the homeostatic balance and thus worsening of the disease.

In this review, we will give an overview of the characteristics and pathological roles of two mediators in fibrosis, i.e., PDGF and IFNγ, and summarize their receptor expressions and counterbalancing activities in various cell types. After that, recent studies on the structure, the receptor interaction, the in vitro and in vivo characteristics of the cell-specific therapeutic compound Fibroferon (previously called BipPB-PEG-IFNγ mimetic, mimγ-BipPB, or BOT191), which contains the most useful parts of both these abovementioned mediators, are summarized.

Platelet-Derived Growth Factor

Platelet-derived growth factor, one of the first growth factors characterized, was discovered in the early 1970s as a key mediator involved in vascular pathologies such as atherosclerosis (15). Studies at that time proved that this growth factor, released from platelets, promoted vascular smooth muscle cell migration and proliferation. Later on, it became clear that PDGF was a potent mitogen for all mesenchymal cells, including fibroblasts and myofibroblasts. In later decades, PDGF was also identified as the most potent mitogen for HSCs in fibrotic livers (16). The distribution of the PDGF receptor correlated closely with the localization of activated mesenchymal cells predominantly present in collagen lesions within the liver (17).

Platelet-derived growth factor belongs to the large cysteine-knot superfamily. This family is characterized by the presence of eight conserved cysteine residues, forming a typical cysteine-knot structure. Within this large family, PDGF is structurally and functionally mostly related to the vascular endothelial growth factor (VEGF) family. The structure of the PDGF-family is highly conserved throughout the animal species (18, 19). Four different chains of PDGF-molecules are known: the traditional A- and B-chains and the more recently discovered C- and D-chains. The PDGF molecule is present in a dimeric form, and four homodimeric (AA, BB, CC, and DD) forms and one heterodimeric (AB) form are described. PDGF-A (211 aa, 24 kDa) and PDGF-B (241 aa, 27 kDa) are each other’s paralogs. Both proteins are processed mainly intracellularly and secreted in the dimeric active form. On the other hand, PDGF-C (345 aa, 39 kDa) and PDGF-D (370 aa, 43 kDa), also each other’s paralogs, are secreted in a dimeric latent form. These PDGF isoforms contain a so-called CUB (Complement C1r/C1s, Uegf, Bmp1) domain at the N-terminal region of these proteins. This CUB domain, mainly found in developmentally regulated extracellular and plasma membrane-associated proteins, causes retention of PDGF-CC and -DD in the extracellular matrix. For interaction with the PDGFR, extracellular activation of the C- and D-chain is necessary and it requires proteolytic cleavage of the CUB-domain by tissue plasminogen activator or urokinase plasminogen activator, respectively (18, 20). All five dimeric PDGF-forms can be found in the extracellular space but also in intracellular compartments, such as the endoplasmic reticulum and Golgi. There are even reports that state that the C-chain can be present in the nucleus (21).

Platelet-derived growth factor exerts its effects via binding to one of the two structurally related PDGF receptors, the PDGF alpha receptor (PDGFαR; Mw = 123 kDa) and beta receptor (PDGFβR; Mw = 124 kDa) that both belong to the class III receptor tyrosine kinases. Similar to the growth factor itself, also the receptor exists in homo (αα and ββ) and hetero (αβ) dimeric forms, all containing an extracellular part to which a particular PDGF-isoform binds and an intracellular signaling part. PDGF-A and C chains bind to the α-receptor, while B and D chains bind to the β-receptor. Both receptors are predominantly expressed on the cell membrane with minor expression in the lysosomes and even nucleus is reported (17, 19). In addition, PDGFαR was detected as a soluble receptor in the extracellular space (22).

Biological Effects

Platelet-derived growth factor has a clear role during embryonic development (18), but after that, in normal healthy tissues the activity and expression of the PDGF system is very limited. During pathological conditions, however, the PDGF system is activated again and diseases that are associated with overactivity of the PDGF system can be divided in three groups: tumors, vascular diseases, and fibrotic diseases. In fibrotic livers, several inflammatory and fibrotic mediators, such as interleukin-1β, tumor necrosis factor alpha (TNFα), TGFβ, and fibroblast growth factor (FGF), are released that can increase PDGF and its receptor expression. In addition, bacterial endotoxin (lipopolysaccharide), which is released in the serum after intestinal leakage and associated with liver fibrosis induction, can upregulate PDGFR expressions in the liver (23). The upregulation of the PDGFβ receptor in human cirrhotic livers was previously shown by Bansal et al. (24).

Various isoforms of PDGF are reported to be present in the fibrotic liver, and all these isoforms have mitogenic and chemoattractive effects on mesenchymal cells. PDGF-BB is the major stimulus for mesenchymal cell proliferation, and the PDGFβ receptor is primarily involved in this cellular effect of PDGF. Also, activation of the β-receptor stimulates chemotaxis, whereas activation of the α-receptor is associated with either stimulation or inhibition of chemotaxis, depending on the mesenchymal cell type. PDGF is known for its effects on intracellular actin reorganization resulting in stimulated cell migration, chemotaxis, and contraction. There is, however, a difference between both PDGF receptors regarding their effects on the actin filament system. Whereas both receptors stimulate edge ruffling and loss of stress fibers, only the β-receptor mediates the formation of circular actin structures on the dorsal surface of the cell. Gu et al. nicely showed that these circular structures are involved in rapid recruitment, internalization, and membrane relocation of surface integrins, and in this way, these circular structures may serve as indicators of cellular transition from static to motile states (25). Together with TGFβ, PDGF-BB also induces fibroblast activation and differentiation, resulting in induction of α-smooth muscle actin (α-SMA) expression in these cells (19, 26). Figure 1 gives an overview of the PDGF-BB-producing and PDGF-BB-responsive cells and its key functions.

FIGURE 1
www.frontiersin.org

Figure 1. Overview of PDGF-BB and IFNγ-producing cells and cells that respond to these mediators of fibrogenesis, illustrating the versatility of both mediators.

The PDGF isoform that is most prominent in liver fibrosis is PDGF-BB (16, 17). PDGF-BB is secreted by both resident and infiltrating cells of the liver, such as platelets, infiltrating monocytes and macrophages, activated Kupffer cells (M2-dominant type), hepatocytes, cholangiocytes, HSCs, and myofibroblasts, and it affects primarily HSCs and myofibroblasts. These latter cells display a highly induced PDGFβ receptor expression in mice and humans, and this expression can be related to the proliferative and migratory activities of these fibroblasts in fibrosis. Of note, activated HSCs and myofibroblasts produce PDGF-BB and thus create an autocrine profibrotic loop without interference of other cells.

Although PDGF production increases profoundly during the disease and thus local concentrations are high, the amounts present in the circulation are generally very low. While concentrations of only 17.5 ± 3.1 ng/ml were detectable in whole human blood, the amount in plasma was undetectable (27). The low concentrations in the circulation were due to a very short plasma half-life of less than 2 min and due to a high binding affinity to several proteins in plasma and extracellular spaces (27). Examples of PDGF-binding proteins are its soluble receptor, α2 macroglobulin, and PDGF-associated protein (PAP). Additionally, various extracellular matrix proteins are able to bind PDGF to form a local storage pool. This implies that the biological activities of PDGF can be modulated by binding to one of these molecules (28, 29).

PDGF-Receptor Expression

Platelet-derived growth factor α-receptor and PDGFβR have overlapping, but also distinct, expression patterns and physiological roles. Generally, PDGFαR signaling is important in embryonic development since it controls gastrulation and the embryonic development of several organs such as lung, intestine, skin, testis, kidneys, bones, and neuroprotective tissues. Some cell types only express the PDGFαR, such as platelets and liver sinusoidal endothelial cells. PDGFβR signaling is recognized as an essential regulator of early hematopoiesis and blood vessel formation. The classical target cells for PDGF, fibroblasts and smooth muscle cells, express both α- and β-receptors, although the expression of the PDGFβR is generally at a much higher level. In fibrotic livers, PDGFβR is mainly expressed on the cell membrane of activated and transformed HSCs or myofibroblasts in the collagenous fibrotic bands (17, 24, 30). Borkham-Kamphorst et al. showed that the PDGFRs in the endothelium and hepatocytes are more of the α-type (30). In addition, PDGFαR was stained outside the area of CCl4-induced hepatocellular fibrosis, and they speculated that this increased expression could be related to hepatocyte survival. Furthermore, they found that the upregulation of PDGFαR was accompanied by PDGF-A and PDGF-C staining in regenerative hepatocytes after CCl4 intoxication.

How PDGF exactly binds to its receptor is the subject of various studies. Insight into this interaction can lead to the generation of PDGF receptor antagonizing drugs or to drug targeting compounds. Already in early 1990s, Clements et al. identified arginine 27 and isoleucine 30 as the PDGF-B chain residues that mediate the binding of the growth factor to the β-receptor in rat and human cells (31). Binding to the α-receptor requires another more cationic part of the molecule (arginine159-lysine160-lysine161-lysine162), and both in the PDGF-A and -B chain this sequence is present (32). More recently, the interaction between the growth factor and the receptor was studied in more detail with modern techniques like crystallography, partly confirming the abovementioned regions in PDGF, but also pointing to additional points of interaction with its receptors (33, 34).

PDGF-Receptor Signaling

After binding of PDGF to one of the receptor chains, dimerization of the receptor is induced and this receptor dimer is then stabilized by the ligand. The subsequent interaction between the cytoplasmic domains of the two receptor chains induces phosphorylation of intracellular tyrosines leading to conformational changes in the PDGF receptor protein and thus to activation of the kinase domains of the receptor. Activation of src-homology-2 (SH2) or phosphotyrosine-binding (PTB) domains will lead to activation of one of the downstream cytoplasmic pathways. Although more pathways are described, activation of the MAPK/ERK pathway, the AKT/PKB pathway, and the PKC pathway are the major routes (19, 35). The MAPK/ERK pathway is also known as the Ras–Raf–MEK–ERK pathway because the GTP-binding protein Ras can activate mitogen-activated protein kinases (MAPK) such as extracellular signal-regulated kinase (ERK). Phosphorylated ERK-1 and -2 form activated ERK dimers and they translocate to the nucleus, where they can activate cellular processes linked to stimulation of cell proliferation and chemotaxis. Phosphoinositol 3-kinase can also be recruited to the activated PDGF receptor leading to stimulation of cell proliferation and migration via the AKT/PKB pathway. AKT, however, regulates multiple other biological processes including glycogen metabolism and cell survival, and it is linked to promotion of tumor growth. Activation of the third pathway involves phospholipase C-γ (PLC) recruitment after stimulation of the PDGFR. This causes mobilization of cellular Ca2+ storage pools and leads to activation of the intracellular signaling molecule protein kinase C (PKC). This route is also associated with increased cell growth and motility.

Intracellular Routing of PDGF and Its Receptor

Much is known about the intracellular signaling and biological effects of PDGFR. However, the cellular kinetics of the PDGFR itself is less well studied and also the fate of the growth factor and its receptor after binding of the ligand at the cell membrane leading to induction of intracellular signaling is poorly described. It is described that the PDGFβR is localized in specialized structures of the cell membrane referred to as caveolae (3638). The morphology of these structures is generated by the protein caveolin and they are enriched in glycosphingolipids and cholesterol (39). Sorkin et al. showed that induction of the tyrosine kinase activity of the PDGFβR promotes ligand-induced internalization and subsequently degradation of the ligand–receptor complex in endosomal and lysosomal compartments of the cell. They showed that the receptors began to migrate out of caveolae after 1 h of exposure to PDGF and that the internalized PDGFβR retained kinase activity and thus could participate in signaling considerable time after internalization (36). The receptor can recycle back to the cell membrane waiting for its next signal. The fate of the growth factor after dissociation from the receptor in the endosomes is degradation in the lysosomal compartment, but other intracellular routes are possible (40).

Interferon Gamma

The involvement of interferons (IFNs) in the defense response against viral infections was first described in 1957. Isaacs and Lindenmann reported that influenza virus-infected cells produced a secreted factor, i.e., interferon, that caused a virus-resistant state in previously uninfected cells (41). The biochemical characterization of this activity was pursued for a long time without success, and it was only in the late 1970s that the interferon protein sequence was ascertained (42, 43). Later, it appeared that the biological activities of IFNs were much broader, and antifibrotic, antitumor, immunoregulatory, and proinflammatory activities were also assigned to the IFN family (44).

Interferons, belonging to the interferon/interleukin-10 cytokine family, can be divided in two groups (type I and II). They are classified according to their receptor specificity and sequence homology (14). The type I IFNs, also described as viral IFNs, include IFNα (of which currently 17 subtypes are described), IFNβ, IFNω, and IFNτ. These type I factors are produced by almost all cell types. The type II IFNs consist only of IFNγ, which shows structurally little to no homology with type I IFN, and this protein is also described as immune IFN (45). In contrast to these type I IFNs, expression and secretion of IFNγ is not the result of the infection itself, but occurs by activated immune cells after recognition of infected cells (46). Therefore, although IFNγ possesses antiviral activity, it functions primarily as an immune modulator responsible for pathogen clearance rather than an antiviral compound (47, 48).

Interferon-γ is a non-covalently bound, homodimeric glycoprotein with subunits consisting of 146 amino acids (17.1 kDa) (49, 50), which are connected in an antiparallel fashion (45). Intracellular processing of this cytokine includes intensive glycosylation, giving rise to a mature form of the molecule that exhibits a predominant molecular mass of 50 kDa (51). Murine and human IFNγ show approximately only 40% sequence homology at the protein level (http://www.uniprot.org). The main cell types that produce IFNγ upon endogenous or exogenous stimulation in the fibrotic liver include T lymphocytes and NK(T) cells, B cells, and other antigen-presenting cells (5258). NK cells were shown to be involved in the resolution of fibrosis in both mice and humans by stimulating the production of IFNγ, but also via killing of activated HSCs (5961). The production of IFNγ in turn increased the number of NK cells (60). Emerging evidence suggests that NK cells and IFNγ are very important in the negative regulation of liver fibrosis (6). This was demonstrated by the amelioration of liver fibrosis in vivo, after activation of NK cells by poly I:C or treatment of rodents with IFNγ (60, 6264).

Biological Effects

Within the liver, IFNγ exerts a variety of sometimes even opposing effects. For example, IFNγ is known to be a potent activator of macrophages, by stimulating them to kill phagocytosed microbes and cancer cells and inducing the production of TNFα and interleukin-12 (65). IFNγ induces an inflammatory (“M1”) activation state of the hepatic macrophages (comprising resident Kupffer cells and infiltrating monocytes and macrophages) (66). The proinflammatory factors produced via this route can activate the HSC to become a profibrotic myofibroblast. In addition, IFNγ induces the expression of both MHC-I and MHC-II in several cells (44), promotes the differentiation of T- and B-lymphocytes, and activates neutrophils, all contributing to promotion of fibrosis. It was also reported that IFNγ inhibits the proliferation of vascular endothelial cells (65). On the other hand, IFNγ has direct antifibrotic effects when it interacts with HSCs. Increased production of IFNγ, as secreted by NK cells in fibrotic livers, is accompanied by an increase in TNF-related apoptosis-inducing ligand (TRAIL) expression in NK cells, which causes both induction of apoptosis and cell cycle arrest in HSCs (60, 64, 67). The interaction between the stimulatory receptor NKG2D on NK cells and its ligand retinoic acid early inducible 1 (RAE1), expressed on activated HSCs, leads to killing of activated HSCs by NK cells and thus to resolution of fibrosis (60). A summary of cells that produce and respond to IFNγ and its main functions is presented in Figure 1.

In addition, IFNγ leads to reduced α-SMA, HSC proliferation, and collagen levels in fibroblasts (24, 62). IFNγ antagonizes the profibrotic activities of TGFβ, most probably by intracellular induction of the expression of the antagonistic Smad7. Smad7 causes impairment of the TGFβ response, via blocking of the interaction of the transcription factor Smad3 with the TGFβ receptor (68).

IFNγ-Receptor Expression

Like many other cytokines, the receptor for IFNγ consists of multiple distinct subunits: the α chain (IFNγR1; 54.4 kDa), necessary for ligand binding and processing, and the β subunit (IFNγR2; 37.8 kDa), needed to induce a biological response by triggering the intracellular signal transduction cascade (14, 69). The complete receptor complex contains two IFNγR1 and two IFNγR2 subunits and these four parts associate after ligand binding. In more detail, IFNγ binds to one IFNγR1 and induces rapid dimerization of two IFNγR1 chains, thereby forming a site that is recognized by the extracellular domain of IFNγR2 (45). The IFNγR2 chain is generally the limiting factor in IFNγ responsiveness, as the IFNγR1 chain is usually in excess (45, 70). The IFNγR2 chain is constitutively expressed, but its expression level is tightly regulated according to the state of cellular differentiation or activation. Of note, the binding of human and murine IFN to their receptors is strictly species specific. They only induce an effect in species-matched cells. Moreover, the interaction of IFNγ with its receptor is not inhibited by type I IFNs (45).

Interferon-γ R1 and R2 are ubiquitously expressed on the cell membrane of virtual all types of cells, but most predominantly on cells of the immune system. In particular, macrophages are strongly positive for the IFNγR. Within the fibrotic liver, expression on infiltrated macrophages and Kupffer cells is high, while other cells like HSCs and hepatocytes weakly express the receptor (71). Bansal et al. previously showed the ubiquitous and increased expression of the IFNγR1 in human cirrhotic livers as compared to healthy livers (72). Although expression of the IFNγR at the plasma membrane varies widely between tissues (200–25,000 sites/cell), no direct correlation between the expression level and the magnitude of IFN-induced responses in cells was found (45).

IFNγ-Receptor Signaling: The Classical Model

In the case of IFNγ, a conformational change in the receptor cytoplasmic domain is induced upon binding of IFNγ to the extracellular domains of two IFNγR1 chains. This causes movement of the inactive Janus activated kinase 2 (Jak2) from IFNγR2 to IFNγR1 and subsequent autophosphorylation and activation (73). After transphosphorylation and activation of Jak1 by Jak2, the recruitment of signal transducer and activator of transcription 1 alpha (Stat1α) is induced by phosphorylation of tyrosine440 residues of both IFNγR1 subunits, forming a homodimeric complex of pStat1α (69, 7476). This Stat1 pair dissociates from the receptor upon phosphorylation at the tyrosine 701 residue, followed by active nuclear translocation via its nuclear localization sequence (NLS) (73, 74). In the nucleus, dimeric Stat1α binds to the IFNγ-activated sequence (GAS element) of the IFNγ-promoter, thereby determining specific gene activation of fibrosis-related genes like procollagen I and III and TGFβ, but also transcription factors like interferon regulatory factor-1 (IRF-1) (7779). The cellular response of IFNγ occurs within 15–30 min after treatment (80). Binding of dimeric Stat1α to the GAS element is regarded as validation of the classical model of cytokine signaling (81).

To a lesser extent, IFNγ-signaling also produces heterodimeric complexes like Stat1:Stat2 and heterotrimeric complexes such as Stat1:Stat1:IRF-9 and Stat1:Stat2:IRF-9 (77, 8284). These complexes are able to bind to IFN-stimulated response element (ISRE) promoter regions in target genes to regulate transcription of, for example, inducible nitric oxide synthase (iNOS), IRF-2, and IFNβ (14).

IFNγ-Receptor Signaling: The Non-Canonical Model

Several reasons led to the recognition that the classical model of cytokine signaling is an oversimplified model. It was shown that upon IFNγ stimulation besides the Jak/Stat pathway, also other pathways were activated or interfered with the Jak/Stat signaling. Examples include the activation of MAPK, phosphoinositide 3-kinase, Ca2+/calmodulin kinase II, and nuclear factor-κB (85). Moreover, it was shown in multiple cell lines that besides activated Stat, also Jak kinases were involved in the epigenetics of gene activation, indicating that Stat1 is not the only key player in the IFNγ signaling (86). In addition to this, many ligands activate the same Stat transcription factors, but have completely different effects in cells, tissues, or organs (87). In the classical model of signaling of IFNγ, it is suggested that the Stat proteins possess intrinsic NLSs, which are responsible for the nuclear translocation of Stat1 and subsequent IFNγ-related gene activation (88). Of note, in this classical model, the internalization of the ligand–receptor complex does not play an active role in the signaling process (87). Different to what was assumed in the classical model, it appeared that ligand, receptor, and activated Jaks and Stats were directly involved in nuclear events leading to specific gene activation. Therefore, the non-canonical model of IFNγ-signaling was developed, which shows large resemblance to that of steroid hormone/steroid receptor signaling (81).

In this model, IFNγ binds to the extracellular domain of the receptor and moves to the cytoplasmic side of the IFNγR1 domain, after which endocytosis of this complex is induced. The movement of Jak2 to the R1 domain results in autoactivation of the Jaks. This causes phosphorylation of R1 and eventually the recruitment and activation of Stat1α. The complex of IFNγ, the IFNγR1, Stat1α, and Jak1 and 2 is actively transported to the nucleus, a process which is under the influence of the NLS of IFNγ (14, 81, 87). The non-canonical model of IFNγ signaling formed the conceptual framework for the development of IFNγ mimetics (81).

Intracellular Routing of IFNγ and Its Receptor

In contrast to the numerous publications on the signal transduction and biological effects of IFNγ similar to the PDGFβ receptor, there are relatively little data available on the trafficking of the IFNγ receptor. The interferon receptor has been localized in clathrin-coated pits (CCPs), lipid rafts, and caveolae (39).

Clathrin-coated pits are specialized regions in the plasma membrane that are engaged in the efficient internalization of receptors from the cell surface. It is known that most transmembrane receptors are endocytosed via clathrin-dependent endocytosis which involves CCPs (89). Both IFNγ and its receptor were shown to be associated with CCPs (90, 91). In general, receptors are recruited into CCPs through direct binding of their cytoplasmic binding motifs to the adaptor-related protein complex 2 (AP-2).

It is suggested that other endocytotic pathways also may participate in the endocytosis of the IFNγ receptor (39). Lipid rafts are microdomains in the plasma membrane, which are enriched in certain lipids, cholesterol, and proteins, caused by redistribution within the lateral plane of cellular membranes. It is hypothesized that rafts exist in a separate phase that diffuses dynamically in a sea of poorly ordered lipids in the plasma membrane. By dynamic diffusion, proteins can be included or excluded in these rafts (91). The IFNγ receptor was shown to be present in lipid rafts. Moreover, the downstream signaling molecules Jak1 and 2 and Stat1 were also associated with lipid rafts in the plasma membrane (9294). Caveolae are considered a subtype of lipid rafts (39). It was shown that IFNγ and its receptor also localize in caveolae, albeit to a lesser extent than in CCPs (95).

When the ligand–receptor complex is internalized via endocytosis, it enters a cellular compartment with acidic pH. Within this compartment, the ligand dissociates from the receptor. Free IFNγ can then be trafficked to the lysosome, where it is degraded. In many cells, such as fibroblasts and macrophages, the receptor eventually recycles back to the cell surface. In most cells, the intracellular pool is approximately two to four times larger when compared with the receptors expressed at the cell surface (96100).

Mimetic IFNγ

It was previously shown that the N-terminal side of IFNγ plays an important role in the recognition of the extracellular domain of its receptor, whereas the C-terminus is involved in endocytosis by binding to the membrane proximal region of the cytoplasmic part of the IFNγR1 subunit (101). Therefore, it is suggested that the N-terminal region of IFNγ is involved in species-specific recognition and binding, whereas the C-terminal region may be involved in binding of internalized IFNγ to the cytoplasmic receptor domain (101). This led to the development of a truncated peptide, which contains the C-terminal region of IFNγ, namely mimetic IFNγ (IFNγ95–132 AA) (101). It appeared that this mimetic peptide possesses similar biological activities as its full length equivalent, despite the fact that it is not able to bind to the extracellular domain of its receptor (102). It was for instance shown that mimetic IFNγ possesses IFNγ-agonistic activity without toxicity, such as the induction of MHCII expression on macrophages (101). Furthermore, mimetic IFNγ displayed antiviral activity in tissue cultures and in mice (103). In order to be able to deliver mimetic IFNγ into the cytoplasm, the peptide was modified with a single N-terminal ε-palmitoyl-lysine residue. This was shown to be suitable to allow penetration across the cell plasma membrane and subsequent cytoplasmic delivery (87, 104), but the use of this palmitoyl residue is unfavorable for in vivo use because it induces non-specific uptake in any cell it encounters. Similar to IFNγ, mimetic IFNγ forms a complex with IFNγR1, Stat1α, and Jak1 and 2 and translocates to the nucleus as directed by its NLS (14, 81, 87). Advantages of mimetic proteins in general are their versatility, the high biological activity and specificity, and their low toxicity (103). There are already several peptide mimetics on the market to treat a broad variety of diseases. One example is the tripeptide mimetic boceprevir for the treatment of hepatitis C viral infections (105). The therapeutic applications of mimetic IFN are nowadays explored (106).

Antifibrotic Compounds

Although in the past decade some progress was made, the development of antifibrotic drugs for the treatment of liver cirrhosis is still in its infancy. Major steps forward in the treatment of liver diseases were made at the level of eradication of the inciting stimulus. The high successes obtained with anti-HCV drugs that effectively clear the virus and thus reverse the chronic pathology also resulted in cases in which regression of the fibrosis in the livers of these patients was ascertained (107). This contributed to the belief that liver fibrosis even in advanced state is reversible and treatable. Parallel to this, experimental therapeutic interventions directly aiming at fibrogenic key cells and processes emerged (4, 108). In 2005, Pinzani et al. showed an impressive list with all possible compounds under (pre)clinical investigation for the treatment of liver fibrosis (109). Later on, more reviews with promising antifibrotic drugs appeared (110112). Currently 370 clinical trials are listed at http://www.clinicaltrials.gov, illustrating the upcoming incidence of clinical trials ongoing in liver cirrhosis, and showing that the pharmaceutical industry is interested to invest in drugs for the treatment of liver cirrhosis, despite the expected long-term trials to assess efficacy in this chronic, slow progressing disease. Of note, about 90% of the clinical trials are still dealing with diagnostics improvements, treatment of cirrhosis-associated complications, or with the application and evaluation of direct antiviral agents. This means that only a minority of the clinical studies really tests novel antifibrotic compounds (108). Remarkably, there are hardly any completed or ongoing clinical trials in patients with liver cirrhosis testing compounds that target PDGF and IFNγ pathways. Only in 2002, a study was started in which IFNγ1b was tested for safety and efficacy in HCV patients with liver fibrosis or cirrhosis. However, although IFNγ was well tolerated, the drug was abandoned and not approved because of lack of antifibrotic activity (113, 114). With regard to intervention at the PDGF level, several experimental approaches aiming at PDGF were described in literature. Examples include inhibition of the PDGF-B chain (115, 116), inhibition of the PDGFβ receptor expressions (117, 118), and inhibition of the PDGFβ receptor signaling (119, 120). However, none of these compounds were found in the clinical trial database on liver fibrosis or cirrhosis.

New targets for direct-acting antifibrotic compounds were identified in the past decades due to a better understanding of the pathological process at molecular level. This includes the specific roles of the different (hepatic) cell types, the mediators involved in the communication between these cells and their intracellular signaling pathways, and the unravelment of the complex process of fibrotic extracellular matrix deposition, during which matrix degrading matrix metalloproteinases (MMPs) and inhibitors of these metalloproteinases (TIMPs) parallel the enhanced production of matrix components. This basic knowledge contributed to the generation of various antifibrotic compounds that can be tested in patients with liver cirrhosis. One of the current promising ongoing clinical trials deals with the application of mesenchymal stem cells to treat liver cirrhosis (121). Mesenchymal stem cells have shown to have the ability to effectively reduce liver fibrosis and improve liver function. The effects are based on various activities of these mesenchymal stem cells. First, these stem cells can differentiate into hepatocytes, allowing the replacement of damaged hepatocytes, and they can promote regeneration of residual hepatocytes. In addition, mesenchymal stem cells also affect HSCs and possess immunomodulatory properties, all adding to their antifibrotic potential. However, some limitations need to be considered. Over the past few years, concerns have been raised about its long-term effectiveness, the potential tumorigenic risk, and the lack of standardized protocols for mesenchymal stem cell transplantation.

The development of cell-specific antifibrotic drugs is another approach that seems promising (122124). Fibroferon, based on the structure of both PDGF-BB and IFNγ, is one of the latest compounds in this context. Fibroferon is an IFNγ-derived mimetic peptide that is specifically delivered to activated HSCs and myofibroblasts. We used the highly upregulated PDGFβR as a target receptor for the HSC/myofibroblast-directed drug targeting approach in fibrosis. We were the first to be able to deliver drugs to HSCs using a series of modified proteins, and one of these drug carriers was directed at the PDGFβR (9, 125, 126). Nowadays, several groups have developed or applied cell-specific proteins to HSCs (124, 127129), recently reviewed by Poelstra et al. (122). In the following paragraphs, we will delineate our approach and show, as depicted in Figure 2, how this has led to the development of the promising antifibrotic PDGFβR-targeted interferon, referred to as Fibroferon.

FIGURE 2
www.frontiersin.org

Figure 2. Design of constructs directed at the PDGFβ receptor in time, finally leading to the development of Fibroferon. The first PDGFβR-targeted product was the carrier pPB-HSA in which several (on average n = 10) groups of pPB were randomly attached to human serum albumin (HSA), in order to allow multivalent interaction with the PDGFR. Several other constructs, comprising a therapeutic entity (drug or protein) coupled to this carrier, were subsequently developed and tested in vivo. In the final step, the structure was simplified by omitting the albumin protein core and applying low-molecular-weight PEG to couple truncated IFNγ to BipPB, to create divalent interaction of truncated IFNγ with the PDGFβR.

PDGFβ Receptor Recognizing Peptide

We designed a cyclic peptide (pPB) that mimics the binding site of PDGF-BB to its endogenous receptor. Similar to PDGF-BB itself, the arginine and isoleucine residues and their adjacent amino acids represent the PDGFβ receptor binding moieties in the pPB peptide (C*SRNLIDC*, where the C* denotes the cyclizing cysteine residues) (31). The peptide was designed in a cyclic conformation because earlier observations revealed that cyclic peptides have stronger interactions with their receptors and have increased stability in plasma as compared to their linear analogs (130, 131).

At first, a number of pPB peptides were coupled to human serum albumin (HSA) yielding the HSC-specific carrier pPB-HSA (see Figure 2) (9). The PDGFβ receptor requires a dimeric interaction for ligand binding and receptor activation (132) and therefore multiple pPB moieties (on average 10 molecules of pPB per 1 molecule HSA at different distances from each other) were necessary to obtain optimal receptor binding.

It was confirmed in vitro that pPB-HSA bound to the PDGFβ receptor on fibroblast cell lines and on primary rat HSCs. This binding was inhibited by PDGF-BB but not by PDGF-AA. Coupling of pPB-peptides to a macromolecule like HSA was necessary for a competitive effect with PDGF-BB, since free pPB did not displace PDGF-BB from its receptor. PDGFβR-induced effects were not found after incubation of fibroblasts with pPB-HSA. Based on these observations, it was hypothesized that coupling of pPB to a macromolecule such as HSA enabled a multivalent interaction with the PDGFβR, but no receptor activation was induced by pPB-based molecules. In vivo, pPB-HSA was shown to accumulate in HSCs in fibrotic human, mouse, and rat livers (9); cells expressing the highest PDGFβR. These results formed the basis for the development of HSC-selective targeting of drugs applying pPB-HSA as drug carrier and for example the antifibrotic agents IFNγ(133) and 15d-prostaglandin-J2 (134) were evaluated. This is particularly relevant for IFNγ, which has a therapeutic application in a wide variety of immunological, viral, and neoplastic diseases, and where high percentages, up to 100%, of patients suffer from general side effects such as headache, malaise, and fever (44). A fundamental question is however whether a cytokine can be delivered to another target receptor, without losing the biological effects elicited by its endogenous receptor.

pPB-Based Delivery of IFNγ to HSCs for the Treatment of Liver Fibrosis

We further developed HSC-selective compounds containing the cyclic peptide pPB and covalently conjugated this peptide to the antifibrotic compound IFNγ (see Figure 2). This led to various compounds either with (pPB-PEG-IFNγ and pPB-HSA-PEG-IFNγ) or without (pPB-IFNγ) polyethylene glycol (PEG) linker in between (24, 133). The choice for a distinct PEG-linker was based on previous experiments, showing that PEGylation of IFNγ with different sizes of PEG-molecules (5, 10, and 20 kDa) improved the pharmacokinetic profile, liver uptake, and antifibrotic effects of IFNγ, with the smallest PEG molecule as most favorable (72). Additionally, improved conformational flexibility of the pPB peptide using a PEG spacer allowed improved presentation of pPB to the PDGFβ receptor (72). Our conjugates were shown to bind to the PDGFβ receptor on fibroblasts and HSCs and inhibited their activation in vitro, as shown by a reduction in collagen type I and α-SMA expression. In addition, the conjugates were shown to inhibit PDGF-induced cell proliferation. A remarkable finding in these studies was that the specific binding to the PDGFβ receptor and subsequent biological effects of the pPB-modified IFNγ-constructs was not species-specific anymore in contrast to IFNγ. This was demonstrated by the absence of both binding and an antifibrotic effect of mouse IFNγ in human cell cultures, whereas mouse IFNγ conjugated to pPB did bind to human cells and showed biological activity (24, 133). It was therefore proposed that following binding to the PDGFβ receptor, the complex was internalized, degraded, and IFNγ (or its signaling moiety) was released into the cytoplasm leading to IFNγ-mediated effects (135).

The targeted constructs (pPB-PEG-IFNγ, pPB-HSA-PEG-IFNγ, and pPB-IFNγ) colocalized with desmin-positive PDGFβR-expressing HSCs in the liver. In addition, they strongly inhibited fibrogenesis and induced fibrolysis in a 2-week treatment study in 8-week-old CCl4 mice, as demonstrated by attenuated collagen type I and α-SMA deposition. Additionally, the balance between collagen-degrading MMP-13 and its endogenous inhibitor TIMP-1 was shown to be shifted toward a fibrolytic state, characterized by a reduced TIMP-1 expression. This effect was not seen in the animals treated with untargeted IFNγ. Furthermore, in contrast to untargeted IFNγ, the targeted IFNγ conjugates did not induce unwanted IFNγ-related side effects. In these mice treated with targeted IFNγ, systemic inflammation, hyperthermia, elevated plasma triglyceride levels, and neurotropic effects, as demonstrated by among others a reduced release of proinflammatory cytokines (IL-1β), were absent. Moreover, targeted IFNγ lowered MHC-II expression in the brain as compared to native IFNγ (24, 133), which makes this compound attractive for long-term treatment.

Redirection of IFNγ to myofibroblasts in the fibrotic liver proves to be a promising approach. We pursued this by creating a smaller, more simplified construct to facilitate clinical application (see Figure 2) (24, 133). This minimized structure combines three basic elements, i.e., IFNγ-activity, PEG, and a PDGFβR-binding peptide (106). In this truncated IFNγ structure (i.e., mimetic IFNγ), the extracellular IFNγ receptor binding sequence was removed, yet the NLS accommodating the biological effects of IFNγ was retained. In this way, interaction with the ubiquitously expressed IFNγ receptor at the membrane of the cells was prevented, leaving only an interaction with the intracellular IFNγR (136, 137). This mimetic IFNγ-structure was redirected to the PDGFβ receptor by coupling it to PDGFβR-recognizing peptides. We also minimized this entity by incorporating only two cyclic PDGFβ receptor binding peptides (BipPB) at a specific distinct distance from each other to allow dimeric ligand–receptor interaction. The PEG linker was minimized to only 2 kDa and BipPB, PEG, and mimetic IFNγ were subsequently coupled in a 1:1:1 ratio, yielding an 8.8 kDa construct (referred to as mimγ-BipPB, BOT191, or Fibroferon). Similar to pPB-PEG-IFNγ, Fibroferon markedly inhibited both early and advanced CCl4-induced liver fibrosis in mice and significantly reduced IFNγ-related side effects (106). The advantages of this targeted mimetic IFNγ-conjugate are the in vivo stability, the cell selectivity, dissociation of effects, the loss of species specificity, the minimal size, and less complexity, making it a better-defined product. This all provides Fibroferon with clear advantages above the therapeutic use of native IFNγ as an antifibrotic compound.

pPB-Based HSC-Selective Drug Carriers for the Treatment of Other Diseases

In addition to the demonstrated antifibrotic properties of redirected truncated IFNγ in a mouse liver fibrosis model, antifibrotic effects of pPB-PEG-IFNγ were also found in the unilateral ureteral obstruction (UUO) mouse model for renal fibrosis. In this model, pPB-PEG-IFNγ specifically accumulated in PDGFβR-overexpressing interstitial myofibroblasts in the kidney. Treatment of mice with pPB-PEG-IFNγ significantly attenuated collagen I, fibronectin, and α-SMA mRNA levels and protein expression in fibrotic kidneys similar to the liver fibrosis studies. Compared to vehicle treatment, pPB-PEG-IFNγ protected the tubular morphology, significantly attenuated interstitial T-cell infiltration and reduced the formation of lymphatic vessels, without affecting the peritubular capillary density. In addition, pPB-PEG-IFNγ reduced IFNγ-mediated side effects, as shown by reduced MHC-II mRNA expression in the brain (135), which was applied as a marker for off-target effects on macrophages.

pPB-HSA-IFNγ was also shown to be effective as antitumor agent (138) because the PDGFβ receptor is also upregulated in various types of cancer (139, 140). The PDGFβ receptor was immunohistochemically shown to be abundantly expressed in stromal cells and pericytes of neoplastic human diseases, like colorectal, pancreatic, and breast cancer (19). Stromal cells, including tumor-associated fibroblasts, endothelial cells, vascular pericytes, and infiltrating inflammatory cells, are increasingly recognized as key players in the induction of growth and progression of tumors (141, 142). Activation and migration of fibroblasts and tube formation in endothelial cells induced by fibroblasts was inhibited by pPB-HSA-IFNγ, suggesting an effect on angiogenic processes. In C26 colon carcinoma tumor-bearing mice, pPB-HSA-IFNγ specifically accumulated into PDGFβ receptor expressing tumor stromal fibroblasts and pericytes and inhibited the tumor growth. Treatment of these mice with pPB-HSA-IFNγ was associated with a significant inhibition of angiogenesis, as reflected by reduced α-SMA and CD31 stainings (138).

Other drugs were also delivered to the PDGFβ receptor using the cyclic peptide pPB as a targeting device. For example, the anticancer agent doxorubicin was coupled to pPB-HSA via an acid-sensitive hydrazine linkage for delivery to the PDGFβ receptor on stromal cells. The targeted construct was shown to significantly reduce tumor growth with high response rate as compared to the untargeted doxorubicin. Therefore, the delivery of anticancer drugs to these stromal cells, nurturing the neighboring tumor cells, is a novel and maybe promising approach to treat cancers effectively (143).

These studies suggest that the PDGFβ receptor-targeted constructs not only exert therapeutic effects in liver fibrosis (24, 106, 133) but also in other diseases associated with high PDGFβ receptor expression, such as kidney fibrosis (135) and some types of cancer (138, 143).

Conclusion

The ever increasing knowledge on molecular mechanisms and mediators that orchestrate the progression of liver fibrosis can be used to design new therapeutic compounds against this lethal disease. The fundamental processes that underlie this disease are similar in many other fibrotic and sclerotic diseases, including stromal cancers, which broadens the scope of applications of such novel compounds. In the past decades, the key cells and key mediators that stimulate and inhibit liver fibrogenesis have been identified and we used this knowledge to create a cell-specific chimeric compound which combines the best of two important mediators: the high disease-induced receptor expression of one mediator (PDGFR) is used to deliver another mediator (IFNγ), endowed with antifibrotic activities, to the HSC, yielding Fibroferon. Further studies on the intracellular trafficking of this compound that induces the activation of the IFNγ-signaling cascade via binding to the PDGFR are in progress. This chimeric molecule might be one of the next-generation antifibrotic therapeutics. These studies also illustrate the surprising adaptability of biological compounds that are the natural inhibitors of this chronic disease.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Venook AP, Papandreou C, Furuse J, de Guevara LL. The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective. Oncologist (2010) 15(Suppl 4):5–13. doi:10.1634/theoncologist.2010-S4-05

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Bruix J, Han KH, Gores G, Llovet JM, Mazzaferro V. Liver cancer: approaching a personalized care. J Hepatol (2015) 62(1 Suppl):S144–56. doi:10.1016/j.jhep.2015.02.007

CrossRef Full Text | Google Scholar

3. Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev (2007) 88(1):125–72. doi:10.1152/physrev.00013.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Trautwein C, Friedman SL, Schuppan D, Pinzani M. Hepatic fibrosis: concept to treatment. J Hepatol (2015) 62(1S):S15–24. doi:10.1016/j.jhep.2015.02.039

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis (2010) 30(3):245–57. doi:10.1055/s-0030-1255354

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Jeong WI, Park O, Suh YG, Byun JS, Park SY, Choi E, et al. Suppression of innate immunity (natural killer cell/interferon-gamma) in the advanced stages of liver fibrosis in mice. Hepatology (2011) 53(4):1342–51. doi:10.1002/hep.24190

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Seki E, Schwabe RF. Hepatic inflammation and fibrosis: functional links and key pathways. Hepatology (2015) 61(3):1066–79. doi:10.1002/hep.27332

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol (2014) 14(3):181–94. doi:10.1038/nri3623

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Beljaars L, Weert B, Geerts A, Meijer DK, Poelstra K. The preferential homing of a platelet derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in fibrotic tissue. Biochem Pharmacol (2003) 66(7):1307–17. doi:10.1016/S0006-2952(03)00445-3

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Baer HU, Friess H, Abou-Shady M, Berberat P, Zimmermann A, Gold LI, et al. Transforming growth factor betas and their receptors in human liver cirrhosis. Eur J Gastroenterol Hepatol (1998) 10(12):1031–9. doi:10.1097/00042737-199812000-00009

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Bedossa P, Peltier E, Terris B, Franco D, Poynard T. Transforming growth factor-beta 1 (TGF-beta 1) and TGF-beta 1 receptors in normal, cirrhotic, and neoplastic human livers. Hepatology (1995) 21(3):760–6. doi:10.1002/hep.1840210325

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Gratchev A, Kzhyshkowska J, Kannookadan S, Ochsenreiter M, Popova A, Yu X, et al. Activation of a TGF-beta-specific multistep gene expression program in mature macrophages requires glucocorticoid-mediated surface expression of TGF-beta receptor II. J Immunol (2008) 180(10):6553–65. doi:10.4049/jimmunol.180.10.6553

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Tilg H, Kaser A, Moschen AR. How to modulate inflammatory cytokines in liver diseases. Liver Int (2006) 26(9):1029–39. doi:10.1111/j.1478-3231.2006.01339.x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol (2004) 75(2):163–89. doi:10.1189/jlb.0603252

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Paul D, Lipton A, Klinger I. Serum factor requirements of normal and simian virus 40-transformed 3T3 mouse fibroplasts. Proc Natl Acad Sci U S A (1971) 68(3):645–52. doi:10.1073/pnas.68.3.645

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Pinzani M, Gesualdo L, Sabbah GM, Abboud HE. Effects of platelet-derived growth factor and other polypeptide mitogens on DNA synthesis and growth of cultured rat liver fat-storing cells. J Clin Invest (1989) 84(6):1786–93. doi:10.1172/JCI114363

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Pinzani M, Milani S, Herbst H, DeFranco R, Grappone C, Gentilini A, et al. Expression of platelet-derived growth factor and its receptors in normal human liver and during active hepatic fibrogenesis. Am J Pathol (1996) 148(3):785–800.

PubMed Abstract | Google Scholar

18. Betsholtz C. Biology of platelet-derived growth factors in development. Birth Defects Res C Embryo Today (2003) 69(4):272–85. doi:10.1002/bdrc.10030

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet-derived growth factor. Physiol Rev (1999) 79(4):1283–316.

PubMed Abstract | Google Scholar

20. Reigstad LJ, Varhaug JE, Lillehaug JR. Structural and functional specificities of PDGF-C and PDGF-D, the novel members of the platelet-derived growth factors family. FEBS J (2005) 272(22):5723–41. doi:10.1111/j.1742-4658.2005.04989.x

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Reigstad LJ, Martinez A, Varhaug JE, Lillehaug JR. Nuclear localisation of endogenous SUMO-1-modified PDGF-C in human thyroid tissue and cell lines. Exp Cell Res (2006) 312(6):782–95. doi:10.1016/j.yexcr.2005.11.035

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Tiesman J, Hart CE. Identification of a soluble receptor for platelet-derived growth factor in cell-conditioned medium and human plasma. J Biol Chem (1993) 268(13):9621–8.

PubMed Abstract | Google Scholar

23. Kim TH, Moon JH, Savard CE, Kuver R, Lee SP. Effects of lipopolysaccharide on platelet-derived growth factor isoform and receptor expression in cultured rat common bile duct fibroblasts and cholangiocytes. J Gastroenterol Hepatol (2009) 24(7):1218–25. doi:10.1111/j.1440-1746.2008.05729.x

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Bansal R, Prakash J, Post E, Beljaars L, Schuppan D, Poelstra K. Novel engineered targeted interferon-gamma blocks hepatic fibrogenesis in mice. Hepatology (2011) 54(2):586–96. doi:10.1002/hep.24395

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Gu Z, Noss EH, Hsu VW, Brenner MB. Integrins traffic rapidly via circular dorsal ruffles and macropinocytosis during stimulated cell migration. J Cell Biol (2011) 193(1):61–70. doi:10.1083/jcb.201007003

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Liu Z, Liu H, Jiang J, Tan S, Yang Y, Zhan Y, et al. PDGF-BB and bFGF ameliorate radiation-induced intestinal progenitor/stem cell apoptosis via akt/p53 signaling in mice. Am J Physiol Gastrointest Liver Physiol (2014) 307(11):G1033–43. doi:10.1152/ajpgi.00151.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Bowen-Pope DF, Malpass TW, Foster DM, Ross R. Platelet-derived growth factor in vivo: levels, activity, and rate of clearance. Blood (1984) 64(2):458–69.

PubMed Abstract | Google Scholar

28. Zafiropoulos A, Fthenou E, Chatzinikolaou G, Tzanakakis GN. Glycosaminoglycans and PDGF signaling in mesenchymal cells. Connect Tissue Res (2008) 49(3):153–6. doi:10.1080/03008200802148702

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Gonias SL, Carmichael A, Mettenburg JM, Roadcap DW, Irvin WP, Webb DJ. Identical or overlapping sequences in the primary structure of human alpha(2)-macroglobulin are responsible for the binding of nerve growth factor-beta, platelet-derived growth factor-BB, and transforming growth factor-beta. J Biol Chem (2000) 275(8):5826–31. doi:10.1074/jbc.275.8.5826

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Borkham-Kamphorst E, Kovalenko E, van Roeyen CR, Gassler N, Bomble M, Ostendorf T, et al. Platelet-derived growth factor isoform expression in carbon tetrachloride-induced chronic liver injury. Lab Invest (2008) 88(10):1090–100. doi:10.1038/labinvest.2008.71

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Clements JM, Bawden LJ, Bloxidge RE, Catlin G, Cook AL, Craig S, et al. Two PDGF-B chain residues, arginine 27 and isoleucine 30, mediate receptor binding and activation. EMBO J (1991) 10(13):4113–20.

PubMed Abstract | Google Scholar

32. Fenstermaker RA, Poptic E, Bonfield TL, Knauss TC, Corsillo L, Piskurich JF, et al. A cationic region of the platelet-derived growth factor (PDGF) A-chain (Arg159-Lys160-Lys161) is required for receptor binding and mitogenic activity of the PDGF-AA homodimer. J Biol Chem (1993) 268(14):10482–9.

PubMed Abstract | Google Scholar

33. Chen PH, Chen X, He X. Platelet-derived growth factors and their receptors: structural and functional perspectives. Biochim Biophys Acta (2013) 1834(10):2176–86. doi:10.1016/j.bbapap.2012.10.015

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Yang Y, Yuzawa S, Schlessinger J. Contacts between membrane proximal regions of the PDGF receptor ectodomain are required for receptor activation but not for receptor dimerization. Proc Natl Acad Sci U S A (2008) 105(22):7681–6. doi:10.1073/pnas.0802896105

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Pinzani M. PDGF and signal transduction in hepatic stellate cells. Front Biosci (2002) 7:d1720–6. doi:10.2741/pinzani

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Sorkin A, Eriksson A, Heldin CH, Westermark B, Claesson-Welsh L. Pool of ligand-bound platelet-derived growth factor beta-receptors remain activated and tyrosine phosphorylated after internalization. J Cell Physiol (1993) 156(2):373–82. doi:10.1002/jcp.1041560221

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Liu P, Ying Y, Ko YG, Anderson RG. Localization of platelet-derived growth factor-stimulated phosphorylation cascade to caveolae. J Biol Chem (1996) 271(17):10299–303. doi:10.1074/jbc.271.17.10299

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Liu P, Ying Y, Anderson RG. Platelet-derived growth factor activates mitogen-activated protein kinase in isolated caveolae. Proc Natl Acad Sci U S A (1997) 94(25):13666–70. doi:10.1073/pnas.94.25.13666

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Claudinon J, Monier MN, Lamaze C. Interfering with interferon receptor sorting and trafficking: impact on signaling. Biochimie (2007) 89(6–7):735–43. doi:10.1016/j.biochi.2007.03.014

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Gill GN. A pit stop at the ER. Science (2002) 295(5560):1654–5. doi:10.1126/science.1070127

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Isaacs A, Lindenmann J. Virus interference. I. The interferon. By A. Isaacs and J. Lindenmann, 1957. J Interferon Res (1987) 7(5):429–38. doi:10.1089/jir.1987.7.429

CrossRef Full Text | Google Scholar

42. Rubinstein M, Rubinstein S, Familletti PC, Miller RS, Waldman AA, Pestka S. Human leukocyte interferon: production, purification to homogeneity, and initial characterization. Proc Natl Acad Sci U S A (1979) 76(2):640–4. doi:10.1073/pnas.76.2.640

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Zoon KC, Smith ME, Bridgen PJ, zur Nedden D, Anfinsen CB. Purification and partial characterization of human lymphoblast interferon. Proc Natl Acad Sci U S A (1979) 76(11):5601–5. doi:10.1073/pnas.76.11.5601

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Younes HM, Amsden BG. Interferon-gamma therapy: evaluation of routes of administration and delivery systems. J Pharm Sci (2002) 91(1):2–17. doi:10.1002/jps.10007

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol (1997) 15:563–91. doi:10.1146/annurev.immunol.15.1.563

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Young HA, Hardy KJ. Interferon-gamma: producer cells, activation stimuli, and molecular genetic regulation. Pharmacol Ther (1990) 45(1):137–51. doi:10.1016/0163-7258(90)90012-Q

CrossRef Full Text | Google Scholar

47. Farrar MA, Schreiber RD. The molecular cell biology of interferon-gamma and its receptor. Annu Rev Immunol (1993) 11:571–611. doi:10.1146/annurev.iy.11.040193.003035

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Billiau A. Interferon-gamma: biology and role in pathogenesis. Adv Immunol (1996) 62:61–130. doi:10.1016/S0065-2776(08)60428-9

CrossRef Full Text | Google Scholar

49. Gray PW, Leung DW, Pennica D, et al. Expression of human immune interferon cDNA in E. coli and monkey cells. Nature (1982) 295(5849):503–8. doi:10.1038/295503a0

CrossRef Full Text | Google Scholar

50. Gray PW, Goeddel DV. Cloning and expression of murine immune interferon cDNA. Proc Natl Acad Sci U S A (1983) 80(19):5842–6. doi:10.1073/pnas.80.19.5842

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Kelker HC, Le J, Rubin BY, Yip YK, Nagler C, Vilcek J. Three molecular weight forms of natural human interferon-gamma revealed by immunoprecipitation with monoclonal antibody. J Biol Chem (1984) 259(7):4301–4.

PubMed Abstract | Google Scholar

52. Carnaud C, Lee D, Donnars O, Park SH, Beavis A, Koezuka Y, et al. Cutting edge: cross-talk between cells of the innate immune system: NKT cells rapidly activate NK cells. J Immunol (1999) 163(9):4647–50.

PubMed Abstract | Google Scholar

53. Frucht DM, Fukao T, Bogdan C, Schindler H, O’Shea JJ, Koyasu S. IFN-gamma production by antigen-presenting cells: mechanisms emerge. Trends Immunol (2001) 22(10):556–60. doi:10.1016/S1471-4906(01)02005-1

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Gessani S, Belardelli F. IFN-gamma expression in macrophages and its possible biological significance. Cytokine Growth Factor Rev (1998) 9(2):117–23. doi:10.1016/S1359-6101(98)00007-0

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Yoshimoto T, Takeda K, Tanaka T, Ohkusu K, Kashiwamura S, Okamura H, et al. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: synergism with IL-18 for IFN-gamma production. J Immunol (1998) 161(7):3400–7.

PubMed Abstract | Google Scholar

56. Flaishon L, Hershkoviz R, Lantner F, Lider O, Alon R, Levo Y, et al. Autocrine secretion of interferon gamma negatively regulates homing of immature B cells. J Exp Med (2000) 192(9):1381–8. doi:10.1084/jem.192.9.1381

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Harris DP, Haynes L, Sayles PC, Duso DK, Eaton SM, Lepak NM, et al. Reciprocal regulation of polarized cytokine production by effector B and T cells. Nat Immunol (2000) 1(6):475–82. doi:10.1038/82717

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Baron S, Tyring SK, Fleischmann WR Jr, Coppenhaver DH, Niesel DW, Klimpel GR, et al. The interferons. Mechanisms of action and clinical applications. JAMA (1991) 266(10):1375–83. doi:10.1001/jama.1991.03470100067035

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Muhanna N, Abu Tair L, Doron S, Amer J, Azzeh M, Mahamid M, et al. Amelioration of hepatic fibrosis by NK cell activation. Gut (2011) 60(1):90–8. doi:10.1136/gut.2010.211136

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Radaeva S, Sun R, Jaruga B, Nguyen VT, Tian Z, Gao B. Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosis-inducing ligand-dependent manners. Gastroenterology (2006) 130(2):435–52. doi:10.1053/j.gastro.2005.10.055

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Melhem A, Muhanna N, Bishara A, Alvarez CE, Ilan Y, Bishara T, et al. Anti-fibrotic activity of NK cells in experimental liver injury through killing of activated HSC. J Hepatol (2006) 45(1):60–71. doi:10.1016/j.jhep.2005.12.025

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Baroni GS, D’Ambrosio L, Curto P, Casini A, Mancini R, Jezequel AM, et al. Interferon gamma decreases hepatic stellate cell activation and extracellular matrix deposition in rat liver fibrosis. Hepatology (1996) 23(5):1189–99. doi:10.1002/hep.510230538

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Rockey DC, Chung JJ. Interferon gamma inhibits lipocyte activation and extracellular matrix mRNA expression during experimental liver injury: implications for treatment of hepatic fibrosis. J Investig Med (1994) 42(4):660–70.

PubMed Abstract | Google Scholar

64. Jeong WI, Park O, Radaeva S, Gao B. STAT1 inhibits liver fibrosis in mice by inhibiting stellate cell proliferation and stimulating NK cell cytotoxicity. Hepatology (2006) 44(6):1441–51. doi:10.1002/hep.21419

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Arai KI, Lee F, Miyajima A, Miyatake S, Arai N, Yokota T. Cytokines: coordinators of immune and inflammatory responses. Annu Rev Biochem (1990) 59:783–836. doi:10.1146/annurev.bi.59.070190.004031

CrossRef Full Text | Google Scholar

66. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci (2008) 13:453–61. doi:10.2741/2692

CrossRef Full Text | Google Scholar

67. Jeong WI, Park O, Gao B. Abrogation of the antifibrotic effects of natural killer cells/interferon-gamma contributes to alcohol acceleration of liver fibrosis. Gastroenterology (2008) 134(1):248–58. doi:10.1053/j.gastro.2007.09.034

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Ulloa L, Doody J, Massague J. Inhibition of transforming growth factor-beta/SMAD signalling by the interferon-gamma/STAT pathway. Nature (1999) 397(6721):710–3. doi:10.1038/17826

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Greenlund AC, Farrar MA, Viviano BL, Schreiber RD. Ligand-induced IFN gamma receptor tyrosine phosphorylation couples the receptor to its signal transduction system (p91). EMBO J (1994) 13(7):1591–600.

PubMed Abstract | Google Scholar

70. Bernabei P, Coccia EM, Rigamonti L, Bosticardo M, Forni G, Pestka S, et al. Interferon-gamma receptor 2 expression as the deciding factor in human T, B, and myeloid cell proliferation or death. J Leukoc Biol (2001) 70(6):950–60.

PubMed Abstract | Google Scholar

71. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Proteomics. Tissue-based map of the human proteome. Science (2015) 347(6220):1260419. doi:10.1126/science.1260419

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Bansal R, Post E, Proost JH, de Jager-Krikken A, Poelstra K, Prakash J. PEGylation improves pharmacokinetic profile, liver uptake and efficacy of interferon gamma in liver fibrosis. J Control Release (2011) 154(3):233–40. doi:10.1016/j.jconrel.2011.05.027

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Briscoe J, Rogers NC, Witthuhn BA, Watling D, Harpur AG, Wilks AF, et al. Kinase-negative mutants of JAK1 can sustain interferon-gamma-inducible gene expression but not an antiviral state. EMBO J (1996) 15(4):799–809.

PubMed Abstract | Google Scholar

74. Greenlund AC, Morales MO, Viviano BL, Yan H, Krolewski J, Schreiber RD. Stat recruitment by tyrosine-phosphorylated cytokine receptors: an ordered reversible affinity-driven process. Immunity (1995) 2(6):677–87. doi:10.1016/1074-7613(95)90012-8

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Heim MH, Kerr IM, Stark GR, Darnell JE Jr. Contribution of STAT SH2 groups to specific interferon signaling by the Jak-STAT pathway. Science (1995) 267(5202):1347–9. doi:10.1126/science.7871432

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Igarashi K, Garotta G, Ozmen L, Ziemiecki A, Wilks AF, Harpur AG, et al. Interferon-gamma induces tyrosine phosphorylation of interferon-gamma receptor and regulated association of protein tyrosine kinases, Jak1 and Jak2, with its receptor. J Biol Chem (1994) 269(20):14333–6.

PubMed Abstract | Google Scholar

77. Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science (1994) 264(5164):1415–21. doi:10.1126/science.8197455

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Schindler C, Darnell JE Jr. Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Annu Rev Biochem (1995) 64:621–51. doi:10.1146/annurev.bi.64.070195.003201

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Ramana CV, Chatterjee-Kishore M, Nguyen H, Stark GR. Complex roles of Stat1 in regulating gene expression. Oncogene (2000) 19(21):2619–27. doi:10.1038/sj.onc.1203525

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Kerr IM, Stark GR. The control of interferon-inducible gene expression. FEBS Lett (1991) 285(2):194–8. doi:10.1016/0014-5793(91)80802-A

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Johnson HM, Noon-Song EN, Dabelic R, Ahmed CM. IFN signaling: how a non-canonical model led to the development of IFN mimetics. Front Immunol (2013) 4:202. doi:10.3389/fimmu.2013.00202

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Matsumoto M, Tanaka N, Harada H, Kimura T, Yokochi T, Kitagawa M, et al. Activation of the transcription factor ISGF3 by interferon-gamma. Biol Chem (1999) 380(6):699–703. doi:10.1515/BC.1999.087

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons. Annu Rev Biochem (1998) 67:227–64. doi:10.1146/annurev.biochem.67.1.227

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Bluyssen AR, Durbin JE, Levy DE. ISGF3 gamma p48, a specificity switch for interferon activated transcription factors. Cytokine Growth Factor Rev (1996) 7(1):11–7. doi:10.1016/1359-6101(96)00005-6

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Gough DJ, Levy DE, Johnstone RW, Clarke CJ. IFNgamma signaling-does it mean JAK-STAT? Cytokine Growth Factor Rev (2008) 19(5–6):383–94. doi:10.1016/j.cytogfr.2008.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Dawson MA, Bannister AJ, Göttgens B, Foster SD, Bartke T, Green AR, et al. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature (2009) 461(7265):819–22. doi:10.1038/nature08448

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Ahmed CM, Johnson HM. The role of a non-canonical JAK-STAT pathway in IFN therapy of poxvirus infection and multiple sclerosis: an example of Occam’s Broom? JAKSTAT (2013) 2(4):e26227. doi:10.4161/jkst.26227

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Reich NC, Liu L. Tracking STAT nuclear traffic. Nat Rev Immunol (2006) 6(8):602–12. doi:10.1038/nri1885

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Brodsky FM, Chen CY, Knuehl C, Towler MC, Wakeham DE. Biological basket weaving: formation and function of clathrin-coated vesicles. Annu Rev Cell Dev Biol (2001) 17:517–68. doi:10.1146/annurev.cellbio.17.1.517

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Filgueira L, Groscurth P, Aguet M. Binding and internalization of gold-labeled IFN-gamma by human Raji cells. J Immunol (1989) 142(10):3436–9.

PubMed Abstract | Google Scholar

91. Sharma P, Varma R, Sarasij RC, Ira K, Gousset K, Krishnamoorthy G, et al. Nanoscale organization of multiple GPI-anchored proteins in living cell membranes. Cell (2004) 116(4):577–89. doi:10.1016/S0092-8674(04)00167-9

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Takaoka A, Mitani Y, Suemori H, Sato M, Yokochi T, Noguchi S, et al. Cross talk between interferon-gamma and -alpha/beta signaling components in caveolar membrane domains. Science (2000) 288(5475):2357–60. doi:10.1126/science.288.5475.2357

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Truchet S, Wietzerbin J, Debey P. Mouse oocytes and preimplantation embryos bear the two sub-units of interferon-gamma receptor. Mol Reprod Dev (2001) 60(3):319–30. doi:10.1002/mrd.1094

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Subramaniam PS, Johnson HM. Lipid microdomains are required sites for the selective endocytosis and nuclear translocation of IFN-gamma, its receptor chain IFN-gamma receptor-1, and the phosphorylation and nuclear translocation of STAT1alpha. J Immunol (2002) 169(4):1959–69. doi:10.4049/jimmunol.169.4.1959

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Sadir R, Lambert A, Lortat-Jacob H, Morel G. Caveolae and clathrin-coated vesicles: two possible internalization pathways for IFN-gamma and IFN-gamma receptor. Cytokine (2001) 14(1):19–26. doi:10.1006/cyto.2000.0854

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Farrar MA, Fernandez-Luna J, Schreiber RD. Identification of two regions within the cytoplasmic domain of the human interferon-gamma receptor required for function. J Biol Chem (1991) 266(29):19626–35.

PubMed Abstract | Google Scholar

97. Hershey GK, Schreiber RD. Biosynthetic analysis of the human interferon-gamma receptor. Identification of N-linked glycosylation intermediates. J Biol Chem (1989) 264(20):11981–8.

PubMed Abstract | Google Scholar

98. Anderson P, Yip YK, Vilcek J. Human interferon-gamma is internalized and degraded by cultured fibroblasts. J Biol Chem (1983) 258(10):6497–502.

PubMed Abstract | Google Scholar

99. Celada A, Schreiber RD. Internalization and degradation of receptor-bound interferon-gamma by murine macrophages. Demonstration of receptor recycling. J Immunol (1987) 139(1):147–53.

PubMed Abstract | Google Scholar

100. Finbloom DS. Internalization and degradation of human recombinant interferon-gamma in the human histiocytic lymphoma cell line, U937: relationship to Fc receptor enhancement and antiproliferation. Clin Immunol Immunopathol (1988) 47(1):93–105. doi:10.1016/0090-1229(88)90148-1

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Szente BE, Johnson HM. Binding of IFN gamma and its C-terminal peptide to a cytoplasmic domain of its receptor that is essential for function. Biochem Biophys Res Commun (1994) 201(1):215–21. doi:10.1006/bbrc.1994.1691

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Ahmed CM, Johnson HM. IFN-gamma and its receptor subunit IFNGR1 are recruited to the IFN-gamma-activated sequence element at the promoter site of IFN-gamma-activated genes: evidence of transactivational activity in IFNGR1. J Immunol (2006) 177(1):315–21. doi:10.4049/jimmunol.177.1.315

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Mujtaba MG, Patel CB, Patel RA, Flowers LO, Burkhart MA, Waiboci LW, et al. The gamma interferon (IFN-gamma) mimetic peptide IFN-gamma (95-133) prevents encephalomyocarditis virus infection both in tissue culture and in mice. Clin Vaccine Immunol (2006) 13(8):944–52. doi:10.1128/CVI.00021-06

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Thiam K, Loing E, Delanoye A, Diesis E, Gras-Masse H, Auriault C, et al. Unrestricted agonist activity on murine and human cells of a lipopeptide derived from IFN-gamma. Biochem Biophys Res Commun (1998) 253(3):639–47. doi:10.1006/bbrc.1998.9831

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Albericio F, Kruger HG. Therapeutic peptides. Future Med Chem (2012) 4(12):1527–31. doi:10.4155/fmc.12.94

CrossRef Full Text | Google Scholar

106. Bansal R, Prakash J, De Ruiter M, Poelstra K. Interferon gamma peptidomimetic targeted to hepatic stellate cells ameliorates acute and chronic liver fibrosis in vivo. J Control Release (2014) 179:18–24. doi:10.1016/j.jconrel.2014.01.022

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Ramachandran P, Iredale JP, Fallowfield JA. Resolution of liver fibrosis: basic mechanisms and clinical relevance. Semin Liver Dis (2015) 35(2):119–31. doi:10.1055/s-0035-1550057

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Schuppan D, Kim YO. Evolving therapies for liver fibrosis. J Clin Invest (2013) 123(5):1887–901. doi:10.1172/JCI66028

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Pinzani M, Rombouts K, Colagrande S. Fibrosis in chronic liver diseases: diagnosis and management. J Hepatol (2005) 42(Suppl(1)):S22–36. doi:10.1016/j.jhep.2004.12.008

CrossRef Full Text | Google Scholar

110. Mehal WZ, Schuppan D. Antifibrotic therapies in the liver. Semin Liver Dis (2015) 35(2):184–98. doi:10.1055/s-0035-1550055

CrossRef Full Text | Google Scholar

111. Hauff P, Gottwald U, Ocker M. Early to phase II drugs currently under investigation for the treatment of liver fibrosis. Expert Opin Investig Drugs (2015) 24(3):309–27. doi:10.1517/13543784.2015.997874

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for fibrotic diseases: nearing the starting line. Sci Transl Med (2013) 5(167):167sr1. doi:10.1126/scitranslmed.3004700

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Muir AJ, Sylvestre PB, Rockey DC. Interferon gamma-1b for the treatment of fibrosis in chronic hepatitis C infection. J Viral Hepat (2006) 13(5):322–8. doi:10.1111/j.1365-2893.2005.00689.x

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Soza A, Heller T, Ghany M, Lutchman G, Jake Liang T, Germain J, et al. Pilot study of interferon gamma for chronic hepatitis C. J Hepatol (2005) 43(1):67–71. doi:10.1016/j.jhep.2005.02.023

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Borkham-Kamphorst E, Stoll D, Gressner AM, Weiskirchen R. Antisense strategy against PDGF B-chain proves effective in preventing experimental liver fibrogenesis. Biochem Biophys Res Commun (2004) 321(2):413–23. doi:10.1016/j.bbrc.2004.06.153

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Ogawa S, Ochi T, Shimada H, Inagaki K, Fujita I, Nii A, et al. Anti-PDGF-B monoclonal antibody reduces liver fibrosis development. Hepatol Res (2010) 40(11):1128–41. doi:10.1111/j.1872-034X.2010.00718.x

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Chen SW, Zhang XR, Wang CZ, Chen WZ, Xie WF, Chen YX. RNA interference targeting the platelet-derived growth factor receptor beta subunit ameliorates experimental hepatic fibrosis in rats. Liver Int (2008) 28(10):1446–57. doi:10.1111/j.1478-3231.2008.01759.x

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Chen SW, Chen YX, Zhang XR, Qian H, Chen WZ, Xie WF. Targeted inhibition of platelet-derived growth factor receptor-beta subunit in hepatic stellate cells ameliorates hepatic fibrosis in rats. Gene Ther (2008) 15(21):1424–35. doi:10.1038/gt.2008.93

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Neef M, Ledermann M, Saegesser H, Schneider V, Widmer N, Decosterd LA, et al. Oral imatinib treatment reduces early fibrogenesis but does not prevent progression in the long term. J Hepatol (2006) 44(1):167–75. doi:10.1016/j.jhep.2005.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Hong F, Chou H, Fiel MI, Friedman SL. Antifibrotic activity of sorafenib in experimental hepatic fibrosis: refinement of inhibitory targets, dosing, and window of efficacy in vivo. Dig Dis Sci (2013) 58(1):257–64. doi:10.1007/s10620-012-2325-y

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Berardis S, Dwisthi Sattwika P, Najimi M, Sokal EM. Use of mesenchymal stem cells to treat liver fibrosis: current situation and future prospects. World J Gastroenterol (2015) 21(3):742–58. doi:10.3748/wjg.v21.i3.742

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Poelstra K, Beljaars L, Melgert BN. Cell-specific delivery of biologicals: problems, pitfalls and possibilities of antifibrotic compounds in the liver. Drug Discov Today (2013) 18(23–24):1237–42. doi:10.1016/j.drudis.2013.05.013

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Jiménez Calvente C, Sehgal A, Popov Y, Kim YO, Zevallos V, Sahin U, et al. Specific hepatic delivery of procollagen alpha1(I) siRNA in lipid-like nanoparticles resolves liver fibrosis. Hepatology (2015). doi:10.1002/hep.27936

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Elrick LJ, Leel V, Blaylock MG, Duncan L, Drever MR, Strachan G, et al. Generation of a monoclonal human single chain antibody fragment to hepatic stellate cells – a potential mechanism for targeting liver anti-fibrotic therapeutics. J Hepatol (2005) 42(6):888–96. doi:10.1016/j.jhep.2005.01.028

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Beljaars L, Molema G, Weert B, Bonnema H, Olinga P, Groothuis GM, et al. Albumin modified with mannose 6-phosphate: a potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic stellate cells. Hepatology (1999) 29(5):1486–93. doi:10.1002/hep.510290526

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Beljaars L, Molema G, Schuppan D, Geerts A, De Bleser PJ, Weert B, et al. Successful targeting to rat hepatic stellate cells using albumin modified with cyclic peptides that recognize the collagen type VI receptor. J Biol Chem (2000) 275(17):12743–51. doi:10.1074/jbc.275.17.12743

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Yang N, Singh S, Mahato RI. Targeted TFO delivery to hepatic stellate cells. J Control Release (2011) 155(2):326–30. doi:10.1016/j.jconrel.2011.06.037

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Li F, Li QH, Wang JY, Zhan CY, Xie C, Lu WY. Effects of interferon-gamma liposomes targeted to platelet-derived growth factor receptor-beta on hepatic fibrosis in rats. J Control Release (2012) 159(2):261–70. doi:10.1016/j.jconrel.2011.12.023

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Bartneck M, Warzecha KT, Tacke F. Therapeutic targeting of liver inflammation and fibrosis by nanomedicine. Hepatobiliary Surg Nutr (2014) 3(6):364–76. doi:10.3978/j.issn.2304-3881.2014.11.02

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Koivunen E, Wang B, Ruoslahti E. Phage libraries displaying cyclic peptides with different ring sizes: ligand specificities of the RGD-directed integrins. Biotechnology (N Y) (1995) 13(3):265–70. doi:10.1038/nbt0395-265

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Marcelino J, McDevitt CA. Attachment of articular cartilage chondrocytes to the tissue form of type VI collagen. Biochim Biophys Acta (1995) 1249(2):180–8. doi:10.1016/0167-4838(95)00026-Q

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Fretto LJ, Snape AJ, Tomlinson JE, Seroogy JJ, Wolf DL, LaRochelle WJ, et al. Mechanism of platelet-derived growth factor (PDGF) AA, AB, and BB binding to alpha and beta PDGF receptor. J Biol Chem (1993) 268(5):3625–31.

PubMed Abstract | Google Scholar

133. Bansal R, Prakash J, de Ruijter M, Beljaars L, Poelstra K. Peptide-modified albumin carrier explored as a novel strategy for a cell-specific delivery of interferon gamma to treat liver fibrosis. Mol Pharm (2011) 8(5):1899–909. doi:10.1021/mp200263q

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Hagens WI, Mattos A, Greupink R, de Jager-Krikken A, Reker-Smit C, van Loenen-Weemaes A, et al. Targeting 15d-prostaglandin J2 to hepatic stellate cells: two options evaluated. Pharm Res (2007) 24(3):566–74. doi:10.1007/s11095-006-9175-2

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Poosti F, Bansal R, Yazdani S, Prakash J, Post E, Klok P, et al. Selective delivery of IFN-gamma to renal interstitial myofibroblasts: a novel strategy for the treatment of renal fibrosis. FASEB J (2015) 29(3):1029–42. doi:10.1096/fj.14-258459

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Subramaniam PS, Flowers LO, Haider SM, Johnson HM. Signal transduction mechanism of a peptide mimetic of interferon-gamma. Biochemistry (2004) 43(18):5445–54. doi:10.1021/bi036213t

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Szente BE, Soos JM, Johnson HW. The C-terminus of IFN gamma is sufficient for intracellular function. Biochem Biophys Res Commun (1994) 203(3):1645–54. doi:10.1006/bbrc.1994.2375

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Bansal R, Tomar T, Ostman A, Poelstra K, Prakash J. Selective targeting of interferon gamma to stromal fibroblasts and pericytes as a novel therapeutic approach to inhibit angiogenesis and tumor growth. Mol Cancer Ther (2012) 11(11):2419–28. doi:10.1158/1535-7163.MCT-11-0758

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev (2008) 22(10):1276–312. doi:10.1101/gad.1653708

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Bonner JC. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev (2004) 15(4):255–73. doi:10.1016/j.cytogfr.2004.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Angeli F, Koumakis G, Chen MC, Kumar S, Delinassios JG. Role of stromal fibroblasts in cancer: promoting or impeding? Tumour Biol (2009) 30(3):109–20. doi:10.1159/000218708

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Mueller MM, Fusenig NE. Friends or foes – bipolar effects of the tumour stroma in cancer. Nat Rev Cancer (2004) 4(11):839–49. doi:10.1038/nrc1477

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Prakash J, de Jong E, Post E, Gouw AS, Beljaars L, Poelstra K. A novel approach to deliver anticancer drugs to key cell types in tumors using a PDGF receptor-binding cyclic peptide containing carrier. J Control Release (2010) 145(2):91–101. doi:10.1016/j.jconrel.2010.03.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: liver fibrosis, drug targeting, therapy, PDGFβ receptor, PDGF-BB, IFNγ, hepatic stellate cell

Citation: van Dijk F, Olinga P, Poelstra K and Beljaars L (2015) Targeted therapies in liver fibrosis: combining the best parts of platelet-derived growth factor BB and interferon gamma. Front. Med. 2:72. doi: 10.3389/fmed.2015.00072

Received: 09 July 2015; Accepted: 18 September 2015;
Published: 05 October 2015

Edited by:

Achim Weber, University of Zurich, Switzerland

Reviewed by:

Francesco Trapasso, University “Magna Græcia” of Catanzaro, Italy
Michael Hölzel, University Hospital Bonn, Germany

Copyright: © 2015 van Dijk, Olinga, Poelstra and Beljaars. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Fransien van Dijk, Department of Pharmacokinetics, Toxicology and Targeting, Rijksuniversiteit Groningen, A. Deusinglaan 1, Groningen 9713 AV, Netherlands, fransien.van.dijk@rug.nl

Download