Hepatitis C virus–cell interactions and their role in pathogenesis

Hepatitis C virus–cell interactions and their role in pathogenesis

Clin Liver Dis 7 (2003) 67 – 88 Hepatitis C virus–cell interactions and their role in pathogenesis Stephen J. Polyak, PhD Departments of Laboratory M...

298KB Sizes 0 Downloads 92 Views

Clin Liver Dis 7 (2003) 67 – 88

Hepatitis C virus–cell interactions and their role in pathogenesis Stephen J. Polyak, PhD Departments of Laboratory Medicine and Microbiology, University of Washington, Box 359690, 325 9th Avenue, Seattle, WA, 98104-2499, USA

At the time a virus infects a cell, it encounters a hostile environment that is unsuited to its survival and propagation. A successful virus therefore must assume control of the cell to promote conditions favorable for an infectious cycle. For persistent viral infections, this control of the host must facilitate longterm (and sometimes attenuated) replication. The genes and gene products the virus brings into the cell not only are required to promote the replication of large quantities of progeny viruses, but they also must interact with or perturb host cell proteins that are required for viral replication or aimed at the virus’ demise. Thus, the study of virus – host interactions provides insight into mechanisms of virus persistence, antiviral resistance, and pathogenesis.

The medical significance of hepatitis C virus infection As covered elsewhere in this issue, hepatitis C virus (HCV) infects an estimated 3% or 170 million of the world’s population and causes an estimated 476,000 deaths per year caused by complications of end– stage liver disease. Of those acutely infected with HCV, most develop chronic infection. Moreover, 70% of patients with chronic viremia develop histological evidence of chronic liver disease. Chronic infection results in a spectrum of liver disease, including chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Hepatitis C is the most frequent indication for liver transplantation [1,2] (see article by Rosen in this issue for more detailed information). In Europe, approximately 9 million people are infected with HCV, 5 million in the European Union, with a variable prevalence ranging from 0.5% in Northern Europe to 2% in Southern Europe. The prevalent genotype in Europe is 1b (47%), followed by 1a (17%), 3 (16%), 2 (13%), and other genotypes (7%) [3]. In the United States, about 1.8% of the general

E-mail address: [email protected] 1089-3261/03/$ – see front matter D 2003, Elsevier Science (USA). All rights reserved. PII: S 1 0 8 9 - 3 2 6 1 ( 0 2 ) 0 0 0 7 5 - 2

68

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

population or 4 million people are infected. About 57% of cases involve genotype 1a, 17% genotype 1b, 14% genotype 2, 7% genotype 3, 0.9% genotype 4, and 3.2% genotype 6 [4,5]. The prevalent genotype in Japan is type 1b.

Acute hepatitis C virus infection As alluded to previously, progression to chronic hepatitis C is the norm for the most acutely infected patients. A growing research area involves the characterization of virus and host factors associated with clearance of acute HCV infection. An impediment to these studies is the difficulty in diagnosing acute HCV infection, because the initial manifestations are often subclinical. The establishment of networks to identify and manage healthcare workers with occupational exposure to HCV has greatly facilitated research on acute HCV [6,7], however. Studies of acutely infected patients have demonstrated that broad, multi-specific cytotoxic T lymphocyte (CTL) immune responses are associated with HCV clearance [6,8,9]. Similar findings also have been made during experimental infection of chimpanzees [10]. Recently, two mechanisms have been described whereby HCV – host interactions appear to influence the progression of acute to chronic hepatitis C. The generation of viruses with mutations in CTL epitopes has been shown to permit acute HCV to persist by escape from the cell-mediated immune response [11]. Moreover, the ability of the HCV NS5A protein to interact with the interferon (IFN) -induced cellular protein, RNA-activated protein kinase (PKR), correlates with progression from acute to chronic HCV infection [12]. Understanding the factors involved in clearance of acute HCV will continue to be an important area of research.

Antiviral therapy for hepatitis C virus Recombinant antiviral IFN-a has been used for more than a decade as a treatment of chronic hepatitis C, despite the fact that most patients still fail to have sustained responses (ie, clearance of viremia) [13] (see article in this issue by McHutchison and Fried for more detailed information). More recent therapies including IFN plus ribavirin [14 – 16], pegylated IFN [17,18], and pegylated IFN plus ribavirin [19], result in significant improvements in sustained response rates. Nonetheless, HCV viremia is not eradicated in more than 60% of treated patients. This is particularly evident in patients with genotype 1a or 1b HCV infections. This is clearly a problem in areas where genotype 1 is common such as in North America, Europe, and Japan. In this light, HCV seems resistant to IFN antiviral therapy. As a consequence, much attention has been focused on characterizing HCV – host interactions that contribute to resistance to IFN therapy. Elucidation of the mechanisms of antiviral resistance is important for improving therapy for chronic hepatitis C. An additional benefit from studies of this nature is that insights into viral pathogenesis often are uncovered simultaneously.

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

69

The modus operandi for studying HCV –host interactions usually involves overexpression of HCV RNA and proteins in cell culture and assessment of the effects on host cell metabolism and signal transduction pathways. It is therefore possible that the level of HCV protein expression in cellular systems is higher than in the HCV-infected liver, raising questions about the biological relevance of the results. To this end, some studies use regulated expression systems that facilitate HCV protein analysis at more physiological concentrations. Importantly, in other research arenas, similar studies have permitted the dissection of molecular mechanisms and the development of specific inhibitors of various pathways. In some cases, the inhibitors have found their way into the clinic, such as the PKC inhibitor CGP41251 [20], the BCR-ABL tyrosine kinase inhibitor STI571 [21], and the natural inhibitor of angiogenesis, angiostatin [22]. Thus, the study of HCV – host interactions also may open up new avenues for therapeutic intervention through targeted disruption or promotion of virus –host protein complexes.

Virus – host interactions during the hepatitis C virus life cycle For HCV, virus – cell interactions can be grouped into three broad categories: entry, replication, and assembly/release. These events are summarized in Fig. 1. In the initial stage of the infection, HCV binds to and enters the target cell. This virus – cell interaction may be mediated by at least two putative cellular receptors for HCV, the low-density lipoprotein receptor, LDLR or CD81. The HCV envelope glycoproteins, E1 and E2, are responsible for binding the cell surface receptors. E1 and E2 are believed to be present on the virion surface in a noncovalently associated complex. The complex is formed during virus assembly, which occurs at the endoplasmic reticulum [23]. HCV particles in human sera are complexed with antibodies and low-density lipoproteins (LDL) [24,25]. Agnello et al demonstrated that HCV – LDL complexes are capable of entry into various human liver cell lines [26]. Although the precise molecular events involved in LDLR-mediated entry await further characterization, up-regulation of LDLR using chemicals such as lovastatin appear to enhance HCV entry in this model system. If LDLR turns out to be a bona fide cellular receptor for LDL-complexed HCV, then the lack of LDLR and the use of LDL-free HCV-infected serum might explain the generalized lack of reproducibility and poor success of serum-based HCV culture systems. The fact that HCV in patient serum is complexed not only with LDL but with antibodies also may influence the outcome of serum-based HCV infection systems, if Fc receptormediated endocytosis is operative. Abrigani et al demonstrated that the HCV E2 glycoprotein bound the tetraspanin molecule, CD81, on human B cells [27]. E2 was shown to bind CD81 present on B cells and various human cell lines including hepatoma Huh7 cells, suggesting that the E2-CD81 interaction is biologically relevant in getting HCV into the liver. Thus, CD81 originally was touted as a receptor for HCV. Several subsequent studies, however, indicated that this interpretation may have been premature, because there seems to be no correlation between the sequence

70

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

Fig. 1. The HCV life cycle. In the first step, HCV binds to and enters cells. This step may be mediated by interaction of HCV glycoproteins with CD81 and LDLR. HCV then is uncoated in acidic endosomes (H+) where replication occurs. Replication involves multiple virus – virus and virus – host interactions that regulate HCV polyprotein production and processing, and genome (+RNA) versus antigenome synthesis. HCV proteins accumulated in the endoplasmic reticulum (ER) and Golgi apparatus, where assembly and release of progeny viruses, occur. Abbreviations: RdRp, RNA dependent RNA polymerase; Rep, HCV replicase complex.

of CD81 from various species and susceptibility of cell lines to HCV infection. For example, E2 binds to tamarin CD81 more tightly than human CD81 in vitro and on cell lines, yet tamarins are not susceptible to HCV infection [28,29]. More recent studies using cells from Tupeia belangeri (tree shrew) suggest an alternative receptor for HCV. Tupeia hepatocytes support HCV binding and infection, and this seems to be independent of CD81 [30]. Furthermore, Baumert’s group has demonstrated that the binding of HCV virus-like proteins (containing core, E1 and E2) bind to human cell lines independent of CD81 and LDLR [31]. These studies suggest that there are additional cell surface molecules that mediate HCV entry into cells. In other viral infections, there are precedents for this concept. For example, HIV uses CD4 and chemokine receptors [32] for virus entry, while herpesviruses (HSV) use herpes virus entry mediator (HVEM; a member of the tumor necrosis factor [TNF] receptor family), heparin sulfate, and nectins 1 and 2 (members of the immunoglobulin superfamily) [33,34]. For HSV, multiple viral glycoproteins are required during binding, fusion, and entry. Specifically, glycoproteins gB and gC mediate high affinity interaction with cell

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

71

surface heparin sulfate, followed by low-affinity interaction of gD with HVEM, nectin 1, or nectin 2 [33,34]. Interaction of gD with any of the receptors seems to trigger fusion. HCV may use E1 and E2 during virus entry, because it has been reported that E1 may interact with LDLR [35], while E2, as described previously, binds to CD81. Thus, HCV entry likely involves E1 and E2 proteins and multiple cell surface molecules. Differential usage of these molecules may be required for HCV entry into nonhepatocyte cells such as lymphocytes, monocytes, and bone marrow [36 – 45] and also may influence HCV-associated pathology. Even if CD81 is not involved in HCV entry directly, CD81 engagement by HCV virions theoretically could affect immune cell activation functions. Indeed, cross-linking of CD81 with monoclonal antibodies or immobilized E2 inhibited NK cytotoxic activity and IFN-g production [46] and granule release, proliferation, and activated mitogen – activated protein kinase (MAPK) signaling cascades [47]. Cross-linking of CD81 also seems to make T cells hyper-responsive in interleukin (IL)-2, IFN-g, and IL-4 production [48]. These studies suggest that the accessory interaction of HCV with CD81 may affect the development of innate and acquired immunity to HCV. HCV infection is not a prerequisite for these biological effects on the immune system. Thus, cell function may be altered as immune cells ‘‘sample’’ the microenvironment through HCV –host interactions that are limited to molecules on the cell surface. As shown in Fig. 1, after HCV enters the cell, it is uncoated. This may occur in acidic endosomes, because lysosomotropic agents that raise the pH of endosomes inhibit the entry of pseudotyped E1 – and E2 – containing VSV particles [35]. Although this effect may be caused by the vesicular stomatitis virus (VSV) backbone used in this study, many other viruses including arenaviruses, Hantaan virus, hepatitis A virus, and Semliki Forest virus, require low pH for viral entry [49 – 53]. Most notably, entry of the flavivirus St. Louis Encephalitis virus into mosquito cells requires low pH [54]. In theory, HCV interaction with other cell surface molecules may be a postreceptor-binding event that directs internalized HCV into the acidic endosome, reinforcing the concept that multiple interactions between HCV glycoproteins and cellular membrane proteins are involved in HCV entry. Following uncoating, the HCV RNA is translated, and the polyprotein is processed proteolytically into the 10 viral proteins. The first third of the polyprotein encodes the structural proteins that form the three-dimensional architecture of the virus. They include the core protein, two envelope proteins (E1, E2), and a protein of unknown function (P7). An additional open reading frame in the core gene recently was described, although the role of this protein in HCV replication and pathogenesis remains to be determined [55]. Nonstructural proteins are derived from the remaining two-thirds of the polyprotein and have functions in HCV replication. They include NS2, NS3 (a serine protease/helicase), NS4A, NS4B, NS5A, and NS5B (the RNA – dependent RNA polymerase) [56]. Successful viral protein translation and genome replication requires the coordination of multiple viral and cellular proteins. For instance, polypyrimidine tract binding protein (PTB) has been shown to bind to the core coding region and 30 UTR [57,58]. PTB

72

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

binding to the core region inhibits HCV translation, which is relieved if RNA transcripts contain the 30UTR. These data suggest that PTB binding to HCV RNA may facilitate the switch from HCV polyprotein translation to negative strand replication. More recent studies suggest that the La antigen, an important cellular translation factor, may also interact with the 50 and 30 UTRs [59,60]. The previous examples underscore the reliance of HCV on cellular factors for successful replication. Continued research into this area likely will uncover other HCV – host interactions that may be suitable targets for therapeutic drug discovery. Specificity is achievable if the engineered molecule can be directed to the HCV – host interaction, thereby disrupting only those cells that contain replicating virus.

The hepatitis C virus core and NS5A proteins As alluded to previously, HCV – host interactions have been investigated intensely despite the lack of a robust viral infection system. As a result, many laboratories have established model cell culture systems to study HCV –host interactions, with a major focus on the core and NS5A proteins. At first glance, these two viral proteins appear different in several aspects. The first is their position in the HCV genome; they are at opposite ends, with core being the first protein produced, while NS5A is penultimate. Second, the encoded proteins are different in size, with the core being 19 to 21 kDa and NS5A being 56 to 58 kDa. The third difference lies in the presumed roles of the proteins in the lifecycle of HCV. The HCV core is a structural protein that binds RNA and ribosomes [61] and presumably promotes HCV capsid formation, while NS5A is probably a component of the HCV polymerase complex [62] and as such, functions in HCV replication. The proteins share several similarities, however. One is in subcellular localization; the predominant forms of the proteins are found in the cytoplasm. This is illustrated in Fig. 2, which depicts immunofluorescent detection of NS5A (left panel) and core (right panel) in human cells. Both proteins are localized predominantly to the perinuclear area in the cytoplasm. This location of HCV proteins to this region of the cell is not unique. Indeed, all HCV proteins have been localized to a similar region within the cell [63 – 65]. The location within the cell to where HCV proteins localize corresponds to the endoplasmic reticulum (ER) and golgi apparatus. This is the region where HCV replication is believed to occur (see Fig. 1) and is consistent the subcellular localization of other flaviviruses including West Nile virus [66] and dengue virus [67]. The targeting of HCV protein expression to the ER membrane likely is facilitated by way of interaction of HCV proteins with cellular proteins. In this regard, NS5A and NS5B interact with hVAP-33, a cellular protein found primarily in the ER [68]. Core and NS5A proteins also share the similarity of colocalizing on cytoplasmic lipid droplets in hepatocytes [69] and having truncated forms that localize to the nucleus [70,71]. Recently, a small amphipathic alpha helix in the amino terminus of NS5A was shown to mediate retention of NS5A on cytoplasmic membranes [72]. The biological implications of the location of core and NS5A in the cell may relate

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

73

Fig. 2. Localization of HCV proteins in human cells. NS5A (left panel) and core (right panel) proteins were expressed in human osteosarcoma and hepatocytes (Huh7) cells, respectively, and immunofluorescent detection of the proteins was performed. As can be seen, both proteins are located primarily in the cytoplasm, although small amounts of NS5A can be seen in the nucleus and core is associated with cytoplasmic lipid droplets.

to the proteins’ abilities to perturb several signal transduction pathways involved in cell cycle, antiviral, apoptotic, and lipid regulation.

HCV NS5A – host interactions For genotype 1a and 1b isolates, NS5A is 448 amino acids in length and is post-translationally modified by a series of phosphorylations on conserved serine residues, resulting in the production of phosphorylated and hyperphosphorylated forms of the protein [73 – 80]. Major sites for phosphorylation of NS5A on Ser-2194 and Ser-2321 have been described recently [81,82], and the cellular serine kinase(s) responsible for NS5A phosphorylation has been characterized partially [77,83,84]. The exact role of NS5A phosphorylation on protein function is not known, but the serines are conserved strictly in vivo, despite the fact that many other regions of NS5A mutate during chronic HCV infection [85]. Conversely, studies using HCV subgenomic replicons suggest that hyperphosphorylation of NS5A is not required for HCV replication [86]. Many studies on the effects of NS5A expression on cellular metabolism and physiology have been performed. During the course of these studies, NS5A has emerged as an important protein in the HCV life cycle. Fig. 3A summarizes some of the structure – function relationships for NS5A that are described in the proceeding section. In general, NS5A may directly regulate HCV replication through interaction with the HCV –encoded polymerase complex [62,87]. NS5A’s role as a direct modulator of viral replication also stems from observations that adaptive mutations in NS5A increase the replication of HCV subgenomic replicons [86,88,89]. Mutations in NS5A also accumulate during IFN therapy of patients

74

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

Fig. 3. NS5A (A) and core (B) structure function relationships. The top panel of each figure depicts the position of the gene in the context of the HCV polyprotein, along with major phosphorylation sites. Below each figure are the regions of each protein that are required for various activities including cytoplasmic retention (cyto) [72], interferon sensitivity determining region (ISDR) [90], PKR binding domain (PKR) [135], nuclear localization signal (NLS) [70], Grb2 binding domain (Grb2) [92], variable domains 3 [149] and 4 [85] (V3, V4). The domains required for binding cellular proteins and RNAs also are indicated.

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

75

with HCV [85,90] and in chimpanzees that develop chronic infection following experimental inoculation with HCV [91]. Although the mutations described in vitro are not found in NS5A genes isolated from patients (data not shown), the fact that NS5A accumulates mutation in vitro and in vivo suggests that NS5A is under selective pressure in diverse environments. NS5A also promotes a cellular environment favorable to HCV replication, a function of the protein usually taking the form of perturbation of a signal transduction pathway. For example, NS5A interacts with Grb2, an adaptor protein involved in MAPK signaling, resulting in inhibition of epidermal growth factor (EGF) signaling by way of reduced extracellular regulated kinase (ERK) 1 and ERK2 phosphorylation [92]. A follow-up study reported that recombinant vaccinia virus expressing NS5A inhibited double-stranded RNA – activated protein kinase (PKR) activity and the subsequent phosphorylation of the eukaryotic initiation factor 2 (eIF2a) [93]. In the same study, NS5A was shown to perturb the p38 MAPK pathway, which normally leads to eIF – 4E phosphorylation, suggesting that NS5A may regulate cellular translation to favor IRESmediated translation during HCV infection [93]. These studies also imply that NS5A can affect the control of cellular proliferation by affecting homeostasis of mRNA translation. In this regard, NS5A inhibits apoptosis in some [94,95], but not all systems [96]. Proposed mechanisms for NS5A inhibition of apoptosis include negative regulation of PKR [94], inhibition of TNF-a induced activation of NF-kB by way of direct interaction with TRAF2 [97], and induction of oxidative stress and activation of NF-kB [98]. Another study suggests NS5A is inhibitory to the cell cycle by way of perturbation of Cdk1/2 complexes [99]. Given the inherent variability of all clinical NS5A isolates [85], the ability of NS5A to localize in the nucleus and cytoplasm [100], and the different cellular systems used, it is not surprising that NS5A has been reported to have such diverse effects. Nonetheless, the modulation of cellular proliferation and apoptosis by NS5A may have implications for the pathogenesis of HCV-related liver damage. In this regard, NS5A-mediated inhibition of apoptosis is associated with transformation of NIH 3T3 cells and tumor formation in nude mice, suggesting that NS5A has an oncogenic potential [94]. Moreover, NS5A associates with apolipoprotein AI (apoAI) on the surface of lipid droplets in Huh7 cells [69]. Because steatosis or lipid accumulation in the liver is a frequent histological finding in chronic hepatitis C, the interaction of NS5A with apoAI may be involved in the pathogenesis of hepatic steatosis. Interestingly, NS5A is not the only HCV protein that interacts with cellular apolipoproteins, as the HCV core protein binds apoAII.

HCV core– host interactions Like NS5A, the HCV core protein also seems to be a major player in the regulation of cellular growth and HCV pathogenesis. Core expression in transgenic mice induces hepatic steatosis and hepatocellular carcinoma [101,102].

76

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

Moreover, transgenic mice expressing HCV structural and nonstructural proteins also develop hepatic steatosis and hepatocellular carcinoma [103]. In this model system, steatosis develops in animals expressing core, E1, and E2 proteins, while the additional expression of the nonstructural proteins is a predisposing factor for developing cancer. The association of core with apolipoprotein A2 on lipid droplets in hepatocytes may be involved in the pathogenesis of liver disease in transgenic animals [104], through altered very low density lipoprotein (VLDL) secretion [105] and by way of core-induced oxidative stress and mitochondrial injury [106]. As described previously, NS5A binds apoAI on cellular lipid droplets in human hepatocytes [69]. Therefore, the interaction of HCV proteins with cellular apolipoproteins in hepatocytes may be mechanistically involved in the development of steatosis. Core protein also has been shown to interact with the cytoplasmic tail of the TNF-a receptor and induce apoptosis [107,108]. Studies from other groups, however, have reported that the HCV core protein inhibits apoptosis, promotes cell growth, and can transform cells to a tumorigenic phenotype [109 – 111]. Similarly, contradictory results on the effects of expression of the HCV core protein on MAPK, p53, p21/WAF, and NF-kB have been reported [112 – 116]. The reasons for these discrepancies are not clear, although the cell lines used may affect the apoptotic responses observed [117], or differences in subcellular localization of the core protein might explain the dichotomous effects of the protein [71]. Disparties could also be due to sequence differences among core quasispecies isolates. In a recent study, core was shown to interact directly with NS5A, inducing caspase-mediated cleavage of NS5A and apoptosis of osteosarcoma cells [118], suggesting that it is important to evaluate the effects of a single HCV protein in the context of additional, or ideally, all HCV proteins. A summary of the structure-function relationships for core is presented in Fig. 3B. In summary, the HCV core and NS5A proteins engage in many interactions within the host cell that have a significant influence on HCV replication and pathogenesis, and on HCV antiviral resistance.

The interferon system Many different signal transduction pathways are engaged in the binding of cytokines and chemokines to their cognate receptors. In general, these pathways involve a series of phosphorylation-dependent protein – protein interactions and small molecule second messengers that culminate in the assembly of transcription factor complexes to induce genes that mediate the biological activity of the cytokine or chemokine. The STAT/JAK (signal transducers and activator of transcription/Janus-associated kinase) pathway is the principle pathway used by IFN-a. At the time IFN-a binds its receptor, two receptor-associated tyrosine kinases, Tyk2 and Jak1, become activated by phosphorylation and phosphorylate STAT-1 and STAT-2 on conserved tyrosine residues [119]. STAT-1 and STAT-2 combine with the p48 protein to form the transcription factor IFN-

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

77

stimulated gene factor 3 (ISGF-3), which enters the nucleus, binds to the IFNstimulated response element (ISRE), inducing transcription of IFN-a– induced genes [119]. Three IFN-induced genes have been studied extensively, the 20 – 50 oligoadenylate synthetase (OAS)/RNAse L system, PKR, and myxovirus resistance proteins (Mx). The constitutive expression of the RNAse L and PKR pathways contribute to the IFN antiviral state as observed by the increased susceptibility of cells to viral infection from mice devoid of these enzymes [120 –122]. The ubiquitous RNase L is activated by 20 –50-oligoadenylate (2-5A); OAS synthesizes 2 – 5A from ATP in response to double-stranded RNA, such as the replicative intermediates of certain RNA viruses [123,124]. RNAse L is believed to degrade viral RNA and is implicated in the control of positive – stranded RNA viruses such as the picornavirus and encephalomyocarditis virus (EMCV). PKR is an IFN-inducible dsRNA-activated serine/threonine kinase. Its action is mediated through inhibition of protein synthesis caused by phosphorylation of eIF2a and has been partially implicated in IFN inhibition of EMCV [125,126]. Mx proteins mediate the inhibition of VSV and orthomyoxoviruses by way of inhibition of viral genome amplification and protein synthesis [127]. Many viruses including HIV, poliovirus, HSV, influenza virus, adenovirus and vaccinia virus encode RNAs or proteins that specifically inhibit the IFN system, either by inhibiting STAT/JAK signal transduction or by inhibiting the enzymatic activity of IFN –induced cellular proteins. HCV can be added to this list of viral inhibitors of the IFN system.

Hepatitis C virus inhibition of the interferon system Hepatitis C virus proteins have been shown to interact with and inhibit the IFN system at several levels. For example, expression of the entire HCV polyprotein inhibited IFN – induced STAT/JAK signaling in human U2-OS osteosarcoma cells [128]. HCV proteins also interact with and inhibit the antiviral functions of IFN – induced host cell proteins. The author’s group initially demonstrated that NS5A partially inhibits the antiviral actions of IFN [129], a finding that has been verified by several other laboratories [94,130 –133]. At present, two mechanisms for inhibiting the antiviral actions of IFN by NS5A have been described. NS5A has been shown to interact with PKR, inhibiting PKR kinase activity [134,135]. These findings provided a molecular explanation for the clinical observation that mutations within a small, 40 amino acid domain in NS5A, termed the IFN sensitivity determining region (ISDR), are associated with response to IFN therapy [90,136]. Gale et al showed that the ISDR was required for binding to PKR, and accumulation of mutations within the ISDR abrogated the interaction of NS5A with PKR. Because PKR induction and activation by IFN inhibit the translation of viral mRNAs, blockade of PKR activity is presumed to permit HCV replication to continue during IFN challenge. Enomoto’s and Gale’s findings were controversial, because other clinical and in vitro studies could not verify the

78

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

association of ISDR mutations and response to therapy [137] or interaction of NS5A with PKR [130]. Some of these differences are most certainly attributable to methodological differences. Because the bulk of ISDR studies limit the analysis to the ISDR and PKR binding region, it is premature to make conclusions on the biological activity of NS5A, especially because it is not known a priori how mutations in other regions of NS5A affect the conformation of the ISDR and PKR binding domains. It is known, however, that NS5A is variable throughout the entire gene and between different patients [85], indicating that future studies should focus on the entire gene. Furthermore, recent meta-analyses of the ISDR clinical studies indicate that low sample size was the predominant reason why previous studies could not find an association between ISDR mutation and IFN response [138]. Thus, the association of the ISDR with IFN response seems to have been validated, and one mechanism by which this occurs involves targeted disruption of PKR. Hopefully, this will provide a modus vivendi for the field. Recently, a novel mechanism for inhibition of the IFN system by the NS5A protein was characterized. NS5A was shown to induce the expression of the proinflammatory chemokine IL-8, which was associated with inhibition of IFN action [139]. NS5A induced transcriptional activation of the IL-8 promoter, possibly involving the transcription factors NF-kB and AP-1. Because NS5A can activate transcription, and NS5A mutants lacking amino termini can enter the nucleus, the precise mechanisms of IL-8 induction await further characterization. Using cDNA microarray analysis, NS5A also was shown to increase the expression of IL-8 in human hepatocytes, thereby extending this finding to the natural reservoir for HCV infection [140]. Mechanistically, the IL-8 anti-IFN action in EMCV-infected cells was associated with decreased OAS activity in a manner that was independent of OAS gene expression [141], suggesting that inhibition of the IFN system occurs at a post-transcriptional level. The IL-8 suppressive action on OAS activity in the presence of IFN-a seems to be linked to the type of the virus, because the antiIFN action of IL-8 was not seen with VSV infection. It appeared that IL-8 had no dramatic effect on ISRE-driven 6 –16 gene expression, supporting the hypothesis that IL-8 apparently blocks the antiviral action of IFN-a at late, such as OAS activity, rather than early stage. This is also the case with HCV NS5A-induced inhibition of IFN antiviral action, whereas NS5A protein expression did not appear to affect IFN-mediated signaling in HeLa cells [139], although it is still possible that NS5A affects STAT/JAK signal transduction at some level. Interleukin-8 also stimulates the replication of several viruses including CMV, HIV, and adenovirus. Although the effect of IL-8 on HCV replication has not been reported, it is intriguing that HCV –infected Saudi Arabian patients have elevated IL-8 and TNF-a (a physiological inducer of IL-8) levels, with biochemical nonresponders to IFN therapy having highest serum levels [142]. Elevated IL-8 also is associated with other viral infections including HIV, rhinovirus, and cytomegalovirus [143]. Another study, however, arrived at exactly the opposite conclusion. Researchers found that elevated IL-8 levels are correlated with IFN response in French patients with HCV [144]. In this

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

79

study, the sample size was low (N = 18) compared with the study on Saudi patients (N = 132), and the five patients who responded to IFN and then relapsed had highly elevated pretreatment IL-8 levels compared with sustained responders or nonresponders. In a follow-up study from the same group in which 47 patients with HCV were analyzed, it was shown that serum levels of TNF-a were highest in nonresponder patients [145], similar to the results of Polyak et al. In addition, different ELISA kits and IL-8 standards were used between the studies of Neuman and Polyak, so it is difficult to compare the actual levels of IL-8. The common theme in these studies, however, is that HCV infection produces elevations in inflammatory cytokines and chemokines such as TNF-a and IL-8. Clearly, additional prospective studies on patients with HCV from different countries need to be performed. The clinical studies described previously also underscore the need for additional characterization of the interaction of HCV with the IL-8 system. Although the precise mechanisms for the proviral properties of IL-8 await further study, one mechanism could involve IL-8’s anti-IFN effects. It is also possible that IL-8 promotes virus replication through IL-8 –initiated signal transduction events. In summary, the interaction of NS5A with the IL-8 system represents a novel mechanism of inhibition of the IFN system by the NS5A protein.

Stimulation of the interferon system by hepatitis C virus core It has been demonstrated that the HCV core protein activates the 2– 5 OAS gene [146] and PKR enzyme activity [147], two key IFN-induced genes that mediate the antiviral effects of IFN. This suggests that core interacts with the innate cellular antiviral response. The authors have found that the HCV core protein activates the IFN system on a more global level involving activation of the STAT/JAK pathway and IFN –stimulated gene promoter activity (Polyak et al, submitted). This effect of core is hypothesized to reflect activation of the innate immune system by the HCV core protein, which is likely to be the first viral protein that a cell encounters during acute infection. It is also possible that stimulation of the IFN system by the HCV core protein is required to balance the IFN inhibitory actions of other HCV proteins such as E2 [148] and NS5A [134,139] to provide fine tuning of HCV replication.

Hepatitis C virus assembly and egress As shown in Fig. 1, the final category of the HCV life cycle is virus maturation and release. This category is as yet an untapped research area, quite likely because so many researchers are trying to determine how the virus enters cells and replicates. The future will provide interesting developments in this important area of the HCV life cycle. This stage of the HCV life cycle likely

80

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

involves multiple interactions between HCV and host cell proteins, which may be an area for development of novel classes of antiviral molecules.

Summary In summary, HCV –cell interactions include those directly involved with the HCV life cycle such as virus attachment, entry, and replication. Included within this broad area of research are the interactions of HCV proteins with the IFN system, cytokine and chemokine pathways such as IL-8, and various other cellular proteins and pathways. The plethora of contradictory and sometimes confusing accessory HCV – host interactions defies precise predictions of their role in HCV biology. It is clear that these virus – cell interactions affect HCV replication, antiviral resistance, persistence, and pathogenesis. Because HCV – host interactions are initiated immediately on infection, they are operative during acute HCV infection, whereby HCV interacts with innate cellular antiviral and immune systems. The magnitude and duration of these HCV – host interactions therefore may influence the development of acquired immunity. Because HCV exists as a quasispecies in all infected individuals, heterogeneity in biological responses to HCV – host interactions is predicted, revealing opportunities for the development of various genotypic and phenotypic prognostic indicators. With the model systems in place, these hypotheses can be tested. The challenge for the future is to determine if there is a hierarchical importance to these interactions, to delineate how these virus –cell interactions affect the patient infected with HCV, and to determine whether any of these interactions represents a target for therapeutic intervention.

Acknowledgments The author would like to thank Stuart Ray, MD and Christoph Sarrazin, MD for providing templates for Figs. 1 and 3, and David Gretch, MD, PhD for insightful discussions.

References [1] Di Bisceglie AM. Liver transplantation for hepatitis C: the promise and the challenge. Hepatology 1995;22:660 – 2. [2] Ferrell LD, Wright TL, Roberts J, Ascher N, Lake J. Hepatitis C viral infection in liver transplant recipients. Hepatology 1992;16:865 – 76. [3] Bellentani S, Miglioli L, Masutti F, Saccoccio G, Tiribelli C. Epidemiology of hepatitis C virus infection in Italy: the slowly unraveling mystery. Microbes and Infection 2000;2:1757 – 63. [4] Alter MJ, Kruszon-Moran D, Nainan OV, McQuillan GM, Gao F, Moyer LA, et al. The prevalence of hepatitis C virus infection in the United States, 1988 through 1994. N Engl J Med 1999;341:556 – 62. [5] Wasley A, Alter MJ. Epidemiology of hepatitis C: geographic differences and temporal trends. Semin Liver Dis 2000;20:1 – 16.

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

81

[6] Thimme R, Oldach D, Chang KM, Steiger C, Ray SC, Chisari FV. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J Exp Med 2001;194: 1395 – 406. [7] Ray SC, Wang YM, Laeyendecker O, Ticehurst JR, Villano SA, Thomas DL. Acute hepatitis C virus structural gene sequences as predictors of persistent viremia: hypervariable region 1 as a decoy. J Virol 1999;73:2938 – 46. [8] Missale G, Bertoni R, Lamonaca V, Valli A, Massari M, Mori C, et al. Different clinical behaviors of acute hepatitis C infection are associated with different vigor of the antiviral cell – mediated immune response. J Clin Invest 1996;98:706 – 14. [9] Nelson DR, Marousis CG, Ohno T, Davis GL, Lau JYN. Intrahepatic hepatitis C virus – specific cytotoxic T-lymphocyte activity and response to interferon alfa therapy in chronic hepatitis C. Hepatology 1998;28:225 – 30. [10] Cooper S, Erickson AL, Adams EJ, Kansopon J, Weiner AJ, Chien DY, et al. Analysis of a successful immune response against hepatitis C virus. Immunity 1999;10:439 – 49. [11] Erickson AL, Kimura Y, Igarashi S, Eichelberger J, Houghton M, Sidney J, et al. The outcome of hepatitis C virus infection is predicted by escape mutations in epitopes targeted by cytotoxic T lymphocytes. Immunity 2001;15:883 – 95. [12] Oldach D, Gale M, Almiroudis D, Ray S, Thomas D, Thimme R, et al. Evolving HCV – NS5A sequence and protein kinase (PKR) – NS5A interaction phenotypes in acute HCV infection. Presented at the 8th International Symposium on Hepatitis C Virus and Related Viruses. Paris, Sept 2 – 5, 2001. [13] Lindsay KL. Therapy of hepatitis C: overview. Hepatology 1997;26:71S – 7S. [14] Davis GL, Esteban-Mur R, Rustgi V, Hoefs J, Gordon SC, Trepo C, et al. Interferon alfa-2b alone or in combination with ribavirin for the treatment of relapse of chronic hepatitis C. N Engl J Med 1998;339:1493 – 9. [15] McHutchison JG, Gordon SC, Schiff ER, Shiffman ML, Lee WM, Rustgi VK, et al. Interferon alfa-2b alone or in combination with ribavirin as initial treatment for chronic hepatitis C. N Engl J Med 1998;339:1485 – 92. [16] Poynard T, Marcellin P, Lee SS, Niederau C, Minuk GS, Ideo G, et al. Randomised trial of interferon alfa-2b plus ribavirin for 48 weeks or for 24 weeks versus interferon alfa-2b plus placebo for 48 weeks for treatment of chronic infection with hepatitis C virus. International Hepatitis Interventional Therapy Group. Lancet 1998;352:1426 – 32. [17] Lindsay KL, Trepo C, Heintges T, Shiffman ML, Gordon SC, Hoefs JC, et al. A randomized, double-blind trial comparing pegylated interferon alfa-2b to interferon alfa-2b as initial treatment for chronic hepatitis C. Hepatology 2001;34:395 – 403. [18] Reddy KR, Wright TL, Pockros PJ, Shiffman M, Everson G, Reindollar R, et al. Efficacy and safety of pegylated (40-kd) interferon alfa-2a compared with interferon alfa-2a in noncirrhotic patients with chronic hepatitis C. Hepatology 2001;33:433 – 8. [19] Glue P, Rouzier-Panis R, Raffanel C, Sabo R, Gupta SK, Salfi M, et al. A dose-ranging study of pegylated interferon alfa-2b and ribavirin in chronic hepatitis C. The Hepatitis C Intervention Therapy Group. Hepatology 2000;32:647 – 53. [20] Thavasu P, Propper D, McDonald A, Dobbs N, Ganesan T, Talbot D, et al. The protein kinase C inhibitor CGP41251 suppresses cytokine release and extracellular signal-regulated kinase 2 expression in cancer patients. Cancer Res 1999;59:3980 – 4. [21] Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med 2001;344: 1038 – 42. [22] O’Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994;79:315 – 28. [23] Dubuisson J. Folding, assembly, and subcellular localization of hepatitis C virus glycoproteins. Curr Top Microbiol Immunol 2000;242:135 – 48.

82

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

[24] Thomssen R, Bonk S, Propfe C, Heermann KH, Kochel HG, Uy A. Association of hepatitis C virus in human sera with beta-lipoprotein. Med Microbiol Immunol (Berl) 1992;181:293 – 300. [25] Thomssen R, Bonk S, Thiele A. Density heterogeneities of hepatitis C virus in human sera due to the binding of beta-lipoproteins and immunoglobulins. Med Microbiol Immunol (Berl) 1993; 182:329 – 34. [26] Agnello V, Abel G, Elfahal M, Knight GB, Zhang QX. Hepatitis C virus and other flaviviridae viruses enter cells via low density lipoprotein receptor. Proc Natl Acad Sci U S A 1999;96: 12766 – 71. [27] Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, et al. Binding of hepatitis C virus to CD81. Science 1998;282:938 – 41. [28] Allander T, Forns X, Emerson SU, Purcell RH, Bukh J. Hepatitis C virus envelope protein E2 binds to CD81 of tamarins. Virology 2000;277:358 – 67. [29] Meola A, Sbardellati A, Ercole BB, Cerretani M, Pezzanera M, Ceccacci A, et al. Binding of hepatitis C virus E2 glycoprotein to CD81 does not correlate with species permissiveness to infection. J Virol 2000;74:5933 – 8. [30] Zhao X, Tang ZY, Klumpp B, Wolff-Vorbeck G, Barth H, Levy S, et al. Primary hepatocytes of Tupaia belangeri as a potential model for hepatitis C virus infection. J Clin Invest 2002;109: 221 – 32. [31] Wellnitz S, Klumpp B, Barth H, Ito S, Depla E, Dubuisson J, et al. Binding of hepatitis C viruslike particles derived from infectious clone H77C to defined human cell lines. J Virol 2002;76: 1181 – 93. [32] Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol 2001;2:108 – 15. [33] Spear PG, Eisenberg RJ, Cohen GH. Three classes of cell surface receptors for alfaherpesvirus entry. Virology 2000;275:1 – 8. [34] Shukla D, Spear PG. Herpesviruses and heparan sulfate: an intimate relationship in aid of viral entry. J Clin Invest 2001;108:503 – 10. [35] Meyer K, Basu A, Ray R. Functional features of hepatitis C virus glycoproteins for pseudotype virus entry into mammalian cells. Virology 2000;276:214 – 26. [36] Young KC, Chang TT, Liou TC, Wu HL. Detection of hepatitis C virus RNA in peripheral blood mononuclear cells and in saliva. J Med Virol 1993;41:55 – 60. [37] Willems M, Peerlinck K, Moshage H, Deleu I, Van DEC, Vermylen J, et al. Hepatitis C virus RNAs in plasma and in peripheral blood mononuclear cells of hemophiliacs with chronic hepatitis C: evidence for viral replication in peripheral blood mononuclear cells. J Med Virol 1994;42:272 – 8. [38] Wang JT, Sheu JC, Lin JT, Wang TH, Chen DS. Detection of replicative form of hepatitis C virus RNA in peripheral blood mononuclear cells. J Infect Dis 1992;166:1167 – 9. [39] Qian C, Camps J, Maluenda MD, Civeira MP, Prieto J. Replication of hepatitis C virus in peripheral blood mononuclear cells. Effect of alfa-interferon therapy. J Hepatol 1992;16:380 – 3. [40] Bartolome J, Castillo I, Quiroga JA, Navas S, Carreno V. Detection of hepatitis C virus RNA in serum and peripheral blood mononuclear cells. J Hepatol 1993;17(Suppl 3):590 – 3. [41] Bouffard P, Hayashi PH, Acevedo R, Levy N, Zeldis JB. Hepatitis C virus is detected in a monocyte/macrophage subpopulation of peripheral blood mononuclear cells of infected patients. J Infect Dis 1992;166:1276 – 80. [42] Ferri C, Monti M, La CL, Longombardo G, Greco F, Pasero G, et al. Infection of peripheral blood mononuclear cells by hepatitis C virus in mixed cryoglobulinemia. Blood 1993;82: 3701 – 4. [43] Gabrielli A, Manzin A, Candela M, Caniglia ML, Paolucci S, Danieli MG, et al. Active hepatitis C virus infection in bone marrow and peripheral blood mononuclear cells from patients with mixed cryoglobulinaemia. Clin Exp Immunol 1994;97:87 – 93. [44] Bronowicki JP, Loriot MA, Thiers V, Grignon Y, Zignego AL, Brechot C. Hepatitis C virus persistence in human hematopoietic cells injected into SCID mice. Hepatology 1998; 28:211 – 8.

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

83

[45] Laskus T, Radkowski M, Wang LF, Vargas H, Rakela J. The presence of active hepatitis C virus replication in lymphoid tissue in patients coinfected with human immunodeficiency virus type 1. J Infect Dis 1998;178:1189 – 92. [46] Tseng CT, Klimpel GR. Binding of the hepatitis C virus envelope protein E2 to CD81 inhibits natural killer cell functions. J Exp Med 2002;195:43 – 9. [47] Crotta S, Stilla A, Wack A, D’Andrea A, Nuti S, D’Oro U, et al. Inhibition of natural killer cells through engagement of CD81 by the major hepatitis C virus envelope protein. J Exp Med 2002; 195:35 – 41. [48] Wack A, Soldaini E, Tseng C, Nuti S, Klimpel G, Abrignani S. Binding of the hepatitis C virus envelope protein E2 to CD81 provides a co-stimulatory signal for human T cells. Eur J Immunol 2001;31:166 – 75. [49] Jin M, Park J, Lee S, Park B, Shin J, Song KJ, et al. Hantaan virus enters cells by clathrindependent receptor – mediated endocytosis. Virology 2002;294:60 – 9. [50] Bishop NE. Examination of potential inhibitors of hepatitis A virus uncoating. Intervirology 1998;41:261 – 71. [51] Borrow P, Oldstone MB. Mechanism of lymphocytic choriomeningitis virus entry into cells. Virology 1994;198:1 – 9. [52] Helenius A, Marsh M, White J. Inhibition of Semliki forest virus penetration by lysosomotropic weak bases. J Gen Virol 1982;58(Pt 1):47 – 61. [53] Polyak SJ, Rawls WE, Harnish DG. Characterization of Pichinde virus infection of cells of the monocytic lineage. J Virol 1991;65:3575 – 82. [54] Randolph VB, Stollar V. Low pH-induced cell fusion in flavivirus-infected Aedes albopictus cell cultures. J Gen Virol 1990;71:1845 – 50. [55] Walewski JL, Keller TR, Stump DD, Branch AD. Evidence for a new hepatitis C virus antigen encoded in an overlapping reading frame. RNA 2001;7:710 – 21. [56] Purcell R. The hepatitis C virus. Overview. Hepatology 1997;26(3 Suppl 1):11S – 4S. [57] Ito T, Lai MM. An internal polypyrimidine tract binding protein binding site in the hepatitis C virus RNA attenuates translation, which is relieved by the 30-untranslated sequence. Virology 1999;254:288 – 96. [58] Ito T, Lai MM. Determination of the secondary structure of and cellular protein binding to the 30-untranslated region of the hepatitis C virus RNA genome. J Virol 1997;71: 8698 – 706. [59] Wood J, Frederickson RM, Fields S, Patel AH. Hepatitis C virus 30X region interacts with human ribosomal proteins. J Virol 2001;75:1348 – 58. [60] Spangberg K, Goobar-Larsson L, Wahren-Herlenius M, Schwartz S. The La protein from human liver cells interacts specifically with the U- rich region in the hepatitis C virus 30 untranslated region. J Hum Virol 1999;2:296 – 307. [61] Santolini E, Migliaccio G, La MN. Biosynthesis and biochemical properties of the hepatitis C virus core protein. J Virol 1994;68:3631 – 41. [62] Shirota Y, Luo H, Qin W, Kaneko S, Yamashita T, Kobayashi K, et al. Hepatitis C virus NS5A binds RNA-dependent RNA polymerase NS5B and modulates RdRP activity. J Biol Chem 2002;277(13):11149 – 55. [63] Hugle T, Fehrmann F, Bieck E, Kohara M, Krausslich HG, Rice CM, et al. The hepatitis C virus nonstructural protein 4B is an integral endoplasmic reticulum membrane protein. Virology 2001;284:70 – 81. [64] Moradpour D, Kary P, Rice CM, Blum HE. Continuous human cell lines inducibly expressing hepatitis C virus structural and nonstructural proteins. Hepatology 1998;28:192 – 201. [65] Selby MJ, Choo QL, Berger K, Kuo G, Glazer E, Eckart M, et al. Expression, identification and subcellular localization of the proteins encoded by the hepatitis C viral genome. J Gen Virol 1993;74:1103 – 13. [66] Wengler G, Nowak T, Castle E. Description of a procedure which allows isolation of viral nonstructural proteins from BHK vertebrate cells infected with the West Nile flavivirus in a state which allows their direct chemical characterization. Virology 1990; 177:795 – 801.

84

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

[67] Cauchi MR, Henchal EA, Wright PJ. The sensitivity of cell-associated dengue virus proteins to trypsin and the detection of trypsin-resistant fragments of the nonstructural glycoprotein NS1. Virology 1991;180:659 – 67. [68] Tu H, Gao L, Shi ST, Taylor DR, Yang T, Mircheff AK, et al. Hepatitis C virus RNA polymerase and NS5A complex with a SNARE-like protein. Virology 1999;263:30 – 41. [69] Shi ST, Polyak SJ, Hong T, Taylor DR, Gretch DR, Lai MMC. Hepatitis C Virus NS5A colocalizes with the core protein on lipid droplets and interacts with apolipoproteins. Virology 2002;292:198 – 210. [70] Ide Y, Zhang LW, Chen M, Inchauspe G, Bahl C, Sasaguri Y, et al. Characterization of the nuclear localization signal and subcellular distribution of hepatitis C virus nonstructural protein NS5A. Gene 1996;182:203 – 11. [71] Suzuki T, Matsuura Y, Harada T, Suzuki R, Saito I, Miyamura T. Molecular basis of subcellular localization of HCV core protein. Liver 1996;16:221 – 4. [72] Brass V, Bieck E, Montserret R, Wolk B, Hellings JA, Blum HE, et al. An amino-terminal amphipathic alfa -helix mediates membrane association of the hepatitis C virus nonstructural protein 5A. J Biol Chem 2002;277:8130 – 9. [73] Tanji Y, Kaneko T, Satoh S, Shimotohno K. Phosphorylation of hepatitis C virus – encoded nonstructural protein NS5A. J Virol 1995;69:3980 – 6. [74] Kaneko T, Tanji Y, Satoh S, Hijikata M, Asabe S, Kimura K, et al. Production of two phosphoproteins from the NS5A region of the hepatitis C viral genome. Biochem Biophys Res Commun 1994;205:320 – 6. [75] Asabe SI, Tanji Y, Satoh S, Kaneko T, Kimura K, Shimotohno K. The N-terminal region of hepatitis C virus-encoded NS5A is important for NS4A-dependent phosphorylation. J Virol 1997;71:790 – 6. [76] Reed KE, Xu J, Rice CM. Phosphorylation of the hepatitis C virus NS5A protein in vitro and in vivo: properties of the NS5A-associated kinase. J Virol 1997;71:7187 – 97. [77] Reed KE, Gorbalenya AE, Rice CM. The NS5A/NS5 proteins of viruses from three genera of the family Flaviviridae are phosphorylated by associated serine/threonine kinases. J Virol 1998;72:6199 – 206. [78] Hirota M, Satoh S, Asabe S, Kohara M, Tsukiyama K-K, Kato N, et al. Phosphorylation of nonstructural 5A protein of hepatitis C virus: HCV group-specific hyperphosphorylation. Virology 1999;257:130 – 7. [79] Koch JO, Bartenschlager R. Modulation of hepatitis C virus NS5A hyperphosphorylation by nonstructural proteins NS3, NS4A, and NS4B. J Virol 1999;73:7138 – 46. [80] Neddermann P, Clementi A, De FR. Hyperphosphorylation of the hepatitis C virus NS5A protein requires an active NS3 protease, NS4A, NS4B, and NS5A encoded on the same polyprotein. J Virol 1999;73:9984 – 91. [81] Reed KE, Rice CM. Identification of the major phosphorylation site of the hepatitis C virus H strain NS5A protein as serine 2321. J Biol Chem 1999;274:28011 – 8. [82] Katze MG, Kwieciszewski B, Goodlett DR, Blakely CM, Neddermann P, Tan SL, et al. Ser(2194) is a highly conserved major phosphorylation site of the hepatitis C virus nonstructural protein NS5A. Virology 2000;278:501 – 13. [83] Ide Y, Tanimoto A, Sasaguri Y, Padmanabhan R. Hepatitis C virus NS5A protein is phosphorylated in vitro by a stably bound protein kinase from HeLa cells and by cAMP-dependent protein kinase A-alfa catalytic subunit. Gene 1997;201:151 – 8. [84] Kim J, Lee D, Choe J. Hepatitis C virus NS5A protein is phosphorylated by casein kinase II. Biochem Biophys Res Commun 1999;257:777 – 81. [85] Nousbaum JB, Polyak SJ, Sullivan DG, Larson AM, Carithers Jr RL, Gretch DR. Prospective Characterization of full – length HCV NS5A quasispecies during induction and combination antiviral therapy. J Virol 2000;74:9028 – 38. [86] Blight KJ, Kolykhalov AA, Rice CM. Efficient initiation of HCV RNA replication in cell culture. Science 2000;290:1972 – 4. [87] Qin W, Yamashita T, Shirota Y, Lin Y, Wei W, Murakami S. Mutational analysis of the structure

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

[88] [89] [90]

[91]

[92]

[93]

[94]

[95] [96]

[97]

[98]

[99] [100]

[101] [102]

[103]

[104]

[105]

[106]

85

and functions of hepatitis C virus RNA – dependent RNA polymerase. Hepatology 2001;33: 728 – 37. Lohmann V, Korner F, Dobierzewska A, Bartenschlager R. Mutations in hepatitis C virus RNAs conferring cell culture adaptation. J Virol 2001;75:1437 – 49. Krieger N, Lohmann V, Bartenschlager R. Enhancement of hepatitis c virus RNA replication by cell culture- adaptive mutations. J Virol 2001;75:4614 – 24. Enomoto N, Sakuma I, Asahina Y, Kurosaki M, Murakami T, Yamamoto C, et al. Mutations in the nonstructural protein 5A and response to interferon in patients with chronic hepatitis C virus 1b infection. N Engl J Med 1996;334:77 – 81. Thomson M, Nascimbeni M, Gonzales S, Murthy KK, Rehermann B, Liang TJ. Emergence of a distinct pattern of viral mutations in chimpanzees infected with a homogeneous inoculum of hepatitis C virus. Gastroenterology 2001;121:1226 – 33. Tan SL, Nakao H, He YP, Vijaysri S, Neddermann P, Jacobs BL, et al. NS5A, a nonstructural protein of hepatitis C virus, binds growth factor receptor – bound protein 2 adaptor protein in a Src homology 3 domain/ligand-dependent manner and perturbs mitogenic signaling. Proc Natl Acad Sci U S A 1999;96:5533 – 8. He Y, Tan SL, Tareen SU, Vijaysri S, Langland JO, Jacobs BL, et al. Regulation of mRNA translation and cellular signaling by hepatitis C virus nonstructural protein NS5A. J Virol 2001; 75:5090 – 8. Gale M, Kwieciszewski B, Dossett M, Nakao H, Katze MG. Antiapoptotic and oncogenic potentials of hepatitis C virus are linked to interferon resistance by viral repression of the PKR protein kinase. J Virol 1999;73:6506 – 16. Ghosh AK, Majumder M, Steele R, Meyer K, Ray R, Ray RB. Hepatitis C virus NSSA protein protects against TNF-alfa – mediated apoptotic cell death. Virus Res 2000;67:173 – 8. Ezelle HJ, Balachandran S, Sicheri F, Polyak SJ, Barber GN. Analyzing the mechanisms of interferon-induced apoptosis using CrmA and hepatitis C virus NS5A. Virology 2001;281: 124 – 37. Park KJ, Choi SH, Lee SY, Hwang SB, Lai MM. Nonstructural 5A protein of hepatitis C virus modulates TNF-alfa – stimulated NF-kappa B activation. J Biol Chem 2002;277: 13122 – 8. Gong G, Waris G, Tanveer R, Siddiqui A. Human hepatitis C virus NS5A protein alters intracellular calcium levels, induces oxidative stress, and activates STAT-3 and NF-kappa B. Proc Natl Acad Sci U S A 2001;98:9599 – 604. Arima N, Kao CY, Licht T, Padmanabhan R, Sasaguri Y. Modulation of cell growth by the hepatitis C virus nonstructural protein NS5A. J Biol Chem 2001;276:12675 – 84. Satoh S, Hirota M, Noguchi T, Hijikata M, Handa H, Shimotohno K. Cleavage of hepatitis C virus nonstructural protein 5A by a caspase-like protease(s) in mammalian cells. Virology 2000;270:476 – 87. Moriya K, Yotsuyanagi H, Shintani Y, Fujie H, Ishibashi K, Matsuura Y, et al. Hepatitis C virus core protein induces hepatic steatosis in transgenic mice. J Gen Virol 1997;78:1527 – 31. Moriya K, Fujie H, Shintani Y, Yotsuyanagi H, Tsutsumi T, Ishibashi K, et al. The core protein of hepatitis C virus induces hepatocellular carcinoma in transgenic mice. Nat Med 1998;4: 1065 – 7. Lerat H, Honda M, Beard MR, Loesch K, Sun J, Yang Y, et al. Steatosis and liver cancer in transgenic mice expressing the structural and nonstructural proteins of hepatitis C virus. Gastroenterology 2002;122:352 – 65. Barba G, Harper F, Harada T, Kohara M, Goulinet S, Matsuura Y, et al. Hepatitis C virus core protein shows a cytoplasmic localization and associates to cellular lipid storage droplets. Proc Natl Acad Sci U S A 1997;94(4):1200 – 5. Perlemuter G, Sabile A, Letteron P, Vona G, Topilco A, Chretien Y, et al. Hepatitis C virus core protein inhibits microsomal triglyceride transfer protein activity and very low density lipoprotein secretion: a model of viral-related steatosis. FASEB J 2002;16:185 – 94. Okuda M, Li K, Beard MR, Showalter LA, Scholle F, Lemon SM, et al. Mitochondrial injury,

86

[107]

[108]

[109] [110]

[111]

[112]

[113]

[114]

[115]

[116] [117] [118] [119] [120]

[121]

[122]

[123]

[124] [125]

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88 oxidative stress, and antioxidant gene expression are induced by hepatitis C virus core protein. Gastroenterology 2002;122:366 – 75. Matsumoto M, Hsieh TY, Zhu NL, VanArsdale T, Hwang SB, Jeng KS, et al. Hepatitis C virus core protein interacts with the cytoplasmic tail of lymphotoxin-beta receptor. J Virol 1997;71: 1301 – 9. Zhu NL, Khoshnan A, Schneider R, Matsumoto M, Dennert G, Ware C, et al. Hepatitis C virus core protein binds to the cytoplasmic domain of tumor necrosis factor (TNF) receptor 1 and enhances TNF-induced apoptosis. J Virol 1998;72:3691 – 7. Ray RB, Meyer K, Ray R. Suppression of apoptotic cell death by hepatitis C virus core protein. Virology 1996;226:176 – 82. Ray RB, Meyer K, Steele R, Shrivastava A, Aggarwal BB, Ray R. Inhibition of tumor necrosis factor (TNF-alfa)-mediated apoptosis by hepatitis C virus core protein. J Biol Chem 1998;273: 2256 – 9. Marusawa H, Hijikata M, Chiba T, Shimotohno K. Hepatitis C virus core protein inhibits Fasand tumor necrosis factor alfa-mediated apoptosis via NF-kappa B activation. J Virol 1999;73: 4713 – 20. Giambartolomei S, Covone F, Levrero M, Balsano C. Sustained activation of the Raf/MEK/Erk pathway in response to EGF in stable cell lines expressing the hepatitis C Virus (HCV) core protein. Oncogene 2001;20:2606 – 10. Fukuda K, Tsuchihara K, Hijikata M, Nishiguchi S, Kuroki T, Shimotohno K. Hepatitis C virus core protein enhances the activation of the transcription factor, Elk1, in response to mitogenic stimuli. Hepatology 2001;33:159 – 65. Hayashi J, Aoki H, Kajino K, Moriyama M, Arakawa Y, Hino O. Hepatitis C virus core protein activates the MAPK/ERK cascade synergistically with tumor promoter TPA, but not with epidermal growth factor or transforming growth factor alfa. Hepatology 2000;32:958 – 61. Tsuchihara K, Hijikata M, Fukuda K, Kuroki T, Yamamoto N, Shimotohno K. Hepatitis C virus core protein regulates cell growth and signal transduction pathway transmitting growth stimuli. Virology 1999;258:100 – 7. Lai MMC. Hepatitis C virus proteins: direct link to hepatic oxidative stress, steatosis, carcinogenesis and more. Gastroenterology 2002;122:568 – 71. Lai MMC, Ware CF. Hepatitis C virus core protein: possible roles in viral pathogenesis. Curr Top Microbiol Immunol 2000;242:117 – 34. Goh PY, Tan YJ, Lim SP, Lim SG, Tan YH. The hepatitis C virus core protein interacts with NS5A and activates its caspase – mediated proteolytic cleavage. Virology 2001;290:224 – 36. Darnell JE, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994;264:1415 – 21. Khabar KSA, Dhalla M, Siddiqui Y, Zhou A. Effect of the deficiency of the double-stranded RNA – dependent protein kinase, PKR, on antiviral resistance in presence or absence of ribonuclease L: HSV-1 replication is particularly sensitive to deficiency of the major IFNmediated enzymes. J Interferon Cytokine Res 2000;20(7):653 – 9. Zhou A, Paranjape J, Brown TL, Nie H, Naik S, Dong B, et al. Interferon action and apoptosis are defective in mice devoid of 20,50- oligoadenylate-dependent RNase L. EMBO J 1997;16: 6355 – 63. Zhou A, Paranjape JM, Der SD, Williams BR, Silverman RH. Interferon action in triply deficient mice reveals the existence of alternative antiviral pathways. Virology 1999;258: 435 – 40. Silverman RH. 2 – 5A dependent RNAse L: a regulated endoribonuclease in the interferon system. In: D’Alessio G, Riordan JF, editors. Ribonucleases: structure and function. New York: Academic Press; 1997. p. 515 – 51. Silverman RH, Williams BR. Translational control perks up. Nature 1999;397:208 – 11. Meurs E, Chong K, Galabru J, Thomas NS, Kerr IM, Williams BRG, et al. Molecular cloning and characterization of the human double-stranded RNA – activated protein kinase induced by interferon. Cell 1990;62:379 – 90.

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

87

[126] Meurs EF, Watanabe Y, Kadereit S, Barber GN, Katze MG, Chong K, et al. Constitutive expression of human double-stranded RNA – activated p68 kinase in murine cells mediates phosphorylation of eukaryotic initiation factor 2 and partial resistance to encephalomyocarditis virus growth. J Virol 1992;66:5804 – 14. [127] Pavlovic J, Zurcher T, Haller O, Staeheli P. Resistance to influenza virus and vesicular stomatitis virus conferred by expression of human MxA protein. J Virol 1990;64:3370 – 5. [128] Heim MH, Moradpour D, Blum HE. Expression of hepatitis C virus proteins inhibits signal transduction through the Jak-STAT pathway. J Virol 1999;73:8469 – 75. [129] Polyak SJ, Paschal D, McArdle S, Gale M, Moradpour D, Gretch DR. Characterization of the effects of hepatitis C virus nonstructural 5A protein expression in human cell lines and on interferon-sensitive virus replication. Hepatology 1999;29:1262 – 71. [130] Francois C, Duverlie G, Rebouillat D, Khorsi H, Castelain S, Blum HE, et al. Expression of hepatitis C virus proteins interferes with the antiviral action of interferon independently of PKR-mediated control of protein synthesis. J Virol 2000;74:5587 – 96. [131] Paterson M, Laxton CD, Thomas HC, Ackrill AM, Foster GR. Hepatitis C virus NS5A protein inhibits interferon antiviral activity, but the effects do not correlate with clinical response. Gastroenterology 1999;117:1187 – 97. [132] Song J, Fujii M, Wang F, Itoh M, Hotta H. The NS5A protein of hepatitis C virus partially inhibits the antiviral activity of interferon. J Gen Virol 1999;80:879 – 86. [133] Aizaki H, Saito S, Ogino T, Miyajima N, Harada T, Matsuura Y, et al. Suppression of interferon – induced antiviral activity in cells expressing hepatitis C virus proteins. J Interferon Cytokine Res 2000;20:1111 – 20. [134] Gale MJ, Korth MJ, Tang NM, Tan SL, Hopkins DA, Dever TE, et al. Evidence that hepatitis C virus resistance to interferon is mediated through repression of the PKR protein kinase by the nonstructural 5A protein. Virology 1997;230:217 – 27. [135] Gale M, Blakely CM, Kwieciszewski B, Tan SL, Dossett M, Tang NM, et al. Control of PKR protein kinase by hepatitis C virus nonstructural 5A protein: molecular mechanisms of kinase regulation. Mol Cell Biol 1998;18:5208 – 18. [136] Enomoto N, Sakuma I, Asahina Y, Kurosaki M, Murakami T, Yamamoto C, et al. Comparison of full-length sequences of interferon-sensitive and resistant hepatitis C virus 1b. Sensitivity to interferon is conferred by amino acid substitutions in the NS5A gene. J Clin Invest 1995;96: 224 – 30. [137] Polyak SJ, Gerotto M. The molecular basis for responsiveness to anti-viral therapy in hepatitis C. Forum (Genova) 2000;10:46 – 58. [138] Witherell GW, Beineke P. Statistical analysis of combined substitutions in nonstructural 5A region of hepatitis C virus and interferon response. J Med Virol 2001;63:8 – 16. [139] Polyak SJ, Khabar KS, Paschal DM, Ezelle HJ, Duverlie G, Barber GN, et al. Hepatitis C virus nonstructural 5A protein induces interleukin-8, leading to partial inhibition of the interferon – induced antiviral response. J Virol 2001;75:6095 – 106. [140] Girard S, Shalhoub P, Lescure P, Sabile A, Misek DE, Hanash S, et al. An altered cellular response to interferon and up-regulation of interleukin-8 induced by hepatitis C viral protein NS5A uncovered by microarray analysis. Virology 2002;295(2):272 – 83. [141] Khabar KS, Al-Zoghaibi F, Murayama T, Matsushima K, Mukaida N, Siddiqui Y, et al. Interleukin-8 selectively enhances cytopathic effect (CPE) induced by positive – strand RNA viruses in the human WISH cell line. Biochem Biophys Res Commun 1997;235:774 – 8. [142] Polyak SJ, Khabar KS, Rezeiq M, Gretch DR. Elevated levels of interleukin-8 in serum are associated with hepatitis C virus infection and resistance to interferon therapy. J Virol 2001;75: 6209 – 11. [143] Mukaida N. Interleukin-8: an expanding universe beyond neutrophil chemotaxis and activation. Int J Hematol 2000;72:391 – 8. [144] Neuman MG, Benhamou JP, Martinot M, Boyer N, Shear NH, Malkiewicz I, et al. Predictors of sustained response to alfa interferon therapy in chronic hepatitis C. Clin Biochem 1999;32: 537 – 45.

88

S.J. Polyak / Clin Liver Dis 7 (2003) 67–88

[145] Neuman MG, Benhamou JP, Malkiewicz IM, Akremi R, Shear NH, Asselah T, et al. Cytokines as predictors for sustained response and as markers for immunomodulation in patients with chronic hepatitis C. Clin Biochem 2001;34:173 – 82. [146] Naganuma A, Nozaki A, Tanaka T, Sugiyama K, Takagi H, Mori M, et al. Activation of the interferon-inducible 20-50-oligoadenylate synthetase gene by hepatitis C virus core protein. J Virol 2000;74:8744 – 50. [147] Delhem N, Sabile A, Gajardo R, Podevin P, Abadie A, Blaton MA, et al. Activation of the interferon – inducible protein kinase PKR by Hepatocellular carcinoma derived-Hepatitis C virus core protein. Oncogene 2001;20:5836 – 45. [148] Taylor DR, Shi ST, Romano PR, Barber GN, Lai MMC. Inhibition of the interferon – inducible protein kinase PKR by HCV E2 protein. Science 1999;285:107 – 10. [149] Duverlie G, Khorsi H, Castelain S, Jaillon O, Izopet J, Lunel F, et al. Sequence analysis of the NS5A protein of European hepatitis C virus 1b isolates and relation to interferon sensitivity. J Gen Virol 1998;79:1373 – 81.