School of Biochemistry & Microbiology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
Correspondence
Mark Harris
mharris{at}bmb.leeds.ac.uk
![]() |
ABSTRACT |
---|
![]() ![]() ![]() |
---|
Published online ahead of print on 8 June 2004 as DOI 10.1099/vir.0.80204-0.
Present address: MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK.
Hepatitis C virus
Hepatitis C virus (HCV) is the only member of the genus Hepacivirus, within the family Flaviviridae. Viral isolates are further classified into six genotypes (16) and 30 subtypes, the most intensively studied of which are genotypes 1a and 1b. It is estimated that 170 million individuals are infected with this virus (Anonymous, 1999), and in 80 % of cases the virus establishes a chronic infection, resulting in fibrosis, cirrhosis and, increasingly, hepatocellular carcinoma (HCC). Current therapy, which consists of a combination of pegylated alpha interferon (IFN-
) and ribavirin, gives a response rate of between 48 % (genotypes 1, 4, 5 and 6) and 88 % (genotypes 2 and 3) (Poynard et al., 2003
). The genome is a positive-sense 9·6 kb RNA molecule, comprising 5' and 3' untranslated regions (UTRs) flanking a single open reading frame encoding a polyprotein of
3000 aa (Fig. 1
). A second translation product, the F protein, is produced by ribosomal frameshift (Xu et al., 2003
). Little is known about the function of this protein, although it has been shown to localize to the endoplasmic reticulum (ER). The 5' UTR contains an internal ribosome entry site (IRES), allowing cap-independent initiation of translation. The polyprotein is cleaved into 10 polypeptides by cellular and viral proteinases. At the N terminus are three structural proteins Core and the glycoproteins E1 and E2. Following E2 is p7, a protein that oligomerizes to form cation channels (Griffin et al., 2003
) and has been speculated to be incorporated into virus particles. The C-terminal two-thirds of the polyprotein comprise the six non-structural proteins NS2, NS3, NS4A, NS4B, NS5A and NS5B. Two of these are functionally well characterized: NS3 complexed with NS4A is a proteinase that cleaves between the non-structural proteins (it also has helicase activity); NS5B is the viral RNA-dependent RNA polymerase. The others presumably play as yet undefined roles in virus replication; however, an increasing body of literature points to a role for these proteins in perturbing cell signalling and mediating immune evasion. In this regard, NS5A is of great interest, and here we will review our current knowledge of this protein.
|
N-terminal amphipathic helix
The N-terminal 30 aa of NS5A were predicted to form a highly conserved amphipathic alpha helix (Brass et al., 2002) (Fig. 2a
), and were shown to be both necessary and sufficient to mediate association of NS5A (or GFP) with the ER membrane. By a combination of in vitro biochemical assays and comparison with other, similarly membrane-anchored, proteins, it was postulated that the N terminus of NS5A lies parallel to the membrane such that the hydrophobic face of the helix was buried within the bilayer; this interaction appeared to confer upon NS5A the properties of an integral membrane protein. Subsequently, this structure was shown to be essential for HCV RNA replication.
|
The p58 form of NS5A is hyperphosphorylated within a serine-rich region in the centre of the protein (Fig. 2b), although there is little consensus as to the identity of the phosphorylated serine residues. Katze et al. (2000)
demonstrated that Ser2194 (polyprotein numbering, residue 221 of NS5A) was the major phosphorylation site in an HCV1b isolate of NS5A expressed in insect or COS cells. In contrast, Tanji et al. (1995)
showed that Ser2197, Ser2201 and Ser2204 were required for hyperphosphorylation of HCV1b NS5A, although they did not conclusively show that these residues were phosphorylation sites. This discrepancy may be explained by the fact that, as discussed below, the presence of the other non-structural proteins modulates hyperphosphorylation; whereas Tanji and co-workers utilized a construct expressing NS2 to NS5A, Katze and co-workers expressed NS5A alone. Reed & Rice (1999)
showed that Ser2321 (residue 348 in NS5A), although poorly conserved throughout different genotypes of HCV, was the major phosphorylation site in an HCV1a NS5A isolate. This residue is flanked by proline-rich sequences [later identified as binding sites for cellular Src-Homology 3 (SH3) domains; see Fig. 2d
], leading to the speculation that, as for basal phosphorylation, a proline-directed kinase (e.g. the CMGC family) might be involved. However, proline-rich sequences show a preference for Mg2+ over Mn2+, whereas in vitro phosphorylation of NS5A was most efficient in Mn2+-containing buffers (Reed et al., 1997
). Thus, the precise identity of the kinase(s) responsible for NS5A phosphorylation remains to be elucidated.
Hyperphosphorylation is dependent on the presence of other non-structural proteins. NS4A (expressed in trans) was required for the production of the p58 NS5A form of an HCV1b isolate (HCV-J) (Kaneko et al., 1994; Asabe et al., 1997
). Furthermore, residues 162166 of NS5A were required for NS4A-dependent phosphorylation. Interestingly, deletion of the N-terminal 162 residues of NS5A resulted in NS4A-independent hyperphosphorylation. The reason for this observation is unclear. Perhaps binding of NS4A might induce a conformational change in NS5A, facilitating hyperphosphorylation? Deletion of the N terminus of NS5A could thus alter the overall conformation of the protein allowing access by the kinase(s). Liu et al. (1999)
, using another HCV1b isolate (HCV-BK), demonstrated a requirement for NS2 generated by autoproteolysis from the NS23 precursor. Two groups (Neddermann et al., 1999
; Koch & Bartenschlager, 1999
) showed that hyperphosphorylation of an HCV1b (Con1) NS5A required its expression as part of a continuous polyprotein with NS3, NS4A and NS4B, and required the protease activity of NS3. These studies were performed following transient transfection: it will be important to verify such data using the subgenomic replicon system in which NS5A will be involved in replication of the viral RNA. Interestingly, in replicon cells, the p58 form of NS5A was less stable than p56 (half-life
7 h compared to 16 h) (Pietschmann et al., 2001
), suggesting that the two forms of the protein play distinct roles in the virus replication cycle.
Other structural features of NS5A
NS5A contains three consensus ProXaaXaaPro motifs: these are present in many signalling proteins, form extended alpha helices and bind to SH3 domains. Two closely spaced motifs near the C terminus (residues 343356) (Tan et al., 1999; Macdonald et al., 2004
) (Fig. 2a and d
) are predicted to be surface-exposed, consistent with the ability of NS5A to bind tightly to SH3 domains. In contrast, an N-terminal motif (residues 2632) is dispensable for SH3 binding and is predicted to lie within the previously discussed amphipathic helix and is thus unlikely to be available for binding to other proteins.
The second C-terminal motif overlaps with a putative nuclear localization signal (Ide et al., 1996). The physiological significance of this signal is unclear, however, as native NS5A is entirely cytoplasmic, being associated with the ER membrane via the N-terminal amphipathic helix. Consistent with this, Satoh et al. (2000)
showed that N-terminal truncations of NS5A localized to the nucleus; they also reported that NS5A could be cleaved by a caspase-like protease and that one of the cleavage products lacked both N and C termini and was located within the nucleus. However, these observations were made in cells expressing NS5A alone and have not been documented in a more physiologically relevant environment such as replicon cells; indeed in the latter NS5A is exclusively cytoplasmic (Mottola et al., 2002
; Shi et al., 2003
).
Functions of NS5A in virus replication
Information derived mainly from the subgenomic replicon system, originally described by Lohmann et al. (1999), suggests that NS5A is involved in genomic RNA replication. Replicons consist of bicistronic RNA transcripts in which the HCV IRES drives translation of neomycin phosphotransferase; a second IRES [from encephalomyocarditis virus (EMCV)] drives translation of NS3NS5B (Fig. 1
). In vitro transcripts are transfected into the human hepatoma cell line Huh7 and stable cell clones selected with G418. These cells then harbour a cytoplasmic, autonomously replicating RNA and express large amounts of NS3NS5B. Although not without its drawbacks, for example the rapid mutation rate due to the error-prone nature of NS5B and the limited range of cells that support replicon replication, the system has allowed the investigation of the biochemistry of HCV RNA replication and points to a key role for NS5A in this process.
NS5A is part of a multi-protein, membrane-bound replication complex
By analogy to other RNA viruses, it has been hypothesized that the HCV non-structural proteins form a complex located on cytoplasmic membranes that catalyses replication of the viral genome. The replicon system has confirmed this, and shown that this complex contains NS5A. Together with the other non-structural proteins, NS5A co-localized with viral RNA in a cytoplasmic membrane structure termed the membranous web (Egger et al., 2002; Mottola et al., 2002
), the generation of which required the NS4B protein. Replicon cell membrane fractions isolated by differential centrifugation contained both p56 and p58 forms of NS5A and were competent for synthesis of HCV RNA in vitro (Hardy et al., 2003
). An RNAprotein complex purified from replicon cells by a two-step affinity procedure with biotin/digoxigenin-labelled oligonucleotides (Waris et al., 2004
) contained all of the non-structural proteins, including NS5A, although hyperphosphorylated NS5A appeared not to be present in this complex. Two studies from Michael Lai's group (Shi et al., 2003
; Gao et al., 2004
) analysed the location of HCV RNA replication by membrane flotation and showed that almost all NS5A (although not the hyperphosphorylated form), and all the viral RNA, co-localized with caveolin-2 in an NP-40-resistant fashion. They proposed, therefore, that viral RNA replication occurred within a detergent-resistant fraction of cellular membranes most likely representing lipid rafts. Further evidence for a critical role of NS5A came from experiments in which the amphipathic membrane-targeting helix was mutated in the context of the replicon (Elazar et al., 2003
). Introduction of three helix-disrupting mutations (Fig. 2a
) completely abrogated the ability of the replicon to establish G418-resistant colonies, implying that NS5A membrane association is an indispensable event during HCV RNA replication.
Evidence for direct interactions between NS5A and other non-structural proteins
The formation of a multi-protein complex requires specific interactions between the individual components. In this regard, NS5A was shown to bind to NS5B, both in vitro and in transiently transfected cells. This binding required residues 105162 and 277334 of NS5A (Shirota et al., 2002) and four discontinuous regions of NS5B (Qin et al., 2001
). However, somewhat paradoxically, when present at a molar ratio of 0·1 or below, NS5A modestly stimulated NS5B polymerase activity, but at higher ratios NS5A inhibited NS5B activity. It is unclear why NS5A should inhibit NS5B; however, it may be that the results obtained in vitro using purified NS5A and NS5B fusion proteins do not accurately reflect the interaction between these two proteins in vivo, when other viral and cellular factors may be involved. It was shown that deletions within NS5A that abrogated the interaction with NS5B also rendered the subgenomic replicon non-functional (Shimakami et al., 2004
), whereas a deletion that had no effect on NS5B binding was replication-competent. The authors conclude that NS5ANS5B interactions are necessary for HCV RNA replication; however, an alternative interpretation that these deletions perturb the structure of NS5A, thereby abrogating another function associated with genomic replication should also be considered.
Dimitrova et al. (2003), using a combination of standard biochemical and genetic proteinprotein interaction assays, demonstrated that NS5A was able to interact independently with all the non-structural proteins, including itself. Taken together with the observation that NS5A hyperphosphorylation required the presence of the other non-structural proteins, it is clear that NS5A does indeed participate in a multi-protein replication complex. However, the molecular details of the structure and function of this complex remain major challenges for the future.
Culture adaptation of the replicon provides further evidence of a role for NS5A in RNA replication
With the development of the replicon system a key question was asked: why was the relative efficiency of establishment of G418-resistant colonies so low, when the level of RNA replication in selected cells was high? The answer lay in the ability of the replicon to undergo mutation, leading to increases in the RNA replication efficiency (Lohmann et al., 2001; Krieger et al., 2001
; Blight et al., 2000
). Thus, although transfection of the replicon only gave rise to G418-resistant colonies at a frequency of 2040 per µg of RNA, RNA extracted from replicon cells and re-transfected into naïve Huh7 cells gave a much higher efficiency of colony formation (Lohmann et al., 2001
). After cDNA cloning of RNA derived from replicon cells, sequencing revealed changes throughout the non-structural region (although interestingly not in the UTRs). These cloned replicons exhibited a much higher efficiency of colony formation compared with the original replicon RNA. When engineered back into the parental replicon, single amino-acid substitutions dramatically enhanced the efficiency of colony formation the most effective was an Arg2884Gly substitution in NS5B (500-fold increase). In NS5A, Glu2163Gly enhanced colony formation 70-fold, whereas Lys2330Glu had no effect. Interestingly, combination of Glu2163Gly (NS5A) with Arg2884Gly (NS5B) rendered the replicon non-viable, whereas Lys2330Glu (NS5A) with Arg2884Gly (NS5B) retained the 400-fold enhancement of colony formation: the explanation for this remains obscure as these residues map outwith the NS5ANS5B interaction sites (Qin et al., 2001
; Shirota et al., 2002
).
Krieger et al. (2001) confirmed that these culture-adaptive mutations increased the efficiency of colony formation by enhancing RNA replication, and they identified a further single amino-acid substitution in NS5A (Ser2197Pro) that enhanced colony formation 1000-fold. Blight et al. (2000)
identified a cluster of mutations in NS5A between residues 2177 and 2204, as well as a deletion of 47 aa (22072254), that each stimulated colony formation. The most effective of these (20 000-fold stimulation) was Ser2204Ile. The most interesting observation from these studies is that both Ser2197 and Ser2204 were shown previously to be required for hyperphosphorylation (Fig. 2b
); indeed, reduced levels of the p58 form of NS5A were observed in cells harbouring replicons with mutations at these residues (Blight et al., 2000
). The implication of these results is that NS5A hyperphosphorylation is dispensable for viral RNA replication and, together with the observations that hyperphosphorylated NS5A is not a component of the membrane-bound RNA replication complex (Shi et al., 2003
; Waris et al., 2004
), these data suggest that this form of NS5A may play a distinct role in the virus life cycle, perhaps during particle assembly. This suggestion is further supported by the observation that, after introduction into a chimpanzee of an RNA transcript derived from an infectious clone containing the Ser2197Pro mutation, within 7 days the virus reverted to the parental sequence at that position (Bukh et al., 2002
). A further possibility might be that hyperphosphorylated NS5A functions during translation perhaps it acts directly to modulate IRES-mediated translation, for example by interacting with ribosomal components or even the IRES itself? In this regard, NS5A has been shown to stimulate IRES-mediated translation (He et al., 2003
) and interact with RNA-binding proteins such as the La antigen (Houshmand & Bergqvist, 2003
) that itself stimulates the IRES. Furthermore, IRES activity is regulated by the cell cycle (Honda et al., 2000
), and the CMGC family of kinases that have been reported to hyperphosphorylate NS5A include the cyclin-dependent kinases whose activity is very tightly linked to the cell cycle. It is tempting to speculate that NS5A hyperphosphorylation might also be linked to the cell cycle; however, this hypothesis remains to be tested.
NS5A and cellular signalling pathways
The interactions between NS5A and cellular signalling pathways have been the subject of intense investigation, a trend that started with the observation that HCV resistance to IFN could be explained by the ability of NS5A to bind to and inhibit the dsRNA-activated, IFN-induced kinase PKR (Gale et al., 1997). Since then NS5A has been shown to bind to a range of cellular signalling molecules (Table 1
), somewhat reminiscent of the Nef protein from human immunodeficiency virus type 1 (HIV-1) (Harris, 1996
; Renkema & Saksela, 2000
).
|
NS5A and the IFN response the role of PKR
The links between NS5A and the IFN response have been reviewed recently (Tan & Katze, 2001) so this section will be brief and highlight more recent observations. NS5A was first linked to IFN-responsiveness in patients by molecular epidemiological studies (Enomoto et al., 1995
, 1996
) that identified a stretch of 40 aa in the centre of NS5A conserved in IFN-resistant HCV isolates (Fig. 2c
). HCV variants with mutations within this region appeared to be more sensitive to therapy, suggesting that NS5A played a role in conferring IFN resistance, thus this region was termed the IFN sensitivity determining region (ISDR). Other groups confirmed these data, showing that expression of NS5A from IFN-resistant HCV1b isolates rendered cells partially resistant to the effects of IFN and allowed growth of either vesicular stomatitis virus (VSV) (Noguchi et al., 2001
) or EMCV (Polyak et al., 1999
). NS5A isolates from genotype 1a or 2a IFN-responders or 1b ISDR-deleted mutants did not inhibit IFN activity in these assays. However, two studies (Paterson et al., 1999
; Aizaki et al., 2000
) failed to find a correlation between ISDR sequence and ability to inhibit IFN activity, and subsequently it has been suggested that sequences towards the C terminus of NS5A, outwith the ISDR, are required to inhibit IFN activity (Nousbaum et al., 2000
).
Biochemical data have shown that NS5A bound to and inactivated PKR (Gale et al., 1997), an IFN-induced gene product that is activated by binding to dsRNA, commonly produced during RNA virus genome replication. PKR phosphorylates the translation initiation factor eIF-2
, shutting down protein translation (Williams, 2001
). The interaction of NS5A with PKR required the ISDR and an additional 26 residues C-terminal to the ISDR (Gale et al., 1998
). Within PKR, the binding site was identified as the dimerization domain. Binding of NS5A to PKR resulted in inhibition of both PKR autophosphorylation and phosphorylation of an exogenous substrate. However, more recently, conflicting results have been presented. Podevin et al. (2001)
and Ezelle et al. (2001)
were unable to observe any effect of NS5A (from either IFN-responders or non-responders) on the activity of PKR in either Huh7 or HeLa cells, although Podevin et al. (2001)
did show that all isolates of NS5A inhibited IFN activity (using both EMCV and VSV). Furthermore, Podevin et al. (2001)
were also unable to detect an NS5APKR interaction either by co-immunoprecipitation or by co-immunofluorescence.
PKR-independent effects of NS5A on the IFN response
As a result of the controversy surrounding the link between NS5A and PKR, a number of groups have sought evidence for effects of NS5A on IFN activity that are independent of PKR. In this regard, it has been reported that NS5A is able to upregulate production of interleukin (IL) 8. This cytokine can attenuate the antiviral properties of IFN (Khabar et al., 1997), suggesting that endogenously produced IL8 may facilitate virus infection. Polyak et al. (2001a)
demonstrated that NS5A upregulated the IL8 promoter, thereby increasing the levels of IL8, an observation confirmed by microarray analysis (Girard et al., 2002
). Importantly, this observation has been verified in vivo as HCV-infected patients showed higher levels of serum IL8 compared with controls; in addition, levels were higher in IFN non-responders compared to responders, suggesting a further mechanism for inhibition of this antiviral pathway (Polyak et al., 2001b
).
Many recent studies have analysed the effects of IFN on the subgenomic replicon (space constraints preclude a detailed review of this literature here). However, it is important to point out that, although IFN- has been shown to inhibit replicon function at the level of both RNA replication (Frese et al., 2001
; Guo et al., 2001
) and IRES-mediated translation (Wang et al., 2003
), this inhibition appeared not to depend on whether the replicons were derived from IFN-resistant or -sensitive HCV isolates. Wang et al. (2003)
suggested that the interaction of NS5A with PKR was associated with a partial rescue of replicon replication. Clearly, further analysis of the effect of NS5A on the IFN response in the context of the replicon is called for. However, such analysis is complicated by the observations that some studies have shown activation of the dsRNA response by replicons (Pflugheber et al., 2002
; McCormick et al., 2004
). In addition, there is evidence that Huh7 cells are defective in some aspects of the dsRNA response (Lanford et al., 2003
).
Expression of the whole HCV genome in an osteosarcoma cell line resulted in an inhibition of IFN signalling through the Jak/Stat pathway (Heim et al., 1999). This inhibition resulted in reductions in both the expression levels and DNA-binding activity of the IFN-
-stimulated transcription factor ISGF3, consistent with reduced activity of the ISGF3 components Stat1 and IRF9 (p48). The mechanisms and HCV proteins involved in this inhibition were not clarified, although Stat1 tyrosine phosphorylation and nuclear translocation were not impaired. This study was supported by in vivo data (Blindenbacher et al., 2003
), showing reduced ISGF3 DNA-binding activity and enhanced susceptibility to infection with lymphochoriomeningitis virus in transgenic mice expressing the complete HCV genome. Although not analysed in these studies, there is growing evidence that serine phosphorylation of Stats plays a pivotal role in the regulation of IFN signalling (Shuai, 2003
). MAPK family members have been shown to serine phosphorylate Stat1 and this has been linked with maximal transcriptional activation (Song et al., 2002
). As discussed below, NS5A inhibits the extracellular signal-regulated kinase (ERK) MAPK, possibly via an interaction with the adaptor protein Grb2 (Tan et al., 1999
; Macdonald et al., 2003
). By inhibiting ERK, NS5A may reduce levels of Stat1 serine phosphorylation and in doing so impair the cellular response to IFN-
(Tan et al., 1999
); however, this hypothesis is not supported by the observation that NS5A from IFN-resistant and IFN-sensitive genotypes of HCV (1a, 1b and 2a) was able to bind Grb2 (Macdonald et al., 2004
).
NS5A, mitogenic signalling pathways and cell growth
To establish a chronic infection, viruses must modulate host cell mitogenic signalling pathways to regulate cell growth and activation. Mitogenic signalling is mediated, for the most part, by the MAPK, a family of serine/threonine kinases that mediate the cellular response to a variety of extracellular stimuli. Three main pathways have been characterized (Chang & Karin, 2001): these are the previously mentioned ERK pathway, the stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) pathway, and the less well understood p38MAPK pathway (Fig. 3
). All of these are based around the principle of a triple-kinase signalling module consisting of a MAPK that is itself phosphorylated on serine and threonine residues by a MAPK kinase (MAPKK), which in turn is activated by a variety of upstream MAPK kinase kinases (MAPKKK) (Chang & Karin, 2001
).
|
It is interesting to note that NS5A inhibits ERK signalling, whereas both hepatitis B virus X protein (Klein & Schneider, 1997) and hepatitis E virus ORF3 (Korkaya et al., 2001
) activate this signalling cascade. Why there should be a disparity between these three hepatotrophic viruses is not understood, although the need for differential activation of ERK may reflect differences in the life cycle of each virus. Indeed, data suggest that activation of ERK by IL1 may inhibit HCV subgenomic replicon replication (Zhu & Liu, 2003
). In this context, it is pertinent to note that poly(I)
poly(C)-mediated induction of IRF1 activation is profoundly inhibited in cells treated with antagonists of ERK (Harcourt & Offermann, 2001
); therefore, an inhibition of ERK may perturb the innate immune response. However, a causative link between NS5A-mediated ERK inhibition and aberrant IFN signalling remains to be proven, and the observation of an antagonistic effect of ERK on RNA replication still requires further verification.
It has also been demonstrated that NS5A is able to inhibit the EGF-stimulated activation of p38MAPK (He et al., 2001). This pathway plays a pivotal role in control of protein translation via a downstream target, Mnk-1, which in turn phosphorylates and regulates the activity of the translation initiation factor eIF4E (Wang et al., 1998
). NS5A expression resulted in decreased eIF4E phosphorylation (He et al., 2001
). Although the mechanism by which NS5A inhibits p38 is unclear, p38 is a substrate for PKR, itself known to be downregulated by NS5A. Phosphorylation of eIF4E increases cap-dependent translation, and NS5A-mediated inhibition of this event might result in a cellular environment that favoured cap-independent (IRES-mediated) initiation of translation, a hypothesis supported by evidence showing that NS5A stimulates HCV IRES-driven translation (He et al., 2003
).
Thirdly, NS5A has also been shown to activate the stress-activated JNK MAPK signalling pathway (Park et al., 2003). At first glance this may seem at odds with the data of Macdonald et al. (2003)
, who found no modulation of JNK activity in replicon cells. However, on closer inspection this discrepancy can be reconciled. NS5A was shown to bind to the adaptor protein TRAF2, which is recruited to the tumour necrosis factor (TNF) receptor upon ligand binding and is involved in activation of JNK (Liu et al., 1996
). NS5A does not directly activate JNK, it merely potentiates activation mediated via TNF-
(Park et al., 2003
): basal levels of JNK activity were unaffected by NS5A. Activation of JNK by NS5A was mediated by the interaction with TRAF2, and overexpression of TRAF2 was sufficient to induce NS5A-mediated activation of JNK in the absence of TNF-
stimulation. Taken together, these data show that NS5A is able to differentially regulate members of the MAPK family in a stimulus-dependent manner. It is pertinent to note that, as discussed above, three MAPKKs (MEK1, MKK6 and MKK7
1) have all been shown to phosphorylate NS5A (Coito et al., 2004
). These three MAPKKs phosphorylate ERK, p38MAPK and JNK, respectively, suggesting that the interaction between NS5A and MAPK signalling may occur at multiple stages.
One of the many effects of the deregulation of MAPK signalling is perturbation of host cell cycle control; indeed, activation of the ERK pathway by growth factors is required for hepatocytes to progress through a restriction point in the late G1 phase (Talarmin et al., 1999). Many viruses perturb the cell cycle in order to facilitate maximal virus replication, in some cases causing a block in G1, for example reovirus HA3 (Saragovi et al., 1999
). Interestingly, a recent study of liver biopsies from HCV-infected patients observed that the proportion of cells in G1 was significantly increased in comparison to samples from patients with alcohol-induced liver injury (Freeman et al., 2003
). In this regard, NS5A has been shown to regulate the cell cycle. However, the results of these studies are ambiguous. For example, a reduction in expression of the cyclin-dependent kinase inhibitor p21WAF1 by NS5A was observed (Ghosh et al., 1999
), and was shown to be due to a direct interaction between NS5A and p53, leading to cytoplasmic sequestration of the latter (Majumder et al., 2001
). Interestingly, NS5A was shown to bind to the amino terminus of p53, which is the region bound by host cellular inhibitors of p53 such as MDM2 (Michael & Oren, 2003
). By reducing p21WAF1 levels NS5A promoted cell growth, suggesting that NS5A was able to subvert cell cycle regulation and bypass checkpoint controls, one outcome of which could be tumour formation: indeed, when NS5A was expressed in nude mice all of the animals developed tumours within 2 weeks (Ghosh et al., 1999
). Many viral proteins, including human papillomavirus 16 E6, hepatitis B virus pX, adenovirus E1B/55 kDa and the simian virus 40 large T antigen (Munger & Howley, 2002
; Murakami, 2001
; Burgert et al., 2002
; Chen & Hahn, 2003
), inhibit p53 and p21WAF1 activity. All of these viruses establish chronic infections and correlate with oncogenesis, and it is interesting to speculate that by subverting cellular growth NS5A may also facilitate the development of HCC, increasingly cited as a long-term consequence of HCV infection. A note of caution must be raised, however, as it has been observed that in a variety of stable cell lines NS5A causes an increase in the G2/M phase of the cell cycle, resulting in growth retardation (Arima et al., 2001
). This was caused by a direct interaction between NS5A and the cyclin-dependent kinase Cdk1 and resulted in the activation of p21WAF1. It is difficult to find consensus between these opposing observations; however, these experiments were to a large degree performed in transformed cell lines of non-hepatic origin. To clarify the effects of NS5A on the cell cycle it is vital that experiments are conducted in primary human hepatocytes; however, such experiments are technically challenging.
NS5A and apoptosis
Apoptosis is an important process for the elimination of virus-infected cells and, consequently, many viruses that establish a chronic infection have evolved mechanisms to evade this response to ensure their persistence. Over recent years there have been numerous publications demonstrating reduced apoptosis in cells expressing various HCV proteins, including NS5A, and data from these studies suggest that NS5A may utilize multiple mechanisms to inhibit both extrinsic and intrinsic apoptotic stimuli.
TNF- is a potent extrinsic apoptotic stimulus (Gaur & Aggarwal, 2003
), and it has been demonstrated that cells expressing NS5A are refractile to TNF-
treatment, undergoing significantly less apoptosis than control cells (Ghosh et al., 2000a
; Miyasaka et al., 2003
). These findings have been confirmed using a transgenic mouse model in which NS5A was expressed from a liver-specific promoter (Majumder et al., 2002
). Biochemical analysis revealed that the inhibition of TNF-
signalling was mediated by an interaction between NS5A and the TNF-
-responsive adaptor protein TRADD. TRADD is recruited to the activated TNF receptor upon ligand binding and forms a multimeric complex with the TNF receptor, a second death domain containing adaptor protein, FADD, and TRAF2. This complex then signals to a variety of downstream effectors, including caspases and NF-
B. By binding to TRADD, NS5A was able to block these signals. However, this study also found that, unlike TNF-
-mediated signalling, NS5A was unable to inhibit apoptosis stimulated by FasL (Majumder et al., 2002
). This may be because NS5A is unable to bind FADD, the death domain adaptor protein recruited to the Fas receptor upon ligand binding.
Intrinsic apoptosis is regulated by a balance between pro- and anti-apoptotic signals, for example the Bcl2 family of proteins contain pro- and anti-apoptotic members that localize to the mitochondrial and nuclear membranes, where they form heterodimers, driven by interactions between Bcl-2 homology (BH) domains. The balance between various members of the family dictates the apoptotic response (Cory & Adams, 2002). One recent study noted that sequences within the ISDR share homology with the BH domains of anti-apoptotic members of the Bcl2 family (Chung et al., 2003
). The putative BH domain within NS5A allowed it to bind to the pro-apoptotic Bcl2 protein, Bax, rendering cells refractile to certain pro-apoptotic agonists (Chung et al., 2003
). An important caveat to this work is the requirement for NS5A to co-localize with Bax within the nucleus. However, intact NS5A resides exclusively within the cytoplasm, although truncated forms have been shown to localize to the nucleus (Satoh et al., 2000
). NS5A also targets another pro-apoptotic Bcl2 family member, Bad (He et al., 2002
; Street et al., 2004
), by a different mechanism (see below).
As discussed previously, NS5A is able to bind to and sequester p53 in the cytoplasm. As well as its role in regulating cell growth, p53 also acts as an intrinsic apoptotic signal following DNA damage or unscheduled DNA synthesis (Fridman & Lowe, 2003). Most viruses that target p53 have tumorigenic properties, highlighting the importance of this protein for cell survival. The ability of NS5A to bind p53 and induce cytosolic sequestration may prevent DNA binding and transactivation of target genes, resulting in impaired apoptosis (Lan et al., 2002
). This may have implications for hepatocarcinogenesis, as any adverse effects on p53 activity could lead to compromised DNA repair, creating a pool of hepatocytes containing unstable genetic material with increased probability of neoplastic transformation. The varied effects of NS5A on apoptosis are illustrated in Fig. 4
.
|
|
|
Subgenomic replicons have been used to ask whether NS5A-mediated elevation of ROS might affect HCV replication. Controversially, two studies showed that ROS either suppressed (Choi et al., 2004) or stimulated (Qadri et al., 2004
) replicon replication; the former study showed that treatment of cells with anti-oxidants restored levels of replicon replication. Further study is clearly warranted to develop our understanding of the role of ROS production in HCV replication.
Other NS5A targets
There are many other NS5A-interacting partners (Table 1), unfortunately space constraints preclude the detailed discussion of these here. However, a number of them are worthy of brief mention as they are involved in protein trafficking and membrane morphology, and thus may have relevance for the formation and function of the membrane-bound replication complex. These include karyopherin
3 (Chung et al., 2000
), the SNARE-like protein hVAP-33 (Tu et al., 1999
; Gao et al., 2004
), apolipoprotein A1 (Shi et al., 2002
) and amphiphysin II (Zech et al., 2003
). It is tempting to speculate that, whilst at the ER, NS5A may interact with many of these proteins to aid in the formation of a membrane-bound replication complex. This idea is supported by recent data (Gao et al., 2004
) showing that siRNA or dominant negative mutants of hVAP-33 resulted in relocation of NS5B from detergent-resistant to detergent-sensitive membranes and reduced levels of HCV RNA and proteins in replicon cells. Over the next few years the use of the replicon system should strengthen our understanding of many of these interactions, and determine whether they play a role in the formation of a replication complex.
Concluding remarks
Although there is much conflicting experimental data in the literature, it is generally accepted that NS5A plays a critical role in the replication of HCV, both directly, with regard to viral RNA replication, and indirectly, by modulating the host cell environment to favour the virus. The molecular details of the involvement of NS5A in the process of HCV genomic RNA replication remain to be characterized. NS5A is clearly associated with replicating RNA in a cytoplasmic replication complex, but we currently have little or no information about precisely what function it is performing. NS5A also perturbs signalling pathways within the cell and there are many convincing arguments to explain why such perturbations would benefit the virus.
What direction should research into NS5A take? A key goal must be to elucidate the three-dimensional structure of the protein. This would provide a rational framework for mutagenesis and facilitate a better understanding of the proteinprotein interactions participated in by NS5A. It is certain that the list of NS5A-interacting proteins, together with the list of NS5A functions, will grow; however, the challenge will be to validate these findings, linking laboratory observations to the pathogenesis of HCV infection, at both the cellular and organismal levels. Our understanding of the role of NS5A in virus replication would be greatly aided by the development of more-robust replication systems for HCV, ideally involving productive infection of primary human hepatocytes, allowing the function of NS5A to be analysed in a truly physiologically relevant setting. The problems of cultivating such cells in the laboratory would seem to preclude such experiments in the conceivable future; however, developments such as the mosaic mouse model (Mercer et al., 2001), full-length replicons (Ikeda et al., 2002
), and the use of replicons in cells other than Huh7 (Zhu et al., 2003
; Ali et al., 2004
) provide optimism for the future.
Will NS5A ever be a target for antiviral therapy? As a protein with no known enzymic activity it is certainly not amenable to current drug screening protocols. However, as inhibitors of the HCV replicative enzymes (NS3 and NS5B) are still in development, attention is being drawn to other potential targets. We believe that NS5A is such a target, and that the challenge ahead is to prove that specific NS5Acellular protein interactions are critical for virus replication. Strategies to disrupt these interactions would then provide the basis for novel, much-needed therapies for HCV. It seems likely that NS5A will continue to keep us busy for many years into the future!
![]() |
ACKNOWLEDGEMENTS |
---|
![]() |
REFERENCES |
---|
![]() ![]() ![]() |
---|
Ali, S., Pellerin, C., Lamarre, D. & Kukolj, G. (2004). Hepatitis C virus subgenomic replicons in the human embryonic kidney 293 cell line. J Virol 78, 491501.
Anonymous (1999). Global surveillance and control of hepatitis C. Report of a WHO Consultation organized in collaboration with the Viral Hepatitis Prevention Board, Antwerp, Belgium. J Viral Hepat 6, 3547.[CrossRef][Medline]
Arima, N., Kao, C. Y., Licht, T., Padmanabhan, R., Sasaguri, Y. & Padmanabhan, R. (2001). Modulation of cell growth by the hepatitis C virus nonstructural protein NS5A. J Biol Chem 276, 1267512684.
Asabe, S. I., Tanji, Y., Satoh, S., Kaneko, T., Kimura, K. & Shimotohno, K. (1997). The N-terminal region of hepatitis C virus-encoded NS5A is important for NS4A-dependent phosphorylation. J Virol 71, 790796.[Abstract]
Ban, K. C., Singh, H., Krishnan, R. & Seow, H. F. (2003). GSK-3 phosphorylation and alteration of
-catenin in hepatocellular carcinoma. Cancer Lett 199, 201208.[CrossRef][Medline]
Blight, K. J., Kolykhalov, A. A. & Rice, C. M. (2000). Efficient initiation of HCV RNA replication in cell culture. Science 290, 19721974.
Blindenbacher, A., Duong, F. H., Hunziker, L. & 9 other authors (2003). Expression of hepatitis C virus proteins inhibits interferon alpha signaling in the liver of transgenic mice. Gastroenterology 124, 14651475.[CrossRef][Medline]
Brass, V., Bieck, E., Montserret, R., Wolk, B., Hellings, J. A., Blum, H. E., Penin, F. & Moradpour, D. (2002). An amino-terminal amphipathic -helix mediates membrane association of the hepatitis C virus nonstructural protein 5A. J Biol Chem 277, 81308139.
Briggs, S. D., Scholtz, B., Jacque, J. M., Swingler, S., Stevenson, M. & Smithgall, T. E. (2001). HIV-1 Nef promotes survival of myeloid cells by a Stat3-dependent pathway. J Biol Chem 276, 2560525611.
Bukh, J., Pietschmann, T., Lohmann, V. & 7 other authors (2002). Mutations that permit efficient replication of hepatitis C virus RNA in Huh-7 cells prevent productive replication in chimpanzees. Proc Natl Acad Sci U S A 99, 1441614421.
Burgert, H. G., Ruzsics, Z., Obermeier, S., Hilgendorf, A., Windheim, M. & Elsing, A. (2002). Subversion of host defense mechanisms by adenoviruses. Curr Top Microbiol Immunol 269, 273318.[Medline]
Cantley, L. C. (2002). The phosphoinositide 3-kinase pathway. Science 296, 16551657.
Chang, L. F. & Karin, M. (2001). Mammalian MAP kinase signalling cascades. Nature 410, 3740.[CrossRef][Medline]
Chen, W. & Hahn, W. C. (2003). SV40 early region oncoproteins and human cell transformation. Histol Histopathol 18, 541550.[Medline]
Choi, J., Lee, K. J., Zheng, Y., Yamaga, A. K., Lai, M. M. & Ou, J. H. (2004). Reactive oxygen species suppress hepatitis C virus RNA replication in human hepatoma cells. Hepatology 39, 8189.[CrossRef][Medline]
Chung, K. M., Lee, J., Kim, J. E., Song, O. K., Cho, S., Lim, J., Seedorf, M., Hahm, B. & Jang, S. K. (2000). Nonstructural protein 5A of hepatitis C virus inhibits the function of karyopherin 3. J Virol 74, 52335241.
Chung, Y. L., Sheu, M. L. & Yen, S. H. (2003). Hepatitis C virus NS5A as a potential viral Bcl-2 homologue interacts with Bax and inhibits apoptosis in hepatocellular carcinoma. Int J Cancer 107, 6573.[CrossRef][Medline]
Coffer, P. J., Jin, J. & Woodgett, J. R. (1998). Protein kinase B (c-Akt): a multifunctional mediator of phosphatidylinositol 3-kinase activation. Biochem J 335, 113.[Medline]
Coito, C., Diamond, D. L., Neddermann, P., Korth, M. J. & Katze, M. G. (2004). High-throughput screening of the yeast kinome: identification of human serine/threonine protein kinases that phosphorylate the hepatitis C virus NS5A protein. J Virol 78, 35023513.
Cooray, S. (2004). The pivotal role of phosphatidylinositol 3-kinaseAkt signal transduction in virus survival. J Gen Virol 85, 10651076.
Cory, S. & Adams, J. M. (2002). The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer 2, 647656.[CrossRef][Medline]
Cui, J., Zhou, X. D., Liu, Y. K., Tang, Z. Y. & Romeih, M. (2003). Wnt signaling in hepatocellular carcinoma: analysis of mutation and expression of -catenin, T-cell factor-4 and glycogen synthase kinase 3-
genes. J Gastroenterol Hepatol 18, 280287.[CrossRef][Medline]
Desbois-Mouthon, C., Blivet-Van Eggelpoel, M. J., Beurel, E., Boissan, M., Delelo, R., Cadoret, A. & Capeau, J. (2002). Dysregulation of glycogen synthase kinase-3 signaling in hepatocellular carcinoma cells. Hepatology 36, 15281536.[CrossRef][Medline]
Dimitrova, M., Imbert, I., Kieny, M. P. & Schuster, C. (2003). Protein-protein interactions between hepatitis C virus nonstructural proteins. J Virol 77, 54015414.
Edamoto, Y., Hara, A., Biernat, W. & 7 other authors (2003). Alterations of RB1, p53 and Wnt pathways in hepatocellular carcinomas associated with hepatitis C, hepatitis B and alcoholic liver cirrhosis. Int J Cancer 106, 334341.[CrossRef][Medline]
Egger, D., Wolk, B., Gosert, R., Bianchi, L., Blum, H. E., Moradpour, D. & Bienz, K. (2002). Expression of hepatitis C virus proteins induces distinct membrane alterations including a candidate viral replication complex. J Virol 76, 59745984.
Elazar, M., Cheong, K. H., Liu, P., Greenberg, H. B., Rice, C. M. & Glenn, J. S. (2003). Amphipathic helix-dependent localization of NS5A mediates hepatitis C virus RNA replication. J Virol 77, 60556061.
Enomoto, N., Sakuma, I., Asahina, Y., Kurosaki, M., Murakami, T., Yamamoto, C., Izumi, N., Marumo, F. & Sato, C. (1995). 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 region. J Clin Invest 96, 224230.[Medline]
Enomoto, N., Sakuma, I., Asahina, Y. & 7 other authors (1996). Mutations in the nonstructural protein 5A gene and response to interferon in patients with chronic hepatitis C virus 1b infection. N Engl J Med 334, 7781.
Ezelle, H. J., Balachandran, S., Sicheri, F., Polyak, S. J. & Barber, G. N. (2001). Analyzing the mechanisms of interferon-induced apoptosis using CrmA and hepatitis C virus NS5A. Virology 281, 124137.[CrossRef][Medline]
Freeman, A., Hamid, S., Morris, L., Vowler, S., Rushbrook, S., Wight, D. G., Coleman, N. & Alexander, G. J. (2003). Improved detection of hepatocyte proliferation using antibody to the pre-replication complex: an association with hepatic fibrosis and viral replication in chronic hepatitis C virus infection. J Viral Hepat 10, 345350.[CrossRef][Medline]
Frese, M., Pietschmann, T., Moradpour, D., Haller, O. & Bartenschlager, R. (2001). Interferon- inhibits hepatitis C virus subgenomic RNA replication by an MxA-independent pathway. J Gen Virol 82, 723733.
Fridman, J. S. & Lowe, S. W. (2003). Control of apoptosis by p53. Oncogene 22, 90309040.[CrossRef][Medline]
Gale, M. J., Jr, Korth, M. J., Tang, N. M., Tan, S. L., Hopkins, D. A., Dever, T. E., Polyak, S. J., Gretch, D. R. & Katze, M. G. (1997). Evidence that hepatitis C virus resistance to interferon is mediated through repression of the PKR protein kinase by the nonstructural 5A protein. Virology 230, 217227.[CrossRef][Medline]
Gale, M., Jr, Blakely, C. M., Kwieciszewski, B. & 7 other authors (1998). Control of PKR protein kinase by hepatitis C virus nonstructural 5a protein: molecular mechanisms of kinase regulation. Mol Cell Biol 18, 52085218.
Gao, L., Aizaki, H., He, J. W. & Lai, M. M. (2004). Interactions between viral nonstructural proteins and host protein hVAP-33 mediate the formation of hepatitis C virus RNA replication complex on lipid raft. J Virol 78, 34803488.
Gaur, U. & Aggarwal, B. B. (2003). Regulation of proliferation, survival and apoptosis by members of the TNF superfamily. Biochem Pharmacol 66, 14031408.[CrossRef][Medline]
Georgopoulou, U., Caravokiri, K. & Mavromara, P. (2003). Suppression of the ERK1/2 signaling pathway from HCV NS5A protein expressed by herpes simplex recombinant viruses. Arch Virol 148, 237251.[CrossRef][Medline]
Ghosh, A. K., Steele, R., Meyer, K., Ray, R. & Ray, R. B. (1999). Hepatitis C virus NS5A protein modulates cell cycle regulatory genes and promotes cell growth. J Gen Virol 80, 11791183.[Abstract]
Ghosh, A. K., Majumder, M., Steele, R., Meyer, K., Ray, R. & Ray, R. B. (2000a). Hepatitis C virus NSSA protein protects against TNF- mediated apoptotic cell death. Virus Res 67, 173178.[CrossRef][Medline]
Ghosh, A. K., Majumder, M., Steele, R., Yaciuk, P., Chrivia, J., Ray, R. & Ray, R. B. (2000b). Hepatitis C virus NS5A protein modulates transcription through a novel cellular transcription factor SRCAP. J Biol Chem 275, 71847188.
Ghosh, A. K., Majumder, M., Steele, R., Ray, R. & Ray, R. B. (2003). Modulation of interferon expression by hepatitis C virus NS5A protein and human homeodomain protein PTX1. Virology 306, 5159.[CrossRef][Medline]
Girard, S., Shalhoub, P., Lescure, P., Sabile, A., Misek, D. E., Hanash, S., Brechot, C. & Beretta, L. (2002). An altered cellular response to interferon and up-regulation of interleukin-8 induced by the hepatitis C viral protein NS5A uncovered by microarray analysis. Virology 295, 272283.[CrossRef][Medline]
Gong, G. Z., Waris, G., Tanveer, R. & Siddiqui, A. (2001). Human hepatitis C virus NS5A protein alters intracellular calcium levels, induces oxidative stress, and activates STAT-3 and NF-B. Proc Natl Acad Sci U S A 98, 95999604.
Griffin, S. D. C., Beales, L. P., Clarke, D. S., Worsfold, O., Evans, S. D., Jaeger, J., Harris, M. P. G. & Rowlands, D. J. (2003). The p7 protein of hepatitis C virus forms an ion channel that is blocked by the antiviral drug, Amantadine. FEBS Lett 535, 3438.[CrossRef][Medline]
Guo, J. T., Bichko, V. V. & Seeger, C. (2001). Effect of alpha interferon on the hepatitis C virus replicon. J Virol 75, 85168523.
Hanafusa, H., Torii, S., Yasunaga, T. & Nishida, E. (2002). Sprouty1 and Sprouty2 provide a control mechanism for the Ras/MAPK signalling pathway. Nat Cell Biol 4, 850858.[CrossRef][Medline]
Harcourt, J. L. & Offermann, M. K. (2001). Multiple signaling cascades are differentially involved in gene induction by double stranded RNA in interferon--primed cells. Eur J Biochem 268, 13731381.
Hardy, R. W., Marcotrigiano, J., Blight, K. J., Majors, J. E. & Rice, C. M. (2003). Hepatitis C virus RNA synthesis in a cell-free system isolated from replicon-containing hepatoma cells. J Virol 77, 20292037.
Harris, M. (1996). From negative factor to a critical role in virus pathogenesis: the changing fortunes of Nef. J Gen Virol 77, 23792392.[Medline]
He, Y. P., Tan, S. L., Tareen, S. U., Vijaysri, S., Langland, J. O., Jacobs, B. L. & Katze, M. G. (2001). Regulation of mRNA translation and cellular signaling by hepatitis C virus nonstructural protein NS5A. J Virol 75, 50905098.
He, Y. P., Nakao, H. H., Tan, S. L., Polyak, P. J., Neddermann, P., Vijaysri, S., Jacobs, B. L. & Katze, M. G. (2002). Subversion of cell signaling pathways by hepatitis C virus nonstructural 5A protein via interaction with Grb2 and P85 phosphatidylinositol 3-kinase. J Virol 76, 92079217.
He, Y. P., Yan, W., Coito, C., Li, Y., Gale, M., Jr & Katze, M. G. (2003). The regulation of hepatitis C virus (HCV) internal ribosome-entry site-mediated translation by HCV replicons and nonstructural proteins. J Gen Virol 84, 535543.
Heim, M. H., Moradpour, D. & Blum, H. E. (1999). Expression of hepatitis C virus proteins inhibits signal transduction through the Jak-STAT pathway. J Virol 73, 84698475.
Honda, M., Kaneko, S., Matsushita, E., Kobayashi, K., Abell, G. A. & Lemon, S. M. (2000). Cell cycle regulation of hepatitis C virus internal ribosomal entry site-directed translation. Gastroenterology 118, 152162.[Medline]
Houshmand, H. & Bergqvist, A. (2003). Interaction of hepatitis C virus NS5A with La protein revealed by T7 phage display. Biochem Biophys Res Commun 309, 695701.[CrossRef][Medline]
Ide, Y., Zhang, L. W., Chen, M., Inchauspe, G., Bahl, C., Sasaguri, Y. & Padmanabhan, R. (1996). Characterization of the nuclear localization signal and subcellular distribution of hepatitis C virus nonstructural protein NS5A. Gene 182, 203211.[CrossRef][Medline]
Ide, Y., Tanimoto, A., Sasaguri, Y. & Padmanabhan, R. (1997). 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- catalytic subunit. Gene 201, 151158.[CrossRef][Medline]
Ikeda, M., Yi, M. K., Li, K. & Lemon, S. M. (2002). Selectable subgenomic and genome-length dicistronic RNAs derived from an infectious molecular clone of the HCV-N strain of hepatitis C virus replicate efficiently in cultured Huh7 cells. J Virol 76, 29973006.
Kaneko, T., Tanji, Y., Satoh, S., Hijikata, M., Asabe, S., Kumura, K. & Shimotohno, K. (1994). Production of two phosphoproteins from the NS5A region of the hepatitis C viral genome. Biochem Biophys Res Commun 205, 320326.[CrossRef][Medline]
Katze, M. G., Kwieciszewski, B., Goodlett, D. R., Blakely, C. M., Neddermann, P., Tan, S. L. & Aebersold, R. (2000). Ser(2194) is a highly conserved major phosphorylation site of the hepatitis C virus nonstructural protein NS5A. Virology 278, 501513.[CrossRef][Medline]
Khabar, K. S., Al-Zoghaibi, F., Al-Ahdal, M. N. & 8 other authors (1997). The chemokine, interleukin 8, inhibits the antiviral action of interferon
. J Exp Med 186, 10771085.
Kim, J., Lee, D. & Choe, J. (1999). Hepatitis C virus NS5A protein is phosphorylated by casein kinase II. Biochem Biophys Res Commun 257, 777781.[CrossRef][Medline]
Klein, N. P. & Schneider, R. J. (1997). Activation of Src family kinases by hepatitis B virus HBx protein and coupled signaling to Ras. Mol Cell Biol 17, 64276436.[Abstract]
Koch, J. O. & Bartenschlager, R. (1999). Modulation of hepatitis C virus NS5A hyperphosphorylation by nonstructural proteins NS3, NS4A, and NS4B. J Virol 73, 71387146.
Korkaya, H., Jameel, S., Gupta, D. & 9 other authors (2001). The ORF3 protein of hepatitis E virus binds to Src homology 3 domains and activates MAPK. J Biol Chem 276, 4238942400.
Krieger, N., Lohmann, V. & Bartenschlager, R. (2001). Enhancement of hepatitis C virus RNA replication by cell culture-adaptive mutations. J Virol 75, 46144624.
Kucharczak, J., Simmons, M. J., Fan, Y. & Gelinas, C. (2003). To be, or not to be: NF-B is the answer role of Rel/NF-
B in the regulation of apoptosis. Oncogene 22, 89618982.[CrossRef][Medline]
Lan, K. H., Sheu, M. L., Hwang, S. J. & 8 other authors (2002). HCV NS5A interacts with p53 and inhibits p53-mediated apoptosis. Oncogene 21, 48014811.[CrossRef][Medline]
Lanford, R. E., Guerra, B., Lee, H., Averett, D. R., Pfeiffer, B., Chavez, D., Notvall, L. & Bigger, C. (2003). Antiviral effect and virus-host interactions in response to alpha interferon, gamma interferon, poly(I)poly(C), tumor necrosis factor alpha, and ribavirin in hepatitis C virus subgenomic replicons. J Virol 77, 10921104.[CrossRef][Medline]
Liu, Q. Y., Bhat, R. A., Prince, A. M. & Zhang, P. (1999). The hepatitis C virus NS2 protein generated by NS2-3 autocleavage is required for NS5A phosphorylation. Biochem Biophys Res Commun 254, 572577.[CrossRef][Medline]
Liu, Z. G., Hsu, H., Goeddel, D. V. & Karin, M. (1996). Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-B activation prevents cell death. Cell 87, 565576.[Medline]
Lohmann, V., Korner, F., Koch, J. O., Herian, U., Theilmann, L. & Bartenschlager, R. (1999). Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science 285, 110113.
Lohmann, V., Korner, F., Dobierzewska, A. & Bartenschlager, R. (2001). Mutations in hepatitis C virus RNAs conferring cell culture adaptation. J Virol 75, 14371449.
Macdonald, A., Crowder, K., Street, A., McCormick, C., Saksela, K. & Harris, M. (2003). The hepatitis C virus non-structural 5A protein inhibits activating protein-1 function by perturbing Ras-ERK pathway signaling. J Biol Chem 278, 1777517784.
Macdonald, A., Crowder, K., Street, A., McCormick, C. & Harris, M. (2004). The hepatitis C virus NS5A protein binds to members of the Src family of tyrosine kinases and regulates kinase activity. J Gen Virol 85, 721729.
Majumder, M., Ghosh, A. K., Steele, R., Ray, R. & Ray, R. B. (2001). Hepatitis C virus NS5A physically associates with p53 and regulates p21/waf1 gene expression in a p53-dependent manner. J Virol 75, 14011407.
Majumder, M., Ghosh, A. K., Steele, R., Zhou, X. Y., Phillips, N. J., Ray, R. & Ray, R. B. (2002). Hepatitis C virus NS5A protein impairs TNF-mediated hepatic apoptosis, but not by an anti-FAS antibody, in transgenic mice. Virology 294, 94105.[CrossRef][Medline]
McCormick, C. J., Challinor, L., Macdonald, A., Rowlands, D. J. & Harris, M. (2004). Introduction of replication-competent hepatitis C virus transcripts using a tetracycline-regulable baculovirus delivery system. J Gen Virol 85, 429439.
Mercer, D. F., Schiller, D. E., Elliott, J. F. & 9 other authors (2001). Hepatitis C virus replication in mice with chimeric human livers. Nat Med 7, 927933.[CrossRef][Medline]
Michael, D. & Oren, M. (2003). The p53-Mdm2 module and the ubiquitin system. Semin Cancer Biol 13, 4958.[CrossRef][Medline]
Miyasaka, Y., Enomoto, N., Kurosaki, M. & 9 other authors (2003). Hepatitis C virus nonstructural protein 5A inhibits tumor necrosis factor--mediated apoptosis in Huh7 cells. J Infect Dis 188, 15371544.[CrossRef][Medline]
Mottola, G., Cardinali, G., Ceccacci, A., Trozzi, C., Bartholomew, L., Torrisi, M. R., Pedrazzini, E., Bonatti, S. & Migliaccio, G. (2002). Hepatitis C virus nonstructural proteins are localized in a modified endoplasmic reticulum of cells expressing viral subgenomic replicons. Virology 293, 3143.[CrossRef][Medline]
Munger, K. & Howley, P. M. (2002). Human papillomavirus immortalization and transformation functions. Virus Res 89, 213228.[CrossRef][Medline]
Murakami, S. (2001). Hepatitis B virus X protein: a multifunctional viral regulator. J Gastroenterol 36, 651660.[CrossRef][Medline]
Neddermann, P., Clementi, A. & De Francesco, R. (1999). Hyperphosphorylation of the hepatitis C virus NS5A protein requires an active NS3 protease, NS4A, NS4B, and NS5A encoded on the same polyprotein. J Virol 73, 99849991.
Noguchi, T., Satoh, S., Noshi, T., Hatada, E., Fukuda, R., Kawai, A., Ikeda, S., Hijikata, M. & Shimotohno, K. (2001). Effects of mutation in hepatitis C virus nonstructural protein 5A on interferon resistance mediated by inhibition of PKR kinase activity in mammalian cells. Microbiol Immunol 45, 829840.[Medline]
Nousbaum, J. B., Polyak, S. J., Ray, S. C., Sullivan, D. G., Larson, A. M., Carithers, R. L., Jr & Gretch, D. R. (2000). Prospective characterization of full-length hepatitis C virus NS5A quasispecies during induction and combination antiviral therapy. J Virol 74, 90289038.
Park, K. J., Choi, S. H., Lee, S. Y., Hwang, S. B. & Lai, M. M. C. (2002). Nonstructural 5A protein of hepatitis C virus modulates tumor necrosis factor -stimulated nuclear factor
B activation. J Biol Chem 277, 1312213128.
Park, K. J., Choi, S. H., Choi, D. H., Park, J. M., Yie, S. W., Lee, S. Y. & Hwang, S. B. (2003). Hepatitis C virus NS5A protein modulates c-Jun N-terminal kinase through interaction with tumor necrosis factor receptor-associated factor 2. J Biol Chem 278, 3071130718.
Paterson, M., Laxton, C. D., Thomas, H. C., Ackrill, A. M. & Foster, G. R. (1999). Hepatitis C virus NS5A protein inhibits inteferon antiviral activity, but the effects do not correlate with clinical response. Gastroenterology 117, 11871197.[Medline]
Pflugheber, J., Fredericksen, B., Sumpter, R., Jr, Wang, C. F., Ware, F., Sodora, D. L. & Gale, M., Jr (2002). Regulation of PKR and IRF-1 during hepatitis C virus RNA replication. Proc Natl Acad Sci U S A 99, 46504655.
Pietschmann, T., Lohmann, V., Rutter, G., Kurpanek, K. & Bartenschlager, R. (2001). Characterization of cell lines carrying self-replicating hepatitis C virus RNAs. J Virol 75, 12521264.
Podevin, P., Sabile, A., Gajardo, R., Delhem, N., Abadie, A., Lozach, P. Y., Beretta, L. & Brechot, C. (2001). Expression of hepatitis C virus NS5A natural mutants in a hepatocytic cell line inhibits the antiviral effect of interferon in a PKR-independent manner. Hepatology 33, 15031511.[CrossRef][Medline]
Polyak, S. J., Paschal, D. M., McArdle, S., Gale, M. J., Jr, Moradpour, D. & Gretch, D. R. (1999). Characterization of the effects of hepatitis C virus nonstructural 5A protein expression in human cell lines and on interferon-sensitive virus replication. Hepatology 29, 12621271.[Medline]
Polyak, S. J., Khabar, K. S., Paschal, D. M., Ezelle, H. J., Duverlie, G., Barber, G. N., Levy, D. E., Mukaida, N. & Gretch, D. R. (2001a). Hepatitis C virus nonstructural 5A protein induces interleukin-8, leading to partial inhibition of the interferon-induced antiviral response. J Virol 75, 60956106.
Polyak, S. J., Khabar, K. S., Rezeiq, M. & Gretch, D. R. (2001b). Elevated levels of interleukin-8 in serum are associated with hepatitis C virus infection and resistance to interferon therapy. J Virol 75, 62096211.
Poynard, T., Yuen, M. F., Ratziu, V. & Lai, C. L. (2003). Viral hepatitis C. Lancet 362, 20952100.[CrossRef][Medline]
Prange, W., Breuhahn, K., Fischer, F., Zilkens, C., Pietsch, T., Petmecky, K., Eilers, R., Dienes, H. P. & Schirmacher, P. (2003). -Catenin accumulation in the progression of human hepatocarcinogenesis correlates with loss of E-cadherin and accumulation of p53, but not with expression of conventional WNT-1 target genes. J Pathol 201, 250259.[CrossRef][Medline]
Qadri, I., Iwahashi, M. & Simon, F. (2002). Hepatitis C virus NS5A protein binds TBP and p53, inhibiting their DNA binding and p53 interactions with TBP and ERCC3. Biochim Biophys Acta 1592, 193204.[Medline]
Qadri, I., Iwahashi, M., Capasso, J. M., Hopken, M. W., Flores, S., Schaack, J. & Simon, F. R. (2004). Induced oxidative stress and activated expression of manganese superoxide dismutase during hepatitis C virus replication: role of JNK, p38 MAPK and AP-1. Biochem J 378, 919928.[CrossRef][Medline]
Qin, W., Yamashita, T., Shirota, Y., Lin, Y., Wei, W. & Murakami, S. (2001). Mutational analysis of the structure and functions of hepatitis C virus RNA-dependent RNA polymerase. Hepatology 33, 728737.[CrossRef][Medline]
Reed, K. E. & Rice, C. M. (1999). Identification of the major phosphorylation site of the hepatitis C virus H strain NS5A protein as serine 2321. J Biol Chem 274, 2801128018.
Reed, K. E., Xu, J. & Rice, C. M. (1997). Phosphorylation of the hepatitis C virus NS5A protein in vitro and in vivo: properties of the NS5A-associated kinase. J Virol 71, 71877197.[Abstract]
Renkema, G. H. & Saksela, K. (2000). Interactions of HIV-1 Nef with cellular signal transducing proteins. Front Biosci 5, D268283.[Medline]
Saragovi, H. U., Rebai, N., Di Guglielmo, G. M., MacLeod, R., Sheng, J. S., Rubin, D. H. & Greene, M. I. (1999). A G1 cell cycle arrest induced by ligands of the reovirus type 3 receptor is secondary to inactivation of p21ras and mitogen-activated protein kinase. DNA Cell Biol 18, 763770.[CrossRef][Medline]
Satoh, S., Hirota, M., Noguchi, T., Hijikata, M., Handa, H. & Shimotohno, K. (2000). Cleavage of hepatitis C virus nonstructural protein 5A by a caspase-like protease(s) in mammalian cells. Virology 270, 476487.[CrossRef][Medline]
Shang, X. Z., Zhu, H., Lin, K., Tu, Z., Chen, J., Nelson, D. R. & Liu, C. (2004). Stabilized -catenin promotes hepatocyte proliferation and inhibits TNF
-induced apoptosis. Lab Invest 84, 332341.[CrossRef][Medline]
Shi, S. T., Polyak, S. J., Tu, H., Taylor, D. R., Gretch, D. R. & Lai, M. M. C. (2002). Hepatitis C virus NS5A colocalizes with the core protein on lipid droplets and interacts with apolipoproteins. Virology 292, 198210.[CrossRef][Medline]
Shi, S. T., Lee, K. J., Aizaki, H., Hwang, S. B. & Lai, M. M. (2003). Hepatitis C virus RNA replication occurs on a detergent-resistant membrane that cofractionates with caveolin-2. J Virol 77, 41604168.
Shimakami, T., Hijikata, M., Luo, H., Ma, Y. Y., Kaneko, S., Shimotohno, K. & Murakami, S. (2004). Effect of interaction between hepatitis C virus NS5A and NS5B on hepatitis C virus RNA replication with the hepatitis C virus replicon. J Virol 78, 27382748.
Shirota, Y., Luo, H., Qin, W. P., Kaneko, S., Yamashita, T., Kobayashi, K. & Murakami, S. (2002). Hepatitis C virus (HCV) NS5A binds RNA-dependent RNA polymerase (RdRP) NS5B and modulates RNA-dependent RNA polymerase activity. J Biol Chem 277, 1114911155.
Shuai, K. (2003). Serine phosphorylation: arming Stat1 against infection. Immunity 19, 771772.[CrossRef][Medline]
Simon, A. R., Rai, U., Fanburg, B. L. & Cochran, B. H. (1998). Activation of the JAK-STAT pathway by reactive oxygen species. Am J Physiol 275, C16401652.[Medline]
Song, J. H., So, E. Y. & Lee, C. E. (2002). Increased serine phosphorylation and activation of STAT1 by oncogenic Ras transfection. Mol Cells 13, 322326.[Medline]
Street, A., Macdonald, A., Crowder, K. & Harris, M. (2004). The hepatitis C virus NS5A protein activates a phosphoinositide 3-kinase-dependent survival signaling cascade. J Biol Chem 279, 1223212241.
Tai, D. I., Tsai, S. L., Chen, Y. M. & 8 other authors (2000). Activation of nuclear factor B in hepatitis C virus infection: implications for pathogenesis and hepatocarcinogenesis. Hepatology 31, 656664.[Medline]
Talarmin, H., Rescan, C., Cariou, S., Glaise, D., Zanninelli, G., Bilodeau, M., Loyer, P., Guguen-Guillouzo, C. & Baffet, G. (1999). The mitogen-activated protein kinase kinase/extracellular signal-regulated kinase cascade activation is a key signalling pathway involved in the regulation of G(1) phase progression in proliferating hepatocytes. Mol Cell Biol 19, 60036011.
Tan, S. L. & Katze, M. G. (2001). How hepatitis C virus counteracts the interferon response: the jury is still out on NS5A. Virology 284, 112.[CrossRef][Medline]
Tan, S. L., Nakao, H., He, Y. P., Vijaysri, S., Neddermann, P., Jacobs, B. L., Mayer, B. J. & Katze, M. G. (1999). 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 96, 55335538.
Tang, E. D., Nunez, G., Barr, F. G. & Guan, K. L. (1999). Negative regulation of the forkhead transcription factor FKHR by Akt. J Biol Chem 274, 1674116746.
Tanji, Y., Kaneko, T., Satoh, S. & Shimotohno, K. (1995). Phosphorylation of hepatitis C virus-encoded nonstructural protein NS5A. J Virol 69, 39803986.[Abstract]
Tari, A. M. & Lopez-Berestein, G. (2001). GRB2: a pivotal protein in signal transduction. Semin Oncol 28, 142147.[CrossRef][Medline]
Tu, H., Gao, L., Shi, S. T. & 7 other authors (1999). Hepatitis C virus RNA polymerase and NS5A complex with a SNARE- like protein. Virology 263, 3041.[CrossRef][Medline]
Wang, C. F., Pflugheber, J., Sumpter, R., Jr, Sodora, D. L., Hui, D., Sen, G. C. & Gale, M., Jr (2003). Alpha interferon induces distinct translational control programs to suppress hepatitis C virus RNA replication. J Virol 77, 38983912.
Wang, X., Flynn, A., Waskiewicz, A. J., Webb, B. L., Vries, R. G., Baines, I. A., Cooper, J. A. & Proud, C. G. (1998). The phosphorylation of eukaryotic initiation factor eIF4E in response to phorbol esters, cell stresses, and cytokines is mediated by distinct MAP kinase pathways. J Biol Chem 273, 93739377.
Waris, G., Livolsi, A., Imbert, V., Peyron, J. F. & Siddiqui, A. (2003). Hepatitis C virus NS5A and subgenomic replicon activate NF-B via tyrosine phosphorylation of I
B
and its degradation by calpain protease. J Biol Chem 278, 4077840787.
Waris, G., Sarker, S. & Siddiqui, A. (2004). Two-step affinity purification of the hepatitis C virus ribonucleoprotein complex. RNA 10, 321329.
Williams, B. R. (2001). Signal integration via PKR. Sci STKE 89, RE2.
Xu, Z., Choi, J., Lu, W. & Ou, J. H. (2003). Hepatitis C virus F protein is a short-lived protein associated with the endoplasmic reticulum. J Virol 77, 15781583.[CrossRef][Medline]
Yoshikawa, H., Matsubara, K., Qian, G. S., Jackson, P., Groopman, J. D., Manning, J. E., Harris, C. C. & Herman, J. G. (2001). SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity. Nat Genet 28, 2935.[CrossRef][Medline]
Zech, B., Kurtenbach, A., Krieger, N. & 10 other authors (2003). Identification and characterization of amphiphysin II as a novel cellular interaction partner of the hepatitis C virus NS5A protein. J Gen Virol 84, 555560.
Zhu, H. & Liu, C. (2003). Interleukin-1 inhibits hepatitis C virus subgenomic RNA replication by activation of extracellular regulated kinase pathway. J Virol 77, 54935498.
Zhu, Q., Guo, J. T. & Seeger, C. (2003). Replication of hepatitis C virus subgenomes in nonhepatic epithelial and mouse hepatoma cells. J Virol 77, 92049210.