Next Article in Journal
Exploration of the Protection of Riboflavin Laurate on Oral Mucositis Induced by Chemotherapy or Radiotherapy at the Cellular Level: What Is the Leading Contributor?
Next Article in Special Issue
Global Gene Expression Profiling Reveals Functional Importance of Sirt2 in Endothelial Cells under Oxidative Stress
Previous Article in Journal
Irradiated Human Dermal Fibroblasts Are as Efficient as Mouse Fibroblasts as a Feeder Layer to Improve Human Epidermal Cell Culture Lifespan
Previous Article in Special Issue
Cyclic Stretch Induces Inducible Nitric Oxide Synthase and Soluble Guanylate Cyclase in Pulmonary Artery Smooth Muscle Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interplay between Hepatitis C Virus and Redox Cell Signaling

1
Department of Infectious, Parasitic and Immune Mediated Diseases, Italian National Institute of Health, 00161 Rome, Italy
2
Department of Public Health and Infectious Diseases, Institute Pasteur, Cenci-Bolognetti Foundation, "Sapienza" University of Rome, 00185 Rome, Italy
3
San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care, 00163 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2013, 14(3), 4705-4721; https://doi.org/10.3390/ijms14034705
Submission received: 17 January 2013 / Revised: 13 February 2013 / Accepted: 17 February 2013 / Published: 26 February 2013
(This article belongs to the Special Issue Redox Signaling in Biology and Patho-Biology)

Abstract

:
Hepatitis C virus (HCV) infects approximately 3% of the world’s population. Currently licensed treatment of HCV chronic infection with pegylated-interferon-α and ribavirin, is not fully effective against all HCV genotypes and is associated to severe side effects. Thus, development of novel therapeutics and identification of new targets for treatment of HCV infection is necessary. Current opinion is orienting to target antiviral drug discovery to the host cell pathways on which the virus relies, instead of against viral structures. Many intracellular signaling pathways manipulated by HCV for its own replication are finely regulated by the oxido-reductive (redox) state of the host cell. At the same time, HCV induces oxidative stress that has been found to affect both virus replication as well as progression and severity of HCV infection. A dual role, positive or negative, for the host cell oxidized conditions on HCV replication has been reported so far. This review examines current information about the effect of oxidative stress on HCV life cycle and the main redox-regulated intracellular pathways activated during HCV infection and involved in its replication.

1. Introduction

According to evaluations of the World Health Organization (WHO), hepatitis C virus (HCV) currently infects at least 130 million people worldwide, which is about 3% of the global population [1]. Following acute HCV infection, a chronic state is established in as many as 80% of infected individuals. Although many subjects carrying the virus remain asymptomatic, chronicity is often accompanied by altered liver function and progressive liver disease that culminates in cirrhosis and hepatocellular carcinoma in up to 20% of chronically infected individuals [2].
HCV is a positive-sense single stranded RNA virus of the Flaviviridae family. The HCV genome is about 9.6 kb in length and consists of the 5′ untranslated region (UTR), the structural (core, E1 and E2) and non-structural (p7, NS2, NS3, NS4 A/B, NS5 A/B) proteins and the 3′ UTR. HCV binds to the CD81 and/or SR-BI receptors, enters the cell through clathrin-mediated endocytosis and is uncoated, releasing the RNA genome into the cytoplasm. The positive-sense RNA first serves as a messenger RNA for translation of a single large polyprotein that is proteolytically cleaved, by host and viral peptidases, to generate individual proteins. Subsequently, HCV RNA acts as a template for viral genome replication, mediated by virus encoded RNA-dependent RNA polymerase, NS5B, in cooperation with NS5A and other viral proteins. The virus replication complex is localized to the membranous web that is formed by modified membranes of the ER and Golgi. After encapsidation of the viral genome, virions are exported through the host secretory pathway. For recent reviews on the HCV life cycle see [3,4].
Current therapy of chronic hepatitis C is based on a combination of PEG-interferon (PEG-IFN)-α and ribavirin. This combination therapy is not fully effective, as it achieves a sustained virological response (SVR) in less than half of treated patients with HCV genotype 1 and about 80% of those with genotype 2, and causes severe side effects [57]. Recent approval, in Europe and in the United States, of the two new drugs targeting the HCV protease (telaprevir and boceprevir) produced an increased SVR rate to 70%–80% in triple combination therapy with PEG-IFN-α and ribavirin [8]. However triple combination treatment has limitations in partial non-responders and null responders to a prior course of PEG-IFN-α and ribavirin [912] and has more severe side effects.
From the above, a pressing need exists for the development of new and alternative therapeutic strategies to combat HCV infection. To circumvent the onset of drug resistance, a major obstacle to therapeutic success, it seems to be particularly important to approach viral infection therapy by considering the interactions between virus and host cells. In fact, current opinion is to target antiviral drug discovery to the host cell pathways on which the virus relies, instead of against viral structures, in order to decrease the likelihood of acquiring drug resistance [13,14].
It is known that many cellular proteins involved in a wide range of signaling pathways that are manipulated by HCV, to promote its own replication [1518], are influenced by the intracellular redox state [1921]. The redox state of a cell results from an equilibrium between the production of reactive oxygen/nitrogen species (ROS/RNS) and the anti-oxidative defenses (antioxidant molecules and enzymes) [21]. An imbalance towards pro-oxidative conditions is referred to as oxidative stress. The intracellular thiols are an important class of antioxidant molecules, with glutathione (GSH) being the most abundant. In cells, GSH can be free or bound to proteins. Free GSH exists mainly in its reduced form (GSH), which can be transformed to the oxidized gluthatione (GSSG) during oxidative stress and it can be reverted to the reduced form by the enzyme GSH reductase. The ratio between GSH/GSSG is considered an index of the antioxidant capacity of the cell [22]. The reduced form of glutathione is an important radical scavenger that directly neutralizes a variety of reactive molecules (such as superoxide anion and hydroxyl radicals) [23]. In addition to its role as a redox buffer, GSH can form mixed disulfides (GSSR) with protein thiols (S-glutathionylation), thus protecting cysteine residues of proteins from irreversible oxidation events. Moreover, S-glutathionylation may alter protein function and is actually considered to be involved in signal transduction [24,25]. It has been demonstrated that infections by several viruses, including HCV, are frequently characterized by host-cell redox changes, characteristic of oxidative stress, that affect the efficacy of viral replication [2633].
This review presents a summary of the current knowledge about redox regulation of HCV life cycle and discusses some of main redox regulated intracellular pathways activated during HCV infection and involved in its replication.

2. HCV Infection Induces Oxidative Stress

Increased oxidative stress is a hallmark of HCV infection (reviewed in [28,29,33]). Clinical studies have shown that HCV patients have elevated levels of reactive aldehydes produced by lipid peroxidation, such as malondialdehyde and 4-hydroxy-2-nonenal, in their serum, peripheral blood mononuclear cells and liver [34,35]. Liver levels of 8-hydroxydeoxyguanosine, a marker of oxidative damage, are also elevated [34,36]. Furthermore, an increased glutathione turnover in the liver, blood and lymphatic system has been suggested by the observation of decreased ratio between GSH/GSSG [34,3739].
Usually, viruses induce an inflammatory response at the site of infection, where release of ROS by immune cells acts as non-specific toxins, kills pathogens and injuries adjacent cells. In the case of persistent infections, chronic inflammation results in overwhelming production of ROS that induces tissues damage and predispose to disease induction [40]. However, although inflammation is an important source of ROS during chronic infection [41,42], HCV infected patients with minimum or no liver disease showed markers of oxidative stress [43] and HCV transgenic mice exhibited signs of increased oxidative damage in the absence of inflammation [44]. These evidences suggest that HCV may directly promote oxidative stress in hepatocytes (Figure 1). Actually, numerous in vitro studies have shown that not only HCV replication, but also expression of the HCV structural and non-structural proteins, directly act as inducers of oxidative stress, mainly through mitochondrial dysfunction of hepatocytes and endoplasmic reticulum (ER) stress [4552]. In particular, direct interaction of the structural HCV core protein with mitochondria decreases the mitochondrial NADPH levels, reduces the activity of the electron transport complex I and increases generation of ROS [4547]; the non-structural proteins NS5A [49,50], NS4B [50] and the structural glycoproteins E1 and E2 [51] induce ROS production by ER stress. Recently, it has been demonstrated that hepatocyte NAD(P)H oxidase (Nox) proteins 1 and 4 represent another prominent source of ROS during HCV infection [53,54], as they were found significantly elevated both in HCV infected cells and in the liver of HCV-infected patients [53]. In addition, HCV replication may induce impairment of host cell antioxidant defenses [5558], and directly alter the endogenous levels of GSH [53,56,59] (Table 1). Although in vivo studies reported a decreased hepatic GSH content in HCV chronically infected patients [34,38,40], however studies in vitro showed conflicting results about the effect of HCV on intracellular GSH as well as on the enzymes regulating GSH homeostasis [55,6062]. In this regard, Roe and collaborators [59] reported a significant raise of GSSG in HCV infected cells; conversely, increased GSH concentration was demonstrated by de Mochel et al. [53] using same in vitro infection system. Analogous contradiction has been described for the effect of HCV on the activity of NF-E2-related factor 2 (Nrf2), a central regulator of the enzymes of glutathione homeostasis [63,64]. Recently, two studies, performed by Burdette et al. [60] and Ivanov et al. [61] reported activation of Nrf2 pathway, respectively in HCV infected Huh-7 cells and in cells expressing the individual viral proteins. Conversely, Carvajal-Yepes and collaborators [62] demonstrated suppression of this pathway upon transfection of cells with HCV clones. The apparently disparate effects on GSH by HCV may be due to different experimental systems used (HCV transfected versus infected cell cultures). However, it has to be considered that viruses producing chronic infections, such as HIV and HCV, cause significant changes of GSH levels only after chronic infection is well established [27,65]. So, it has been proposed [61] that data by Burdette et al. [60] and by Ivanov et al. [61] may be representative of the acute phase of HCV infection, when the cells enhance the expression of antioxidant genes to protect themselves against the virus induced oxidative stress. In contrast, data from the study of Carvajal-Yepes [62] could be related to chronic virus infection. Further studies about the changes of GSH levels during HCV acute and chronic infections are required to clarify this point.

3. Oxidative Stress Affects HCV Replication

HCV related oxidative stress is involved in the development of liver injury, and in the progression of liver disease towards fibrosis, cirrhosis and HCC [28,29,66,67]. Therefore, antioxidants have been proposed as new potential treatment of HCV patients. For example, in a phase I clinical trial testing a combination of seven different antioxidants, 44% of the patients had normalization of liver enzymes levels and 36.1% of the treated patients had histological improvement [68]. Alongside this positive effect of the anti-oxidants, however, it remains unclear how modulation of oxidative stress can affect HCV replication. In general, treatment with drugs that amplify the effects of endogenous ROS, generated during normal cellular metabolism or in response to HCV, has suppressive effect on HCV replication. Using replicon cells (that are human hepatoma cell lines constitutively replicating HCV RNA but not producing infectious virus particles), Choi and collaborators found that H2O2 and t-butylhydroperoxide (t-BOOH), which generates low levels of intracellular ROS, reduced HCV RNA by a mechanism involving the disruption of HCV replication complex on cell membranes [69,70]. Similar suppression of intracellular HCV was obtained upon ROS induction with glucose oxidase (GO) transfection in cell lines [71]. In addition, modulation of intracellular glutathione, by arsenic trioxide or by BSO, negatively affected HCV replication in vitro, either in replicon cells as well as in cells infected with HCV infectious clone (JFH1-infected cells) [71,72]. According to the above-reported negative effect of oxidative stress on HCV in vitro, some anti-oxidants (such as vitamins A, vitamin E and resveratrol) have been shown to enhance HCV replication in replicon model, with yet unknown mechanisms [73,74]. On the other side, opposite effect of some antioxidant treatments on HCV has also been reported. For examples, N-acetylcysteine (NAC) [75], pyrrolidine dithiocarbamate (PDTC) [76,77], sylimarin [78,79], naringerin [8082], quercetin [82,83], curcumin [84] and also the acetylsalicylic acid (ASA) [77], have been found to decrease HCV replication and HCV proteins level in vitro. In addition, transient transfection of the anti-oxidant enzyme, Mn-superoxide dismutase, in replicon cells lowered viral replication [76]. The steps in virus life cycle targeted by these antioxidants and the mechanisms related to their action are mostly unknown, although in some cases they have been described. As examples, PDTC is reported to act through inhibition of MAPK pathways [76]; sylimarin inhibits virus entry and infectious virions production into culture supernatants through the inhibition of microsomal triglyceride transfer protein (MTP) activity and apolipoprotein B secretion [78]; naringerin blocks virus assembly, partly by the activation of PPARα, and consequent decrease in VLDL production [80]. Few clinical studies describing the effect of antioxidant supplementation on HCV titer are available and have yielded so far inconsistent results [68,85,86]. This lack of a clear correlation between oxidative stress and HCV replication remains an open question and may in part be explained by the fact that the same antioxidant molecules can display pro- or anti-oxidant functions, depending on their own oxidative status, which in turn reflects the specific redox potential of the microenvironment [87,88]. In support of these considerations, Wang and collaborators found that pre-treatment of mice with NAC, a GSH precursor, prevents liver from acute ethanol-induced damage, via counteracting ethanol-induced oxidative stress; whereas, when administered after ethanol, NAC behaves as a pro-oxidant and exacerbates acute ethanol-induced liver damage [89]. Furthermore, we have shown that resveratrol (RV) can decrease GSH in a different virus and cell system, thus functioning as oxidant molecule [90]. This fact has been explained considering that, although RV can quench reactive free radicals by donating hydrogen atoms, this process generates also phenoxyl radicals that can oxidize GSH to GS·. Moreover, oxidation of the RV-phenoxyl radical produces an RV-quinone form, which can alkylate GSH, further diminishing the intracellular concentrations of free GSH [91]. A similar behaviour has been shown by vitamin E (tocopherol), a chain-breaking antioxidants that interferes with radical chain propagation by trapping radicals. During its metabolism, vitamin E is converted into tocopheroxyl radical with low reactivity that needs to be removed by a secondary antioxidant, such as ascorbic acid (vitamin C). If the removal of tocopheroxyl radical by a secondary antioxidant is delayed, it may promote lipid peroxidation, functioning as pro-oxidant [92].
Therefore antioxidants are considered intriguing molecules to be used as antivirals. However most of the studies so far available, evaluate the antiviral potential of antioxidant molecules, without concurrent measure of their effect on the cellular redox state, which could be the reason behind reported controversies in literature.

4. Redox-Regulated Signaling Pathways: MAPK and PI3K/Akt Signaling Pathways Are Critical Controllers of HCV Replication

An abundance of scientific literature exists demonstrating that oxidative stress influences several signaling pathways [25,9395], among which the two mostly affected, MAPK (Mitogen Activated Protein Kinase) and PI3K/Akt pathways, have a pivotal role on replication of several viruses, such as influenza A virus [9698], HIV [99], human cytomegalovirus [100], varicella-zoster [101] and also HCV [15,17]. MAP kinases (comprising the three best characterized members ERK, JNK and p38 MAPK) and Akt are activated by general induction of intracellular ROS [93,94,102] and are inhibited by the antioxidants [17,76,103]. Moreover, exposure of cells to exogenous H2O2, that mimic oxidative stress, also leads to activation of MAPK and PI3K/Akt pathways in multiple cell types (vascular smooth muscle cell, cardiac myocytes, A431 cell, CHO cell line, hepatocytes) [94,104109]. Different mechanisms have been proposed for activation of the two pathways in response to ROS [19,25,9395] (Figure 1). In normal redox conditions, thioredoxin (TRX) association with ASK1, the MAKKK for JNK and p38, maintains the pathways inactive [110,111]; upon oxidative stress thiol modification of TRX, promotes its dissociation from ASK1 [93,94] thus activating p38 and JNK [112]. A second proposed mechanism involves ROS induced oligomerization of glutathione-S-transferase Pi (GSTp), resulting in its dissociation from the complex with JNK and the subsequent activation of the pathway [113]. A further mechanism for activation of MAPK and Akt is by degrading the protein tyrosine phosphatases, PtPases, that maintain the pathways in inactive state [19,25]. An example of this latter is oxidation and consequent inactivation of PTEN, a negative regulator of Akt that results in PI3K/Akt activation [114,115].
It is known that during its life cycle, HCV activates MAPK and Akt pathways that have a role in the pathogenesis of inflammation, fibrosis, HCC [116120] and in viral immune evasion strategies occurring during HCV infection [17,121]. With regard to the role of MAPK cascades in HCV replication, virus binding to the CD81 receptor has been shown to activate the Raf/MEK/ERK pathway that was necessary for post-entry events [122]. Consistently, it has recently been proposed that the blockade of ERK cascade with the inhibitor of MEK 1/2 (U0126) prevents HCV assembly and virion release [16,123]. Furthermore, the inhibitors of JNK and p38MAPK (SP600125 and SB203580 respectively) blocked HCV replication in replicon systems, suggesting that these kinases are tightly involved in positive control of HCV life cycle [76]. However, some authors suggested a negative role of ERK1/2 pathway on HCV in vitro, as activation of this pathway was shown to suppress viral replication in replicon system, whereas its inhibition promoted HCV replication [124126]. The discrepancies on the effects of the diverse cascades of the MAPK pathway (ERK, JNK and p38) on HCV replication, are probably due to the different in vitro system used by the authors, therefore further research would need to elucidate this issue.
With regard to the role of PI3K/Akt pathway several reports have shown that it is activated by HCV [15,119,120] and that this event is critical for viral replication, as suggested by studies with small interfering RNA (siRNA) [127] or compounds that specifically block Akt activity [18,84,127]. Among these latter, triciribine, that blocks Akt activity without affecting its upstream activators, was found to inhibit both basal HCV replication, as well as that enhanced by epithelial growth factor (EGF) [127]. Conversely, little is known about the steps of HCV replication cycle influenced by PI3K/Akt. Recently, Liu et al.[18] demonstrated that HCV rapidly and transiently activated Akt, to enhance its entry into the cells, via the interaction between HCV E2 envelope protein and its co-receptors, CD81 and claudin-1. Interestingly, PI3K/Akt pathway positively affects HCV replication by modulating lipid metabolism; the mechanisms involve enhanced expression of SREBP-1 (sterol regulatory element binding protein-1) [84], an important transcription factor of lipogenic gene expression, and inhibition of the AMPK (AMP-activated protein kinase) activity [128], that results in stimulation of cholesterol and triglyceride synthesis. Increasing evidences show that HCV is critically dependent on cellular lipids throughout its life cycle [4,129,130]. In fact viral RNA replication complexes localize to lipid rafts (membranous structures derived from the ER) rich in sphingomyelin and cholesterol [131]; moreover a crucial role for lipid droplets (LDs) in HCV assembly has also been demonstrated [132].
On the basis of all the above reported data it could be reasonably speculated that PI3K/Akt pathway, through modulation of hepatic lipid metabolism, could have a role both in early and late stages of HCV life cycle. However, further investigations would need to disclose the exact steps of HCV life cycle influenced by Akt pathway.

5. Conclusions

Although HCV induces oxidative stress in host cells, all current revised literature points to a dual role of the host cell oxidized conditions on HCV replication. Several studies are in favor of a positive control of HCV replication by oxidative stress; however, it cannot be ruled out that virus induced oxidative stress can negatively modulate its own replication. This could be a mechanism to control the interaction at equilibrium between virus and host cell, which is the basis for establishment of chronicity. Although much has yet to be disclosed, the redox-mediated host cell mechanisms so far identified, together with those that will be further recognized, could uncover new effective approaches for HCV treatment.

Acknowledgments

This work was partially supported by Istituto Superiore di Sanità intramural fundings, by the Italian Ministry of Instruction, Universities and Research (PON Project, International FIRB and PRIN 2010–2011) and Ateneo grants.

Conflict of Interest

The authors declare no conflict of interest.

References

  1. Lavanchy, D. The global burden of hepatitis C. Liver Int 2009, 29, 74–81. [Google Scholar]
  2. Liang, T.J.; Heller, T. Pathogenesis of hepatitis C-associated hepatocellular carcinoma. Gastroenterology 2004, 127, S62–S71. [Google Scholar]
  3. Moradpour, D.; Penin, F.; Rice, C.M. Replication of hepatitis C virus. Nat. Rev. Microbiol 2007, 5, 453–463. [Google Scholar]
  4. Herker, E.; Ott, M. Unique ties between hepatitis C virus replication and intracellular lipids. Trends Endocrinol. Metab 2011, 22, 241–248. [Google Scholar]
  5. Manns, M.P.; Wedemeyer, H.; Cornberg, M. Treating viral hepatitis C: Efficacy, side effects, and complications. Gut 2006, 55, 1350–1359. [Google Scholar]
  6. Hadziyannis, S.J.; Sette, H., Jr; Morgan, T.R.; Balan, V.; Diago, M.; Marcellin, P.; Ramadori, G.; Bodenheimer, H., Jr; Bernstein, D.; Rizzetto, M.; et al. Peginterferon-alpha2a and ribavirin combination therapy in chronic hepatitis C: A randomized study of treatment duration and ribavirin dose. Ann. Intern. Med. 2004, 140, 346–355. [Google Scholar]
  7. Manns, M.P.; McHutchison, J.G.; Gordon, S.C.; Rustgi, V.K.; Shiffman, M.; Reindollar, R.; Goodman, Z.D.; Koury, K.; Ling, M.; Albrecht, J.K. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: A randomised trial. Lancet 2001, 358, 958–965. [Google Scholar]
  8. Welsch, C.; Jesudian, A.; Zeuzem, S.; Jacobson, I. New direct-acting antiviral agents for the treatment of hepatitis C virus infection and perspectives. Gut 2012, 61, i36–i46. [Google Scholar]
  9. Jacobson, I.M.; McHutchison, J.G.; Dusheiko, G.; Di Bisceglie, A.M.; Reddy, K.R.; Bzowej, N.H.; Marcellin, P.; Muir, A.J.; Ferenci, P.; Flisiak, R.; et al. Telaprevir for previously untreated chronic hepatitis C virus infection. N. Engl. J. Med 2011, 364, 2405–2416. [Google Scholar]
  10. Poordad, F.; McCone, J., Jr; Bacon, B.R.; Bruno, S.; Manns, M.P.; Sulkowski, M.S.; Jacobson, I.M.; Reddy, K.R.; Goodman, Z.D.; Boparai, N.; et al. Boceprevir for untreated chronic HCV genotype 1 infection. N. Engl. J. Med. 2011, 364, 1195–1206. [Google Scholar]
  11. Sherman, K.E.; Flamm, S.L.; Afdhal, N.H.; Nelson, D.R.; Sulkowski, M.S.; Everson, G.T.; Fried, M.W.; Adler, M.; Reesink, H.W.; Martin, M.; et al. Response-guided telaprevir combination treatment for hepatitis C virus infection. N. Engl. J. Med 2011, 365, 1014–1024. [Google Scholar]
  12. Sarrazin, C.; Hézode, C.; Zeuzem, S.; Pawlotsky, J.M. Antiviral strategies in hepatitis C virus infection. J. Hepatol 2012, 56, S88–S100. [Google Scholar]
  13. Bühler, S.; Bartenschlager, R. New targets for antiviral therapy of chronic hepatitis C. Liver Int 2012, 32, 9–16. [Google Scholar]
  14. Gallay, P.A. Cyclophilin inhibitors: A novel class of promising host-targeting anti-HCV agents. Immunol. Res 2012, 52, 200–210. [Google Scholar]
  15. Bode, J.G.; Brenndörfer, E.D.; Karthe, J.; Häussinger, D. Interplay between host cell and hepatitis C virus in regulating viral replication. Biol. Chem 2009, 390, 1013–1032. [Google Scholar]
  16. Menzel, N.; Fischl, W.; Hueging, K.; Bankwitz, D.; Frentzen, A.; Haid, S.; Gentzsch, J.; Kaderali, L.; Bartenschlager, R.; Pietschmann, T. MAP-kinase regulated cytosolic phospholipase A2 activity is essential for production of infectious hepatitis C virus particles. PLoS Pathog 2012, 8, e1002829. [Google Scholar]
  17. Mannová, P.; Beretta, L. Activation of the N-Ras-PI3K-Akt-mTOR pathway by hepatitis C virus: Control of cell survival and viral replication. J. Virol 2005, 79, 8742–8749. [Google Scholar]
  18. Liu, Z.; Tian, Y.; Machida, K.; Lai, M.M.; Luo, G.; Foung, S.K.; Ou, J.H. Transient activation of the PI3K-AKT pathway by HCV to enhance viral entry. J. Biol. Chem 2012, 287, 41922–41930. [Google Scholar]
  19. Cross, J.V.; Templeton, D.J. Regulation of signal transduction through protein cysteine oxidation. Antioxid. Redox Signal 2006, 8, 1819–1827. [Google Scholar]
  20. Filomeni, G.; Rotilio, G.; Ciriolo, M.R. Disulfide relays and phosphorylative cascades: Partners in redox-mediated signaling pathways. Cell Death Differ 2005, 12, 1555–1563. [Google Scholar]
  21. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.D.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell. Biol 2007, 39, 44–84. [Google Scholar]
  22. Meister, A.; Anderson, M.E. Glutathione. Ann. Rev. Biochem 1983, 52, 711–760. [Google Scholar]
  23. Forman, H.J.; Dickinson, D.A. Oxidative signaling and glutathione synthesis. Biofactors 2003, 17, 1–12. [Google Scholar]
  24. Dalle-Donne, I.; Milzani, A.; Gagliano, N.; Colombo, R.; Giustarini, D.; Rossi, R. Molecular mechanisms and potential clinical significance of S-glutathionylation. Antioxid. Redox Signal 2008, 10, 445–473. [Google Scholar]
  25. Pastore, A.; Piemonte, F. S-Glutathionylation signaling in cell biology: Progress and prospects. Eur. J. Pharm. Sci 2012, 46, 279–292. [Google Scholar]
  26. Nencioni, L.; Sgarbanti, R.; Amatore, D.; Checconi, P.; Celestino, I.; Limongi, D.; Anticoli, S.; Palamara, A.T.; Garaci, E. Intracellular redox signaling as therapeutic target for novel antiviral strategy. Curr. Pharm. Des 2011, 17, 3898–3904. [Google Scholar]
  27. Fraternale, A.; Paoletti, M.F.; Casabianca, A.; Oiry, J.; Clayette, P.; Vogel, J.U.; Cynatl, J., Jr.; Palamara, A.T.; Sgarbanti, R.; Garaci, E.; et al. Antiviral and immunomodulatory properties of new pro-glutathione (GSH) molecules. Curr. Med. Chem. 2006, 13, 1749–1755. [Google Scholar]
  28. Choi, J. Oxidative stress, endogenous antioxidants, alcohol, and hepatitis C: Pathogenic interactions and therapeutic considerations. Free Radic. Biol. Med 2012, 52, 1135–1150. [Google Scholar]
  29. Choi, J.; Ou, J.H. Mechanisms of liver injury. III. Oxidative stress in the pathogenesis of hepatitis C virus. Am. J. Physiol. Gastrointest. Liver Physiol 2006, 290, G847–G851. [Google Scholar]
  30. Sgarbanti, R.; Nencioni, L.; Amatore, D.; Coluccio, P.; Fraternale, A.; Sale, P.; Mammola, C.L.; Carpino, G.; Gaudio, E.; Magnani, M.; et al. Redox regulation of the influenza hemagglutinin maturation process: A new cell-mediated strategy for anti-influenza therapy. Antioxid. Redox Signal 2011, 15, 593–606. [Google Scholar]
  31. Kavouras, J.H.; Prandovszky, E.; Valyi-Nagy, K.; Kovacs, S.K.; Tiwari, V.; Kovacs, M.; Shukla, D.; Valyi-Nagy, T. Herpes simplex virus type 1 infection induces oxidative stress and the release of bioactive lipid peroxidation by-products in mouse P19N neural cell cultures. J. Neurovirol 2007, 13, 416–425. [Google Scholar]
  32. Peterhans, E. Oxidants and antioxidants in viral diseases: Disease mechanisms and metabolic regulation. J. Nutr 1997, 127, S962–S965. [Google Scholar]
  33. Seronello, S.; Sheikh, M.Y.; Choi, J. Redox regulation of hepatitis C in nonalcoholic and alcoholic liver. Free Radic. Biol. Med 2007, 43, 869–882. [Google Scholar]
  34. Mahmood, S.; Kawanaka, M.; Kamei, A.; Izumi, A.; Nakata, K.; Niiyama, G.; Ikeda, H.; Hanano, S.; Suehiro, M.; Togawa, K.; et al. Immunohistochemical evaluation of oxidative stress markers in chronic hepatitis C. Antioxid. Redox Signal 2004, 6, 19–24. [Google Scholar]
  35. Romero, M.J.; Bosch-Morell, F.; Romero, B.; Rodrigo, J.M.; Serra, M.A.; Romero, F.J. Serum malondialdehyde: Possible use for the clinical management of chronic hepatitis C patients. Free Radic. Biol. Med 1998, 25, 993–997. [Google Scholar]
  36. Fujita, N.; Sugimoto, R.; Ma, N.; Tanaka, H.; Iwasa, M.; Kobayashi, Y.; Kawanishi, S.; Watanabe, S.; Kaito, M.; Takei, Y. Comparison of hepatic oxidative DNA damage in patients with chronic hepatitis B and C. J. Viral Hepat 2008, 15, 498–507. [Google Scholar]
  37. Farias, M.S.; Budni, P.; Ribeiro, C.M.; Parisotto, E.B.; Santos, C.E.; Dias, J.F.; Dalmarco, E.M.; Fröde, T.S.; Pedrosa, R.C.; Wilhelm Filho, D. Antioxidant supplementation attenuates oxidative stress in chronic hepatitis C patients. Gastroenterol. Hepatol 2012, 35, 386–394. [Google Scholar]
  38. Lin, C.C.; Liu, W.H.; Wang, Z.H.; Yin, M.C. Vitamins B status and antioxidative defense in patients with chronic hepatitis B or hepatitis C virus infection. Eur. J. Nutr 2011, 50, 499–506. [Google Scholar]
  39. Barbaro, G.; Di Lorenzo, G.; Soldini, M.; Parrotto, S.; Bellomo, G.; Belloni, G.; Grisorio, B.; Barbarini, G. Hepatic glutathione deficiency in chronic hepatitis C: Quantitative evaluation in patients who are HIV positive and HIV negative and correlations with plasmatic and lymphocytic concentrations and with the activity of the liver disease. Am. J. Gastroenterol 1996, 91, 2569–2573. [Google Scholar]
  40. Loguercio, C.; Federico, A. Oxidative stress in viral and alcoholic hepatitis. Free Radic. Biol. Med 2003, 34, 1–10. [Google Scholar]
  41. Hosomura, N.; Kono, H.; Tsuchiya, M.; Ishii, K.; Ogiku, M.; Matsuda, M.; Fujii, H. HCV-related proteins activate Kupffer cells isolated from human liver tissues. Dig. Dis. Sci 2011, 56, 1057–1064. [Google Scholar]
  42. Thoren, F.; Romero, A.; Lindh, M.; Dahlgren, C.; Hellstrand, K. A hepatitis C virus-encoded, nonstructural protein (NS3) triggers dysfunction and apoptosis in lymphocytes: Role of NADPH oxidase-derived oxygen radicals. J. Leukoc. Biol 2004, 76, 1180–1186. [Google Scholar]
  43. Vendemiale, G.; Grattagliano, I.; Portincasa, P.; Serviddio, G.; Palasciamo, G.; Altomare, E. Oxidative stress in symptom-free HCV carriers: Relation with ALT flare-up. Eur. J. Clin. Invest 2001, 31, 54–63. [Google Scholar]
  44. Moriya, K.; Nakagawa, K.; Santa, T.; Shintani, Y.; Fujie, H.; Miyoshi, H.; Tsutsumi, T.; Miyazawa, T.; Ishibashi, K.; Horie, T.; et al. Oxidative stress in the absence of inflammation in a mouse model for hepatitis C virus-associated hepatocarcinogenesis. Cancer Res 2001, 61, 4365–4370. [Google Scholar]
  45. Korenaga, M.; Wang, T.; Li, Y.; Showalter, L.A.; Chan, T.; Sun, J.; Weinman, S.A. Hepatitis C virus core protein inhibits mitochondrial electron transport and increases reactive oxygen species (ROS) production. J. Biol. Chem 2005, 280, 37481–37488. [Google Scholar]
  46. Li, Y.; Boehning, D.F.; Qian, T.; Popov, V.L.; Weinman, S.A. Hepatitis C virus core protein increases mitochondrial ROS production by stimulation of Ca2+ uniporter activity. FASEB J 2007, 21, 2474–2485. [Google Scholar]
  47. Machida, K.; Cheng, K.T.; Lai, C.K.; Jeng, K.S.; Sung, V.M.; Lai, M.M. Hepatitis C virus triggers mitochondrial permeability transition with production of reactive oxygen species, leading to DNA damage and STAT3 activation. J. Virol 2006, 80, 7199–7207. [Google Scholar]
  48. Tardif, K.D.; Waris, G.; Siddiqui, A. Hepatitis C virus, ER stress, and oxidative stress. Trends Microbiol 2005, 13, 159–163. [Google Scholar]
  49. Robinson, L.C.; Marchant, J.S. Enhanced Ca2+ leak from ER Ca2+ stores induced by hepatitis C NS5A protein. Biochem. Biophys. Res. Commun 2008, 368, 593–599. [Google Scholar]
  50. Tong, W.Y.; Nagano-Fujii, M.; Hidajat, R.; Deng, L.; Takigawa, Y.; Hotta, H. Physical interaction between hepatitis C virus NS4B protein and CREB-RP/ATF6beta. Biochem. Biophys. Res. Commun 2002, 299, 366–372. [Google Scholar]
  51. Chan, S.W.; Egan, P.A. Hepatitis C virus envelope proteins regulate CHOP via induction of the unfolded protein response. FASEB J 2005, 19, 1510–1512. [Google Scholar]
  52. Santos, C.X.; Tanaka, L.Y.; Wosniak, J.; Laurindo, F.R. Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: Roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid. Redox. Signal 2009, 11, 2409–2427. [Google Scholar]
  53. De Mochel, N.S.R.; Seronello, S.; Wang, S.H. Hepatocyte NAD(P)H oxidases as an endogenous source of reactive oxygen species during hepatitis C virus infection. Hepatology 2010, 52, 47–59. [Google Scholar]
  54. Boudreau, H.E.; Emerson, S.U.; Korzeniowska, A.; Jendrysik, M.A.; Leto, T.L. Hepatitis C virus (HCV) proteins induce NADPH oxidase 4 expression in a transforming growth factor beta-dependent manner: A new contributor to HCV-induced oxidative stress. J. Virol 2009, 83, 12934–12946. [Google Scholar]
  55. Diamond, D.L.; Syder, A.J.; Jacobs, J.M.; Sorensen, C.M.; Walters, K.A.; Proll, S.C.; McDermott, J.E.; Gritsenko, M.A.; Zhang, Q.; Zhao, R.; et al. Temporal proteome and lipidome profiles reveal hepatitis C virus-associated reprogramming of hepatocellular metabolism and bioenergetics. PLoS Pathog 2010, 6, e1000719. [Google Scholar]
  56. Abdalla, M.Y.; Ahmad, I.M.; Spitz, D.R.; Schmidt, W.N.; Britigan, B.E. Hepatitis C virus-core and non structural proteins lead to different effects on cellular antioxidant defenses. J. Med. Virol 2005, 76, 489–497. [Google Scholar]
  57. Qadri, I.; Iwahashi, M.; Capasso, J.M.; Hopken, M.W.; Flores, S.; Schaack, J.; Simon, F.R. 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 2004, 378, 919–928. [Google Scholar]
  58. Li, K.; Prow, T.; Lemon, S.M.; Beard, M.R. Cellular response to conditional expression of hepatitis C virus core protein in Huh7 cultured human hepatoma cells. Hepatology 2002, 35, 1237–1246. [Google Scholar]
  59. Roe, B.; Kensicki, E.; Mohney, R.; Hall, W.W. Metabolomic profile of hepatitis C virus-infected hepatocytes. PLoS One 2011, 6, e23641. [Google Scholar]
  60. Burdette, D.; Olivarez, M.; Waris, G. Activation of transcription factor Nrf2 by hepatitis C virus induces the cell-survival pathway. J. Gen. Virol 2010, 91, 681–690. [Google Scholar]
  61. Ivanov, A.V.; Smirnova, O.A.; Ivanova, O.N.; Masalova, O.V.; Kochetkov, S.N.; Isaguliants, M.G. Hepatitis C virus proteins activate NRF2/ARE pathway by distinct ROS-dependent and independent mechanisms in HUH7 cells. PLoS One 2011, 6, e24957. [Google Scholar]
  62. Carvajal-Yepes, M.; Himmelsbach, K.; Schaedler, S.; Ploen, D.; Krause, J.; Ludwig, L.; Weiss, T.; Klingel, K.; Hildt, E. Hepatitis C virus impairs the induction of cytoprotective Nrf2 target genes by delocalization of small Maf proteins. J. Biol. Chem 2011, 286, 8941–8951. [Google Scholar]
  63. Motohashi, H.; Yamamoto, M. Nrf2-Keap1 defines a physiologically important stress response mechanism. Trends Mol. Med 2004, 10, 549–557. [Google Scholar]
  64. Rushmore, T.H.; Morton, M.R.; Pickett, C.B. The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity. J. Biol. Chem 1991, 266, 11632–11639. [Google Scholar]
  65. Garaci, E.; Palamara, A.T.; Ciriolo, M.R.; D’Agostini, C.; Abdel-Latif, M.S.; Aquaro, S.; Lafavia, E.; Rotilio, G. Intracellular GSH content and HIV replication in human macrophages. J. Leukoc. Biol 1997, 62, 54–59. [Google Scholar]
  66. Clement, S.; Pascarella, S.; Negro, F. Hepatitis C Virus infection: Molecular pathways to steatosis, insulin resistance and oxidative stress. Viruses 2009, 1, 126–143. [Google Scholar]
  67. Tanikawa, K.; Torimura, T. Studies on oxidative stress in liver diseases: Important future trends in liver research. Med. Mol. Morphol 2006, 39, 22–27. [Google Scholar]
  68. Melhem, A.; Stern, M.; Shibolet, O.; Israeli, E.; Ackerman, Z.; Pappo, O.; Hemed, N.; Rowe, M.; Ohana, H.; Zabrecky, G.; et al. Treatment of chronic hepatitis C virus infection via antioxidants: Results of a phase I clinical trial. J. Clin. Gastroenterol 2005, 39, 737–742. [Google Scholar]
  69. Choi, J.; Lee, K.J.; Zheng, Y.; Yamaga, A.K.; Lai, M.M.; Ou, J.H. Reactive oxygen species suppress hepatitis C virus RNA replication in human hepatoma cells. Hepatology 2004, 39, 81–89. [Google Scholar]
  70. Choi, J.; Forman, H.J.; Ou, J.H.; Lai, M.M.; Seronello, S.; Nandipati, A. Redox modulation of the hepatitis C virus replication complex is calcium dependent. Free Radic. Biol. Med 2006, 41, 1488–1498. [Google Scholar]
  71. Seronello, S.; Ito, C.; Wakita, T.; Choi, J. Ethanol enhances hepatitis C virus replication through lipid metabolism and elevated NADH/NAD+. J. Biol. Chem 2010, 285, 845–854. [Google Scholar]
  72. Kuroki, M.; Ariumi, Y.; Ikeda, M.; Dansako, H.; Wakita, T.; Kato, N. Arsenic trioxide inhibits hepatitis C virus RNA replication through modulation of the glutathione redox system and oxidative stress. J. Virol. 2009, 83, 2338–2348. [Google Scholar]
  73. Yano, M.; Ikeda, M.; Abe, K.; Dansako, H.; Ohkoshi, S.; Aoyagi, Y.; Kato, N. Comprehensive analysis of the effects of ordinary nutrients on hepatitis C virus RNA replication in cell culture. Antimicrob. Agents Chemother 2007, 51, 2016–2027. [Google Scholar]
  74. Nakamura, M.; Saito, H.; Ikeda, M.; Hokari, R.; Kato, N.; Hibi, T.; Miura, S. An antioxidant resveratrol significantly enhanced replication of hepatitis C virus. World J. Gastroenterol 2010, 16, 184–192. [Google Scholar]
  75. McCartney, E.M.; Beard, M.R. Impact of alcohol on hepatitis C virus replication and interferon signaling. World J. Gastroenterol 2010, 16, 1337–1343. [Google Scholar]
  76. Waris, G.; Turkson, J.; Hassanein, T.; Siddiqui, A. Hepatitis C virus (HCV) constitutively activates STAT-3 via oxidative stress: Role of STAT-3 in HCV replication. J. Virol 2005, 79, 1569–1580. [Google Scholar]
  77. Rivas-Estilla, A.M.; Bryan-Marrugo, O.L.; Trujillo-Murillo, K.; Pérez-Ibave, D.; Charles-Niño, C.; Pedroza-Roldan, C.; Ríos-Ibarra, C.; Ramírez-Valles, E.; Ortiz-López, R.; Islas-Carbajal, M.C.; et al. Cu/Zn superoxide dismutase (SOD1) induction is implicated in the antioxidative and antiviral activity of acetylsalicylic acid in HCV-expressing cells. Am. J. Physiol. Gastrointest. Liver Physiol 2012, 302, G1264–G1273. [Google Scholar]
  78. Wagoner, J.; Negash, A.; Kane, O.J.; Martinez, L.E.; Nahmias, Y.; Bourne, N.; Owen, D.M.; Grove, J.; Brimacombe, C.; McKeating, J.A.; et al. Multiple effects of silymarin on the hepatitis C virus life cycle. Hepatology 2010, 51, 1912–1921. [Google Scholar]
  79. Wagoner, J.; Morishima, C.; Graf, T.N.; Oberlies, N.H.; Teissier, E.; Pécheur, E.I.; Tavis, J.E.; Polyak, S.J. Differential in vitro effects of intravenous versus oral formulations of silibinin on the HCV life cycle and inflammation. PLoS One 2011, 6, e16464. [Google Scholar]
  80. Goldwasser, J.; Cohen, P.Y.; Lin, W.; Kitsberg, D.; Balaguer, P.; Polyak, S.J.; Chung, R.T.; Yarmush, M.L.; Nahmias, Y. Naringenin inhibits the assembly and long-term production of infectious hepatitis C virus particles through a PPAR-mediated mechanism. J. Hepatol 2011, 55, 963–971. [Google Scholar]
  81. Nahmias, Y.; Goldwasser, J.; Casali, M.; van Poll, D.; Wakita, T.; Chung, R.T.; Yarmush, M.L. Apolipoprotein B-dependent hepatitis C virus secretion is inhibited by the grapefruit flavonoid naringenin. Hepatology 2008, 47, 1437–1445. [Google Scholar]
  82. Khachatoorian, R.; Arumugaswami, V.; Raychaudhuri, S.; Yeh, G.K.; Maloney, E.M.; Wang, J.; Dasgupta, A.; French, S.W. Divergent antiviral effects of bioflavonoids on the hepatitis C virus life cycle. Virology 2012, 433, 346–355. [Google Scholar]
  83. Bachmetov, L.; Gal-Tanamy, M.; Shapira, A.; Vorobeychik, M.; Giterman-Galam, T.; Sathiyamoorthy, P.; Golan-Goldhirsh, A.; Benhar, I.; Tur-Kaspa, R.; Zemel, R. Suppression of hepatitis C virus by the flavonoid quercetin is mediated by inhibition of NS3 protease activity. J. Viral. Hepat 2012, 19, e81–e88. [Google Scholar]
  84. Kim, K.; Kim, K.H.; Kim, H.Y.; Cho, H.K.; Sakamoto, N.; Cheong, J. Curcumin inhibits hepatitis C virus replication via suppressing the Akt-SREBP-1 pathway. FEBS Lett 2010, 584, 707–712. [Google Scholar]
  85. Guedj, J.; Dahari, H.; Pohl, R.T.; Ferenci, P.; Perelson, A.S. Understanding silibinin’s modes of action against HCV using viral kinetic modeling. J. Hepatol 2012, 56, 1019–1024. [Google Scholar]
  86. Falasca, K.; Ucciferri, C.; Mancino, P.; Vitacolonna, E.; De Tullio, D.; Pizzigallo, E.; Conti, P.; Vecchiet, J. Treatment with silybin-vitamin E-phospholipid complex in patients with hepatitis C infection. J. Med. Virol 2008, 80, 1900–1906. [Google Scholar]
  87. Powis, G. Free radical formation by antitumor quinones. Free Radic. Biol. Med 1989, 6, 63–101. [Google Scholar]
  88. Monks, T.J.; Hanzlik, R.P.; Cohen, G.M.; Ross, D.; Graham, D.G. Quinone chemistry and toxicity. Toxicol. Appl. Pharmacol 1992, 112, 2–16. [Google Scholar]
  89. Wang, A.L.; Wang, J.P.; Wang, H.; Chen, Y.H.; Zhao, L.; Wang, L.S.; Wei, W.; Xu, D.X. A dual effect of N-acetylcysteine on acute ethanol-induced liver damage in mice. Hepatol. Res 2006, 34, 199–206. [Google Scholar]
  90. Palamara, A.T.; Nencioni, L.; Aquilano, K; De Chiara, G.; Hernandez, L.; Cozzolino, F.; Ciriolo, M.R.; Garaci, E. Inhibition of influenza A virus replication by resveratrol. J. Infect. Dis. 2005, 191, 1719–1729. [Google Scholar]
  91. Burkitt, M.J.; Duncan, J. Effects of trans-resveratrol on copper-dependent hydroxyl-radical formation and DNA damage: Evidence for hydroxyl-radical scavenging and a novel, glutathione-sparing mechanism of action. Archiv. Biochem. Biophys 2000, 381, 253–263. [Google Scholar]
  92. Chang, Y.C.; Chuang, L.M. The role of oxidative stress in the pathogenesis of type 2 diabetes: From molecular mechanism to clinical implication. Am. J. Transl. Res 2010, 2, 316–331. [Google Scholar]
  93. Torres, M.; Forman, H.J. Redox signaling and the MAP kinase pathways. Biofactors 2003, 17, 287–296. [Google Scholar]
  94. McCubrey, J.A.; Lahair, M.M.; Franklin, R.A. Reactive oxygen species-induced activation of the MAP kinase signaling pathways. Antioxid. Redox. Signal 2006, 8, 1775–1789. [Google Scholar]
  95. Leslie, N.R. The redox regulation of PI 3-kinase-dependent signaling. Antioxid. Redox. Signal 2006, 8, 1765–1774. [Google Scholar]
  96. Nencioni, L.; de Chiara, G.; Sgarbanti, R.; Amatore, D.; Aquilano, K.; Marcocci, M.E.; Serafino, A.; Torcia, M.; Cozzolino, F.; Ciriolo, M.R.; et al. Bcl-2 expression and p38MAPK activity in cells infected with influenza A virus: Impact on virally induced apoptosis and viral replication. J. Biol. Chem 2009, 284, 16004–16015. [Google Scholar]
  97. Nencioni, L.; Sgarbanti, R.; De Chiara, G.; Garaci, E.; Palamara, A.T. Influenza virus and redox mediated cell signaling: A complex network of virus/host interaction. New Microbiol 2007, 30, 367–375. [Google Scholar]
  98. Shin, Y.K.; Liu, Q.; Tikoo, S.K.; Babiuk, L.A.; Zhou, Y. Effect of the phosphatidylinositol 3-kinase/Akt pathway on influenza A virus propagation. J. Gen. Virol 2007, 88, 942–950. [Google Scholar]
  99. Muthumani, K.; Wadsworth, S.A.; Dayes, N.S.; Hwang, D.S.; Choo, A.Y.; Abeysinghe, H.R.; Siekierka, J.J.; Weiner, D.B. Suppression of HIV-1 viral replication and cellular pathogenesis by a novel p38/JNK kinase inhibitor. AIDS 2004, 18, 739–748. [Google Scholar]
  100. Yu, Y.; Alwine, J.C. Human cytomegalovirus major immediate-early proteins and simian virus 40 large T antigen can inhibit apoptosis through activation of the phosphatidylinositide 3′-OH kinase pathway and the cellular kinase Akt. J. Virol 2002, 76, 3731–3738. [Google Scholar]
  101. Rahaus, M.; Desloges, N.; Wolff, M.H. Varicella-zoster virus requires a functional PI3K/Akt/GSK-3alpha/beta signaling cascade for efficient replication. Cell Signal 2007, 19, 312–320. [Google Scholar]
  102. Lahair, M.M.; Howe, C.J.; Rodriguez-Mora, O.; McCubrey, J.A.; Franklin, R.A. Molecular pathways leading to oxidative stress-induced phosphorylation of Akt. Antioxid. Redox Signal 2006, 8, 1749–1756. [Google Scholar]
  103. Waris, G.; Siddiqui, A. Hepatitis C virus stimulates the expression of cyclooxygenase-2 via oxidative stress: Role of prostaglandin E2 in RNA replication. J. Virol. 2005, 79, 9725–9734. [Google Scholar]
  104. Blanc, A.; Pandey, N.R.; Srivastava, A.K. Distinct roles of Ca2+, calmodulin, and protein kinase C in H2O2-induced activation of ERK1/2, p38 MAPK, and protein kinase B signaling in vascular smooth muscle cells. Antioxid. Redox Signal 2004, 6, 353–366. [Google Scholar]
  105. Dossumbekova, A.; Berdyshev, E.V.; Gorshkova, I.; Shao, Z.; Li, C.; Long, P.; Joshi, A.; Natarajan, V.; Vanden Hoek, T.L. Akt activates NOS3 and separately restores barrier integrity in H2O2-stressed human cardiac microvascular endothelium. Am. J. Physiol. Heart Circ. Physiol 2008, 295, H2417–H2426. [Google Scholar]
  106. Mehdi, M.Z.; Pandey, N.R.; Pandey, S.K.; Srivastava, A.K. H2O2-induced phosphorylation of ERK1/2 and PKB requires tyrosine kinase activity of insulin receptor and c-Src. Antioxid. Redox Signal 2005, 7, 1014–1020. [Google Scholar]
  107. Conde de la Rosa, L.; Schoemaker, M.H.; Vrenken, T.E.; Buist-Homan, M.; Havinga, R.; Jansen, P.L.; Moshage, H. Superoxide anions and hydrogen peroxide induce hepatocyte death by different mechanisms: Involvement of JNK and ERK MAP kinases. J. Hepatol 2006, 44, 918–929. [Google Scholar]
  108. Aikawa, R.; Komuro, I.; Yamazaki, T.; Zou, Y.; Kudoh, S.; Tanaka, M.; Shiojima, I.; Hiroi, Y.; Yazaki, Y. Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J. Clin. Invest 1997, 100, 1813–1821. [Google Scholar]
  109. Lee, S.R.; Kwon, K.S.; Kim, S.R.; Rhee, S.G. Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor. J. Biol. Chem 1998, 273, 15366–15372. [Google Scholar]
  110. Liu, H.; Nishitoh, H.; Ichijo, H.; Kyriakis, J.M. Activation of apoptosis signal-regulating kinase 1 (ASK1) by tumor necrosis factor receptor-associated factor 2 requires prior dissociation of the ASK1 inhibitor thioredoxin. Mol. Cell Biol 2000, 20, 2198–2208. [Google Scholar]
  111. Saitoh, M.; Nishitoh, H.; Fujii, M.; Takeda, K.; Tobiume, K.; Sawada, Y.; Kawabata, M.; Miyazono, K.; Ichijo, H. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. EMBO J 1998, 17, 2596–2606. [Google Scholar]
  112. Matsukawa, J.; Matsuzawa, A.; Takeda, K.; Ichijo, H. The ASK1-MAP kinase cascades in mammalian stress response. J. Biochem 2004, 136, 261–265. [Google Scholar]
  113. Adler, V.; Yin, Z.; Fuchs, S.Y.; Benezra, M.; Rosario, L.; Tew, K.D.; Pincus, M.R.; Sardana, M.; Henderson, C.J.; Wolf, C.R.; et al. Regulation of JNK signaling by GSTp. EMBO J 1999, 18, 1321–1334. [Google Scholar]
  114. Lee, S.R.; Yang, K.S.; Kwon, J.; Lee, C.; Jeong, W.; Rhee, S.G. Reversible inactivation of the tumor suppressor PTEN by H2O2. J. Biol. Chem 2002, 277, 20336–20342. [Google Scholar]
  115. Leslie, N.R.; Bennett, D.; Lindsay, Y.E.; Stewart, H.; Gray, A.; Downes, C.P. Redox regulation of PI 3-kinase signalling via inactivation of PTEN. EMBO J 2003, 22, 5501–5510. [Google Scholar]
  116. Mazzocca, A.; Sciammetta, S.C.; Carloni, V.; Cosmi, L.; Annunziato, F.; Harada, T.; Abrignani, S.; Pinzani, M. Binding of hepatitis C virus envelope protein E2 to CD81 up-regulates matrix metalloproteinase-2 in human hepatic stellate cells. J. Biol. Chem 2005, 280, 11329–11339. [Google Scholar]
  117. Ming-Ju, H.; Yih-Shou, H.; Tzy-Yen, C.; Hui-Ling, C. Hepatitis C virus E2 protein induce reactive oxygen species (ROS)-related fibrogenesis in the HSC-T6 hepatic stellate cell line. J. Cell Biochem 2011, 112, 233–243. [Google Scholar]
  118. Lin, W.; Tsai, W.L.; Shao, R.X.; Wu, G.; Peng, L.F.; Barlow, L.L.; Chung, W.J.; Zhang, L.; Zhao, H.; Jang, J.Y.; et al. Hepatitis C virus regulates transforming growth factor beta1 production through the generation of reactive oxygen species in a nuclear factor kappaB-dependent manner. Gastroenterology 2010, 138, 2509–2518. [Google Scholar]
  119. Street, A.; Macdonald, A.; McCormick, C.; Harris, M. Hepatitis C virus NS5A-mediated activation of phosphoinositide 3-kinase results in stabilization of cellular beta-catenin and stimulation of beta-catenin-responsive transcription. J. Virol 2005, 79, 5006–5016. [Google Scholar]
  120. Street, A.; Macdonald, A.; Crowder, K.; Harris, M. The Hepatitis C virus NS5A protein activates a phosphoinositide 3-kinase-dependent survival signaling cascade. J. Biol. Chem 2004, 279, 12232–12241. [Google Scholar]
  121. Frelin, L.; Brenndörfer, E.D.; Ahlén, G.; Weiland, M.; Hultgren, C.; Alheim, M.; Glaumann, H.; Rozell, B.; Milich, D.R.; Bode, J.G.; et al. The hepatitis C virus and immune evasion: Non-structural 3/4A transgenic mice are resistant to lethal tumour necrosis factor alpha mediated liver disease. Gut 2006, 55, 1475–1483. [Google Scholar]
  122. Brazzoli, M.; Bianchi, A.; Filippini, S.; Weiner, A.; Zhu, Q.; Pizza, M.; Crotta, S. CD81 is a central regulator of cellular events required for hepatitis C virus infection of human hepatocytes. J. Virol 2008, 82, 8316–8329. [Google Scholar]
  123. Pei, R.; Chen, H.; Lu, L.; Zhu, W.; Beckebaum, S.; Cicinnati, V.; Lu, M.; Chen, X. Hepatitis C virus infection induces the expression of amphiregulin, a factor related to the activation of cellular survival pathways and required for efficient viral assembly. J. Gen. Virol 2011, 92, 2237–2248. [Google Scholar]
  124. Murata, T.; Hijikata, M.; Shimotohno, K. Enhancement of internal ribosome entry site-mediated translation and replication of hepatitis C virus by PD98059. Virology 2005, 340, 105–115. [Google Scholar]
  125. Huang, Y.; Chen, X.C.; Konduri, M.; Fomina, N.; Lu, J.; Jin, L.; Kolykhalov, A.; Tan, S.L. Mechanistic link between the anti-HCV effect of interferon gamma and control of viral replication by a Ras-MAPK signaling cascade. Hepatology 2006, 43, 81–90. [Google Scholar]
  126. Yano, M.; Ikeda, M.; Abe, K.; Kawai, Y.; Kuroki, M.; Mori, K.; Dansako, H.; Ariumi, Y.; Ohkoshi, S.; Aoyagi, Y.; et al. Oxidative stress induces anti-hepatitis C virus status via the activation of extracellular signal-regulated kinase. Hepatology 2009, 50, 678–688. [Google Scholar]
  127. Brenndörfer, E.D.; Karthe, J.; Frelin, L.; Cebula, P.; Erhardt, A.; Schulteam Esch, J.; Hengel, H.; Bartenschlager, R.; Sällberg, M.; Häussinger, D.; et al. Nonstructural 3/4A protease of hepatitis C virus activates epithelial growth factor-induced signal transduction by cleavage of the T-cell protein tyrosine phosphatase. Hepatology 2009, 49, 1810–1820. [Google Scholar]
  128. Mankouri, J.; Tedbury, P.R.; Gretton, S.; Hughes, M.E.; Griffin, S.D.; Dallas, M.L.; Green, K.A.; Hardie, D.G.; Peers, C.; Harris, M. Enhanced hepatitis C virus genome replication and lipid accumulation mediated by inhibition of AMP-activated protein kinase. Proc. Natl. Acad. Sci. USA 2010, 107, 11549–11554. [Google Scholar]
  129. Fukuhara, T.; Matsuura, Y. Role of miR-122 and lipid metabolism in HCV infection. J. Gastroenterol 2012, 48, 169–176. [Google Scholar]
  130. Syed, G.H.; Amako, Y.; Siddiqui, A. Hepatitis C virus hijacks host lipid metabolism. Trends Endocrinol. Metab 2010, 21, 33–40. [Google Scholar]
  131. Aizaki, H.; Lee, K.J.; Sung, V.M.; Ishiko, H.; Lai, M.M. Characterization of the hepatitis C virus RNA replication complex associated with lipid rafts. Virology 2004, 324, 450–461. [Google Scholar]
  132. Miyanari, Y.; Atsuzawa, K.; Usuda, N.; Watashi, K.; Hishiki, T.; Zayas, M.; Bartenschlager, R.; Wakita, T.; Hijikata, M.; Shimotohno, K. The lipid droplet is an important organelle for hepatitis C virus production. Nat. Cell. Biol 2007, 9, 1089–1097. [Google Scholar]
Figure 1. Schematic representation of ROS sources during Hepatitis C virus (HCV) infection and redox mechanisms for activation of MAPKs and PI3K/Akt.
Figure 1. Schematic representation of ROS sources during Hepatitis C virus (HCV) infection and redox mechanisms for activation of MAPKs and PI3K/Akt.
Ijms 14 04705f1
Table 1. Mechanisms of oxidative stress induction by HCV infection and viral proteins involved.
Table 1. Mechanisms of oxidative stress induction by HCV infection and viral proteins involved.
Source of ROS/RNSHCV proteinsReferences
Transcriptional up-regulation of iNOScore, NS3[47]
Activation of Nox2 in PBMCsNS3[41]
GSH depletionCore[56]
Increased production of mitochondrial ROS by the electron transport chainCore[45]
Activation of Nox2 in Kupffer cellsHCV[29,40]
Endoplasmic reticulum stress (ER stress)NS4B, NS5A, E1, E2[4951]
Increase of proinflammatory cytokinesHCV, core[54]
Activation of Nox1 and 4 proteins in hepatocytesHCV[53,54]

Share and Cite

MDPI and ACS Style

Ruggieri, A.; Anticoli, S.; Nencioni, L.; Sgarbanti, R.; Garaci, E.; Palamara, A.T. Interplay between Hepatitis C Virus and Redox Cell Signaling. Int. J. Mol. Sci. 2013, 14, 4705-4721. https://doi.org/10.3390/ijms14034705

AMA Style

Ruggieri A, Anticoli S, Nencioni L, Sgarbanti R, Garaci E, Palamara AT. Interplay between Hepatitis C Virus and Redox Cell Signaling. International Journal of Molecular Sciences. 2013; 14(3):4705-4721. https://doi.org/10.3390/ijms14034705

Chicago/Turabian Style

Ruggieri, Anna, Simona Anticoli, Lucia Nencioni, Rossella Sgarbanti, Enrico Garaci, and Anna Teresa Palamara. 2013. "Interplay between Hepatitis C Virus and Redox Cell Signaling" International Journal of Molecular Sciences 14, no. 3: 4705-4721. https://doi.org/10.3390/ijms14034705

Article Metrics

Back to TopTop