Abstract
Hepatocellular carcinoma (HCC) is the most common liver cancer and a leading cause of cancer-related mortality in the world. Hepatitis C virus (HCV) is a major etiologic agent of HCC. A majority of HCV infections lead to chronic infection that can progress to cirrhosis and eventually, HCC and liver failure. A common pathogenic feature present in HCV infection, and other conditions leading to HCC, is oxidative stress. HCV directly increases superoxide and H2O2 formation in hepatocytes by elevating Nox protein expression and sensitizing mitochondria to reactive oxygen species generation while decreasing glutathione. Nitric oxide synthesis and hepatic iron are also elevated. Furthermore, activation of phagocytic NADPH oxidase 2 (Nox2) of host immune cells is likely to exacerbate oxidative stress in HCV-infected patients. Key mechanisms of HCC include: genome instability, epigenetic regulation, inflammation with chronic tissue injury and sustained cell proliferation, and modulation of cell growth and death. Oxidative stress, or Nox proteins, plays various roles in these mechanisms. Nox proteins also function in hepatic fibrosis, which commonly precedes HCC, and Nox4 elevation by HCV was mediated by transforming growth factor beta. This review summarizes mechanisms of oncogenesis by HCV, highlighting the role of oxidative stress and hepatic Nox enzymes in HCC.
Keywords: cancer, HCV, inflammation, NADPH oxidase, reactive oxygen species
I. Introduction
Oxygen is essential for the life of aerobic organisms. During aerobic respiration, O2 is reduced to H2O through a series of electron transfer reactions occurring in mitochondria that drive ATP synthesis. In addition, reactive oxygen species (ROS) are formed. While ROS are often associated with toxicity, not all biological ROS generation are harmful, and ROS participate in specific, reversible biochemical reactions in redox signaling. For example, reversible thiol oxidation of protein tyrosine phosphatases and peroxiredoxins by H2O2 that produces cysteine sulfenic acid intermediates is well-characterized (1). Even an over-production of reactive species is not necessarily harmful and could confer some benefit to the organism producing them. A clear example would be the burst of superoxide anion (O2−˙) produced by NADPH oxidase 2 (Nox2) in phagocytes during inflammation, which is critical for the destruction of microbes within phagosomes; absence of a functional Nox2 protein complex leads to chronic and frequent infections, as well as dysregulated inflammation in patients with chronic granulomatous disease (2). Nevertheless, aberrant levels of ROS that overwhelm the body’s antioxidant defenses can result in pathogenic changes and tissue injury, and a shift in redox status is associated with diverse disease conditions.
Hepatitis C virus (HCV) is a major etiologic agent of severe liver diseases, including cancer. HCV perturbs the host redox status by increasing production of ROS and reactive nitrogen species (RNS), and oxidative stress has emerged as a common pathogenic feature in numerous in vitro and in vivo studies on HCV. Several sources of ROS contribute to hepatic oxidative stress during HCV infection, including hepatocyte and non-hepatocyte sources, and oxidative stress is likely to contribute to HCV-associated liver cancer through multiple mechanisms. This review summarizes the mechanisms of hepatocarcinogenesis by HCV, highlighting the role of oxidative stress and hepatic Nox enzymes in hepatocellular carcinoma (HCC).
A. Hepatocellular carcinoma
HCC is a malignant epithelial tumor of the parenchymal liver cell that accounts for 70 – 85 % of primary liver cancers (3). A majority of HCC cases occur in developing countries, with the highest prevalence in sub-Saharan Africa and Eastern Asia. HCC rates are low in developed countries, with the exception of Japan. HCC is the second leading cause of cancer-related mortality in the world (4). Disease prognosis is poor; five year survival rates of ~11 % have been reported, even in developed countries (5–7). Unlike many cancers that have declined over the years, HCC is still on the rise (8). In the U.S., the incidence rates of HCC tripled in both men and women from 1995 – 1997 to 2005 – 2007; death rates for liver cancer also increased (9). Increases in HCC incidence rates were most prominent among Hispanic, black, and white middle-aged men between 2000 – 2005 (9, 10). Higher overall HCC incidence and mortality rates were also found among Asians/Pacific Islanders compared to other groups tested, indicating significant health disparities among populations (9). A ten-fold increase in the prevalence of HCC was reported among U.S. military veterans with HCV infection from 1996 to 2006 (11). In 2009, 22,620 new cases of liver and intrahepatic bile duct cancers and 18,160 deaths were estimated in the U.S. (8). Worldwide, an estimated 748,300 new liver cancer cases and 695,900 liver cancer-related deaths occurred in 2008 (12).
Risk factors for HCC include viral hepatitis, alcohol abuse, aflatoxin exposure, obesity, as well as nonalcoholic fatty liver disease (4). Among these, viral hepatitis is the most important risk factor for HCC; together, hepatitis B virus (HBV) and HCV are responsible for 78 % of HCC worldwide (13). While the highest rates of HCC are found in areas where HBV is endemic, the increasing incidence of HCC, particularly in developed countries, has been largely attributed to the prevalence of HCV infection. For example, HCV infection is found in approximately 80% of HCC cases in Japan and about half of HCC cases in the U.S. (14, 15). Unlike HBV, for which there is a vaccine, there is no vaccine that can prevent the spread of HCV infection, and this is likely contributing to the rising incidence of HCV-associated HCC. The odds ratio for developing HCC is 11.5 among HCV-infected individuals (16).
B. HCV induces HCC
HCV is an enveloped, positive sense RNA virus of the Flaviviridae family that was discovered in 1989 (17). HCV is transmitted through blood, and common risk factors for HCV infection include having received blood, blood products, or organs prior to 1992, injection drug use, birth to an infected mother, sex with an infected person, and occupational exposure. A majority of HCV infected individuals do not clear the virus and become chronically infected. It is estimated that approximately 170 million individuals are chronically infected with HCV worldwide, including 4 million in the U.S. (18, 19). Baby boomers, or people who were born between 1945 to 1965, are more likely to have hepatitis C than others; a National Health and Nutrition Examination Survey estimated that among the non-institutionalized, civilian U.S. population who had chronic HCV infection, approximately 66 % were baby boomers who would now have been living with chronic hepatitis C for several decades (20). It has also been estimated that approximately 50 % of individuals with chronic HCV infections are undiagnosed in the U.S. (20).
Chronic HCV infection occurs in about 80 % of HCV-infected individuals, which increases the risk of developing severe liver diseases. About 20 – 30 % of chronic hepatitis C patients develop cirrhosis in 20 – 30 years, and 4 – 7 % of these individuals progress to HCC and end stage liver disease each year (21). Extrahepatic manifestations, such as lymphoproliferative disorders, have also been associated with chronic HCV infection (22). HCV is the leading cause for liver transplantation in the U.S., and HCV-related deaths rose by 123 % between 1994 and 2005 in the U.S. (23). By 2020, the proportion of infected and untreated patients with cirrhosis is estimated to increase from 16 to 32 %, hepatic decompensation by 106 %, HCC by 81 %, and liver-related deaths by 180 % (24).
The previous standard of care for HCV infection, consisting of pegylated interferon (IFN) alpha and ribavirin, achieved sustained virological response (SVR) in 50 – 60 % of individuals undergoing treatment and produced severe side effects (25). SVR was as low as 30 % in patients with genotype 1, the most prevalent HCV genotype, and a high viral load. Recently developed direct-acting antivirals that target specific HCV factors, such as NS3/4A protease and NS5A protein, have shown strong promise, and these drugs are rapidly changing how HCV infection is managed clinically (26). SVR following anti-HCV therapy generally decreases the rate of HCC, supporting the importance of eradicating the virus to decrease HCC (27, 28). However, HCC can still form even after achieving SVR in subjects with pretreatment cirrhosis, elevated alanine aminotransferase (ALT) and steatosis, or prior HCC diagnosis, necessitating continued screening for HCC and development of therapy targeting cancer formation (29–31). In addition, as mentioned, many individuals with chronic hepatitis C do not know that they are infected; by the time they are diagnosed, they have already progressed to an advanced disease state, which complicates antiviral therapy.
Development of various in vitro and in vivo HCV models has greatly increased our understanding of HCV and its mechanism of pathogenesis (32–35). Briefly, the HCV genome is 9.6 kb in length and contains one large open reading frame that codes for structural and nonstructural (NS) proteins. The open reading frame is flanked by untranslated regions (UTR) that function in viral gene expression and replication (Figure 1A). Structural genes code for core protein, which forms the viral capsid, and the envelope glycoproteins, E1 and E2. p7 and NS genes function in viral replication, polyprotein processing, and/or morphogenesis and viral release. HCV enters the cell through receptor-mediated endocytosis, which is facilitated by a series of molecular interactions that involve glycosaminoglycans (GAG), cluster of differentiation 81 (CD81), occludin (OCLN), claudin-1 (CLDN1), low-density lipoprotein receptors (LDLR), and scavenger receptor class B type I (SR-BI) (36) (Figure 1B). Upon entry, the positive-sense RNA genome is released in the cytoplasm, where translation of HCV RNA, initiated using an internal ribosomal entry site located in the 5’ UTR, produces a large polyprotein. The polyprotein is then cleaved by host signal peptidase and NS2 and NS3/4A viral proteases to generate individual protein products, including NS5B, the viral replicase. The alternate reading frame of the core protein-coding sequence can also support the production of frameshift/alternate reading frame protein (F/ARFP), among others (37, 38). Then, NS5B and various proteins that comprise the replication complex replicate the genome through generation of a negative strand RNA. The newly synthesized positive sense RNA genome is then encapsidated, and virions are exported through the host secretory pathway (Figure 1B). Detailed review of the HCV replication cycle may be found elsewhere (36).
Fig. 1. HCV genome and replication cycle.
(A) Genome organization of HCV. SP indicate serine protease. (B) Replication cycle of HCV.
Many studies have demonstrated that HCV is capable of inducing tumors in vivo. Transgenic mice that express HCV genotype 1 core or core plus E1 and E2 under the control of eukaryotic elongation factor 1α, a cytomegalovirus promoter, or in the liver under the control of an HBV promoter, spontaneously develop hepatic tumors, including HCC (35, 39, 40). HCC also forms spontaneously in transgenic mice expressing the entire open reading frame of genotype 1b HCV in the liver, or only NS5A (41, 42). Other studies reported an increased tumor rate in HCV core or NS5A transgenic mice treated with CCl4, ethanol, diethylnitrosamine (DEN)/Phenobarbital, or co-expressing HBV X protein (39, 43–45). Spontaneous hepatic tumors in HCV transgenic mouse models generally occurred in the absence of hepatitis or cirrhosis, pointing to the direct oncogenic effects of HCV (35, 46, 47). This conclusion was supported by observations that core, NS3, NS4A, NS4B, NS5A, or polyprotein expression also transformed cells in vitro, either alone or in cooperation with Ras (48–53). Likewise, different HCV proteins accelerated tumor growth in nude mice (51, 54, 55).
II. HCV induces oxidative stress
A. Iron overload and nitric oxide generation
Chronic hepatitis C is characterized by pronounced increases in lipid peroxidation products (46, 56–60). Iron overload, independent of hemochromatosis gene mutations, has been reported among hepatitis C patients (56, 59, 61–63). Hepatocytes and liver macrophages act as a substantial reservoir of iron, where 20 – 30% is bound to ferritin and hemosiderin (64). Decreased expression of hepcidin, a negative regulator of iron absorption, by virus-induced oxidative stress and HCV F/ARFP, may underlie elevated iron deposition (65, 66). Furthermore, IFN therapy decreased 4-hydroxy-2-nonenal protein adducts and hepatic iron deposits in responders (59).
HCV also increases the generation of nitric oxide (·NO), an RNS, by transcriptional up-regulation of inducible nitric oxide synthase (iNOS) through a mechanism involving c-Jun (39, 67). Increased levels of nitrotyrosine were detected in liver biopsies from patients with chronic viral hepatitis, including hepatitis C, that co-localized with iNOS (68). These findings suggest increased nitrative stress during natural HCV infection
B. Mitochondrial ROS
In addition, HCV increases ROS generation. Specifically, elevated levels of superoxide and/or H2O2 have been demonstrated with HCV gene expression or infectious HCV in hepatocytes in vitro, as well as in hepatitis C patient liver (34, 69–74). HCV infection has been associated with significant mitochondrial abnormalities, and the mitochondrial electron transport chain was identified as a source of ROS in various HCV protein-expressing cells (Figure 2A). HCV-associated increases in oxidative stress or ROS were mediated by mitochondrial Ca2+ uptake and could be inhibited by diphenyleneiodonium (DPI), determined by monitoring ethidium generation from dihydroethidium, dihydrodichlorocarboxyfluorescein (DCF) diacetate fluorescence in isolated mitochondria and hepatocytes, and MitoSox fluorescence assay, (46, 60, 69, 75–79). HCV core localized to the mitochondria where Ca2+ uptake increased through the action of a mitochondrial Ca2+ uniporter; this then sensitized the organelle to mitochondrial membrane permeability transition (MPT) and increased superoxide generation at complex I (60, 69, 76, 78, 79) (Figure 2A). Mitochondrial glutathione (GSH), NADPH, and ATP synthesis decreased in the presence of HCV proteins (69, 76). Additionally, HCV core decreased the levels of prohibitin, a mitochondrial protein chaperon, as well as cytochrome c oxidase activity (80). Japanese fulminant hepatitis-1 (JFH1) strain, a genomic replicon, and HCV core likewise sensitized mitochondria to membrane depolarization or MPT induction by thapsigargin, which elevates cytosolic Ca2+ concentration by triggering a passive release of Ca2+ from the endoplasmic reticulum (ER) (73, 78). Exogenous peroxide, which has been shown to elevate cytosolic calcium content in hepatocytes, produced similar effects as thapsigargin on mitochondria in the presence of HCV (73, 78, 81). A spontaneous decrease in mitochondrial membrane potential in Raji cells, which could be inhibited by Bcl2, N-acetylcysteine (NAC), and an inhibitor of iNOS, was also noted with HCV (77).
Fig. 2. Mechanisms of HCV-induced oxidative/nitrosative stress and hepatic Nox proteins.
(A) Sources of ROS and RNS during HCV infection. HCV increases superoxide (O2−˙) and H2O2 formation in hepatocytes by Nox enzymes, Complex I of mitochondria, and CYP enzymes in combination with alcohol. Superoxide generation by phagocytic Nox2 from activated Kupffer cells and decreased antioxidant defense will exacerbate oxidative stress. Increased nitric oxide (·NO) generation through iNOS and subsequent formation of peroxynitrite (ONOO−) from nitric oxide and superoxide, and hydroxyl radical (HO·) from H2O2 and Fe2+ are also shown. Oxidative stress will contribute to pathogenesis of HCC by DNA, lipid, and protein damage as well as modification of cell signaling. (B) Schematics of Nox1, Nox2, and Nox4 protein complexes. (C) Nox4 protein domain map. FAD and NADPH binding sites are marked.
Indeed, mitochondrial Ca2+ loading can increase mitochondrial ROS generation in hepatocytes by augmenting NAD(P)H synthesis by Ca2+-sensitive mitochondrial dehydrogenases (82–84). Furthermore, excessive Ca2+ accumulation in mitochondria may trigger MPT, leading to osmotic swelling, breakage of the outer membrane, and release of cytochrome c, that has been associated with increased mitochondrial ROS (69, 85). Other mechanisms whereby Ca2+ loading has been shown to accelerate mitochondrial ROS generation involve nitric oxide production and cardiolipin peroxidation (84). In addition, HCV has been shown to induce ER stress (75). These findings suggest that HCV increases mitochondrial ROS generation secondary to the mobilization of intracellular Ca2+, although the detailed mechanism awaits further investigation.
C. Phagocytic Nox2
The Nox family enzymes consist of Nox and Dual oxidase (Duox) proteins (Nox1, 2, 3, 4, and 5 and Duox 1 and 2) that catalyze the transfer of electrons from NADPH to O2 to generate superoxide and H2O2 (88). Unlike many other sources of ROS, such as mitochondria that produce ROS as a byproduct of aerobic metabolism, the primary function of Nox/Duox proteins is superoxide and H2O2 generation. Among the Nox proteins, the best characterized is Nox2 of the immune cells (2). Upon stimulation, Nox2 complex consisting of gp91phox, p47phox, p67phox, p22phox, Ras-related C3 botulinum toxin substrate (Rac), and p40phox, assembles with translocation of the cytosolic components, p47phox, p67phox, Rac, and p40phox (2) (Figure 2B). This transocation leads to the respiratory burst, characterized by increased oxygen consumption and direct production of superoxide.
HCV has been shown to activate the phagocytic Nox2 (89, 90). Incubating human monocytes isolated from healthy blood donors with recombinant HCV NS3 protein, but not core, NS4, or NS5 proteins, resulted in superoxide production (89). NS3-stimulated superoxide generation was associated with phosphorylation and translocation of p47phox and required Ca2+ influx, tyrosine kinase, and stress-activated protein kinase 2 p38 (SAPK2/p38) activities. This transient increase in superoxide generation observed in human monocytes was followed by an inhibition of phorbol 12-myristate 13-acetate-stimulated respiratory burst by NS3 (89). NS3 also induced a prolonged generation of ROS in mononuclear and polymorphonuclear phagocytes, that was inhibited by DPI and histamine (89, 90), and HCV proteins induced a prolonged activation of Kupffer cells in HCV-infected liver (91). In addition, HCV peptide (C5A), derived from the NS5A membrane anchor domain, could function as a chemoattractant and activated human phagocytic leukocytes, leading to massive phagocyte infiltration (92). The activated phagocytes in turn triggered dysfunction and/or apoptosis of T cells and natural killer cells, suggesting the role of HCV-associated oxidative stress in immune dysfunction (89, 90). Recently, it was also shown that Nox2 could be involved in the myeloid-derived suppressor cell-dependent T cell suppression in HCV infected patients (93). The liver contains 80 % of the host mononuclear phagocytic system (94). These reports suggest a significant role of phagocytic Nox2 in hepatic oxidative stress in chronic hepatitis C (Figure 2A).
D. Hepatocyte Nox1 and Nox4
In addition, hepatocytes, which are the primary site of HCV replication, express Nox family proteins (95–98). Recently, HCV was shown to increase Nox1 and Nox4 mRNA and protein levels in hepatocytes (71, 72) (Figure 2A, 2B). Nox1 and Nox4 served as a prominent source of ROS in hepatocytes producing infectious HCV of genotypes 2a and 1b or expressing HCV proteins (71, 72). Dominant negative Nox4, containing a C-terminus deletion, Nox4 or Nox1 knockdown, and DPI significantly decreased HCV-induced superoxide and/or H2O2 production in hepatocytes, compared to controls (71, 72). In fact, DPI is an inhibitor of flavoproteins that inhibits Nox enzymes as well as complex I of the electron transport chain (86, 87). Similar increases in p22phox, p67phox, NOXA1, and NOXA2 mRNAs were also found (72). Nox4 and Nox1 proteins and Nox activity were significantly elevated in HCV-infected human liver (72). Likewise, human hepatoma (Huh7) cells inoculated with genotype 3 HCV-infected patient serum showed an increase in Nox4 mRNA (99). HCV core protein was sufficient to increase Nox4 levels in vitro (71).
Nox4 is thought to be constitutively active and regulated at the level of gene expression (see (100) for a review). Transforming growth factor beta (TGFβ), a fibrogenic cytokine, upregulates Nox4 expression in hepatocytes and hepatic stellate cells (HSCs) (96–98, 101). HCV activated human and murine Nox4 promoter activity in hepatocytes; furthermore, HCV increased levels of TGFβ in these cells and induced Nox4 transcription through TGFβ (71, 72). Increased levels of TGFβ have also been detected in patients with chronic hepatitis C (71, 72, 102). HCV was shown to induce TGFβ transcription through a redox-sensitive mechanism involving nuclear factor kappa B (NFκB) in hepatocytes, and through NFκB, activator protein 1 (AP-1), specificity protein 1 (Sp1), and signal transducer and activator of transcription 3 (STAT3) in HSCs (103–105). Hepatocyte Nox4 induction by TGFβ likely involves sma and mad related family (SMAD) proteins, which were shown to mediate TGFβ-induced Nox4 elevation in myofibroblasts (106). While the mechanism of how HCV increases Nox1 is unknown, TGFβ-stimulated increases in hepatocyte Nox1 and Rac proteins, or activities, have been reported (96–98).
Nox4, like other Nox enzymes, is composed of six predicted transmembrane domains and an intracellular C-terminal region that contains FAD and NADPH binding sites (107) (Figures 2B and 2C). The C-terminal FAD and NADPH binding sites and intracellular B loop are essential for its catalytic activity. It has also been reported that the D loop allows proper interaction between Nox4 and p22phox, and the E loop is involved in H2O2 generation by Nox4 (107, 108). Nox4 has been localized to the cytoplasm as well as the plasma membrane and nuclear or peri-nuclear regions in various cell types (109–115). Nox4 was detected in both the inner and outer membrane of hepatic cell nuclei by immunogold electron microscopy, where Nox4 nuclear inclusions were also observed (116). The nuclear/peri-nuclear localization of Nox4 also increased with HCV in hepatocytes (72), with corresponding increases in nuclear nitrotyrosine and Nox activity that could be decreased by Nox4 small interfering RNA and an inhibitor of nitric oxide synthase (72). A recent study by Anilkumar et al. also described an N-terminus truncated Nox4, lacking transmembrane domains, that localized to nucleolus and produced superoxide in an NADPH-dependent manner (100, 117, 118). The nature of Nox proteins’ association with nuclear compartments and the specific functions of nuclear Nox4, however, remain to be uncovered. TGFβ, which mediated Nox4 elevation by HCV in hepatocytes (71, 72), could increase nuclear Nox4 protein in other cell types (110, 111). Therefore, nuclear or peri-nuclear Nox4 could represent a normal cellular process that is enhanced by HCV. Nuclear location of Nox1 and Nox2 has also been reported in other studies (115, 119, 120). Together, evidence points to Nox1, Nox4, and Nox2 as major endogenous sources of ROS responsible for the HCV- and immune cell-mediated oxidative stress in hepatitis C patients.
E. Cytochrome P450 enzymes and decreased antioxidant defense
Another potential source of oxidative stress in HCV-infected individuals, particularly during alcohol intake, includes the cytochrome P450 (CYP) family enzymes (121, 122) (Figure 2A). CYP2E1 is involved in the hepatic detoxification of ethanol that leads to ROS generation (123, 124). HCV has been suggested to increase the level of CYP2E1 and potentiate CYP2E1-dependent ROS production in response to ethanol (46, 121, 125, 126). A recent study also reported that sulfaphenazole, which inhibits CYP2C9, decreased HCV-induced ROS, associated with an elevated expression of c-Myc (127). Interestingly, ethanol and its metabolite, acetaldehyde, activated H2O2 and superoxide generation in rat HSCs, and raloxifene, a non-specific inhibitor of Nox, decreased ROS elevation by acetaldehyde (128).
In addition, HCV can blunt the host antioxidant defense (see (129) and (130) for reviews). For example, hepatic, blood, and lymphocytic GSH is depleted with HCV, along with decreases in plasma antioxidant potential, vitamins A, B, and E, zinc, and selenium levels (57) (131) (46, 63, 132, 133). Such decline in antioxidant defense mechanisms would increase susceptibility to redox-mediated damage. It has been suggested that HCV directly interferes with the upregulation of antioxidant defense that normally occurs in response to sublethal oxidative stress, for example, by delocalizing small Maff proteins from the nucleus to the viral replication complex in the ER (134, 135). While GSH is clearly suppressed in patients, however, the extent to which HCV interferes with upregulation of the antioxidant defense is unclear (72, 136, 137).
III. Oxidative stress and pathogenesis of HCC
A. Oxidative stress is a key mediator of HCC
Evidence strongly pointing to a redox mechanism of hepatocarcinogenesis comes from in vivo studies that altered the levels of antioxidants or antioxidant gene expression to examine their effects. Mice deficient in copper, zinc superoxide dismutase (CuZn SOD, sod1−/−), which converts superoxide to H2O2, exhibited reduced lifespan and increased incidence of nodular hyperplasia and HCC (138). Liver-specific deletion of nuclear factor-erythroid-2 related transcription factor-1 (Nrf1), involved in the regulation of antioxidant and phase II enzyme gene expression, likewise increased lipid peroxidation, oxidative DNA damage, and spontaneous liver cancer that occurred in 100 % of both male and female Nrf1-knockout mice compared to 0 % in littermate controls (139). These studies suggested that oxidative stress could be sufficient to induce liver cancer.
In addition, a recent study that examined the mechanism of hepatocarcinogenesis by a well-known tumor promoter, 12-o-tetradecanoylphorbol-13-acetate (TPA), found that TPA increased mitochondrial ROS and DCF fluorescence (140). The TPA-induced tumor cell invasion could be decreased by addition of NAC and by decreasing the expression of ROS modulator 1 (Romo1) (140). TPA-associated ROS elevation was also suppressed by myxothiazol, an inhibitor of electron transport at complex III, and by DPI. TPA induced matrix metalloproteinase (MMP) promoter activity that was suppressed by non-specific inhibitors of Nox proteins, such as DPI and apocynin. In the DEN model of HCC in rats, DEN increased the oxidation of DCF in isolated liver mitochondria as well as conjugated dienes, while decreasing catalase and SOD levels in the liver; nanocapsulated curcumin protected animals from histopathologic changes induced by DEN (141). In a different HCC model where the loss of Toll-like receptor 2 (TLR2), a pattern-recognition receptor, promoted DEN-induced HCC, pretreating the animals with NAC attenuated carcinogenesis and HCC progression (142); here, the TLR2 deficiency was thought to enhance HCC formation by decreasing activation of NFκB, a key mediator of cell survival and neoplastic progression in the liver (142, 143). Other studies found a suppression of DEN-induced liver damage or HCC by butylated hydroxyanisole (BHA) (144, 145). NAC also attenuated D-galactosamine/lipopolysaccharide (LPS)-stimulated liver injury (146).
Likewise, double-transgenic mice bearing liver-specific expression of TGFα and c-Myc developed HCC with elevated DCF fluorescence and decreased GSH in hepatocytes (147–149). Preneoplastic and neoplastic lesions from the c-Myc, TGF-κ and c-Myc/TGF-α transgenic mice contained increased levels of Nox subunits, p47phox, p67phox, and Rac1, 70 kilodalton heat shock protein, as well as heme oxygenase-1; vitamin E reduced cell proliferation, apoptosis, and chromosomal alterations, and produced a 65% reduction in tumor incidence in animals (148, 149). These effects were associated with decreased levels of Nox subunits and iNOS, in addition to changes in DCF fluorescence. 2-Acetylaminofluorene induced mitochondrial redox cycling and increased superoxide levels concomitant with Rac activation in human hepatoma cells (150, 151). It is interesting that aflatoxin, alcohol, and HBV and HCV infections, which are the major etiologic agents of liver cancer, are each associated with oxidative stress (124, 152, 153). These studies indicate that oxidative stress is a common pathogenic feature in diverse HCC models. Note, however, that DCF oxidation is not specific to any particular reactive species and can be affected by diverse molecules including, cytochrome c; thus, changes in DCF fluorescence should be interpreted with caution, particularly during apoptosis (154).
Several lines of evidences also support the role of oxidative/nitrosative stress in HCV-associated liver cancer. ROS, detected using an electron paramagnetic resonance spin probe, were positively correlated with histological disease activity in chronic hepatitis C patients (74). Iron depletion therapy reduced oxidative damage, improved ALT levels, reduced inflammation and progression of fibrosis, and decreased the risk of HCC among hepatitis C patients (133, 155–157). Conversely, increasing hepatic iron enhanced liver steatosis and HCC formation in transgenic mice expressing HCV polyprotein (47, 158). Mitoquinone and antioxidant formulations containing silymarin, ascorbic acid, lipoic acid, L-GSH, or α-tocopherol generally had favorable effects on HCV-associated liver damage in patients (130, 159–161). Furthermore, the addition of BHA in the drinking water of HCV core transgenic mice significantly decreased DEN/Phenobarbital-driven hepatic tumor formation (39).
Cancer is characterized by sustained proliferative signaling, resistance to cell death, replicative immortality, angiogenesis, and activation of invasion and metastasis, and the processes underlying these hallmarks involve genome instability and inflammation (162). Specific mechanisms whereby oxidative stress contributes to HCC are summarized in the following sections and Figures 3A and 3B.
Fig. 3. Oxidative stress in the pathogenesis of HCC.
(A) Oxidative stress in the pathogenesis of HCV-associated HCC. A majority of HCV infection results in chronic infection that can progress to cirrhosis in 10 – 30 years. HCC forms in 4 – 7% of chronic hepatitis C patients with cirrhosis per year (see I-B). HCV induces oxidative stress and chronic inflammation that can lead to liver tumors/HCC through DNA damage, increased cell proliferation, regulation of cell death, and fibrosis that increases the risk for HCC. (B) Inflammation-driven pathways to HCC in chronic hepatitis C. Some of the main events contributing to HCV-induced HCC involving inflammation and redox-active processes are shown. (i) HCV activates Nox2 of monocytes/Kupffer cells and polymorphonuclear cells (PMN). (ii) Lymphotoxin β contributes to HCC formation in an NFκB-dependent manner. AP-1 and Stat3 also mediate cytokine elevation. Activation of NALP3 inflammasome in hepatocytes and Kupffer cells by HCV contributes to increased IL-1β secretion. (iii) TLR4-mediated inflammation and liver injury mediated by Kupffer cells require p47phox, a component of Nox2. (iv) TLR4 stimulation by LPS, CXCL10, and alcohol mediates transformation of hepatic progenitor cells and increases proliferation of tumor-initiating cells through Yap, while inhibiting tumor suppressive effects of TGFβ.
B. Oxidative DNA damage and inhibition of DNA repair
Chemical mutagenesis of DNA, leading to the mutation of critical cellular genes, is likely to be a key mechanism whereby oxidative stress contributes to hepatocarcinogenesis (Figure 3A). DNA strand breaks and other DNA modifications occur when highly reactive species such as hydroxyl radical (HȮ) attack bases and the deoxyribosyl backbone of DNA (163, 164). Chemical mutagenesis is likely to be random, but can facilitate the development of cancer when critical cellular regulatory genes are affected, such as activation of a proto-oncogene. 8-Hydroxydeoxyguanosine (8-OHdG), a product of redox-mediated DNA damage, for example, can induce G:C to T:A transversions frequently observed in cancers. Hepatic 8-OHdG is increased in HCC and has been positively correlated with HCC grades (165, 166). In patients with HCC, high levels of 8-OHdG in non-cancerous regions at the time of hepatectomy have been associated with an increased risk of tumor recurrence (167).
Increased levels of 8-OHdG and double strand DNA breaks were also found in hepatitis C patients as well as various cell culture and animal models of HCV, suggesting significant alteration of the host redox status with ongoing DNA damage (56, 59, 61–63, 67, 77, 131–133, 168–170). Higher levels of 8-OHdG were observed with HCV compared to HBV (131). Mutations in p53, β-catenin, B-cell lymphoma-6, and immunoglobulin heavy chain genes were elevated in HCV-infected B cells and HCV-associated peripheral blood mononuclear cells, lymphomas, and HCC (67, 168). In addition, significant membrane alterations with loss of normal chromatin organization could be found in HCV transgenic hepatocytes (40). HCV core, the primary factor responsible for virus-induced changes in the host redox status, functioned as a tumor initiator in phenobarbital-treated mice (171).
Hydroxyl radical, which induces 8-OHdG and other DNA damage products, is produced by a non-enzymatic reaction between H2O2 and ferrous iron (Fe2+), referred to as the Fenton reaction (Figure 2A). Because of its high reactivity and propensity to react with whatever molecule near the site of its generation, hydroxyl radical needs to be generated in close proximity to DNA to directly induce DNA damage. HCV-induced DNA damage was reduced by NAC, as well as strategies that decreased iron or the production of nitric oxide (77). 8-OHdG decreased with iron reduction therapy in hepatitis C patients and increased with iron overload in HCV transgenic mice (47, 133). These findings suggest that excess iron and sources of superoxide/H2O2 and nitric oxide (see section II) all contribute to genotoxicity during HCV infection via hydroxyl radical and peroxynitrite formation (Figure 2A). The discovery that Nox4 is present in or around the nucleus of hepatocytes actively replicating HCV is intriguing as it would increase the probability of DNA damage (72) (Figure 2A). For example, a shift in ROS generation from the cytosol to the nucleus accelerated DNA damage in peroxyredoxin 1-knockout mice (172). Nucleolar localization of the 28kD Nox4 splice variant was associated with increased DNA damage, as determined by phosphorylated histone family H2A, member X (H2AX).
HCV also interferes with DNA repair, which would exacerbate its mutagenic effects. 8-OHdG is eliminated mainly through base excision repair initiated by 8-oxoguanine glycosylase (OGG1), a DNA glycosylase. Human OGG1 polymorphism was associated with some cancers and could predict survival in HCC patients (173, 174). A positive correlation was observed between 8-OHdG levels, disease stage, telomerase activity, OGG1 polymorphisms and ALT/gamma-glutamyl transpeptidase levels in HCC patients (175). OGG1 activity could be modulated by reversible cysteine modification and inhibited by nitric oxide; the latter was associated with the formation of S-nitrosothiol adducts and loss or ejection of zinc ions (176, 177). Recently, hXRCC3, a different DNA repair protein that participates in homologous recombination, was also suggested to be redox-regulated (178). HCV was found to induce error-prone DNA polymerases while decreasing DNA glycosylase activity in an iNOS-dependent manner (39, 168). HCV also interfered with nonhomologous end-joining repair. Interestingly, HCV core interacted with 14-3-3 protein (179). H2AX, involved in DNA damage response and repair, was suggested to affect Nox1 activity indirectly by binding to 14-3-3 zeta in HCC cells (180). Over-expression of H2AX triggered dissociation of 14-3-3 zeta from NoxA1, leading to Nox1 activation and induction of oxidative stress (181, 182).
C. Redox regulation of epigenetics in HCC
The pathogenesis of HCC involves epigenetic as well as genetic modifications. Previously, H2O2 was shown to decrease the expression of E-cadherin through epigenetic silencing (166). Loss of E-cadherin, a cell adhesion molecule that regulates epithelial-mesenchymal transition (EMT), occurs in metastasis and is associated with poor prognosis of HCC. Exogenous H2O2 and menadione, a redox-cycling quinone, increased hypermethylation of the E-cadherin promoter by histone deacetylase 1 and DNA (cytosine-5)-methyltransferase 1 (DNMT) through increased expression of snail, which silenced the expression of E-cadherin (166, 183). CuZn SOD, catalase, and GSH peroxidase 1 activities also declined with HCC grade, and prolonged exposure to H2O2 increased methylation of the catalase promoter, downregulating its transcription in HCC cell lines (166, 184).
Decreased expression of E-cadherin occurred as an early marker of HCC in HCV-related cirrhosis (185). HCV triggered EMT, with loss of E-cadherin, and elevated snail and twist in primary human hepatocytes and in HCV-infected liver biopsies (186). Hypermethylation of the E-cadherin promoter, and increased the expression of DNA (cytosine-5)-methyltransferase 1 and 3 by HCV core have also been described (187, 188). In a recent study, HCV increased DNA methylation in severe combined immunodeficient (SCID) mice carrying a urokinase-type plasminogen activator transgene controlled by an albumin promoter (or, uPA/SCID mice), where the liver was repopulated with human hepatocytes (189). Changes in DNA methylation were accompanied by increased dihydroethidium stain and induction of IFNγ (189).
D. Inflammation, HCC, and Nox enzymes
Currently, there is no established HCV small animal model that shows spontaneous liver tumors, as well as chronic hepatitis and hepatic fibrosis, which has limited our understanding of the role that inflammation plays in HCV-induced pathogenesis. For example, a recently described chimeric mouse model with a humanized liver and reconstituted T cell immunity can be infected by HCV and show signs of inflammation, hepatitis, and fibrosis, but whether hepatic tumors form in these animals is unknown (190). In addition, transgenic mice that express human HCV receptors CD81, OCLN, CLDN1, and SRB-1, referred to as four HCV entry factors, were developed. These mice have complete host immunity and can be infected with HCV and show increases in IFNγ, tumor necrosis factor alpha (TNFα), and IFN stimulated genes, but whether these mice show natural disease progression is unclear (191).
Nevertheless, emerging data from various in vitro, animal, and clinical studies suggest an important role of inflammation in HCC. Indeed, although spontaneous tumors/HCC can form without hepatitis in several HCV transgenic animal models (46, 47), natural HCV infection is marked by chronic hepatitis (90–92, 192–194). Kupffer cells, which are resident liver macrophages, represent 15 to 20% of the total liver cell population. Studies indicate that Kupffer cells/Nox2 are activated in chronic hepatitis C (91, 92, 192, 195) (also, refer to section IIC) (Figure 3B). In addition, myeloperoxidase genotype was associated with the progression of hepatic fibrosis and HCC in individuals with chronic hepatitis C, suggesting the involvement of neutrophils in pathogenesis (196, 197). Peripheral blood neutrophils from children with chronic hepatitis C demonstrated an increased expression of TLR2 and TLR4 that was associated with increased ALT and intensified hepatic necrosis (198). Rises in malondialdehyde and protein carbonyls, and decreases in GSH and sulfhydryl proteins in the plasma preceded ALT flare-ups in hepatitis C patients, supporting a role of oxidative stress in inflammation and tissue damage (199).
• Lymphotoxin and NFκB
Recently, a dramatic upregulation in lymphotoxin-β levels was identified in tumors from HCV transgenic mice, which was accompanied by activation of NF-κB, elevated chemokines, and intra-tumoral recruitment of mononuclear cells (200) (Figure 3B). Inactivation of IKKβ reduced the tumor rate in these animals (200). Previously, liver-specific expression of lymphotoxin was shown to trigger liver inflammation and HCC in mice, and the study supported the importance of NFκB in HCC (201, 202). NFκB, a chief regulator of pro-inflammatory responses and apoptosis, is regulated in the cytoplasm as well as nucleus by redox mechanisms (203). The mechanism of NFκB activation in the cytoplasm involves protein kinase A-mediated serine phosphorylation as well as H2O2-induced disulfide bond formation in NEMO that inhibits the IκB kinase complex (204, 205). Cysteine oxidation of the p50 subunit, on the other hand, inhibits DNA binding of NFκB; thus, NFκB requires reduction by redox factor 1 (Ref-1) in order to function (206, 207). For example, Nox2 and Nox1 activated c-Src and NFκB and increased hepatic production of TNFα during ischemia/reperfusion (208).
• Pro-inflammatory cytokines
Serum cytokines, such as TNFα and interleukin-1 beta (IL-1β), are elevated in chronic hepatitis C (121, 209, 210) (Figure 3B). Kupffer cells acted as the main source of IL-1β during HCV infection (211). These cells are also likely to be a major source of TNFα in chronic hepatitis C (91, 121). Increased levels of IL-1/IL-1β, IL-6, and TNFα were also demonstrated in HCV core transgenic mice that developed liver tumors (39, 212). The mitogen-activated protein kinase (MAPK) pathway is a well known target of redox signaling (213). c-Jun N-terminal kinase (JNK) and AP-1 were activated in the core transgenic mouse liver (39, 214), and cytokine elevation was found to be mediated by c-Jun and STAT3; genetic deletion of c-Jun and STAT3 decreased tumor rates in these mice (39) (Figure 3B). HCV NS5A-induced activation of JNK, p38 MAPK, and AP-1 could also be inhibited by NAC and dithiothreitol (215, 216). NAC and pyrrolidine dithiocarbamate (PDTC) also affected NFκB and STAT3 activation by HCV (75).
Secretion of IL-1β requires inflammsome-mediated activation of caspase 1 (217). HCV increased the secretion of IL-1β by activating NACHT, LRR and PYD domains-containing protein 3 (NALP3) inflammasomes in Kupffer cells, as well as human hepatoma cells (136, 211) (Figure 3B). While NALP3 inflammasome/caspase 1 activation has been suggested to require ROS and to regulate Nox2 under certain conditions, HCV-stimulated IL-1β secretion in hepatocytes could be blocked by PDTC (136, 217, 218). These studies suggest that NFκB and cytokine modulation by HCV could be redox-mediated.
• DEN-induced liver damage and p47phox
Previously, inflammation, apoptosis, and regenerative DNA synthesis subsequent to DEN-induced liver damage were shown to be diminished in mice deficient in p47phox (219). DEN directly activated Kupffer cells to produce superoxide and TNFα, and these responses were attenuated in the p47phox knockout mice. Furthermore, DEN induced DNA damage in hepatocytes in vivo, but the DNA damage was reduced in p47phox-deficient animals. These studies suggested that superoxide generation by phagocytes during inflammation/liver injury produces genotoxic effects in hepatocytes, which may contribute to tumor initiation and promotion (219).
• TLR4 and HCC
TLR4, a pattern recognition receptor, is activated by endotoxins such as LPS present in gram negative bacteria. Alcohol is a major cofactor in HCV-associated liver disease (220). In alcohol-induced endotoxemia, gut permeability is increased, resulting in bacterial translocation and activation of TLR4. Nox2/p47phox is a key mediator of LPS/TLR4-stimulated ROS generation in polymorphonuclear cells and macrophages (221–224) (Figure 3B). Early effects of alcohol on liver injury also involve LPS-stimulation of Kupffer cells that serve as the predominant source of TNFα (225). The p47phox knockout mice failed to show an increase in free radical detection in bile by electron spin resonance, activation of NFκB, increase in TNFα mRNA, or liver pathology in response to alcohol treatment (225). Also, ethanol and LPS activated Rac1-GTPase and translocation of p67phox in rat Kuffper cells (226). TLR4 stimulation activated Rac1 through IRAK-1 in macrophages (227).
Recent studies indicate that TLR4 is a key factor in HCC formation; TLR4 increased cell proliferation in response to carcinogens, and depleting the intestinal microbiota decreased the incidence of HCC (145, 228). TLR4 is also important in HCV-induced HCC. For example, HCV can also up-regulate TLR4 (229); TLR4, LPS, as well as C-X-C motif chemokine 10 (CXCL10), an endogenous chemokine that activates TLR4, are likewise elevated in hepatitis C patients (230–233), although a downregulation of TLR4 and associated apoptosis was also reported in one study (234). A recent study further indicated that alcohol and LPS treatments synergized with HCV NS5A to enhance the rate of hepatic tumors through a progenitor cell marker, Nanog (44) (Figure 3B). LPS increased tumorigenesis in p53-deficient and TLR4-transduced hepatic progenitor cells, and enhanced the proliferation of tumor initiating cells (39, 235). Intestinal colonization by the gram negative bacteria, H. hepaticus, promoted aflatoxin-and HCV transgene-induced HCC in mice, that was associated with increased hepatocyte turnover, Wnt/β–catenin activation, oxidative injury, and activation of NFκB-regulated networks in the lower bowel and liver (236). Clinically, the rs4986791 T allele of TLR4 was associated with a decreased risk of HCC in chronic hepatitis C patients (237). Concanavalin A-induced liver injury and cell death were also attenuated by antagonism of CXCL10 (232). These studies suggest that Nox2 could be a key mediator of hepatocarcinogenesis mediated by the TLR4 pathway during HCV infection, and other conditions leading to HCC. A recent study reported that macrophages from p47phox knockout mice showed greater NFκB activation by LPS than control mice (238); although it is not clear why p47phox exhibited different effects on NFκB in the study, but the results are consistent with the regulation of NFκB in the nucleus by Ref-1 (203).
LPS stimulation was also shown to up-regulate Nox1 transcription in bone marrow-derived macrophages; here, Nox1 transcription was associated with activation of IRAK-1 and increased recruitment of p65 and C/EBP to the Nox1 promoter (227). LPS-stimulated Nox1 expression has also been associated with accumulation and nuclear translocation of β-catenin in macrophages (239). In addition, TLR4 signaling required Nox4 in the Raw264.7 murine macrophage cell line, HEK293 cells, and human aortic endothelial cells (240–242). Whether Nox proteins are involved in TLR4 signaling in hepatocytes, however, is unknown.
• TNFa and hepatocyte proliferation
Increased expression of pro-inflammatory cytokines such as TNFα is associated with apoptosis but has also been shown to be essential for hepatocyte proliferation and liver regeneration (243–245). TNFα increased cell-cycle progression and 8-oxo-dG levels in murine hepatocytes without stabilizing p53 (246, 247). TNFα also up-regulated hypoxia-inducible factor 1-mediated Forkhead box M1 (FoxMI) expression that could be inhibited by BHA; these changes were associated with HCC proliferation and resistance to apoptosis (245). FoxMI is a cell cycle regulator that is upregulated in human HCC tissues, and FoxMI expression has been associated with HCC formation in mice (248–250). Furthermore, TNFα treatment of hepatocytes increased mitochondrial ROS generation at complex I and complex III that in turn, activated NF-κB and increased cell migration (251) (Figure 2). TNFα also up-regulated Nox2, Nox4, Nox1, p67phox, p47phox, p22phox, and/or Duox2 gene expression in other systems (252–254). Nox1 and Nox4, which are elevated by HCV (71, 72), also contain conserved gamma activated sequence elements in their promoters, and were activated by IFNγ in human large intestinal epithelial cells and aortic smooth muscle cells; likewise, Duox2 was activated by IFNγ in primary respiratory tract epithelial cell cultures (255–257). IFNγ levels are elevated in the liver and serum in the presence of HCV (189, 191, 210). Whether Nox proteins are regulated by TNFα and IFNγ and mediate their effects in hepatocytes, remains to be tested.
E. Hepatic fibrosis, TGFβ, and Nox enzymes
Liver fibrosis is characterized by accumulation of collagen and other extracellular matrix components in response to chronic liver injury (258). Cirrhosis is an end-stage process of fibrotic liver degeneration (259). HSCs and Kupffer cells are major fibrogenic cell types in the liver. During hepatic fibrosis, quiescent HSCs transdifferentiate to myofibroblasts and produce cytokines, extracellular matrix proteins, and glycoproteins (260). Oxidative stress is a key mediator of hepatic fibrosis, and ROS and lipid peroxidation can stimulate the production of type I collagen (258, 259, 261, 262). Increased iron levels have also been shown to induce inflammatory cytokines and subsequent activation of HSCs, leading to fibrosis (263). A majority of HCV-associated HCC cases are preceded by cirrhosis (21, 264). Thus, the induction of hepatic fibrosis could be another process through which oxidative stress facilitates HCC formation (Figure 3A).
Accumulating data indicate the importance of Nox2, Nox1, and Nox4 proteins in hepatic fibrosis. Nox2 and Nox1 were upregulated, in addition to Nox4, in activated HSCs (265). Bile duct ligation (BDL)-induced liver fibrosis was attenuated in p47phox knockout mice compared to controls, where Nox1 and Nox2 were shown to play a role (265–267). Also, phagocytosis of apoptotic hepatocytes activated HSCs and collagen production through Nox2 activation (268, 269). CCl4-induced liver fibrosis and hydroxyproline were similarly reduced in gp91phox knockout mice (270). Furthermore, pharmacological inhibition of Nox1 and Nox4 with GKT137831 significantly reduced liver fibrosis induced by bile duct ligation or CCl4 (271, 272). Fibrogenic responses to angiotensin II were blunted by NAC and DPI treatments, and angiotensin II-stimulated DNA synthesis and cell migration of HSCs were reduced in p47phox knockout mice (266). On the other hand, decreased functions of neutrophils, including repiratory burst, are commonly observed in decompensated patients with cirrhosis (273).
TGFβ, which activated Nox4 and Nox1/Rac in hepatocytes (71, 72, 96–98), is a major factor accelerating the progression of organ fibrosis (274). TGFβ stimulates type I collagen transcription and regulates the expression of MMPs and their inhibitors (274). The fibrogenic effects of TGFβ are mediated by ROS (259). Nox4 was also increased by TGFβ in fibroblasts, and Nox4 mediated TGFβ functions in hepatocytes, fibroblasts, as well as HSCs (106, 111, 271, 275, 276). The discovery that HCV induces Nox4 in a TGFβ-dependent manner, therefore, is potentially significant and suggests a role of hepatocyte Nox4 and oxidative stress in hepatitis C-associated fibrosis. Increased expression of Nox4 was found in HCV-infected patient liver, which increased with fibrosis degree (72, 101). Duox1, Duox2, and Nox4 were expressed at higher frequencies in tumor specimens from patients (277). Furthermore, treatment with DPI abrogated changes in fibrogenic gene expression induced by HCV and human immunodeficiency virus in HSCs and hepatoma cells (278). HSC activation by hepatocyte apoptotic bodies, which is likely Nox2-dependent (269), was also found to be amplified when the apoptotic bodies contained HCV nonstructural proteins (279). Supplemental treatment of chronic hepatitis C patients with CuZn SOD reduced TGF-β mediated liver fibrosis (280). Other enhancers of fibrosis, increased in response to HCV, include other growth factors, chemokines, and cytokines, such as TNF-α (258, 281).
TGFβ, on the other hand, has a tumor suppressive effect on hepatocytes. A recent study showed that Nox4 expression was significantly downregulated during active hepatocyte proliferation during DEN-induced hepatocarcinogensis, and after partial hepatectomy in mice (282). In the same study, silencing of Nox4 was associated with increased proliferation of liver tumors and untransformed cells as well as decreased activation of caspase 3 by TGFβ. These results are consistent with previous studies that reported a role of Nox4 in TGFβ-induced senescence arrest/apoptosis in hepatocytes (276, 283–285). However, loss of the tumor suppressive function of TGFβ is frequently observed in cancers (274). In chronic HCV infection, an inflammatory environment, mediated by JNK-activated IL1β, was also suggested to shift the tumor-suppressive signaling of TGFβ to a migratory and mesenchymal phenotype associated with tumor progression (286). HCV itself suppressed TGFβ-induced apoptosis (287, 288). TLR4 signaling also interfered with the tumor suppressive effects of TGFβ in tumor-initiating cells through Yap1, and the TGFβ pathway in turn suppressed TLR4 signaling (235) (Figure 3B). In addition, TGFβ treatment of rat pluripotent liver progenitor cell (LPC)-like cells impaired the LPC potential but conferred tumor initiating cell properties (289). TGFβ level was positively correlated with LPC and tumor initiating cell markers in rat models of HCC as well as cirrhotic livers of human HCC patients (289). Furthermore, VAS2870, a Nox inhibitor, decreased serum-dependent growth and phosphorylation of AKT and ERK and enhanced TGFβ-induced apoptosis in HCC cell lines (290). Nox1, which was co-elevated with Nox4 by HCV (71, 72), was also suggested to provide a partial resistance to hepatocyte apoptosis induced by TGFβ (290, 291), and excessive stimulation of the MAPK/ERK pathway produced similar effects (284). Furthermore, NS5A was recently found to interact with mixed lineage kinase 3 that activated p38 MAPK to combat apoptosis induced by sulfhydryl oxidizing agent, dithiodipyridine (292). HCV core protein suppressed cellular senescence stimulated by H2O2 by downregulating p16 (293). Mutation or allelic loss of caspase-8, an initiator caspase, is also frequently observed in HCC (294). In advanced HCC associated with chronic hepatitis C, apoptosis was decreased with downregulation of Ras (295). On the other hand, recent studies using HCV infection models suggest that HCV is pro-apoptotic (70, 296–298). Thus, HCV most likely regulates apoptosis differently at various stages of pathogenesis, and the role of hepatocyte Nox proteins in the regulation of cell apoptosis and proliferation should be further defined.
F. Hepatic steatosis and other mechanisms
The incidence of hepatic steatosis, observed in almost half of the HCV infected patients (1, 299), strongly correlates with the development of liver fibrosis, cirrhosis and eventually HCC (300–302). Various in vivo studies show that HCV core and NS5A are associated with steatosis (303–306), and HCV core, NS2 and NS4B proteins also induced accumulation of lipid droplets in vitro (307–309).
HCV can upregulate sterol regulatory element binding protein 1 (SREBP1) and peroxisome proliferators-activated receptor (PPAR) γ, which in turn increase hepatic fatty acid synthesis (310). PPARα and proteasome activator 28γ were necessary for the induction of steatosis as well as hepatocarcinogenesis in HCV core transgenic mice (304, 305). Activation of SREBP-1 by HCV was sensitive to PDTC, Ca2+ chelator, and an inhibitor of phosphoinositide 3-kinase (PI3K) (311). Other metabolic factors such as insulin resistance and hyperglycemia associated with HCV may also contribute to lipid accumulation indirectly. Thus, the increased accumulation of polyunsaturated fatty acids in steatosis resulting from the combination of viral and metabolic factors, together with the increased hepatic iron reserves detected in the liver of chronic hepatitis C patients may make the steatotic liver an easy target for lipid peroxidation (312, 313) (see II-A). Lipid peroxidation can result in the formation of DNA adducts (314).
HCV also modulates regulators of cell proliferation and cell death that include retinoblastoma protein (pRb), p53, Wnt/β-catenin, and MAPK pathways (see (315–317) for reviews). pRb is a G1/S regulator of the cell cycle that functions by inhibiting E2F-dependent transcription. p53 is a tumor suppressor protein that functions in cell cycle regulation, induction of apoptosis, and DNA repair in the presence of DNA damage. Aberrant activation of the Wnt signaling pathway stabilizes β-catenin allowing entrance into the nucleus to activate transcription of Wnt-target genes that can mediate hepatocarcinogenesis (318). HCV can decrease the level of pRb and p53 and interact with these factors (315–317). HCV also up-regulated β–catenin-mediated transcription via PI3K (319), which may contribute to HCC by increasing hepatocyte proliferation, tumorigenesis, and EMT (127, 320, 321). Several studies reported that some of these effects of HCV could be inhibited by NAC or PDTC, including PI3K and osteopontin that regulated β-catenin (307, 321, 322).
Moreover, ROS and Nox proteins have been shown to mediate angiogenesis (323, 324) (325), but whether Nox enzymes mediate angiogenesis in HCV-induced HCC remains to be tested. H2O2 could increase iron uptake in murine fibroblasts (326, 327), and it would be interesting to test whether H2O2 contributes to hepatic iron accumulation in chronic hepatitis C. Another potential mechanism of hepatocarcinogenesis involves increased survival of the infected cells through activation of Nrf2 by reactive species, although an attenuation of Nrf2 and cellular antioxidant response by HCV has also been reported (134, 135, 328, 329) (see IIE).
IV. Oxidative stress and HCC – conclusions and considerations for therapy
Chronic hepatitis C is a global health concern. Accumulating evidence suggests that oxidative stress plays an important role in HCC induction by HCV as well as other conditions leading to HCC. Cancer is a multi-step process, and the mutagenic effects of HCV-associated oxidative stress are likely to cooperate with other pathogenic mechanisms of HCV, such as inflammation, to induce HCC (Figure 3). These mechanisms may include redox signaling as well as oxidative damage (Figure 2A).
The multikinase inhibitor, sorafenib, was approved for therapeutic use in advanced, non-resectable HCC (330). Treatment with sorafenib could improve the one-year survival rate to 44% versus 33% in the control group, and lengthened time to radiological progression to 5.5 months versus 2.8 months in the control group (330). However, the limited success, growing increase in HCC cases, as well as reports of serious adverse events resulting in treatment discontinuation, warrant the investigation of adjunct therapies to enhance treatment efficacy. In vitro and in vivo studies combining sorafenib with mammalian target of rapamycin inhibitors suggest enhanced benefit over monotherapy (331). Indeed, HCC is heterogeneous, and the pathogenesis likely involves multiple mechanisms, calling for continuation of basic and clinical studies aimed at investigating the therapeutic benefit of multiple drug combinations in the treatment of HCC, in addition to development of an HCV vaccine.
The common occurrence of increased reactive species in diverse HCC models suggests that strategies designed to reduce the levels of reactive species could be beneficial in preventing HCC. Antioxidant strategies generally produced favorable effects in various in vitro and in vivo models of HCC and in HCV-associated liver damage/HCC, although data on human HCC cases are still limited (130). A recent in vitro study further suggested that oxidative stress may contribute to the high baseline sequence variability of HCV that complicates antiviral therapy (332–336); iron chelator and GSH ester could decrease HCV sequence variability (332). In clinical studies, some antioxidant strategies enhanced the virological outcome of antiviral therapy in patients whereas other studies showed either no effect or inconsistent benefit (130); possible explanations include limited bioavailability, efficacy, and non-specific effects of traditional antioxidants. Altogether, these studies suggest that combining antioxidant strategies with antivirals directly targeting HCV may also improve antiviral efficacy. In this regard, Nox proteins can be inhibited pharmacologically, and this may be more effective at suppressing ROS levels than blunting the damage once reactive species are already generated. Additional studies, however, will be needed to determine whether strategies that target oxidative stress would be beneficial in the treatment of HCC and HCV infection. Possible effects on antiviral host immunity will also need to be evaluated. Recent development of specific Nox-deleted mice and mouse models that support complete HCV replication with partial, or complete adaptive immunity, provide new tools to test these hypotheses (190, 191, 324). Finally, tipping the metabolic balance further towards greater oxidative stress has been associated with cancer cell death, and has been suggested to aid in cancer treatment once cancer has established (337). Thus, the possibility of increasing oxidative stress to enhance the destruction of HCC presents a different perspective that may, too, need to be considered.
Acknowledgments
This work was supported by NIH grant R01CA158271 to J.C.
Abbreviations
- 8-OHdG
8-hydroxydeoxyguanosine
- 8-OHG
8-hydroxyguanine
- ALT
alanine aminotransferase
- AP-1
activator protein 1
- BHA
butylated hydroxyanisole
- CD81
cluster of differentiation 81
- CXCL10
C-X-C motif chemokine 10
- CYP
cytochrome P450
- DCF
dihydrodichlorocarboxyfluorescein
- DEN
diethylnitrosamine
- DPI
diphenyleneiodonium
- DUOX
dual oxidase
- EMT
epithelial-mesenchymal transition
- ER
endoplasmic reticulum
- F/ARFP
frameshift/alternate reading frame protein
- FoxMI
Forkhead box M1
- GAG
glycosaminoglycans
- GSH
glutathione
- H2AX
H2A histone family, member X
- HBV
hepatitis B virus
- HCC
hepatocellular carcinoma
- HCV
hepatitis C virus
- HSCs
Hepatic stellate cells
- Huh7
human hepatoma
- IFN
interferon
- IL-1β
interleukin-1 beta
- iNOS
inducible nitric oxide synthase
- JFH1
Japanese fulminant hepatitis-1
- LDLR
low density lipoprotein receptor
- LPC
liver progenitor cell
- LPS
lipopolysaccharide
- MAPK
mitogen-activated protein kinase
- MMP
matrix metalloproteinase
- MPT
membrane permeability transition
- NAC
N-acetylcysteine
- NALP3
NACHT, LRR and PYD domains-containing protein 3
- NFκB
nuclear factor kappa B
- Nox
NADPH oxidase
- Nrf
nuclear factor-erythroid-2 related transcription factor
- NS
nonstructural
- OCLN
occludin
- OGG1
8-oxoguanine glycosylase
- PDTC
pyrrolidine dithiocarbamate
- PI3K
phosphoinositide 3-kinase
- PPAR
peroxisome proliferator-activated receptor
- pRb
retinoblastoma protein
- Rac
ras-related C3 botulinum toxin substrate
- RNS
reactive nitrogen species
- Romo1
ROS modulator 1
- ROS
reactive oxygen species
- SCID
severe combined immunodeficient
- SMAD
sma and mad related family
- SOD
superoxide dismutase
- Sp1
specificity protein 1
- SR-B1
scavenger receptor class B type I
- STAT
signal transducer and activator of transcription
- SVR
sustained virological response
- TGF
transforming growth factor
- TLR
Toll-like receptor
- TNFα
tumor necrosis factor alpha
- TPA
12-O-tetradecanoylphorbol-13-acetate
- UTR
untranslated region
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Woo HA, Chae HZ, Hwang SC, Yang KS, Kang SW, Kim K, Rhee SG. Reversing the inactivation of peroxiredoxins caused by cysteine sulfinic acid formation. Science. 2003;300:653–656. doi: 10.1126/science.1080273. [DOI] [PubMed] [Google Scholar]
- 2.Babior BM. NADPH oxidase: an update. Blood. 1999;93:1464–1476. [PubMed] [Google Scholar]
- 3.Okuda K, Okuda H. Malignant tumours. In: Bircher J, J-P B, McIntyre N, Rizzetto M, Rodes J, editors. Oxford Textbook of Clinical Hepatology. 2nd ed. Volume II. Oxford, New York, Tokyo: Oxford University Press; 1999. pp. 1491–1550. [Google Scholar]
- 4.Mittal S, El-Serag HB. Epidemiology of hepatocellular carcinoma: consider the population. J Clin Gastroenterol. 2013;47(Suppl):S2–S6. doi: 10.1097/MCG.0b013e3182872f29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.London WT, McGlynn KA. Liver cancer. In: Schottenfeld D, Fraumeni JFJ, editors. Cancer Epidemiology and Prevention. 3 ed. New York, NY: Oxford University Press; 2006. pp. 763–786. [Google Scholar]
- 6.THE CANCER OF THE LIVER ITALIAN PROGRAM (CLIP) INVESTIGATORS. A new prognostic system for hepatocellular carcinoma: a retrospective study of 435 patients: the Cancer of the Liver Italian Program (CLIP) investigators. Hepatology. 1998;28:751–755. doi: 10.1002/hep.510280322. [DOI] [PubMed] [Google Scholar]
- 7.Houben KW, McCall JL. Liver transplantation for hepatocellular carcinoma in patients without underlying liver disease: a systematic review. Liver Transpl Surg. 1999;5:91–95. doi: 10.1002/lt.500050201. [DOI] [PubMed] [Google Scholar]
- 8.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59:225–249. doi: 10.3322/caac.20006. [DOI] [PubMed] [Google Scholar]
- 9.Altekruse SF, McGlynn KA, Reichman ME. Hepatocellular carcinoma incidence, mortality, and survival trends in the United States from 1975 to 2005. J Clin Oncol. 2009;27:1485–1491. doi: 10.1200/JCO.2008.20.7753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.El-Serag HB, Davila JA, Petersen NJ, McGlynn KA. The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med. 2003;139:817–823. doi: 10.7326/0003-4819-139-10-200311180-00009. [DOI] [PubMed] [Google Scholar]
- 11.Kanwal F, Hoang T, Kramer JR, Asch SM, Goetz MB, Zeringue A, Richardson P, et al. Increasing prevalence of HCC and cirrhosis in patients with chronic hepatitis C virus infection. Gastroenterology. 2011;140:1182–1188. e1181. doi: 10.1053/j.gastro.2010.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
- 13.Perz JF, Armstrong GL, Farrington LA, Hutin YJ, Bell BP. The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol. 2006;45:529–538. doi: 10.1016/j.jhep.2006.05.013. [DOI] [PubMed] [Google Scholar]
- 14.El-Serag HB. Hepatocellular carcinoma: recent trends in the United States. Gastroenterology. 2004;127:S27–S34. doi: 10.1053/j.gastro.2004.09.013. [DOI] [PubMed] [Google Scholar]
- 15.Koike K, Tsutsumi T, Miyoshi H, Shinzawa S, Shintani Y, Fujie H, Yotsuyanagi H, et al. Molecular basis for the synergy between alcohol and hepatitis C virus in hepatocarcinogenesis. J Gastroenterol Hepatol. 2008;23(Suppl 1):S87–S91. doi: 10.1111/j.1440-1746.2007.05292.x. [DOI] [PubMed] [Google Scholar]
- 16.Donato F, Boffetta P, Puoti M. A meta-analysis of epidemiological studies on the combined effect of hepatitis B and C virus infections in causing hepatocellular carcinoma. Int J Cancer. 1998;75:347–354. doi: 10.1002/(sici)1097-0215(19980130)75:3<347::aid-ijc4>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
- 17.Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science. 1989;244:359–362. doi: 10.1126/science.2523562. [DOI] [PubMed] [Google Scholar]
- 18.Centers for Disease Control. Recommendations for prevention and control of hepatitis C virus (HCV) infection and HCV-related chronic disease. MMWR. 1998;47(RR19):1–39. [PubMed] [Google Scholar]
- 19.World Health Organization. Hepatitis C: global prevalence (update) Wkly Epidemiol Rec. 2000;75:18–19. [PubMed] [Google Scholar]
- 20.El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264–1273. e1261. doi: 10.1053/j.gastro.2011.12.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tong MJ, el-Farra NS, Reikes AR, Co RL. Clinical outcomes after transfusion-associated hepatitis C. N Engl J Med. 1995;332:1463–1466. doi: 10.1056/NEJM199506013322202. [DOI] [PubMed] [Google Scholar]
- 22.Zignego AL, Ferri C, Giannelli F, Giannini C, Caini P, Monti M, Marrocchi ME, et al. Prevalence of bcl-2 rearrangement in patients with hepatitis C virus-related mixed cryoglobulinemia with or without B-cell lymphomas. Ann Intern Med. 2002;137:571–580. doi: 10.7326/0003-4819-137-7-200210010-00008. [DOI] [PubMed] [Google Scholar]
- 23.Wise M, Bialek S, Finelli L, Bell BP, Sorvillo F. Changing trends in hepatitis C-related mortality in the United States, 1995–2004. Hepatology. 2008;47:1128–1135. doi: 10.1002/hep.22165. [DOI] [PubMed] [Google Scholar]
- 24.Davis GL, Albright JE, Cook SF, Rosenberg DM. Projecting future complications of chronic hepatitis C in the United States. Liver Transpl. 2003;9:331–338. doi: 10.1053/jlts.2003.50073. [DOI] [PubMed] [Google Scholar]
- 25.National Institutes of Health. NIH Consensus Statement on Management of Hepatitis C: 2002. NIH Consens State Sci Statements. 2002;19:1–46. [PubMed] [Google Scholar]
- 26.Vermehren J, Sarrazin C. The role of resistance in HCV treatment. Best Pract Res Clin Gastroenterol. 2012;26:487–503. doi: 10.1016/j.bpg.2012.09.011. [DOI] [PubMed] [Google Scholar]
- 27.Morgan RL, Baack B, Smith BD, Yartel A, Pitasi M, Falck-Ytter Y. Eradication of hepatitis C virus infection and the development of hepatocellular carcinoma: a meta-analysis of observational studies. Ann Intern Med. 2013;158:329–337. doi: 10.7326/0003-4819-158-5-201303050-00005. [DOI] [PubMed] [Google Scholar]
- 28.van der Meer AJ, Veldt BJ, Feld JJ, Wedemeyer H, Dufour JF, Lammert F, Duarte-Rojo A, et al. Association between sustained virological response and all-cause mortality among patients with chronic hepatitis C and advanced hepatic fibrosis. JAMA. 2012;308:2584–2593. doi: 10.1001/jama.2012.144878. [DOI] [PubMed] [Google Scholar]
- 29.Chang KC, Wu YY, Hung CH, Lu SN, Lee CM, Chiu KW, Tsai MC, et al. Clinical-guide risk prediction of hepatocellular carcinoma development in chronic hepatitis C patients after interferon-based therapy. Br J Cancer. 2013;109:2481–2488. doi: 10.1038/bjc.2013.564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.George SL, Bacon BR, Brunt EM, Mihindukulasuriya KL, Hoffmann J, Di Bisceglie AM. Clinical, virologic, histologic, and biochemical outcomes after successful HCV therapy: a 5-year follow-up of 150 patients. Hepatology. 2009;49:729–738. doi: 10.1002/hep.22694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Nojiri K, Sugimoto K, Shiraki K, Kusagawa S, Tanaka J, Beppu T, Yamamoto N, et al. Development of hepatocellular carcinoma in patients with chronic hepatitis C more than 10 years after sustained virological response to interferon therapy. Oncol Lett. 2010;1:427–430. doi: 10.3892/ol_00000075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lohmann V, Korner F, Koch J, Herian U, Theilmann L, Bartenschlager R. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science. 1999;285:110–113. doi: 10.1126/science.285.5424.110. [DOI] [PubMed] [Google Scholar]
- 33.Blight KJ, Kolykhalov AA, Rice CM. Efficient initiation of HCV RNA replication in cell culture. Science. 2000;290:1972–1974. doi: 10.1126/science.290.5498.1972. [DOI] [PubMed] [Google Scholar]
- 34.Wakita T, Pietschmann T, Kato T, Date T, Miyamoto M, Zhao Z, Murthy K, et al. Production of infectious hepatitis C virus in tissue culture from a cloned viral genome. Nat Med. 2005;11:791–796. doi: 10.1038/nm1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Moriya K, Fujie H, Shintani Y, Yotsuyanagi H, Tsutsumi T, Ishibashi K, Matsuura Y, et al. The core protein of hepatitis C virus induces hepatocellular carcinoma in transgenic mice. Nat Med. 1998;4:1065–1067. doi: 10.1038/2053. [DOI] [PubMed] [Google Scholar]
- 36.Buhler S, Bartenschlager R. Promotion of hepatocellular carcinoma by hepatitis C virus. Dig Dis. 2012;30:445–452. doi: 10.1159/000341688. [DOI] [PubMed] [Google Scholar]
- 37.Xu Z, Choi J, Yen TS, Lu W, Strohecker A, Govindarajan S, Chien D, et al. Synthesis of a novel hepatitis C virus protein by ribosomal frameshift. Embo J. 2001;20:3840–3848. doi: 10.1093/emboj/20.14.3840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Walewski JL, Keller TR, Stump DD, Branch AD. Evidence for a new hepatitis C virus antigen encoded in an overlapping reading frame. Rna. 2001;7:710–721. doi: 10.1017/s1355838201010111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Machida K, Tsukamoto H, Liu JC, Han YP, Govindarajan S, Lai MM, Akira S, et al. c-Jun mediates hepatitis C virus hepatocarcinogenesis through signal transducer and activator of transcription 3 and nitric oxide-dependent impairment of oxidative DNA repair. Hepatology. 2010;52:480–492. doi: 10.1002/hep.23697. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 40.Naas T, Ghorbani M, Alvarez-Maya I, Lapner M, Kothary R, De Repentigny Y, Gomes S, et al. Characterization of liver histopathology in a transgenic mouse model expressing genotype 1a hepatitis C virus core and envelope proteins 1 and 2. J Gen Virol. 2005;86:2185–2196. doi: 10.1099/vir.0.80969-0. [DOI] [PubMed] [Google Scholar]
- 41.Lerat H, Honda M, Beard MR, Loesch K, Sun J, Yang Y, Okuda M, et al. Steatosis and liver cancer in transgenic mice expressing the structural and nonstructural proteins of hepatitis C virus. Gastroenterology. 2002;122:352–365. doi: 10.1053/gast.2002.31001. [DOI] [PubMed] [Google Scholar]
- 42.Wang AG, Lee DS, Moon HB, Kim JM, Cho KH, Choi SH, Ha HL, et al. Non-structural 5A protein of hepatitis C virus induces a range of liver pathology in transgenic mice. J Pathol. 2009;219:253–262. doi: 10.1002/path.2592. [DOI] [PubMed] [Google Scholar]
- 43.Kato T, Miyamoto M, Date T, Yasui K, Taya C, Yonekawa H, Ohue C, et al. Repeated hepatocyte injury promotes hepatic tumorigenesis in hepatitis C virus transgenic mice. Cancer Sci. 2003;94:679–685. doi: 10.1111/j.1349-7006.2003.tb01502.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Machida K, Tsukamoto H, Mkrtchyan H, Duan L, Dynnyk A, Liu HM, Asahina K, et al. Toll-like receptor 4 mediates synergism between alcohol and HCV in hepatic oncogenesis involving stem cell marker Nanog. Proc Natl Acad Sci U S A. 2009;106:1548–1553. doi: 10.1073/pnas.0807390106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Keasler VV, Lerat H, Madden CR, Finegold MJ, McGarvey MJ, Mohammed EM, Forbes SJ, et al. Increased liver pathology in hepatitis C virus transgenic mice expressing the hepatitis B virus X protein. Virology. 2006;347:466–475. doi: 10.1016/j.virol.2005.11.050. [DOI] [PubMed] [Google Scholar]
- 46.Moriya K, Nakagawa K, Santa T, Shintani Y, Fujie H, Miyoshi H, Tsutsumi 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. [PubMed] [Google Scholar]
- 47.Furutani T, Hino K, Okuda M, Gondo T, Nishina S, Kitase A, Korenaga M, et al. Hepatic iron overload induces hepatocellular carcinoma in transgenic mice expressing the hepatitis C virus polyprotein. Gastroenterology. 2006;130:2087–2098. doi: 10.1053/j.gastro.2006.02.060. [DOI] [PubMed] [Google Scholar]
- 48.Ray RB, Lagging LM, Meyer K, Ray R. Hepatitis C virus core protein cooperates with ras and transforms primary rat embryo fibroblasts to tumorigenic phenotype. J Virol. 1996;70:4438–4443. doi: 10.1128/jvi.70.7.4438-4443.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ray RB, Meyer K, Ray R. Hepatitis C virus core protein promotes immortalization of primary human hepatocytes. Virology. 2000;271:197–204. doi: 10.1006/viro.2000.0295. [DOI] [PubMed] [Google Scholar]
- 50.Akkari L, Gregoire D, Floc'h N, Moreau M, Hernandez C, Simonin Y, Rosenberg AR, et al. Hepatitis C viral protein NS5A induces EMT and participates in oncogenic transformation of primary hepatocyte precursors. J Hepatol. 2012;57:1021–1028. doi: 10.1016/j.jhep.2012.06.027. [DOI] [PubMed] [Google Scholar]
- 51.Sakamuro D, Furukawa T, Takegami T. Hepatitis C virus nonstructural protein NS3 transforms NIH 3T3 cells. J Virol. 1995;69:3893–3896. doi: 10.1128/jvi.69.6.3893-3896.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Park JS, Yang JM, Min MK. Hepatitis C virus nonstructural protein NS4B transforms NIH3T3 cells in cooperation with the Ha-ras oncogene. Biochem Biophys Res Commun. 2000;267:581–587. doi: 10.1006/bbrc.1999.1999. [DOI] [PubMed] [Google Scholar]
- 53.Einav S, Sklan EH, Moon HM, Gehrig E, Liu P, Hao Y, Lowe AW, et al. The nucleotide binding motif of hepatitis C virus NS4B can mediate cellular transformation and tumor formation without Ha-ras co-transfection. Hepatology. 2008;47:827–835. doi: 10.1002/hep.22108. [DOI] [PubMed] [Google Scholar]
- 54.Sun BS, Pan J, Clayton MM, Liu J, Yan X, Matskevich AA, Strayer DS, et al. Hepatitis C virus replication in stably transfected HepG2 cells promotes hepatocellular growth and tumorigenesis. J Cell Physiol. 2004;201:447–458. doi: 10.1002/jcp.20083. [DOI] [PubMed] [Google Scholar]
- 55.Ghosh AK, Steele R, Meyer K, Ray R, Ray RB. Hepatitis C virus NS5A protein modulates cell cycle regulatory genes and promotes cell growth. J Gen Virol. 1999;80(Pt 5):1179–1183. doi: 10.1099/0022-1317-80-5-1179. [DOI] [PubMed] [Google Scholar]
- 56.Farinati F, Cardin R, De Maria N, Della Libera G, Marafin C, Lecis E, Burra P, et al. Iron storage, lipid peroxidation and glutathione turnover in chronic anti-HCV positive hepatitis. J Hepatol. 1995;22:449–456. doi: 10.1016/0168-8278(95)80108-1. [DOI] [PubMed] [Google Scholar]
- 57.Barbaro G, Di Lorenzo G, Ribersani M, Soldini M, Giancaspro G, Bellomo G, Belloni G, et al. Serum ferritin and hepatic glutathione concentrations in chronic hepatitis C patients related to the hepatitis C virus genotype. J Hepatol. 1999;30:774–782. doi: 10.1016/s0168-8278(99)80128-7. [DOI] [PubMed] [Google Scholar]
- 58.Higueras V, Raya A, Rodrigo JM, Serra MA, Roma J, Romero FJ. Interferon decreases serum lipid peroxidation products of hepatitis C patients. Free Radic Biol Med. 1994;16:131–133. doi: 10.1016/0891-5849(94)90250-x. [DOI] [PubMed] [Google Scholar]
- 59.Kageyama F, Kobayashi Y, Kawasaki T, Toyokuni S, Uchida K, Nakamura H. Successful interferon therapy reverses enhanced hepatic iron accumulation and lipid peroxidation in chronic hepatitis C. Am J Gastroenterol. 2000;95:1041–1050. doi: 10.1111/j.1572-0241.2000.01979.x. [DOI] [PubMed] [Google Scholar]
- 60.Okuda M, Li K, Beard MR, Showalter LA, Scholle F, Lemon SM, Weinman SA. Mitochondrial injury, oxidative stress, and antioxidant gene expression are induced by hepatitis C virus core protein. Gastroenterology. 2002;122:366–375. doi: 10.1053/gast.2002.30983. [DOI] [PubMed] [Google Scholar]
- 61.Hezode C, Cazeneuve C, Coue O, Roudot-Thoraval F, Lonjon I, Bastie A, Duvoux C, et al. Liver iron accumulation in patients with chronic active hepatitis C: prevalence and role of hemochromatosis gene mutations and relationship with hepatic histological lesions. J Hepatol. 1999;31:979–984. doi: 10.1016/s0168-8278(99)80308-0. [DOI] [PubMed] [Google Scholar]
- 62.Thorburn D, Curry G, Spooner R, Spence E, Oien K, Halls D, Fox R, et al. The role of iron and haemochromatosis gene mutations in the progression of liver disease in chronic hepatitis C. Gut. 2002;50:248–252. doi: 10.1136/gut.50.2.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fujita N, Horiike S, Sugimoto R, Tanaka H, Iwasa M, Kobayashi Y, Hasegawa K, et al. Hepatic oxidative DNA damage correlates with iron overload in chronic hepatitis C patients. Free Radic Biol Med. 2007;42:353–362. doi: 10.1016/j.freeradbiomed.2006.11.001. [DOI] [PubMed] [Google Scholar]
- 64.Tandara L, Salamunic I. Iron metabolism: current facts and future directions. Biochem Med (Zagreb) 2012;22:311–328. doi: 10.11613/bm.2012.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Miura K, Taura K, Kodama Y, Schnabl B, Brenner DA. Hepatitis C virus-induced oxidative stress suppresses hepcidin expression through increased histone deacetylase activity. Hepatology. 2008;48:1420–1429. doi: 10.1002/hep.22486. [DOI] [PubMed] [Google Scholar]
- 66.Kotta-Loizou I, Vassilaki N, Pissas G, Kakkanas A, Bakiri L, Bartenschlager R, Mavromara P. Hepatitis C virus core+1/ARF protein decreases hepcidin transcription through an AP1 binding site. J Gen Virol. 2013;94:1528–1534. doi: 10.1099/vir.0.050328-0. [DOI] [PubMed] [Google Scholar]
- 67.Machida K, Cheng KT, Sung VM, Lee KJ, Levine AM, Lai MM. Hepatitis C Virus Infection Activates the Immunologic (Type II) Isoform of Nitric Oxide Synthase and Thereby Enhances DNA Damage and Mutations of Cellular Genes. J Virol. 2004;78:8835–8843. doi: 10.1128/JVI.78.16.8835-8843.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Garcia-Monzon C, Majano PL, Zubia I, Sanz P, Apolinario A, Moreno-Otero R. Intrahepatic accumulation of nitrotyrosine in chronic viral hepatitis is associated with histological severity of liver disease. J Hepatol. 2000;32:331–338. doi: 10.1016/s0168-8278(00)80080-x. [DOI] [PubMed] [Google Scholar]
- 69.Piccoli C, Scrima R, Quarato G, D'Aprile A, Ripoli M, Lecce L, Boffoli D, et al. Hepatitis C virus protein expression causes calcium-mediated mitochondrial bioenergetic dysfunction and nitro-oxidative stress. Hepatology. 2007;46:58–65. doi: 10.1002/hep.21679. [DOI] [PubMed] [Google Scholar]
- 70.Deng L, Adachi T, Kitayama K, Bungyoku Y, Kitazawa S, Ishido S, Shoji I, et al. Hepatitis C virus infection induces apoptosis through a Bax-triggered, mitochondrion-mediated, caspase 3-dependent pathway. J Virol. 2008;82:10375–10385. doi: 10.1128/JVI.00395-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Boudreau HE, Emerson SU, Korzeniowska A, Jendrysik MA, Leto TL. 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. doi: 10.1128/JVI.01059-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.de Mochel NS, Seronello S, Wang SH, Ito C, Zheng JX, Liang TJ, Lambeth JD, et al. Hepatocyte NAD(P)H oxidases as an endogenous source of reactive oxygen species during hepatitis C virus infection. Hepatology. 2010;52:47–59. doi: 10.1002/hep.23671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wang T, Campbell RV, Yi MK, Lemon SM, Weinman SA. Role of Hepatitis C virus core protein in viral-induced mitochondrial dysfunction. J Viral Hepat. 2010;17:784–793. doi: 10.1111/j.1365-2893.2009.01238.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Valgimigli M, Valgimigli L, Trere D, Gaiani S, Pedulli GF, Gramantieri L, Bolondi L. Oxidative stress EPR measurement in human liver by radical-probe technique. Correlation with etiology, histology and cell proliferation. Free Radic Res. 2002;36:939–948. doi: 10.1080/107156021000006653. [DOI] [PubMed] [Google Scholar]
- 75.Gong G, Waris G, Tanveer R, Siddiqui A. Human hepatitis C virus NS5A protein alters intracellular calcium levels, induces oxidative stress, and activates STAT-3 and NF-kappa B. Proc Natl Acad Sci U S A. 2001;98:9599–9604. doi: 10.1073/pnas.171311298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Korenaga M, Wang T, Li Y, Showalter LA, Chan T, Sun J, Weinman SA. Hepatitis C virus core protein inhibits mitochondrial electron transport and increases ROS production. J Biol Chem. 2005;280:37481–37488. doi: 10.1074/jbc.M506412200. [DOI] [PubMed] [Google Scholar]
- 77.Machida K, Cheng KT, Lai CK, Jeng KS, Sung VM, Lai MM. 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. doi: 10.1128/JVI.00321-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Li Y, Boehning DF, Qian T, Popov VL, Weinman SA. Hepatitis C virus core protein increases mitochondrial ROS production by stimulation of Ca2+ uniporter activity. Faseb J. 2007;21:2474–2485. doi: 10.1096/fj.06-7345com. [DOI] [PubMed] [Google Scholar]
- 79.Benali-Furet NL, Chami M, Houel L, De Giorgi F, Vernejoul F, Lagorce D, Buscail L, et al. Hepatitis C virus core triggers apoptosis in liver cells by inducing ER stress and ER calcium depletion. Oncogene. 2005;24:4921–4933. doi: 10.1038/sj.onc.1208673. [DOI] [PubMed] [Google Scholar]
- 80.Tsutsumi T, Matsuda M, Aizaki H, Moriya K, Miyoshi H, Fujie H, Shintani Y, et al. Proteomics analysis of mitochondrial proteins reveals overexpression of a mitochondrial protein chaperon, prohibitin, in cells expressing hepatitis C virus core protein. Hepatology. 2009;50:378–386. doi: 10.1002/hep.22998. [DOI] [PubMed] [Google Scholar]
- 81.Choi J, Forman HJ, Ou JH, Lai MM, Seronello S, Nandipati A. Redox modulation of the hepatitis C virus replication complex is calcium dependent. Free Radic Biol Med. 2006;41:1488–1498. doi: 10.1016/j.freeradbiomed.2006.08.008. [DOI] [PubMed] [Google Scholar]
- 82.Gaspers LD, Memin E, Thomas AP. Calcium-dependent physiologic and pathologic stimulus-metabolic response coupling in hepatocytes. Cell Calcium. 2012;52:93–102. doi: 10.1016/j.ceca.2012.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417:1–13. doi: 10.1042/BJ20081386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kowaltowski AJ, de Souza-Pinto NC, Castilho RF, Vercesi AE. Mitochondria and reactive oxygen species. Free Radic Biol Med. 2009;47:333–343. doi: 10.1016/j.freeradbiomed.2009.05.004. [DOI] [PubMed] [Google Scholar]
- 85.Ruggieri V, Mazzoccoli C, Pazienza V, Andriulli A, Capitanio N, Piccoli C. Hepatitis C virus, mitochondria and auto/mitophagy: Exploiting a host defense mechanism. World J Gastroenterol. 2014;20:2624–2633. doi: 10.3748/wjg.v20.i10.2624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.O'Donnell BV, Tew DG, Jones OT, England PJ. Studies on the inhibitory mechanism of iodonium compounds with special reference to neutrophil NADPH oxidase. Biochem J. 1993;290(Pt 1):41–49. doi: 10.1042/bj2900041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Li Y, Trush MA. Diphenyleneiodonium, an NAD(P)H oxidase inhibitor, also potently inhibits mitochondrial reactive oxygen species production. Biochem Biophys Res Commun. 1998;253:295–299. doi: 10.1006/bbrc.1998.9729. [DOI] [PubMed] [Google Scholar]
- 88.Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol. 2004;4:181–189. doi: 10.1038/nri1312. [DOI] [PubMed] [Google Scholar]
- 89.Bureau C, Bernad J, Chaouche N, Orfila C, Beraud M, Gonindard C, Alric L, et al. Nonstructural 3 protein of hepatitis C virus triggers an oxidative burst in human monocytes via activation of NADPH oxidase. J Biol Chem. 2001;276:23077–23083. doi: 10.1074/jbc.M100698200. [DOI] [PubMed] [Google Scholar]
- 90.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. doi: 10.1189/jlb.0704387. [DOI] [PubMed] [Google Scholar]
- 91.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. doi: 10.1007/s10620-010-1395-y. [DOI] [PubMed] [Google Scholar]
- 92.Lin Q, Fang D, Hou X, Le Y, Fang J, Wen F, Gong W, et al. HCV peptide (C5A), an amphipathic alpha-helical peptide of hepatitis virus C, is an activator of N-formyl peptide receptor in human phagocytes. J Immunol. 2011;186:2087–2094. doi: 10.4049/jimmunol.1002340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Tacke RS, Lee HC, Goh C, Courtney J, Polyak SJ, Rosen HR, Hahn YS. Myeloid suppressor cells induced by hepatitis C virus suppress T-cell responses through the production of reactive oxygen species. Hepatology. 2012;55:343–353. doi: 10.1002/hep.24700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Saba TM. Physiology and physiopathology of the reticuloendothelial system. Arch Intern Med. 1970;126:1031–1052. [PubMed] [Google Scholar]
- 95.Reinehr R, Becker S, Eberle A, Grether-Beck S, Haussinger D. Involvement of NADPH oxidase isoforms and Src family kinases in CD95-dependent hepatocyte apoptosis. J Biol Chem. 2005;280:27179–27194. doi: 10.1074/jbc.M414361200. [DOI] [PubMed] [Google Scholar]
- 96.Murillo MM, Carmona-Cuenca I, Del Castillo G, Ortiz C, Roncero C, Sanchez A, Fernandez M, et al. Activation of NADPH oxidase by transforming growth factor-beta in hepatocytes mediates up-regulation of epidermal growth factor receptor ligands through a nuclear factor-kappaB-dependent mechanism. Biochem J. 2007;405:251–259. doi: 10.1042/BJ20061846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Carmona-Cuenca I, Roncero C, Sancho P, Caja L, Fausto N, Fernandez M, Fabregat I. Upregulation of the NADPH oxidase NOX4 by TGF-beta in hepatocytes is required for its proapoptotic activity. J Hepatol. 2008;49:965–976. doi: 10.1016/j.jhep.2008.07.021. [DOI] [PubMed] [Google Scholar]
- 98.Sancho P, Bertran E, Caja L, Carmona-Cuenca I, Murillo MM, Fabregat I. The inhibition of the epidermal growth factor (EGF) pathway enhances TGF-beta-induced apoptosis in rat hepatoma cells through inducing oxidative stress coincident with a change in the expression pattern of the NADPH oxidases (NOX) isoforms. Biochim Biophys Acta. 2009;1793:253–263. doi: 10.1016/j.bbamcr.2008.09.003. [DOI] [PubMed] [Google Scholar]
- 99.Tariq M, Manzoor S, Ahmed QL, Khalid M, Ashraf W. NOX4 induces oxidative stress and apoptosis through upregulation of caspases 3 and 9 and downregulation of TIGAR in HCV-infected Huh-7 cells. FUTURE VIROLOGY. 2013;8:707–716. [Google Scholar]
- 100.Lambeth JD, Kawahara T, Diebold B. Regulation of Nox and Duox enzymatic activity and expression. Free Radic Biol Med. 2007;43:319–331. doi: 10.1016/j.freeradbiomed.2007.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sancho P, Mainez J, Crosas-Molist E, Roncero C, Fernandez-Rodriguez CM, Pinedo F, Huber H, et al. NADPH oxidase NOX4 mediates stellate cell activation and hepatocyte cell death during liver fibrosis development. PLoS One. 2012;7:e45285. doi: 10.1371/journal.pone.0045285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Nelson DR, Gonzalez-Peralta RP, Qian K, Xu Y, Marousis CG, Davis GL, Lau JY. Transforming growth factor-beta 1 in chronic hepatitis C. J Viral Hepat. 1997;4:29–35. doi: 10.1046/j.1365-2893.1997.00124.x. [DOI] [PubMed] [Google Scholar]
- 103.Taniguchi H, Kato N, Otsuka M, Goto T, Yoshida H, Shiratori Y, Omata M. Hepatitis C virus core protein upregulates transforming growth factor-beta 1 transcription. J Med Virol. 2004;72:52–59. doi: 10.1002/jmv.10545. [DOI] [PubMed] [Google Scholar]
- 104.Lin W, Tsai WL, Shao RX, Wu G, Peng LF, Barlow LL, Chung WJ, 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. doi: 10.1053/j.gastro.2010.03.008. 2518 e2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Presser LD, McRae S, Waris G. Activation of TGF-beta1 promoter by hepatitis C virus-induced AP-1 and Sp1: role of TGF-beta1 in hepatic stellate cell activation and invasion. PLoS One. 2013;8:e56367. doi: 10.1371/journal.pone.0056367. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 106.Hecker L, Vittal R, Jones T, Jagirdar R, Luckhardt TR, Horowitz JC, Pennathur S, et al. NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med. 2009;15:1077–1081. doi: 10.1038/nm.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.von Lohneysen K, Noack D, Hayes P, Friedman JS, Knaus UG. Constitutive NADPH oxidase 4 activity resides in the composition of the B-loop and the penultimate C terminus. J Biol Chem. 2012;287:8737–8745. doi: 10.1074/jbc.M111.332494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Takac I, Schroder K, Zhang L, Lardy B, Anilkumar N, Lambeth JD, Shah AM, et al. The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4. J Biol Chem. 2011;286:13304–13313. doi: 10.1074/jbc.M110.192138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kuroda J, Nakagawa K, Yamasaki T, Nakamura K, Takeya R, Kuribayashi F, Imajoh-Ohmi S, et al. The superoxide-producing NAD(P)H oxidase Nox4 in the nucleus of human vascular endothelial cells. Genes Cells. 2005;10:1139–1151. doi: 10.1111/j.1365-2443.2005.00907.x. [DOI] [PubMed] [Google Scholar]
- 110.Sturrock A, Huecksteadt TP, Norman K, Sanders K, Murphy TM, Chitano P, Wilson K, et al. Nox4 mediates TGF-beta1-induced retinoblastoma protein phosphorylation, proliferation, and hypertrophy in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1543–L1555. doi: 10.1152/ajplung.00430.2006. [DOI] [PubMed] [Google Scholar]
- 111.Liu RM, Choi J, Wu JH, Gaston Pravia KA, Lewis KM, Brand JD, Mochel NS, et al. Oxidative modification of nuclear mitogen-activated protein kinase phosphatase 1 is involved in transforming growth factor beta1-induced expression of plasminogen activator inhibitor 1 in fibroblasts. J Biol Chem. 2010;285:16239–16247. doi: 10.1074/jbc.M110.111732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gordillo G, Fang H, Park H, Roy S. Nox-4-dependent nuclear H2O2 drives DNA oxidation resulting in 8-OHdG as urinary biomarker and hemangioendothelioma formation. Antioxid Redox Signal. 2010;12:933–943. doi: 10.1089/ars.2009.2917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Pendergrass KD, Gwathmey TM, Michalek RD, Grayson JM, Chappell MC. The angiotensin II-AT1 receptor stimulates reactive oxygen species within the cell nucleus. Biochem Biophys Res Commun. 2009;384:149–154. doi: 10.1016/j.bbrc.2009.04.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang L, Nguyen MV, Lardy B, Jesaitis AJ, Grichine A, Rousset F, Talbot M, et al. New insight into the Nox4 subcellular localization in HEK293 cells: First monoclonal antibodies against Nox4. Biochimie. 2011;93:457–468. doi: 10.1016/j.biochi.2010.11.001. [DOI] [PubMed] [Google Scholar]
- 115.Sipkens JA, Hahn N, van den Brand CS, Meischl C, Cillessen SA, Smith DE, Juffermans LJ, et al. Homocysteine-induced apoptosis in endothelial cells coincides with nuclear NOX2 and peri-nuclear NOX4 activity. Cell Biochem Biophys. 2011;67:341–352. doi: 10.1007/s12013-011-9297-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Spencer NY, Yan Z, Boudreau RL, Zhang Y, Luo M, Li Q, Tian X, et al. Control of hepatic nuclear superoxide production by glucose 6-phosphate dehydrogenase and NADPH oxidase-4. J Biol Chem. 2011;286:8977–8987. doi: 10.1074/jbc.M110.193821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Anilkumar N, San Jose G, Sawyer I, Santos CX, Sand C, Brewer AC, Warren D, et al. A 28-kDa splice variant of NADPH oxidase-4 is nuclear-localized and involved in redox signaling in vascular cells. Arterioscler Thromb Vasc Biol. 2013;33:e104–e112. doi: 10.1161/ATVBAHA.112.300956. [DOI] [PubMed] [Google Scholar]
- 118.Goyal P, Weissmann N, Rose F, Grimminger F, Schafers HJ, Seeger W, Hanze J. Identification of novel Nox4 splice variants with impact on ROS levels in A549 cells. Biochem Biophys Res Commun. 2005;329:32–39. doi: 10.1016/j.bbrc.2005.01.089. [DOI] [PubMed] [Google Scholar]
- 119.Choi DH, Cristovao AC, Guhathakurta S, Lee J, Joh TH, Beal MF, Kim YS. NADPH oxidase 1-mediated oxidative stress leads to dopamine neuron death in Parkinson's disease. Antioxid Redox Signal. 2012;16:1033–1045. doi: 10.1089/ars.2011.3960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Chamulitrat W, Schmidt R, Tomakidi P, Stremmel W, Chunglok W, Kawahara T, Rokutan K. Association of gp91phox homolog Nox1 with anchorage-independent growth and MAP kinase-activation of transformed human keratinocytes. Oncogene. 2003;22:6045–6053. doi: 10.1038/sj.onc.1206654. [DOI] [PubMed] [Google Scholar]
- 121.Gochee PA, Jonsson JR, Clouston AD, Pandeya N, Purdie DM, Powell EE. Steatosis in chronic hepatitis C: association with increased messenger RNA expression of collagen I, tumor necrosis factor-alpha and cytochrome P450 2E1. J Gastroenterol Hepatol. 2003;18:386–392. doi: 10.1046/j.1440-1746.2003.02984.x. [DOI] [PubMed] [Google Scholar]
- 122.Anderson GR, Hasan A, Yin H, Qadri I, Quattrochi LC. Regulation of the CYP1A1 gene by 2,3,7,8-tetrachlorodibenzo-p-dioxin but not by beta-naphthoflavone or 3-methylcholanthrene is altered in hepatitis C virus replicon-expressing cells. Mol Pharmacol. 2006;70:1062–1070. doi: 10.1124/mol.106.024125. [DOI] [PubMed] [Google Scholar]
- 123.Cederbaum AI, Defeng W, Montserrat M, Jingxiang B. CYP2E1-dependent toxicity and oxidative stress in HepG2 cells. Free Radic Biol Med. 2001;31:1539–1543. doi: 10.1016/s0891-5849(01)00743-2. [DOI] [PubMed] [Google Scholar]
- 124.Lieber CS. Role of oxidative stress and antioxidant therapy in alcoholic and nonalcoholic liver diseases. Adv Pharmacol. 1997;38:601–628. doi: 10.1016/s1054-3589(08)61001-7. [DOI] [PubMed] [Google Scholar]
- 125.Otani K, Korenaga M, Beard MR, Li K, Qian T, Showalter LA, Singh AK, et al. Hepatitis C virus core protein, cytochrome P450 2E1, and alcohol produce combined mitochondrial injury and cytotoxicity in hepatoma cells. Gastroenterology. 2005;128:96–107. doi: 10.1053/j.gastro.2004.10.045. [DOI] [PubMed] [Google Scholar]
- 126.Rigamonti C, Mottaran E, Reale E, Rolla R, Cipriani V, Capelli F, Boldorini R, et al. Moderate alcohol consumption increases oxidative stress in patients with chronic hepatitis C. Hepatology. 2003;38:42–49. doi: 10.1053/jhep.2003.50275. [DOI] [PubMed] [Google Scholar]
- 127.Higgs MR, Lerat H, Pawlotsky JM. Hepatitis C virus-induced activation of beta-catenin promotes c-Myc expression and a cascade of pro-carcinogenetic events. Oncogene. 2013;32:4683–4693. doi: 10.1038/onc.2012.484. [DOI] [PubMed] [Google Scholar]
- 128.Novitskiy G, Traore K, Wang L, Trush MA, Mezey E. Effects of ethanol and acetaldehyde on reactive oxygen species production in rat hepatic stellate cells. Alcohol Clin Exp Res. 2006;30:1429–1435. doi: 10.1111/j.1530-0277.2006.00171.x. [DOI] [PubMed] [Google Scholar]
- 129.Seronello S, Sheikh MY, Choi J. Redox regulation of hepatitis C in nonalcoholic and alcoholic liver. Free Radic Biol Med. 2007;43:869–882. doi: 10.1016/j.freeradbiomed.2007.05.036. [DOI] [PubMed] [Google Scholar]
- 130.Choi J. Oxidative stress, endogenous antioxidants, alcohol, and hepatitis C: pathogenic interactions and therapeutic considerations. Free Radic Biol Med. 2012;52:1135–1150. doi: 10.1016/j.freeradbiomed.2012.01.008. [DOI] [PubMed] [Google Scholar]
- 131.Farinati F, Cardin R, Degan P, De Maria N, Floyd RA, Van Thiel DH, Naccarato R. Oxidative DNA damage in circulating leukocytes occurs as an early event in chronic HCV infection. Free Radic Biol Med. 1999;27:1284–1291. doi: 10.1016/s0891-5849(99)00161-6. [DOI] [PubMed] [Google Scholar]
- 132.Cardin R, Saccoccio G, Masutti F, Bellentani S, Farinati F, Tiribelli C. DNA oxidative damage in leukocytes correlates with the severity of HCV- related liver disease: validation in an open population study. J Hepatol. 2001;34:587–592. doi: 10.1016/s0168-8278(00)00098-2. [DOI] [PubMed] [Google Scholar]
- 133.Kato J, Kobune M, Nakamura T, Kuroiwa G, Takada K, Takimoto R, Sato Y, et al. Normalization of elevated hepatic 8-hydroxy-2'-deoxyguanosine levels in chronic hepatitis C patients by phlebotomy and low iron diet. Cancer Res. 2001;61:8697–8702. [PubMed] [Google Scholar]
- 134.Moriya K, Miyoshi H, Shinzawa S, Tsutsumi T, Fujie H, Goto K, Shintani Y, et al. Hepatitis C virus core protein compromises iron-induced activation of antioxidants in mice and HepG2 cells. J Med Virol. 2010;82:776–792. doi: 10.1002/jmv.21661. [DOI] [PubMed] [Google Scholar]
- 135.Carvajal-Yepes M, Himmelsbach K, Schaedler S, Ploen D, Krause J, Ludwig L, Weiss T, et al. Hepatitis C virus impairs the induction of cytoprotective nrf2 target genes by delocalization of small maf proteins. J Biol Chem. 2011;286:8941–8951. doi: 10.1074/jbc.M110.186684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Burdette D, Haskett A, Presser L, McRae S, Iqbal J, Waris G. Hepatitis C virus activates interleukin-1beta via caspase-1-inflammasome complex. J Gen Virol. 2012;93:235–246. doi: 10.1099/vir.0.034033-0. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 137.Abdalla MY, Ahmad IM, Spitz DR, Schmidt WN, Britigan BE. Hepatitis C virus-core and non structural proteins lead to different effects on cellular antioxidant defenses. J Med Virol. 2005;76:489–497. doi: 10.1002/jmv.20388. [DOI] [PubMed] [Google Scholar]
- 138.Elchuri S, Oberley TD, Qi W, Eisenstein RS, Jackson Roberts L, Van Remmen H, Epstein CJ, et al. CuZnSOD deficiency leads to persistent and widespread oxidative damage and hepatocarcinogenesis later in life. Oncogene. 2005;24:367–380. doi: 10.1038/sj.onc.1208207. [DOI] [PubMed] [Google Scholar]
- 139.Xu Z, Chen L, Leung L, Yen TS, Lee C, Chan JY. Liver-specific inactivation of the Nrf1 gene in adult mouse leads to nonalcoholic steatohepatitis and hepatic neoplasia. Proc Natl Acad Sci U S A. 2005;102:4120–4125. doi: 10.1073/pnas.0500660102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Chung JS, Park S, Park SH, Park ER, Cha PH, Kim BY, Chung YM, et al. Overexpression of Romo1 promotes production of reactive oxygen species and invasiveness of hepatic tumor cells. Gastroenterology. 2012;143:1084–1094. doi: 10.1053/j.gastro.2012.06.038. e1087. [DOI] [PubMed] [Google Scholar]
- 141.Ghosh D, Choudhury ST, Ghosh S, Mandal AK, Sarkar S, Ghosh A, Saha KD, et al. Nanocapsulated curcumin: oral chemopreventive formulation against diethylnitrosamine induced hepatocellular carcinoma in rat. Chem Biol Interact. 2011;195:206–214. doi: 10.1016/j.cbi.2011.12.004. [DOI] [PubMed] [Google Scholar]
- 142.Lin H, Liu XB, Yu JJ, Hua F, Hu ZW. Antioxidant N-acetylcysteine attenuates hepatocarcinogenesis by inhibiting ROS/ER stress in TLR2 deficient mouse. PLoS One. 2013;8:e74130. doi: 10.1371/journal.pone.0074130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Arsura M, Cavin LG. Nuclear factor-kappaB and liver carcinogenesis. Cancer Lett. 2005;229:157–169. doi: 10.1016/j.canlet.2005.07.008. [DOI] [PubMed] [Google Scholar]
- 144.Tsuda H, Fukushima S, Imaida K, Sakata T, Ito N. Modification of carcinogenesis by antioxidants and other compounds. Acta Pharmacol Toxicol (Copenh) 1984;55(Suppl 2):125–143. doi: 10.1111/j.1600-0773.1984.tb02486.x. [DOI] [PubMed] [Google Scholar]
- 145.Yu LX, Yan HX, Liu Q, Yang W, Wu HP, Dong W, Tang L, et al. Endotoxin accumulation prevents carcinogen-induced apoptosis and promotes liver tumorigenesis in rodents. Hepatology. 2010;52:1322–1333. doi: 10.1002/hep.23845. [DOI] [PubMed] [Google Scholar]
- 146.Sakurai T, Kudo M, Watanabe T, Itoh K, Higashitsuji H, Arizumi T, Inoue T, et al. Hypothermia Protects against Fulminant Hepatitis in Mice by Reducing Reactive Oxygen Species Production. Dig Dis. 2013;31:440–446. doi: 10.1159/000355242. [DOI] [PubMed] [Google Scholar]
- 147.Vafa O, Wade M, Kern S, Beeche M, Pandita TK, Hampton GM, Wahl GM. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol Cell. 2002;9:1031–1044. doi: 10.1016/s1097-2765(02)00520-8. [DOI] [PubMed] [Google Scholar]
- 148.Factor VM, Laskowska D, Jensen MR, Woitach JT, Popescu NC, Thorgeirsson SS. Vitamin E reduces chromosomal damage and inhibits hepatic tumor formation in a transgenic mouse model. Proc Natl Acad Sci U S A. 2000;97:2196–2201. doi: 10.1073/pnas.040428797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Calvisi DF, Ladu S, Hironaka K, Factor VM, Thorgeirsson SS. Vitamin E down-modulates iNOS and NADPH oxidase in c-Myc/TGF-alpha transgenic mouse model of liver cancer. J Hepatol. 2004;41:815–822. doi: 10.1016/j.jhep.2004.07.030. [DOI] [PubMed] [Google Scholar]
- 150.Klohn PC, Massalha H, Neumann HG. A metabolite of carcinogenic 2-acetylaminofluorene, 2-nitrosofluorene, induces redox cycling in mitochondria. Biochim Biophys Acta. 1995;1229:363–372. doi: 10.1016/0005-2728(95)00019-f. [DOI] [PubMed] [Google Scholar]
- 151.Kuo MT, Liu Z, Wei Y, Lin-Lee YC, Tatebe S, Mills GB, Unate H. Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-kappaB signaling. Oncogene. 2002;21:1945–1954. doi: 10.1038/sj.onc.1205117. [DOI] [PubMed] [Google Scholar]
- 152.Shen HM, Shi CY, Shen Y, Ong CN. Detection of elevated reactive oxygen species level in cultured rat hepatocytes treated with aflatoxin B1. Free Radic Biol Med. 1996;21:139–146. doi: 10.1016/0891-5849(96)00019-6. [DOI] [PubMed] [Google Scholar]
- 153.Swietek K, Juszczyk J. Reduced glutathione concentration in erythrocytes of patients with acute and chronic viral hepatitis. J Viral Hepat. 1997;4:139–141. doi: 10.1111/j.1365-2893.1997.tb00217.x. [DOI] [PubMed] [Google Scholar]
- 154.Eruslanov E, Kusmartsev S. Identification of ROS using oxidized DCFDA and flow-cytometry. Methods Mol Biol. 2010;594:57–72. doi: 10.1007/978-1-60761-411-1_4. [DOI] [PubMed] [Google Scholar]
- 155.Yano M, Hayashi H, Wakusawa S, Sanae F, Takikawa T, Shiono Y, Arao M, et al. Long term effects of phlebotomy on biochemical and histological parameters of chronic hepatitis C. Am J Gastroenterol. 2002;97:133–137. doi: 10.1111/j.1572-0241.2002.05436.x. [DOI] [PubMed] [Google Scholar]
- 156.Kato J, Miyanishi K, Kobune M, Nakamura T, Takada K, Takimoto R, Kawano Y, et al. Long-term phlebotomy with low-iron diet therapy lowers risk of development of hepatocellular carcinoma from chronic hepatitis C. J Gastroenterol. 2007;42:830–836. doi: 10.1007/s00535-007-2095-z. [DOI] [PubMed] [Google Scholar]
- 157.Hayashi H, Takikawa T, Nishimura N, Yano M, Isomura T, Sakamoto N. Improvement of serum aminotransferase levels after phlebotomy in patients with chronic active hepatitis C and excess hepatic iron. Am J Gastroenterol. 1994;89:986–988. [PubMed] [Google Scholar]
- 158.Nishina S, Korenaga M, Hidaka I, Shinozaki A, Sakai A, Gondo T, Tabuchi M, et al. Hepatitis C virus protein and iron overload induce hepatic steatosis through the unfolded protein response in mice. Liver Int. 2010;30:683–692. doi: 10.1111/j.1478-3231.2010.02210.x. [DOI] [PubMed] [Google Scholar]
- 159.Melhem A, Stern M, Shibolet O, Israeli E, Ackerman Z, Pappo O, Hemed N, 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. doi: 10.1097/01.mcg.0000174023.73472.29. [DOI] [PubMed] [Google Scholar]
- 160.Falasca K, Ucciferri C, Mancino P, Vitacolonna E, De Tullio D, Pizzigallo E, Conti P, et al. Treatment with silybin-vitamin E-phospholipid complex in patients with hepatitis C infection. J Med Virol. 2008;80:1900–1906. doi: 10.1002/jmv.21292. [DOI] [PubMed] [Google Scholar]
- 161.Gane EJ, Weilert F, Orr DW, Keogh GF, Gibson M, Lockhart MM, Frampton CM, et al. The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase II study of hepatitis C patients. Liver Int. 2010;30:1019–1026. doi: 10.1111/j.1478-3231.2010.02250.x. [DOI] [PubMed] [Google Scholar]
- 162.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2012;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 163.Valko M, Izakovic M, Mazur M, Rhodes CJ, Telser J. Role of oxygen radicals in DNA damage and cancer incidence. Mol Cell Biochem. 2004;266:37–56. doi: 10.1023/b:mcbi.0000049134.69131.89. [DOI] [PubMed] [Google Scholar]
- 164.Burney S, Caulfield JL, Niles JC, Wishnok JS, Tannenbaum SR. The chemistry of DNA damage from nitric oxide and peroxynitrite. Mutat Res. 1999;424:37–49. doi: 10.1016/s0027-5107(99)00006-8. [DOI] [PubMed] [Google Scholar]
- 165.Schwarz KB, Kew M, Klein A, Abrams RA, Sitzmann J, Jones L, Sharma S, et al. Increased hepatic oxidative DNA damage in patients with hepatocellular carcinoma. Dig Dis Sci. 2001;46:2173–2178. doi: 10.1023/a:1011958814371. [DOI] [PubMed] [Google Scholar]
- 166.Lim SO, Gu JM, Kim MS, Kim HS, Park YN, Park CK, Cho JW, et al. Epigenetic changes induced by reactive oxygen species in hepatocellular carcinoma: methylation of the Ecadherin promoter. Gastroenterology. 2008;135:2128–2140. doi: 10.1053/j.gastro.2008.07.027. 2140 e2121–2128. [DOI] [PubMed] [Google Scholar]
- 167.Matsumoto K, Satoh Y, Sugo H, Takamori S, Kojima K, Fukasawa M, Beppu T, et al. Immunohistochemical study of the relationship between 8-hydroxy-2'-deoxyguanosine levels in noncancerous region and postoperative recurrence of hepatocellular carcinoma in remnant liver. Hepatol Res. 2003;25:435–441. doi: 10.1016/s1386-6346(02)00313-3. [DOI] [PubMed] [Google Scholar]
- 168.Machida K, Cheng KT, Sung VM, Shimodaira S, Lindsay KL, Levine AM, Lai MY, et al. Hepatitis C virus induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes. Proc Natl Acad Sci U S A. 2004;101:4262–4267. doi: 10.1073/pnas.0303971101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Hara Y, Hino K, Okuda M, Furutani T, Hidaka I, Yamaguchi Y, Korenaga M, et al. Hepatitis C virus core protein inhibits deoxycholic acid-mediated apoptosis despite generating mitochondrial reactive oxygen species. J Gastroenterol. 2006;41:257–268. doi: 10.1007/s00535-005-1738-1. [DOI] [PubMed] [Google Scholar]
- 170.Mahmood S, Kawanaka M, Kamei A, Izumi A, Nakata K, Niiyama G, Ikeda H, et al. Immunohistochemical evaluation of oxidative stress markers in chronic hepatitis C. Antioxid Redox Signal. 2004;6:19–24. doi: 10.1089/152308604771978318. [DOI] [PubMed] [Google Scholar]
- 171.Machida K, McNamara G, Cheng KT, Huang J, Wang CH, Comai L, Ou JH, et al. Hepatitis C virus inhibits DNA damage repair through reactive oxygen and nitrogen species and by interfering with the ATM-NBS1/Mre11/Rad50 DNA repair pathway in monocytes and hepatocytes. J Immunol. 2010;185:6985–6998. doi: 10.4049/jimmunol.1000618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Egler RA, Fernandes E, Rothermund K, Sereika S, de Souza-Pinto N, Jaruga P, Dizdaroglu M, et al. Regulation of reactive oxygen species, DNA damage, and c-Myc function by peroxiredoxin 1. Oncogene. 2005;24:8038–8050. doi: 10.1038/sj.onc.1208821. [DOI] [PubMed] [Google Scholar]
- 173.Kershaw RM, Hodges NJ. Repair of oxidative DNA damage is delayed in the Ser326Cys polymorphic variant of the base excision repair protein OGG1. Mutagenesis. 2012;27:501–510. doi: 10.1093/mutage/ges012. [DOI] [PubMed] [Google Scholar]
- 174.Jung SW, Park NH, Shin JW, Park BR, Kim CJ, Lee JE, Shin ES, et al. Polymorphisms of DNA repair genes in Korean hepatocellular carcinoma patients with chronic hepatitis B: possible implications on survival. J Hepatol. 2012;57:621–627. doi: 10.1016/j.jhep.2012.04.039. [DOI] [PubMed] [Google Scholar]
- 175.Romilda C, Marika P, Alessandro S, Enrico L, Marina B, Andromachi K, Umberto C, et al. Oxidative DNA damage correlates with cell immortalization and mir-92 expression in hepatocellular carcinoma. BMC Cancer. 2012;12:177. doi: 10.1186/1471-2407-12-177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Bravard A, Vacher M, Gouget B, Coutant A, de Boisferon FH, Marsin S, Chevillard S, et al. Redox regulation of human OGG1 activity in response to cellular oxidative stress. Mol Cell Biol. 2006;26:7430–7436. doi: 10.1128/MCB.00624-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Jaiswal M, LaRusso NF, Nishioka N, Nakabeppu Y, Gores GJ. Human Ogg1, a protein involved in the repair of 8-oxoguanine, is inhibited by nitric oxide. Cancer Res. 2001;61:6388–6393. [PubMed] [Google Scholar]
- 178.Girard PM, Graindorge D, Smirnova V, Rigolet P, Francesconi S, Scanlon S, Sage E. Oxidative stress in mammalian cells impinges on the cysteines redox state of human XRCC3 protein and on its cellular localization. PLoS One. 2013;8:e75751. doi: 10.1371/journal.pone.0075751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Aoki H, Hayashi J, Moriyama M, Arakawa Y, Hino O. Hepatitis C virus core protein interacts with 14-3-3 protein and activates the kinase Raf-1. J Virol. 2000;74:1736–1741. doi: 10.1128/jvi.74.4.1736-1741.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Yang X, Zou P, Yao J, Yun D, Bao H, Du R, Long J, et al. Proteomic dissection of cell type-specific H2AX-interacting protein complex associated with hepatocellular carcinoma. J Proteome Res. 2010;9:1402–1415. doi: 10.1021/pr900932y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Kim JS, Diebold BA, Babior BM, Knaus UG, Bokoch GM. Regulation of Nox1 activity via protein kinase A-mediated phosphorylation of NoxA1 and 14-3-3 binding. J Biol Chem. 2007;282:34787–34800. doi: 10.1074/jbc.M704754200. [DOI] [PubMed] [Google Scholar]
- 182.Kang MA, So EY, Simons AL, Spitz DR, Ouchi T. DNA damage induces reactive oxygen species generation through the H2AX-Nox1/Rac1 pathway. Cell Death Dis. 2012;3:e249. doi: 10.1038/cddis.2011.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Strathdee G. Epigenetic versus genetic alterations in the inactivation of E-cadherin. Semin Cancer Biol. 2002;12:373–379. doi: 10.1016/s1044-579x(02)00057-3. [DOI] [PubMed] [Google Scholar]
- 184.Min JY, Lim SO, Jung G. Downregulation of catalase by reactive oxygen species via hypermethylation of CpG island II on the catalase promoter. FEBS Lett. 2011;584:2427–2432. doi: 10.1016/j.febslet.2010.04.048. [DOI] [PubMed] [Google Scholar]
- 185.Llovet JM, Chen Y, Wurmbach E, Roayaie S, Fiel MI, Schwartz M, Thung SN, et al. A molecular signature to discriminate dysplastic nodules from early hepatocellular carcinoma in HCV cirrhosis. Gastroenterology. 2006;131:1758–1767. doi: 10.1053/j.gastro.2006.09.014. [DOI] [PubMed] [Google Scholar]
- 186.Bose SK, Meyer K, Di Bisceglie AM, Ray RB, Ray R. Hepatitis C virus induces epithelial-mesenchymal transition in primary human hepatocytes. J Virol. 2012;86:13621–13628. doi: 10.1128/JVI.02016-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ripoli M, Barbano R, Balsamo T, Piccoli C, Brunetti V, Coco M, Mazzoccoli G, et al. Hypermethylated levels of E-cadherin promoter in Huh-7 cells expressing the HCV core protein. Virus Res. 2011;160:74–81. doi: 10.1016/j.virusres.2011.05.014. [DOI] [PubMed] [Google Scholar]
- 188.Benegiamo G, Vinciguerra M, Mazzoccoli G, Piepoli A, Andriulli A, Pazienza V. DNA methyltransferases 1 and 3b expression in Huh-7 cells expressing HCV core protein of different genotypes. Dig Dis Sci. 2012;57:1598–1603. doi: 10.1007/s10620-012-2160-1. [DOI] [PubMed] [Google Scholar]
- 189.Okamoto Y, Shinjo K, Shimizu Y, Sano T, Yamao K, Gao W, Fujii M, et al. Hepatitis Virus Infection Affects DNA Methylation in Mice with Humanized Livers. Gastroenterology. 2013;146:562–572. doi: 10.1053/j.gastro.2013.10.056. [DOI] [PubMed] [Google Scholar]
- 190.Bility MT, Zhang L, Washburn ML, Curtis TA, Kovalev GI, Su L. Generation of a humanized mouse model with both human immune system and liver cells to model hepatitis C virus infection and liver immunopathogenesis. Nat Protoc. 2012;7:1608–1617. doi: 10.1038/nprot.2012.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Dorner M, Horwitz JA, Donovan BM, Labitt RN, Budell WC, Friling T, Vogt A, et al. Completion of the entire hepatitis C virus life cycle in genetically humanized mice. Nature. 2013;501:237–241. doi: 10.1038/nature12427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Mengshol JA, Golden-Mason L, Arikawa T, Smith M, Niki T, McWilliams R, Randall JA, et al. A crucial role for Kupffer cell-derived galectin-9 in regulation of T cell immunity in hepatitis C infection. PLoS One. 2010;5:e9504. doi: 10.1371/journal.pone.0009504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Roberts JM, Searle JW, Cooksley WG. Histological patterns of prolonged hepatitis C infection. Gastroenterol Jpn. 1993;28(Suppl 5):37–41. doi: 10.1007/BF02989202. [DOI] [PubMed] [Google Scholar]
- 194.Heydtmann M. Macrophages in hepatitis B and hepatitis C virus infections. J Virol. 2009;83:2796–2802. doi: 10.1128/JVI.00996-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Dolganiuc A, Norkina O, Kodys K, Catalano D, Bakis G, Marshall C, Mandrekar P, et al. Viral and host factors induce macrophage activation and loss of toll-like receptor tolerance in chronic HCV infection. Gastroenterology. 2007;133:1627–1636. doi: 10.1053/j.gastro.2007.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Reynolds WF, Patel K, Pianko S, Blatt LM, Nicholas JJ, McHutchison JG. A genotypic association implicates myeloperoxidase in the progression of hepatic fibrosis in chronic hepatitis C virus infection. Genes Immun. 2002;3:345–349. doi: 10.1038/sj.gene.6363880. [DOI] [PubMed] [Google Scholar]
- 197.Nahon P, Sutton A, Rufat P, Charnaux N, Mansouri A, Moreau R, Ganne-Carrie N, et al. A variant in myeloperoxidase promoter hastens the emergence of hepatocellular carcinoma in patients with HCV-related cirrhosis. J Hepatol. 2012;56:426–432. doi: 10.1016/j.jhep.2011.08.010. [DOI] [PubMed] [Google Scholar]
- 198.Wisniewska-Ligier M, Wozniakowska-Gesicka T, Glowacka E, Lewkowicz P, Banasik M, Tchorzewski H. Involvement of innate immunity in the pathogenesis of chronic hepatitis C in children. Scand J Immunol. 2006;64:425–432. doi: 10.1111/j.1365-3083.2006.01800.x. [DOI] [PubMed] [Google Scholar]
- 199.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. doi: 10.1046/j.1365-2362.2001.00747.x. [DOI] [PubMed] [Google Scholar]
- 200.Simonin Y, Vegna S, Akkari L, Gregoire D, Antoine E, Piette J, Floc'h N, et al. Lymphotoxin signaling is initiated by the viral polymerase in HCV-linked tumorigenesis. PLoS Pathog. 2013;9:e1003234. doi: 10.1371/journal.ppat.1003234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Haybaeck J, Zeller N, Wolf MJ, Weber A, Wagner U, Kurrer MO, Bremer J, et al. A lymphotoxin-driven pathway to hepatocellular carcinoma. Cancer Cell. 2009;16:295–308. doi: 10.1016/j.ccr.2009.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Tai DI, Tsai SL, Chang YH, Huang SN, Chen TC, Chang KS, Liaw YF. Constitutive activation of nuclear factor kappaB in hepatocellular carcinoma. Cancer. 2000;89:2274–2281. [PubMed] [Google Scholar]
- 203.Staal FJ, Roederer M, Herzenberg LA. Intracellular thiols regulate activation of nuclear factor kappa B and transcription of human immunodeficiency virus. Proc Natl Acad Sci U S A. 1990;87:9943–9947. doi: 10.1073/pnas.87.24.9943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Jamaluddin M, Wang S, Boldogh I, Tian B, Brasier AR. TNF-alpha-induced NF-kappaB/RelA Ser(276) phosphorylation and enhanceosome formation is mediated by an ROS-dependent PKAc pathway. Cell Signal. 2007;19:1419–1433. doi: 10.1016/j.cellsig.2007.01.020. [DOI] [PubMed] [Google Scholar]
- 205.Herscovitch M, Comb W, Ennis T, Coleman K, Yong S, Armstead B, Kalaitzidis D, et al. Intermolecular disulfide bond formation in the NEMO dimer requires Cys54 and Cys347. Biochem Biophys Res Commun. 2008;367:103–108. doi: 10.1016/j.bbrc.2007.12.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Matthews JR, Kaszubska W, Turcatti G, Wells TN, Hay RT. Role of cysteine62 in DNA recognition by the P50 subunit of NF-kappa B. Nucleic Acids Res. 1993;21:1727–1734. doi: 10.1093/nar/21.8.1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Gloire G, Piette J. Redox regulation of nuclear post-translational modifications during NF-kappaB activation. Antioxid Redox Signal. 2009;11:2209–2222. doi: 10.1089/ars.2009.2463. [DOI] [PubMed] [Google Scholar]
- 208.Spencer NY, Zhou W, Li Q, Zhang Y, Luo M, Yan Z, Lynch TJ, et al. Hepatocytes produce TNF-alpha following hypoxia-reoxygenation and liver ischemia-reperfusion in a NADPH oxidase- and c-Src-dependent manner. Am J Physiol Gastrointest Liver Physiol. 2013;305:G84–G94. doi: 10.1152/ajpgi.00430.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Knobler H, Schattner A. TNF-{alpha}, chronic hepatitis C and diabetes: a novel triad. Qjm. 2005;98:1–6. doi: 10.1093/qjmed/hci001. [DOI] [PubMed] [Google Scholar]
- 210.Capone F, Costantini S, Guerriero E, Calemma R, Napolitano M, Scala S, Izzo F, et al. Serum cytokine levels in patients with hepatocellular carcinoma. Eur Cytokine Netw. 2010;21:99–104. doi: 10.1684/ecn.2010.0192. [DOI] [PubMed] [Google Scholar]
- 211.Negash AA, Ramos HJ, Crochet N, Lau DT, Doehle B, Papic N, Delker DA, et al. IL-1beta production through the NLRP3 inflammasome by hepatic macrophages links hepatitis C virus infection with liver inflammation and disease. PLoS Pathog. 2013;9:e1003330. doi: 10.1371/journal.ppat.1003330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Tsutsumi T, Suzuki T, Moriya K, Yotsuyanagi H, Shintani Y, Fujie H, Matsuura Y, et al. Alteration of intrahepatic cytokine expression and AP-1 activation in transgenic mice expressing hepatitis C virus core protein. Virology. 2002;304:415–424. doi: 10.1006/viro.2002.1702. [DOI] [PubMed] [Google Scholar]
- 213.Liu RM, Gaston Pravia KA. Oxidative stress and glutathione in TGF-beta-mediated fibrogenesis. Free Radic Biol Med. 2010;48:1–15. doi: 10.1016/j.freeradbiomed.2009.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Yoshida T, Hanada T, Tokuhisa T, Kosai K, Sata M, Kohara M, Yoshimura A. Activation of STAT3 by the hepatitis C virus core protein leads to cellular transformation. J Exp Med. 2002;196:641–653. doi: 10.1084/jem.20012127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Qadri I, Iwahashi M, Capasso JM, Hopken MW, Flores S, Schaack J, Simon FR. 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. doi: 10.1042/BJ20031587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.McCubrey JA, Lahair MM, Franklin RA. Reactive oxygen species-induced activation of the MAP kinase signaling pathways. Antioxid Redox Signal. 2006;8:1775–1789. doi: 10.1089/ars.2006.8.1775. [DOI] [PubMed] [Google Scholar]
- 217.Rubartelli A. Redox control of NLRP3 inflammasome activation in health and disease. J Leukoc Biol. 2012;92:951–958. doi: 10.1189/jlb.0512265. [DOI] [PubMed] [Google Scholar]
- 218.Sokolovska A, Becker CE, Ip WK, Rathinam VA, Brudner M, Paquette N, Tanne A, et al. Activation of caspase-1 by the NLRP3 inflammasome regulates the NADPH oxidase NOX2 to control phagosome function. Nat Immunol. 2013;14:543–553. doi: 10.1038/ni.2595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Teufelhofer O, Parzefall W, Kainzbauer E, Ferk F, Freiler C, Knasmuller S, Elbling L, et al. Superoxide generation from Kupffer cells contributes to hepatocarcinogenesis: studies on NADPH oxidase knockout mice. Carcinogenesis. 2005;26:319–329. doi: 10.1093/carcin/bgh320. [DOI] [PubMed] [Google Scholar]
- 220.Szabo G, Wands JR, Eken A, Osna NA, Weinman SA, Machida K, Joe Wang H. Alcohol and hepatitis C virus--interactions in immune dysfunctions and liver damage. Alcohol Clin Exp Res. 2010;34:1675–1686. doi: 10.1111/j.1530-0277.2010.01255.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.DeLeo FR, Renee J, McCormick S, Nakamura M, Apicella M, Weiss JP, Nauseef WM. Neutrophils exposed to bacterial lipopolysaccharide upregulate NADPH oxidase assembly. J Clin Invest. 1998;101:455–463. doi: 10.1172/JCI949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Dang PM, Stensballe A, Boussetta T, Raad H, Dewas C, Kroviarski Y, Hayem G, et al. A specific p47phox -serine phosphorylated by convergent MAPKs mediates neutrophil NADPH oxidase priming at inflammatory sites. J Clin Invest. 2006;116:2033–2043. doi: 10.1172/JCI27544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Kim H, Jung Y, Shin BS, Song H, Bae SH, Rhee SG, Jeong W. Redox regulation of lipopolysaccharide-mediated sulfiredoxin induction, which depends on both AP-1 and Nrf2. J Biol Chem. 2010;285:34419–34428. doi: 10.1074/jbc.M110.126839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Kim SY, Lee JG, Cho WS, Cho KH, Sakong J, Kim JR, Chin BR, et al. Role of NADPH oxidase-2 in lipopolysaccharide-induced matrix metalloproteinase expression and cell migration. Immunol Cell Biol. 2010;88:197–204. doi: 10.1038/icb.2009.87. [DOI] [PubMed] [Google Scholar]
- 225.Kono H, Rusyn I, Yin M, Gabele E, Yamashina S, Dikalova A, Kadiiska MB, et al. NADPH oxidase-derived free radicals are key oxidants in alcohol-induced liver disease. J Clin Invest. 2000;106:867–872. doi: 10.1172/JCI9020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Thakur V, Pritchard MT, McMullen MR, Wang Q, Nagy LE. Chronic ethanol feeding increases activation of NADPH oxidase by lipopolysaccharide in rat Kupffer cells: role of increased reactive oxygen in LPS-stimulated ERK1/2 activation and TNF-alpha production. J Leukoc Biol. 2006;79:1348–1356. doi: 10.1189/jlb.1005613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Maitra U, Singh N, Gan L, Ringwood L, Li L. IRAK-1 contributes to lipopolysaccharide-induced reactive oxygen species generation in macrophages by inducing NOX-1 transcription and Rac1 activation and suppressing the expression of antioxidative enzymes. J Biol Chem. 2009;284:35403–35411. doi: 10.1074/jbc.M109.059501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, Caviglia JM, et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell. 21:504–516. doi: 10.1016/j.ccr.2012.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Machida K, Cheng KT, Sung VM, Levine AM, Foung S, Lai MM. Hepatitis C virus induces toll-like receptor 4 expression, leading to enhanced production of beta interferon and interleukin-6. J Virol. 2006;80:866–874. doi: 10.1128/JVI.80.2.866-874.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Caradonna L, Mastronardi ML, Magrone T, Cozzolongo R, Cuppone R, Manghisi OG, Caccavo D, et al. Biological and clinical significance of endotoxemia in the course of hepatitis C virus infection. Curr Pharm Des. 2002;8:995–1005. doi: 10.2174/1381612024606983. [DOI] [PubMed] [Google Scholar]
- 231.Vespasiani-Gentilucci U, Carotti S, Onetti-Muda A, Perrone G, Ginanni-Corradini S, Latasa MU, Avila MA, et al. Toll-like receptor-4 expression by hepatic progenitor cells and biliary epithelial cells in HCV-related chronic liver disease. Mod Pathol. 2012;25:576–589. doi: 10.1038/modpathol.2011.197. [DOI] [PubMed] [Google Scholar]
- 232.Sahin H, Borkham-Kamphorst E, do ON, Berres ML, Kaldenbach M, Schmitz P, Weiskirchen R, et al. Proapoptotic effects of the chemokine, CXCL 10 are mediated by the noncognate receptor TLR4 in hepatocytes. Hepatology. 2013;57:797–805. doi: 10.1002/hep.26069. [DOI] [PubMed] [Google Scholar]
- 233.Brownell J, Polyak SJ. Molecular pathways: hepatitis C virus, CXCL10, and the inflammatory road to liver cancer. Clin Cancer Res. 2013;19:1347–1352. doi: 10.1158/1078-0432.CCR-12-0928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Tamura R, Kanda T, Imazeki F, Wu S, Nakamoto S, Tanaka T, Arai M, et al. Hepatitis C Virus nonstructural 5A protein inhibits lipopolysaccharide-mediated apoptosis of hepatocytes by decreasing expression of Toll-like receptor 4. J Infect Dis. 2011;204:793–801. doi: 10.1093/infdis/jir381. [DOI] [PubMed] [Google Scholar]
- 235.Chen CL, Tsukamoto H, Liu JC, Kashiwabara C, Feldman D, Sher L, Dooley S, et al. Reciprocal regulation by TLR4 and TGF-beta in tumor-initiating stem-like cells. J Clin Invest. 2013;123:2832–2849. doi: 10.1172/JCI65859. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 236.Fox JG, Feng Y, Theve EJ, Raczynski AR, Fiala JL, Doernte AL, Williams M, et al. Gut microbes define liver cancer risk in mice exposed to chemical and viral transgenic hepatocarcinogens. Gut. 2010;59:88–97. doi: 10.1136/gut.2009.183749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Agundez JA, Garcia-Martin E, Devesa MJ, Carballo M, Martinez C, Lee-Brunner A, Fernandez C, et al. Polymorphism of the TLR4 gene reduces the risk of hepatitis C virus-induced hepatocellular carcinoma. Oncology. 2012;82:35–40. doi: 10.1159/000335606. [DOI] [PubMed] [Google Scholar]
- 238.Han W, Li H, Cai J, Gleaves LA, Polosukhin VV, Segal BH, Yull FE, et al. NADPH oxidase limits lipopolysaccharide-induced lung inflammation and injury in mice through reduction-oxidation regulation of NF-kappaB activity. J Immunol. 2013;190:4786–4794. doi: 10.4049/jimmunol.1201809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Kim JS, Yeo S, Shin DG, Bae YS, Lee JJ, Chin BR, Lee CH, et al. Glycogen synthase kinase 3beta and beta-catenin pathway is involved in toll-like receptor 4-mediated NADPH oxidase 1 expression in macrophages. FEBS J. 2010;277:2830–2837. doi: 10.1111/j.1742-4658.2010.07700.x. [DOI] [PubMed] [Google Scholar]
- 240.Park HS, Jung HY, Park EY, Kim J, Lee WJ, Bae YS. Cutting edge: direct interaction of TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced production of reactive oxygen species and activation of NF-kappa B. J Immunol. 2004;173:3589–3593. doi: 10.4049/jimmunol.173.6.3589. [DOI] [PubMed] [Google Scholar]
- 241.Chiang E, Dang O, Anderson K, Matsuzawa A, Ichijo H, David M. Cutting edge: apoptosis-regulating signal kinase 1 is required for reactive oxygen species-mediated activation of IFN regulatory factor 3 by lipopolysaccharide. J Immunol. 2006;176:5720–5724. doi: 10.4049/jimmunol.176.10.5720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Park HS, Chun JN, Jung HY, Choi C, Bae YS. Role of NADPH oxidase 4 in lipopolysaccharide-induced proinflammatory responses by human aortic endothelial cells. Cardiovasc Res. 2006;72:447–455. doi: 10.1016/j.cardiores.2006.09.012. [DOI] [PubMed] [Google Scholar]
- 243.Yamada Y, Webber EM, Kirillova I, Peschon JJ, Fausto N. Analysis of liver regeneration in mice lacking type 1 or type 2 tumor necrosis factor receptor: requirement for type 1 but not type 2 receptor. Hepatology. 1998;28:959–970. doi: 10.1002/hep.510280410. [DOI] [PubMed] [Google Scholar]
- 244.Kirillova I, Chaisson M, Fausto N. Tumor necrosis factor induces DNA replication in hepatic cells through nuclear factor kappaB activation. Cell Growth Differ. 1999;10:819–828. [PubMed] [Google Scholar]
- 245.Xia L, Mo P, Huang W, Zhang L, Wang Y, Zhu H, Tian D, et al. The TNF-alpha/ROS/HIF-1-induced upregulation of FoxMI expression promotes HCC proliferation and resistance to apoptosis. Carcinogenesis. 2012;33:2250–2259. doi: 10.1093/carcin/bgs249. [DOI] [PubMed] [Google Scholar]
- 246.Wheelhouse NM, Chan YS, Gillies SE, Caldwell H, Ross JA, Harrison DJ, Prost S. TNF-alpha induced DNA damage in primary murine hepatocytes. Int J Mol Med. 2003;12:889–894. [PubMed] [Google Scholar]
- 247.Perlemuter G, Letteron P, Carnot F, Zavala F, Pessayre D, Nalpas B, Brechot C. Alcohol and hepatitis C virus core protein additively increase lipid peroxidation and synergistically trigger hepatic cytokine expression in a transgenic mouse model. J Hepatol. 2003;39:1020–1027. doi: 10.1016/s0168-8278(03)00414-8. [DOI] [PubMed] [Google Scholar]
- 248.Sun HC, Li M, Lu JL, Yan DW, Zhou CZ, Fan JW, Qin XB, et al. Overexpression of Forkhead box M1 protein associates with aggressive tumor features and poor prognosis of hepatocellular carcinoma. Oncol Rep. 2011;25:1533–1539. doi: 10.3892/or.2011.1230. [DOI] [PubMed] [Google Scholar]
- 249.Kalinichenko VV, Major ML, Wang X, Petrovic V, Kuechle J, Yoder HM, Dennewitz MB, et al. Foxm1b transcription factor is essential for development of hepatocellular carcinomas and is negatively regulated by the p19ARF tumor suppressor. Genes Dev. 2004;18:830–850. doi: 10.1101/gad.1200704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Gusarova GA, Wang IC, Major ML, Kalinichenko VV, Ackerson T, Petrovic V, Costa RH. A cell-penetrating ARF peptide inhibitor of FoxM1 in mouse hepatocellular carcinoma treatment. J Clin Invest. 2007;117:99–111. doi: 10.1172/JCI27527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Kastl L, Sauer SW, Ruppert T, Beissbarth T, Becker MS, Suss D, Krammer PH, et al. TNF-alpha mediates mitochondrial uncoupling and enhances ROS-dependent cell migration via NF-kappaB activation in liver cells. FEBS Lett. 2013;588:175–183. doi: 10.1016/j.febslet.2013.11.033. [DOI] [PubMed] [Google Scholar]
- 252.Moe KT, Aulia S, Jiang F, Chua YL, Koh TH, Wong MC, Dusting GJ. Differential upregulation of Nox homologues of NADPH oxidase by tumor necrosis factor-alpha in human aortic smooth muscle and embryonic kidney cells. J Cell Mol Med. 2006;10:231–239. doi: 10.1111/j.1582-4934.2006.tb00304.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Manea A, Manea SA, Gafencu AV, Raicu M, Simionescu M. AP-1-dependent transcriptional regulation of NADPH oxidase in human aortic smooth muscle cells: role of p22phox subunit. Arterioscler Thromb Vasc Biol. 2008;28:878–885. doi: 10.1161/ATVBAHA.108.163592. [DOI] [PubMed] [Google Scholar]
- 254.Kim YS, Morgan MJ, Choksi S, Liu ZG. TNF-induced activation of the Nox1 NADPH oxidase and its role in the induction of necrotic cell death. Mol Cell. 2007;26:675–687. doi: 10.1016/j.molcel.2007.04.021. [DOI] [PubMed] [Google Scholar]
- 255.Manea A, Tanase LI, Raicu M, Simionescu M. Jak/STAT signaling pathway regulates nox1 and nox4-based NADPH oxidase in human aortic smooth muscle cells. Arterioscler Thromb Vasc Biol. 2010;30:105–112. doi: 10.1161/ATVBAHA.109.193896. [DOI] [PubMed] [Google Scholar]
- 256.Kuwano Y, Kawahara T, Yamamoto H, Teshima-Kondo S, Tominaga K, Masuda K, Kishi K, et al. Interferon-gamma activates transcription of NADPH oxidase 1 gene and upregulates production of superoxide anion by human large intestinal epithelial cells. Am J Physiol Cell Physiol. 2006;290:C433–C443. doi: 10.1152/ajpcell.00135.2005. [DOI] [PubMed] [Google Scholar]
- 257.Harper RW, Xu C, Eiserich JP, Chen Y, Kao CY, Thai P, Setiadi H, et al. Differential regulation of dual NADPH oxidases/peroxidases, Duox1 and Duox2, by Th1 and Th2 cytokines in respiratory tract epithelium. FEBS Lett. 2005;579:4911–4917. doi: 10.1016/j.febslet.2005.08.002. [DOI] [PubMed] [Google Scholar]
- 258.Sanchez-Valle V, Chavez-Tapia NC, Uribe M, Mendez-Sanchez N. Role of oxidative stress and molecular changes in liver fibrosis: a review. Curr Med Chem. 2012;19:4850–4860. doi: 10.2174/092986712803341520. [DOI] [PubMed] [Google Scholar]
- 259.Poli G. Pathogenesis of liver fibrosis: role of oxidative stress. Mol Aspects Med. 2000;21:49–98. doi: 10.1016/s0098-2997(00)00004-2. [DOI] [PubMed] [Google Scholar]
- 260.Paik YH, Kim J, Aoyama T, De Minicis S, Bataller R, Brenner DA. Role of NADPH Oxidases in Liver Fibrosis. Antioxid Redox Signal. 2014 doi: 10.1089/ars.2013.5619. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Nieto N, Greenwel P, Friedman SL, Zhang F, Dannenberg AJ, Cederbaum AI. Ethanol and arachidonic acid increase alpha 2(I) collagen expression in rat hepatic stellate cells overexpressing cytochrome P450 2E1. Role of H2O2 and cyclooxygenase-2. J Biol Chem. 2000;275:20136–20145. doi: 10.1074/jbc.M001422200. [DOI] [PubMed] [Google Scholar]
- 262.Nieto N. Ethanol and fish oil induce NFkappaB transactivation of the collagen alpha2(I) promoter through lipid peroxidation-driven activation of the PKC-PI3K-Akt pathway. Hepatology. 2007;45:1433–1445. doi: 10.1002/hep.21659. [DOI] [PubMed] [Google Scholar]
- 263.Rigamonti C, Andorno S, Maduli E, Morelli S, Pittau S, Nicosia G, Boldorini R, et al. Iron, hepatic stellate cells and fibrosis in chronic hepatitis C. Eur J Clin Invest. 2002;32(Suppl 1):28–35. doi: 10.1046/j.1365-2362.2002.0320s1028.x. [DOI] [PubMed] [Google Scholar]
- 264.Colombo M, de Franchis R, Del Ninno E, Sangiovanni A, De Fazio C, Tommasini M, Donato MF, et al. Hepatocellular carcinoma in Italian patients with cirrhosis. N Engl J Med. 1991;325:675–680. doi: 10.1056/NEJM199109053251002. [DOI] [PubMed] [Google Scholar]
- 265.Paik YH, Iwaisako K, Seki E, Inokuchi S, Schnabl B, Osterreicher CH, Kisseleva T, et al. The nicotinamide adenine dinucleotide phosphate oxidase (NOX) homologues NOX1 and NOX2/gp91(phox) mediate hepatic fibrosis in mice. Hepatology. 2011;53:1730–1741. doi: 10.1002/hep.24281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Bataller R, Schwabe RF, Choi YH, Yang L, Paik YH, Lindquist J, Qian T, et al. NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic fibrosis. J Clin Invest. 2003;112:1383–1394. doi: 10.1172/JCI18212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.De Minicis S, Seki E, Paik YH, Osterreicher CH, Kodama Y, Kluwe J, Torozzi L, et al. Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis. Hepatology. 2010;52:1420–1430. doi: 10.1002/hep.23804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Zhan SS, Jiang JX, Wu J, Halsted C, Friedman SL, Zern MA, Torok NJ. Phagocytosis of apoptotic bodies by hepatic stellate cells induces NADPH oxidase and is associated with liver fibrosis in vivo. Hepatology. 2006;43:435–443. doi: 10.1002/hep.21093. [DOI] [PubMed] [Google Scholar]
- 269.Jiang JX, Venugopal S, Serizawa N, Chen X, Scott F, Li Y, Adamson R, et al. Reduced nicotinamide adenine dinucleotide phosphate oxidase 2 plays a key role in stellate cell activation and liver fibrogenesis in vivo. Gastroenterology. 2010;139:1375–1384. doi: 10.1053/j.gastro.2010.05.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Novitskiy G, Potter JJ, Wang L, Mezey E. Influences of reactive oxygen species and nitric oxide on hepatic fibrogenesis. Liver Int. 2006;26:1248–1257. doi: 10.1111/j.1478-3231.2006.01364.x. [DOI] [PubMed] [Google Scholar]
- 271.Jiang JX, Chen X, Serizawa N, Szyndralewiez C, Page P, Schroder K, Brandes RP, et al. Liver fibrosis and hepatocyte apoptosis are attenuated by GKT137831, a novel NOX4/NOX1 inhibitor in vivo. Free Radic Biol Med. 2012;53:289–296. doi: 10.1016/j.freeradbiomed.2012.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Aoyama T, Paik YH, Watanabe S, Laleu B, Gaggini F, Fioraso-Cartier L, Molango S, et al. Nicotinamide adenine dinucleotide phosphate oxidase in experimental liver fibrosis: GKT137831 as a novel potential therapeutic agent. Hepatology. 2012;56:2316–2327. doi: 10.1002/hep.25938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Rolas L, Makhezer N, Hadjoudj S, El-Benna J, Djerdjouri B, Elkrief L, Moreau R, et al. Inhibition of mammalian target of rapamycin aggravates the respiratory burst defect of neutrophils from decompensated patients with cirrhosis. Hepatology. 2013;57:1163–1171. doi: 10.1002/hep.26109. [DOI] [PubMed] [Google Scholar]
- 274.Inagaki Y, Okazaki I. Emerging insights into Transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut. 2007;56:284–292. doi: 10.1136/gut.2005.088690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Hu T, Ramachandrarao SP, Siva S, Valancius C, Zhu Y, Mahadev K, Toh I, et al. Reactive oxygen species production via NADPH oxidase mediates TGF-beta-induced cytoskeletal alterations in endothelial cells. Am J Physiol Renal Physiol. 2005;289:F816–F825. doi: 10.1152/ajprenal.00024.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Senturk S, Mumcuoglu M, Gursoy-Yuzugullu O, Cingoz B, Akcali KC, Ozturk M. Transforming growth factor-beta induces senescence in hepatocellular carcinoma cells and inhibits tumor growth. Hepatology. 2010;52:966–974. doi: 10.1002/hep.23769. [DOI] [PubMed] [Google Scholar]
- 277.Lu CL, Qiu JL, Huang PZ, Zou RH, Hong J, Li BK, Chen GH, et al. NADPH oxidase DUOX1 and DUOX2 but not NOX4 are independent predictors in hepatocellular carcinoma after hepatectomy. Tumour Biol. 2011;32:1173–1182. doi: 10.1007/s13277-011-0220-3. [DOI] [PubMed] [Google Scholar]
- 278.Lin W, Wu G, Li S, Weinberg EM, Kumthip K, Peng LF, Mendez-Navarro J, et al. HIV and HCV cooperatively promote hepatic fibrogenesis via induction of reactive oxygen species and NFkappaB. J Biol Chem. 2011;286:2665–2674. doi: 10.1074/jbc.M110.168286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Gieseler RK, Marquitan G, Schlattjan M, Sowa JP, Bechmann LP, Timm J, Roggendorf M, et al. Hepatocyte apoptotic bodies encasing nonstructural HCV proteins amplify hepatic stellate cell activation: implications for chronic hepatitis C. J Viral Hepat. 2011;18:760–767. doi: 10.1111/j.1365-2893.2010.01362.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Emerit J, Samuel D, Pavio N. Cu-Zn super oxide dismutase as a potential antifibrotic drug for hepatitis C related fibrosis. Biomed Pharmacother. 2006;60:1–4. doi: 10.1016/j.biopha.2005.09.002. [DOI] [PubMed] [Google Scholar]
- 281.Jiang JX, Chen X, Fukada H, Serizawa N, Devaraj S, Torok NJ. Advanced glycation endproducts induce fibrogenic activity in nonalcoholic steatohepatitis by modulating TNF-alpha-converting enzyme activity in mice. Hepatology. 2013;58:1339–1348. doi: 10.1002/hep.26491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Crosas-Molist E, Bertran E, Sancho P, Lopez-Luque J, Fernando J, Sanchez A, Fernandez M, et al. The NADPH oxidase NOX4 inhibits hepatocyte proliferation and liver cancer progression. Free Radic Biol Med. 2014;69:338–347. doi: 10.1016/j.freeradbiomed.2014.01.040. [DOI] [PubMed] [Google Scholar]
- 283.Wang F, Kaur S, Cavin LG, Arsura M. Nuclear-factor-kappaB (NF-kappaB) and radical oxygen species play contrary roles in transforming growth factor-beta1 (TGF-beta1)-induced apoptosis in hepatocellular carcinoma (HCC) cells. Biochem Biophys Res Commun. 2008;377:1107–1112. doi: 10.1016/j.bbrc.2008.10.130. [DOI] [PubMed] [Google Scholar]
- 284.Caja L, Sancho P, Bertran E, Iglesias-Serret D, Gil J, Fabregat I. Overactivation of the MEK/ERK pathway in liver tumor cells confers resistance to TGF-{beta}-induced cell death through impairing up-regulation of the NADPH oxidase NOX4. Cancer Res. 2009;69:7595–7602. doi: 10.1158/0008-5472.CAN-09-1482. [DOI] [PubMed] [Google Scholar]
- 285.Yu JH, Zhu BM, Riedlinger G, Kang K, Hennighausen L. The liver-specific tumor suppressor STAT5 controls expression of the reactive oxygen species-generating enzyme NOX4 and the proapoptotic proteins PUMA and BIM in mice. Hepatology. 2012;56:2375–2386. doi: 10.1002/hep.25900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Matsuzaki K, Murata M, Yoshida K, Sekimoto G, Uemura Y, Sakaida N, Kaibori M, et al. Chronic inflammation associated with hepatitis C virus infection perturbs hepatic transforming growth factor beta signaling, promoting cirrhosis and hepatocellular carcinoma. Hepatology. 2007;46:48–57. doi: 10.1002/hep.21672. [DOI] [PubMed] [Google Scholar]
- 287.Cheng PL, Chang MH, Chao CH, Lee YH. Hepatitis C viral proteins interact with Smad3 and differentially regulate TGF-beta/Smad3-mediated transcriptional activation. Oncogene. 2004;23:7821–7838. doi: 10.1038/sj.onc.1208066. [DOI] [PubMed] [Google Scholar]
- 288.Choi SH, Hwang SB. Modulation of the transforming growth factor-beta signal transduction pathway by hepatitis C virus nonstructural 5A protein. J Biol Chem. 2006;281:7468–7478. doi: 10.1074/jbc.M512438200. [DOI] [PubMed] [Google Scholar]
- 289.Wu K, Ding J, Chen C, Sun W, Ning BF, Wen W, Huang L, et al. Hepatic transforming growth factor beta gives rise to tumor-initiating cells and promotes liver cancer development. Hepatology. 2012;56:2255–2267. doi: 10.1002/hep.26007. [DOI] [PubMed] [Google Scholar]
- 290.Sancho P, Fabregat I. The NADPH oxidase inhibitor VAS2870 impairs cell growth and enhances TGF-beta-induced apoptosis of liver tumor cells. Biochem Pharmacol. 2011;81:917–924. doi: 10.1016/j.bcp.2011.01.007. [DOI] [PubMed] [Google Scholar]
- 291.Ortiz C, Caja L, Bertran E, Gonzalez-Rodriguez A, Valverde AM, Fabregat I, Sancho P. Protein-tyrosine phosphatase 1B (PTP1B) deficiency confers resistance to transforming growth factor-beta (TGF-beta)-induced suppressor effects in hepatocytes. J Biol Chem. 2012;287:15263–15274. doi: 10.1074/jbc.M111.303958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Amako Y, Igloi Z, Mankouri J, Kazlauskas A, Saksela K, Dallas M, Peers C, et al. Hepatitis C virus NS5A inhibits mixed lineage kinase 3 to block apoptosis. J Biol Chem. 2013;288:24753–24763. doi: 10.1074/jbc.M113.491985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Lim JS, Park SH, Jang KL. Hepatitis C virus Core protein overcomes stress-induced premature senescence by down-regulating p16 expression via DNA methylation. Cancer Lett. 2012;321:154–161. doi: 10.1016/j.canlet.2012.01.044. [DOI] [PubMed] [Google Scholar]
- 294.Soung YH, Lee JW, Kim SY, Sung YJ, Park WS, Nam SW, Kim SH, et al. Caspase-8 gene is frequently inactivated by the frameshift somatic mutation 1225_1226delTG in hepatocellular carcinomas. Oncogene. 2005;24:141–147. doi: 10.1038/sj.onc.1208244. [DOI] [PubMed] [Google Scholar]
- 295.Baddour N, Farrag E, Zeid A, Bedewy E, Taher Y. Decreased apoptosis in advanced-stage/high-grade hepatocellular carcinoma complicating chronic hepatitis C is mediated through the downregulation of p21 ras. Chin J Cancer Res. 2013;25:281–288. doi: 10.3978/j.issn.1000-9604.2013.04.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Mateu G, Donis RO, Wakita T, Bukh J, Grakoui A. Intragenotypic JFH1 based recombinant hepatitis C virus produces high levels of infectious particles but causes increased cell death. Virology. 2008;376:397–407. doi: 10.1016/j.virol.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Walters KA, Syder AJ, Lederer SL, Diamond DL, Paeper B, Rice CM, Katze MG. Genomic analysis reveals a potential role for cell cycle perturbation in HCV-mediated apoptosis of cultured hepatocytes. PLoS Pathog. 2009;5:e1000269. doi: 10.1371/journal.ppat.1000269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Joyce MA, Walters KA, Lamb SE, Yeh MM, Zhu LF, Kneteman N, Doyle JS, et al. HCV induces oxidative and ER stress, and sensitizes infected cells to apoptosis in SCID/Alb-uPA mice. PLoS Pathog. 2009;5:e1000291. doi: 10.1371/journal.ppat.1000291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Lonardo A, Loria P, Adinolfi LE, Carulli N, Ruggiero G. Hepatitis C and steatosis: a reappraisal. J Viral Hepat. 2006;13:73–80. doi: 10.1111/j.1365-2893.2005.00669.x. [DOI] [PubMed] [Google Scholar]
- 300.Pekow JR, Bhan AK, Zheng H, Chung RT. Hepatic steatosis is associated with increased frequency of hepatocellular carcinoma in patients with hepatitis C-related cirrhosis. Cancer. 2007;109:2490–2496. doi: 10.1002/cncr.22701. [DOI] [PubMed] [Google Scholar]
- 301.Leandro G, Mangia A, Hui J, Fabris P, Rubbia-Brandt L, Colloredo G, Adinolfi LE, et al. Relationship between steatosis, inflammation, and fibrosis in chronic hepatitis C: a meta-analysis of individual patient data. Gastroenterology. 2006;130:1636–1642. doi: 10.1053/j.gastro.2006.03.014. [DOI] [PubMed] [Google Scholar]
- 302.Ohata K, Hamasaki K, Toriyama K, Matsumoto K, Saeki A, Yanagi K, Abiru S, et al. Hepatic steatosis is a risk factor for hepatocellular carcinoma in patients with chronic hepatitis C virus infection. Cancer. 2003;97:3036–3043. doi: 10.1002/cncr.11427. [DOI] [PubMed] [Google Scholar]
- 303.Lerat H, Honda M, Beard MR, Loesch K, Sun J, Yang Y, Okuda M, et al. Steatosis and liver cancer in transgenic mice expressing the structural and nonstructural proteins of hepatitis C virus. Gastroenterology. 2002;122:352–365. doi: 10.1053/gast.2002.31001. [DOI] [PubMed] [Google Scholar]
- 304.Moriishi K, Mochizuki R, Moriya K, Miyamoto H, Mori Y, Abe T, Murata S, et al. Critical role of PA28gamma in hepatitis C virus-associated steatogenesis and hepatocarcinogenesis. Proc Natl Acad Sci U S A. 2007;104:1661–1666. doi: 10.1073/pnas.0607312104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Tanaka N, Moriya K, Kiyosawa K, Koike K, Gonzalez FJ, Aoyama T. PPARalpha activation is essential for HCV core protein-induced hepatic steatosis and hepatocellular carcinoma in mice. J Clin Invest. 2008;118:683–694. doi: 10.1172/JCI33594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Naas T, Ghorbani M, Alvarez-Maya I, Lapner M, Kothary R, De Repentigny Y, Gomes S, et al. Characterization of liver histopathology in a transgenic mouse model expressing genotype 1a hepatitis C virus core and envelope proteins 1 and 2. Journal of General Virology. 2005;86:2185–2196. doi: 10.1099/vir.0.80969-0. [DOI] [PubMed] [Google Scholar]
- 307.Waris G, Felmlee DJ, Negro F, Siddiqui A. Hepatitis C virus induces proteolytic cleavage of sterol regulatory element binding proteins and stimulates their phosphorylation via oxidative stress. J Virol. 2007;81:8122–8130. doi: 10.1128/JVI.00125-07. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 308.Oem J-K, Jackel-Cram C, Li Y-P, Zhou Y, Zhong J, Shimano H, Babiuk LA, et al. Activation of sterol regulatory element-binding protein 1c and fatty acid synthase transcription by hepatitis C virus non-structural protein 2. Journal of General Virology. 2008;89:1225–1230. doi: 10.1099/vir.0.83491-0. [DOI] [PubMed] [Google Scholar]
- 309.Harris C, Herker E, Farese RV, Ott M. Hepatitis C Virus Core Protein Decreases Lipid Droplet Turnover: A MECHANISM FOR CORE-INDUCED STEATOSIS. Journal of Biological Chemistry. 2011;286:42615–42625. doi: 10.1074/jbc.M111.285148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Kim KH, Hong SP, Kim K, Park MJ, Kim KJ, Cheong J. HCV core protein induces hepatic lipid accumulation by activating SREBP1 and PPARγ. Biochemical and Biophysical Research Communications. 2007;355:883–888. doi: 10.1016/j.bbrc.2007.02.044. [DOI] [PubMed] [Google Scholar]
- 311.Waris G, Felmlee DJ, Negro F, Siddiqui A. Hepatitis C Virus Induces Proteolytic Cleavage of Sterol Regulatory Element Binding Proteins and Stimulates Their Phosphorylation via Oxidative Stress. Journal of Virology. 2007;81:8122–8130. doi: 10.1128/JVI.00125-07. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 312.Farinati F, Cardin R, De Maria N, Della Libera G, Marafin C, Lecis E, Burra P, et al. Iron storage, lipid peroxidation and glutathione turnover in chronic anti-HCV positive hepatitis. Journal of Hepatology. 1995;22:449–456. doi: 10.1016/0168-8278(95)80108-1. [DOI] [PubMed] [Google Scholar]
- 313.Macdonald GA, Bridle KR, Ward PJ, Walker NI, Houglum K, George DK, Smith JL, et al. Lipid peroxidation in hepatic steatosis in humans is associated with hepatic fibrosis and occurs predominately in acinar zone 3. Journal of Gastroenterology and Hepatology. 2001;16:599–606. doi: 10.1046/j.1440-1746.2001.02445.x. [DOI] [PubMed] [Google Scholar]
- 314.Bartsch H, Nair J. Chronic inflammation and oxidative stress in the genesis and perpetuation of cancer: role of lipid peroxidation, DNA damage, and repair. Langenbecks Arch Surg. 2006;391:499–510. doi: 10.1007/s00423-006-0073-1. [DOI] [PubMed] [Google Scholar]
- 315.Jeong SW, Jang JY, Chung RT. Hepatitis C virus and hepatocarcinogenesis. Clin Mol Hepatol. 2012;18:347–356. doi: 10.3350/cmh.2012.18.4.347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Banerjee A, Ray RB, Ray R. Oncogenic potential of hepatitis C virus proteins. Viruses. 2010;2:2108–2133. doi: 10.3390/v2092108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.McGivern DR, Lemon SM. Virus-specific mechanisms of carcinogenesis in hepatitis C virus associated liver cancer. Oncogene. 2011;30:1969–1983. doi: 10.1038/onc.2010.594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.de La Coste A, Romagnolo B, Billuart P, Renard CA, Buendia MA, Soubrane O, Fabre M, et al. Somatic mutations of the beta-catenin gene are frequent in mouse and human hepatocellular carcinomas. Proc Natl Acad Sci U S A. 1998;95:8847–8851. doi: 10.1073/pnas.95.15.8847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.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. doi: 10.1128/JVI.79.8.5006-5016.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Zhang Y, Wei W, Cheng N, Wang K, Li B, Jiang X, Sun S. Hepatitis C virus-induced up-regulation of microRNA-155 promotes hepatocarcinogenesis by activating Wnt signaling. Hepatology. 2012;56:1631–1640. doi: 10.1002/hep.25849. [DOI] [PubMed] [Google Scholar]
- 321.Iqbal J, McRae S, Banaudha K, Mai T, Waris G. Mechanism of HCV-induced Osteopontin and its role in epithelial to mesenchymal transition of hepatocytes. J Biol Chem. 2013;288:36994–37009. doi: 10.1074/jbc.M113.492314. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 322.Saito M, Kohara M, Tsukiyama-Kohara K. Hepatitis C virus promotes expression of the 3beta-hydroxysterol delta24-reductase through Sp1. J Med Virol. 2012;84:733–746. doi: 10.1002/jmv.23250. [DOI] [PubMed] [Google Scholar]
- 323.Arbiser JL, Petros J, Klafter R, Govindajaran B, McLaughlin ER, Brown LF, Cohen C, et al. Reactive oxygen generated by Nox1 triggers the angiogenic switch. Proc Natl Acad Sci U S A. 2002;99:715–720. doi: 10.1073/pnas.022630199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Zhang M, Brewer AC, Schroder K, Santos CX, Grieve DJ, Wang M, Anilkumar N, et al. NADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesis. Proc Natl Acad Sci U S A. 2010;107:18121–18126. doi: 10.1073/pnas.1009700107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Garrido-Urbani S, Jemelin S, Deffert C, Carnesecchi S, Basset O, Szyndralewiez C, Heitz F, et al. Targeting vascular NADPH oxidase 1 blocks tumor angiogenesis through a PPARalpha mediated mechanism. PLoS One. 2011;6:e14665. doi: 10.1371/journal.pone.0014665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Caltagirone A, Weiss G, Pantopoulos K. Modulation of cellular iron metabolism by hydrogen peroxide. Effects of H2O2 on the expression and function of iron-responsive element-containing mRNAs in B6 fibroblasts. J Biol Chem. 2001;276:19738–19745. doi: 10.1074/jbc.M100245200. [DOI] [PubMed] [Google Scholar]
- 327.Pantopoulos K, Hentze MW. Activation of iron regulatory protein-1 by oxidative stress in vitro. Proc Natl Acad Sci U S A. 1998;95:10559–10563. doi: 10.1073/pnas.95.18.10559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.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. doi: 10.1099/vir.0.014340-0. [DOI] [PubMed] [Google Scholar]
- 329.Ivanov AV, Smirnova OA, Ivanova ON, Masalova OV, Kochetkov SN, Isaguliants MG. Hepatitis C virus proteins activate NRF2/ARE pathway by distinct ROS-dependent and independent mechanisms in HUH7 cells. PLoS One. 2011;6:e24957. doi: 10.1371/journal.pone.0024957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
- 331.Kelley RK, Nimeiri HS, Munster PN, Vergo MT, Huang Y, Li CM, Hwang J, et al. Temsirolimus combined with sorafenib in hepatocellular carcinoma: a phase I dose-finding trial with pharmacokinetic and biomarker correlates. Ann Oncol. 2013;24:1900–1907. doi: 10.1093/annonc/mdt109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332.Seronello S, Montanez J, Presleigh K, Barlow M, Park SB, Choi J. Ethanol and reactive species increase basal sequence heterogeneity of hepatitis C virus and produce variants with reduced susceptibility to antivirals. PLoS ONE. 2011;6:e27436. doi: 10.1371/journal.pone.0027436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333.Halfon P, Locarnini S. Hepatitis C virus resistance to protease inhibitors. J Hepatol. 2007;55:192–206. doi: 10.1016/j.jhep.2011.01.011. [DOI] [PubMed] [Google Scholar]
- 334.Fridell RA, Qiu D, Wang C, Valera L, Gao M. Resistance analysis of the hepatitis C virus NS5A inhibitor BMS-790052 in an in vitro replicon system. Antimicrob Agents Chemother. 2010;54:3641–3650. doi: 10.1128/AAC.00556-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335.Scheel TK, Gottwein JM, Mikkelsen LS, Jensen TB, Bukh J. Recombinant HCV variants with NS5A from genotypes 1–7 have different sensitivities to an NS5A inhibitor but not interferon-alpha. Gastroenterology. 2011;140:1032–1042. doi: 10.1053/j.gastro.2010.11.036. [DOI] [PubMed] [Google Scholar]
- 336.Sun JH, O'Boyle Ii DR, Zhang Y, Wang C, Nower P, Valera L, Roberts S, et al. Impact of a baseline polymorphism on the emergence of resistance to the hepatitis C virus nonstructural protein 5A replication complex inhibitor, BMS-790052. Hepatology. 2012;55:1692–1699. doi: 10.1002/hep.25581. [DOI] [PubMed] [Google Scholar]
- 337.Gong K, Li W. Shikonin, a Chinese plant-derived naphthoquinone, induces apoptosis in hepatocellular carcinoma cells through reactive oxygen species: A potential new treatment for hepatocellular carcinoma. Free Radic Biol Med. 2011;51:2259–2271. doi: 10.1016/j.freeradbiomed.2011.09.018. [DOI] [PubMed] [Google Scholar]