Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Endoplasmic Reticulum Stress Activates the Hepatic Activator Protein 1 Complex via Mitogen Activated Protein Kinase-Dependent Signaling Pathways

  • Shantel Olivares,

    Affiliation Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Richard M. Green,

    Affiliation Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Anne S. Henkel

    a-henkel2@northwestern.edu

    Affiliation Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

Abstract

Background and Aims

Endoplasmic reticulum (ER) stress is induced in many forms of chronic liver disease and may promote the development of hepatocellular carcinoma. The activator protein 1 (AP-1) complex is a transcription factor that promotes hepatic carcinogenesis in response to cellular stress. The aim of this study was to determine the role of ER stress in the regulation of the hepatic AP-1 complex.

Methods

Human hepatocellular carcinoma (HepG2) cells and C57BL/6J mice were subjected to pharmacologic ER stress and the expression of AP-1-associated genes and proteins was assessed. To determine the role of MAPK signaling in ER stress-induced AP-1 activation, ER stress was induced in JNK- and ERK-inhibited HepG2 cells.

Results

Induction of ER stress promoted the activation of both Jun- and Fos-related genes and proteins of the AP-1 complex in HepG2 cells and murine liver. Inhibition of ERK phosphorylation in HepG2 cells completely prevented ER stress-induced activation of the fos-related components of AP-1 whereas activation of Jun-related components was only partially attenuated. Conversely, inhibition of JNK phosphorylation in HepG2 cells reduced ER stress-induced activation of Jun-related components but did not prevent activation of fos-related components.

Conclusions

ER stress activates the hepatic AP-1 complex via MAPK-dependent signaling pathways. ER stress-induced activation of Fos-related components is dependent primarily on ERK activation whereas ER stress-induced activation of Jun-related components is dependent primarily on JNK activation, although there is interplay between these regulatory pathways. These data implicate a novel signaling pathway by which sustained ER stress, as observed in many chronic liver diseases, may promote hepatic carcinogenesis.

Introduction

The incidence of hepatocellular carcinoma (HCC) has more than doubled over the past 20 years attributable in large part to the high rate of new hepatitis C virus infection 30 to 40 years ago [1], [2]. Cirrhosis is the primary risk factor for HCC, however, the underlying cause of cirrhosis impacts the risk of progressing to HCC. It is becoming increasingly evident that patients with cirrhosis secondary to nonalcoholic fatty liver disease (NAFLD) are at particularly high risk for developing HCC. In fact, recent observations suggest that NAFLD is a risk factor for HCC even in the absence of cirrhosis [3], [4]. Although significant progress is being made toward curing chronic hepatitis C, the prevalence of NAFLD is rapidly escalating in the United States. As such, the burden of HCC is unlikely to decline in the foreseeable future [5], [6].

Endoplasmic reticulum (ER) stress and the ensuing unfolded protein response are strongly implicated in the pathogenesis of many forms of chronic liver disease, including hepatitis C infection, alcoholic liver disease, and NAFLD [7][14]. The ER functions to maintain protein homeostasis by regulating protein synthesis, folding and processing. Under conditions of ER stress, such as glucose deprivation, aberrant calcium signaling, viral infection, lipotoxicity, and disruption of redox regulation, normal ER function becomes compromised leading to the accumulation of unfolded or misfolded proteins [15]. The accumulation of unfolded proteins triggers an evolutionarily conserved intracellular signal transduction pathway known as the unfolded protein response (UPR) [16]. The UPR initially aims to restore homeostasis and allow the cell to adapt to the stressor [17]. If homeostasis is not adequately restored, however, pathways leading to apoptosis are initiated [18], [19]. Mitogen activated protein kinases (MAPKs) are activated in response to ER stress and may mediate, in part, the critical switch from restoration of homeostasis to initiation of apoptosis. In particular, cJun N-terminal kinase (JNK), a well-established downstream target of the IRE1α branch of the UPR, is thought to promote ER stress-induced apoptosis [20], [21]. The role of extracellular signal-regulated protein kinase (ERK) activation in mediating the ER stress response is less well-characterized but may function to enhance cell survival [22], [23]. The mechanism by which ER stress induces ERK is incompletely understood but is thought to be at least partially mediated by PI3K and adaptor protein Nck [22], [23].

Recent data suggest that ER stress and the UPR may be important in the development of hepatocellular carcinoma [24][31]. Grp78/BiP, a molecular chaperone and master regulator of the UPR, has been posited to promote malignant transformation in numerous tissues including the liver [32][34]. ER stress has also been implicated in the pathogenesis of liver cancer resulting from chronic alcohol use [35]. Sorafenib, the only approved chemotherapeutic agent to treat HCC, has been shown to modulate the UPR [26], [27]. Yet, it remains unknown whether induction of ER stress contributes to the development of HCC in chronic liver diseases.

Activation of the activator protein 1 (AP-1) complex has been shown to be a critical event in the development of HCC [36]. The AP-1 complex is a dimer composed of protooncogenes from the Jun family (cJun, JunD, and JunB) and the Fos family (cFos, FosB, Fra-1, and Fra-2) [37], [38]. In response to various types of cellular stress, the AP-1 complex is activated to function as a transcription factor targeting genes involved in cellular differentiation, proliferation, and apoptosis [37], [38]. MAPKs are key regulators of numerous protooncogenes, including elements of the AP-1 complex [39], [40].

In the present study we aim to determine the impact of ER stress on the regulation of the hepatic AP-1 complex. Specifically we will test our hypothesis that ER stress activates the hepatic AP-1 complex via activation of MAPK-dependent signaling pathways.

Materials and Methods

Cell culture

Human hepatocellular carcinoma (HepG2) cells (ATCC, Mannasas, VA) were cultured in DMEM with 10% fetal bovine serum and maintained at 37°C in 5% CO2. Cells were grown to 80% confluence in 6-well plates and treated with 12 µM tunicamycin, 100 nM thapsigargin (Sigma-Aldrich, St. Louis, MO), 5 mM DL-homocysteine (Sigma-Aldrich) or vehicle (DMSO/saline) in serum-free DMEM for 6 hours. RNA isolation was performed using TRIzol Reagent (Invitrogen, Carlsbad, CA) per protocol. To determine whether the effects of tunicamycin are dependent on ERK1/2 signaling, HepG2 cells were treated with the MEK inhibitor, PD184352 (Santa Cruz Biotechnology) at a concentration of 1 µM or vehicle (DMSO/saline). To determine whether the effects of tunicamycin are dependent on cJun-N-terminal kinase (JNK) signaling, HepG2 cells were treated with the JNK inhibitor, SP600125 (Sigma-Aldrich) at a concentration of 25 µM. After one hour of exposure to PD184352 or SP600125, cells were treated with tunicamycin (10 µg/mL) (Sigma-Aldrich) in serum-free DMEM and incubated for an additional 6 hours. Cytotoxicity was assessed by measuring LDH release in cell culture media using a Cytotox 96 Nonradioactive Cytotoxicity Assay (Promega, Madison, WI). Cellular proliferation was assessed using a BrdU Cell Proliferation ELISA kit (Abcam, Cambridge, MA). In a separate experiment, cells were treated with siRNA targeting JNK1 or control siRNA (Santa Cruz Biotechnology). HepG2 cells grown to 80% confluence were treated with JNK1 siRNA or control siRNA per protocol. Briefly, for each transfection, 6 µL of control or JNK1 siRNA were diluted in 100 µL of siRNA Transfection Medium (Solution A) and 6 µL of siRNA Transfection Reagent was diluted in 100 µL of siRNA Transfection Medium (Solution B). Fifty microliters of Solutions A was added to 100 µL of Solution B and allowed to incubate at room temperature for 30 minutes. After washing the cells once with Transfection Medium, 0.8 mL of the siRNA in Transfection Reagent was added to each well. Cells were incubated for 6 hours at 37°C. The transfection medium was then removed, the cells were washed and incubated in normal growth medium (DMEM with 10% FBS) for 18 hours. The cells were then treated with tunicamycin (10 µg/mL) (Sigma-Aldrich) in serum-free DMEM for 6 hours. The doses of the inhibitors/siRNA were experimentally determined to achieve near complete inhibition of ERK- and JNK-phosphorylation with PD184352 and SP600125, respectively, and an approximately 50% inhibition of JNK1 using JNK1 siRNA as confirmed by western blot (Figure S1).

Animals and Treatments

Male C57BL/6J mice (8 weeks of age) were purchased from Jackson Laboratories (Bar Harbor, ME). Mice underwent 14/10-hour light/dark cycling and were given free access to standard laboratory chow and water. Mice were treated with a single intraperitoneal injection of tunicamycin (2 mg/kg) or vehicle (20% DMSO/PBS) and sacrificed 6 or 72 hours post-injection. To examine the effects of prolonged, sublethal exposure to ER stress, an additional cohort of mice was treated with a daily injection of low-dose tunicamycin (0.1 mg/kg I.P.) for 5 days to achieve a cumulative dose of 0.5 mg/kg. At the end of the treatment protocols, mice were sacrificed by CO2 inhalation followed by cardiac puncture. The collected blood was immediately centrifuged to collect the plasma. The livers were rapidly excised, flushed with ice-cold saline and sectioned. An aliquot of liver was fixed in formalin and the remaining liver was snap-frozen in liquid nitrogen. H&E staining, TUNEL staining, and immunohistochemical staining for Ki67 were performed on liver tissue by the Northwestern University Pathology Core. Plasma ALT was measured using a colorimetric assay per manufacturer's protocol (Teco Diagnostics, Anaheim, CA). All animal protocols were approved by the Northwestern University Animal Care and Use Committee (ACUC).

Analysis of Gene Expression and Protein expression

Total RNA from frozen liver samples or HepG2 cells was isolated using TRIzol reagent and real-time quantitative PCR was performed as previously described [11], [41]. Primer sequences for qPCR are shown in Table S1. Total protein was isolated from frozen liver samples and HepG2 cells and Western blotting was performed as previously described [11], [41]. Protein detection was performed using polyclonal rabbit antibodies to total and phosphorylated cFos, cJun, ERK, and JNK (Cell Signaling Technology, Danvers, MA). Bound antibody was detected using goat anti-rabbit polyclonal HRP antibody (Cell Signaling Technology) and developed using ECL Western Blotting Substrate (Cell Signaling Technology). Representative Western blots of pooled samples are shown.

Statistical Analysis

Data are presented as mean ± standard deviation (SD). Comparisons between groups were performed using Student's t-test analysis.

Results

Endoplasmic reticulum stress activates AP-1-associated genes in human hepatocellular carcinoma cells

To determine the effect of ER stress on activation of the AP-1 complex, human hepatocellular carcinoma cells (HepG2) were treated with three well-established ER stress inducing agents, tunicamycin, thapsigargin, and homocysteine. As expected, all treatments resulted in robust activation of the unfolded protein response as evidenced by elevations in GRP78/BIP and CHOP (Figure 1 A,B). We then examined the effect of ER stress on the expression of the fos-related genes (CFOS and fos-related antigen 1 (FRA1)), and jun-related genes (CJUN and JUND) of the AP-1 complex. As shown in Figure 1, induction of ER stress by any of the three pharmacologic methods stimulated transcription of both Jun- and Fos-related genes of the AP-1 complex.

thumbnail
Figure 1. Induction of ER stress transcriptionally activates genes of the AP-1 complex in HepG2 cells.

Relative mRNA expression of A) GRP78/BIP, B) CHOP, C) CFOS, D) FRA-1, E) CJUN, and F) JUND in HepG2 cells treated with tunicamycin (Tm), homocysteine (Hcy) or thapsigargin (Tg) for 6 hours. Mean (n = 6) ± SD. * p<0.05 vs vehicle-treated control cells.

https://doi.org/10.1371/journal.pone.0103828.g001

Endoplasmic reticulum stress activates AP-1 in murine liver

We next sought to determine whether ER stress activates the AP-1 complex in vivo. Mice were treated with a standard dose of tunicamycin (2 mg/kg I.P.) and sacrificed 6 or 72 hours later. Tunicamycin treatment resulted in significant induction of the unfolded protein response in murine liver at both time points (Figure 2 A,B). There was significant induction of hepatic cFos, Fra-1, cJun, and JunD mRNA 6 hours after exposure to tunicamycin (Figure 2A). The AP-1-associated genes were induced to an even greater degree 72 hours after tunicamycin administration (Figure 2B).

thumbnail
Figure 2. Induction of ER stress in mice activates the hepatic AP-1 complex.

Relative hepatic mRNA expression of Grp78/Bip, Chop, cFos, Fra-1, cJun, and JunD in mice treated with tunicamycin (Tm) at doses of A) 2 mg/kg for 6 hours, B) 2 mg/kg for 72 hours, or C) 0.5 mg/kg over 5 days. Hepatic protein expression of total and phosphorylated CFOS and CJUN in mice treated with Tm at doses of D) 2 mg/kg for 6 hours, E) 2 mg/kg for 72 hours, or F) 0.5 mg/kg over 5 days. Quantitative PCR shown as mean (n = 6) ± SD. * p<0.05 vs vehicle-treated. Representative Western blot of pooled samples, n = 2–3.

https://doi.org/10.1371/journal.pone.0103828.g002

A standard experimental dose of tunicamycin (2 mg/kg) induces overwhelming ER stress from which mice are unable to restore homeostasis and do not typically survive beyond 4 days. Chronic liver diseases, however, induce low-grade ER stress over prolonged periods. Therefore, to examine the effects of prolonged, sublethal exposure to ER stress, mice were treated with daily low-dose injections of tunicamycin over 5 days to achieve a cumulative dose of 0.5 mg/kg. Robust UPR activation was achieved with this dosing strategy (Figure 2C). Mice subjected to prolonged, low-dose tunicamycin showed induction of both fos- and jun-related genes of the AP-1 complex (Figure 2C).

The AP-1 complex is also subject to regulation at the translational and post-translational levels. As shown in Figure 2, hepatic CJUN was consistently increased at the translational and post-translational level in response to 6 hours, 72 hours, or 5 days of ER stress. Total and phosphorylated CFOS were only faintly detectable at 6 hours but did demonstrate modestly enhanced expression with exposure to tunicamycin (Figure 2D). At the 72-hour and 5-day time points we observed greatly increased levels of both total and phosphorylated CFOS in response to ER stress (Figure 2 E,F).

ER stress and AP-1 activation have been shown to promote both apoptosis and cellular proliferation. We found that mice exposed to low-dose tunicamycin for 5 days developed hepatocyte ballooning and increased apoptosis in the liver but no significant increase in cellular proliferation (Figure 3A). Plasma ALT, a common marker of hepatic injury, was modestly increased in mice treated with tunicamycin (Figure 3B).

thumbnail
Figure 3. Prolonged low-grade ER stress promotes hepatocyte injury and apoptosis.

Markers of liver injury, apoptosis, and cellular proliferation in mice treated with a cumulative dose of 0.5/kg tunicamycin over 5 days. A) Representative H&E, TUNEL, and Ki67 staining of liver sections. B) Plasma ALT level (IU/L). Mean (n = 6) ± SD, *p<0.05.

https://doi.org/10.1371/journal.pone.0103828.g003

Induction of Fos-related components of AP-1 by ER stress is dependent on ERK activation

We next sought to determine the mechanism by which ER stress activates the AP-1 complex in human hepatocellular carcinoma cells. ER stress has been shown to activate MAPK signaling pathways [20], [21], [23]. Furthermore, ERK activation is known to promote the transcription of fos-related genes [42], [43]. We therefore explored whether ER stress activates AP-1-associated genes via an ERK-dependent signaling pathway. HepG2 cells were pretreated with the potent MEK inhibitor, PD184352, which prevents ERK1/2 phosphorylation (activation), followed by treatment with tunicamycin to induce ER stress. As shown in Figure 4, treatment with tunicamycin induced ERK phosphorylation, which was completely abolished by pre-treatment with PD184352. Inhibition of ERK phosphorylation did not prevent tunicamycin-induced upregulation of ER stress markers, GRP78/BIP and CHOP, however, there was a trend toward a reduction in the degree of upregulation of these markers (Figure 4 B,C). PD184352 completely abolished the activation of CFOS and FRA-1 by tunicamycin at the transcriptional level (Figure 4 D,E). Tunicamycin-induced transcription of CJUN and JUND was attenuated by ERK inhibition (Figure 4 F,G).

thumbnail
Figure 4. ER stress-induced activation of CFOS and FRA-1 is mediated by ERK1/2 signaling.

HepG2 cells pretreated with MEK1 inhibitor (PD) or vehicle followed by treatment with tunicamycin (Tm) or vehicle (control) for 6 hours. A) Representative Western blot of total and phosphorylated ERK, JNK, CFOS, and CJUN. Relative mRNA expression of B) GRP78/BIP, C) CHOP, D) CFOS, E) FRA-1, F) CJUN, and G) JUND. Mean (n = 6) ± SD. * p<0.05.

https://doi.org/10.1371/journal.pone.0103828.g004

We found that ERK signaling also regulates AP-1 at the translational and post-translational levels (Figure 4A). Tunicamycin increased total and phosphorylated CFOS protein levels, which was completely abolished by ERK-inhibition, thus correlating with CFOS gene expression. Conversely, inhibition of ERK phosphorylation accentuated tunicamycin-induced CJUN phosphorylation. We also found that inhibition of ERK-phosphorylation increased levels of phosphorylated JNK, a key activator of CJUN.

As expected, treatment of HepG2 cells with tunicamycin increased cytotoxicity as measured by LDH release (Figure 5A). We found that ERK-inhibition did not attenuate tunicamycin-induced cell death. Induction of ER stress inhibited cellular proliferation which was also unaffected by ERK-inhibition (Figure 5B).

thumbnail
Figure 5. Effects of ERK- and JNK-inhibition on tunicamycin-induced cytotoxicity and cellular proliferation.

Cytoxicity as measured by LDH release (A,C) and cellular proliferation as measured by BrdU incorporation (B,D) in HepG2 cells pretreated with MEK1 inhibitor (PD184352) or JNK inhibitor (SP600125) followed by treatment with tunicamycin or vehicle (control) for 6 hours. Mean (n = 6) ± SD. *p<0.05.

https://doi.org/10.1371/journal.pone.0103828.g005

Induction of Jun-related components of AP-1 by ER stress is dependent on JNK activation

JNK, a MAP kinase that mediates multiple stress signaling pathways in the liver, is a well-established downstream target of the ER stress response [20], [21]. Furthermore, JNK is a critical mediator of hepatic injury in fatty liver diseases and has been implicated in the development of HCC [44][46]. Given our observation that ER stress-induced activation of Jun-related genes is not completely abolished by inhibition of ERK phosphorylation, we next explored whether ER stress activates Jun-related genes via JNK activation. HepG2 cells were treated with the JNK inhibitor, SP600125, followed by treatment with tunicamycin. As expected, treatment of HepG2 cells with tunicamycin markedly induced JNK phosphorylation, which was attenuated by pretreatment with SP600125 (Figure 6A). Inhibition of JNK phosphorylation did not significantly impact tunicamycin-induced GRP78/BIP or CHOP activation however there was a trend toward reduced GRP78/BIP transcription in JNK-inhibited cells (Figure 6 B,C). Inhibition of JNK phosphorylation reduced tunicamycin-induced CJUN transcription and completely abolished upregulation of JUND transcription (Figure 6 D,E). CFOS and FRA-1 expression was induced by tunicamycin in JNK–inhibited cells (Figure 6 C,D); however, the degree of CFOS induction was attenuated by JNK-inhibition, indicating that ER stress-induced CFOS elevation is at least partially mediated by JNK signaling.

thumbnail
Figure 6. ER stress-induced activation of CJUN and JUND is mediated by JNK signaling.

HepG2 cells pretreated with JNK inhibitor (SP600125) or vehicle followed by treatment with tunicamycin or vehicle (control) for 6 hours. A) Representative Western blot of total and phosphorylated JNK, ERK, CJUN, and CFOS. Relative mRNA expression of B) GRP78/BIP, C) CHOP, D) CJUN, E) JUND, F) CFOS, and G) FRA-1. Mean (n = 6) ± SD. * p<0.05.

https://doi.org/10.1371/journal.pone.0103828.g006

As shown in Figure 6A, inhibition of JNK phosphorylation attenuated the induction of total and phosphorylated CJUN protein by tunicamycin. Conversely, ER stress-induced CFOS activation was not attenuated by JNK-inhibition. ER stress-induced ERK phosphorylation was enhanced in JNK-inhibited cells.

Similar to inhibition of ERK phosphorylation, inhibition of JNK phosphorylation did not attenuate tunicamycin-induced cytotoxicity (Figure 5C). Inhibition of JNK activation did, however, enhance the effects of tunicamycin on inhibition of cellular proliferation (Figure 5D).

Given that sustained activation of the JNK1 isoform, in particular, has been associated with steatohepatitis and the development of HCC [44][46] we confirmed the findings observed in JNK-inhibited cells using JNK1-silenced cells. Silencing of JNK1 did not effect tunicamycin-induced GRP78/BiP transcription but accentuated CHOP expression (Figure 7 A,B). Silencing JNK1 prevented tunicamycin-induced transcription of CJUN and JUND but not CFOS or FRA-1 (Figure 7 C–F).

thumbnail
Figure 7. ER stress-induced transcriptional activation of CJUN and JUND is mediated by JNK1 signaling.

Relative mRNA expression of A) GRP78/BIP, B) CHOP, C) CJUN, D) JUND, E) CFOS, and F) FRA-1 in HepG2 cells pretreated with JNK1 siRNA or control siRNA followed by treatment with tunicamycin or vehicle (control) for 6 hours. Mean (n = 6) ± SD. * p<0.05.

https://doi.org/10.1371/journal.pone.0103828.g007

Discussion

Endoplasmic reticulum stress and the ensuing UPR are now recognized as mediators of numerous pathologic processes in the liver. The UPR is activated in several chronic liver diseases that ultimately promote the development of HCC including NAFLD, alcohol-induced liver disease and hepatitis C [7][13]. However, it is unknown whether activation of the UPR in these chronic liver diseases contributes to the increased risk of HCC.

The AP-1 complex is a critical regulator of hepatic carcinogenesis [36]. Here we demonstrate that induction of ER stress in human hepatocellular carcinoma cells and murine liver activates the AP-1 complex at the transcriptional, translational, and post-translational levels. We show that ER stress regulates fos-related components of the AP-1 complex primarily via activation of ERK signaling whereas the activation of jun-related components is primarily mediated by JNK. There is, however, an element of cross-talk between these pathways as evidenced by attenuated induction of fos-related genes in JNK-inhibited cells and attenuated induction of jun-related genes in ERK-inhibited cells in response to ER stress. Given the established role of AP-1 in promoting hepatic carcinogenesis, we speculate that ER stress induced AP-1 activation may be a mechanism by which chronic liver diseases promote malignant transformation.

The effect of JNK1-silencing on the expression of AP-1-associated genes paralleled that of HepG2 cells treated with SP600125 indicating that specifically the JNK1 isoform is critical for AP-1 activation. Of note, the degree of tunicamycin-induced UPR activation and resultant AP-1 gene transcription was modestly attenuated in HepG2 cells pre-treated with control (scrambled) siRNA relative to non-transfected control cells (e.g. Figures 1, 4, and 6). Although we cannot exclude the possibility that the blunted UPR was an effect of scrambled siRNA, these differences could also be due to differing cell culture conditions required for transfection (e.g. duration of serum starvation prior to tunicamycin exposure).

Mice exhibit robust activation of the unfolded protein response within 6 hours of exposure to a standard experimental dose of tunicamycin (2 mg/kg). However, beyond 72 hours this dose of tunicamycin is typically fatal. This raised the possibility that ER stress-induced AP-1 activation may occur only in the setting of overwhelming cellular stress. As such, the physiologic relevance of our observations with respect to chronic liver disease was initially uncertain. Therefore, we also subjected mice to sustained, sublethal ER stress and demonstrate robust activation of hepatic AP-1-associated genes and proteins.

AP-1 has numerous critical functions in the liver with respect to cellular differentiation, proliferation, and apoptosis. The function of AP-1 is determined in part by the relative activation of the various components of this heterodimeric complex. The UPR is a highly dynamic signal transduction pathway in which the various branches are preferentially activated depending on the degree and duration of ER stress. We show that the degree of induction of AP-1-associated genes in murine liver varies depending on the duration and severity of ER stress. As such, the degree and duration of ER stress may impact the transcriptional activity of the AP-1 complex by shifting the relative induction of ERK and JNK and the resultant induction of jun- and fos-related genes. Although we hypothesize that ER stress and the resultant AP-1 activation may promote hepatic carcinogenesis, it is also conceivable that under conditions of chronic low-grade ER stress, activation of the UPR may confer protection against carcinogenesis (e.g. via promoting apoptosis.) The duration of this experiment was insufficient to observe tumor formation in mice, however, future studies are warranted to determine whether inhibition of ER stress may protect against the development of HCC in mice.

At the chosen dose and time points we demonstrate that tunicamycin promotes hepatocyte injury, apoptosis, and cell death and inhibits cellular proliferation. Importantly we found that inhibition of MAPK signaling pathways did not prevent tunicamycin-induced activation of ER stress markers and did not attenuate ER stress-induced cytotoxicity. These findings indicate that the observed reduction in AP-1 activation in ERK- and JNK-inhibited cells is not a consequence of reduced cellular injury.

We propose that coordinated activation of ERK1/2 and JNK1 signaling pathways by ER stress promotes activation of the hepatic AP-1 complex. These data identify a novel pathway by which ER stress, a common feature of many chronic liver diseases, may promote hepatic carcinogenesis. Currently there are limited pharmacotherapeutic options to treat HCC. This work raises the possibility that targeting the unfolded protein response may have therapeutic benefit in the treatment or prevention of HCC in patients with chronic liver disease.

Supporting Information

Figure S1.

Successful silencing of JNK1 in HepG2 cells treated with siRNA targeting JNK1. Representative Western blot of pooled samples (n = 6).

https://doi.org/10.1371/journal.pone.0103828.s001

(TIF)

Table S1.

Primer sequences for quantitative PCR.

https://doi.org/10.1371/journal.pone.0103828.s002

(DOC)

Author Contributions

Conceived and designed the experiments: AH RG SO. Performed the experiments: AH SO. Analyzed the data: AH RG SO. Contributed reagents/materials/analysis tools: AH RG. Wrote the paper: AH.

References

  1. 1. El-Serag HB, Mason AC (1999) Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med 340: 745–750.
  2. 2. Gomaa AI, Khan SA, Toledano MB, Waked I, Taylor-Robinson SD (2008) Hepatocellular carcinoma: epidemiology, risk factors and pathogenesis. World J Gastroenterol 14: 4300–4308.
  3. 3. Kawada N, Imanaka K, Kawaguchi T, Tamai C, Ishihara R, et al. (2009) Hepatocellular carcinoma arising from non-cirrhotic nonalcoholic steatohepatitis. J Gastroenterol 44: 1190–1194.
  4. 4. Ertle J, Dechene A, Sowa JP, Penndorf V, Herzer K, et al. (2011) Non-alcoholic fatty liver disease progresses to hepatocellular carcinoma in the absence of apparent cirrhosis. Int J Cancer 128: 2436–2443.
  5. 5. El-Serag HB, Rudolph KL (2007) Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132: 2557–2576.
  6. 6. Baffy G, Brunt EM, Caldwell SH (2012) Hepatocellular carcinoma in non-alcoholic fatty liver disease: an emerging menace. J Hepatol 56: 1384–1391.
  7. 7. Tardif KD, Mori K, Siddiqui A (2002) Hepatitis C virus subgenomic replicons induce endoplasmic reticulum stress activating an intracellular signaling pathway. J Virol 76: 7453–7459.
  8. 8. Li S, Ye L, Yu X, Xu B, Li K, et al. (2009) Hepatitis C virus NS4B induces unfolded protein response and endoplasmic reticulum overload response-dependent NF-kappaB activation. Virology 391: 257–264.
  9. 9. Puri P, Mirshahi F, Cheung O, Natarajan R, Maher JW, et al. (2008) Activation and dysregulation of the unfolded protein response in nonalcoholic fatty liver disease. Gastroenterology 134: 568–576.
  10. 10. Wei Y, Wang D, Topczewski F, Pagliassotti MJ (2006) Saturated fatty acids induce endoplasmic reticulum stress and apoptosis independently of ceramide in liver cells. Am J Physiol Endocrinol Metab 291: E275–281.
  11. 11. Henkel AS, Elias MS, Green RM (2009) Homocysteine supplementation attenuates the unfolded protein response in a murine nutritional model of steatohepatitis. J Biol Chem 284: 31807–31816.
  12. 12. Werstuck GH, Lentz SR, Dayal S, Hossain GS, Sood SK, et al. (2001) Homocysteine-induced endoplasmic reticulum stress causes dysregulation of the cholesterol and triglyceride biosynthetic pathways. J Clin Invest 107: 1263–1273.
  13. 13. Ji C, Kaplowitz N (2003) Betaine decreases hyperhomocysteinemia, endoplasmic reticulum stress, and liver injury in alcohol-fed mice. Gastroenterology 124: 1488–1499.
  14. 14. Wang D, Wei Y, Pagliassotti MJ (2006) Saturated fatty acids promote endoplasmic reticulum stress and liver injury in rats with hepatic steatosis. Endocrinology 147: 943–951.
  15. 15. Xu C, Bailly-Maitre B, Reed JC (2005) Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest 115: 2656–2664.
  16. 16. Zhang K, Kaufman RJ (2004) Signaling the unfolded protein response from the endoplasmic reticulum. J Biol Chem 279: 25935–25938.
  17. 17. Wek RC, Cavener DR (2007) Translational control and the unfolded protein response. Antioxid Redox Signal 9: 2357–2371.
  18. 18. Marciniak SJ, Ron D (2006) Endoplasmic reticulum stress signaling in disease. Physiol Rev 86: 1133–1149.
  19. 19. Walter P, Ron D (2011) The unfolded protein response: from stress pathway to homeostatic regulation. Science 334: 1081–1086.
  20. 20. Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, et al. (2000) Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287: 664–666.
  21. 21. Hu P, Han Z, Couvillon AD, Kaufman RJ, Exton JH (2006) Autocrine tumor necrosis factor alpha links endoplasmic reticulum stress to the membrane death receptor pathway through IRE1alpha-mediated NF-kappaB activation and down-regulation of TRAF2 expression. Mol Cell Biol 26: 3071–3084.
  22. 22. Nguyen DT, Kebache S, Fazel A, Wong HN, Jenna S, et al. (2004) Nck-dependent activation of extracellular signal-regulated kinase-1 and regulation of cell survival during endoplasmic reticulum stress. Molecular Biology of the Cell 15: 4248–4260.
  23. 23. Hu P, Han Z, Couvillon AD, Exton JH (2004) Critical role of endogenous Akt/IAPs and MEK1/ERK pathways in counteracting endoplasmic reticulum stress-induced cell death. J Biol Chem 279: 49420–49429.
  24. 24. Dai R, Chen R, Li H (2009) Cross-talk between PI3K/Akt and MEK/ERK pathways mediates endoplasmic reticulum stress-induced cell cycle progression and cell death in human hepatocellular carcinoma cells. Int J Oncol 34: 1749–1757.
  25. 25. Tang J, Guo YS, Zhang Y, Yu XL, Li L, et al. (2012) CD147 induces UPR to inhibit apoptosis and chemosensitivity by increasing the transcription of Bip in hepatocellular carcinoma. Cell Death Differ 19: 1779–1790.
  26. 26. Honma Y, Harada M (2013) Sorafenib enhances proteasome inhibitor-mediated cytotoxicity via inhibition of unfolded protein response and keratin phosphorylation. Exp Cell Res
  27. 27. Yi P, Higa A, Taouji S, Bexiga MG, Marza E, et al. (2012) Sorafenib-mediated targeting of the AAA(+) ATPase p97/VCP leads to disruption of the secretory pathway, endoplasmic reticulum stress, and hepatocellular cancer cell death. Mol Cancer Ther 11: 2610–2620.
  28. 28. Xu M, Lu N, Zhang H, Dai Q, Wei L, et al. (2012) Wogonin induced cytotoxicity in human hepatocellular carcinoma cells by activation of unfolded protein response and inactivation of AKT. Hepatol Res
  29. 29. Duan Q, Wang X, Gong W, Ni L, Chen C, et al. (2012) ER stress negatively modulates the expression of the miR-199a/214 cluster to regulates tumor survival and progression in human hepatocellular cancer. PLoS One 7: e31518.
  30. 30. Yang F, Zhang L, Wang F, Wang Y, Huo XS, et al. (2011) Modulation of the unfolded protein response is the core of microRNA-122-involved sensitivity to chemotherapy in hepatocellular carcinoma. Neoplasia 13: 590–600.
  31. 31. Chen R, Dai RY, Duan CY, Liu YP, Chen SK, et al. (2011) Unfolded protein response suppresses cisplatin-induced apoptosis via autophagy regulation in human hepatocellular carcinoma cells. Folia Biol (Praha) 57: 87–95.
  32. 32. Belfi CA, Chatterjee S, Gosky DM, Berger SJ, Berger NA (1999) Increased sensitivity of human colon cancer cells to DNA cross-linking agents after GRP78 up-regulation. Biochem Biophys Res Commun 257: 361–368.
  33. 33. Fernandez PM, Tabbara SO, Jacobs LK, Manning FC, Tsangaris TN, et al. (2000) Overexpression of the glucose-regulated stress gene GRP78 in malignant but not benign human breast lesions. Breast Cancer Res Treat 59: 15–26.
  34. 34. Shuda M, Kondoh N, Imazeki N, Tanaka K, Okada T, et al. (2003) Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J Hepatol 38: 605–614.
  35. 35. Han H, Hu J, Lau MY, Feng M, Petrovic LM, et al. (2013) Altered methylation and expression of ER-associated degradation factors in long-term alcohol and constitutive ER stress-induced murine hepatic tumors. Front Genet 4: 224.
  36. 36. Liu P, Kimmoun E, Legrand A, Sauvanet A, Degott C, et al. (2002) Activation of NF-kappa B, AP-1 and STAT transcription factors is a frequent and early event in human hepatocellular carcinomas. J Hepatol 37: 63–71.
  37. 37. Hess J, Angel P, Schorpp-Kistner M (2004) AP-1 subunits: quarrel and harmony among siblings. J Cell Sci 117: 5965–5973.
  38. 38. Shaulian E, Karin M (2002) AP-1 as a regulator of cell life and death. Nat Cell Biol 4: E131–136.
  39. 39. Karin M (1995) The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem 270: 16483–16486.
  40. 40. Johnson GL, Lapadat R (2002) Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science 298: 1911–1912.
  41. 41. Henkel AS, Anderson KA, Dewey AM, Kavesh MH, Green RM (2011) A chronic high-cholesterol diet paradoxically suppresses hepatic CYP7A1 expression in FVB/NJ mice. J Lipid Res 52: 289–298.
  42. 42. Gille H, Kortenjann M, Thomae O, Moomaw C, Slaughter C, et al. (1995) ERK phosphorylation potentiates Elk-1-mediated ternary complex formation and transactivation. EMBO J 14: 951–962.
  43. 43. Whitmarsh AJ, Davis RJ (1996) Transcription factor AP-1 regulation by mitogen-activated protein kinase signal transduction pathways. J Mol Med (Berl) 74: 589–607.
  44. 44. Chang Q, Zhang Y, Beezhold KJ, Bhatia D, Zhao H, et al. (2009) Sustained JNK1 activation is associated with altered histone H3 methylations in human liver cancer. J Hepatol 50: 323–333.
  45. 45. Hui L, Zatloukal K, Scheuch H, Stepniak E, Wagner EF (2008) Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation. J Clin Invest 118: 3943–3953.
  46. 46. Schattenberg JM, Singh R, Wang Y, Lefkowitch JH, Rigoli RM, et al. (2006) JNK1 but not JNK2 promotes the development of steatohepatitis in mice. Hepatology 43: 163–172.