Skip to main content

Glucose 6-phosphate dehydrogenase knockdown enhances IL-8 expression in HepG2 cells via oxidative stress and NF-κB signaling pathway

Abstract

Background

This study was designed to investigate the effect of glucose 6-phosphate dehydrogenase (G6PD) deficiency on pro-inflammatory cytokine secretion using a palmitate-induced inflammation HepG2 in vitro model. The modulation of cellular pro-inflammatory cytokine expression under G6PD deficiency during chronic hepatic inflammation has never been investigated before.

Methods

The culture medium of untreated and palmitate-treated G6PD-scramble (Sc) and G6PD-knockdown (Gi) HepG2 cells were subjected to cytokine array analysis, followed by validation with ELISA and qRT-PCR of the target cytokine. The mechanism of altered cytokine secretion in palmitate-treated Sc and Gi HepG2 cells was examined in the presence of anti-oxidative enzyme (glutathione peroxidase, GPX), anti-inflammatory agent (curcumin), NF-κB inhibitor (BAY11-7085) and specific SiRNA against NF-κB subunit p65.

Results

Cytokine array analysis indicated that IL-8 is most significantly increased in G6PD-knockdown HepG2 cells. The up-regulation of IL-8 caused by G6PD deficiency in HepG2 cells was confirmed in other G6PD-deficient cells by qRT-PCR. The partial reduction of G6PD deficiency-derived IL-8 due to GPX and NF-κB blockers indicated that G6PD deficiency up-regulates pro-inflammatory cytokine IL-8 through oxidative stress and NF-κB pathway.

Conclusions

G6PD deficiency predisposes cells to enhanced production of pro-inflammatory cytokine IL-8. Mechanistically, G6PD deficiency up-regulates IL-8 through oxidative stress and NF-κB pathway. The palmitate-induced inflammation in G6PD-deficient HepG2 cells could serve as an in vitro model to study the role of altered redox homeostasis in chronic hepatic inflammation.

Background

Glucose 6-phosphate dehydrogenase (G6PD) catalyzes the rate-limiting step in the hexose monophosphate shunt with the concomitant generation of reduced nicotinamide adenine dinucleotide phosphate (NADPH), which is involved in cellular reductive biosynthesis and redox homeostasis [1]. G6PD deficiency, also known as favism, is one of the most common genetic disorders in the world affecting an estimated four hundred million people worldwide [2]. G6PD is the only enzyme in erythrocytes to regenerate NADPH and subsequently glutathione (GSH), which protects red cells against oxidative attacks. G6PD-deficient erythrocytes are particularly susceptible to hemolysis upon exposure to oxidants such as fava beans or primaquine. Classically, since G6PD deficiency has been associated with hemolytic crise as a major clinical manifestation [3], most studies on G6PD deficiency have been focused on erythrocytes [4]. Recently, an increasing number of studies have shown that G6PD deficiency not only affects erythrocytes, but also induces aberrations of cellular functions in nucleated cells [5-11].

Redox status plays an essential role in the modulation of inflammation and immune response [12]. During inflammation, the production of intracellular reactive oxygen species (ROS) is involved in triggering inflammatory responses via the secretion of pro-inflammatory cytokines, which can directly affect the course of inflammation-associated diseases [13,14]. Among the pro-inflammatory cytokines, Interleukin-8 (IL-8) has received much attention owing to its capacity to mediate polymorphonuclear neutrophils (PMN) chemotaxis through CXC chemokine receptor 1 (CXCR1) and 2 (CXCR2) [15]. Several stimulating factors are known to up-regulate the expression of IL-8, including lipopolysaccharide (LPS) [16], phytohemagglutinin (PHA) [17], aggregated immune complex (IC) [18], tumor necrosis factor (TNF) [19], Interleukin-1β (IL-1 β) [20] and palmitate [21-24]. The activation of nuclear factor kappa B (NF-κB), NF-IL-6 (C/EBP β) or activator protein-1 (AP-1) is required for the transcription of IL-8 [25-27]. Although redox status is known to modulate cytokines, the relationship between G6PD and pro-inflammatory cytokines, such as IL-8, has been overlooked. Further investigation is warranted for how G6PD modulates pro-inflammatory cytokines and affects the inflammatory response.

Inflammation is a critical component in different types of acute and chronic liver disorders, which consequently progress to hepatitis and fibrosis [28]. In this study, we have adopted a palmitate-induced inflammation HepG2 in vitro model [29] and performed cytokine array analysis to investigate the role of G6PD during chronic hepatic inflammation. We have shown that G6PD modulates the secretion of the pro-inflammatory cytokine IL-8 via oxidative stress and NF-κB pathway because the elevated IL-8 due to G6PD deficiency is partially blocked by exogenous antioxidant and NF-κB inhibitors. These findings suggest that G6PD may play an important role in the modulation of chronic hepatic inflammation.

Methods

Materials

Dulbecco’s modified Eagle’s medium (DMEM), fetal calf serum (FCS), streptomycin, and penicillin were purchased from Invitrogen (CA, USA). Hydrogen peroxide was obtained from Merck (Darmstadt, Germany). BAY11-7085 (NF-κB inhibitor) was purchased from Calbiochem (Millipore, MA, USA). Glutathione peroxidase, curcumin and human SiRNA of scrambled control and p65, were purchased from Sigma (MO, USA)

Cell culture

Human hepatocarcinoma cell lines HepG2, SK-Hep1 and the primary human foreskin fibroblast (HFF) were grown in DMEM supplemented with 10% FCS, antibiotics (100 units/ml penicillin and 100 mg/ml streptomycin) and 5% CO2 at 37°C. The construction and generation of the G6PD scramble control (Sc, G6PD normal) and G6PD shRNA (Gi, G6PD-deficient) HepG2 cells [7,30] as well as G6PD scramble control (SK-i-Sc, G6PD normal) and G6PD shRNA (SK-i-Gi, G6PD-deficient) SK-Hep1 cells [11] were described previously. The isolation and the characterization of normal primary HFF (HFF3, G6PD normal) and its G6PD-deficient HFF counterpart (HFF1, G6PD-deficient) were reported previously [31].

Induction of palmitate overload in HepG2 cells

The induction of palmitate overload in HepG2 cells was performed according to a published protocol [29]. In brief, 3×106 HepG2 cells were cultured in petridish for overnight at 37°C. The culture medium was then replaced with fresh medium containing palmitate and the cells were continued to culture for 24 hr. The palmitate-containing medium was prepared by diluting 150 mM palmitate stock solution (dissolved in isopropanol) to DMEM supplemented with 1% fatty acid free bovine serum albumin (Sigma) followed by overnight incubation at 37°C.

Cell viability assay

The HepG2 cell viability was measured based on the Neutral Red uptake assay described previously [6]. In brief, 1.2×105 untreated or palmitate-treated Sc and Gi HepG2 cells were cultured in 24-well plates for overnight at 37°C. At the end of treatment, cells were incubated with 0.5 ml of 0.033% Neutral Red solution for 2 hr at 37°C followed by fixation with 0.5 ml of 0.1% CaCl2 and 0.5% formaldehyde for 5 min. Subsequently, the incorporated dye was solubilized in 0.5 ml of 1% acetic acid and 50% ethanol solution. The absorbance of Neutral Red was detected at 540 nm with a reference wavelength at 690 nm using a SpectraMax 340PC384 microplate reader (Molecular Devices, CA, USA).

ROS measurement and ROS treatment

The ROS level of palmitate-treated Sc and Gi HepG2 cells was analyzed by flow cytometry based on a previous protocol [6]. In brief, 3×106 HepG2 cells were cultured in petridish for overnight at 37°C. After palmitate treatment, cells were incubated with 20 μM 2′, 7′-dichlorodihydrofluoroscein diacetate (DCF-DA) solution (Molecular Probes, OR, USA) for 30 min at 37°C followed by Trypsin-EDTA treatment. The trypsinized cells were then detected for ROS production by a FACS Calibur flow cytometer (Becton Dickson, CA, USA) (excitation 490 nm, emission 520 nm). The data was analyzed by Cell Quest Pro software (Becton Dickson). For ROS treatment, 3×106 Sc and Gi HepG2 cells were seeded separately in petridish. After 24 hr, the medium was replaced by fresh medium with or without 0.5 mM H2O2 and continued to culture for 24 hr. After the treatment, the medium of Sc and Gi HepG2 cells were collected for the determination of IL-8 secretion.

Gene expression by quantitative real-time PCR

The quantitative real-time PCR (qRT-PCR) was conducted by using SYBR Green PCR Premix reagent (Yeastern Biotechnology, Taipei, Taiwan) with iQ5 real-time thermal cycler (Bio-Rad, CA, USA) according to a previous publication [32]. Primers were designed using Beacon designer software (Bio-Rad). In brief, HepG2 (3×106 cells), SK-Hep1 (3×106 cells), and primary human foreskin fibroblasts (5×105 cells) were seeded separately in petridish and cultured for 3 days until harvesting for RNA isolation. The RNA was extracted by using TRIzol reagent (Life Technologies, CA, USA) according to the instructions provided by the manufacturer. The concentration of RNA was quantified by Nanophotometer (Implen, Munich, Germany). 1 μg of RNA was reversely transcribed to cDNA by using SuperScript III Reverse Transcriptase (Life Technologies) with 0.5 μg of oligo (dT)18 primer (Bioman Scientific, Taipei, Taiwan). The qRT-PCR reaction contained 1 μg of cDNA, primers, and SYBR Green PCR Premix. The thermal cycle program was set as followed: 95°C for 10 min, 40 cycles of 95°C for 15 sec and 60°C for 1 min. The gene expression of IL-8 (forward primer: 5′-CTTTCAGAGACAGCAGAG-3′; reverse primer: 5′-CTAAGTTCTTTAGCACTCC-3′) was normalized against threshold cycle (Ct) values of the housekeeping gene actin (forward primer: 5′-TCCACCTTCCAGCAGATG-3′; reverse primer: 5′-GTGTAACGCAACTAAGTCATAG-3′). The relative index (2-ΔΔCt) was calculated by comparing the average expression level for control samples with the index defined as 1.00.

Cytokine profiling

The cytokine profiling of Sc and Gi HepG2 cells with or without palmitate treatment was determined by using Human Cytokine Array Panel A, Proteome Profiler™ Array (R&D systems). In brief, 3×106 HepG2 cells were cultured in petridish for overnight followed by treating with or without palmitate for 24 hr at 37°C. The culture medium of each sample was collected and concentrated with 5 kDa centrifugal filter units (Millipore, MA, USA). The volume of concentrated medium was adjusted to 1 ml followed by mixing with Cytokine Array Detection Antibody Cocktail for 1 hr at room temperature. The concentrated medium/antibody mixture was then incubated with the nitrocellulose membrane, which contains 36 different anti-cytokine antibodies printed in duplicate, for overnight at 4°C on a rocking shaker (Firstek Scientific, Taiwan). The washed membrane was incubated with diluted Streptavidin-Horseradish Peroxidase (HRP) for 30 min at room temperature on a rocking shaker. The membrane was washed and incubated with chemiluminescent detection reagent for 1 min. Lastly, the membrane was covered with plastic wrap in a X-ray film cassette and exposed to X-ray film (Fujifilm, Japan). The cytokine array data on developed a X-ray film was quantified by scanning the film on a transmission-mode scanner. The array image was analyzed by image analysis software (Image J). The average signal (pixel density) of duplicate spots representing each cytokine was obtained by subtracting averaged background signal on the array. In order to obtain the relative level of each cytokine between different samples, the IL-8 protein level of each sample was first quantified by ELISA. The level of IL-8 was then defined as a reference to which other cytokines were normalized in each array image. After normalization to IL-8, comparison of the signal intensity of each cytokine between array images can be used to determine the relative level of each cytokine in different samples.

Determination of IL-8 by ELISA

The IL-8 protein level in the culture medium secreted by Sc and Gi HepG2 cells with or without palmitate treatment was quantified by the human IL-8 enzyme-linked immunosorbent assay (ELISA) kit (R&D systems). In brief, 3×106 HepG2 cells were cultured in petridish for overnight followed by treating with or without palmitate for 24 hr at 37°C. The culture medium of each sample was collected and concentrated with 5 kDa centrifugal filter units (Millipore). The concentrated medium of each sample and IL-8 standard (50 μl) were transferred to the wells containing 100 μl assay diluent in a flat-bottomed 96-well plate coated with IL-8 antibody and incubated for 2 hr at room temperature. The plate was aspirated and washed followed by adding 100 μl IL-8 conjugate (IL-8 antibody conjugated to HRP) for 1 hr at room temperature. The plate was then aspirated and washed for four times. The working solution of Streptavidin-HRP was added subsequently and incubated for 30 min at room temperature followed by aspiration. Lastly, the stabilized chromogen was added to the plate for 30 min at room temperature in the dark followed by adding stop solution. The absorbance of each well was detected at 450 nm with a reference at 490 nm in a microplate reader (Versa MAX, Molecular Devices). The recombinant human IL-8 protein (R&D systems) was used for the generation of a standard curve for IL-8.

Small interfering RNA (SiRNA)

3×105 HepG2 cells were cultured in a 6-well plate for overnight at 37°C. The culture medium was subsequently replaced with fresh DMEM and incubated for 1 hr before SiRNA transfection. The cells were transfected with SiRNA oligonucleotides using 7.5 μl PolyJet In Vitro DNA Transfection Reagent (SignaGen Laboratories, MD, USA). Two different SiRNA oligonucleotides were used: (1) SiRNA against non-targeting scramble control, (2) SiRNA against human NF-κB subunit p65. Both Sc and Gi HepG2 cells were transfected with SiRNA-control and SiRNA-p65 in different concentrations (50, 100, 150 nM) in a total volume of 1 ml DMEM per well. After 72 hr incubation, the culture medium of each well was collected for the determination of IL-8 secretion by ELISA. The cells in each well were harvested for isolation of RNA and protein for measuring IL-8 mRNA and p65 level by qRT-PCR and Western blotting, respectively.

Western blot analysis

The protein lysate isolated from cells was resolved by SDS-PAGE (TGX Fastcast, Bio-Rad) and electrotransferred by semi-dry Western blotting (Trans-Blot Turbo blotting system, Bio-Rad). The immunoblotting was conducted by using antibody against human p65 (Cell Signaling technology, MA, USA), actin (Santa Cruz Biotechnology, TX, USA) according to the protocols provided by manufacturers.

Statistical analysis

Data were presented as means ± S.D. Student’s t-test was used to analyze the statistical difference between G6PD-scramble (G6PD normal) and G6PD-knockdown (G6PD-deficient) HepG2 cells. Comparisons between different concentrations or time course of palmitate treatment were evaluated by one-way analysis of variance followed by Tukey’s multiple comparison test. Value of P < 0.05 was considered statistically significant.

Results

Establishment of a G6PD-knockdown HepG2 cell model with or without palmitate treatment

To investigate the role of G6PD in chronic hepatic inflammation, we adopted a stable G6PD-knockdown HepG2 cell model [6] undergoing lipid-induced inflammation [29]. Consistent with previous report, the G6PD-knockdown (Gi) HepG2 cells displayed a significant reduction (10-fold, P < 0.05) of G6PD catalytic activity (see Additional file 1 for all supplementary materials; Additional file 2: Figure S1a) as well as protein expression (Additional file 2: Figure S1b) compared to its G6PD normal counterpart, G6PD-scramble (Sc) HepG2 cells. Upon palmitate treatment, both Sc and Gi HepG2 cells showed an increase of lipid deposition as indicated by microscopic examination with Sudan Red staining (Additional file 3: Figure S2). This observation was corroborated by flow cytometry analysis with Nile Red staining (Additional file 4: Figure S3). To address whether palmitate treatment induced cytotoxicity in HepG2 cell upon G6PD deficiency, the cell viability was determined in control and palmitate-treated Sc and Gi HepG2 cells by Neutral Red uptake assay (Table 1). The result showed that the viability of Sc and Gi HepG2 cells was reduced by palmitate treatment in a dose-dependent fashion. At 0.4 and 0.6 mM palmitate, there was a significant difference of viability between Sc and Gi HepG2 cells (P < 0.05). These data demonstrate that in the palmitate-treated cell model, G6PD deficiency does not affect lipid accumulation, but renders cells more susceptible to cytotoxic effect at higher palmitate concentrations.

Table 1 The effect of palmitate treatments on the viability of HepG2 cells

Enhancement of pro-inflammatory cytokine secretion in G6PD-knockdown HepG2 cells

Since palmitate induces pro-inflammatory cytokine IL-8 secretion in hepatocytes [21], we investigated whether G6PD deficiency affected pro-inflammatory cytokine secretion in the palmitate-treated cell model by cytokine array analysis. Among 36 different cytokines tested, several cytokines (IL-8, IL-1ra, MIF, sICAM-1, and Serpin E1) were detected (Additional file 5: Figure S4 and see the array format in Additional file 6: Table S1). Interestingly, G6PD deficiency enhanced secretion of these cytokines at basal and palmitate treatment conditions (Table 2). In particular, the IL-8 secretion showed a 9-fold increase in palmitate-treated Gi HepG2 cells compared with untreated Sc HepG2 cells, whereas the secretion of other high level cytokines showed a 5-fold increase. Since IL-8 showed a most dramatic increase in cytokine array, we validated its up-regulation in Gi HepG2 cells by ELISA (Figure 1a) and qRT-PCR (Figure 2). The ELISA data confirmed that IL-8 secretion was significantly increased (P < 0.05) in palmitate-treated Gi HepG2 cells (Figure 1a) in a palmitate-dependent manner (Figure 1b). Likewise, the qRT-PCR result also validated that G6PD deficiency significantly up-regulated IL-8 gene expression upon palmitate treatment in a palmitate-dependent manner (Figure 2).

Table 2 List of differentially expressed cytokines of Sc and Gi HepG2 cells in basal and palmitate-treated conditions identified by cytokine array
Figure 1
figure 1

The effect of G6PD deficiency on IL-8 secretion in palmitate-treated HepG2 cells. (a) The IL-8 secretion of Sc and Gi HepG2 cells with or without palmitate treatment (0.6 mM, 24 hr) was measured by ELISA. (b) The IL-8 secretion of Sc and Gi HepG2 cells treated with different concentrations of palmitate (0.2, 0.3, 0.4 mM) for 24 hr was analyzed by ELISA. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells. #indicates a significant difference (P < 0.05) between control and palmitate-treated HepG2 cells.

Figure 2
figure 2

The effect of G6PD deficiency on IL-8 mRNA level in palmitate-treated HepG2 cells. The mRNA level of IL-8 in Sc and Gi HepG2 cells treated with different concentrations of palmitate (0.2, 0.4, 0.6, 0.8 mM) for 24 hr was analyzed by qRT-PCR. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells.

Enhancement of IL-8 expression in other G6PD-deficient cells

To rule out the up-regulation of IL-8 induced by G6PD deficiency in HepG2 cell was merely a cell-specific observation, the IL-8 level of other G6PD-deficient cells (SK-Hep1 hepatoma cells and HFF fibroblasts) was analyzed by qRT-PCR (Figure 3). Significantly elevated IL-8 gene expression in G6PD-deficient SK-Hep1 and HFF cells was also observed. These findings suggest that the up-regulation of IL-8 induced by G6PD deficiency is not a cell-specific, but a general phenomenon.

Figure 3
figure 3

The effect of G6PD deficiency on IL-8 mRNA level in cells. The mRNA level of IL-8 in G6PD normal and G6PD-deficient cells (HepG2, SK-Hep1, and HFF) was determined by qRT-PCR. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between G6PD normal and G6PD-deficient cells.

Involvement of NF-κB in G6PD deficiency induced IL-8 secretion in HepG2 cells

It has been reported that G6PD activates NF-κB in β-cells and adipocytes [33,34]. To investigate whether NF-κB pathway was involved in the enhancement of IL-8 secretion in G6PD-knockdown cells, Sc and Gi HepG2 cells with or without 0.6 mM palmitate treatment were exposed to 10 μM NF-κB inhibitor (BAY11-7085) followed by quantification of IL-8 secretion. As shown in Figure 4a, IL-8 secretion was significantly reduced by the NF-κB inhibitor in Sc HepG2 cells without palmitate treatment (75% decrease, P < 0.05) compared to that in untreated control cells. Similarly, the inhibitor reduced the IL-8 secretion in Gi HepG2 cells compared to that in untreated Gi HepG2 cells without palmitate treatment (64% decrease, P < 0.05). In the palmitate-treated cells, the NF-κB inhibitor significantly reduced IL-8 secretion (80% decrease, P < 0.05) in Sc HepG2 cells. Likewise, the NF-κB inhibitor significantly reduced the palmitate-induced IL-8 secretion in Gi HepG2 cells (72% decrease, P < 0.05) compared with palmitate-treated only Gi HepG2 cells. To further investigate the association of G6PD and NF-κB signaling on IL-8 secretion, a G6PD/ NF-κB(p65) double-knockdown cell line was established (Figure 4b). Because the immunoblotting result showed that the protein level of p65 was diminished markedly by SiRNA against p65 at 150 nM, such condition was used for the measurement of IL-8 level. The knockdown of NF-κB(p65) in Sc and Gi HepG2 cells resulted in a significant reduction of IL-8 mRNA (60% decrease, P < 0.05) compared with Sc and Gi HepG2 cells treated with control SiRNA (Figure 4c). In addition, Sc and Gi HepG2 cells treated with p65 SiRNA exhibited diminished IL-8 secretion (40% decrease, P < 0.05) compared to Sc and Gi HepG2 cells treated with control SiRNA (Figure 4d). The blockade of NF-κB due to selective inhibitor or specific SiRNA against NF-κB confirms that the up-regulation of IL-8 caused by G6PD deficiency is linked to NF-κB signaling. Nevertheless, the finding that the inhibition of NF-κB can not fully suppress the elevated IL-8 in Gi HepG2 cells suggests that besides NF-κB pathway there maybe alternative regulatory mechanism modulated by G6PD.

Figure 4
figure 4

The effect of NF-κB inhibition on IL-8 secretion in HepG2 cells. (a) The effect of NF-κB inhibitor on IL-8 secretion with or without palmitate treatment. The IL-8 secretion of Sc and Gi HepG2 cells treated with 10 μM NF-κB inhibitor (BAY11-7085) with or without 0.6 mM palmitate for 24 hr was measured by ELISA. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells. #indicates a significant difference (P < 0.05) between control and inhibitor (BAY)-treated HepG2 cells without palmitate. α indicates a significant difference (P < 0.05) between palmitate (PA)-treated cells without inhibitor (BAY) and palmitate (PA)-inhibitor (BAY) co-treated HepG2 cells. (b) The effect of p65 knockdown in Sc and Gi HepG2 cells on p65 protein level determined by Western blotting. The blot (cell lysate: 25 μg protein content) was incubated with anti-p65 antibody and subsequently stripped for incubation with anti-actin antibody as loading control. (c) The effect of p65 knockdown on IL-8 gene expression by qRT-PCR and (d) IL-8 secretion by ELISA. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between SiRNA- control and SiRNA- p65.

Enhancement of IL-8 production by H2O2

G6PD-deficient HepG2 cells are extremely sensitive to cytotoxic effects induced by oxidants with the concomitant production of oxidative stress [6,7]. To determine whether the enhanced IL-8 expression in palmitate-treated Gi HepG2 cells was due to oxidative stress, the ROS level of Sc and Gi HepG2 cells in control and palmitate-treated conditions was determined. As shown in Figure 5, Gi HepG2 cells without palmitate treatment displayed a 1.4-fold increase of ROS level (P < 0.05) compared with that of control Sc HepG2 cells, while in palmitate-treated Sc and Gi HepG2 cells there was an increase in ROS over that in control with a similar ratio between the two cell types. To examine the effect of oxidative stress on IL-8 secretion, both Sc and Gi HepG2 cells were treated with 0.5 mM H2O2 added exogenously according to a previous protocol [6] followed by quantification of IL-8 secretion. As shown in Figure 6, H2O2 treatment significantly enhanced IL-8 secretion in both Sc and Gi HepG2 cells (5-fold increase, P < 0.05) compared with that in control cells. The dramatic increased in IL-8 secretion by H2O2, particularly in Gi HepG2 cells, is consistent with the notion that IL-8 secretion in HepG2 cells is highly sensitive to oxidative stress.

Figure 5
figure 5

The effect of G6PD deficiency on ROS production in HepG2 cells with or without palmitate treatment. The ROS production of palmitate-treated (0.3 mM, 24 hr) Sc and Gi HepG2 cells was determined by DCF-DA staining and analyzed by flow cytometry. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells. #indicates a significant difference (P < 0.05) between control and palmitate-treated HepG2 cells.

Figure 6
figure 6

The effect of exogenous hydrogen peroxide on IL-8 secretion in HepG2 cells. The IL-8 secretion of Sc and Gi HepG2 cells with or without treatment of 0.5 mM H2O2 for 24 hr was measured by ELISA. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells. #indicates a significant difference (P < 0.05) between control and H2O2-treated HepG2 cells.

Modulation of IL-8 production by glutathione peroxidase and curcumin

To corroborate that oxidative stress is a causative agent of IL-8 up-regulation in palmitate-treated Sc and Gi HepG2 cells, anti-oxidative enzyme glutathione peroxidase (GPX) was examined for its effect on IL-8 level. As shown in Figure 7a, exogenous addition of GPX did not affect IL-8 secretion in Sc and Gi HepG2 cells compared with untreated control cells (P = 0.18, P = 0.25 respectively). Despite GPX did not affect palmitate-induced IL-8 in Sc HepG2 cells (P = 0.35), GPX partially suppressed palmitate-induced IL-8 (21% decrease, P < 0.05) in Gi HepG2 cells compared with that in palmitate-treated Gi HepG2 cells without GPX (Figure 7a). The finding that there is a significant difference between Sc and Gi cells in palmitate/GPX co-treatment but not in GPX treatment alone, indicating that G6PD is required for lowering pro-inflammatory cytokine secretion caused by excess ROS due to palmitate stimulation.

Figure 7
figure 7

The effect of GPX and curcumin on IL-8 secretion in Sc and Gi HepG2 cells with or without palmitate treatment. (a) The IL-8 secretion of Sc and Gi HepG2 cells co-treated with 2 U/ml glutathione peroxidase (GPX) and 0.6 mM palmitate (PA) for 24 hr was measured by ELISA. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells. α indicates a significant difference (P < 0.05) between PA without GPX and PA-GPX co-treated HepG2 cells. (b) The IL-8 mRNA of Sc and Gi HepG2 cells co-treated with 50 or 100 μΜ curcumin (referred to cur50 and cur100, respectively) and 0.6 mM palmitate (PA) for 6 hr was measured by qRT-PCR. These results are representative of at least three separate experiments. *indicates a significant difference (P < 0.05) between Sc and Gi HepG2 cells. #indicates a significant difference between control and curcumin-treated HepG2 cells without palmitate. α indicates a significant difference (P < 0.05) between PA without curcumin and PA-curcumin co-treated HepG2 cells.

On the other hand, curcumin was used as an anti-inflammatory agent in this study. As shown in Figure 7b, curcumin treatments (50 and 100 μM) significantly reduced the IL-8 level of Sc and Gi HepG2 cells without palmitate treatment (50 μM: Sc, 71% decrease, Gi, 67% decrease; 100 μM: Sc, 70% decrease, Gi, 61% decrease, P < 0.05) compared with control cells without palmitate treatment (Figure 7b). Similarly, the IL-8 level of Sc and Gi HepG2 cells with palmitate treatment was significantly decreased by 50 μM curcumin (Sc, 55% decrease, Gi, 67% decrease, P < 0.05) when compared with that of palmitate-treated cells without curcumin. Further inhibition of palmitate-induced IL-8 in both cells occurred when treated with 100 μM curcumin (Sc, 84% decrease; Gi, 80% decrease, P < 0.05) compared to that of palmitate-treated cells without curcumin. These data demonstrate that curcumin significantly suppresses palmitate-induced IL-8 level in a dose dependent manner in both Sc and Gi HepG2 cells.

Discussion

As the culprit causing many abnormal cellular functions, G6PD deficiency is well-known for inducing redox imbalance [2,3,6,7,10,11,31,35], which exacerbates cellular inflammatory response during disease progression [36,37]. Spolarics et al. have shown that ROS detoxifying enzymes, including G6PD, superoxide dismutase (SOD) and glutathione peroxidase (GPX) are up-regulated in hepatic endothelial cells of LPS-treated rats [38]. The increased antioxidant capacity indicates a protective role of G6PD against oxidative stress during hepatic inflammatory response [39]. Using G6PD-knockdown HepG2 cells, we have demonstrated in this study that elevated IL-8 secretion is paralleled with enhanced production of ROS (Figure 5). Such IL-8 elevation can be further enhanced by externally applied H2O2 (Figure 6). Our finding of ROS-induced IL-8 is in accord with previous reports in MKN28 cells [40] and HepG2 cells [41].

H2O2 is one of the well-known ROS species that modulates many signaling pathways [42]. H2O2 activates tumor necrosis factor-alpha (TNF-α) induced-NF-κB, whereas the antioxidant N-acetyl-L-cysteine (NAC) suppresses TNF-α induced-NF-κB [40]. G6PD can activate NF-κB in different cell types, including adipocytes in obesity and β-cells in type-two diabetes [33,34]. Indeed, NF-κB is involved in the ROS-induced IL-8 transcription [43]. Additional support to the notion that NF-κB is involved in the up-regulation of IL-8 either by palmitate treatment or G6PD knockdown in HepG2 cells comes from our study using the NF-κB inhibitor (BAY11-7085), which decreases IL-8 secretion at both basal and palmitate treated conditions (Figure 4a). The fact that knockdown of NF-κB subunit p65 by SiRNA causes a marked drop in IL-8 level (Figure 4c-d) further implicates a close relationship between NF-κB signaling and G6PD deficiency-derived oxidative stress in inflammation.

Antioxidants have been shown to exert the inhibitory effect on oxidative stress and inflammatory response caused by NF-κB activation [43-46]. In our current study, the exogenous addition of ROS scavenging enzyme GPX partially suppresses the up-regulation of IL-8 in palmitate-treated G6PD-knockdown HepG2 cells (Figure 7a). The suppression of IL-8 by GPX is similar to a previous finding that ROS scavenging agent NAC or dimethylsulfoxide (DMSO) inhibits IL-8 expression induced by TNF-α or IL-1β [40]. The possible explanation of IL-8 suppression by external addition of GPX in the culture medium is that GPX can protect against intracellular H2O2 readily crosses cell membrane similar to the action of catalase [47]. Thus extracellular GPX can dissipate H2O2 out of the cells and down-regulate H2O2-induced IL-8.

Curcumin (diferuloylmethane) is the pharmacologically active ingredient found in the spice turmeric (Curcuma longa) [48]. Recent studies suggest that curcumin exhibits anti-oxidative [49-51] and anti-inflammatory [52-55] activities. In addition, curcumin modulates several transcription factors, including AP-1, PPAR-γ, STAT, Nrf-2 and Wnt/β-catenin, [56], protein kinases MAP kinase p38 and ERK [57,58] and inflammatory cytokines TNF-α, IL-6 and IL-1β [59,60]. In our current study, we show that curcumin effectively reduces palmitate-induced IL-8 mRNA level (Figure 7b). Curcumin can ameliorate pro-inflammatory cytokine production [61], however, it has been reported that exposure of curcumin significantly induces apoptosis and cytochrome c release in HepG2 cells as early as 6 hours after treatment [62]. In our experimental condition, IL-8 secretion by short term palmitate-treated HepG2 cells is too low to be detected, whereas significantly increased IL-8 mRNA level in palmitate-treated HepG2 cells can be detected at 6 hours. Hence, we determined the effect of curcumin on IL-8 level in palmitate-treated HepG2 cells at 6 hours by qRT-PCR instead of ELISA.

The inactivation of NF-κB is a well-established mechanism of curcumin described in the literature [55,57,63,64]. It has been shown that curcumin suppresses the phosphorylation of IκBα (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha) through inactivation of IKK (IkappaB kinase) activity [65]. Moreover, curcumin down-regulates the expression of pro-inflammatory gene products regulated by NF-κB, including IL-8, through inhibiting IKK activity in intestinal epithelial cells [66]. A recent study in sepsis-induced acute lung injury rats shows that curcumin significantly enhances SOD activity and reduces lipid peroxidation in the lung [51]. Furthermore, curcumin down-regulates inflammatory cytokines TNF-α, IL-8 and MIF levels in the lung, suggesting a protective role in counteracting inflammation through down-regulation of pro-inflammatory cytokines and oxidative stress. Given that curcumin exerts its inhibitory actions through multiple targets, it is reasonable to speculate that curcumin may act as a non-specific anti-inflammatory agent in our study. Such speculation may justify its superior IL-8 inhibition capacity compared with GPX and NF-κB inhibitor in this study.

Several reports suggest that G6PD deficiency modulates cytokine response during inflammatory and immune responses. In G6PD mutant endotoxemic mice, altered cytokines, including elevated blood IL-6 level, has been documented [67,68]. Clinical studies have indicated that G6PD deficiency correlates with increased incidence of sepsis [69,70]. Moreover, diminished IL-10 and IFN-γ and increased IL-6 are present in African and Mediterranean forms of G6PD-deficient trauma patients [71]. Similarly, reduced monocyte IL-10 in G6PD-deficient trauma patients has been documented [72]. In contrast to the findings in G6PD-deficient adults, a more recent study has reported that the toll-like receptor (TLR) agonists-induced cytokine response in peripheral blood mononuclear cells (PBMCs) isolated from G6PD-deficient infants, including TNF-α, IL-6 and IL-10, is not different from PBMCs of G6PD normal subjects [73]. The discrepancy between G6PD-deficient adults and infants may lie in the relative immature innate immune response during infancy [74,75]. Additionally, the age of the subjects may also contribute to the disparity, because G6PD activity has been suggested to be inversely proportional to age [76].

Conclusion

We have found that the secretion of pro-inflammatory cytokine IL-8 is most significantly increased in G6PD-deficient HepG2 cells by utilizing a cytokine array. Adopting a palmitate-induced inflammation HepG2 cell model, we have found that G6PD deficiency exacerbates pro-inflammatory cytokine IL-8 secretion in HepG2 cells. Mechanistically, G6PD deficiency up-regulates IL-8 through oxidative stress and NF-κB pathway. The palmitate-induced inflammation in G6PD-deficient HepG2 cells could serve as an in vitro model to study the role of altered redox homeostasis in chronic hepatic inflammation.

Abbreviations

G6PD:

Glucose 6-phosphate dehydrogenase

ROS:

Reactive oxygen species

IL-8:

Interleukin-8

NADPH:

Nicotinamide adenine dinucleotide phosphate

ELISA:

Enzyme-linked immunosorbent assay

mRNA:

Messenger RNA

qRT-PCR:

Quantitative real time polymerase chain reaction

SiRNA:

Small interference RNA

DCFDA:

2′, 7′ dichlorodihydrofluoroscein diacetate

H2O2 :

Hydrogen peroxide

GPX:

Glutathione peroxidase

NF-κB:

Nuclear factor-kappa B

Reference

  1. Arese P, Gallo V, Pantaleo A, Turrini F. Life and Death of Glucose-6-Phosphate Dehydrogenase (G6PD) Deficient Erythrocytes - Role of Redox Stress and Band 3 Modifications. Transfus Med Hemother. 2012;39:328–34.

    Article  PubMed Central  PubMed  Google Scholar 

  2. Ho HY, Cheng ML, Chiu DT. Glucose-6-phosphate dehydrogenase - beyond the realm of red cell biology. Free Radic Res. 2014;48:1028–48.

    Article  CAS  PubMed  Google Scholar 

  3. Scott MD, Zuo L, Lubin BH, Chiu DT. NADPH, not glutathione, status modulates oxidant sensitivity in normal and glucose-6-phosphate dehydrogenase-deficient erythrocytes. Blood. 1991;77:2059–64.

    CAS  PubMed  Google Scholar 

  4. Beutler E. Glucose-6-phosphate dehydrogenase deficiency: a historical perspective. Blood. 2008;111:16–24.

    Article  CAS  PubMed  Google Scholar 

  5. Lin HR, Wu CC, Wu YH, Hsu CW, Cheng ML, Chiu DT. Proteome-wide dysregulation by glucose-6-phosphate dehydrogenase (G6PD) reveals a novel protective role for G6PD in aflatoxin B(1)-mediated cytotoxicity. J Proteome Res. 2013;12:3434–48.

    Article  CAS  PubMed  Google Scholar 

  6. Lin CJ, Ho HY, Cheng ML, You TH, Yu JS, Chiu DT. Impaired dephosphorylation renders G6PD-knockdown HepG2 cells more susceptible to H(2)O(2)-induced apoptosis. Free Radic Biol Med. 2010;49:361–73.

    Article  CAS  PubMed  Google Scholar 

  7. Gao LP, Cheng ML, Chou HJ, Yang YH, Ho HY, Chiu DT. Ineffective GSH regeneration enhances G6PD-knockdown Hep G2 cell sensitivity to diamide-induced oxidative damage. Free Radic Biol Med. 2009;47:529–35.

    Article  CAS  PubMed  Google Scholar 

  8. Cheng ML, Ho HY, Liang CM, Chou YH, Stern A, Lu FJ, et al. Cellular glucose-6-phosphate dehydrogenase (G6PD) status modulates the effects of nitric oxide (NO) on human foreskin fibroblasts. FEBS Lett. 2000;475:257–62.

    Article  CAS  PubMed  Google Scholar 

  9. Cheng ML, Ho HY, Lin HY, Lai YC, Chiu DT. Effective NET formation in neutrophils from individuals with G6PD Taiwan-Hakka is associated with enhanced NADP(+) biosynthesis. Free Radic Res. 2013;47:699–709.

    Article  CAS  PubMed  Google Scholar 

  10. Cheng ML, Ho HY, Wu YH, Chiu DT. Glucose-6-phosphate dehydrogenase-deficient cells show an increased propensity for oxidant-induced senescence. Free Radic Biol Med. 2004;36:580–91.

    Article  CAS  PubMed  Google Scholar 

  11. Ho HY, Cheng ML, Shiao MS, Chiu DT. Characterization of global metabolic responses of glucose-6-phosphate dehydrogenase-deficient hepatoma cells to diamide-induced oxidative stress. Free Radic Biol Med. 2013;54:71–84.

    Article  CAS  PubMed  Google Scholar 

  12. Gostner JM, Becker K, Fuchs D, Sucher R. Redox regulation of the immune response. Redox Rep. 2013;18:88–94.

    Article  CAS  PubMed  Google Scholar 

  13. Padgett LE, Broniowska KA, Hansen PA, Corbett JA, Tse HM. The role of reactive oxygen species and proinflammatory cytokines in type 1 diabetes pathogenesis. Ann N Y Acad Sci. 2013;1281:16–35.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Rolo AP, Teodoro JS, Palmeira CM. Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis. Free Radic Biol Med. 2012;52:59–69.

    Article  CAS  PubMed  Google Scholar 

  15. Russo RC, Garcia CC, Teixeira MM, Amaral FA. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev Clin Immunol. 2014;10:593–619.

    Article  CAS  PubMed  Google Scholar 

  16. He W, Qu T, Yu Q, Wang Z, Lv H, Zhang J, et al. LPS induces IL-8 expression through TLR4, MyD88, NF-kappaB and MAPK pathways in human dental pulp stem cells. Int Endod J. 2013;46:128–36.

    Article  CAS  PubMed  Google Scholar 

  17. Liebler JM, Kunkel SL, Burdick MD, Standiford TJ, Rolfe MW, Strieter RM. Production of IL-8 and monocyte chemotactic peptide-1 by peripheral blood monocytes. Disparate responses to phytohemagglutinin and lipopolysaccharide. J Immunol. 1994;152:241–9.

    CAS  PubMed  Google Scholar 

  18. Xiao S, Xu C, Jarvis JN. C1q-bearing immune complexes induce IL-8 secretion in human umbilical vein endothelial cells (HUVEC) through protein tyrosine kinase- and mitogen-activated protein kinase-dependent mechanisms: evidence that the 126 kD phagocytic C1q receptor mediates immune complex activation of HUVEC. Clin Exp Immunol. 2001;125:360–7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. O’Hara AM, Bhattacharyya A, Bai J, Mifflin RC, Ernst PB, Mitra S, et al. Tumor necrosis factor (TNF)-alpha-induced IL-8 expression in gastric epithelial cells: role of reactive oxygen species and AP endonuclease-1/redox factor (Ref)-1. Cytokine. 2009;46:359–69.

    Article  PubMed Central  PubMed  Google Scholar 

  20. Liu X, Ye F, Xiong H, Hu D, Limb GA, Xie T, et al. IL-1beta Upregulates IL-8 Production in Human Muller Cells Through Activation of the p38 MAPK and ERK1/2 Signaling Pathways. Inflammation. 2014;37:1486–95.

    Article  CAS  PubMed  Google Scholar 

  21. Joshi-Barve S, Barve SS, Amancherla K, Gobejishvili L, Hill D, Cave M, et al. Palmitic acid induces production of proinflammatory cytokine interleukin-8 from hepatocytes. Hepatology. 2007;46:823–30.

    Article  CAS  PubMed  Google Scholar 

  22. Igoillo-Esteve M, Marselli L, Cunha DA, Ladriere L, Ortis F, Grieco FA, et al. Palmitate induces a pro-inflammatory response in human pancreatic islets that mimics CCL2 expression by beta cells in type 2 diabetes. Diabetologia. 2010;53:1395–405.

    Article  CAS  PubMed  Google Scholar 

  23. Choi SE, Kim TH, Yi SA, Hwang YC, Hwang WS, Choe SJ, et al. Capsaicin attenuates palmitate-induced expression of macrophage inflammatory protein 1 and interleukin 8 by increasing palmitate oxidation and reducing c-Jun activation in THP-1 (human acute monocytic leukemia cell) cells. Nutr Res. 2011;31:468–78.

    Article  CAS  PubMed  Google Scholar 

  24. Quan J, Liu J, Gao X, Liu J, Yang H, Chen W, et al. Palmitate induces interleukin-8 expression in human aortic vascular smooth muscle cells via Toll-like receptor 4/nuclear factor-kappaB pathway (TLR4/NF-kappaB-8). J Diabetes. 2014;6:33–41.

    Article  CAS  PubMed  Google Scholar 

  25. Yasumoto K, Okamoto S, Mukaida N, Murakami S, Mai M, Matsushima K. Tumor necrosis factor alpha and interferon gamma synergistically induce interleukin 8 production in a human gastric cancer cell line through acting concurrently on AP-1 and NF-kB-like binding sites of the interleukin 8 gene. J Biol Chem. 1992;267:22506–11.

    CAS  PubMed  Google Scholar 

  26. Edwards MR, Mukaida N, Johnson M, Johnston SL. IL-1beta induces IL-8 in bronchial cells via NF-kappaB and NF-IL6 transcription factors and can be suppressed by glucocorticoids. Pulm Pharmacol Ther. 2005;18:337–45.

    Article  CAS  PubMed  Google Scholar 

  27. Yang BC, Yang ZH, Pan XJ, Xiao FJ, Liu XY, Zhu MX, et al. Crotonaldehyde-exposed macrophages induce IL-8 release from airway epithelial cells through NF-kappaB and AP-1 pathways. Toxicol Lett. 2013;219:26–34.

    Article  CAS  PubMed  Google Scholar 

  28. Szabo G, Mandrekar P, Dolganiuc A. Innate immune response and hepatic inflammation. Semin Liver Dis. 2007;27:339–50.

    Article  CAS  PubMed  Google Scholar 

  29. Gomez-Lechon MJ, Donato MT, Martinez-Romero A, Jimenez N, Castell JV, O’Connor JE. A human hepatocellular in vitro model to investigate steatosis. Chem Biol Interact. 2007;165:106–16.

    Article  CAS  PubMed  Google Scholar 

  30. Ho HY, Cheng ML, Wang YH, Chiu DT. Flow cytometry for assessment of the efficacy of siRNA. Cytometry A. 2006;69:1054–61.

    Article  PubMed  Google Scholar 

  31. Ho HY, Cheng ML, Lu FJ, Chou YH, Stern A, Liang CM, et al. Enhanced oxidative stress and accelerated cellular senescence in glucose-6-phosphate dehydrogenase (G6PD)-deficient human fibroblasts. Free Radic Biol Med. 2000;29:156–69.

    Article  CAS  PubMed  Google Scholar 

  32. Yang HC, Chen TL, Wu YH, Cheng KP, Lin YH, Cheng ML, et al. Glucose 6-phosphate dehydrogenase deficiency enhances germ cell apoptosis and causes defective embryogenesis in Caenorhabditis elegans. Cell Death Dis. 2013;4, e616.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  33. Lee JW, Choi AH, Ham M, Kim JW, Choe SS, Park J, et al. G6PD up-regulation promotes pancreatic beta-cell dysfunction. Endocrinology. 2011;152:793–803.

    Article  CAS  PubMed  Google Scholar 

  34. Park J, Choe SS, Choi AH, Kim KH, Yoon MJ, Suganami T, et al. Increase in glucose-6-phosphate dehydrogenase in adipocytes stimulates oxidative stress and inflammatory signals. Diabetes. 2006;55:2939–49.

    Article  CAS  PubMed  Google Scholar 

  35. Ho HY, Cheng ML, Chiu DT. Glucose-6-phosphate dehydrogenase–from oxidative stress to cellular functions and degenerative diseases. Redox Rep. 2007;12:109–18.

    Article  CAS  PubMed  Google Scholar 

  36. Moret I, Cerrillo E, Navarro-Puche A, Iborra M, Rausell F, Tortosa L, et al. Oxidative stress in Crohn’s disease. Gastroenterol Hepatol. 2014;37:28–34.

    Article  PubMed  Google Scholar 

  37. Crowley SD. The Cooperative Roles of Inflammation and Oxidative Stress in the Pathogenesis of Hypertension. Antioxid Redox Signal. 2014;20:102–20.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Spolarics Z, Stein DS, Garcia ZC. Endotoxin stimulates hydrogen peroxide detoxifying activity in rat hepatic endothelial cells. Hepatology. 1996;24:691–6.

    Article  CAS  PubMed  Google Scholar 

  39. Spolarics Z. Endotoxemia, pentose cycle, and the oxidant/antioxidant balance in the hepatic sinusoid. J Leukoc Biol. 1998;63:534–41.

    CAS  PubMed  Google Scholar 

  40. Shimada T, Watanabe N, Hiraishi H, Terano A. Redox regulation of interleukin-8 expression in MKN28 cells. Dig Dis Sci. 1999;44:266–73.

    Article  CAS  PubMed  Google Scholar 

  41. DeForge LE, Preston AM, Takeuchi E, Kenney J, Boxer LA, Remick DG. Regulation of interleukin 8 gene expression by oxidant stress. J Biol Chem. 1993;268:25568–76.

    CAS  PubMed  Google Scholar 

  42. Sies H. Role of metabolic H2O2 generation: redox signaling and oxidative stress. J Biol Chem. 2014;289:8735–41.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  43. Li YJ, Shimizu T, Hirata Y, Inagaki H, Takizawa H, Azuma A, et al. EM, EM703 inhibit NF-kB activation induced by oxidative stress from diesel exhaust particle in human bronchial epithelial cells: importance in IL-8 transcription. Pulm Pharmacol Ther. 2013;26:318–24.

    Article  CAS  PubMed  Google Scholar 

  44. Amore A, Formica M, Giacchino F, Gigliola G, Bonello F, Conti G, et al. N-Acetylcysteine in hemodialysis diabetic patients resets the activation of NF-kB in lymphomonocytes to normal values. J Nephrol. 2013;26:778–86.

    Article  CAS  PubMed  Google Scholar 

  45. Li DY, Xue MY, Geng ZR, Chen PY. The suppressive effects of Bursopentine (BP5) on oxidative stress and NF-kB activation in lipopolysaccharide-activated murine peritoneal macrophages. Cell Physiol Biochem. 2012;29:9–20.

    Article  PubMed  Google Scholar 

  46. Wilkins R, Tucci M, Benghuzzi H. Role of plant-derived antioxidants on NF-kb expression in LPS-stimulated macrophages - biomed 2011. Biomed Sci Instrum. 2011;47:222–7.

    CAS  PubMed  Google Scholar 

  47. Halliwell B. Cell culture, oxidative stress, and antioxidants: avoiding pitfalls. Biomed J. 2014;37:99–105.

    PubMed  Google Scholar 

  48. Chainani-Wu N. Safety and anti-inflammatory activity of curcumin: a component of tumeric (Curcuma longa). J Altern Complement Med. 2003;9:161–8.

    Article  PubMed  Google Scholar 

  49. Trujillo J, Chirino YI, Molina-Jijon E, Anderica-Romero AC, Tapia E, Pedraza-Chaverri J. Renoprotective effect of the antioxidant curcumin: Recent findings. Redox Biol. 2013;1:448–56.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Derochette S, Franck T, Mouithys-Mickalad A, Deby-Dupont G, Neven P, Serteyn D. Intra- and extracellular antioxidant capacities of the new water-soluble form of curcumin (NDS27) on stimulated neutrophils and HL-60 cells. Chem Biol Interact. 2013;201:49–57.

    Article  CAS  PubMed  Google Scholar 

  51. Xiao X, Yang M, Sun D, Sun S. Curcumin protects against sepsis-induced acute lung injury in rats. J Surg Res. 2012;176:e31–9.

    Article  CAS  PubMed  Google Scholar 

  52. Vaughan RA, Garcia-Smith R, Dorsey J, Griffith JK, Bisoffi M, Trujillo KA. Tumor necrosis factor alpha induces Warburg-like metabolism and is reversed by anti-inflammatory curcumin in breast epithelial cells. Int J Cancer. 2013;133:2504–10.

    Article  CAS  PubMed  Google Scholar 

  53. Kloesch B, Becker T, Dietersdorfer E, Kiener H, Steiner G. Anti-inflammatory and apoptotic effects of the polyphenol curcumin on human fibroblast-like synoviocytes. Int Immunopharmacol. 2013;15:400–5.

    Article  CAS  PubMed  Google Scholar 

  54. Klawitter M, Quero L, Klasen J, Gloess AN, Klopprogge B, Hausmann O, et al. Curcuma DMSO extracts and curcumin exhibit an anti-inflammatory and anti-catabolic effect on human intervertebral disc cells, possibly by influencing TLR2 expression and JNK activity. J Inflamm (Lond). 2012;9:29.

    Article  CAS  Google Scholar 

  55. Henrotin Y, Clutterbuck AL, Allaway D, Lodwig EM, Harris P, Mathy-Hartert M, et al. Biological actions of curcumin on articular chondrocytes. Osteoarthritis Cartilage. 2010;18:141–9.

    Article  CAS  PubMed  Google Scholar 

  56. Shanmugam MK, Rane G, Kanchi MM, Arfuso F, Chinnathambi A, Zayed ME, et al. The Multifaceted Role of Curcumin in Cancer Prevention and Treatment. Molecules. 2015;20:2728–69.

    Article  PubMed  Google Scholar 

  57. Shakibaei M, Mobasheri A, Lueders C, Busch F, Shayan P, Goel A. Curcumin enhances the effect of chemotherapy against colorectal cancer cells by inhibition of NF-kappaB and Src protein kinase signaling pathways. PLoS One. 2013;8, e57218.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  58. Um MY, Hwang KH, Ahn J, Ha TY. Curcumin attenuates diet-induced hepatic steatosis by activating AMP-activated protein kinase. Basic Clin Pharmacol Toxicol. 2013;113:152–7.

    Article  CAS  PubMed  Google Scholar 

  59. Mei X, Xu D, Xu S, Zheng Y, Xu S. Novel role of Zn(II)-curcumin in enhancing cell proliferation and adjusting proinflammatory cytokine-mediated oxidative damage of ethanol-induced acute gastric ulcers. Chem Biol Interact. 2012;197:31–9.

    Article  CAS  PubMed  Google Scholar 

  60. Kanitkar M, Gokhale K, Galande S, Bhonde RR. Novel role of curcumin in the prevention of cytokine-induced islet death in vitro and diabetogenesis in vivo. Br J Pharmacol. 2008;155:702–13.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Jain SK, Rains J, Croad J, Larson B, Jones K. Curcumin supplementation lowers TNF-alpha, IL-6, IL-8, and MCP-1 secretion in high glucose-treated cultured monocytes and blood levels of TNF-alpha, IL-6, MCP-1, glucose, and glycosylated hemoglobin in diabetic rats. Antioxid Redox Signal. 2009;11:241–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  62. Cao J, Liu Y, Jia L, Zhou HM, Kong Y, Yang G, et al. Curcumin induces apoptosis through mitochondrial hyperpolarization and mtDNA damage in human hepatoma G2 cells. Free Radic Biol Med. 2007;43:968–75.

    Article  CAS  PubMed  Google Scholar 

  63. Becatti M, Prignano F, Fiorillo C, Pescitelli L, Nassi P, Lotti T, et al. The involvement of Smac/DIABLO, p53, NF-kB, and MAPK pathways in apoptosis of keratinocytes from perilesional vitiligo skin: Protective effects of curcumin and capsaicin. Antioxid Redox Signal. 2010;13:1309–21.

    Article  CAS  PubMed  Google Scholar 

  64. Kim YS, Ahn Y, Hong MH, Joo SY, Kim KH, Sohn IS, et al. Curcumin attenuates inflammatory responses of TNF-alpha-stimulated human endothelial cells. J Cardiovasc Pharmacol. 2007;50:41–9.

    Article  CAS  PubMed  Google Scholar 

  65. Bharti AC, Donato N, Singh S, Aggarwal BB. Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor-kappa B and IkappaBalpha kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis. Blood. 2003;101:1053–62.

    Article  CAS  PubMed  Google Scholar 

  66. Jobin C, Bradham CA, Russo MP, Juma B, Narula AS, Brenner DA, et al. Curcumin blocks cytokine-mediated NF-kappa B activation and proinflammatory gene expression by inhibiting inhibitory factor I-kappa B kinase activity. J Immunol. 1999;163:3474–83.

    CAS  PubMed  Google Scholar 

  67. Wilmanski J, Siddiqi M, Deitch EA, Spolarics Z. Augmented IL-10 production and redox-dependent signaling pathways in glucose-6-phosphate dehydrogenase-deficient mouse peritoneal macrophages. J Leukoc Biol. 2005;78:85–94.

    Article  CAS  PubMed  Google Scholar 

  68. Wilmanski J, Villanueva E, Deitch EA, Spolarics Z. Glucose-6-phosphate dehydrogenase deficiency and the inflammatory response to endotoxin and polymicrobial sepsis. Crit Care Med. 2007;35:510–18.

    Article  CAS  PubMed  Google Scholar 

  69. Spolarics Z, Siddiqi M, Siegel JH, Garcia ZC, Stein DS, Ong H, et al. Increased incidence of sepsis and altered monocyte functions in severely injured type A- glucose-6-phosphate dehydrogenase-deficient African American trauma patients. Crit Care Med. 2001;29:728–36.

    Article  CAS  PubMed  Google Scholar 

  70. Abu-Osba YK, Mallouh AA, Hann RW. Incidence and causes of sepsis in glucose-6-phosphate dehydrogenase-deficient newborn infants. J Pediatr. 1989;114:748–52.

    Article  CAS  PubMed  Google Scholar 

  71. Upperman JS, Pillage G, Siddiqi MQ, Zeevi A, Kelly N, Ford HR, et al. Dominance of high-producing interleukin 6 and low-producing interleukin 10 and interferon gamma alleles in glucose-6-phosphate dehydrogenase-deficient trauma patients. Shock. 2005;23:197–201.

    CAS  PubMed  Google Scholar 

  72. Liese AM, Siddiqi MQ, Siegel JH, Deitch EA, Spolarics Z. Attenuated monocyte IL-10 production in glucose-6-phosphate dehydrogenase-deficient trauma patients. Shock. 2002;18:18–23.

    Article  PubMed  Google Scholar 

  73. Liao SL, Lai SH, Tsai MH, Weng YH. Cytokine Responses of TNF-alpha, IL-6, and IL-10 in G6PD-Deficient Infants. Pediatr Hematol Oncol. 2014;31:87–94.

    Article  CAS  PubMed  Google Scholar 

  74. Timens W, Boes A, Rozeboom-Uiterwijk T, Poppema S. Immaturity of the human splenic marginal zone in infancy. Possible contribution to the deficient infant immune response. J Immunol. 1989;143:3200–6.

    CAS  PubMed  Google Scholar 

  75. Saito F, Kuwata H, Oiki E, Koike M, Uchiyama Y, Honda K, et al. Inefficient phagosome maturation in infant macrophages. Biochem Biophys Res Commun. 2008;375:113–18.

    Article  CAS  PubMed  Google Scholar 

  76. Mohrenweiser HW, Fielek S, Wurzinger KH. Characteristics of enzymes of erythrocytes from newborn infants and adults: activity, thermostability, and electrophoretic profile as a function of cell age. Am J Hematol. 1981;11:125–36.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was carried out with financial support from Ministry of Science and Technology of Taiwan (NSC100-2320-B-182-010-MY3 to DTYC), from Ministry of Education of Taiwan (EMRPD1D0241, EMRPD1D0871 to Chang Gung University), and from Chang Gung Memorial Hospital (CMRPD1B0331, CMRPD1B0332 to Healthy Aging Research Center; CMRPD190423, CMRPD1C0771 to DTYC; CMRPD1C0441, CMRPD1C0761 to MLC).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel Tsun-Yee Chiu.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HCY designed the experiments, performed SiRNA knockdown, qRT-PCR, analyzed data and wrote the 1st draft of the paper. MLC designed the experiments, analyzed data and participated in the writing of the paper. YSH carried out the cell culture, immunoassays and analyzed data. YHW analyzed data and participated in the writing of the paper. HRL carried out the immunoassays and analyzed data. HYL assisted in cell culture and experiment. HYH provided advices for experimental design and prepared G6PD-deficient cells. DTYC obtained funding for this project, conceived and designed the experiments as well as participated in the writing of the paper. All authors read and approved the final manuscript.

Hung-Chi Yang and Mei-Ling Cheng contributed equally to this work.

Additional files

Additional file 1:

Supplementary methods, reference and figure legends.

Additional file 2: Figure S1.

G6PD knockdown reduced G6PD activity and expression in HepG2 cells. (a) The G6PD activity of G6PD-scramble (Sc) and G6PD-knockdown (Gi) HepG2 cells were determined by enzymatic assay. The unit was expressed as U/mg of protein lysate. These results were representative of at least three separate experiments. *indicates a significant difference (P<0.05) between Sc and Gi HepG2 cells. (b) G6PD protein expression of Sc and Gi HepG2 cells were detected by Western blotting, the amount of G6PD protein was normalized to Actin in the respective sample. The blot shown was a representative of three separate experiments.

Additional file 3: Figure S2.

The morphology of palmitate-treated HepG2 cells. The morphology of control and palmitate-treated (0.3 mM) Sc and Gi HepG2 cells were visualized by Sudan Red (orange) and hematoxylin staining (blue).

Additional file 4: Figure S3.

The effect of palmitate treatment on lipid accumulation in HepG2 cells. The lipid levels of control and palmitate-treated (0.3 mM) Sc and Gi HepG2 cells were quantified by flow cytometry after Nile Red staining. These results were representative of at least three separate experiments. # indicates significant difference (P<0.05) between control and palmitate treatment.

Additional file 5: Figure S4.

Cytokine profile of Sc and Gi HepG2 cells with or without 0.3 mM of palmitate treatment for 24 hr. The result was a representative of two separate experiments. The normalization of relative cytokine level was described in the method section. The quantification result was shown in Table 2.

Additional file 6: Table S1.

Format of cytokine array.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, HC., Cheng, ML., Hua, YS. et al. Glucose 6-phosphate dehydrogenase knockdown enhances IL-8 expression in HepG2 cells via oxidative stress and NF-κB signaling pathway. J Inflamm 12, 34 (2015). https://doi.org/10.1186/s12950-015-0078-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12950-015-0078-z

Keywords