Curcumin upregulates Nrf2 nuclear translocation and protects rat hepatic stellate cells against oxidative stress

  • Authors:
    • Zhenxiong Liu
    • Weijia Dou
    • Yuanyuan Zheng
    • Qinsheng Wen
    • Ming Qin
    • Xuxia Wang
    • Hua Tang
    • Rong Zhang
    • Dandan Lv
    • Jingjie Wang
    • Shuguang Zhao
  • View Affiliations

  • Published online on: December 17, 2015     https://doi.org/10.3892/mmr.2015.4690
  • Pages: 1717-1724
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to investigate the protective role of curcumin against oxidative stress in rat hepatic stellate cells (HSCs)-T6, and to determine the possible underlying mechanisms. HSC‑T6 cells were divided into three groups: Negative control group, oxidant‑treated group and curcumin‑treated group. Flow cytometry and spectrophotometry were used to measure the production of reactive oxygen species (ROS), and the levels of malondialdehyde (MDA) and glutathione (GSH). Immunocytochemistry and a radioimmunoassay were used to determine the expression of smooth muscle α‑actin (α‑SMA) and the secretion of extracellular matrix (ECM) molecules. In addition, western blotting and immunocytochemistry were used to determine the expression levels of nuclear factor‑erythroid 2‑related factor (Nrf2). Treatment with glucose oxidase (GO) significantly stimulated the formation of ROS and increased the production of MDA, as compared with the control cells; however, the production of GSH was only slightly increased. In addition, treatment with GO significantly promoted the expression of α‑SMA and the secretion of ECM molecules. Conversely, treatment with curcumin significantly decreased the levels of ROS and MDA, and significantly increased the levels of GSH. Curcumin significantly inhibited the expression of α‑SMA and decreased the secretion of ECM molecules. Furthermore, treatment with curcumin significantly increased the nuclear expression levels of Nrf2. These results indicated that curcumin may protect rat HSCs against oxidative stress and inhibit the GO‑induced activation and secretion of ECM molecules in vitro. These effects were mediated by the upregulation of Nrf2 nuclear translocation.

Introduction

Previous studies have demonstrated that oxidative stress has a significant role in the occurrence and progression of hepatitis and hepatic fibrosis (14). Oxidative stress results from the excessive production of reactive oxygen species (ROS), and the inability of an organism to eliminate them. Excessive ROS lead to lipid peroxidation, protein and DNA damage, and injury to cellular structure and function (5). Numerous studies have reported the important role of ROS in various types of hepatic injury (610). In addition to ROS-induced inflammation, ROS may lead to loss of normal regulatory functions, resulting in tissue injury and excessive repair, and the development of hepatitis and hepatic fibrosis (11,12). It has previously been demonstrated that activation of hepatic stellate cells (HSCs) has a key role in the progression of hepatic fibrosis; therefore, HSCs are considered important target cells in hepatic fibrosis research (13). Furthermore, HSCs can be activated by oxidative stress and transformed into myofibroblasts. Myofibroblasts synthesize abundant extracellular matrix (ECM) molecules, which may lead to hepatic fibrosis (14); therefore, how to inhibit the activation of HSCs exposed to oxidative stress requires further investigation. Nuclear factor-erythroid 2-related factor (Nrf2) is a transcription factor that activates numerous antioxidant enzymes and phase II detoxifying enzymes (15). In addition, Nrf2 has an important role regulating oxidative stress (16,17); however, the majority of studies regarding Nrf2 have focused on the nervous and respiratory systems. The effects of Nrf2 on HSCs, and the underlying molecular mechanisms, have seldom been reported.

The present study hypothesized that upregulation of Nrf2 nuclear translocation would promote the expression of antioxidant enzymes and phase II detoxifying enzymes, thus protecting the liver against injury. The present study investigated the alterations and regulatory mechanisms of the Nrf2 pathway, which is of great significance for understanding the pathogenesis of hepatic fibrosis and developing novel preventative strategies and curative therapies.

Curcumin, which is an ingredient of the spice turmeric, is present in the rhizomes of Curcuma longa Linn (Zingiberaceae). Curcumin has been reported to exert antioxidant, anti-inflammatory, anticancer and hepatoprotective effects (18). Furthermore, curcumin functions as an exogenous Nrf2 agonist, and can promote the nuclear translocation and biological effects of Nrf2 (19). Therefore, the present study used curcumin to upregulate Nrf2, and subsequently investigated the effects of Nrf2 on HSCs.

To investigate the possible regulatory mechanisms that underlie the Nrf2 pathway, the present study examined the effects of curcumin on Nrf2. In addition, alterations in the levels of ROS, malondialdehyde (MDA) and glutathione (GSH) were detected. As an index of HSC activation, smooth muscle α-actin (α-SMA) and desmin levels were measured. Furthermore, ECM-secreted proteins, including type III procollagen (PCIII), type IV collagen (CIV), laminin (LN) and hyaluronic acid (HA), were measured following treatment of HSCs with curcumin. The results of the present study indicated that curcumin was able to protect HSCs against oxidative stress, and inhibit the activation of HSCs via induction of Nrf2 nuclear translocation.

Materials and methods

Materials

Curcumin and GO were purchased from Sigma-Aldrich (St. Louis, MO, USA). Anti-α-SMA (1:200; cat. no. sc-53142) and anti-β-actin (1:500; cat. no. sc-47778) antibodies were obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Anti-Nrf2 (1:500; cat. no. BS6286) and anti-desmin (1:500; cat. no. BS1712) antibodies were purchased from Bioworld Technology, Inc. (St. Louis Park, MN, USA). Horseradish peroxidase-conjugated anti-rabbit (1:10,000; cat. no. bs-0295G-HRP) and anti-mouse (1:10,000; cat. no. bs-0296G-HRP) immunoglobulin (Ig)G, and fluorescein isothiocyanate (FITC)-conjugated anti-rabbit (1:1,000; cat. no. bs-0295G-FITC) and anti-mouse (1:1,000; cat. no. bs-0296G-FITC) IgG secondary antibodies were obtained from Beijing Biosynthesis Biotechnology Co., Ltd. (Beijing, China). Dihydroethidium (DHE) was purchased from Beyotime Institute of Biotechnology (Jiangsu, China). MDA and GSH kits were obtained from Nanjing Jiancheng Biotechnology, Inc. (Nanjing, China). PCIII, CIV, LN and HA kits were purchased from the Shanghai Naval Medical Institute (Shanghai, China).

Cell culture

The HSC-T6 immortalized rat HSC line exhibits a stable phenotype and biochemical characteristics (20). HSC-T6 cells were a generous gift from Dr Ding of the Medical College of Xi'an Jiaotong University (Xi'an, China). The cells were grown under standard conditions in a normoxic atmosphere in high-glucose Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Sijiqing, Hangzhou, China) in a humidified incubator containing 5% CO2 at 37°C. All subsequent experiments were conducted using cells at the exponential stage of growth. Cells were seeded into a 25 cm2 plastic culture flask at a density of 1×106 cells. The cells were separated into three groups: The negative control cells were incubated with 5 ml culture medium; the oxidant-treated cells were incubated with 5 ml culture medium supplemented with 100 mU/ml GO for 2 h before each experimental manipulation; and the curcumin-treated cells were pre-treated with 5 ml culture medium containing 0.15 µmol curcumin for 3 h, and then incubated in the same manner as the oxidant-treated cells.

Western blot analysis

Total, cytoplasmic and nuclear proteins were obtained from the cells using protein extraction kits (Beyotime Institute of Biotechnology, Shanghai, China), according to the manufacturer's protocol. The concentration of the protein samples was quantified using a Bradford Protein Assay kit (Beyotime Institute of Biotechnology), according to the manufacturer's protocol. Subsequently, the protein samples (15 µg) were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and were transferred to nitrocellulose membranes (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The membranes were blocked in buffer containing skim milk for 2 h, and were then incubated with anti-Nrf2 primary antibody at 4°C overnight, and washed with phosphate-buffered saline (PBS). The membranes were then incubated with a horseradish peroxidase-conjugated secondary antibody at room temperature for 1 h. Pierce Enhanced Chemiluminescence Western Blotting substrate (Thermo Fisher Scientific, Inc.) was used to develop the blots, and immunore-activity was visualized following exposure to X-OMAT BT Film (Beyotime Institute of Biotechnology). β-actin was used as an internal control. The results were quantified and normalized to β-actin using Molecular Analyst software (version 1.4.1; Bio-Rad Laboratories, Inc.)

Immunocytochemistry

Following a 24 h cell culture, in which the cells became adherent, immunocytochemistry was conducted. Cell-coated dishes (1×105 cells/ml) were fixed with 4% paraformaldehyde for 30 min and washed with PBS. The cells were permeabilized using 0.3% Triton X-100 (Sigma-Aldrich) for 15 min. Endogenous peroxidases and biotins were quenched using 3% H2O2. The cells were then blocked with FBS and incubated with the indicated primary antibodies at 4°C overnight. Sections stained for Nrf2 protein expression were washed and incubated with a horseradish peroxidase-conjugated secondary antibody at room temperature for 30 min. Bound secondary antibodies were detected using Histostain-SP kits (ZSbio, Beijing, China), according to the manufacturer's protocol. The reaction products were visualized using diaminobenzidine tetrahydro-chloride (Tiangen Biotech Co., Ltd., Beijing, China). Sections stained for α-SMA and desmin protein expression were incubated with FITC-conjugated secondary antibodies at room temperature for 30 min in the dark. The nuclei were stained using Evans Blue (Sigma-Aldrich) in sections stained for α-SMA. In the negative control group, primary antibodies were substituted with PBS. Stained sections were viewed under a Nikon Eclipse 800 fluorescent microscope (Nikon Corporation, Tokyo, Japan).

Flow cytometry

HSC-T6 cells were maintained under standard conditions, and were then transferred to culture dishes. Following a 24 h culture, 1×106 cells from each group were placed in tubes. The cells were washed with PBS and separated by centrifugation at 432 x g for 10 min at 4°C. Serum-free media supplemented with 2-µM DHE were added, and the tubes were incubated at 4°C for 30 min in the dark. The remaining cells were washed with PBS and subjected to further centrifugation at 432 x g for 10 min at 4°C. Paraformaldehyde (2 ml; 4%) was added to each tube and the tubes were incubated at 4°C for 30 min in the dark, followed by centrifugation at 432 x g for 10 min at 4°C. Subsequently, 300 µl PBS was added to the preparations and mixed gently. DHE fluorescence was measured by flow cytometry (FACSCanto II; BD Biosciences, Franklin Lakes, NJ, USA).

MDA and GSH assays

HSC-T6s were incubated in culture dishes and the supernatants were collected for the detection of MDA and GSH by spectrophotometry. Measurement of the product (MDA-TBA adduct) of a reaction between MDA and 2-thiobarbituric acid, and of the product (2-nitro-5-sulphur benzoic acid) of a reaction between GSH and dithiodinitrobenzoic acid allow the levels of MDA and GSH to be analyzed using colorimetric assays. MDA and GSH levels were determined using kits, according to the manufacturers' protocols. Absorbance was measured at 532 and 412 nm using a spectrophotometer (UV-2450; Shimadzu Corporation, Kyoto, Japan) for MDA and GSH, respectively. The concentrations of MDA and GSH were calculated according to the equation provided in the kits.

Analysis of ECM secretion

Cells were initally separated into three groups and cultured in serum-free medium overnight. Subsequently, the cells were treated as mentioned previously (negative control, oxidant-treated and curcumin-treated cells). Following centrifugation at 1,000 x g for 20 min at 4°C, the supernatant was collected and maintained at −80°C until further analysis. The levels of PCIII, CIV, LN and HA secreted into the supernatant were analyzed using commercially available radio-immunoassay kits, according to the manufacturers' protocols.

Statistical analysis

The data are presented as the mean ± standard deviation and significance was assessed using SPSS 12.0 software (SPSS, Inc., Chicago, IL, USA). Statistical comparisons were performed using one-way analysis of variance. Paired comparisons were conducted using Student Newman Keuls-q test. P<0.05 was considered to indicate a statistically significant difference.

Results

Curcumin protects HSCs against GO-induced oxidative stress injury

As a marker of oxidative stress, GO may react with glucose in the culture media and subsequently promote the generation of glucuronic acid and hydrogen peroxide (H2O2). It is well known that DHE, which is a superoxide radical-specific fluorescent probe, is able to enter viable cells freely where it is oxidized by superoxide to form ethidium, which binds to DNA and exhibits red fluorescence. Therefore, the present study detected fluorescence intensity in order to estimate the relative levels of ROS production. As shown in Fig. 1A, compared with the negative control cells, treatment of the cells with GO significantly increased fluorescence intensity, whereas pretreatment with curcumin significantly decreased fluorescence intensity. However, the fluorescence intensity in the curcumin-treated cells was stronger, as compared with in the negative control cells.

Furthermore, ROS may react with lipids in cell and mitochondrial membranes, resulting in the production of MDA, which can be used to measure lipid peroxidation. Due to its strong cytotoxicity, MDA is able to alter membrane permeability or disrupt membrane integrity, leading to oxidative injury. However, even in the case of increased lipid peroxidation, oxidative stress only occurs in cells that are unable to defend and protect against free radical injury or chemically induced damage. GSH is an important endogenous antioxidant, which reduces levels of ROS. GSH acts as a free radical scavenger, a coenzyme for various antioxidant enzymes, a regulator of thioldisulfide status, and is involved in the detoxification of electrophilic xenobiotics via conjugation. Therefore, the present study aimed to detect the levels of MDA and GSH in the supernatant. Significantly enhanced levels of MDA were detected in the oxidant-treated cells, as compared with the negative control cells, which were correlated with the levels of ROS. The MDA levels were markedly attenuated in response to curcumin pretreatment, however they remained higher than in the control cells. Furthermore, compared with the negative control cells, the levels of GSH were slightly increased in the oxidant-treated cells, whereas they were significantly elevated in the curcumin-treated cells (Fig. 1B).

These results suggest that an oxidative stress model of HSC-T6 was established with GO treatment, as demonstrated in the increased levels of ROS and MDA. Furthermore, pretreatment with curcumin was able to significantly suppress the degree of oxidative stress, at least partially due to the induced expression of endogenous GSH.

Curcumin promotes the nuclear translocation of Nrf2

GSH synthesis is governed by Nrf2 via the regulation of the rate-limiting enzymes glutamate cysteine ligase (GCL) catalytic subunit (GCLC) and GCL modifier subunit (GCLM). To elucidate the molecular mechanism by which curcumin protects HSCs against oxidative stress injury, the present study detected the expression of Nrf2 in HSC-T6 cells by immunocytochemistry (Fig. 2A) and western blot analysis (Fig. 2B and C). As expected, the expression of total Nrf2 did not differ between the groups; however, the expression levels of cytoplasmic Nrf2 were slightly decreased in the oxidant-treated cells, and significantly decreased in the curcumin-treated cells, as compared with the negative control cells. Conversely, the expression of nuclear Nrf2 was absent in the negative control cells and was only slightly expressed in the oxidant-treated cells; however, the expression levels of nuclear Nrf2 were markedly increased in the curcumin-treated cells, as compared with in the oxidant-treated cells. Immunocytochemistry detected similar results to the western blot analysis; only minimal positive brown staining was detected in the nuclei of the oxidant-treated cells, as compared with the control cells, in which no positive brown nuclear staining was detected. However, in the curcumin-treated cells, the amount of positive brown staining was abundant in the nucleus, as compared with in the oxidant-treated cells.

These results indicate that oxidative stress may activate the Nrf2 regulatory pathway, and pretreatment with curcumin could protect HSCs against oxidative stress via promoting the translocation of Nrf2 from the cytoplasm to the nucleus.

Curcumin blocks GO-induced α-SMA expression and HSC activation

Desmin, which is a type of cytoskeletal intermediate filament, has been widely used as a marker for distinguishing HSCs. Desmin was expressed in all cell groups, as detected using immunofluorescence (Fig. 3A), and there were no differences in desmin expression between the groups, as determined by western blot analysis (Fig. 3B). α-SMA is produced by activated HSCs, and is a characteristic signal of HSC activation. As shown in Fig. 3A, no fluorescence expression of α-SMA was detected in the negative control cells, whereas following treatment with GO for 2 h abundant fluorescence expression of α-SMA was observed. Furthermore, the fluorescence expression of α-SMA in the cells pretreated with curcumin was markedly decreased, as compared with the oxidant-treated cells. Consistent with the alterations in immunofluorescence, the protein expression levels of α-SMA were significantly increased in the oxidant-treated cells, as compared with the negative control cells, whereas the expression levels of α-SMA were significantly decreased in the curcumin-treated cells, as compared with the oxidant-treated cells. However, the expression levels remained higher in the curcumin-treated cells, as compared with in the negative control cells (Fig. 3B and C).

These results suggest that GO-induced oxidative stress may enhance α-SMA expression in HSCs, and the transformation of HSCs to myofibroblast-like cells. Furthermore, treatment with curcumin may activate the Nrf2 regulatory pathway and subsequently suppress α-SMA expression and HSC activation, which may be associated with its antifibrotic effects.

Curcumin inhibits the expression of ECM molecules in GO-treated HSCs

Serum levels of PCIII, CIV, LN and HA are often considered indices of liver fibrosis; therefore, the present study aimed to detect the levels of PCIII, CIV, LN and HA in the supernatant (Fig. 4A–D). Compared with the negative control cells, HSCs treated with GO for 2 h exhibited significantly increased levels of ECM molecules. Conversely, the levels of ECM molecules were markedly reduced following pretreatment with curcumin; however, the levels remained higher, as compared with in the negative control cells.

These results indicate that treatment with GO may markedly increase the expression of ECM molecules in HSCs. Furthermore, the activation of Nrf2 by curcumin may exert inhibitory effects on the expression of ECM molecules in HSC, which may be associated with its antifibrogenic effects.

Discussion

The Nrf2 pathway is regarded as the most important pathway with regards to cellular protection against oxidative stress (21). As a pivotal modulator of the response to oxidative stress, the activation of Nrf2 induces the expression of various protective antioxidant genes (22,23). Under normal conditions, Nrf2 is sequestered in the cytoplasm where it is bound to the cytoskeleton-associated protein Keap1 (24). Oxidative stress promotes the dissociation of the Nrf2-Keap1 complex, thus resulting in Nrf2 stabilization and translocation to the nucleus (25). In the nucleus, Nrf2 associates with dimerization partners and binds antioxidant-response element sequences, in order to induce the expression of various detoxification and antioxidant enzyme genes that contribute to the protective response (26).

Curcumin is a natural polyphenol product that is derived from the rhizome of Curcuma longa. Numerous studies have detected the various bioactivities of curcumin, including antioxidant, anti-inflammatory, cell apoptosis-inducing and cell proliferation-inhibiting activities (2730). In addition, it has been suggested that curcumin may be used clinically against numerous types of cancer, inflammatory bowel disease, irritable bowel syndrome (IBS), rheumatoid arthritis and atherosclerosis (31). Cheng et al (32) detected the effects of curcumin on 25 patients with various types of high-risk or premalignant lesions; following 3 months of treatment with curcumin, some patients exhibited histological improvement in premalignant lesions. Dhillon et al (33) studied the efficacy of curcumin on patients with advanced pancreatic cancer, and demonstrated that oral curcumin administration exerted biological activity in some patients. Hanai et al (34) conducted a double-blind, placebo-controlled trial in 89 patients with ulcerative colitis (UC); curcumin was shown to reduce the incidence of UC and may be considered a promising treatment for UC. In addition, a blind pilot study demonstrated that abdominal pain and the discomfort score of patients with IBS were significantly reduced following treatment with curcumin (35).

Previous studies have detected various functions of curcumin. Notably, curcumin has been shown to function as an exogenous agonist of Nrf2 (19). A previous study suggested that curcumin may alter the conformation of Keap1, and promote the dissociation of Nrf2 from Keap1 and its subsequent nuclear translocation (36). The present study evaluated the effects of curcumin on Nrf2 regulation in HSCs; Nrf2 was localized to the cytoplasm under normal conditions and little Nrf2 was localized to the nucleus following the induction of oxidative stress. However, pretreatment with curcumin induced a substantial localization of Nrf2 to the nucleus in HSCs.

Increased levels of GSH are an index of Nrf2 activation, and increased GSH may be considered a major factor underlying the protection associated with Nrf2 activation (37,38). GSH not only produces reducing equivalents, which are necessary for the conversion of H2O2 and lipid peroxides to water and lipid alcohols (39), but also has an important role in the protection of protein sulfhydration against oxidation (40). The rate-limiting reaction in GSH biosynthesis is catalyzed by GCL, which comprises two subunits: GCLC and GCLM. Nrf2 is able to increase the expression of GCLC and GCLM (41); therefore, the preferential activation of Nrf2 leads to more efficient GSH biosynthesis and improved antioxidant status (42). In the present study, the levels of GSH were increased alongside increasing Nrf2 nuclear translocation following curcumin treatment. GSH elevates the antioxidant ability of cells against oxidative stress.

The pathogenesis of hepatic fibrosis has yet to be completely clarified; however, it is generally accepted that the activation of HSCs is central to the process (43). In the prophase of liver fibrosis, quiescent HSCs transform into myofibroblasts, which are characterized by the assembly of α-SMA stress fibers, loss of cytosolic retinol and increased proliferation (44). This activation subsequently results in the synthesis of cytokines and the accumulation of ECM molecules (45). Considerable attention has been focused on elucidating the mechanistic triggers of HSC myofibroblast transformation. In addition, it is hypothesized that oxidative stress may contribute to HSC activation (46).

In the present study, GO reacted with glucose in the culture media resulting in the generation of glucuronic acid and H2O2. These increased levels of ROS may stimulate HSCs to undergo oxidative stress (47). The results of the present study demonstrated that following treatment with GO, the levels of ROS in the oxidant-treated cells were significantly increased, as compared with in the negative control cells. The negative control cells were quiescent, whereas the oxidant-treated cells were activated by oxidative stress. In addition to the upregulation of Nrf2 nuclear translocation in the curcumin-treated cells, the levels of ROS and HSC activation were decreased, as compared with in the oxidant-treated cells.

In-depth research regarding the mechanisms underlying liver fibrosis has revealed the role of free radicals and membrane lipid peroxidation in the process of liver injury (48). Oxygen radicals attack unsaturated fatty acids in cellular membranes and initiate lipid peroxidation. Lipid peroxidation leads to alterations in the permeability of cellular membranes, subsequently aggravating cell dysfunction and promoting the secretion of ECM molecules (49,50). Not only does oxidative stress stimulate HSC proliferation and collagen synthesis, it also further damages cells by promoting lipid peroxidation. MDA and other peroxidation products increase collagen synthesis via HSC activation, and stimulate Kupffer cells to release cytokines that promote fibrosis (51). Lipid peroxidation has an important role in regulating collagen gene expression, and is associated with cell injury and fibrosis. ROS and lipid peroxidation have been implicated as profibrogenic mediators (52), whereas Nrf2 is effective at suppressing cell damage resulting from lipid peroxidation (53). In the present study, the levels of MDA were increased alongside ROS levels in the oxidant-treated cells; however, following pretreatment with curcumin, the levels of MDA were reduced, as compared with in the oxidant-treated cells.

Liver fibrosis is the excessive deposition of ECM molecules following liver injury. ECM accumulation is associated with increased collagen synthesis and decreased matrix degradation, contributing to liver fibrosis and remodeling (54). Active HSCs are the primary source of the excessive production of ECM components (55), and the ECM enhanced density leads to increased matrix stiffness, which is a significant stimulus for the activation of HSCs (56). In the present study, oxidative stress induced HSC activation, which was followed by the enhanced synthesis of ECM components. Following curcumin pretreatment, the activation of HSCs was inhibited, and the secretion of ECM components was suppressed.

In conclusion, the present study demonstrated that curcumin-induced Nrf2 activation may protect HSCs against oxidative stress-induced injury, and this effect was characterized by enhanced Nrf2 nuclear translocation and antioxidant capacity. The underlying mechanism remains to be elucidated; however, the present study proposes a broader application for Nrf2 in the prevention and treatment of hepatic damage.

Acknowledgments

The present study was supported by the National Natural Science Foundation of China (grant nos. 30800515, 81270485 and 81170376) and the Natural Science Foundation of Shaanxi Province (grant no. 2013JM4021).

Abbreviations:

Nrf2

nuclear factor-erythroid 2-related factor

HSC

hepatic stellate cell

GO

glucose oxidase

ROS

reactive oxygen species

MDA

malondialdehyde

GSH

glutathione

α-SMA

smooth muscle α-actin

ECM

extracellular matrix

PCIII

type III procollagen

C IV

type IV collagen

LN

laminin

HA

hyaluronic acid

DHE

dihydroethidium

References

1 

Parola M and Pinzani M: Hepatic wound repair. Fibrogenesis Tissue Repair. 2:42009. View Article : Google Scholar : PubMed/NCBI

2 

Loguercio C and Federico A: Oxidative stress in viral and alcoholic hepatitis. Free Radic Biol Med. 34:1–10. 2003. View Article : Google Scholar

3 

Lin X, Zhang S, Huang R, Wei L, Tan S, Liang S, Tian Y, Wu X, Lu Z and Huang Q: Helenalin attenuates alcohol-induced hepatic fibrosis by enhancing ethanol metabolism, inhibiting oxidative stress and suppressing HSC activation. Fitoterapia. 95:203–213. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Lee BH, Hsu WH, Hsu YW and Pan TM: Suppression of dimerumic acid on hepatic fibrosis caused from carboxymethyllysine (CML) by attenuating oxidative stress depends on Nrf2 activation in hepatic stellate cells (HSCs). Food Chem Toxicol. 62:413–419. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Bisbal C, Lambert K and Avignion A: Antioxidants and glucose metabolism disorders. Curr Opin Clin Nutr Metab Care. 13:439–446. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Anoush M, Eghbal MA, Fathiazad F, Hamzeiy H and Kouzehkonani NS: The protective effects of garlic extract against acetaminophen-induced oxidative stress and glutathione depletion. Park J Biol Sci. 12:765–771. 2009. View Article : Google Scholar

7 

Sun Q, Long Z, Wu H, Liu Y, Wang L, Zhang X, Wang X and Hai C: Effect of alcohol on diethylnitrosamine-induced hepatic toxicity: Critical role of ROS, lipid accumulation, and mitochondrial dysfunction. Exp Toxicol Pathol. 67:491–498. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Takemoto K, Hatano E, Iwaisako K, Takeiri M, Noma N, Ohmae S, Toriguchi K, Tanabe K, Tanaka H, Seo S, et al: Necrostatin-1 protects against reactive oxygen species (ROS)-induced hepatotoxicity in acetaminophen-induced acute liver failure. FEBS Open Bio. 4:777–787. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Liu J, Wang X, Liu R, Liu Y, Zhang T, Fu H and Hai C: Oleanolic acid co-administration alleviates ethanol-induced hepatic injury via Nrf-2 and ethanol-metabolizing modulating in rats. Chem Biol Interact. 221:88–98. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Browning JD and Horton JD: Molecular mediators of hepatic steatosis and liver injury. J Clin Invest. 114:147–152. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Singh S, Vrishni S, Singh BK, Rahman I and Kakkar P: Nrf2-ARE stress response mechanism: A control point in oxidative stress-mediated dysfunctions and chronic inflammatory diseases. Free Radic Rec. 44:1267–1288. 2010. View Article : Google Scholar

12 

Matsunami T, Sato Y, Ariga S, Sato T, Kashimura H, Haseqawa Y and Yukawa M: Regulation of oxidative stress and inflammation by hepatic adiponectin receptor 2 in an animal model of nonalcoholic steatohepatitis. Int J Clin Exp Pathol. 3:472–481. 2010.PubMed/NCBI

13 

De Minicis S, Candelaresi C, Agostinelli L, Taffetani S, Saccomanno S, Rychlicki C, Trozzi L, Marzioni M, Benedetti A and Svegliati-Baroni G: Endoplasmic Reticulum stress induces hepatic stellate cell apoptosis and contributes to fibrosis resolution. Liver Int. 32:1574–1584. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Tacke F and Weiskirchen R: Liver fibrosis - pathogenesis and novel therapeutic approaches. Internist (Berl). 51:21–29. 2010.In German. View Article : Google Scholar

15 

Vargas MR and Johnson JA: The Nrf2-ARE cytoprotective pathway in astrocytes. Expert Rev Mol Med. 11:e172009. View Article : Google Scholar : PubMed/NCBI

16 

Klaassen CD and Reisman SA: Nrf2 the rescue: Effects of the antioxidative/electrophilic response on the liver. Toxicol Appl Pharmacol. 244:57–65. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Lamlé J, Marhenke S, Borlak J, von Wasielewski R, Eriksson CJ, Geffers R, Manns MP, Yamamoto M and Vogel A: Nuclear factor-eythroid 2-related factor 2 prevents alcohol-induced fulminant liver injury. Gastrornterology. 134:1159–1168. 2008. View Article : Google Scholar

18 

Bar-Sela G, Epelbaum R and Schaffer M: Curcumin as an anti-cancer agent: Review of the gap between basic and clinical applications. Curr Med Chem. 17:190–197. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Rahman I: Antioxidant therapeutic advances in COPD. Ther Adv Respir Dis. 2:351–374. 2008. View Article : Google Scholar

20 

Ishiqaki N, Yamamoto N, Jin H, Uchida K, Terai S and Sakaida I: Continuos intravenous infusion of atrial natriuretic peptide (ANP) prevented liver fibrosis in rat. Biochem Biophys Res Commun. 378:354–359. 2009. View Article : Google Scholar

21 

Copple IM, Goldring CE, Kitteringham NR and Park BK: The Nrf2-Keap1 defense pathway: Role in protection against drug-induced toxicity. Toxicology. 246:24–33. 2008. View Article : Google Scholar

22 

Mazur W, Lindholm P, Vuorinen K, Myllärniemi M, Salmenkivi K and Kinnula VL: Cell-specific elevation of NRF2 and sulfiredoxin-1 as markers of oxidative stress in the lungs of idiopathic pulmonary fibrosis and non-specific interstitial pneumonia. APMIS. 118:703–712. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Churchman AT, Anwar AA, Li FY, Sato H, Ishii T, Mann GE and Siow RC: Transforming growth factor-beta1 elicits Nrf2-mediated antioxidant responses in aortic smooth muscle cells. J Cell Mol Med. 13:2282–2292. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Chen W, Sun Z, Wang XJ, Jiang T, Huang Z, Fang D and Zhang DD: Direct interaction between Nrf2 and p21 (Cip1/WAF1) upregulates the Nrf2-mediated antioxidant response. Mol Cell. 34:663–673. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Kaspar JW, Niture SK and Jaiswal AK: Nrf2: INrf2 (Keap1) signaling in oxidative stress. Free Radic Biol Med. 47:1304–1309. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Katsuoka F, Motohashi H, Ishii T, Aburatani H, Engel JD and Yamamoto M: Genetic evidence that small maf proteins are essential for the activation of antioxidant response element-dependent genes. Mol Cell Biol. 25:8044–8051. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Ruby AJ, Kuttan G, Babu KD, Rajasekharan KN and Kuttan R: Anti-tumor and antioxidant activity of natural curcuminoids. Cancer Lett. 94:79–83. 1995. View Article : Google Scholar : PubMed/NCBI

28 

Joe B, Rao UJ and Lokesh BR: Presence of acidic glycoprotein in the serum of arthritic rats: Modulation by capsaicin and curcumin. Mol Cell Biochem. 169:125–134. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Xu YX, Pindolina KR, Janakiraman N, Noth C, Chapman RA and Gautam SC: Curcumin, a compound with anti-inflammatory and anti-oxidant properties, down-regulates chemokine expression in bone marrow stromal cell. Exp Hematol. 25:413–422. 1997.PubMed/NCBI

30 

Dujic J, Kippenberger S, Ramirez-Bosca A, Diaz-Alperi J, Bereiter-Hahn J, Kaufmann R, Bernd A and Hofmann M: Curcumin in combination with visible light inhibits tumor growth in a xenograft tumor model. Int J Cancer. 124:1422–1428. 2009. View Article : Google Scholar

31 

Fan X, Zhang C, Liu DB, Yan J and Liang HP: The clinical applications of curcumin: Current state and the future. Curr Pharm Des. 19:2011–2031. 2013.

32 

Cheng AL, Hsu CH, Lin JK, Hsu MM, Ho YF, Shen TS, Ko JY, Lin JT, Lin BR, Ming-Shiang W, et al: Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 21:2895–2900. 2001.PubMed/NCBI

33 

Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, Ng CS, Badmaev V and Kurzrock R: Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res. 14:4491–4499. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Hanai H, Iida T, Takeuchi K, Watanabe F, Maruyama Y, Andoh A, Tsujikawa T, Fujiyama Y, Mitsuyama K, Sata M, et al: Curcumin maintenance therapy for ulcerative colitis: Randomized, multi-center, double-blind, placebo-controlled trial. Clin Gastroenterol Hepatol. 4:1502–1506. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Bundy R, Walker AF, Middleton RW and Booth J: Turmeric extract may improve irritable bowel syndrome symptomology in otherwise healthy adults: A pilot study. J Altern Complement Med. 10:1015–1018. 2004. View Article : Google Scholar

36 

Balogun E, Hoque M, Gong P, Killeen E, Green CJ, Foresti R, Alam J and Motterlini R: Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Biochem J. 371:887–895. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Ma ZC, Hong Q, Wang YG, Tan HL, Xiao CR, Liang QD, Zhang BL and Gao Y: Ferulic acid protects human umbilical vein endothelial cells from radiation induced oxidative stress by phosphatidylinositol 3-kinase and extracelluar signal-regulated kinase pathways. Biol Pharm Bull. 33:29–34. 2010. View Article : Google Scholar

38 

Stridh MH, Correa F, Nodin C, Weber SG, Blomstrand F, Nilsson M and Sandberg M: Enhanced glutathione efflux from astrocytes in culture by low extracellular Ca2+ and curcumin. Neurochem Res. 35:1231–1238. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Ballatori N, Krance SM, Notenboom S, Shi S, Tieu K and Hammond CL: Glutathione dysregulation and the etiology and progression of human disease. Biol Chem. 390:191–214. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Bindoli A, Fukuto JM and Forman HJ: Thiol chemistry in peroxidase catalysis and redox signaling. Antioxid Redox Signal. 10:1549–1564. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Johnson JA, Johnson DA, Kraft AD, Calkins MJ, Jakel RJ, Vargas MR and Chen PC: The Nrf2-ARE pathway: An indicator and modulator of oxidative stress in neurodegeneration. Ann N Y Acad Sci. 1147:61–69. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Kraft AD, Johnson DA and Johnson JA: Nuclear factor E2-related factor 2-dependent antioxidant response element activation by tert-butylhydroquinone and sulforaphane occurring preferentially in astrocytes conditions neurons against oxidative insult. J Neurosci. 24:1101–1112. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Reichard JF and Petersen DR: Hepatic stellate cells lack AP-1 responsiveness to electrophiles and phorbol 12-myristate-13-acetate. Biochem Biophys Res Commun. 322:842–853. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Maeda K, Koda M, Matono T, Sugihara T, Yamamoto S, Ueki M, Murawaki Y, Yamashita N and Nishiyama S: Preventive effects of ME3738 on hepatic fibrosis induced by bile duct ligation in rats. Hepatol Res. 38:727–735. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Urtasun R, Conde de la Rosa L and Nieto N: Oxidative and nitrosative stress and fibrogenic response. Clin Liver Dis. 12:769–790. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Wang J, Leclercq I, Brymora JM, Xu N, Ramezani-Moghadam M, London RM, Brigstock D and George J: Kupffer cells mediate leptin-induced liver fibrosis. Gastroenterology. 137:713–723. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Beyer TA, Xu W, Teupser D, auf dem Keller U, Bugnon P, Hildt E, Thiery J, Kan YW and Werner S: Impaired liver regeneration in Nrf2 knockout mice: Role of ROS-mediated insulin/IGF-1 resistance. EMBO J. 27:212–223. 2008. View Article : Google Scholar

48 

Qin Y and Tian YP: Preventive effects of chronic exogenous growth hormone levels on diet-induced hepatic steatosis in rats. Lipids Health Dis. 9:782010. View Article : Google Scholar : PubMed/NCBI

49 

Kulbacka J, Saczko J and Chwiłkowska A: Oxidative stress in cells damage processes. Pol Merkur Lekarski. 27:44–47. 2009.In Polish. PubMed/NCBI

50 

Moselhy SS and Ali HK: Hepatoprotective effect of cinnamon extracts against carbon tetrachloride induced oxidative stress and liver injury in rats. Biol Res. 42:93–98. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Sener G, Kabasakal L, Yüksel M, Gedik N and Alican Y: Hepatic fibrosis in biliary-obstructed rats is prevented by Ginkgo biloba treatment. World J Gastroenterol. 11:5444–5449. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Kim J, Seok YM, Jung KJ and Park KM: Reactive oxygen species/oxidative stress contributes to progression of kidney fibrosis following transient ischemic injury in mice. Am J Physiol Renal Physoil. 297:F461–F470. 2009. View Article : Google Scholar

53 

Osburn WO, Wakabayashi N, Misra V, Nilles T, Biswal S, Trush MA and Kensler TW: Nrf2 regulates an adaptive response protecting against oxidative damage following diquat-mediated formation of superoxide anion. Arch Biochem Biophys. 454:7–15. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Snyder JC, Zemke AC and Stripp BR: Reparative capacity of airway epithelium impacts deposition and remodeling of extracellular matrix. Am J Respir Cell Mol Biol. 40:633–642. 2009. View Article : Google Scholar :

55 

Friedman SL: Mechanisms of hepatic fibrogenesis. Gastroenterology. 134:1655–1669. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Wells RG: The role of matrix stiffness in regulating cell behavior. Hepatology. 47:1394–1400. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 13 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Z, Dou W, Zheng Y, Wen Q, Qin M, Wang X, Tang H, Zhang R, Lv D, Wang J, Wang J, et al: Curcumin upregulates Nrf2 nuclear translocation and protects rat hepatic stellate cells against oxidative stress. Mol Med Rep 13: 1717-1724, 2016
APA
Liu, Z., Dou, W., Zheng, Y., Wen, Q., Qin, M., Wang, X. ... Zhao, S. (2016). Curcumin upregulates Nrf2 nuclear translocation and protects rat hepatic stellate cells against oxidative stress. Molecular Medicine Reports, 13, 1717-1724. https://doi.org/10.3892/mmr.2015.4690
MLA
Liu, Z., Dou, W., Zheng, Y., Wen, Q., Qin, M., Wang, X., Tang, H., Zhang, R., Lv, D., Wang, J., Zhao, S."Curcumin upregulates Nrf2 nuclear translocation and protects rat hepatic stellate cells against oxidative stress". Molecular Medicine Reports 13.2 (2016): 1717-1724.
Chicago
Liu, Z., Dou, W., Zheng, Y., Wen, Q., Qin, M., Wang, X., Tang, H., Zhang, R., Lv, D., Wang, J., Zhao, S."Curcumin upregulates Nrf2 nuclear translocation and protects rat hepatic stellate cells against oxidative stress". Molecular Medicine Reports 13, no. 2 (2016): 1717-1724. https://doi.org/10.3892/mmr.2015.4690