Saikosaponin‑d alleviates carbon‑tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL‑7702 cell line

  • Authors:
    • Liubing Lin
    • Renye Que
    • Yanting Shen
    • Yirong Chen
    • Ni Yan
    • Yong Li
  • View Affiliations

  • Published online on: April 5, 2018     https://doi.org/10.3892/mmr.2018.8849
  • Pages: 7939-7946
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Saikosaponin‑d (SSd) the primary active component of triterpene saponin derived from Bupleurum falcatum L., possesses anti‑inflammatory and antioxidant properties. The present study aimed to examine the potential therapeutic effects of SSd on carbon tetrachloride (CCl4)‑induced acute hepatocellular injury in the HL‑7702 cell line and its underlying mechanisms. HL‑7702 cells were treated with SSd at different doses (0.5, 1 or 2 µmol/l). Cell viability was determined using an MTT assay. Injury was assessed by the levels of serum alanine aminotransferase (ALT) and aspartate transaminase (AST). Oxidative stress was assessed using malondialdehyde (MDA) content and total‑superoxide dismutase (T‑SOD) activity. The expression of nucleotide‑binding domain, leucine‑rich‑containing family, pyrin domain‑containing‑3 (NLRP3), apoptosis‑associated speck‑like protein (ASC), caspase‑1 and high mobility group protein B1 (HMGB1) was assessed by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis. Interleukin (IL)‑1β and IL‑18 were determined by RT‑qPCR and ELISA. SSd attenuated the inhibition of cell viability and the high AST and ALT levels induced by CCl4 in HL‑7702 cells. Oxidative stress was induced in HL‑7702 cells by CCl4, as demonstrated by the increase of MDA and the decrease of T‑SOD activity. These changes were reversed by SSd. SSd significantly downregulated the mRNA and protein expression of NLRP3, ASC, caspase‑1, IL‑1β, IL‑18 and HMGB1 induced by CCl4. In conclusion SSd alleviated CCl4‑induced acute hepatocellular injury, possibly by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL‑7702 cell line.

Introduction

The liver is important for metabolic homeostasis, detoxification, immunity, and secretory functions (1). Sustained and progressive liver disorders will result in severe liver injury due to increasing cellular, tissue, and function disruption, and is associated with high mortality (2). Management of liver injury remains among the most challenging issues of contemporary medicine (3) and the development of new therapies for acute liver injury is needed.

Carbon tetrachloride (CCl4) is one of the most potent hepatotoxic compounds (4). Acute liver injury models produced using CCl4 are good models of acute chemical liver injury in humans. CCl4 result in the generation of trichloromethyl free radicals (•CCl3) by cytochrome P450. The •CCl3 causes lipid peroxidation and excessive production of reactive oxygen species (ROS), resulting in liver injury (5,6). The imbalance between ROS production and the antioxidant defense leads to oxidative stress, as shown by increased lipid peroxidation and decreased activities of antioxidant enzymes such as the superoxide dismutase (SOD), catalase, and glutathione peroxidase (7).

The nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome has been reported to be involved in the pathogenesis of acute liver injury (8). It is composed of three proteins: NLRP3, apoptosis-associated speck-like protein (ASC), and caspase-1. Once activated by stimuli, the NLRP3 binds to the adaptor protein ASC, and in turn leads to oligomerization of caspase-1, subsequently promoting the activation of caspase-1. Active caspase-1 initiates the cleavage of pro-interleukin (IL)-1β and pro-IL-18 and then promotes the secretion of IL-1β, IL-18, and high mobility group protein B1 (HMGB1) (9). These inflammatory cytokines are critical in various liver diseases such as hepatic ischemia reperfusion injury, alcoholic steatohepatitis, non-alcoholic steatohepatitis, and drug-induced liver injury (1013).

Many herbs exhibit anti-inflammatory and anti-oxidative capacity, and are being used as adjunctive therapy for liver injury. Some compounds extracted from traditional Chinese medicine herbs possess anti-inflammatory and antioxidants effects against CCl4-induced oxidative stress, such as cardamomum, esculentoside A, Ocimum gratissimum, resveratrol, and quercetin (1418). Among these herbs, Radix bupleuri is one of the most commonly used for many diseases such as viral hepatitis and chronic hepatic inflammation (19). Saikosaponin-d (SSd) is the major active component of Bupleurum falcatum L. and has been reported to have anti-inflammatory, antiviral, immunomodulatory, and antioxidant activity (20,21). SSd inhibits the proliferation of rat hepatic stellate cells via decreasing lipid peroxidation (22) and restrains the proliferation of activated T lymphocyte via the NF-AT, NF-κB, and AP-1 pathways (23). In addition, SSd alleviates ventilator-induced lung injury via inhibiting inflammatory responses and oxidative stress (24). Nevertheless, the hepatoprotective effects of SSd against CCl4-induced acute hepatocellular injury remain unclear.

Based on the known effects of SSd, we hypothesized that SSd alleviates the effects of CCl4 on hepatocytes by reducing inflammation and oxidative stress. Therefore, the aim of this study was to investigate the protective effects of SSd on CCl4-induced acute hepatocellular injury in the HL-7702 cell line and whether the effects were related to oxidative stress and NLRP3 inflammasome activation.

Materials and methods

Cell culture

HL-7702 cells (the human normal liver cell line from FuDan IBS Cell Center, Shanghai, China) were routinely cultured in RPMI-1640 medium (Wuhan Boster Biological Technology, Ltd., Wuhan, China) containing 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in 5% CO2 at 37°C. The cells were treated with CCl4 (Beijing Chemical Reagent Company, Beijing, China), SSd (batch no. 110778-201409; China's Drug Supervision, Beijing, China), and N-acetyl-L-cysteine (NAC; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). SSd, CCl4, and NAC were dissolved in dimethyl sulphoxide (DMSO) immediately before use (final concentration of DMSO: <0.1%). DMSO (<0.1%) alone was included as a control in all experiments and did not have any effect on the parameters measured.

MTT assay

Cell proliferation was analyzed by the MTT assay. HL-7702 cells were washed twice with phosphate-buffered saline solution (PBS) and counted. Then, 1×104 cells were seeded on 96-well plates and cultured for 24 h. The cells were exposed to SSd at various concentrations for 24 h. Other cells were seeded onto 96-well plates, incubated for 24 h, and preincubated with NAC (100 µmol/l) or the indicated doses of SSd for 1 h. After that, CCl4 was added at 10 mmol/l to induce acute injury for 24 h (25,26). Then, 100 µl of MTT solution (0.5 mg/ml; Sigma-Aldrich; Merck KGaA) were added to each well and incubated for 4 h. The medium was discarded and 150 µl of DMSO was added for 24 h. The absorbance of each well was measured at 570 nm with an ELx800 universal microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The relative cell viability was calculated as: (absorbance of drug treated group)/(absorbance of untreated group) × 100 (%).

Alanine aminotransferase (ALT) and aspartate transaminase (AST) in supernatants

HL-7702 cells were seeded onto 6-well plates at 8×105 cells/well and cultured in RPMI-1640 with 10% FBS for 24 h. After HL-7702 cells were pretreated with NAC or the indicated doses of SSd for 1 h, CCl4 was added at 10 mmol/l to induce acute hepatocellular injury for 24 h (25,26). The supernatants were collected and stored at −20°C. ALT and AST levels were measured using a Var10skan Flash fluorescence plate reader (Thermo Fisher Scientific, Inc.) using the ALT and AST assay kits (C009-1 and C010-1; Nanjing Jiancheng Institute of Biotechnology, Nanjing, China).

Total-superoxide dismutase (T-SOD) activity and malondialdehyde (MDA) levels

HL-7702 cells were seeded into 6-well plates and cultured in RPMI-1640 with 10% FBS for 24 h. The cells were pretreated with NAC or the indicated doses of SSd for 1 h, followed by CCl4 treatment for another 24 h (25,26). The supernatants were collected and stored at −20°C. MDA was detected by the thiobarbituric acid (TBA) assay using a MDA assay kit (A003-1; Nanjing Jiancheng Institute of Biotechnology). T-SOD activity was determined using the hydroxylamine method with the T-SOD assay kit (A001-1-1; Nanjing Jiancheng Institute of Biotechnology).

Western blotting

NLRP3, caspase-1, ASC, and HMGB1 proteins were measured by western blot. Whole cellular proteins were extracted using the M-PER lysis buffer (Thermo Fisher Scientific, Inc.). Lysates were centrifuged at 12,000 × g for 15 min at 4°C to remove debris. Proteins (50 µg) were separated by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to polyvinylidene difluoride membranes (Immobilon-P; EMD Millipore, Billerica, MA, USA). Membranes were blocked with 5% skimmed milk and probed using NLRP3 (1:500; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), ASC (1:500; Santa Cruz Biotechnology, Inc.), pro-caspase-1 (1:1,000; Abcam, Cambridge, MA, USA), caspase-1 (1:1,000; Abcam), HMGB1 (1:1,000; Abcam), or β-actin (1:2,000; Santa Cruz Biotechnology, Inc.) antibodies overnight at 4°C. Primary antibodies were detected with the corresponding horseradish peroxidase-conjugated secondary antibodies (Cell Signaling Technology, Inc., Danvers, MA, USA) and visualized using an enhanced chemiluminescence (ECL) kit (EMD Millipore) western blotting detection system (Amersham, GE Healthcare, Waukesha, WI, USA). The band intensity was quantified using a Bio-Rad GS-690 Scanner (Bio-Rad Laboratories, Inc.). The relative expression level of each protein was normalized to β-actin or pro-caspase-1.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.). RNA (3 µg) was reverse-transcribed to cDNA with the ReverTra Ace-α-® RT kit (Toyobo, Inc., Tokyo, Japan), according to the manufacturer's protocols. RT-qPCR was performed using the SYBR-Green PCR Master Mix (Toyobo, Inc.) in a PCR detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primers were: NLRP3 forward 5′-TTGACTTCCGCAAGGACCTCGG-3′ and reverse 5′-GCGCCCGGGTTATGCTGCTGGT-3′; ASC forward 5′-GGCTGCTGGATGCTCTGTA-3′ and reverse 5′-AGGCTGGTGTGAAACTGAAGA-3′; caspase-1 forward 5′-CAGACAAGGGTGCTGAACAA-3′ and reverse 5′-TCGGAATAACGGAGTCAATCA-3′; IL-1β forward 5′-TGGCAATGAGGATGACTTGT-3′ and reverse 5′-TGGTGGTCGGAGATTCGTA-3′; IL-18 forward 5′-GACCTTCCAGATCGCTTCCTC-3′ and reverse 5′-GATGCAATTGTCTTCTACTGGTTC-3′; and HMGB1 forward 5′-TATCGCCCAAAAATCAAAGG-3′ and reverse 5′-TAAGGCTGCTTGTCATCTGC-3′. The amplification process was the same for all genes and was 35 cycles of denaturation at 95°C for 45 sec, annealing at 50°C for 45 sec, and elongation at 72°C for 45 sec. β-actin was used as internal control and the primers were: forward 5′-CTCCATCCTGGCCTCGCTGT-3′ and reverse 5′-GCTGTCACCTTCACCGTTCC-3′. The 2−ΔΔCq method was used to represent the relative mRNA expression of the target genes.

Enzyme-linked immunosorbent assay (ELISA)

The levels of IL-18 and IL-1β in the supernatants of HL-7702 cells were measured by ELISA (BMS224HS and BMS267-2; eBioscience, San Diego, CA, USA), according to the manufacturer's instructions.

Statistical analysis

Statistical analysis was conducted using SPSS 21.0 (IBM Corp., Armonk, NY, USA). Results were expressed as mean ± standard deviation. The differences among groups were analyzed using one way analysis of variance (ANOVA) with the Student-Newman-Keuls test for post hoc analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of SSd on the viability of HL-7702 cells

The MTT cell viability assay was performed to examine whether SSd produced cytotoxic effects on HL-7702 cell line. The treatment of HL-7702 cells with SSd at 0.5–2 µmol/l for 24 h did not affect cell viability (Fig. 1), while cell viability was decreased using 2–24 µmol/l (Fig. 1). Therefore, SSd at 0.5, 1, and 2 µmol/l were selected as the low-dose, moderate-dose, and high-dose groups in the subsequent experiments.

SSd alleviates CCl4-induced acute hepatocellular injury

The MTT assay was used to investigate the effects of SSd on CCl4-induced acute hepatocellular injury. As shown in Fig. 2A, CCl4 alone significantly decreased cell viability and SSd reversed the phenomenon in a dose-dependent manner (all P<0.01; Fig. 2A). NAC, as a positive control, also attenuated the decreased cell viability induced by CCl4 (P<0.01; Fig. 2A).

The effects of SSd on CCl4-induced serum AST and ALT levels were examined. CCl4 significantly increased the levels of serum ALT and AST. SSd treatment prevented CCl4-induced increase of AST level in a dose-dependent manner (P<0.05 for low-dose and P<0.01 for the other doses, Fig. 2B). The levels of ALT in the SSd groups were decreased significantly at the moderate and high doses compared to the CCl4 group (P>0.05 for low-dose and P<0.01 for the other doses, Fig. 2C). NAC almost completely reversed the increases of AST and ALT induced by CCl4 (P<0.01; Fig. 2B-C).

SSd attenuates CCl4-induced oxidative stress

To explore the mechanisms by which SSd alleviates CCl4-induced acute hepatocellular injury, the anti-oxidative effects of SSd on CCl4-induced acute hepatocellular injury in HL-7702 cells were explored. We examined the levels of T-SOD and MDA in the cell culture supernatants. The decreased T-SOD induced by CCl4 was attenuated in a dose-dependent manner by SSd (all P<0.01; Fig. 3A), while MDA showed opposite changes (P<0.05 for low-dose and P<0.01 for the other doses, Fig. 3B). CCl4-induced oxidative stress was blocked by NAC (P<0.01; Fig. 3A and B). Thus, the suppressive effect of SSd on oxidative stress could be related to the increase of T-SOD and the reduction of MDA.

SSd inhibits CCl4-induced NLRP3 inflammasome activation

The NLRP3 inflammasome is composed of the NLRP3, caspase-1, and ASC (27,28). To verify whether SSd may affect the NLRP3 inflammasome, NLRP3, ASC, caspase-1, and HMGB1 were assessed by RT-qPCR and western blotting. As shown in Fig. 4A, CCl4 significantly increased NLRP3, caspase-1, ASC, and HMGB1 mRNA levels. Treatment with SSd decreased CCl4-induced NLRP3, ASC, and HMGB1 mRNA expression in a dose-dependent manner (all P<0.01; Fig. 4A), and decreased the level of caspase-1 mRNA at the moderate and high doses (P>0.05 for low-dose, P<0.05 for the moderate dose, and P<0.01 for the high dose; Fig. 4C). The effects of CCl4 injury were blocked by the positive control drug NAC (P<0.01; Fig. 4A).

Similar to the qPCR results, western blotting showed that the expressions of NLRP3, ASC, caspase-1 and HMGB1 were increased by CCl4, but these effects were blocked by NAC (P<0.01; Fig. 4B). The protein expressions of NLRP3, ASC, caspase-1, and HMGB1 were gradually reduced as the dosage of SSd increased (P<0.05 for the low-dose group for ASC, and all other P<0.01; Fig. 4B). Therefore, the anti-inflammatory effects of SSd could be associated with the inhibition of the NLRP3 inflammasome.

SSd inhibits the secretion of proinflammatory cytokines

The production of inflammatory cytokines IL-1β and IL-18 in culture supernatant was examined by qPCR and ELISA. CCl4 significantly increased IL-1β and IL-18 expression, which could be attenuated by NAC. More importantly, treatment with SSd gradually attenuated the expressions of IL-1β and IL-18 as the dosage of SSd increased (all P<0.01; Fig. 5).

Discussion

The CCl4-induced acute liver injury model is frequently used to examine the efficacy of liver protective agents. SSd has been found to attenuate CCl4-induced hepatic injury in rats by inhibiting lipid peroxidation (22). Nevertheless, the downstream molecular mechanism remains unclear. Therefore, this study aimed to examine the effects of SSd on CCl4-induced acute injury in HL-7702 cells and whether the mechanisms could be related to oxidative stress and NLRP3 inflammasome activation. The results suggest that SSd attenuated CCl4-induced acute injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL-7702 cell line.

In the present study, SSd at 0.5–2 µmol/l was protective against CCL4-induced injury, but SSd was toxic at doses >2 µmol/l. Chen et al showed that mitochondrial apoptosis was the main toxic effect of SSd at high concentration (21). Li et al found that saikosaponins at 200 µg/ml induced hepatotoxicity through oxidative stress and lipid metabolism dysregulation (29). On the other hand, Zhao et al found that SSd at 45–60 µmol/l could protect renal tubular epithelial cell against high-glucose-induced injury by regulation of SIRT3 (30). Yao et al found that SSd inhibited the proliferation of prostate cancer cells at 50 µM (31). These results suggest that SSd is toxic at high concentrations, but protective at lower doses.

Oxidative stress is known to be related to the pathogenesis of acute liver injury (3234). CCl4, is widely used to induce acute hepatic injury in animals, has strong hepatotoxic effects that leads to the excessive generation of free radicals associated with oxidative stress, and ultimately results in acute liver injury with functional impairment. Serum AST and ALT are widely used as makers of acute hepatic injury. Decreased levels of AST and ALT associated with fewer necrotic lesions and lipid peroxidation could imply protection against CCl4-induced injury (35). Therefore, we first verified that SSd could attenuate CCl4-induced AST and ALT increases. MDA and T-SOD are indicators of CCl4-induced oxidative stress (36). The increase of MDA, a product of lipid peroxidation, is considered to be a direct indicator of abnormal peroxidation and impaired antioxidant defenses (36). As an antioxidant enzyme, T-SOD catalyzes the dismutation of superoxide anions into hydrogen peroxide and oxygen. Studies showed that CCl4 could lead to excessive generation of free radicals and oxidative stress in the liver by increasing the levels of MDA and decreasing the levels of T-SOD, ultimately leading to acute liver injury (3739). In the present study, SSd played an antioxidant role by inhibiting the production of MDA and improving T-SOD levels in CCl4-induced acute injury in liver cells.

The activation of the NLRP3 inflammasome results in the maturation of the inflammatory cytokines IL-1β and IL-18, and also leads to the release of HMGB1 (9). These potent proinflammatory cytokines further aggravate the inflammatory progress initiated by oxidative stress. IL-1β and IL-18 are members of the IL-1 superfamily and contribute to inflammation (40,41). HMGB1 is a nuclear protein and proinflammatory mediator, and promotes inflammation and necrotic cell death. IL-1β, IL-18, and HMGB1 may be involved in acute hepatocellular injury (4246). The effect of the NLRP3 inflammasome has been explored in many liver conditions such as drug-induced liver injury, non-alcoholic steatohepatitis, alcoholic steatohepatitis, hepatic ischemia reperfusion injury, and fibrosis (1013,47,48). Kim et al (49) showed that NLRP3 inflammasome activation play a central role in GalN/LPS-induced inflammatory responses and the development of hepatic injury. Gong et al (50) showed that the activation of the NLRP3 inflammasome contributes to the induction of inflammation, which might be associated with BDL-induced fibrosis in non-alcoholic and alcoholic steatohepatitis. In the present study, SSd decreased the mRNA and protein expression of the NLRP3 inflammasome components after induction by CCl4, which is consistent with the role of the NLRP3 inflammasome in liver injury. In addition, the levels of IL-1β and IL-18 in the culture supernatants were reduced.

Oxidative stress induced by the overproduction of ROS is an important cause of organ and tissue injury in various inflammatory diseases (33,34). In addition, ROS play crucial roles in the activation of the NLRP3 inflammasome (51,52). Recent evidence also indicate that some herb extracts have anti-inflammatory effects by suppressing ROS production (53). Moreover, it has been confirmed that oxidative stress and inflammatory cytokines are frequently identified as the main factors contributing to acute liver injury, and anti-inflammatory and antioxidant compounds are thought to prevent acute hepatocellular injury (5456). Dang et al (57) showed that SSd attenuated CCl4-induced liver fibrosis in rats through the downregulation of TNF-α, IL-6, and NF-κBp65, and the upregulation of I-κBα. Wu et al (55) also showed the beneficial effects of SSd against liver fibrosis in CCl4 rats models. Taken together, ROS-induced NLRP3 inflammasome activation may be involved in CCl4-induced acute hepatocellular injury. Furthermore, the anti-inflammatory effect of SSd may depend on the modulation the inflammation through the NLRP3 inflammasome. Nevertheless, ROS and factors such as TNF-α, IL-6, NF-κBp65, and I-κBα were not evaluated in this study, and further experiments are needed to confirm those mechanisms. In addition, NLRP3 inhibitors should be used to confirm the role of the NLRP3 inflammasome in CCl4-induced injury and the beneficial effects of SSd.

In conclusion, this study suggests that the suppressive effects of SSd on CCl4-induced acute hepatocellular injury may depend on the inhibition of oxidative stress and NLRP3 inflammasome activation. This study suggests new evidence for the potential efficacy of SSd for the treatment of acute hepatocellular injury.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant no. 81573775), the Shanghai Natural Science Foundation (grant no. 09ZR1429700) and the Shanghai Outstanding Academic Leader of Health System (grant no. XBR2013120).

Availability of data and materials

The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LL made substantial contributions to the acquisition of the data, data analysis and interpretation of the data and writing the manuscript. RQ and YL participated in the conception and design of the study and the revision of the manuscript. LL, YS, YC and NY performed the experiments. YL was primarily responsible for the revision of the manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

SSd

saikosaponin-d

CCl4

carbon tetrachloride

NLRP3

nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3

ALT

alanine aminotransferase

AST

aspartate transaminase

T-SOD

total-superoxide dismutase

MDA

malondialdehyde

HMGB1

high mobility group protein B1

IL

interleukin

References

1 

Zhao P, Qi C, Wang G, Dai X and Hou X: Enrichment and purification of total flavonoids from Cortex Juglandis Mandshuricae extracts and their suppressive effect on carbon tetrachloride-induced hepatic injury in Mice. J Chromatogr B Analyt Technol Biomed Life Sci. 1007:8–17. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Tuñón MJ, Miguel San B, Crespo I, Jorquera F, Santamaria E, Alvarez M, Prieto J and González-Gallego J: Melatonin attenuates apoptotic liver damage in fulminant hepatic failure induced by the rabbit hemorrhagic disease virus. J Pineal Res. 50:38–45. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Auzinger G and Wendon J: Intensive care management of acute liver failure. Curr Opin Crit Care. 14:179–188. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Assayed ME, Khalaf AA and Salem HA: Protective effects of garlic extract and vitamin C against in vivo cypermethrin-induced teratogenic effects in rat offspring. Food Chem Toxicol. 48:3153–3158. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Kodai S, Takemura S, Minamiyama Y, Hai S, Yamamoto S, Kubo S, Yoshida Y, Niki E, Okada S, Hirohashi K and Suehiro S: S-allyl cysteine prevents CCl(4)-induced acute liver injury in rats. Free Radic Res. 41:489–497. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Recknagel RO, Glende EA Jr, Dolak JA and Waller RL: Mechanisms of carbon tetrachloride toxicity. Pharmacol Ther. 43:139–154. 1989. View Article : Google Scholar : PubMed/NCBI

7 

Zelen I, Djurdjevic P, Popovic S, Stojanovic M, Jakovljevic V, Radivojevic S, Baskic D and Arsenijevic N: Antioxidant enzymes activities and plasma levels of oxidative stress markers in B-chronic lymphocytic leukemia patients. J BUON. 15:330–336. 2010.PubMed/NCBI

8 

Pan CW, Pan ZZ, Hu JJ, Chen WL, Zhou GY, Lin W, Jin LX and Xu CL: Mangiferin alleviates lipopolysaccharide and D-galactosamine-induced acute liver injury by activating the Nrf2 pathway and inhibiting NLRP3 inflammasome activation. Eur J Pharmacol. 770:85–91. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Keyel PA: How is inflammation initiated? Individual influences of IL-1, IL-18 and HMGB1. Cytokine. 69:136–145. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Arteel G, Marsano L, Mendez C, Bentley F and McClain CJ: Advances in alcoholic liver disease. Best Pract Res Clin Gastroenterol. 17:625–647. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Neuman MG: Cytokines-central factors in alcoholic liver disease. Alcohol Res Health. 27:307–316. 2003.PubMed/NCBI

12 

Petrasek J, Bala S, Csak T, Lippai D, Kodys K, Menashy V, Barrieau M, Min SY, Kurt-Jones EA and Szabo G: IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J Clin Invest. 122:3476–3489. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Zhu P, Duan L, Chen J, Xiong A, Xu Q, Zhang H, Zheng F, Tan Z, Gong F and Fang M: Gene silencing of NALP3 protects against liver ischemia-reperfusion injury in mice. Hum Gene Ther. 22:853–864. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Zhang F, Wang X, Qiu X, Wang J, Fang H, Wang Z, Sun Y and Xia Z: The protective effect of Esculentoside A on experimental acute liver injury in mice. PLoS One. 9:e1131072014. View Article : Google Scholar : PubMed/NCBI

15 

Lim DW, Kim H, Park JY, Kim JE, Moon JY, Park SD and Park WH: Amomum cardamomum L. ethyl acetate fraction protects against carbon tetrachloride-induced liver injury via an antioxidant mechanism in rats. BMC Complement Altern Med. 16:1552016. View Article : Google Scholar : PubMed/NCBI

16 

Chiu YW, Chao PY, Tsai CC, Chiou HL, Liu YC, Hung CC, Shih HC, Lai TJ and Liu JY: Ocimum gratissimum is effective in prevention against liver fibrosis in vivo and in vitro. Am J Chin Med. 42:833–852. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Chan CC, Lee KC, Huang YH, Chou CK, Lin HC and Lee FY: Regulation by resveratrol of the cellular factors mediating liver damage and regeneration after acute toxic liver injury. J Gastroenterol Hepatol. 29:603–613. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Zhang JQ, Shi L, Xu XN, Huang SC, Lu B, Ji LL and Wang ZT: Therapeutic detoxification of quercetin against carbon tetrachloride-induced acute liver injury in mice and its mechanism. J Zhejiang Univ Sci B. 15:1039–1047. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Lee CH, Wang JD and Chen PC: Risk of liver injury associated with Chinese herbal products containing radix bupleuri in 639,779 patients with hepatitis B virus infection. PLoS One. 6:e160642011. View Article : Google Scholar : PubMed/NCBI

20 

Wong VK, Zhang MM, Zhou H, Lam KY, Chan PL, Law CK, Yue PY and Liu L: Saikosaponin-d enhances the anticancer potency of TNF-α via overcoming its undesirable response of activating NF-Kappa B signalling in cancer cells. Evid Based Complement Alternat Med. 2013:7452952013. View Article : Google Scholar : PubMed/NCBI

21 

Chen L, Zhang F, Kong D, Zhu X, Chen W, Wang A and Zheng S: Saikosaponin D disrupts platelet-derived growth factor-β receptor/p38 pathway leading to mitochondrial apoptosis in human LO2 hepatocyte cells: A potential mechanism of hepatotoxicity. Chem Biol Interact. 206:76–82. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Fan J, Li X, Li P, Li N, Wang T, Shen H, Siow Y, Choy P and Gong Y: Saikosaponin-d attenuates the development of liver fibrosis by preventing hepatocyte injury. Biochem Cell Biol. 85:189–195. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Lu CN, Yuan ZG, Zhang XL, Yan R, Zhao YQ, Liao M and Chen JX: Saikosaponin a and its epimer saikosaponin d exhibit anti-inflammatory activity by suppressing activation of NF-κB signaling pathway. Int Immunopharmacol. 14:121–126. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Wang HW, Liu M, Zhong TD and Fang XM: Saikosaponin-d attenuates ventilator-induced lung injury in rats. Int J Clin Exp Med. 8:15137–4512. 2015.PubMed/NCBI

25 

Lu Q, Yang L, Zhao HY, Jiang JG and Xu XL: Protective effect of compounds from the flowers of Citrus aurantium L. var. amara Engl against carbon tetrachloride-induced hepatocyte injury. Food Chem Toxicol. 62:432–435. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Han WJ, Shi HB, Shi HL, Song JY, Ren F, Duan ZP and Chen Y: Augmenter of liver regeneration promotes the proliferation of HL-7702 cells in carbon tetrachloride-induced acute liver injury via increasing autophag. Zhonghua Gan Zang Bing Za Zhi. 24:761–766. 2016.(In Chinese). PubMed/NCBI

27 

DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, Mangal D, Yu KH, Yeo CJ, Calhoun ES, et al: Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 475:106–109. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Schroder K and Tschopp J: The inflammasomes. Cell. 140:821–832. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Li X, Li X, Lu J, Huang Y, Lv L, Luan Y, Liu R and Sun R: Saikosaponins induced hepatotoxicity in mice via lipid metabolism dysregulation and oxidative stress: A proteomic study. BMC Complement Altern Med. 17:2192017. View Article : Google Scholar : PubMed/NCBI

30 

Zhao L, Zhang H, Bao J, Liu J and Ji Z: Saikosaponin-d protects renal tubular epithelial cell against high glucose induced injury through modulation of SIRT3. Int J Clin Exp Med. 8:6472–6481. 2015.PubMed/NCBI

31 

Yao M, Yang J, Cao L, Zhang L, Qu S and Gao H: Saikosaponind inhibits proliferation of DU145 human prostate cancer cells by inducing apoptosis and arresting the cell cycle at G0/G1 phase. Mol Med Rep. 10:365–372. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Kadiiska MB, Gladen BC, Baird DD, Germolec D, Graham LB, Parker CE, Nyska A, Wachsman JT, Ames BN, Basu S, et al: Biomarkers of oxidative stress study II: Are oxidation products of lipids, proteins, and DNA markers of CCl4 poisoning? Free Radic Biol Med. 38:698–710. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Alfadda AA and Sallam RM: Reactive oxygen species in health and disease. J Biomed Biotechnol. 2012:9364862012. View Article : Google Scholar : PubMed/NCBI

34 

Brüne B, Dehne N, Grossmann N, Jung M, Namgaladze D, Schmid T, von Knethen A and Weigert A: Redox control of inflammation in macrophages. Antioxid Redox Signal. 19:595–637. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Kim WR, Flamm SL, Di Bisceglie AM and Bodenheimer HC: Public Policy Committee of the American Association for the Study of Liver Disease: Serum activity of alanine aminotransferase (ALT) as an indicator of health and disease. Hepatology. 47:1363–1370. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Hoek JB and Pastorino JG: Ethanol, oxidative stress, and cytokine-induced liver cell injury. Alcohol. 27:63–68. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Weber LW, Boll M and Stampfl A: Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol. 33:105–136. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Cheng N, Ren N, Gao H, Lei X, Zheng J and Cao W: Antioxidant and hepatoprotective effects of Schisandra chinensis pollen extract on CCl4-induced acute liver damage in mice. Food Chem Toxicol. 55:234–240. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Tirkey N, Pilkhwal S, Kuhad A and Chopra K: Hesperidin, a citrus bioflavonoid, decreases the oxidative stress produced by carbon tetrachloride in rat liver and kidney. BMC Pharmacol. 5:22005. View Article : Google Scholar : PubMed/NCBI

40 

Dinarello CA: Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 27:519–550. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Davis BK, Wen H and Ting JP: The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol. 29:707–735. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Petrilli V, Papin S, Dostert C, Mayor A, Martinon F and Tschopp J: Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ. 14:1583–1589. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Franchi L, Eigenbrod T, Muñoz-Planillo R and Nuñez G: The inflammasome: A caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 10:241–247. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Yang H, Antoine DJ, Andersson U and Tracey KJ: The many faces of HMGB1: Molecular structure-functional activity in inflammation, apoptosis, and chemotaxis. J Leukoc Biol. 93:865–873. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Chen Q, Yin YX, Wei J, Tong M, Shen F, Zhao M and Chamley L: Increased expression of high mobility group box 1 (HMGB1) in the cytoplasm of placental syncytiotrophoblast from preeclamptic placentae. Cytokine. 85:30–36. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Zhu L, Zhang Z, Zhang L, Shi Y, Qi J, Chang A, Gao J, Feng Y and Yang X: HMGB1-RAGE signaling pathway in severe preeclampsia. Placenta. 36:1148–1152. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Imaeda AB, Watanabe A, Sohail MA, Mahmood S, Mohamadnejad M, Sutterwala FS, Flavell RA and Mehal WZ: Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9 and the Nalp3 inflammasome. J Clin Invest. 119:305–314. 2009.PubMed/NCBI

48 

Artlett CM: The Role of the NLRP3 Inflammasome in Fibrosis. Open Rheumatol J. 6:80–86. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Kim SJ and Lee SM: NLRP3 inflammasome activation in D-galactosamine and lipopolysaccharide-induced acute liver failure: Role of heme oxygenase-1. Free Radic Biol Med. 65:997–1004. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Gong Z, Zhou J, Zhao S, Tian C, Wang P, Xu C, Chen Y, Cai W and Wu J: Chenodeoxycholic acid activates NLRP3 inflammasome and contributes to cholestatic liver fibrosis. Oncotarget. 7:83951–83963. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT and Tschopp J: Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 320:674–677. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Sayan M and Mossman BT: The NLRP3 inflammasome in pathogenic particle and fibre-associated lung inflammation and diseases. Part Fibre Toxicol. 13:512016. View Article : Google Scholar : PubMed/NCBI

53 

Shen YC, Chou CJ, Wang YH, Chen CF, Chou YC and Lu MK: Anti-inflammatory activity of the extracts from mycelia of Antrodia camphorata cultured with water-soluble fractions from five different Cinnamomum species. FEMS Microbiol Lett. 231:137–43. 2004. View Article : Google Scholar : PubMed/NCBI

54 

Jaeschke H: Reactive oxygen and mechanisms of inflammatory liver injury. J Gastroenterol Hepatol. 15:718–724. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Wu SJ, Lin YH, Chu CC, Tsai YH and Chao JC: Curcumin or saikosaponin a improves hepatic antioxidant capacity and protects against CCl4-induced liver injury in rats. J Med Food. 11:224–229. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Yu H, Zheng L, Yin L, Xu L, Qi Y, Han X, Xu Y, Liu K and Peng J: Protective effects of the total saponins from Dioscorea nipponica Makino against carbon tetrachloride-induced liver injury in mice through suppression of apoptosis and inflammation. Int Immunopharmacol. 19:233–244. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Dang SS, Wang BF, Cheng YA, Song P, Liu ZG and Li ZF: Inhibitory effects of saikosaponin-d on CCl4-induced hepatic fibrogenesis in rats. World J Gastroenterol. 13:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 17 Issue 6

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
Lin L, Que R, Shen Y, Chen Y, Yan N and Li Y: Saikosaponin‑d alleviates carbon‑tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL‑7702 cell line. Mol Med Rep 17: 7939-7946, 2018
APA
Lin, L., Que, R., Shen, Y., Chen, Y., Yan, N., & Li, Y. (2018). Saikosaponin‑d alleviates carbon‑tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL‑7702 cell line. Molecular Medicine Reports, 17, 7939-7946. https://doi.org/10.3892/mmr.2018.8849
MLA
Lin, L., Que, R., Shen, Y., Chen, Y., Yan, N., Li, Y."Saikosaponin‑d alleviates carbon‑tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL‑7702 cell line". Molecular Medicine Reports 17.6 (2018): 7939-7946.
Chicago
Lin, L., Que, R., Shen, Y., Chen, Y., Yan, N., Li, Y."Saikosaponin‑d alleviates carbon‑tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL‑7702 cell line". Molecular Medicine Reports 17, no. 6 (2018): 7939-7946. https://doi.org/10.3892/mmr.2018.8849