Open Access

IL‑22 alleviates the fibrosis of hepatic stellate cells via the inactivation of NLRP3 inflammasome signaling

  • Authors:
    • Zhuyun Xing
    • Yayun Wu
    • Na Liu
  • View Affiliations

  • Published online on: July 30, 2021     https://doi.org/10.3892/etm.2021.10522
  • Article Number: 1088
  • Copyright: © Xing et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Persistent and progressive liver injury causes liver fibrosis due to the inability of the liver to regenerate. Interleukin (IL)‑22 serves an important role in liver fibrosis. However, the underlying mechanism by which IL‑22 exerts its effects on liver fibrosis has not been fully elucidated. The aim of the present study was to investigate the underlying mechanism by which IL‑22 affects the development of liver fibrosis. Following activation of the hepatic stellate cells (HSCs) using transforming growth factor β (TGF‑β), HSC proliferation was measured using the Cell Counting Kit‑8 assay. The indicators of oxidative stress were detected using specific kits. In addition, the mRNA and protein expression levels of fibrosis‑associated genes were determined using reverse transcription‑quantitative polymerase chain reaction and western blot analysis, respectively. Subsequently, the protein expression levels of the NOD‑like receptor protein 3 (NLRP3), caspase-1 and IL‑1β were examined using western blotting. Following addition of Nigericin, a NLRP3 activator, the levels of oxidative stress and fibrosis were measured. IL‑22 increased the viability of HSCs, which were activated by TGF‑β. The malondialdehyde content was significantly decreased, whereas superoxide dismutase and glutathione levels were increased following IL‑22 treatment. Moreover, IL‑22 markedly downregulated the expression levels of fibrosis‑associated genes, including α‑smooth muscle actin, type I collagen and TIMP metallopeptidase inhibitor 1. Furthermore, the expression levels of NLRP3, caspase-1 and IL‑1β were decreased in the IL‑22‑treated groups. However, the NLRP3 activator Nigericin reversed the inhibitory effects of IL‑22 on the induction of oxidative stress and fibrosis of HSCs induced by TGF‑β. In conclusion, the present study indicated that IL‑22 alleviated the fibrosis of HSCs by inactivation of NLRP3 inflammasome signaling, which may provide further insight on the underlying mechanism by which IL‑22 exerts protective effects on liver fibrosis.

Introduction

Persistent and progressive liver injury results in the inability of the liver to regenerate and leads to liver fibrosis (1). Liver fibrosis is a wound healing response that is closely associated with hepatic stellate cells (HSCs) and Kupffer cells (2). The activation and accumulation of a large number of HSCs is a pivotal step in the formation and progression of liver fibrosis (3). Initiation and perpetuation are two stages that constitute the activation of HSCs (4). Persistent activation of HSCs can contribute to cellular events, including extracellular matrix degradation and cytokine release, which result in the production and accumulation of fibrotic extracellular matrix components, including α-smooth muscle actin (α-SMA) and collagen type I α1 (COL1A1) (5). Additionally, HSCs can synthesize and secrete TIMP metallopeptidase inhibitor 1 (TIMP1), which is a powerful inhibitor of enzymes that degrade matrix molecules and serves a critical role in fibrosis (6). Overall, liver fibrosis has become an obstacle in clinical trials or during treatment of patients with chronic liver diseases (7). Therefore, there is an urgent need to identify novel and effective treatment strategies for this disease.

Interleukin (IL)-22 was originally identified as an IL-10-related T cell-derived inducible factor (8). IL-22 is an actively secreted protein of 146 amino acids, which belongs to the IL-10 family of cytokines (9,10). Despite the differences noted in primary sequences among IL-10 members, they share a common gene structure containing six helices (11). IL-22 expression is downregulated in the serum of mice and in diabetic nephropathy patients (12). By binding to the IL-22 receptor subunit α-1 (IL-22RA1) and IL-10R2, IL-22 exerts protective effects on hepatocytes (13). It has been shown that the binding of IL-22 to IL-22RA1 can induce senescence of HSCs and ameliorate liver fibrosis (14). However, the activation of several signaling pathways, such as STAT3, SOCS3, p53 and Notch, induced by IL-22 inhibits the progression of liver damage (15).

Oxidative stress serves a crucial role in inducing HSC activation and fibrogenic potential (16). Superoxide dismutase (SOD), one of the most important protective enzymes, provides the first antioxidant defense system in various organs and tissues, including the liver (17). Glutathione (GSH) is a key reactive oxygen species scavenger to protect liver cells from oxidative stress (18). SOD and GSH serve an important role in the antioxidant defense system, while malondialdehyde (MDA) is considered to be the main marker of lipid peroxidation in tissues (19). Inflammation serves a critical role in the progression of liver injury and fibrosis. NOD-like receptor protein 3 (NLRP3) is a well-established inflammasome that consists of apoptosis-associated speck-like protein and the effector molecule pro-caspase-1(20). Recent studies have confirmed the role of NLRP3 in a variety of liver diseases, such as alcoholic and non-alcoholic fatty liver diseases and non-alcoholic steatohepatitis (21,22). Inhibition of the NLRP3 inflammasome can alleviate renal injury and fibrosis (23). NLRP3 is aberrantly activated in vivo and in vitro following exposure to aristolochic acid, and inhibition of NLRP3 ameliorates renal fibrosis and renal failure (24). However, whether IL-22 ameliorates liver fibrosis by directly inhibiting the activation of NLRP3 has not been previously investigated.

In the present study, the role of IL-22 was investigated in transforming growth factor β (TGF-β)-induced HSCs. Whether IL-22 exerted its effects on liver fibrosis via the regulation of NLRP3 inflammasome signaling was also explored. The current study may guide the future exploration of the pathogenesis of liver fibrosis.

Materials and methods

Cell culture and treatment

HSCs were purchased from the American Type Culture Collection and Nigericin (cat. no. N7143) was purchased from Sigma-Aldrich (Merck KGaA). HSCs were cultured in DMEM supplemented with 10% FBS (both Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin in a PBS buffer (R&D Systems, Inc.) at 37˚C in an incubator with 95% air/5% CO2. The cells were treated with 5 ng/ml TGF-β (Sigma-Aldrich; Merck KGaA) at 37˚C for 24 h and subsequently cultured in the presence of 250, 500, 750 and 1,000 pg/ml IL-22 (Sigma-Aldrich; Merck KGaA) for 48 h at 37˚C (25,26). Finally, the cells were treated with Nigericin (40 µM) for 30 min at 37˚C to activate NLRP3 for subsequent experiments (27).

Cell Counting Kit-8 (CCK-8) assay

The CCK-8 assay was conducted for the determination of cell viability. Briefly, HSCs (3x104 cells/well) were inoculated into a 96-well plate at 37˚C. At 48 h after IL-22 treatment, 10 µl CCK-8 reagent (Dojindo Molecular Technologies, Inc.) was added to each well and incubated for 3 h. The absorbance at 450 nm was read using a microplate reader (Bio-Rad Laboratories, Inc.). Three replicates were conducted for each sample.

Apoptosis assay

Apoptosis was evaluated via flow cytometry. HSCs were collected and then re-suspended in 1X binding buffer to a concentration of 1x104 cells/well. An Annexin V-FITC Staining Cell Apoptosis Detection kit (Nanjing KeyGen Biotech Co., Ltd.) was employed to determine apoptosis in accordance with the manufacturer's protocol. Briefly, cells were stained with Annexin V-FITC and PI, and incubated in the dark for 15 min at 37˚C. Cell apoptosis of each sample was assessed by flow cytometry (BD Accuri™ C6; BD Biosciences). Apoptosis was analyzed using FlowJo software (version 7.6.3; FlowJo LLC).

Detection of oxidative stress

The contents of MDA and GSH, as well as the activity of SOD, were respectively detected using the MDA assay kit (cat. no. A003-4-1), GSH assay kit (cat. no. A006-2-1) and SOD assay kit (cat. no. A001-3-2; all from Nanjing Jiancheng Bioengineering Institute) in accordance with the manufacturer's instructions.

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

Total RNA was isolated from cells using TRIzol® reagent (Sigma-Aldrich; Merck KGaA). cDNA was synthesized using a Sensiscript RT kit (Qiagen GmbH) following manufacturer's recommendations. According to the manufacturer's recommendations, PCR assays were conducted using SYBR-Green® Realtime PCR Master Mix and Premix Ex Taq (Takara Bio., Inc.) on ViiA™ 7 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The following thermocycling conditions were used: 5 min at 95˚C, followed by 35 cycles at 95˚C for 15 sec, 40 sec at 55˚C and 72˚C for 1 min. Relative quantification of gene expression was performed using the 2−ΔΔCq method (28). The relative gene expression levels were normalized to those of GAPDH. Three independent experiments were conducted for each target gene. The sequences of the gene-specific primers used in the present study were as follows: α-SMA forward, 5'-TCCAGAGTC CAGCACAATACCAG-3' and reverse, 5'-AATGACCCAGAT TATGTTTGAGACC-3'; COL1A1 forward, 5'-TCAGGGGCG AAGGCAACAGT-3' and reverse, 5'-TTGGGATGGAGG GAGTTTACACGA-3'; TIMP1 forward, 5'-ACCACCTTA TACCAGCGTTATGA-3' and reverse, 5'-GGTGTAGACGAA CCGGATGTC-3'; GAPDH forward, 5'-CTCACCGGATGC ACCAATGTT-3' and reverse, 5'-CGCGTTGCTCACAAT GTTCAT-3'.

Western blot analysis

HSCs were lysed in radioimmunoprecipitation (RIPA) lysis buffer (Beyotime Institute of Biotechnology) and the concentrations of the proteins were measured using a BCA protein assay (Shanghai Yeasen Biotechnology Co., Ltd.). Proteins (40 µg/lane) were separated via 10% SDS-PAGE and were transferred to PVDF membranes, which were then blocked with 5% skimmed milk for 1 h at room temperature. Subsequently, the membranes were incubated at 4˚C overnight with primary antibodies (all 1:1,000) against NLRP3 (cat. no. 15101S), IL-1β (cat. no. 12703S), caspase-1 (cat. no. 3866T), α-SMA (cat. no. 19245T), COL1A1 (cat. no. 72026T), TIMP1 (cat. no. 8946S) and GAPDH (cat. no. 5174T; all from Cell Signaling Technology, Inc.). The membranes were washed three times with TBS-0.2% Tween 20 before incubation with goat anti-rabbit HRP-conjugated secondary antibody (1:3,000; cat. no. 7074S; Cell Signaling Technology, Inc.) for 1 h at room temperature. The expression levels of the targeted proteins were determined using a Bio-Rad ChemiDoc MP imaging system (Bio-Rad Laboratories, Inc.). The bands were detected using an enhanced chemiluminescence reagent (EMD Millipore). The grayscale values of the membranes were semi-quantified using ImageJ software (version 1.52r; National Institutes of Health). GAPDH was used as an internal control.

Statistical analysis

Data were analyzed using GraphPad Prism version 6.0 (GraphPad Software, Inc.). Data were expressed as the mean ± SD. All experiments were performed three times. Comparisons between two groups were conducted by unpaired Student's t-test, while comparisons among multiple groups were performed by one-way ANOVA followed by Turkey's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

IL-22 attenuates HSC viability and oxidative stress activated by TGF-β

Following incubation of HSCs with TGF-β, their viability was significantly increased, while concomitant IL-22 treatment led to significant decreases in HSC viability in a dose-dependent manner (Fig. 1A). As shown in Fig. 1B and C, TGF-β treatment significantly inhibited the apoptosis of HSCs compared with the untreated group. By contrast, IL-22 significantly increased TGF-β-induced apoptosis in a dose-dependent manner (Fig. 1B and C). Subsequently, factors associated with oxidative stress were measured using commercially available kits. As shown in Fig. 1D, TGF-β stimulation caused a significant increase in the relative content of MDA, whereas increasing concentrations of IL-22 led to a gradual decrease in MDA levels. Conversely, the levels of SOD and GSH were significantly inhibited following TGF-β treatment, whereas IL-22 restored their levels in a dose-dependent manner (Fig. 1E and F). Overall, these findings revealed that IL-22 inhibited viability and oxidative stress in HSCs induced by TGF-β.

IL-22 alleviates fibrosis of HSCs activated by TGF-β

Subsequently, the ability of IL-22 to inhibit HSC fibrosis activated by TGF-β was assessed. To confirm this hypothesis, RT-qPCR and western blot analyses were conducted to assess the expression levels of fibrosis-associated genes. TGF-β activation in HSCs significantly enhanced the mRNA and protein expression levels of α-SMA, COL1A1 and TIMP1 compared with the levels in untreated HSCs (Fig. 2A-D). In addition, IL-22 treatment of HSCs activated by TGF-β caused a downregulation in the expression levels of the aforementioned fibrosis-associated factors (Fig. 2A-D). Therefore, the results indicated that IL-22 suppressed HSC fibrosis activated by TGF-β.

IL-22 inhibits NLRP3 inflammasome signaling in TGF-β-induced HSCs

Subsequently, the expression levels of key proteins involved in the NLRP3 inflammasome signaling pathway were detected by western blot analysis in TGF-β-induced HSCs in order to confirm whether IL-22 could alleviate inflammation of TGF-β-induced HSCs. It was observed that TGF-β caused a significant increase in the expression levels of NLRP3, caspase-1 and IL-1β, and this effect was reversed by IL-22 (Fig. 3). Therefore, the results suggested that IL-22 inhibited inflammation of TGF-β-induced HSCs. Treatment of the cells with 500, 750 and 1,000 ng/ml IL-22 resulted in a significant decrease of the expression levels of the inflammatory factors. Therefore, IL-22 was used at a concentration of 750 ng/ml in subsequent experiments.

Nigericin reverses the inhibitory effects of IL-22 on oxidative stress and fibrosis in HSCs stimulated by TGF-β

To further investigate the exact mechanism by which IL-22 exerts its cytoprotective effects, the NLRP3 activator Nigericin was used to treat HSCs. TGF-β significantly upregulated the protein expression levels of NLRP3, caspase-1 and IL-1β, and this effect was reversed by co-treatment with TGF-β and IL-22 (Fig. 4). However, the addition of Nigericin in HSCs co-treated with TGF-β and IL-22 significantly increased the expression levels of NLRP3, caspase-1 and IL-1β compared with HSCs treated with TGF-β and IL-22 (Fig. 4). Similarly, CCK-8 and flow cytometry assays indicated that Nigericin treatment reversed the inhibition of IL-22 on cell viability and apoptosis of TGF-β-induced HSCs (Fig. 5A-C). In addition, the levels of the oxidative stress indices measured in the present study were significantly reversed following Nigericin treatment in TGF-β- and IL-22-induced HSCs compared with TGF-β and IL-22 treatment (Fig. 5D-F). Moreover, the results from the RT-qPCR and western blot analyses suggested that the IL-22-induced decreases in the mRNA and protein expression levels of α-SMA, COL1A and TIMP1 in TGF-β-induced HSCs were significantly reversed by Nigericin (Fig. 6A-D). The present results indicated that Nigericin reversed the inhibitory effects of IL-22 on the induction of oxidative stress and fibrosis in HSCs stimulated by TGF-β.

Discussion

HSCs are a type of non-parenchymal cells that are important members in the occurrence and development of liver fibrosis and cirrhosis (29). The exposure of HSCs to TGF-β transforms their still state into an activated cell form, where HSCs transdifferentiate into proliferative myofibroblasts with pro-fibrogenic transcriptional and secretory properties that are vital for the development of liver fibrosis and collagen deposition (30,31). Therefore, the present study used HSCs activated by TGF-β for the simulation of liver fibrosis in vitro.

IL-22 is a factor that protects liver tissues from the development of various chronic diseases, such as nonalcoholic steatohepatitis, liver fibrosis and hepatocellular carcinoma (15,32). IL-22 can promote liver repair and tissue regeneration by stimulating the proliferation and survival of hepatocytes (33). A previous study has suggested that IL-22 exerts inhibitory effects on liver fibrosis and that the attenuation of HSC activation and inhibition of inflammatory factors by IL-22 can restrict liver fibrogenesis (34). Overexpression of IL-22 protects mice from liver injury and liver apoptosis and necrosis stimulated by various factors (35). In the present study, IL-22 inhibited the proliferation of HSCs and the development of liver fibrosis in vitro, which was consistent with the aforementioned studies. Immune response and inflammation in the liver are contributors of hepatocyte injury and activation of HSCs (29). The induction of oxidative stress and inflammation activated by TGF-β in HSCs was attenuated by IL-22 in the present study.

NLRP3 is an intracellular multi-protein complex, and its activation, which occurs by various types of stimuli, such as endogenous danger signals and environmental irritants, can lead to host defense against microbial infections, activation of caspase-1 and further induction of maturation and secretion of IL-1β and IL-18. This process initiates inflammation (36,37). Inhibition or inactivation of the NLRP3 inflammasome is suggested to enhance lipid metabolism and decrease the levels of inflammation, pyroptosis and the infiltration of immune cells into plaques, thereby alleviating inflammatory responses (38-40). An increased number of studies have reported that the inflammasome and its downstream effectors can trigger liver fibrosis (41-43). The activation of the NLRP3 inflammasome leads to liver inflammation, hepatocyte pyroptosis and liver tissue fibrosis in mice, while pharmacological inhibition of NLRP3 decreases hepatocyte injury and liver inflammation (42,44). Moreover, activation of the NLRP3 signaling pathway triggers the activation of HSCs, which is of great importance for the initiation and development of liver fibrosis (45,46). A previous study focused on the potential drug therapies of liver fibrosis and revealed that the suppression of NLRP3 by oridonin could effectively alleviate this process (47). The selective inhibitor of NLRP3, MCC950, can markedly inhibit liver injury in mice induced by bile duct ligation (41). In the present study, the NLRP3 activator Nigericin was used to confirm the specific role of NLRP3 in liver fibrosis. The addition of Nigericin markedly reversed the inhibitory effect of IL-22 on liver fibrosis of TGF-β-induced HSCs, which indirectly demonstrated that inhibition of the NLRP3 inflammasome may be an effective and potential therapeutic strategy used in the treatment of liver injury and fibrosis.

In conclusion, the current data indicated that IL-22 may serve an important role in inhibiting liver fibrosis by inactivation of NLRP3 inflammasome signaling, which may provide further insight on the underlying mechanism by which IL-22 exerts protective effects on liver fibrosis. However, in vivo experiments were not performed and the effects of IL-22-knockdown on NLRP3 inflammasome signaling were not investigated in the present study. Therefore, further experiments should be performed in future studies to confirm the present conclusions.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

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

Authors' contributions

ZX and YW searched the literature, designed the experiments and performed the experiments. NL analyzed, interpreted the data and wrote the manuscript. ZX revised the manuscript. ZX and YW confirmed the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Seki E and Brenner DA: Recent advancement of molecular mechanisms of liver fibrosis. J Hepatobiliary Pancreat Sci. 22:512–518. 2015.PubMed/NCBI View Article : Google Scholar

2 

Matsuda M and Seki E: The liver fibrosis niche: Novel insights into the interplay between fibrosis-composing mesenchymal cells, immune cells, endothelial cells, and extracellular matrix. Food Chem Toxicol. 143(111556)2020.PubMed/NCBI View Article : Google Scholar

3 

Zhang CY, Yuan WG, He P, Lei JH and Wang CX: Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol. 22:10512–10522. 2016.PubMed/NCBI View Article : Google Scholar

4 

Friedman SL: Mechanisms of disease: Mechanisms of hepatic fibrosis and therapeutic implications. Nat Clin Pract Gastroenterol Hepatol. 1:98–105. 2004.PubMed/NCBI View Article : Google Scholar

5 

Wu S, Liu L, Yang S, Kuang G, Yin X, Wang Y, Xu F, Xiong L, Zhang M, Wan J, et al: Paeonol alleviates CCl4-induced liver fibrosis through suppression of hepatic stellate cells activation via inhibiting the TGF-β/Smad3 signaling. Immunopharmacol Immunotoxicol. 41:438–445. 2019.PubMed/NCBI View Article : Google Scholar

6 

Kumar P, Smith T, Rahman K, Thorn NE and Anania FA: Adiponectin agonist ADP355 attenuates CCl4-induced liver fibrosis in mice. PLoS One. 9(e110405)2014.PubMed/NCBI View Article : Google Scholar

7 

Schuppan D: Liver fibrosis: Common mechanisms and antifibrotic therapies. Clin Res Hepatol Gastroenterol. 39 (Suppl 1):S51–S59. 2015.PubMed/NCBI View Article : Google Scholar

8 

Kunkl M, Amormino C, Frascolla S, Sambucci M, De Bardi M, Caristi S, Arcieri S, Battistini L and Tuosto L: CD28 autonomous signaling orchestrates IL-22 expression and IL-22-regulated epithelial barrier functions in human T lymphocytes. Front Immunol. 11(590964)2020.PubMed/NCBI View Article : Google Scholar

9 

Xie MH, Aggarwal S, Ho WH, Foster J, Zhang Z, Stinson J, Wood WI, Goddard AD and Gurney AL: Interleukin (IL)-22, a novel human cytokine that signals through the interferon receptor-related proteins CRF2-4 and IL-22R. J Biol Chem. 275:31335–31339. 2000.PubMed/NCBI View Article : Google Scholar

10 

Nagem RA, Colau D, Dumoutier L, Renauld JC, Ogata C and Polikarpov I: Crystal structure of recombinant human interleukin-22. Structure. 10:1051–1062. 2002.PubMed/NCBI View Article : Google Scholar

11 

Wolk K, Witte E, Witte K, Warszawska K and Sabat R: Biology of interleukin-22. Semin Immunopathol. 32:17–31. 2010.PubMed/NCBI View Article : Google Scholar

12 

Wang S, Li Y, Fan J, Zhang X, Luan J, Bian Q, Ding T, Wang Y, Wang Z, Song P, et al: Interleukin-22 ameliorated renal injury and fibrosis in diabetic nephropathy through inhibition of NLRP3 inflammasome activation. Cell Death Dis. 8(e2937)2017.PubMed/NCBI View Article : Google Scholar

13 

Xing WW, Zou MJ, Liu S, Xu T, Gao J, Wang JX and Xu DG: Hepatoprotective effects of IL-22 on fulminant hepatic failure induced by d-galactosamine and lipopolysaccharide in mice. Cytokine. 56:174–179. 2011.PubMed/NCBI View Article : Google Scholar

14 

Kim DK, Jo A, Lim HS, Kim JY, Eun KM, Oh J, Kim JK, Cho SH and Kim DW: Enhanced type 2 immune reactions by increased IL-22/IL-22Ra1 signaling in chronic rhinosinusitis with nasal polyps. Allergy Asthma Immunol Res. 12:980–993. 2020.PubMed/NCBI View Article : Google Scholar

15 

Wu Y, Min J, Ge C, Shu J, Tian D, Yuan Y and Zhou D: Interleukin-22 in liver injury, inflammation and cancer. Int J Biol Sci. 16:2405–2413. 2020.PubMed/NCBI View Article : Google Scholar

16 

Que R, Shen Y, Ren J, Tao Z, Zhu X and Li Y: Estrogen receptor β dependent effects of saikosaponin d on the suppression of oxidative stress induced rat hepatic stellate cell activation. Int J Mol Med. 41:1357–1364. 2018.PubMed/NCBI View Article : Google Scholar

17 

Pan X, Shao Y, Wang F, Cai Z, Liu S, Xi J, He R, Zhao Y and Zhuang R: Protective effect of apigenin magnesium complex on H2O2-induced oxidative stress and inflammatory responses in rat hepatic stellate cells. Pharm Biol. 58:553–560. 2020.PubMed/NCBI View Article : Google Scholar

18 

Novo E, Busletta C, Bonzo LV, Povero D, Paternostro C, Mareschi K, Ferrero I, David E, Bertolani C, Caligiuri A, et al: Intracellular reactive oxygen species are required for directional migration of resident and bone marrow-derived hepatic pro-fibrogenic cells. J Hepatol. 54:964–974. 2011.PubMed/NCBI View Article : Google Scholar

19 

Li C, Meng M, Guo MZ, Wang MY, Ju AN and Wang CL: The polysaccharides from Grifola frondosa attenuate CCl4-induced hepatic fibrosis in rats via the TGF-beta/Smad signaling pathway. RSC Advances. 9:33684–33692. 2019.

20 

Ting JP, Lovering RC, Alnemri ES, Bertin J, Boss JM, Davis BK, Flavell RA, Girardin SE, Godzik A, Harton JA, et al: The NLR gene family: A standard nomenclature. Immunity. 28:285–287. 2008.PubMed/NCBI View Article : Google Scholar

21 

Wu X, Dong L, Lin X and Li J: Relevance of the NLRP3 inflammasome in the pathogenesis of chronic liver disease. Front Immunol. 8(1728)2017.PubMed/NCBI View Article : Google Scholar

22 

Feng D, Mukhopadhyay P, Qiu J and Wang H: Inflammation in liver diseases. Mediators Inflamm. 2018(3927134)2018.PubMed/NCBI View Article : Google Scholar

23 

Wang S, Li Y, Fan J, Zhang X, Luan J, Bian Q, Ding T, Wang Y, Wang Z, Song P, et al: Interleukin-22 ameliorated renal injury and fibrosis in diabetic nephropathy through inhibition of NLRP3 inflammasome activation. Cell Death Dis. 8(e2937)2017.PubMed/NCBI View Article : Google Scholar

24 

Wang S, Fan J, Mei X, Luan J, Li Y, Zhang X, Chen W, Wang Y, Meng G and Ju D: Interleukin-22 attenuated renal tubular injury in aristolochic acid nephropathy via suppressing activation of NLRP3 inflammasome. Front Immunol. 10(2277)2019.PubMed/NCBI View Article : Google Scholar

25 

Chen E, Cen Y, Lu D, Luo W and Jiang H: IL-22 inactivates hepatic stellate cells via downregulation of the TGF-β1/Notch signaling pathway. Mol Med Rep. 17:5449–5453. 2018.PubMed/NCBI View Article : Google Scholar

26 

Mu M, Zuo S, Wu RM, Deng KS, Lu S, Zhu JJ, Zou GL, Yang J, Cheng ML and Zhao XK: Ferulic acid attenuates liver fibrosis and hepatic stellate cell activation via inhibition of TGF-β/Smad signaling pathway. Drug Des Devel Ther. 12:4107–4115. 2018.PubMed/NCBI View Article : Google Scholar

27 

Martine P, Chevriaux A, Derangère V, Apetoh L, Garrido C, Ghiringhelli F and Rébé C: HSP70 is a negative regulator of NLRP3 inflammasome activation. Cell Death Dis. 10(256)2019.PubMed/NCBI View Article : Google Scholar

28 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

29 

Aslamazova EB, Stroganova EV and Dzhinanian VL: Pathogenesis of liver cirrhosis. Ter Arkh. 47:30–34. 1975.PubMed/NCBI(In Russian).

30 

Lakner AM, Steuerwald NM, Walling TL, Ghosh S, Li T, McKillop IH, Russo MW, Bonkovsky HL and Schrum LW: Inhibitory effects of microRNA 19b in hepatic stellate cell-mediated fibrogenesis. Hepatology. 56:300–310. 2012.PubMed/NCBI View Article : Google Scholar

31 

Friedman SL: Hepatic stellate cells: Protean, multifunctional, and enigmatic cells of the liver. Physiol Rev. 88:125–172. 2008.PubMed/NCBI View Article : Google Scholar

32 

Hwang S, He Y, Xiang X, Seo W, Kim SJ, Ma J, Ren T, Park SH, Zhou Z, Feng D, et al: Interleukin-22 ameliorates neutrophil-driven nonalcoholic steatohepatitis through multiple targets. Hepatology. 72:412–429. 2020.PubMed/NCBI View Article : Google Scholar

33 

Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ, Karow M and Flavell RA: Interleukin-22 but not interleukin-17 provides protection to hepatocytes during acute liver inflammation. Immunity. 27:647–659. 2007.PubMed/NCBI View Article : Google Scholar

34 

Lu DH, Guo XY, Qin SY, Luo W, Huang XL, Chen M, Wang JX, Ma SJ, Yang XW and Jiang HX: Interleukin-22 ameliorates liver fibrogenesis by attenuating hepatic stellate cell activation and downregulating the levels of inflammatory cytokines. World J Gastroenterol. 21:1531–1545. 2015.PubMed/NCBI View Article : Google Scholar

35 

Xu K, Sun J, Chen S and Li Y, Peng X, Li M and Li Y: Hydrodynamic delivery of IL-38 gene alleviates obesity-induced inflammation and insulin resistance. Biochem Biophys Res Commun. 508:198–202. 2019.PubMed/NCBI View Article : Google Scholar

36 

Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM and Dixit VD: The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 17:179–188. 2011.PubMed/NCBI View Article : Google Scholar

37 

Swanson KV, Deng M and Ting JPY: The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat Rev Immunol. 19:477–489. 2019.PubMed/NCBI View Article : Google Scholar

38 

Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nuñez G, Schnurr M, et al: NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 464:1357–1361. 2010.PubMed/NCBI View Article : Google Scholar

39 

Wang R, Wang Y, Mu N, Lou X, Li W, Chen Y, Fan D and Tan H: Activation of NLRP3 inflammasomes contributes to hyperhomocysteinemia-aggravated inflammation and atherosclerosis in apoE-deficient mice. Lab Invest. 97:922–934. 2017.PubMed/NCBI View Article : Google Scholar

40 

Abderrazak A, Couchie D, Mahmood DF, Elhage R, Vindis C, Laffargue M, Matéo V, Büchele B, Ayala MR, El Gaafary M, et al: Anti-inflammatory and antiatherogenic effects of the NLRP3 inflammasome inhibitor arglabin in ApoE2.Ki mice fed a high-fat diet. Circulation. 131:1061–1070. 2015.PubMed/NCBI View Article : Google Scholar

41 

Qu J, Yuan Z, Wang G, Wang X and Li K: The selective NLRP3 inflammasome inhibitor MCC950 alleviates cholestatic liver injury and fibrosis in mice. Int Immunopharmacol. 70:147–155. 2019.PubMed/NCBI View Article : Google Scholar

42 

Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, Hoffman HM and Feldstein AE: NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology. 59:898–910. 2014.PubMed/NCBI View Article : Google Scholar

43 

Wu X, Zhang F, Xiong X, Lu C, Lian N, Lu Y and Zheng S: Tetramethylpyrazine reduces inflammation in liver fibrosis and inhibits inflammatory cytokine expression in hepatic stellate cells by modulating NLRP3 inflammasome pathway. IUBMB Life. 67:312–321. 2015.PubMed/NCBI View Article : Google Scholar

44 

Mridha AR, Wree A, Robertson AAB, Yeh MM, Johnson CD, Van Rooyen DM, Haczeyni F, Teoh NC, Savard C, Ioannou GN, et al: NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J Hepatol. 66:1037–1046. 2017.PubMed/NCBI View Article : Google Scholar

45 

Watanabe A, Sohail MA, Gomes DA, Hashmi A, Nagata J, Sutterwala FS, Mahmood S, Jhandier MN, Shi Y, Flavell RA, et al: Inflammasome-mediated regulation of hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 296:G1248–G1257. 2009.PubMed/NCBI View Article : Google Scholar

46 

Inzaugarat ME, Johnson CD, Holtmann TM, McGeough MD, Trautwein C, Papouchado BG, Schwabe R, Hoffman HM, Wree A and Feldstein AE: NLR family pyrin domain-containing 3 inflammasome activation in hepatic stellate cells induces liver fibrosis in mice. Hepatology. 69:845–859. 2019.PubMed/NCBI View Article : Google Scholar

47 

Liu D, Qin H, Yang B, Du B and Yun X: Oridonin ameliorates carbon tetrachloride-induced liver fibrosis in mice through inhibition of the NLRP3 inflammasome. Drug Dev Res. 81:526–533. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2021
Volume 22 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xing Z, Wu Y and Liu N: IL‑22 alleviates the fibrosis of hepatic stellate cells via the inactivation of NLRP3 inflammasome signaling. Exp Ther Med 22: 1088, 2021
APA
Xing, Z., Wu, Y., & Liu, N. (2021). IL‑22 alleviates the fibrosis of hepatic stellate cells via the inactivation of NLRP3 inflammasome signaling. Experimental and Therapeutic Medicine, 22, 1088. https://doi.org/10.3892/etm.2021.10522
MLA
Xing, Z., Wu, Y., Liu, N."IL‑22 alleviates the fibrosis of hepatic stellate cells via the inactivation of NLRP3 inflammasome signaling". Experimental and Therapeutic Medicine 22.4 (2021): 1088.
Chicago
Xing, Z., Wu, Y., Liu, N."IL‑22 alleviates the fibrosis of hepatic stellate cells via the inactivation of NLRP3 inflammasome signaling". Experimental and Therapeutic Medicine 22, no. 4 (2021): 1088. https://doi.org/10.3892/etm.2021.10522