Open Access

N‑acetyl cysteine inhibits the lipopolysaccharide‑induced inflammatory response in bone marrow mesenchymal stem cells by suppressing the TXNIP/NLRP3/IL‑1β signaling pathway

  • Authors:
    • Xuemei Wang
    • Mengyi Jiang
    • Xiaoping He
    • Bo Zhang
    • Wei Peng
    • Ling Guo
  • View Affiliations

  • Published online on: August 13, 2020     https://doi.org/10.3892/mmr.2020.11433
  • Pages: 3299-3306
  • Copyright: © Wang 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

N-acetyl cysteine (NAC) has been used to inhibit lipopolysaccharide (LPS)-induced inflammation. However, the molecular mechanism underlying its anti‑inflammatory effects remains to be elucidated. The present study aimed to determine the effect of NAC on the LPS‑induced inflammatory response in bone marrow mesenchymal stem cells (BMSCs) and elucidate the underlying molecular mechanism. First, BMSCs were stimulated by LPS following pretreatment with NAC (0, 0.1, 0.5, 1 or 2 mM). A Cell Counting Kit 8 assay was used to determine the number of viable cells and 1 mM NAC was selected as the experimental concentration. Then, the secretion of inflammatory factors, including interleukin (IL)‑1β, IL‑6 and tumor necrosis factor‑α was evaluated by enzyme‑linked immunosorbent assay. Finally, the expression levels of mRNA and proteins, including apoptosis‑associated speck‑like protein containing a CARD (ASC), nucleotide‑binding oligomerization domain‑like receptor protein 3 (NLRP3), caspase‑1, thioredoxin‑interacting protein (TXNIP), and thioredoxin (TRX), were evaluated by reverse transcription‑quantitative PCR and western blot analysis, respectively. The results demonstrated that the secretion of inflammatory factors, which was increased by the administration of LPS, was reduced by pretreatment with NAC. Furthermore, NAC reduced the expression of ASC, NLRP3, caspase‑1 and TXNIP, but enhanced that of TRX. To conclude, NAC had anti‑inflammatory effects on LPS‑stimulated BMSCs, which was closely associated with the TXNIP/NLRP3/IL‑1β signaling pathway. Thus, NAC may be a promising treatment to attenuate the inflammatory response in LPS‑induced BMSCs.

Introduction

Oral inflammation, including periodontitis and peri-implant disease, has become a common challenge encountered in the dental clinic. It directly leads to bone resorption around the tooth or implant, subsequently causing loosening of the tooth and implant failure (1,2). There is a high prevalence of periodontitis and peri-implant disease in the United States (3,4). The National Health and Nutrition Examination Survey conducted between 2009 and 2014 reporterd that 42.2% of US dentate adults aged ≥30 years had periodontitis (5). Derks et al (6) found that 45% of 588 patients who had received implant-supported oral rehablitation therapy 9 years earlier presented with peri-implant disease. The principal etiological factor for these inflammatory diseases is plaque accumulation (7,8). Among the vast array of oral bacteria, Porphyromonas gingivalis serves a particularly important role in both periodontitis and peri-implant diseases, resulting from the expression of various virulence factors, the most recognizable of which is lipopolysaccharide (LPS) (9,10). LPS, also known as endotoxin, induces a variety of cell types, such as osteoblasts (11), human umbilical vein endothelial cells (12) and gingival fibroblasts (13), to overexpress related inflammatory factors, including interleukin (IL)-1β, IL-6 and tumor necrosis factor α (TNF-α). Recently, it was demonstrated that LPS could induce the overexpression of TNF-α, IL-1β and IL-6 in bone marrow mesenchymal stem cells (BMSCs) (14). To identify the molecular mechanism underlying LPS-mediated inflammation, the majority of studies have focused on the vital role served by nuclear factor (NF)-κB, as LPS can mediate the phosphorylation of p65, which is critical for the transactivation of NF-κB (15).

By contrast, less attention has been paid to the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in studies on LPS-induced inflammation. LPS is widely known to stimulate the NLRP3 inflammasome in different cell types (1618). Additionally, the NLRP3 inflammasome serves a significant role in LPS-mediated inflammation, as its upregulation is conducive to the secretion of IL-1β and the excessive release of IL-1β contributes to the cellular inflammatory response (1921). Thus, there is an urgent need for a novel therapeutic approach that blocks activation of the NLRP3 inflammasome and controls inflammation triggered by LPS, thereby eradicating the infection that occurs in both soft and hard tissue and eventually facilitating the development of bone around the tooth or implant.

N-acetyl cysteine (NAC), a precursor of glutathione, is a typical oxygen-free radical scavenger and anti-inflammatory agent (22). It is used as a tool for inhibiting the release of inflammatory factors and reducing the inflammation triggered by LPS (11), as well as in the treatment of different diseases (2325), including cardiovascular diseases, respiratory diseases and cancer. In our previous studies (10,11,26), human umbilical vein endothelial cells, osteoblasts and periodontal ligament fibroblasts were pretreated with NAC prior to their stimulation with LPS. The results demonstrated that this pretreatment led to the inhibition of IL-1β, IL-6 and TNF-α, and the inflammatory response initiated by LPS.

Based on these findings, it was hypothesized that NAC may inhibit the inflammatory response in LPS-induced BMSCs. To test this hypothesis and to clarify the molecular mechanism underlying the anti-inflammatory action of NAC, the effect of NAC on inflammation stimulated by LPS was evaluated. Whether NAC could control the inflammatory response in LPS-mediated BMSCs was also assessed and the associated anti-inflammatory mechanisms were determined.

Materials and methods

Cell culture

BMSCs (cat. no. BNCC340947; BeNa Culture Collection; Beijing Beina Chunglian Biotechnology Research Institute) were maintained in Dulbecco's modified Eagle's medium (DMEM; HyClone; Cytiva) supplemented with 1% (v/v) penicillin-streptomycin (Beyotime Institute of Biotechnology) and 10% (v/v) fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and then cultured at 37°C with 5% CO2. BMSCs were used from the fifth to eighth generations.

Four experimental groups were established (Table I): i) Control; ii) NAC + LPS; iii) resveratrol (Res, 50 µM; Sigma-Aldrich; Merck KGaA) + LPS; and iv) LPS. Cells in the NAC + LPS group were incubated with 1 mM NAC (Sigma-Aldrich; Merck KGaA) for 1 h, while cells in the Res + LPS group were cultured for 2 h in the dark. Next, all groups, excluding the control group, were stimulated with LPS (1 µg/ml; Sigma-Aldrich; Merck KGaA) for 24 h. All experiments were performed at 37°C with 5% CO2.

Table I.

Experimental groups.

Table I.

Experimental groups.

GroupsControlNAC + LPSRes + LPSLPS
NAC+
LPS+++
Res+

[i] NAC, N-acetyl cysteine; LPS, lipopolysaccharide; Res, resveratrol.

Cell proliferation assay

The effects of NAC on BMSC proliferation were determined by a Cell Counting Kit-8 (CCK-8) assay. First, 1×105/cm2 BMSCs were seeded into a 96-well plate and incubated for 24 h at 37°C. Next, BMSCs were pretreated with NAC at concentrations of 0, 0.1, 0.5, 1 and 2 mM for 1 h prior to stimulation with LPS at 37°C. The cells were then cultured for 24 h at 37°C. Subsequently, the original medium was carefully removed prior to the addition of 10 µl CCK-8 reagent (Beyotime Institute of Biotechnology) and 100 µl DMEM to each well. Following incubation at 37°C for 1 h, a microplate reader (Thermo Fisher Scientific, Inc.) was used to detect the absorbance of each well at 450 nm. The appropriate concentration of NAC for cell proliferation was selected with three replicates and the assay was conducted according to the manufacturer's protocols.

Reverse transcription-quantitative (RT-q) PCR

BMSCs (1×105/cm2) were seeded into four plates. Subsequently, the cells were collected and total RNA of apoptosis-associated speck-like protein containing a CARD (ASC), NLRP3, caspase-1, thioredoxin-interacting protein (TXNIP) and thioredoxin (TRX) was isolated from the BMSCs using TRIzol® reagent (Thermo Fisher Scientific, Inc.). The SYBR-Green Real-Time PCR Master Mix (KAPA Biosystems; Roche Diagnostics) was used for qPCR, and cDNA synthesis was performed using the PrimeScript RT Reagent kit (Takara Bio, Inc.) in a reaction that included 1 µl cDNA, 0.5 µl forward primer, 0.5 µl reverse primer, 10 µl SYBR FAST qPCR Master Mix and 8 µl ddH2O to a total volume of 20 µl. All reactions were performed according to the manufacturer's protocols. The PCR protocol was as follows: 95°C for 3 min, followed by 40 cycles of 95°C for 5 sec, 56°C for 10 sec, and 72°C for 25 sec. Assays were conducted in triplicate and β-actin served as the reference gene. The primers are shown in Table II. Finally, the values of 2−ΔΔCq reflected the mRNA abundance (27).

Table II.

Primers for reverse transcription-quantitative PCR.

Table II.

Primers for reverse transcription-quantitative PCR.

Gene namePrimer sequence (5′→3′)Amplicon length (bp)
ASCF: AGCATCCAGCAAACCA259
R: GGACCCCATAGACCTCA
NLRP3F: CATCTTAGTCCTGCCAA94
R: CAACAGACGCTACACCC
Caspase-1F: TTGAAGAGCAGAAAGCA105
R: CAGTAGGAAACTCCGAAG
TXNIPF: CAAGGTAAGTGTGCCG105
R: GATTCTGTGAAGGTGATGA
TRXF: CCAACCTTTTGACCCTTT143
R: CCCTTCTTTCATTCCCTC
β-actinF: TAGGAGCCAGGGCAGTA110
R: CGTTGACATCCGTAAAGAC

[i] F, forward; R, reverse; ASC, apoptosis-associated speck-like protein containing a CARD; NLRP3, nucleotide-binding oligomerization domain-like receptor protein 3; TXNIP, thioredoxin-interacting protein; TRX, thioredoxin.

Western blot analysis

Western blot analysis was conducted to calculate the levels of related proteins, including ASC, NLRP3, caspase-1, TXNIP and TRX, in the BMSCs. The BMSCs were treated as aforementioned, and the total protein was collected with radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, Inc.). The total protein concentration was normalized following quantification with the BCA Protein Assay (Beijing Solarbio Science & Technology Co., Ltd). The proteins (20 µg) were then separated via 12% SDS-PAGE (Sigma-Aldrich; Merck KGaA) and electrotransferred to polyvinylidene fluoride membranes. The membranes were blocked in 5% non-fat milk at room temperature for 2 h and then incubated with the following rabbit primary antibodies for ≥8 h at 4°C: ASC (1:1,000; cat. no. PAB30696; Bioswamp), caspase-1 (1:1,000; cat. no. PAB36756; Bioswamp), TXNIP (1:1,000; cat. no. PAB43948; Bioswamp), TRX (1:1,000; cat. no. PAB32168; Bioswamp), NLRP3 (1:1,000; cat. no. PAB37930; Bioswamp) and β-actin (1:1,000; cat. no. PAB36265; Bioswamp). Subsequently, the membranes were treated at room temperature for 1 h using goat anti-rabbit IgG (1:20,000; cat. no. SAB43714; Bioswamp) labeled with horseradish peroxidase. The bands were detected by enhanced chemiluminescence (Analytik Jena AG). Tanon GIS software version 4.2 (Tanon Science & Technology Co., Ltd.) was used to determine the band intensity.

Enzyme-linked immunoassay (ELISA)

The BMSCs were treated as aforementioned, then the supernatants were collected, which were centrifuged at 1,000 × g at room temperature for 10 min. The expression of inflammatory mediators, including IL-1β, IL-6 and TNF-α, was evaluated. All steps were in accordance with the protocols of rat IL-1β (cat. no. RLB00), IL-6 (cat. no. R6000B) and TNF-α (cat. no. RTA00) ELISA kits (R&D Systems, Inc.). The absorbance of each group was evaluated at 450 nm with a microplate reader. Finally, the concentrations of the inflammatory mediators were measured according to a standard curve.

Statistics analysis

All data are expressed as the mean ± standard deviation. Data analysis was performed using SPSS 17.0 (SPSS, Inc.). For multiple comparisons, the differences in variables among groups were analyzed using one-way ANOVA followed by a Tukey's post hoc test. For comparisons between two groups, one-way ANOVA was used. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of NAC on the proliferation rate of LPS-mediated BMSCs

The cytotoxicity of NAC is shown in Fig. 1A. The results demonstrated no significant changes in the viability of BMSCs following pretreatment with NAC alone at various concentrations (0, 0.1, 0.5, 1 or 2 mM) for 24 h. In order to determine the most suitable concentration for BMSC proliferation to use in subsequent experiments, BMSCs were pretreated with NAC (0, 0.1, 0.5, 1, 2 mM) and incubated for 1 h prior to stimulation with LPS for 24 h. The results are shown in Fig. 1B. An NAC concentration of 1 mM had the strongest regulatory effect on the proliferation rate of BMSCs compared with no treatment (P<0.05 vs. control group); an increase in concentration from 1 to 2 mM NAC did not lead to a significant difference [LPS + NAC (1 mM) group vs. LPS + NAC (2 mM) group, P>0.05, P=0.957]. Thus, 1 mM NAC was selected for the subsequent assays.

NAC and Res decrease the levels of inflammatory mediators in LPS-stimulated BMSCs

To investigate whether NAC could inhibit inflammation in LPS-induced BMSCs, inflammatory mediators in response to NAC or Res, which is a signaling pathway inhibitor, were detected by ELISA kits. As shown in Fig. 2, expression of IL-1β, IL-6 and TNF-α was higher in BMSCs in the LPS group compared with the control group (P<0.05). At the same time, pretreatment with NAC for 1 h led to a significant decrease in inflammatory mediators (P<0.05). Similar results were observed in the Res + LPS and NAC + LPS groups (NAC + LPS vs. Res + LPS groups, P>0.05; IL-β, P=0.980; IL-6, P=0.961; TNF-α, P=0.876). Together, the results demonstrated that LPS could induce an inflammatory response, whereas NAC or Res pretreatment clearly suppressed LPS-triggered inflammation.

TXNIP/NLRP3 mediates the regulatory effects of NAC on inflammation in LPS-treated BMSCs

To further identify the potential molecular mechanism underlying the regulatory effects of NAC on LPS-mediated BMSCs, RT-qPCR and western blotting were conducted to evaluate the expression of mRNA and proteins, respectively, in LPS-treated BMSCs. According to the RT-qPCR results (Fig. 3), compared with the other groups, the LPS group (P<0.05) had the highest expression of ASC, NLRP3, caspase-1 and TXNIP, but the lowest expression of TRX. However, the effects of LPS on BMSCs were reversed with NAC pretreatment (P<0.05). Specifically, NAC pretreatment downregulated the expression of mRNAs increased by LPS and only upregulated the expression of TRX. In addition, when BMSCs were pretreated with Res, the same result as treatment with NAC was observed (NAC + LPS vs. Res + LPS, P>0.05; NLRP3, P=0.892; ASC, P=1.000; caspase-1, P=0.999; TXNIP, P=1.000; TRX, P=0.927). As shown in Fig. 4, the western blotting results were consistent with the mRNA data (P<0.05 vs. LPS group) (NAC + LPS vs. Res + LPS, P>0.05; NLRP3, P=0.394; ASC, P=0.998; caspase-1, P=0.954; TXNIP, P=0.912; TRX, P=1.000) (control vs. NAC + LPS, P>0.05; NLRP3, P=0.932; ASC, P=0.986; caspase-1, P=0.913; TXNIP, P=1.000; TRX, P=0.945) (control vs. Res + LPS, P>0.05; NLRP3, P=0.708; ASC, P=0.998; caspase-1, P=0.666; TXNIP, P=0.898; TRX, P=0.922). Briefly, pretreatment with NAC or Res reduced the expression of ASC, NLRP3, caspase-1 and TXNIP, but increased expression of TRX compared with cells only treated with LPS.

Discussion

The results of the present study demonstrated that NAC inhibited the LPS-induced inflammatory response in BMSCs by significantly mediating the downregulation of IL-1β, IL-6 and TNF-α, which were upregulated in response to LPS treatment. In addition, it was identified that following treatment with LPS, NLRP3, ASC, caspase-1 and TXNIP were upregulated, but TRX was downregulated in BMSCs. It was also identified that NAC downregulated the expression of ASC, NLRP3, caspase-1 and TXNIP, but enhanced the expression of TRX in LPS-induced BMSCs, which was similar to the effect of Res. Consequently, the results suggested that LPS induced inflammatory responses in BMSCs, which could be reversed by the anti-inflammatory activity of NAC via inhibition of the TXNIP/NLRP3/IL-1β signaling pathway (Fig. 5).

Recently, BMSCs have attracted increasing attention, due to their self-renewal ability and multidirectional differentiation potential (28). BMSCs are located in alveolar bone and are strongly associated with bone formation and resorption (29). It has been confirmed that the majority of osteoblasts colonizing the implant surface are generated from BMSCs, contributing significantly to osseointegration (30,31). Meanwhile, several studies have reported that BMSCs can be effectively used for the treatment of periodontitis (14,32,33). However, the viability and osteogenic differentiation of BMSCs can be downregulated in an inflammatory environment, which directly results in treatment failure (34). Thereby, controlling inflammation is essential for diseases associated with BMSCs.

A number of studies have demonstrated that LPS-induced overproduction of IL-1β, IL-6 and TNF-α contributes to the majority of inflammatory responses that are involved in the pathogenesis of inflammatory-stimulated oral bone diseases, including periodontitis (35), peri-implantitis (36) and apical periodontitis (37). By contrast, our previous studies identified that NAC inhibited LPS-mediated synthesis of IL-1β, IL-6 and TNF-α (10,26). Thus the present study focused on the secretion of IL-1β, IL-6 and TNF-α. It was demonstrated that in BMSCs, IL-1β, IL-6 and TNF-α were overexpressed following administration of LPS, leading to generation of inflammatory responses. Pretreatment with NAC reduced the secretion of IL-1β, IL-6 and TNF-α, and inhibited the inflammatory response. These findings revealed that NAC could be used to manage the inflammatory response.

The NLRP3 inflammasome is critical for secretion of the proinflammatory cytokine IL-1β (38). The present study demonstrated that NAC can reduce the secretion of IL-1β. Subsequent experiments determined whether NAC reduced IL-1β levels by inhibiting the NLRP3 inflammasome. The NLRP3 inflammasome, which contains NLRP3, ASC and caspase-1 (39), can initiate inflammation in response to infection of microbial products (40,41). It is widely recognized that LPS results in activation of the NLRP3 inflammasome (4244). Once activated, NLRP3 interacts with ASC, inducing the reciprocal identification of NLRP3 and pro-caspase-1, thereby leading to activation of caspase-1 (45,46). Immediately, caspase-1, also termed inflammatory caspase, mediates the maturation and secretion of IL-1β (47,48), which serves a significant role in inflammation (49,50). In the present study, protein and mRNA expression levels indicated that LPS mediated overexpression of the NLRP3 inflammasome in BMSCs. However, pretreatment with NAC attenuated the expression of mRNA and target proteins. These results clearly demonstrated that NAC downregulated the NLRP3 inflammasome and inhibited inflammatory responses caused by LPS.

The specific molecular mechanism by which NAC acts on the NLRP3 inflammasome and exhibits an anti-inflammatory effect remains to be elucidated. TXNIP is an essential multi-functional protein, which interferes with the function of TRX (51). The overexpression of TXNIP activates inflammatory pathways, in part since TRX-mediated inhibition of inflammation is reversed by TXNIP (51). Additionally, TXNIP directly activates the NLRP3 inflammasome, further stimulating inflammation (5254). Previous studies have suggested that, in response to stimulation, TXNIP dissociates from TRX and then directly combines with a specific region of NLRP3 (55,56). In addition, the incorporation of TXNIP and NLRP3 accelerates inflammasome aggregation and oligomerization, inducing transformation of caspase-1 and maturation of IL-1β (57). In the present study, LPS treatment induced the overproduction of TXNIP and decreased expression of TRX at the mRNA and protein levels. However, the expression of TXNIP was downregulated and that of TRX was upregulated by pretreatment with NAC. These observations further indicated that TXNIP and TRX are involved in the anti-inflammatory effect of NAC on LPS-induced BMSCs.

To clarify the underlying signaling pathway, Res was used as a pathway blocker. Previous studies have indicated that Res, a natural plant polyphenolic compound, can restrain NLRP3 inflammasome activation, which prevents the over-release of inflammatory cytokines (58,59). It was reported that Res can significantly inhibit the TXNIP/TRX cascade, restrain the expression of TXNIP, inhibit the incorporation of TXNIP and NLRP3 and finally block the inflammasome aggregation and oligomerization (60). As expected, in the present study, the effect of NAC was identified to be consistent with that of Res. Therefore, it was concluded that the inflammatory regulatory effects of NAC on LPS-induced BMSCs were closely associated with the TXNIP/NLRP3/IL-1β pathway.

On one hand, the results of the present study provided novel evidence that NAC can inhibit the inflammatory response in BMSCs stimulated by LPS; on the other hand, it demonstrated that one of the molecular mechanisms underlying the inflammatory effects of NAC on LPS-induced BMSCs was closely associated with the TXNIP/NLRP3/IL-1β pathway. These findings not only broadened our view of how NAC affects LPS-induced BMSCs but also provided an improved understanding of the underlying molecular mechanism that may promote the development of anti-inflammation strategies. However, in vivo experiments are required to validate these findings so we plan to establish animal models to further explore the anti-inflammatory effects of NAC in future research.

Acknowledgements

Not applicable.

Funding

This study was funded by a grant from Luzhou-Southwest Medical University (grant no. 2016LZXNYD-J21).

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

XW and LG were primarily responsible for the research protocol design. XW and MJ performed the experiments. XW, XH, BZ and WP analyzed and interpreted the data. XW drafted the manuscript. XW, LG and XH revised the manuscript. 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 

Berglundh T, Armitage G, Araujo MG, Avila-Ortiz G, Blanco J, Camargo PM, Chen S, Cochran D, Derks J, Figuero E, et al: Peri-implant diseases and conditions: Consensus report of workgroup 4 of the 2017 world workshop on the classification of periodontal and peri-implant diseases and conditions. J Clin Periodontol. 45 (Suppl 20):3299–S291. 2018. View Article : Google Scholar

2 

Kinane DF, Stathopoulou PG and Papapanou PN: Periodontal diseases. Nat Rev Dis Primers. 3:170382017. View Article : Google Scholar : PubMed/NCBI

3 

Eke PI, Borgnakke WS and Genco RJ: Recent epidemiologic trends in periodontitis in the USA. Periodontol 2000. 82:257–267. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Dreyer H, Grischke J, Tiede C, Eberhard J, Schweitzer A, Toikkanen SE, Glöckner S, Krause G and Stiesch M: Epidemiology and risk factors of peri-implantitis: A systematic review. J Periodontal Res. 53:657–681. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS, Dye BA and Genco RJ: Periodontitis in US adults: National health and nutrition examination survey 2009–2014. J Am Dent Assoc. 149:576–588.e6. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Derks J, Schaller D, Hakansson J, Wennstrom JL, Tomasi C and Berglundh T: Effectiveness of implant therapy analyzed in a Swedish population: Prevalence of peri-implantitis. J Dent Res. 95:43–49. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Monje A, Insua A and Wang HL: Understanding peri-implantitis as a plaque-associated and site-specific entity: On the local predisposing factors. J Clin Med. 8:2792019. View Article : Google Scholar

8 

Lamont RJ, Koo H and Hajishengallis G: The oral microbiota: Dynamic communities and host interactions. Nat Rev Microbiol. 16:745–759. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Singhrao SK and Olsen I: Assessing the role of Porphyromonas gingivalis in periodontitis to determine a causative relationship with Alzheimer's disease. J Oral Microbiol. 11:15634052019. View Article : Google Scholar : PubMed/NCBI

10 

Wang L, Yang Y, Xiong X, Yu T, Wang X, Meng W, Wang H, Luo G and Ge L: Oral lichen-planus-associated fibroblasts acquire myofibroblast characteristics and secrete pro-inflammatory cytokines in response to Porphyromonas gingivalis lipopolysaccharide stimulation. BMC Oral Health. 18:1972018. View Article : Google Scholar : PubMed/NCBI

11 

Guo L, Zhang H, Li W, Zhan D and Wang M: N-acetyl cysteine inhibits lipopolysaccharide-mediated induction of interleukin-6 synthesis in MC3T3-E1 cells through the NF-κB signaling pathway. Arch Oral Biol. 93:149–154. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Zhang ZZ, Xiong T, Zheng R, Huang JL and Guo L: N-acetyl cysteine protects HUVECs against lipopolysaccharide-mediated inflammatory reaction by blocking the NF-κB signaling pathway. Mol Med Rep. 20:4349–4357. 2019.PubMed/NCBI

13 

Huang JL, Xiong T, Zhang ZZ, Tan YJ and Guo L: Inhibition of the receptor for advanced glycation inhibits lipopolysaccharide-mediated High mobility group protein B1 and Interleukin-6 synthesis in human gingival fibroblasts through the NF-κB signaling pathway. Arch Oral Biol. 105:81–87. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Yu X, Quan J, Long W, Chen H, Wang R, Guo J, Lin X and Mai S: LL-37 inhibits LPS-induced inflammation and stimulates the osteogenic differentiation of BMSCs via P2X7 receptor and MAPK signaling pathway. Exp Cell Res. 372:178–187. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Čebatariūnienė A, Kriaučiūnaitė K, Prunskaitė J, Tunaitis V and Pivoriūnas A: Extracellular vesicles suppress basal and lipopolysaccharide-induced NFκB activity in human periodontal ligament stem cells. Stem Cells Dev. 28:1037–1049. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Lian D, Dai L, Xie Z, Zhou X, Liu X, Zhang Y, Huang Y and Chen Y: Periodontal ligament fibroblasts migration injury via ROS/TXNIP/Nlrp3 inflammasome pathway with Porphyromonas gingivalis lipopolysaccharide. Mol Immunol. 103:209–219. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Qiu Z, He YH, Ming H, Lei SQ, Leng Y and Xia ZY: Lipopolysaccharide (LPS) aggravates high glucose- and hypoxia/reoxygenation-induced injury through activating ROS-dependent NLRP3 inflammasome-mediated pyroptosis in H9C2 cardiomyocytes. J Diabetes Res. 2019:81518362019. View Article : Google Scholar : PubMed/NCBI

18 

Tang YS, Zhao YH, Zhong Y, Li XZ, Pu JX, Luo YC and Zhou QL: Neferine inhibits LPS-ATP-induced endothelial cell pyroptosis via regulation of ROS/NLRP3/caspase-1 signaling pathway. Inflamm Res. 68:727–738. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Raudales JLM, Yoshimura A, Ziauddin SM, Kaneko T, Ozaki Y, Ukai T, Miyazaki T, Latz E and Hara Y: Dental calculus stimulates interleukin-1β secretion by activating NLRP3 inflammasome in human and mouse phagocytes. PLoS One. 11:e01628652016. View Article : Google Scholar : PubMed/NCBI

20 

Shibata K: Historical aspects of studies on roles of the inflammasome in the pathogenesis of periodontal diseases. Mol Oral Microbiol. 33:203–211. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C, McDowell J, Paskind M, Rodman L, Salfeld J, et al: Mice deficient in IL-l beta-converting enzyme are defective in production of mature IL-l beta and resistant to endotoxic shock. Cell. 80:401–411. 1995. View Article : Google Scholar : PubMed/NCBI

22 

Neuwelt EA, Pagel MA, Hasler BP, Deloughery TG and Muldoon LL: Therapeutic efficacy of aortic administration of N-acetylcysteine as a chemoprotectant against bone marrow toxicity after intracarotid administration of alkylators, with or without glutathione depletion in a rat model. Cancer Res. 61:7868–7874. 2001.PubMed/NCBI

23 

Rosic G, Selakovic D, Joksimovic J, Srejovic I, Zivkovic V, Tatalović N, Orescanin-Dusic Z, Mitrovic S, Ilic M and Jakovljevic V: The effects of N-acetylcysteine on cisplatin-induced changes of cardiodynamic parameters within coronary autoregulation range in isolated rat hearts. Toxicol Lett. 242:34–46. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Kelly GS: Clinical applications of N-acetylcysteine. Altern Med Rev. 3:114–127. 1998.PubMed/NCBI

25 

Shih WL, Chang CD, Chen HT and Fan KK: Antioxidant activity and leukemia initiation prevention in vitro and in vivo by N-acetyl-L-cysteine. Oncol Lett. 16:2046–2052. 2018.PubMed/NCBI

26 

Zheng R, Tan YJ, Gu MQ, Kang T, Zhang H and Guo L: N-acetyl cysteine inhibits lipopolysaccharide-mediated synthesis of interleukin-1β and tumor necrosis factor-α in human periodontal ligament fibroblast cells through nuclear factor-kappa B signaling. Medicine (Baltimore). 98:e171262019. View Article : Google Scholar : PubMed/NCBI

27 

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. View Article : Google Scholar : PubMed/NCBI

28 

Huang C, Geng J and Jiang S: MicroRNAs in regulation of osteogenic differentiation of mesenchymal stem cells. Cell Tissue Res. 368:229–238. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Arvidson K, Abdallah BM, Applegate LA, Baldini N, Cenni E, Gomez-Barrena E, Granchi D, Kassem M, Konttinen YT, Mustafa K, et al: Bone regeneration and stem cells. J Cell Mol Med. 15:718–746. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Hu D, Li K, Xie Y, Pan H, Zhao J, Huang L and Zheng X: The combined effects of nanotopography and Sr ion for enhanced osteogenic activity of bone marrow mesenchymal stem cells (BMSCs). J Biomater Appl. 31:1135–1147. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Heng BC, Cao T, Stanton LW, Robson P and Olsen B: Strategies for directing the differentiation of stem cells into the osteogenic lineage in vitro. J Bone Miner Res. 19:1379–1394. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Lu L, Liu Y, Zhang X and Lin J: The therapeutic role of bone marrow stem cell local injection in rat experimental periodontitis. J Oral Rehabil. Jun 20–2019.(Epub ahead of print). doi: 10.1111/joor.12843. View Article : Google Scholar

33 

Han N, Zhang F, Li G, Zhang X, Lin X, Yang H, Wang L, Cao Y, Du J and Fan Z: Local application of IGFBP5 protein enhanced periodontal tissue regeneration via increasing the migration, cell proliferation and osteo/dentinogenic differentiation of mesenchymal stem cells in an inflammatory niche. Stem Cell Res Ther. 8:2102017. View Article : Google Scholar : PubMed/NCBI

34 

Wu L, Zhang G, Guo C, Zhao X, Shen D and Yang N: MiR-128-3p mediates TNF-α-induced inflammatory responses by regulating Sirt1 expression in bone marrow mesenchymal stem cells. Biochem Biophys Res Commun. 521:98–105. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Zhang L and Deng S: Effects of astragaloside IV on inflammation and immunity in rats with experimental periodontitis. Braz Oral Res. 33:e0322019. View Article : Google Scholar : PubMed/NCBI

36 

Li H, Chen Z, Zhong X, Li J and Li W: Mangiferin alleviates experimental peri-implantitis via suppressing interleukin-6 production and toll-like receptor 2 signaling pathway. J Orthop Surg Res. 14:3252019. View Article : Google Scholar : PubMed/NCBI

37 

Cosme-Silva L, Dal-Fabbro R, Cintra LTA, dos Santos VR, Duque C, Ervolino E, Bomfim SM and Gomes-Filho JE: Systemic administration of probiotics reduces the severity of apical periodontitis. Int Endod J. 52:1738–1749. 2019. View Article : Google Scholar : PubMed/NCBI

38 

He Y, Hara H and Núñez G: Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 41:1012–1021. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Guo H, Callaway JB and Ting JP: Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat Med. 21:677–687. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Shin S and Brodsky IE: The inflammasome: Learning from bacterial evasion strategies. Semin Immunol. 27:102–110. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Place DE and Kanneganti TD: Recent advances in inflammasome biology. Curr Opin Immunol. 50:32–38. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD and Latz E: Targeting the NLRP3 inflammasome in inflammatory diseases. Nat Rev Drug Discov. 17:588–606. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Baker PJ, Boucher D, Bierschenk D, Tebartz C, Whitney PG, D'Silva DB, Tanzer MC, Monteleone M, Robertson AAB, Cooper MA, et al: NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5. Eur J Immunol. 45:2918–2926. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Feng H, Gu J, Gou F, Huang W, Gao C, Chen G, Long Y, Zhou X, Yang M, Liu S, et al: High glucose and lipopolysaccharide prime NLRP3 inflammasome via ROS/TXNIP pathway in mesangial cells. J Diabetes Res. 2016:69731752016. View Article : Google Scholar : PubMed/NCBI

45 

Xiaoyu H, Si H, Li S, Wang W, Guo J, Li Y, Cao Y, Fu Y and Zhang N: Induction of heme oxygenas-1 attenuates NLRP3 inflammasome activation in lipopolysaccharide-induced mastitis in mice. Int Immunopharmacol. 52:185–190. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Bolívar BE, Vogel TP and Bouchier-Hayes L: Inflammatory caspase regulation: Maintaining balance between inflammation and cell death in health and disease. FEBS J. 286:2628–2644. 2019.PubMed/NCBI

47 

Martinon F, Burns K and Tschopp Jr: The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 10:417–426. 2002. View Article : Google Scholar : PubMed/NCBI

48 

Lu WL, Song DZ, Yue JL, Wang TT, Zhou XD, Zhang P, Zhang L and Huang DM: NLRP3 inflammasome may regulate inflammatory response of human periodontal ligament fibroblasts in an apoptosis-associated speck-like protein containing a CARD (ASC)-dependent manner. Int Endod J. 50:967–975. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Dinarello CA: A clinical perspective of IL-1β as the gatekeeper of inflammation. Eur J Immunol. 41:1203–1217. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Mantovani A, Dinarello CA, Molgora M and Garlanda C: Interleukin-1 and related cytokines in the regulation of inflammation and immunity. Immunity. 50:778–795. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Devi TS, Lee I, Hüttemann M, Kumar A, Nantwi KD and Singh LP: TXNIP links innate host defense mechanisms to oxidative stress and inflammation in retinal Muller glia under chronic hyperglycemia: Implications for diabetic retinopathy. Exp Diabetes Res. 2012:1–19. 2012. View Article : Google Scholar

52 

Wang CY, Xu Y, Wang X, Guo C, Wang T and Wang ZY: Dl-3-n-Butylphthalide inhibits NLRP3 inflammasome and mitigates Alzheimer's-like pathology via Nrf2-TXNIP-TrX axis. Antioxid Redox Signal. 30:1411–1431. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Hou Y, Wang Y, He Q, Li L, Xie H, Zhao Y and Zhao J: Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury. Behav Brain Res. 336:32–39. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Chen W, Zhao MJ, Zhao SZ, Lu QY, Ni LS, Zou C, Lu L, Xu X, Guan HJ, Zheng Z and Qiu QH: Activation of the TXNIP/NLRP3 inflammasome pathway contributes to inflammation in diabetic retinopathy: A novel inhibitory effect of minocycline. Inflamm Res. 66:157–166. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Liu Y, Lian K, Zhang L, Wang R, Yi F, Gao C, Xin C, Zhu D, Li Y, Yan W, et al: TXNIP mediates NLRP3 inflammasome activation in cardiac microvascular endothelial cells as a novel mechanism in myocardial ischemia/reperfusion injury. Basic Res Cardiol. 109:4152014. View Article : Google Scholar : PubMed/NCBI

56 

Zhang X, Zhang JH, Chen XY, Hu QH, Wang MX, Jin R, Zhang QY, Wang W, Wang R, Kang LL, et al: Reactive oxygen species-induced TXNIP drives fructose-mediated hepatic inflammation and lipid accumulation through NLRP3 inflammasome activation. Antioxid Redox Signal. 22:848–870. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Ma Z and Damania B: Editorial: NLRP3: Immune activator or modulator? J Leukoc Biol. 99:641–643. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Jiang L, Zhang L, Kang K, Fei D, Gong R, Cao Y, Pan S and Zhao M and Zhao M: Resveratrol ameliorates LPS-induced acute lung injury via NLRP3 inflammasome modulation. Biomed Pharmacother. 84:130–138. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Chang YP, Ka SM, Hsu WH, Chen A, Chao LK, Lin CC, Hsieh CC, Chen MC, Chiu HW, Ho CL, et al: Resveratrol inhibits NLRP3 inflammasome activation by preserving mitochondrial integrity and augmenting autophagy. J Cell Physiol. 230:1567–1579. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Feng L and Zhang L: Resveratrol suppresses Aβ-induced microglial activation through the TXNIP/TRX/NLRP3 signaling pathway. DNA Cell Biol. 38:874–879. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 22 Issue 4

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
Wang X, Jiang M, He X, Zhang B, Peng W and Guo L: N‑acetyl cysteine inhibits the lipopolysaccharide‑induced inflammatory response in bone marrow mesenchymal stem cells by suppressing the TXNIP/NLRP3/IL‑1β signaling pathway. Mol Med Rep 22: 3299-3306, 2020
APA
Wang, X., Jiang, M., He, X., Zhang, B., Peng, W., & Guo, L. (2020). N‑acetyl cysteine inhibits the lipopolysaccharide‑induced inflammatory response in bone marrow mesenchymal stem cells by suppressing the TXNIP/NLRP3/IL‑1β signaling pathway. Molecular Medicine Reports, 22, 3299-3306. https://doi.org/10.3892/mmr.2020.11433
MLA
Wang, X., Jiang, M., He, X., Zhang, B., Peng, W., Guo, L."N‑acetyl cysteine inhibits the lipopolysaccharide‑induced inflammatory response in bone marrow mesenchymal stem cells by suppressing the TXNIP/NLRP3/IL‑1β signaling pathway". Molecular Medicine Reports 22.4 (2020): 3299-3306.
Chicago
Wang, X., Jiang, M., He, X., Zhang, B., Peng, W., Guo, L."N‑acetyl cysteine inhibits the lipopolysaccharide‑induced inflammatory response in bone marrow mesenchymal stem cells by suppressing the TXNIP/NLRP3/IL‑1β signaling pathway". Molecular Medicine Reports 22, no. 4 (2020): 3299-3306. https://doi.org/10.3892/mmr.2020.11433