Open Access

The c‑Jun signaling pathway has a protective effect on nucleus pulposus cells in patients with intervertebral disc degeneration

  • Authors:
    • Ming Lei
    • Kun Wang
    • Shuai Li
    • Kangcheng Zhao
    • Wenbin Hua
    • Xinghuo Wu
    • Cao Yang
  • View Affiliations

  • Published online on: September 21, 2020     https://doi.org/10.3892/etm.2020.9251
  • Article Number: 123
  • Copyright: © Lei 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

Among a range of diverse clinical symptoms, intervertebral disc degeneration (IDD) contributes mostly to the onset of lower back pain. The present study aimed to investigate the effects of c‑Jun on nucleus pulposus (NP) cells of IDD and its regulation on molecular mechanisms. Intervertebral disc (IVD) tissues were collected from patients suffering from IDD disease, and NP cells were subsequently isolated and cultured. By overexpressing c‑Jun in NP cells, expression levels of mRNAs and proteins of IDD‑related genes and inflammatory cytokines were subjected to reverse transcription‑quantitative PCR, western blot and ELISA assays. Additional transforming growth factor‑β (TGF‑β) antibodies were administrated to suppress the function of TGF‑β. Cell proliferation and apoptosis were determined via Cell Counting Kit‑8 and TUNEL assays, respectively. The results demonstrated that the overexpression of c‑Jun robustly upregulated both mRNA and protein expression of TGF‑β, TIMP metallopeptidase inhibitor 3, aggrecan and collagen type II alpha 1 chain and simultaneously downregulated the expression of the inflammatory cytokines TNF‑α, interleukin (IL)‑1β, IL‑6 and IL‑17. Furthermore, following c‑Jun overexpression, survival rates of NP cells were increased while apoptosis rates were decreased. However, the addition of a TGF‑β antibody significantly promoted apoptosis and restricted cell survival, which differed from the results of the c‑Jun overexpression group. The present study hypothesized therefore that c‑Jun may positively regulate TGF‑β expression within NP cells of IDD, which could promote the proliferation of IDD‑NP cells and accelerate cell viability via reducing apoptosis and the inflammatory response.

Introduction

The intervertebral disc (IVD) is composed of nucleus pulposus (NP), anulus fibrosus and cartilage endplate (1). Back pain is mostly attributable to the degeneration of IVD, which may cause nerve damage, poor labor capacity and low quality of life in some severe cases (1). Despite intervertebral disc degeneration (IDD) development being a natural process that comes with aging, some evidence suggests that the development of IDD stems from both genetic and environmental factors (2). For example, certain factors, including the decrease of NP cells, imbalance between the synthesis and degradation of the extracellular matrix (ECM) and overexpression of inflammatory cytokines, are known to be responsible for accelerating the development of IDD (3).

NP cell-produced collagen type II alpha 1 chain (COL2A1), aggrecan (ACAN) and some ECM components are believed to be crucial to maintain the integrity of IVD (4,5). It has been reported that some molecules, such as insulin-like growth factor-1, epidermal growth factor and transforming growth factor-β (TGF-β), can promote anabolic metabolism of ECM (6,7). Tumor necrosis factor (TNF)-α, interleukins (IL), matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin type I motifs (ADAMTS) work synergistically and facilitate the catabolism of the ECM (8-10). Catabolic activities of MMPs and ADAMTSs were leveraged by inhibitory actions and through tissue inhibitors of metalloproteinases (TIMPs) (10). In NP cells, TGF-β can delay the process of IDD via promoting the synthesis of ACAN and COL2A1, which enhances cell proliferation and inhibits MMPs expression (6,11). IDD NP cells also produced diverse inflammatory mediators which caused the aggravation of IDD, such as TNF-α, IL-1 α/β, IL-6 and IL-17(12).

As a major component of activator protein-1 (AP-1), c-Jun expression is increased when cells enter the logarithmic phase of growth in the presence of growth-factor-induced stimulation (13). Wisdom et al (14) demonstrated that c-Jun stimulates fibroblast growth and inhibits its apoptosis. Furthermore, c-Jun was found to protect cells from TNF-α-induced apoptosis, which is required for cell proliferation (14). Conversely, the presence and phosphorylation-based activation of c-Jun are necessary for the execution of apoptosis in both neuronal cells and thymocytes (15). In the intestinal ischemia-reperfusion injured autograft model, activation of both c-Fos and c-Jun genes can trigger cell proliferation and apoptosis (16).

Behrens et al (17) reported that a lack of c-Jun in mice can lead to the impairment of hepatocyte proliferation and liver regeneration. Notochordal cells in the absence of c-Jun have also been demonstrated to experience an increase in apoptosis, leading to impairment of IVD formation (18). Subsequently, it was speculated that c-Jun expression may have an essential role in IDD process. However, the function and underlying mechanisms of c-Jun in IDD remain unknown. Because the present study aimed to explore the role of c-Jun in NP cells, NP cells were transduced with a c-Jun-overexpressing lentivirus, and changes in IVD-related genes on a molecular level were detected. This study attempted to further elucidate the pathogenesis of IDD and provide a novel addition to valuable clinical information for the treatment of IDD.

Materials and methods

IVD tissue collection

IVD tissues were collected as surgical waste from 10 patients with IDD (age, 35-58 years). In addition, IVD tissues from 10 patients with lumbar fractures (age, 26-52 years), excluding those with spinal tumors, infections and rheumatic immune diseases, were collected as controls. This study was approved by the Institutional Review Board of Tongji Medical College and followed the Declaration of Helsinki. Written informed consent was obtained from each patient. According to the MRI scanning techniques reported by Pfirrmann et al (19), the obtained IVD tissues were graded by T2-weighted images to determine degrees of degeneration. Relative normal nondegenerated discs from patients with lumbar fractures were graded I-II (Control), whereas degenerative discs from patients with IDD were graded III-V. Subsequently, NP cells were isolated from IVD tissues of patients with IDD and control subjects.

Isolation and culture NP cells

Primary NP cells were isolated and cultured as previously reported (5). The IDD and control NP tissue samples were washed three times with D-Hanks solution under aseptic conditions. These specimens were cut into 1-mm3 pieces, and digested with 0.25% trypsin (Beyotime Institute of Biotechnology) and 0.2% collagenase II (Beyotime Institute of Biotechnology) for 3 h at 37˚C. NP cells were filtered through a 200-mesh sieve, washed three times with PBS and the supernatant was discarded following centrifugation at 2,000 x g for 5 min (37˚C). Cells were washed with DMEM-F12 (Gibco; Thermo Fisher Scientific, Inc.) medium containing 10% FBS to terminate digestion. After centrifugation at 2,000 x g for 5 min (37˚C), NP cells were counted and seeded into 25 cm2 culture dishes. DMEM-F12 medium was supplemented with 15% FBS (Gibco; Thermo Fisher Scientific, Inc.), 10 µg/ml insulin and 1% penicillin/streptomycin (Invitrogen; Thermo Fisher Scientific, Inc.) was used to culture NP cells under conventional incubation conditions (37˚C, 5% CO2 and 95% humidity), and the medium was refreshed twice a week. When NP confluency reached 80%, cells were passaged at a ratio of 1:2. Cells in passage P2 were used for subsequent experiments.

Lentiviral vector construction and lentivirus infection of NP cells

The c-Jun gene was inserted into green fluorescence protein (GFP)-labeled LV5 plasmids obtained from Boaimaidisen Biotechnology Co., Ltd. and the lentivirus was packaged by four plasmid systems namely LV5-c-Jun, PG-p1-VSVG, PG-P2-REV and PG-P3-RRE (Boaimaidisen Biotechnology Co., Ltd.). Transfections were performed into 293T packaging cell lines (American Type Culture Collection). Lentiviral packaging enrichment was completed by Chongqing Biomedicine Biotechnology Co., Ltd. The GFP-labeled blank LV5-empty vector was used as the negative control. The viral titer of LV5-c-Jun and LV5-empty vector lentivirus was 1x108 TU/ml.

Before lentiviral transfection, a preliminary test was carried out to estimate the efficiency of lentiviral infection in target cells. The multiplicity of infection (MOI) refers to the proportion of infectious viruses per cells and is calculated as follows: MOI=(virus titer x virus volume)/number of cells. NP cells were seeded into 24-well culture plate at the density of 3x104/well. 293T cells served as the control cells in a parallel experiment to determine the affinity of lentivirus to target cells. Transfection was conducted when the confluency reached 70%. The infectious lentiviruses were incubated with the NP cells and control cells at a final MOI of 0, 10, 20, 40 and 100. The mixed lentivirus and cells were incubated in a incubator (37˚C, 5% CO2) overnight. After 24 h, culture medium containing lentivirus was replaced with normal culture medium. At 4 days following infection, the transduction efficiency was tested via flow cytometry according to GFP-positive cells (BD Diagnostics). GFP expression was observed in cellSens 1.12 software (Olympus Corporation) at a magnification of x100 using an inverted fluorescence microscope (CKX53, Olympus Corporation).

Western blotting

NP cells were lysed in RIPA reagent (Beyotime Institute of Biotechnology), and the concentration of extracted proteins was measured by BCA standard method (Takara Biotechnology Co., Ltd.). Proteins (30 µg) were separated by 10% SDS-PAGE and transferred onto PVDF membranes (EMD Millipore). PVDF membranes were washed with TBST (0.05% Tween-20) three times before being blocked with 5% skimmed milk at room temperature for 2 h. Primary antibodies (1:500) were incubated overnight at 4˚C. Subsequently, the HRP-conjugated goat anti-rabbit antibody was incubated for 1.5 h at room temperature (1:1,500, ab205718; Abcam). The proteins were visualized by chemiluminescence (Thermo Fisher Scientific, Inc.,). Densitometry was semi-quantified with ImageJ 1.8.0 software (National Institutes of Health). Results were normalized by GAPDH. The following primary antibodies were used: Anti-c-Jun (cat. no. ab40766), anti-TGF-β (cat. no. ab31013), anti-TIMP-1 (cat. no. ab211926), anti-TIMP-3 (cat. no. ab39184), anti-ADAMTS-4 (cat. no. ab185722), anti-ADAMTS-5 (cat. no. ab41037), anti-ACAN (cat. no. ab3778), anti-COL1A1 (cat. no. ab34710), anti-COL2A1 (cat. no. ab34712), anti-IL-1β (cat. no. ab2105), anti-IL-6 (cat. no. ab6672), anti-IL-17 (cat. no. ab79056), anti-TNF-α (cat. no. ab6671), anti-GAPDH (cat. no. ab181602) (all from Abcam).

Reverse transcription-quantitative PCR (RT-qPCR) assay

Total RNA was isolated from NP cells using TRIzol® reagent (Beyotime Institute of Biotechnology) according to the manufacturer's instructions. Reverse transcription was carried out using the PrimeScript™ RT reagent kit with gDNA Eraser (Takara Bio, Inc.), according to the manufacturer's instructions. The reverse transcription reactions occurred as follows: 37˚C for 15 min, 85˚C for 5 sec, 4˚C for 5 min. RT-qPCR analysis was performed on an ABI 7500 instrument (Thermo Fisher Scientific, Inc.). All reaction systems and procedures of RT-qPCR were conducted following the manufacturer's protocol of TB Green Premix Ex Taq II (Takara Bio, Inc.). Reaction protocol was set with three-step cycling conditions: 95˚C for 30 sec, followed by 40 cycles of 95˚C for 5 sec, 60˚C for 30 sec and 72˚C for 15 sec. The melting curve stage was included, ramping from 65 to 95˚C (increment 0.5˚C/5 sec) to verify the specificity of the primer amplification based on the presence of a single and sharp peak. All primer sequences are listed in Table SI. The 2-ΔΔCq method was used to calculate the relative expression levels (20).

ELISA assay

After the c-Jun- and empty vector-transfected NP cells were cultured, the supernatants were collected into sterile centrifuge tube. The expression levels of inflammatory factors, including TNF-α (cat. no. P01P0087; Shanghai BlueGene Biotech Co., Ltd.), IL-1β (cat. no. E01I0010; Shanghai BlueGene Biotech Co., Ltd.), IL-6 (cat. no. E01I0006; Shanghai BlueGene Biotech Co., Ltd.) and IL-17 (cat. no. E01I0362; Shanghai BlueGene Biotech Co., Ltd.) were measured by ELISA according to the manufacturer's instructions.

Cell Counting Kit-8 (CCK8) and TUNEL assay

According to the CCK-8 (Sigma-Aldrich; Merck KGaA) manufacturer's instructions, 100 µl/well cell suspension was seeded into a 96-well plate (1x104 cells/well). After being cultured for 24 h at 37˚C, c-Jun-mediated cells were treated with TGF-β neutralizing antibody (2 µg/ml; cat. no. AB-100-NA; R&D Systems, Inc.) and isotype-matched control IgG (2 µg/ml; cat. no. AB-108-C; R&D Systems, Inc.). Upon 12 h of incubation, 10 µl CCK8 solution was added to each well and for 2 h. The absorbance was measured at 450 nm on a microplate reader.

For the TUNEL assay, cells were fixed with 4% paraformaldehyde for 20 min at room temperature. Cells were washed three times with PBS and cells were stained according to the manufacturer's instructions of the TUNEL kit [cat. no. 11684817910; Roche Diagnostics (Shanghai) Co., Ltd.]. Apoptotic cell was observed at a magnification of x100 using an inverted fluorescence microscope (cat. no. CKX53; Olympus Corporation), each sample was randomly counted in four fields, with red representing apoptotic cells and blue representing nucleus pulposus. The apoptosis rate was the percentage of red fluorescent cells to blue fluorescent cells, and the average value was taken as the final apoptosis rate of nucleus pulposus cells.

Statistical analysis

Data were presented as the means ± standard deviation. Each experiment was repeated three times independently. Statistical analyses were performed using SPSS 23.0 (IBM Corp.). Comparisons among multiple groups were made with one-way analysis of variance (ANOVA) followed by a Tukey's post hoc test and differences between two groups were identified with a Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Cell isolation and lentivirus transfection

The present study demonstrated that NP cells were successfully isolated from both normal and IDD tissues. Briefly, following c-Jun overexpression, GFP expression in normal NP cells displayed 25, 63, 78 and 93% efficiencies at MOI values of 10, 20, 40 and 100, respectively. When MOI values were set at 10, 20, 40 and 100, the GFP expression efficiency in the IDD NP cells exhibited 26, 47, 74 and 88%, respectively. These results indicated that cell expressed the transfected GFP most efficiently when the MOI value was set at 100 (Fig. 1A and B).

Detection of c-Jun expression by western blotting and RT-qPCR

The results from western blotting indicated that c-Jun expression was the highest when the MOI was set at 100 for both normal and IDD NP cells (Fig. 1C and D). A similar increase in c-Jun expression was observed by RT-qPCR between the normal and IDD groups (Fig. 1E). These results demonstrated a successful overexpression of c-Jun in NP cells. Notably, the data demonstrated that c-Jun expression was lower in IDD NP cells compared with normal NP cells (Fig. 1C-E).

c-Jun overexpression upregulates the genes associated with ameliorating IDD

The expression levels of IDD-related factors were detected by western blotting and RT-qPCR following transduction (MOI 100 PFU/cell) and subsequent cell culture for 3 and 7 days in the normal and IDD NP cells. On day 3, a comparison was made between the c-Jun-overexpressed IDD NP cells and the empty vector-transfected cells. The results demonstrated that c-Jun-overexpression groups exhibited significant increases in the mRNA expression of TGF-β, TIMP-3, ADAMTS-4, ACAN and COL2A1, and a significant decrease in COL1A1 expression, while no significant changes were detected in TIMP-1 and ADAMTS-5 expression (Fig. 2A). On day 7, expression levels of TGF-β, ACAN and COL2A1 were significantly upregulated after c-Jun overexpression when compared with the control. The effect of c-Jun on NP cells was most evident on the third day; consequently, the cells transfected for 3 days were selected for subsequent protein detection (Fig. 2B and C). The results suggested that expression levels of TGF-β, TIMP-3, ACAN and COL2A1 were significantly increased after c-Jun overexpression in the degenerative NP cells. Taken together, these data demonstrated that c-Jun overexpression may promote the expression of proteins associated with IDD amelioration.

c-Jun overexpression downregulates the expression of inflammatory cytokines

The expression levels of TNF-α, IL-1β, IL-6 and IL-17 were detected by RT-qPCR, western blotting and ELISA. The results indicated a significant decrease in mRNA and protein expression of TNF-α, IL-1β, IL-6 and IL-17 following c-Jun overexpression in normal and IDD NP cells (Figs. 2B, 3A-E). In addition, the expression levels of inflammatory cytokines in IDD NP cells were higher than those in normal healthy NP cells. Similar concentration-dependent trends were observed in the ELISA assays. Furthermore, both in normal and IDD NP cells, the concentration of inflammatory factors was significantly lower than those in the empty-vector transfected cells following c-Jun overexpression (Fig. 3F-I). These findings suggested that the overexpression of c-Jun inhibited the expressions of inflammatory cytokines.

Effects of TGF-β antibody on c-Jun-regulated cell

A previous study reported that c-Jun can activate TGF-β-induced transcription (21). To clarify the roles of TGF-β in c-Jun-regulated cell proliferation and apoptosis, cells were treated with TGF-β antibody to inhibit TGF-β activity, and the the survival and apoptosis rates of degenerated cells were analyzed by CCK8 and TUNEL assays, respectively. Survival rates in the empty vector-transfected and the c-Jun transfected cells were 100 and 103.15%, respectively. The survival rates manifested differently following the addition of TGF-β antibody, which significantly decreased cell survival to 86.42%, compared with 103.28% in the c-Jun + IgG1 group (Fig. 4A). These results indicated that TGF-β antibody was capable of decreasing cell proliferation and inducing cell death in c-Jun-overexpressed NP cells. Consequently, the apoptosis rates of empty vector-transfected group, c-Jun transfected group, c-Jun + TGF-β antibody group and c-Jun + IgG1 control group were evaluated, and the results demonstrated that the apoptosis rates were 8.15, 7.15, 21.25 and 5.48%, respectively (Fig. 4B and C). The apoptosis rates increased significantly following TGF-β antibody addition, indicating that TGF-β could inhibit the apoptosis following c-Jun overexpression within IDD NP cells.

Discussion

As a member of the AP-1 transcription factor family, c-Jun has a functional role in various cellular responses to extracellular stimuli (13,22). By functioning as a key transcription factor, AP-1 was reported to regulate cell survival and cell death pathways, initiate gene transcription as a molecular switch, and was demonstrated to be involved in relevant cellular processes including proliferation, apoptosis and inflammation (22). In c-Jun-deficient mice, the IVD formation was impaired (18). In addition, IVD cell clusters produce certain ECM components, allowing the repair of damaged tissues with c-Jun as a major protein expressed (23). It was therefore hypothesized that c-Jun could have crucial effects on IVD tissues. However, the role of c-Jun in the progression of IDD remains unknown, in particular the its effects on NP cell proliferation, apoptosis and target gene regulation in vitro. The results from the present study demonstrated that the mRNA and protein expression of c-Jun in normal NP cells was higher compared with that in IDD NP cells. Furthermore, c-Jun had a positive effect on NP cells in promoting cell proliferation, reducing cell apoptosis, increasing ECM synthesis and downregulating inflammatory factors.

A previous study demonstrated that inflammatory mediators serve crucial roles in IVD and could contribute to the IDD process (24). The expression levels of inflammatory factors IL-1β, IL-6, IL-17 and TNF-α in the degenerated disc tissue were distinctly increased compared with normal disc tissue, and their expressions was positively correlated with the degree of disc degeneration, including IL-1β, IL-6, IL-17 and TNF-α (25). In addition, AP-1 is considered as a pro-inflammatory factor that can directly regulate the expression of ILs and MMPs (26). The results from the present study suggested that NP cells overexpressin c-Jun exhibited a dramatic decrease in the expression of TNF-α, IL-1β, IL-6 and IL-17, suggesting that c-Jun may exert an anti-inflammatory function in NP cells. However, AP-1 dimers composed of different protein subunits processed different biological functions, and Jun family members exhibit different functional properties as transcription factors (27). Li et al (28) reported that IL-17a can enhance the expression of cyclooxygenase 2 and prostaglandin E2 in NP cells, leading to the regulation of inflammatory responses through p38/c-Fos and c-Jun n-terminal kinase (JNK)/c-Jun pathways. Duval et al (29) reported that the JNK pathway could downregulate the production of TNF-α. JNK is mainly known to regulate the phosphorylation of c-Jun, which regulates the production of more c-Jun, along with other genes (30). Inflammatory cytokines induce the expression of c-Jun; however, the specific mechanism of how the inflammatory cytokines control the overexpression of c-Jun requires further research.

Previous studies reported that the cytokine TGF-β can delay and repair IDD by upregulating ACAN and COL2A1 expression, therefore promoting NP cell proliferation and increasing ECM synthesis (11,31,32). Furthermore, the proto-oncogene c-myc serves an active role in NP cell proliferation and cycle progression under TGF-β stimulation (33). Interestingly, c-Jun belongs to proto-oncogene family of nuclear transcription factors, and it forms a complex with promoters of different genes to regulate transcription (23). Numerous growth-related cytokines contain 12-O-tetradecanoylphorbol-13-acetate response element (TRE) binding sites, including basic fibroblast growth factor and TGF-β, the dephosphorylated AP-1 is able to recognize the TRE binding sites and initiate transcription of these cytokines (34). The Smad3-Smad4 heterodimeric complex cooperate with c-Jun/c-Fos to induce transcriptional activation in response to TGF-β (21,35). Significant increase in TGF-β mRNA and protein expression was observed in NP cells following c-Jun overexpression, and an upregulation was also observed for the cytokines TIMP-3, ACAN and COL2A1, which are associated with amelioration of IDD. It has been reported that TIMP3 can increase synthesis of ECM (36). COL2A1 and ACAN, which act as main components of matrix of NP (37), are essential to maintaining the integrity of IVD (4). The results from the present study suggested that c-Jun was positively related to IDD in NP cells, and that TGF-β may act as a key regulator in c-Jun signaling pathway. Hiyama et al (38) reported that when NP cells are stimulated by TGF-β, there is a concomitant increase in c-Jun expression and activity (38). In the present study, when patients' NP cells were transduced to overexpress c-Jun, cell survival rate was increased and apoptosis rate was decreased. However, there was no significant difference between the control group and the overexpressed c-Jun group. Treatment with antibody against TGF-β significantly increased the rate of apoptosis and decreased the survival rate, suggesting that c-Jun might promote cell proliferation and inhibit cell apoptosis, thereby minimizing the function of c-Jun after inhibition of TGF-β. This result was consistent with previous studies showing that c-Jun promotes cell proliferation and protects cells from apoptosis (39-42).

In the present study, c-Jun increased the expression of TGF-β, TIMP-3, ACAN and COL2A1 which may promote ECM synthesis, and suppressed the inflammatory response by decreasing the expression of TNF-α, IL-1β, IL-6 and IL-17. Furthermore, c-Jun facilitated cell proliferation and decreased cell apoptosis by upregulating TGF-β expression. These results suggested that c-Jun may alleviate IDD by regulating TGF-β. The results from this study allowed a better understanding of the molecular mechanisms of IDD, which may help the development of novel treatment of IDD disease. To the best of our knowledge, only a few studies have elucidated the role of c-Jun in IDD or additional degenerative diseases. The present study evaluated the effects of c-Jun on NP cells in vitro; however, whether c-Jun could delay disc degeneration in vivo, or directly interact with TGF-β remain unknown. Further investigation is therefore required.

Supplementary Material

Sequences of the primers used for reverse transcription quantitative PCR.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

ML performed the experiments, collected the results and wrote the manuscript. KW, SL, KZ, WH and XW contributed to data analysis and manuscript revision. CY conceived the study and contributed to reviewing/editing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study complied with the Declaration of Helsinki and was approved by the Institutional Review Board Ethics Committee of Union Hospital of Tongji Medical College. Written informed consent was obtained from each patient.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Freemont AJ: The cellular pathobiology of the degenerate intervertebral disc and discogenic back pain. Rheumatology (Oxford). 48:5–10. 2009.PubMed/NCBI View Article : Google Scholar

2 

Hanaei S, Abdollahzade S, Khoshnevisan A, Kepler CK and Rezaei N: Genetic aspects of intervertebral disc degeneration. Rev Neurosci. 26:581–606. 2015.PubMed/NCBI View Article : Google Scholar

3 

Risbud MV and Shapiro IM: Role of cytokines in intervertebral disc degeneration: Pain and disc content. Nat Rev Rheumatol. 10:44–56. 2014.PubMed/NCBI View Article : Google Scholar

4 

Zheng L, Jianxiong S, Wu WK, Xin Y, Jinqian L, Guixing Q and Jiaming L: The role of leptin on the organization and expression of cytoskeleton elements in nucleus pulposus cells. J Orthop Res. 31:847–857. 2013.PubMed/NCBI View Article : Google Scholar

5 

Li Z, Liang J, WK WK, Yu X, Yu J, Weng X and Shen J: Leptin activates RhoA/ROCK pathway to induce cytoskeleton remodeling in nucleus pulposus cells. Int J Mol Sci. 15:1176–1188. 2014.PubMed/NCBI View Article : Google Scholar

6 

Pattison ST, Melrose J, Ghosh P and Taylor TKF: Regulation of gelatinase-a (mmp-2) production by ovine intervertebral disc nucleus pulposus cells grown in alginate bead culture by transforming growth factor-β and insulin like growth factor-1. Cell Biol Int. 25:679–689. 2001.PubMed/NCBI View Article : Google Scholar

7 

Gruber HE and Hanley EN Jr: Recent advances in disc cell biology. Spine (Phila Pa 1976). 28:186–193. 2003.PubMed/NCBI View Article : Google Scholar

8 

Jianru W, Dessislava M, D Greg A, Zhaomin Z, Irving MS and Makarand VR: TNF-α and IL-1β promote a disintegrin-like and metalloprotease with thrombospondin type I motif-5-mediated aggrecan degradation through syndecan-4 in intervertebral disc. J Biol Chem. 286:39738–39749. 2011.PubMed/NCBI View Article : Google Scholar

9 

Wang WJ, Yu XH, Wang C, Yang W, He WS, Zhang SJ, Yan YG and Zhang J: MMPs and ADAMTSs in intervertebral disc degeneration. Clin Chim Acta. 448:238–246. 2015.PubMed/NCBI View Article : Google Scholar

10 

Vo NV, Hartman RA, Yurube T, Jacobs LJ, Sowa GA and Kang JD: Expression and regulation of metalloproteinases and their inhibitors inintervertebral disc aging and degeneration. Spine J. 13:331–341. 2013.PubMed/NCBI View Article : Google Scholar

11 

Jin H, Shen J, Wang B, Wang M, Shu B and Chen D: TGF-β signaling plays an essential role in the growth and maintenance of intervertebral disc tissue. FEBS Lett. 585:1209–1215. 2011.PubMed/NCBI View Article : Google Scholar

12 

Feng C, Liu H, Yang M, Zhang Y, Huang B and Zhou Y: Disc cell senescence in intervertebral disc degeneration: Causes and molecular pathways. Cell Cycle. 15:1674–1684. 2016.PubMed/NCBI View Article : Google Scholar

13 

Angel P and Karin M: The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. Biochim Biophys Acta. 1072:129–157. 1991.PubMed/NCBI View Article : Google Scholar

14 

Wisdom R, Johnson RS and Moore C: c-Jun regulates cell cycle progression and apoptosis by distinct mechanisms. EMBO J. 18:188–197. 1999.PubMed/NCBI View Article : Google Scholar

15 

Behrens A, Sibilia M and Wagner EF: Amino-terminal phosphorylation of c-Jun regulates stress-induced apoptosis and cellular proliferation. Nat Genet. 21:326–329. 1999.PubMed/NCBI View Article : Google Scholar

16 

Santos MM, Tannuri AC, Coelho MC, Gonçalves JD, Serafini S, Da Silva LF and Tannuri U: Immediate expression of c-Fos and c-jun mRNA in a model of intestinal autotransplantation and ischemia-reperfusion in situ. Clinics (Sao Paulo). 70:373–379. 2015.PubMed/NCBI View Article : Google Scholar

17 

Behrens A, Sibilia M, David JP, Möhle-Steinlein U, Tronche F, Schütz G and Wagner EF: Impaired postnatal hepatocyte proliferation and liver regeneration in mice lacking c-jun in the liver. EMBO J. 21:1782–1790. 2002.PubMed/NCBI View Article : Google Scholar

18 

Behrens A, Haigh J, Mechta-Grigoriou F, Nagy A, Yaniv M and Wagner EF: Impaired intervertebral disc formation in the absence of Jun. Development. 130:103–109. 2003.PubMed/NCBI View Article : Google Scholar

19 

Pfirrmann CW, Metzdorf A, Zanetti M, Hodler J and Boos N: Magnetic resonance classification of lumbar intervertebral disc degeneration. Spine (Phila Pa 1976). 26:1873–1878. 2001.PubMed/NCBI View Article : Google Scholar

20 

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

21 

Zhang Y, Feng XH and Derynck R: Smad3 and Smad4 cooperate with c-Jun/c-Fos to mediate TGF-beta-induced transcription. Nature. 394:909–913. 1998.PubMed/NCBI View Article : Google Scholar

22 

Ye N, Ding Y, Wild C, Shen Q and Jia Z: Small molecule inhibitors targeting activator protein 1 (AP-1). J Med Chem. 57:6930–6948. 2014.PubMed/NCBI View Article : Google Scholar

23 

Tolonen J, Grönblad M, Virri J, Seitsalo S, Rytömaa T and Karaharju E: Oncoprotein c-Fos and c-Jun immunopositive cells and cell clusters in herniated intervertebral disc tissue. Eur Spine J. 11:452–458. 2002.PubMed/NCBI View Article : Google Scholar

24 

Monchaux M, Forterre S, Spreng D, Karol A, Forterre F and Wuertz-Kozak K: Inflammatory processes associated with canine intervertebral disc herniation. Front Immunol. 8(1681)2017.PubMed/NCBI View Article : Google Scholar

25 

Liu XG, Hou HW and Liu YL: Expression levels of IL-17 and TNF-α in degenerated lumbar intervertebral discs and their correlation. Exp Ther Med. 11:2333–2340. 2016.PubMed/NCBI View Article : Google Scholar

26 

Liu Y, Du Y, Wang H, Li D and Feng WH: Porcine reproductive and respiratory syndrome virus (PRRSV) up-regulates IL-8 expression through TAK-1/JNK/AP-1 pathways. Virology. 506:64–72. 2017.PubMed/NCBI View Article : Google Scholar

27 

Shaulian E and Karin M: AP-1 as a regulator of cell life and death. Nat Cell Biol. 4:E131–E136. 2002.PubMed/NCBI View Article : Google Scholar

28 

Li JK, Nie L, Zhao YP, Zhang YQ, Wang X, Wang SS, Liu Y, Zhao H and Cheng L: IL-17 mediates inflammatory reactions via p38/c-Fos and JNK/c-Jun activation in an AP-1-dependent manner in human nucleus pulposus cells. J Transl Med. 14(77)2016.PubMed/NCBI View Article : Google Scholar

29 

Duval H, Mbatchi SF, Grandadam S, Legendre C, Loyer P, Ribault C, Piquet-Pellorce C, Guguen-Guillouzo C, Boudjema K and Corlu A: Reperfusion stress induced during intermittent selective clamping accelerates rat liver regeneration through JNK pathway. J Hepatol. 52:560–569. 2010.PubMed/NCBI View Article : Google Scholar

30 

Koba S, Pakala R, Watanabe T, Katagiri T and Benedict CR: Vascular smooth muscle proliferation-a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. J Am Coll Cardiol. 34:1644–1651. 1999.

31 

Lee YJ, Kong MH, Song KY, Lee KH and Heo SH: The relation between Sox9, TGF-beta1, and proteoglycan in human intervertebral disc cells. J Korean Neurosurg Soc. 43:149–154. 2008.PubMed/NCBI View Article : Google Scholar

32 

Wang SL, Yu YL, Tang CL and Lv FZ: Effects of TGF-β1 and IL-1β on expression of ADAMTS enzymes and TIMP-3 in human intervertebral disc degeneration. Exp Ther Med. 6:1522–1526. 2013.PubMed/NCBI View Article : Google Scholar

33 

Nakai T, Mochida J and Sakai D: Synergistic role of c-Myc and ERK1/2 in the mitogenic response to TGF beta-1 in cultured rat nucleus pulposus cells. Arthritis Res Ther. 10(R140)2008.PubMed/NCBI View Article : Google Scholar

34 

Iwata A, Masago A and Yamada K: Expression of basic fibroblast growth factor mRNA after transient focal ischemia: Comparison with expression of c-fos, c-jun, and hsp 70 mRNA. J Neurotrauma. 14:201–210. 1997.PubMed/NCBI View Article : Google Scholar

35 

Qing J, Zhang Y and Derynck R: Structural and functional characterization of the transforming growth factor-beta-induced Smad3/c-Jun transcriptional cooperativity. J Biol Chem. 275:38802–38812. 2000.PubMed/NCBI View Article : Google Scholar

36 

Yan L, Kang L, Han X, Mao C, Kai Z, Zhao T and Jie Z: The imbalance between TIMP3 and matrix-degrading enzymes plays an important role in intervertebral disc degeneration. Biochem Biophys Res Commun. 469:507–514. 2016.PubMed/NCBI View Article : Google Scholar

37 

Chung SA, Khan SN and Diwan AD: The molecular basis of intervertebral disk degeneration. Orthop Clin North Am. 34:209–219. 2003.PubMed/NCBI View Article : Google Scholar

38 

Hiyama A, Gogate SS, Gajghate S, Mochida J, Shapiro IM and Risbud MV: BMP-2 and TGF-beta stimulate expression of beta 1,3-glucuronosyl transferase 1 (GlcAT-1) in nucleus pulposus cells through AP1, TonEBP, and Sp1: Role of MAPKs. J Bone Miner Res. 25:1179–1190. 2010.PubMed/NCBI View Article : Google Scholar

39 

Gaudet P, Livstone MS, Lewis SE and Thomas PD: Phylogenetic-based propagation of functional annotations within the gene ontology consortium. Brief Bioinform. 12:449–462. 2011.PubMed/NCBI View Article : Google Scholar

40 

Qian Z, Li Y, Chen J, Li X and Gou D: MiR-4632 mediates PDGFBB-induced proliferation and anti-apoptosis of human pulmonary artery smooth muscle cells via targeting cJUN. Am J Physiol Cell Physiol. 313:C380–C391. 2017.PubMed/NCBI View Article : Google Scholar

41 

Khachigian LM, Fahmy RG, Zhang G, Bobryshev YV and Kaniaros A: c-Jun regulates vascular smooth muscle cell growth and neointima formation after arterial injury. Inhibition by a novel DNA enzyme targeting c-Jun. J Biol Chem. 277:22985–22991. 2002.PubMed/NCBI View Article : Google Scholar

42 

Kappelmann M, Bosserhoff A and Kuphal S: AP-1/c-Jun transcription factors: Regulation and function in malignant melanoma. Eur J Cell Biol. 93:76–81. 2014.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2020
Volume 20 Issue 5

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
Lei M, Wang K, Li S, Zhao K, Hua W, Wu X and Yang C: The c‑Jun signaling pathway has a protective effect on nucleus pulposus cells in patients with intervertebral disc degeneration. Exp Ther Med 20: 123, 2020
APA
Lei, M., Wang, K., Li, S., Zhao, K., Hua, W., Wu, X., & Yang, C. (2020). The c‑Jun signaling pathway has a protective effect on nucleus pulposus cells in patients with intervertebral disc degeneration. Experimental and Therapeutic Medicine, 20, 123. https://doi.org/10.3892/etm.2020.9251
MLA
Lei, M., Wang, K., Li, S., Zhao, K., Hua, W., Wu, X., Yang, C."The c‑Jun signaling pathway has a protective effect on nucleus pulposus cells in patients with intervertebral disc degeneration". Experimental and Therapeutic Medicine 20.5 (2020): 123.
Chicago
Lei, M., Wang, K., Li, S., Zhao, K., Hua, W., Wu, X., Yang, C."The c‑Jun signaling pathway has a protective effect on nucleus pulposus cells in patients with intervertebral disc degeneration". Experimental and Therapeutic Medicine 20, no. 5 (2020): 123. https://doi.org/10.3892/etm.2020.9251