Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Apr 3;23(1):395.
doi: 10.1186/s12967-024-05913-1.

SOX4 accelerates intervertebral disc degeneration via EZH2/NRF2 pathway in response to mitochondrial ROS-dependent NLRP3 inflammasome activation in nucleus pulposus cells

Affiliations

SOX4 accelerates intervertebral disc degeneration via EZH2/NRF2 pathway in response to mitochondrial ROS-dependent NLRP3 inflammasome activation in nucleus pulposus cells

Wenzhi Zhao et al. J Transl Med. .

Abstract

Objective: The transcription factor SRY-related HMG-box 4 (SOX4) has been implicated in intervertebral disc diseases. This study aimed to investigate the role of SOX4 in intervertebral disc degeneration (IDD) and explore the underlying molecular mechanisms.

Methods: We established an IDD rat model via surgery and analyzed SOX4 expression using qRT-PCR and Western blotting. Histological evaluation, immunohistochemistry, and Safranin O staining assessed IDD progression. In vitro, an IDD cellular model was constructed using IL-1β-stimulated nucleus pulposus (NP) cells. SOX4 knockdown and overexpression experiments in NP cells examined SOX4 effects on ECM degradation, NLRP3-mediated pyroptosis, and mitochondrial ROS-dependent NLRP3 inflammasome activation. The involvement of the EZH2/NRF2 pathway in SOX4-mediated NLRP3 activation was also examined.

Results: SOX4 expression was significantly increased in IDD rats and promoted IDD progression. Knockdown of SOX4 inhibited ECM degradation and NLRP3-mediated pyroptosis in NP cells. In vitro experiments showed that SOX4 promoted ECM degradation by upregulating MMPs and ADAMTS-5 expression, and suppressed collagen II and aggrecan synthesis. SOX4 knockdown inhibited NLRP3-mediated pyroptosis, while overexpression accelerated it in NP cells. Additionally, SOX4 was found to exacerbate mitochondrial ROS-dependent NLRP3 inflammasome activation in NP cells. Further investigation revealed that SOX4 enhanced NLRP3 inflammasome activation by upregulating EZH2 expression and modulating the EZH2/NRF2 pathway, with EZH2 inhibition attenuating SOX4-induced NLRP3 activation.

Conclusion: Our findings suggest that SOX4 accelerates IDD progression by promoting NLRP3 inflammasome activation via modulating the EZH2/NRF2 pathway, leading to NP cell pyroptosis and ECM degradation. Targeting SOX4 may represent a potential therapeutic strategy for treating IDD.

Keywords: EZH2/NRF2 pathway; Extracellular matrix degradation; Intervertebral disc degeneration; NLRP3 inflammasome activation; Pyroptosis; SRY-related HMG-box 4.

PubMed Disclaimer

Conflict of interest statement

Declarations. Ethics approval and consent to participate: This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of Yangzhou University (No. 202306014). Consent for publication: Not applicable. Conflict of interest: The authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1
SOX4 expression is increased in intervertebral disc degeneration (IDD) rats and promotes IDD progression. (AB) Quantitative real-time PCR (qRT-PCR) and Western blot analysis showing the expression of SOX4 in the sh-SOX4 and sh-NC groups. (C) T2-weighted MRI and Pfirrmann grading at 8 weeks post-surgery for the sh-SOX4 and sh-NC groups. (DE) HE staining and safranin O staining of nucleus pulposus (NP) tissue at week 8 post-surgery in the sh-SOX4 and sh-NC groups; magnification: 5× and 20×. **P < 0.01 versus the sh-NC group
Fig. 2
Fig. 2
Knockdown of SOX4 inhibits ECM degradation and NLRP3-mediated pyroptosis in IDD rats. (A) Immunohistochemical staining of ADAMTS-5, MMP-13, and caspase-3 in NP tissues from the IDD, control, IDD + sh-SOX4, and IDD + sh-NC groups; scale bar: 20 μm. (B) ELISA analysis of the expression of MMP-1, MMP-2, TIMP-1, and TIMP-2 in NP cell supernatants. (C) Western blot analysis indicating the protein expression of NLRP3, caspase-1, and IL-1β in NP tissues from the IDD, control, IDD + sh-SOX4, and IDD + sh-NC groups. **P < 0.01 versus the control group; #P < 0.05 and ##P < 0.01 versus the IDD + sh-NC group
Fig. 3
Fig. 3
Evaluation of apoptosis in IL-1β-stimulated NP cells with SOX4 knockdown and overexpression. (A-B) Detection of SOX4 expression in NP cells using qRT-PCR and Western blotting. (C) Cell viability test (CCK-8) showing the effect of SOX4 knockdown and overexpression in IL-1β-stimulated NP cells. (D) Flow cytometry analysis of cell apoptosis in IL-1β-stimulated NP cells after knockdown and overexpression of SOX4. (E) Western blot analysis of apoptosis markers (caspase-3, Bcl-2, and Bax) in IL-1β-induced NP cells after sh-SOX4 and oe-SOX4 transfection. **P < 0.01 versus the sh-NC group; ##P < 0.01 versus the oe-NC group
Fig. 4
Fig. 4
SOX4 exacerbates NLRP3 inflammasome activation in NP cells. (A) Immunofluorescence analysis to assess the expression of GSDMD (pyroptosis marker), MMP13, and NLRP3 in IL-1β-treated NP cells with SOX4 knockdown and overexpression; scale bar: 25 μm. (B) Transmission Electron Microscopy (TEM) images showing the mitochondrial morphology in NP cells with SOX4 knockdown and overexpression; scale bar: 500 nm. (CD) qRT-PCR or Western blot analysis of NLRP3 inflammasome activation proteins (NLPR3 and p20), ADAMTS-5, and MMP-13 in IL-1β-induced NP cells with SOX4 knockdown and overexpression. **P < 0.01 versus the sh-NC group; ##P < 0.01 versus the oe-NC group
Fig. 5
Fig. 5
SOX4 overexpression induces the mitochondrial damage in NP cells. (A) The H2DCF-DA assay analyzed the intracellular ROS levels of NP cells following Mito-tempo treatment and/or SOX4 overexpression; scale bar: 25 μm. (B) MitoSOX Red staining demonstrating mitochondrial ROS levels in NP cells; scale bar: 25 μm. (C) TEM images displaying mitochondrial morphology in NP cells; scale bar: 100 nm. (D) Levels of 8-OHdG in NP cells were measured by ELISA. **P < 0.01 versus the model group; ##P < 0.01 versus the Mito-tempo + oe-NC group
Fig. 6
Fig. 6
SOX4 overexpression induces the mitochondrial damage in NP cells by promoting NLRP3 inflammasome activation. (A) JC-1 assay results displaying the mitochondrial membrane potential in NP cells pre-treated with Mito-tempo and/or oe-SOX4/NC transfection; scale bar: 25 μm. (B) Western blot analysis indicating the protein expression of NLPR3, p20, ADAMTS-5, MMP-13, and mitochondrial morphology-related proteins (Drp1, OPA1, Mfn1/2) following Mito-tempo treatment and/or SOX4 overexpression. **P < 0.01 versus the model group; ##P < 0.01 versus the Mito-tempo + oe-NC group
Fig. 7
Fig. 7
SOX4 promotes NLRP3 inflammasome activation in NP cells through upregulating EZH2 expression. (A) NP cells were infected with indicated sh-NC or sh-SOX4. Cells were collected for ChIP-qRT-PCR analysis using the IgG or SOX4 antibody in NP cells. (B) Immunofluorescence analysis displaying co-localization of SOX4 and EZH2 in the nucleus of NP cells; scale bar: 25 μm. (CD) qRT-PCR and Western blot analysis demonstrating the expression of SOX4 and EZH2 after oe-EZH2 and/or sh-SOX4 transfection in IL-1β-induced NP cells. (E) MitoSOX Red staining showing mitochondrial ROS production in NP cells after oe-EZH2 and/or sh-SOX4 transfection; scale bar: 25 μm. (F) Western blot analysis indicating the protein expression of NLRP3, p20, Drp1, and Mfn2 following oe-EZH2 and/or sh-SOX4 transfection. *P < 0.05 and **P < 0.01 versus the sh-NC group; #P < 0.05 and ##P < 0.01 versus the oe-EZH2 group
Fig. 8
Fig. 8
SOX4 enhances NLRP3 inflammasome activation in NP cells via the EZH2/Nrf2 pathway. (AB) qRT-PCR and Western blot analysis showing the expression of SOX4, EZH2, and Nrf2 in NP cells after treatment with ML385 and/or sh-SOX4. (C) CCK8 assay results demonstrating cell viability after treatment with ML385 and/or sh-SOX4. (D) Levels of 8-OHdG in NP cells were measured by ELISA. (E) JC-1 assay indicating the mitochondrial membrane potential after treatment with ML385 and/or sh-SOX4; scale bar: 25 μm. (F) DCFH-DA probe showing ROS levels in NP cells after treatment with ML385 and/or sh-SOX4; scale bar: 25 μm. (G) Western blot analysis of NLPR3, p20, Drp1, OPA1, Mfn1, and Mfn2 protein expression in NP cells following treatment with ML385 and/or sh-SOX4. *P < 0.05 and **P < 0.01 versus the Model group; #P < 0.05 and ##P < 0.01 versus the ML385 + sh-SOX4 group
Fig. 9
Fig. 9
A schematic diagram illustrating the mechanism of SOX4 in intervertebral disc degeneration

Similar articles

Cited by

References

    1. Zhang GZ, et al. NF-κB signalling pathways in nucleus pulposus cell function and intervertebral disc degeneration. Cell Prolif. 2021;54(7):e13057. - PMC - PubMed
    1. Wang J, et al. The Regulatory Effect of MicroRNA-101-3p on Disc Degeneration by the STC1/VEGF/MAPK pathway. Oxid Med Cell Longev. 2021;2021:p1073458. - PMC - PubMed
    1. Gao S, et al. A potential target gene CD63 for different degrees of intervertebral disc degeneration. Sci Rep. 2022;12(1):957. - PMC - PubMed
    1. Liao C-R, et al. Advanced oxidation protein products increase TNF-α and IL-1β expression in chondrocytes via NADPH oxidase 4 and accelerate cartilage degeneration in osteoarthritis progression. Redox Biol. 2020;28:101306–101306. - PMC - PubMed
    1. Guo HY, et al. Emerging evidence on noncoding-RNA regulatory machinery in intervertebral disc degeneration: a narrative review. Arthritis Res Ther. 2020;22(1):270. - PMC - PubMed

MeSH terms

LinkOut - more resources