Open Access

Inflammatory response and oxidative stress attenuated by sulfiredoxin‑1 in neuron‑like cells depends on nuclear factor erythroid‑2‑related factor 2

  • Authors:
    • Zhiliang Wu
    • Zhenghao Lu
    • Jun Ou
    • Xiaotao Su
    • Jingnan Liu
  • View Affiliations

  • Published online on: September 28, 2020     https://doi.org/10.3892/mmr.2020.11545
  • Pages: 4734-4742
  • Copyright: © Wu 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

Sulfiredoxin‑1 (SRX1) is a conserved endogenous antioxidative protein, which is involved in the response to cellular damage caused by oxidative stress. Oxidative stress and inflammation are the primary pathological changes in spinal cord injuries (SCI). The aim of present study was to explore the roles of SRX1 in SCI. Using reverse transcription‑quantitative PCR and western blotting, the present study discovered that the expression levels of SRX1 were downregulated in the spinal cord tissues of SCI model rats. Massive irregular cavities and decreased Nissl bodies were observed in the model group compared with the sham group. Thus, to determine the underlying mechanisms, neuron‑like PC12 cells were cultured in vitro. Western blotting analysis indicated that SRX1 expression levels were downregulated following the exposure of cells to lipopolysaccharide (LPS). Following the transfection with the SRX1 overexpression plasmid and stimulation with LPS, the results of the Cell Counting Kit‑8 assay indicated that the cell viability was increased compared with LPS stimulation alone. Furthermore, the expression levels of proinflammatory cytokines secreted by LPS‑treated PC12 cells were downregulated following SRX1 overexpression. Increased malondialdehyde content, decreased superoxide dismutase activity and reactive oxygen species production were also identified in PC12 cells treated with LPS using commercial detection kits, whereas the overexpression of SRX1 partially reversed the effects caused by LPS stimulation. The aforementioned results were further verified by determining the expression levels of antioxidative proteins using western blotting analysis. In addition, nuclear factor erythroid‑2‑related factor 2 (NRF2), a transcription factor known to regulate SRX1, was indicated to participate in the protective effect of SRX1 against oxidative stress. Inhibition of NRF2 further downregulated the expression levels of SRX1, NAD(P)H dehydrogenase quinone 1 and heme oxygenase‑1 in the presence of LPS, while activation of NRF2 reversed the effects of LPS on the expression levels of these proteins. In conclusion, the results of the present study indicated that the anti‑inflammatory and antioxidative effects of SRX1 may depend on NRF2, providing evidence that SRX1 may serve as a novel molecular target to exert a neuroprotective effect in SCI.

Introduction

Spinal cord injury (SCI) affects millions of individuals worldwide (1), with between 16 and 19.4 new cases per million annually in Western European countries (2), and characteristically causes life-long neurological consequences (different degrees of paralysis and sensory impairment of limbs and trunk) with substantial socioeconomic implications (3). SCI is divided into primary injury and secondary injury; primary injury refers to the initial physical damage of the spinal cord caused by an indirect or direct external force, while the secondary injury is characterized by a series of physiological and pathological changes to the spinal cord, including inflammation, oxidative stress, necrosis and neuronal apoptosis on the basis of the primary damage, further deepening and expanding the degree and scope of the damage (46). The secondary injury is largely responsible for the neurological dysfunction associated with SCI. The current therapeutic strategies for SCI include surgical intervention (grafts and bridges), neural stem cell transplantation and the administration of high-dose methylprednisolone (7,8), the neurological recovery remains limited since there is no consensus about the beneficial effects. Both molecular therapies (modulation of inflammatory response and administration of growth-stimulating factors), rehabilitative training and combinatorial therapies (tissue engineering and searching for synergistic effects) have raised hopes in developing novel therapies for attenuating secondary damage (9). Previous studies have demonstrated that the pathophysiological processes of SCI involved neuronal inflammation, oxidative stress, neuronal degeneration and apoptosis, as well as reactive changes in the glia (1013). Therefore, further investigations into the pathogenesis may improve the current understanding of the molecular mechanisms of SCI.

The small-molecule protein sulfiredoxin-1 (SRX1), a conserved endogenous antioxidative protein, is found widely distributed in eukaryotes (14). It was previously reported that SRX1 served an important role in maintaining the pulmonary antioxidative defense against cigarette smoke via nuclear factor erythroid-2-related factor 2 (NRF2)-dependent transcriptional regulation (15). SRX1 was also identified to have a crucial role in cellular damage triggered by oxidative stress (16,17). Moreover, SRX1 protected cardiac progenitor cells from oxidative stress and promoted their survival via ERK/NRF2 signaling (18). Another study also illustrated the cardioprotective effect of SRX1 via the inhibition of mitochondrial apoptosis through the PI3K/AKT signaling pathway (19). However, to the best of our knowledge, the effects of SRX1 on nerve damage in SCI remain poorly understood. Therefore, the present study aimed to determine whether the functions of SRX1 in SCI were associated with oxidative stress and inflammation. In addition, the roles of NRF2 and the downstream target genes were investigated.

Materials and methods

Establishment of SCI model rats

All experiments were approved by the Institutional Animal Ethics Committees of University of South China (Hengyang, China) and were performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals (NIH Publications no. 8023, revised 1978). In total, 32 male Sprague Dawley rats (6 weeks old, 200–220 g) were purchased from the Central South University Experiment Center. The animals were provided with free access to food and water, and maintained in standardized conditions with a 12-h light/dark cycle, at a temperature of 22±2°C and humidity of 55±10%. Following one week of adaptive feeding, all rats received thoracic laminectomies at the 7, 8 and 9th thoracic vertebrae under aseptic conditions; 5% isoflurane was used to induce anesthesia and rats were maintained at a surgical plane by continuous inhalation of 2% isoflurane for the duration of surgery. Subsequently, the rats were randomly divided into two groups (16 rats per group): Sham and model. The model group was established by the clip compression technique as previously described (20,21) and the wound was sutured. The sham group contained rats who underwent a laminectomy without clip compression.

During the 8 weeks following surgery, the animal health and behavior were monitored and recorded once a day (behavioral assessment data not disclosed). In the sham group, two rats died within 3 days of the sham operation and one died within 8 to 14 days; no deaths were recorded after 2 weeks. In the model group, two rats died within 3 days following spinal trauma, none died from day 4 to day 7, three died on day 8 to day 14 and one died from day 15 to day 30; no deaths were reported after 31 days (Table SI). Therefore, there were 10 rats in the model group and 13 rats in the sham group. Autopsy was performed to identify the cause of death. Subsequently, 8 weeks after surgery, rats were euthanized through inhalation of 5% isoflurane until respiration ceased (within 5 min) and decapitation was used as a secondary method of euthanasia to ensure a humane death. Spinal cord tissues were then extracted after confirming cardiac arrest. The following humane endpoints were adhered to through the study: Emaciation, lethargy, abdominal swelling and self-mutilation.

Histological analysis

Spinal cord tissues were fixed in 4% formaldehyde at 4°C for 24 h and embedded in paraffin. The paraffin-embedded tissues were cut into 5-µm thick sections and stained with hematoxylin for 10 min and eosin for 5 min at room temperature (H&E).

Spinal cord tissues were fixed in 4% formaldehyde at 4°C for 24 h and embedded in paraffin. A Nissl staining kit (Beijing Solarbio Science & Technology Co., Ltd.) was used to stain 8-µm thick spinal cord sections, according to the manufacturer's instructions. Tissues sections were observed under a light microscope (magnification, ×200).

Cell culture and treatments

Rat pheochromocytoma PC12 cells (China Center for Type Culture Collection) were cultured in DMEM (Hyclone; Cytiva), supplemented with 10% FBS (Hyclone; Cytiva), and maintained at 37°C under humid conditions with 5% CO2 and 95% air. For differentiation, PC12 cells were treated with 50 ng/ml nerve growth factor (Sigma-Aldrich; Merck KGaA) every other day for 6 days at 37°C, which is a widely used method for the in vitro study of nervous system diseases, including SCI (2225).

To investigate the function of NRF2 in the differentiated PC12 cells, the cells were pretreated with the NRF2 activator tert-butylhydroquinone (25 µM, TBHQ; Sigma-Aldrich; Merck KGaA) or inhibitor ML385 (5 µM; MedChemExpress) for 24 h prior to the experiments (2628), then PC12 cells were exposed to 5 µg/ml lipopolysaccharide (LPS; MedChemExpress) for 18 h.

Cell transfection

PC12 cells were seeded into a 6-well plate (5×105 cells per well). At a confluence of 60%, 8 µg SRX1 overexpression (Ov) pEX-1 plasmid (Ov-SRX1; Shanghai GenePharma Co., Ltd.) or an empty control pEX-1 plasmid (Ov-NC) was transfected into cells using 5 µl Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). PC12 cells were transfected with Ov-SRX1 or Ov-NC according to the manufacturer's protocol. After successful transfection, cells were used in subsequent experiments.

Cell viability assay

The viability of PC12 cells was analyzed using a Cell Counting Kit-8 (CCK-8) assay (MedChemExpress), according to the manufacturer's protocols. Briefly, cells at a density of 5×103 cells/ml were plated into a 96-well plate (100 µl per well) and treated with 0.625, 1.25, 2.5 and 5 µg/ml LPS for 18 h. Subsequently, 10 µl CCK-8 solution was added/well and incubated for 4 h. Following the incubation, the absorbance was measured at a wavelength of 450 nm using a microplate reader (Bio-Rad Laboratories, Inc.).

Western blotting

Total protein was extracted from spinal cord tissues or PC12 cells using RIPA lysis buffer (Beyotime Institution of Biotechnology), while nuclear protein was extracted using a Nuclear and Cytoplasmic Extraction kit (Beyotime Institute of Biotechnology). Total protein was quantified using a bicinchoninic acid assay kit (Abcam) and 25 µg protein was separated via 10% SDS-PAGE. The separated proteins were subsequently transferred onto PVDF membranes (EMD Millipore) and then membranes were blocked with 5% BSA Blocking Buffer (Beijing Solarbio Science & Technology Co., Ltd.). The membranes were then incubated with the following primary antibodies at 4°C overnight: Anti-SRX1 (cat. no. ab203613; 1:1,000; Abcam), anti-peroxiredoxin (PRDX)1 (cat. no. ab15571; 1:1,000; Abcam), anti-PRDX6 (cat. no. ab59543; 1:1,000; Abcam), anti-thioredoxin reductase (TXNRD) 1 (cat. no. ab16840; 1:1000; Abcam), anti-superoxide dismutase (SOD)2 (cat. no. ab13534; 1:1,000; Abcam), anti-NRF2 (cat. no. ab89443; 1:1,000; Abcam), anti-NAD(P)H dehydrogenase quinone (NQO) 1 (cat. no. ab28947; 1:1,000; Abcam), anti-heme oxygenase 1 (cat. no. ab13248; 1:1,000; HO-1; Abcam), anti-GAPDH (cat. no. ab181603; 1:1,000; Abcam) and anti-Lamin B1 (cat. no. ab16048; 1:1,000; Abcam). Following the primary antibody incubation, the membranes were washed with TBS with Tween-20 (0.05%) and incubated with a goat anti-mouse IgG HRP-conjugated secondary antibody (cat. no. 31430; 1:10,000; Thermo Fisher Scientific, Inc.) or a goat anti-rabbit IgG HRP-conjugated secondary antibody (cat. no. 31460; 1:10,000; Thermo Fisher Scientific, Inc.) at room temperature for 2 h. Protein bands were visualized using Immobilon Crescendo Western HRP substrate (cat. no. WBLUR0100; Merck KGaA) and the expression levels were semi-quantified using ImageJ v1.8.0 software (National Institutes of Health).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the spinal cord tissues or PC12 cells using TRI reagent® (Sigma-Aldrich; Merck KGaA). The reaction templates in PrimeScript™ RT reagent kit (Takara Bio, Inc.) were added into the tube according to the manufacturer's protocol, centrifuged for 5 sec (500 × g) at room temperature and total RNA was reversed transcribed into cDNA at 37°C for 15 min and 85°C for 5 sec. qPCR was subsequently performed using a SYBR® Premix Ex Taq II kit (Takara Bio, Inc.), and the following thermocycling conditions were used: One cycle of 95°C for 30 sec, 40 cycles of 95°C for 5 sec and 60°C for 34 sec. The following primers sequences were used for the qPCR: SRX1 forward, 5′-AATCCCCAACCCCTGACTTT-3′ and reverse, 5′-AATCCCCAACCCCTGACTTT-3′; NQO1 forward, 5′-GCGTCTGGAGACTGTCTGGG-3′ and reverse, 5′-CGGCTGGAATGGACTTGC-3′; HO-1 forward, 5′-GCGAAACAAGCAGAACCCA-3′ and reverse, 5′-GCTCAGGATGAGTACCTCCCA-3′; and GAPDH forward, 5′-TGGCCTCCAAGGAGTAAGAAAC-3′ and reverse, 5′-GGCCTCTCTCTTGCTCTCAGTATC-3′. The expression levels were calculated using the 2−∆∆Cq method and normalized to the loading control GAPDH (29).

Determination of malondialdehyde (MDA) content, and the levels of SOD, reactive oxygen species (ROS) and inflammatory cytokines

The cell medium of PC12 cells was collected and centrifugated at 500 × g for 5 min at room temperature. The concentrations of tumor necrosis factor (TNF)-α (cat. no. PT516; Beyotime Institute of Biotechnology), interleukin (IL)-1β (cat. no. PI303; Beyotime Institute of Biotechnology), IL-18 (cat. no. SEKR-0054; Beijing Solarbio Science & Technology Co., Ltd.) and IL-10 (cat. no. PI525; Beyotime Institute of Biotechnology) in the medium supernatants were analyzed using ELISA kits. MDA and SOD in the cellular supernatants were analyzed using commercial detection kits (Beyotime Institute of Biotechnology), according to the manufacturers' protocols. PC12 cells (2×105) were seeded into a 24-well plate, and loaded with 10 µM 2′,7′-dichlorofluorescin diacetate (DCFH-DA; Invitrogen; Thermo Fisher Scientific, Inc.) for 20 min in the dark at room temperature. Intracellular ROS production was observed under a fluorescence microscope (magnification, ×100; Olympus Corporation) and analyzed using ImageJ v1.8.0 software (National Institutes of Health).

Statistical analysis

Statistical analysis was performed using GraphPad Prism 6 software (GraphPad Software, Inc.) and data was presented as the mean ± SD. Each experiment was repeated three times. Statistical differences between groups were determined using a one-way ANOVA, followed by a Tukey's post hoc test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression levels of SRX1 are downregulated in SCI model rats

SCI model rats were established as previously reported (20,21). The pathological changes in the spinal cord tissues and the expression levels of SRX1 were subsequently investigated. The expression levels of SRX1 in the model group were significantly downregulated at the mRNA and protein level compared with the sham group (Fig. 1A and B). H&E staining also revealed numerous irregular cavities in the injured spinal cords of the model group compared with the sham group (Fig. 1C). In addition, Nissl bodies in the sham group were larger in shape and more abundant compared with those in the SCI model group, indicating a weaker protein synthesis ability of Nissl bodies in the SCI model rats (Fig. 1D). Collectively, these results suggested that the rat model of SCI was successfully established, and that the expression levels of SRX1 may be downregulated in damaged spinal cord tissues.

SRX1 expression levels are downregulated in PC12 cells stimulated with LPS

Subsequently, in vitro experiments were performed using the PC12 cell line, a common cell model used for researching neurobiological events, including SCI (2225). A range of concentrations of LPS (0.625–5 µg/ml) were used to stimulate PC12 cells for 18 h as previously described (26) and the viability of PC12 cells was subsequently determined. The results revealed that 1.25, 2.5 and 5 µg/ml LPS significantly impaired the viability of the PC12 cells compared with the control cells (Fig. 2A). Subsequently, SRX1 expression levels were analyzed following the stimulation with LPS; the expression levels of SRX1 were downregulated by LPS in a dose-dependent manner at both the protein and mRNA level (Fig. 2B and C). These results suggested that SRX1 expression levels may be downregulated in PC12 cells challenged with LPS.

Overexpression of SRX1 inhibits the inflammatory response in PC12 cells

To determine the association between LPS-induced inflammation and SRX1 in PC12 cells, Ov-SRX1 was constructed and transfected into PC12 cells; a significant upregulation in the mRNA expression levels of SRX1 were identified after the cells were transfected with Ov-SRX1 compared with the control and Ov-NC groups (Fig. 3A), indicating that the transfection with the overexpression plasmid was successful. Cell viability was significantly impaired after stimulation with 5 µg/ml LPS, so 5 µg/ml LPS was chosen for subsequent experiments. There was a significant increase in the expression of SRX1 mRNA and protein after cells were transfected with Ov-SRX1 (Fig. 3B and C). In addition, the results of the CCK-8 assay revealed that the Ov-SRX1 plasmid significantly increased the viability of LPS-induced cells compared with cells treated with LPS only (Fig. 3D).

Subsequently, the levels of inflammatory cytokines secreted by PC12 cells following the overexpression of SRX1 and LPS stimulation were analyzed. The levels of the proinflammatory factors, TNF-α, IL-1β and IL-18, were all significantly increased following the stimulation of LPS compared with the control group, while the simultaneous transfection of cells with Ov-SRX1 reversed this effect (Fig. 3E). Conversely, the secretion of IL-10 was significantly decreased following LPS treatment compared with the control group, while the simultaneous transfection with Ov-SRX1 significantly increased the secretion of IL-10 compared with the LPS treatment group (Fig. 3E). Taken together, these results indicated that the inflammatory response in PC12 cells may be attenuated by the overexpression of SRX1.

Overexpression of SRX1 reduces oxidative stress in PC12 cells

The activation of the inflammatory response results in the production of ROS, which further exacerbates inflammation and leads to tissue damage (30). To investigate the antioxidative role of SRX1 in damaged neuron-like cells, the intracellular production of ROS was determined using the fluorescent probe DCFH-DA. Following the treatment of the cells with 5 µg/ml LPS, the levels of ROS were significantly increased compared with the control group, whereas this effect was significantly attenuated following the overexpression of SRX1 (Fig. 4A). Furthermore, the MDA content and SOD activity were also analyzed; LPS treatment significantly increased the generation of MDA compared with the control group; however, this effect was significantly impeded by the overexpression of SRX1 (Fig. 4B). The overexpression of SRX1 also significantly reversed the inhibition over SOD activity induced by LPS (Fig. 4C). The expression levels of PRDX1, PRDX6, TXNRD1 and SOD2 were significantly downregulated in LPS-stimulated cells compared with the control group; however, a significant upregulation in the expression levels of these antioxidative proteins was observed in cells transfected with the Ov-SRX1 plasmid and stimulated with LPS compared with LPS treatment alone (Fig. 4D). Altogether, these findings suggested that SRX1 may exert an antioxidative role in injured PC12 cells.

NRF2 controls the expression levels of downstream target genes and SRX1

A previous study indicated that SRX1 exerted an antioxidative role, which relied on the activation of NRF2. SRX1 was discovered to protect neurons from ischemia/reperfusion-induced oxidative stress injury, which was regulated by NRF2 (31). The expression levels of NRF2 and its downstream target proteins NQO1 and HO-1 were all significantly downregulated in PC12 cells exposed to LPS compared with the control group (Fig. 5A), suggesting that NRF2 may participate in the neuronal damage triggered by LPS.

Subsequently, the NRF2 inducer, TBHQ, and inhibitor, ML385, were used to confirm the regulatory effects of NRF2 on SRX1. The mRNA expression levels of SRX1, NQO1 and HO-1 were then analyzed using RT-qPCR; TBHQ significantly reversed the suppressive effects of LPS on the expression levels of SRX1, NQO1 and HO-1, while ML385 enhanced the inhibitory effects of LPS treatment (Fig. 5B). In addition, the protein expression levels of SRX1, NQO1 and HO-1, as well as nuclear NRF2, were significantly upregulated following the co-treatment of TBHQ and LPS compared to LPS treatment alone (Fig. 5C). However, ML385 treatment further downregulated the protein expression levels of SRX1, NQO1, HO-1 and NRF2 in PC12 cells exposed to LPS compared with LPS treatment alone. Overall, these findings suggested that the alleviation of the inflammatory response and oxidative stress by SRX1 may be modulated by NRF2.

Discussion

SCI is the most serious complication of spinal injury and often leads to severe dysfunction of the limb below the injury segment, which brings devastating physical and psychological harm to patients and imposes huge economic burdens to society (32). Currently, there are >1,000,000 patients with SCI in the United States and >12,000 new cases annually (33). Therefore, it remains an urgent requirement to determine effective therapeutic strategies for SCI. Inflammation and oxidative stress in neuron-like cells are commonly considered pathological alterations of SCI (10,11). However, the precise regulatory mechanisms of inflammation and oxidative stress in SCI remain poorly understood.

In the present study, SCI model rats were established by surgery in an aseptic environment. A large amount of cavities and decreased Nissl bodies were observed in the injured spinal cords of the model group compared with the sham group. In addition, the upregulated expression levels of SRX1 identified in the spinal cord tissues were of great significance in the present study. A previous study illustrated that SRX1 protected against cardiomyocyte injury upon simulated ischemia/reperfusion by inhibiting mitochondrial apoptosis (19). SRX1 was also discovered to relieve apoptosis and oxidative stress in primary rat cortical astrocytes stimulated by oxygen-glucose deprivation or H2O2, which involved the activation of the mitochondrial apoptotic pathway (34). However, to the best of our knowledge, the definite role of SRX1 in SCI has remained elusive.

LPS, which is composed of lipids and polysaccharides, is a constituent of the outer cell wall of Gram-negative bacteria and typically induces oxidative stress and an inflammatory response in cells (35,36). In the present study, the mRNA and protein expression levels of SRX1 were downregulated in PC12 cells exposed to 0.625–5 µg/ml LPS in a dose-dependent manner. Therefore, to determine the role of SRX1 in SCI, SRX1 was overexpressed in PC12 cells. The secretion of the inflammatory cytokines TNF-α, IL-1β and IL-18 was reduced to differing degrees following the transfection of Ov-SRX1 and LPS stimulation, while the levels of IL-10 demonstrated the opposite trend.

In addition, the levels of oxidative stress were analyzed in PC12 cells transfected with Ov-SRX1 and stimulated with LPS; ROS production and MDA content were both inhibited following the overexpression of SRX1 and stimulation of LPS, whereas SOD activity was increased. ROS scavenging heavily relies on manganese-dependent SOD, a mitochondrial enzyme encoded by the SOD2 gene (37). PRDX1 and PRDX6 are members of the PRDX family that reduce the oxidative load and remove ROS (38,39), thus serving an important role in the antioxidant processes. In addition, TXNRD1 is an antioxidant enzyme, which is involved in antioxidant defense and redox regulation (40). SOD, PRDXs, glutathione and catalase are major ROS detoxification enzymes (14). In the current study, the production of glutathione and catalase was not investigated, thus future work should aim to investigate this. NRF2 is known to stimulate anti-inflammatory effects and redox homeostasis, conferring resistance to oxidative damage induced by exogenous chemicals and thereby promoting cell survival (41). SRX1 was identified as a pivotal NRF2-regulated gene responsible for the defense against oxidative injury in the lung induced by cigarette smoke (15). Another study demonstrated that SRX1 protected human cardiac stem/progenitor cells against oxidative stress-induced apoptosis through the activation of the ERK/NRF2 signaling pathway (18). NRF2 has also been reported to activate the antioxidant response element-dependent gene expression of HO-1, NQO1 and SOD through evading Kelch-like ECH-associated protein 1 (KEAP1)-mediated ubiquitination-proteasomal degradation and subsequent nuclear translocation (42). Thus, the precise regulatory mechanism between SRX1 and NRF2 in damaged PC12 cells was investigated in the present study. The results indicated that LPS reduced the expression levels of NRF2 in the nucleus, as well as the transcription and translation of the downstream target genes, NQO1 and HO-1. Conversely, the activation of NRF2 with TBHQ reduced the impact of LPS, while the inhibition of NRF2 with ML385 aggravated the effect caused by LPS, suggesting that the neuroprotective effect of SRX1 on PC12 cells challenged by LPS may depend on NRF2.

In conclusion, the findings of the present study discovered that the expression levels of SRX1 were downregulated in the injured spinal cord of rats. In addition, the data suggested that SRX1 may alleviate the inflammatory response and oxidative stress in neuron-like cells, which was dependent on the nuclear translocation of NRF2, providing a novel therapeutic target for nervous system damage. Neuron-like PC12 cells are commonly used to investigate neuronal damage resulting from spinal cord injury (2224,43,44); however, primary spinal neurons derived from the rat spinal cord may be more appropriate for the study of SCI and should be used in further investigations. Moreover, the influence of SRX1-related drugs on the SCI model rats and the influence of the ubiquitination of KEAP1 on NRF2 activation should be further studied to determine the functional recovery of the rats. Further studies addressing these limitations will help to validate the conclusions of the present study.

Supplementary Material

Supporting Data

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

ZW and ZL made substantial contributions to conception and design of the study, JO and XS analyzed and interpreted data, and JL conducted the experiments and drafted the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All experiments were approved by the Institutional Animal Ethics Committees of University of South China (Hengyang, China) and were performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Fakhoury M: Spinal cord injury: Overview of experimental approaches used to restore locomotor activity. Rev Neurosci. 26:397–405. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Scivoletto G, Miscusi M, Forcato S, Ricciardi L, Serrao M, Bellitti R and Raco A: The rehabilitation of spinal cord injury patients in Europe. Acta Neurochir Suppl. 124:203–210. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Friedli L, Rosenzweig ES, Barraud Q, Schubert M, Dominici N, Awai L, Nielson JL, Musienko P, Nout-Lomas Y, Zhong H, et al: Pronounced species divergence in corticospinal tract reorganization and functional recovery after lateralized spinal cord injury favors primates. Sci Transl Med. 7:302ra1342015. View Article : Google Scholar : PubMed/NCBI

4 

Cramer SC, Lastra L, Lacourse MG and Cohen MJ: Brain motor system function after chronic, complete spinal cord injury. Brain. 128:2941–2950. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Sekhon LH and Fehlings MG: Epidemiology, demographics, and pathophysiology of acute spinal cord injury. Spine (Phila Pa 1976). 26 (Suppl 24):S2–S12. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Beattie MS: Inflammation and apoptosis: Linked therapeutic targets in spinal cord injury. Trends Mol Med. 10:580–583. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Lin XY, Lai BQ, Zeng X, Che MT, Ling EA, Wu W and Zeng YS: Cell Transplantation and neuroengineering approach for spinal cord injury treatment: A summary of current laboratory findings and review of literature. Cell Transplant. 25:1425–1438. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Ordikhani F, Sheth S and Zustiak SP: Polymeric particle-mediated molecular therapies to treat spinal cord injury. Int J Pharm. 516:71–81. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Silva NA, Sousa N, Reis RL and Salgado AJ: From basics to clinical: A comprehensive review on spinal cord injury. Prog Neurobiol. 114:25–57. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Chen S, Ye J, Chen X, Shi J, Wu W, Lin W, Lin W, Li Y, Fu H and Li S: Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-κB pathway dependent of HDAC3. J Neuroinflammation. 15:1502018. View Article : Google Scholar : PubMed/NCBI

11 

Lin X, Zhu J, Ni H, Rui Q, Sha W, Yang H, Li D and Chen G: Treatment with 2-BFI attenuated spinal cord injury by inhibiting oxidative stress and neuronal apoptosis via the Nrf2 signaling pathway. Front Cell Neurosci. 13:5672019. View Article : Google Scholar : PubMed/NCBI

12 

Ying X, Tu W, Li S, Wu Q, Chen X, Zhou Y, Hu J, Yang G and Jiang S: Hyperbaric oxygen therapy reduces apoptosis and dendritic/synaptic degeneration via the BDNF/TrkB signaling pathways in SCI rats. Life Sci. 229:187–199. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Zanuzzi CN, Nishida F, Sisti MS, Barbeito CG and Portiansky EL: Reactivity of microglia and astrocytes after an excitotoxic injury induced by kainic acid in the rat spinal cord. Tissue Cell. 56:31–40. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Findlay VJ, Tapiero H and Townsend DM: Sulfiredoxin: A potential therapeutic agent? Biomed Pharmacother. 59:374–379. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Singh A, Ling G, Suhasini AN, Zhang P, Yamamoto M, Navas-Acien A, Cosgrove G, Tuder RM, Kensler TW, Watson WH and Biswal S: Nrf2-dependent sulfiredoxin-1 expression protects against cigarette smoke-induced oxidative stress in lungs. Free Radic Biol Med. 46:376–386. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Biteau B, Labarre J and Toledano MB: ATP-dependent reduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin. Nature. 425:980–984. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Li Q, Yu S, Wu J, Zou Y and Zhao Y: Sulfiredoxin-1 protects PC12 cells against oxidative stress induced by hydrogen peroxide. J Neurosci Res. 91:861–870. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Li X, He P, Wang XL, Zhang S, Devejian N, Bennett E and Cai C: Sulfiredoxin-1 enhances cardiac progenitor cell survival against oxidative stress via the upregulation of the ERK/NRF2 signal pathway. Free Radic Biol Med. 123:8–19. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Zhang J, He Z, Guo J, Li Z, Wang X, Yang C and Cui X: Sulfiredoxin-1 protects against simulated ischaemia/reperfusion injury in cardiomyocyte by inhibiting PI3K/AKT-regulated mitochondrial apoptotic pathways. Biosci Rep. 36:e003252016. View Article : Google Scholar : PubMed/NCBI

20 

Soubeyrand M, Badner A, Vawda R, Chung YS and Fehlings MG: Very high resolution ultrasound imaging for real-time quantitative visualization of vascular disruption after spinal cord injury. J Neurotrauma. 31:1767–1775. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Can H, Aydoseli A, Gömleksiz C, Göker B, Altunrende ME, Dolgun M and Sencer A: Combined and individual use of pancaspase inhibitor Q-VD-OPh and NMDA receptor antagonist riluzole in experimental spinal cord injury. Ulus Travma Acil Cerrahi Derg. 23:452–458. 2017.PubMed/NCBI

22 

Goldshmit Y, Tang J, Siegel AL, Nguyen PD, Kaslin J, Currie PD and Jusuf PR: Different Fgfs have distinct roles in regulating neurogenesis after spinal cord injury in zebrafish. Neural Dev. 13:242018. View Article : Google Scholar : PubMed/NCBI

23 

Zhong ZX, Feng SS, Chen SZ, Chen ZM and Chen XW: Inhibition of MSK1 promotes inflammation and apoptosis and inhibits functional recovery after spinal cord injury. J Mol Neurosci. 68:191–203. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Shen LM, Song ZW, Hua Y, Chao X and Liu JB: miR-181d-5p promotes neurite outgrowth in PC12 Cells via PI3K/Akt pathway. CNS Neurosci Ther. 23:894–906. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Martin TF and Grishanin RN: PC12 cells as a model for studies of regulated secretion in neuronal and endocrine cells. Methods Cell Biol. 71:267–286. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Khodagholi F and Tusi SK: Stabilization of Nrf2 by tBHQ prevents LPS-induced apoptosis in differentiated PC12 cells. Mol Cell Biochem. 354:97–112. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Gallorini M, Petzel C, Bolay C, Hiller KA, Cataldi A, Buchalla W, Krifka S and Schweikl H: Activation of the Nrf2-regulated antioxidant cell response inhibits HEMA-induced oxidative stress and supports cell viability. Biomaterials. 56:114–128. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Singh A, Venkannagari S, Oh KH, Zhang YQ, Rohde JM, Liu L, Nimmagadda S, Sudini K, Brimacombe KR, Gajghate S, et al: Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in KEAP1-deficient NSCLC tumors. ACS Chem Biol. 11:3214–3225. 2016. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Mittal M, Siddiqui MR, Tran K, Reddy SP and Malik AB: Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 20:1126–1167. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Wu J, Chen Y, Yu S, Li L, Zhao X, Li Q, Zhao J and Zhao Y: Neuroprotective effects of sulfiredoxin-1 during cerebral ischemia/reperfusion oxidative stress injury in rats. Brain Res Bull. 132:99–108. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Li X, Zhan J, Hou Y, Hou Y, Chen S, Luo D, Luan J, Wang L and Lin D: Coenzyme Q10 regulation of apoptosis and oxidative stress in H2O2 Induced BMSC death by modulating the Nrf-2/NQO-1 signaling pathway and its application in a model of spinal cord injury. Oxid Med Cell Longev. 2019:64930812019. View Article : Google Scholar : PubMed/NCBI

33 

Hachem LD, Ahuja CS and Fehlings MG: Assessment and management of acute spinal cord injury: From point of injury to rehabilitation. J Spinal Cord Med. 40:665–675. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Zhou Y, Zhou Y, Yu S, Wu J, Chen Y and Zhao Y: Sulfiredoxin-1 exerts anti-apoptotic and neuroprotective effects against oxidative stress-induced injury in rat cortical astrocytes following exposure to oxygen-glucose deprivation and hydrogen peroxide. Int J Mol Med. 36:43–52. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Lee JY, Joo B, Nam JH, Nam HY, Lee W, Nam Y, Seo Y, Kang HJ, Cho HJ, Jang YP, et al: An aqueous extract of herbal medicine ALWPs enhances cognitive performance and inhibits LPS-induced neuroinflammation via FAK/NF-κb signaling pathways. Front Aging Neurosci. 10:2692018. View Article : Google Scholar : PubMed/NCBI

36 

Shah SA, Khan M, Jo MH, Jo MG, Amin FU and Kim MO: Melatonin stimulates the SIRT1/Nrf2 signaling pathway counteracting lipopolysaccharide (LPS)-induced oxidative stress to rescue postnatal rat brain. CNS Neurosci Ther. 23:33–44. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Ashtekar A, Huk D, Magner A, La Perle KMD, Boucai L and Kirschner LS: Alterations in Sod2-induced oxidative stress affect endocrine cancer progression. J Clin Endocrinol Metab. 103:4135–4145. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Kubo E, Chhunchha B, Singh P, Sasaki H and Singh DP: Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci Rep. 7:141302017. View Article : Google Scholar : PubMed/NCBI

39 

Ding C, Fan X and Wu G: Peroxiredoxin 1-an antioxidant enzyme in cancer. J Cell Mol Med. 21:193–202. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Tuo L, Xiang J, Pan X, Gao Q, Zhang G, Yang Y, Liang L, Xia J, Wang K and Tang N: PCK1 downregulation promotes TXNRD1 expression and hepatoma cell growth via the Nrf2/Keap1 pathway. Front Oncol. 8:6112018. View Article : Google Scholar : PubMed/NCBI

41 

Wu S, Lu H and Bai Y: Nrf2 in cancers: A double-edged sword. Cancer Med. 8:2252–2267. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Magesh S, Chen Y and Hu L: Small molecule modulators of Keap1-Nrf2-are pathway as potential preventive and therapeutic agents. Med Res Rev. 32:687–726. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Lin CW, Chen B, Huang KL, Dai YS and Teng HL: Inhibition of autophagy by estradiol promotes locomotor recovery after spinal cord injury in rats. Neurosci Bull. 32:137–144. 2016. View Article : Google Scholar : PubMed/NCBI

44 

He Z, Zhou Y, Huang Y, Wang Q, Zheng B, Zhang H, Li J, Liu Y, Wu F, Zhang X, et al: Dl-3-n-butylphthalide improves functional recovery in rats with spinal cord injury by inhibiting endoplasmic reticulum stress-induced apoptosis. Am J Transl Res. 9:1075–1087. 2017.PubMed/NCBI

Related Articles

Journal Cover

December-2020
Volume 22 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu Z, Lu Z, Ou J, Su X and Liu J: Inflammatory response and oxidative stress attenuated by sulfiredoxin‑1 in neuron‑like cells depends on nuclear factor erythroid‑2‑related factor 2. Mol Med Rep 22: 4734-4742, 2020
APA
Wu, Z., Lu, Z., Ou, J., Su, X., & Liu, J. (2020). Inflammatory response and oxidative stress attenuated by sulfiredoxin‑1 in neuron‑like cells depends on nuclear factor erythroid‑2‑related factor 2. Molecular Medicine Reports, 22, 4734-4742. https://doi.org/10.3892/mmr.2020.11545
MLA
Wu, Z., Lu, Z., Ou, J., Su, X., Liu, J."Inflammatory response and oxidative stress attenuated by sulfiredoxin‑1 in neuron‑like cells depends on nuclear factor erythroid‑2‑related factor 2". Molecular Medicine Reports 22.6 (2020): 4734-4742.
Chicago
Wu, Z., Lu, Z., Ou, J., Su, X., Liu, J."Inflammatory response and oxidative stress attenuated by sulfiredoxin‑1 in neuron‑like cells depends on nuclear factor erythroid‑2‑related factor 2". Molecular Medicine Reports 22, no. 6 (2020): 4734-4742. https://doi.org/10.3892/mmr.2020.11545